1
|
Song Y, Li Y, Zhou J, Yu J, Hu Q, Yang F, Yin Z, Wang Y, Wang Y, Zhang X, Tao Y, Peng C, Liu S. IL-8-NF-κB-ALDH1A1 loop promotes the progression of intrahepatic cholangiocarcinoma. Hepatol Commun 2025; 9:e0664. [PMID: 40008905 PMCID: PMC11868433 DOI: 10.1097/hc9.0000000000000664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/02/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is a poor prognosis of malignant cancer with high lymph node metastasis and resistance to systemic therapies. Recent studies suggested that the involvement of IL-8 could promote ICC metastasis through epithelial-mesenchymal transition while the ICC-ALDH1A1high subtype is clarified by multi-omics study. The correlation between ALDH1A1 and IL-8 in ICC remains elusive. This study aims to further explore the roles and regulatory mechanisms of ALDH1A1 and IL-8 in ICC. METHODS We analyzed IL-8 and ALDH1A1 expression in ICC patients and cells. CXCR2 inhibitor (SB225002) was applied to inhibit the function of IL-8, and JSH-23 was applied to inhibit the NF-κB signaling pathway. We examined the effects of IL-8 inhibition on NF-κB, ALDH1A1 expression, and cell growth, migration, invasion, and stemness. Moreover, we examined the effects of ALDH1A1 on NF-κB, IL-8 expression, and cell growth, migration, invasion, and stemness. The effects of IL-8 and ALDH1A1 on tumor growth and NF-κB expression were validated using subcutaneous tumors in nude mice. RESULTS IL-8-derived tumor cells could promote ICC progression. The high expression of IL-8 in serum was associated with lymph node metastasis. IL-8 could upregulate ALDH1A1 expression by activating the NF-κB signaling pathway, promoting tumor progression. Upregulation of ALDH1A1 could activate NF-κB to promote IL-8 secretion, forming a positive feedback loop to promote tumor invasiveness and cell stemness in ICC. CONCLUSIONS IL-8-derived tumor cells could upregulate ALDH1A1 expression by activating the NF-κB signaling pathway, promoting tumor progression. Upregulation of ALDH1A1 could activate NF-κB to promote IL-8 secretion, forming a positive feedback loop to promote tumor invasiveness and cell stemness in ICC.
Collapse
Affiliation(s)
- Yinghui Song
- Central Laboratory, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Yufeng Li
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Jia Zhou
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Jianfeng Yu
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Qianwei Hu
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Feicheng Yang
- Department of Pathology, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Zexi Yin
- Department of Pediatrics, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Yizhi Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Yueren Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Xinling Zhang
- Central Laboratory, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Yuewei Tao
- School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Chuang Peng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| | - Sulai Liu
- Central Laboratory, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P. R. China
| |
Collapse
|
2
|
Kucheryavenko AS, Muzyko EA, Perfilova VN, Kaplanov KD, Frolov MY. The role of the PPM1D gene in tumor pathogenesis. BIOMEDITSINSKAIA KHIMIIA 2025; 71:19-28. [PMID: 40045720 DOI: 10.18097/pbmcr1495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The PPM1D gene and its protein product (serine-threonine protein phosphatase, PPM1D or Wip1) are involved in regulation of cell's DNA damage response, cell cycle control, and repair. Amplification, overexpression, or mutations of the PPM1D gene have a significant impact on cell responses to stress factors and genetic instability as well as impairments of processes of double-strand break repair, nucleotide excision repair, base excision repair, cell cycle, and apoptosis. PPM1D dephosphorylates and thus inactivates p53, proteins that respond to DNA strand integrity damage, cell cycle checkpoint proteins, and apoptotic proteins. This contributes to tumor development, growth, and maintenance of the tumor phenotype. In this review we consider data on the role of the PPM1D gene in the formation and maintenance of various oncological processes, including tumors of the mammary glands, ovaries, prostate gland, esophagus, stomach, intestines, liver and pancreas, hemoblastoses, and others.
Collapse
Affiliation(s)
| | - E A Muzyko
- Volgograd State Medical University, Volgograd, Russia
| | - V N Perfilova
- Volgograd State Medical University, Volgograd, Russia; Volgograd Medical Research Center, Volgograd, Russia
| | | | - M Yu Frolov
- Volgograd State Medical University, Volgograd, Russia
| |
Collapse
|
3
|
Liu Y, Xia F, Zhu C, Song J, Tang B, Zhang B, Huang Z. Protein serine/threonine phosphatases in tumor microenvironment: a vital player and a promising therapeutic target. Theranostics 2025; 15:1164-1184. [PMID: 39776803 PMCID: PMC11700861 DOI: 10.7150/thno.104529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
The tumor microenvironment (TME) is involved in cancer initiation and progression. With advances in the TME field, numerous therapeutic approaches, such as antiangiogenic treatment and immune checkpoint inhibitors, have been inspired and developed. Nevertheless, the sophisticated regulatory effects on the biological balance of the TME remain unclear. Decoding the pathological features of the TME is urgently needed to understand the tumor ecosystem and develop novel antitumor treatments. Protein serine/threonine phosphatases (PSPs) are responsible for inverse protein phosphorylation processes. Aberrant expression and dysfunction of PSPs disturb cellular homeostasis, reprogram metabolic processes and reshape the immune landscape, thereby contributing to cancer progression. Some therapeutic implications, such as the use of PSPs as targets, have drawn the attention of researchers and clinicians. To date, the effects of PSP inhibitors are less satisfactory in real-world practice. With breakthroughs in sequencing technologies, scientists can decipher TME investigations via multiomics and higher resolution. These benefits provide an opportunity to explore the TME in a more comprehensive manner and inspire more findings concerning PSPs in the TME. The current review starts by introducing the canonical knowledge of PSPs, including their members, structures and posttranslational modifications for activities. We then summarize the functions of PSPs in regulating cellular homeostasis. In particular, we specified the up-to-date roles of PSPs in modulating the immune microenvironment, adopting hypoxia, reprogramming metabolic processes, and responding to extracellular matrix remodeling. Finally, we introduce preclinical PSP inhibitors with translational value and conclude with clinical trials of PSP inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Yiyang Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Xia
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Song
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; Key Laboratory of Organ Transplantation, National Health Commission; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Hsu YC, Chiu YC, Lu TP, Hsiao TH, Chen Y. Predicting drug response through tumor deconvolution by cancer cell lines. PATTERNS (NEW YORK, N.Y.) 2024; 5:100949. [PMID: 38645769 PMCID: PMC11026976 DOI: 10.1016/j.patter.2024.100949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 04/23/2024]
Abstract
Large-scale cancer drug sensitivity data have become available for a collection of cancer cell lines, but only limited drug response data from patients are available. Bridging the gap in pharmacogenomics knowledge between in vitro and in vivo datasets remains challenging. In this study, we trained a deep learning model, Scaden-CA, for deconvoluting tumor data into proportions of cancer-type-specific cell lines. Then, we developed a drug response prediction method using the deconvoluted proportions and the drug sensitivity data from cell lines. The Scaden-CA model showed excellent performance in terms of concordance correlation coefficients (>0.9 for model testing) and the correctly deconvoluted rate (>70% across most cancers) for model validation using Cancer Cell Line Encyclopedia (CCLE) bulk RNA data. We applied the model to tumors in The Cancer Genome Atlas (TCGA) dataset and examined associations between predicted cell viability and mutation status or gene expression levels to understand underlying mechanisms of potential value for drug repurposing.
Collapse
Affiliation(s)
- Yu-Ching Hsu
- Bioinformatics Program, Taiwan International Graduate Program, National Taiwan University, Taipei 115, Taiwan
- Bioinformatics Program, Institute of Statistical Science, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan
- Institute of Health Data Analytics and Statistics, Department of Public Health, College of Public Health, National Taiwan University, Taipei 100, Taiwan
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Yu-Chiao Chiu
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Tzu-Pin Lu
- Institute of Health Data Analytics and Statistics, Department of Public Health, College of Public Health, National Taiwan University, Taipei 100, Taiwan
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
5
|
Wang T, Li X, Liao G, Wang Z, Han X, Gu J, Mu X, Qiu J, Qian Y. AFB1 Triggers Lipid Metabolism Disorders through the PI3K/Akt Pathway and Mediates Apoptosis Leading to Hepatotoxicity. Foods 2024; 13:163. [PMID: 38201191 PMCID: PMC10778638 DOI: 10.3390/foods13010163] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
As the most prevalent mycotoxin in agricultural products, aflatoxin B1 not only causes significant economic losses but also poses a substantial threat to human and animal health. AFB1 has been shown to increase the risk of hepatocellular carcinoma (HCC) but the underlying mechanism is not thoroughly researched. Here, we explored the toxicity mechanism of AFB1 on human hepatocytes following low-dose exposure based on transcriptomics and lipidomics. Apoptosis-related pathways were significantly upregulated after AFB1 exposure in all three hES-Hep, HepaRG, and HepG2 hepatogenic cell lines. By conducting a comparative analysis with the TCGA-LIHC database, four biomarkers (MTCH1, PPM1D, TP53I3, and UBC) shared by AFB1 and HCC were identified (hazard ratio > 1), which can be used to monitor the degree of AFB1-induced hepatotoxicity. Simultaneously, AFB1 induced abnormal metabolism of glycerolipids, sphingolipids, and glycerophospholipids in HepG2 cells (FDR < 0.05, impact > 0.1). Furthermore, combined analysis revealed strong regulatory effects between PIK3R1 and sphingolipids (correlation coefficient > 0.9), suggesting potential mediation by the phosphatidylinositol 3 kinase (PI3K) /protein kinase B (AKT) signaling pathway within mitochondria. This study revealed the dysregulation of lipid metabolism induced by AFB1 and found novel target genes associated with AFB-induced HCC development, providing reliable evidence for elucidating the hepatotoxicity of AFB as well as assessing food safety risks.
Collapse
Affiliation(s)
- Tiancai Wang
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.W.); (X.L.); (G.L.); (Z.W.); (J.G.); (X.M.); (J.Q.)
- Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiabing Li
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.W.); (X.L.); (G.L.); (Z.W.); (J.G.); (X.M.); (J.Q.)
- Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Guangqin Liao
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.W.); (X.L.); (G.L.); (Z.W.); (J.G.); (X.M.); (J.Q.)
- Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zishuang Wang
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.W.); (X.L.); (G.L.); (Z.W.); (J.G.); (X.M.); (J.Q.)
- Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiaoxu Han
- National Center of Technology Innovation for Dairy, Hohhot 010100, China;
| | - Jingyi Gu
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.W.); (X.L.); (G.L.); (Z.W.); (J.G.); (X.M.); (J.Q.)
- Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiyan Mu
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.W.); (X.L.); (G.L.); (Z.W.); (J.G.); (X.M.); (J.Q.)
- Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Jing Qiu
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.W.); (X.L.); (G.L.); (Z.W.); (J.G.); (X.M.); (J.Q.)
- Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Yongzhong Qian
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.W.); (X.L.); (G.L.); (Z.W.); (J.G.); (X.M.); (J.Q.)
- Key Laboratory of Agri-Food Quality and Safety, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
6
|
Shen J, Wang Q, Mao Y, Gao W, Duan S. Targeting the p53 signaling pathway in cancers: Molecular mechanisms and clinical studies. MedComm (Beijing) 2023; 4:e288. [PMID: 37256211 PMCID: PMC10225743 DOI: 10.1002/mco2.288] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 06/01/2023] Open
Abstract
Tumor suppressor p53 can transcriptionally activate downstream genes in response to stress, and then regulate the cell cycle, DNA repair, metabolism, angiogenesis, apoptosis, and other biological responses. p53 has seven functional domains and 12 splice isoforms, and different domains and subtypes play different roles. The activation and inactivation of p53 are finely regulated and are associated with phosphorylation/acetylation modification and ubiquitination modification, respectively. Abnormal activation of p53 is closely related to the occurrence and development of cancer. While targeted therapy of the p53 signaling pathway is still in its early stages and only a few drugs or treatments have entered clinical trials, the development of new drugs and ongoing clinical trials are expected to lead to the widespread use of p53 signaling-targeted therapy in cancer treatment in the future. TRIAP1 is a novel p53 downstream inhibitor of apoptosis. TRIAP1 is the homolog of yeast mitochondrial intermembrane protein MDM35, which can play a tumor-promoting role by blocking the mitochondria-dependent apoptosis pathway. This work provides a systematic overview of recent basic research and clinical progress in the p53 signaling pathway and proposes that TRIAP1 is an important therapeutic target downstream of p53 signaling.
Collapse
Affiliation(s)
- Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouZhejiangChina
| | - Qurui Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouZhejiangChina
| | - Yunan Mao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouZhejiangChina
| | - Wei Gao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouZhejiangChina
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouZhejiangChina
| |
Collapse
|
7
|
Qi Y, Song Y, Cai M, Li J, Yu Z, Li Y, Huang J, Jiang Y, Peng C, Jiang B, Liu S. Vascular endothelial growth factor A is a potential prognostic biomarker and correlates with immune cell infiltration in hepatocellular carcinoma. J Cell Mol Med 2023; 27:538-552. [PMID: 36729917 PMCID: PMC9930434 DOI: 10.1111/jcmm.17678] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/30/2022] [Accepted: 01/06/2023] [Indexed: 02/03/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related deaths among cancer patients. Vascular endothelial growth factor A (VEGFA) is involved in regulating biological processes, such as angiogenesis and vascular permeability, and is very closely related to the pathogenesis of various tumours, especially vascular-rich, solid tumours. Clinical data of patients with HCC and other tumours were analysed through public databases, such as the TCGA database, Gene Expression Omnibus database, Human Protein Atlas database, STRING, Tumour Immune Estimation Resource and Kaplan-Meier Plotter. The tumour tissues and adjacent normal tissues of patients with HCC from Hunan Provincial People's Hospital were collected to verify the expression of VEGFA by immunohistochemistry, immunofluorescence, Western blotting and qPCR. VEGFA expression is elevated in multiple tumour types and correlates with the prognosis of tumour patients. VEGFA is involved in regulating the tumour microenvironment and immune cell function in tumour development. Inhibition of VEGFA reduces proliferation, invasion, and migration and promotes apoptosis in HCC cells. VEGFA is a potential predictive biomarker for the diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- Yuchen Qi
- Department of Hepatobiliary SurgeryHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
- Department of CardiologyXiangdong Hospital Affiliated to Hunan Normal UniversityLilingChina
- Central Laboratory of Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Yinghui Song
- Department of Hepatobiliary SurgeryHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
- Central Laboratory of Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Mengting Cai
- Department of Nuclear MedicineHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Jianwen Li
- Department of CardiologyXiangdong Hospital Affiliated to Hunan Normal UniversityLilingChina
| | - Zhangtao Yu
- Department of Hepatobiliary SurgeryHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Yuhang Li
- Department of Hepatobiliary SurgeryHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Junkai Huang
- Department of Hepatobiliary SurgeryHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Yu Jiang
- Institute of Emergency Medicine/Hunan Provincial Key Laboratory of Emergency and Critical Care MetabonomicsHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Chuang Peng
- Department of Hepatobiliary SurgeryHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Bo Jiang
- Department of Hepatobiliary SurgeryHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Sulai Liu
- Department of Hepatobiliary SurgeryHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
- Central Laboratory of Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| |
Collapse
|
8
|
Dai W, Wang J, Wang Z, Xiao Y, Li J, Hong L, Pei M, Zhang J, Yang P, Wu X, Tang W, Jiang X, Jiang P, Xiang L, Li A, Lin J, Liu S, Wang J. Comprehensive Analysis of the Prognostic Values of the TRIM Family in Hepatocellular Carcinoma. Front Oncol 2022; 11:767644. [PMID: 35004288 PMCID: PMC8733586 DOI: 10.3389/fonc.2021.767644] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/30/2021] [Indexed: 12/13/2022] Open
Abstract
Background Accumulating studies have demonstrated the abnormal expressions and prognostic values of certain members of the tripartite motif (TRIM) family in diverse cancers. However, comprehensive prognostic values of the TRIM family in hepatocellular carcinoma (HCC) are yet to be clearly defined. Methods The prognostic values of the TRIM family were evaluated by survival analysis and univariate Cox regression analysis based on gene expression data and clinical data of HCC from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The expression profiles, protein–protein interaction among the TRIM family, prediction of transcription factors (TFs) or miRNAs, genetic alterations, correlations with the hallmarks of cancer and immune infiltrates, and pathway enrichment analysis were explored by multiple public databases. Further, a TRIM family gene-based signature for predicting overall survival (OS) in HCC was built by using the least absolute shrinkage and selection operator (LASSO) regression. TCGA–Liver Hepatocellular Carcinoma (LIHC) cohort was used as the training set, and GSE76427 was used for external validation. Time-dependent receiver operating characteristic (ROC) and survival analysis were used to estimate the signature. Finally, a nomogram combining the TRIM family risk score and clinical parameters was established. Results High expressions of TRIM family members including TRIM3, TRIM5, MID1, TRIM21, TRIM27, TRIM32, TRIM44, TRIM47, and TRIM72 were significantly associated with HCC patients’ poor OS. A novel TRIM family gene-based signature (including TRIM5, MID1, TRIM21, TRIM32, TRIM44, and TRIM47) was built for OS prediction in HCC. ROC curves suggested the signature’s good performance in OS prediction. HCC patients in the high-risk group had poorer OS than the low-risk patients based on the signature. A nomogram integrating the TRIM family risk score, age, and TNM stage was established. The ROC curves suggested that the signature presented better discrimination than the similar model without the TRIM family risk score. Conclusion Our study identified the potential application values of the TRIM family for outcome prediction in HCC.
Collapse
Affiliation(s)
- Weiyu Dai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhi Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yizhi Xiao
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaying Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Linjie Hong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Miaomiao Pei
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jieming Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Yang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaosheng Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weimei Tang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoling Jiang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Jiang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Xiang
- Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianjiao Lin
- Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, China
| | - Jide Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Gastroenterology, Longgang District People's Hospital, Shenzhen, China
| |
Collapse
|