1
|
Zhou Y, Wei S, Xu M, Wu X, Dou W, Li H, Zhang Z, Zhang S. CAR-T cell therapy for hepatocellular carcinoma: current trends and challenges. Front Immunol 2024; 15:1489649. [PMID: 39569202 PMCID: PMC11576447 DOI: 10.3389/fimmu.2024.1489649] [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: 09/01/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent cancers worldwide, highlighting the urgent need for improved diagnostic and therapeutic methodologies. The standard treatment regimen generally involves surgical intervention followed by systemic therapies; however, the median survival rates for patients remain unsatisfactory. Chimeric antigen receptor (CAR) T-cell therapy has emerged as a pivotal advancement in cancer treatment. Both clinical and preclinical studies emphasize the notable efficacy of CAR T cells in targeting HCC. Various molecules, such as GPC3, c-Met, and NKG2D, show significant promise as potential immunotherapeutic targets in liver cancer. Despite this, employing CAR T cells to treat solid tumors like HCC poses considerable challenges within the discipline. Numerous innovations have significant potential to enhance the efficacy of CAR T-cell therapy for HCC, including improvements in T cell trafficking, strategies to counteract the immunosuppressive tumor microenvironment, and enhanced safety protocols. Ongoing efforts to discover therapeutic targets for CAR T cells highlight the need for the development of more practical manufacturing strategies for CAR-modified cells. This review synthesizes recent findings and clinical advancements in the use of CAR T-cell therapies for HCC treatment. We elucidate the therapeutic benefits of CAR T cells in HCC and identify the primary barriers to their broader application. Our analysis aims to provide a comprehensive overview of the current status and future prospects of CAR T-cell immunotherapy for HCC.
Collapse
Affiliation(s)
- Yexin Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- The General Hospital of Western Theater Command, Chengdu, China
| | - Shanshan Wei
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Menghui Xu
- The General Hospital of Western Theater Command, Chengdu, China
| | - Xinhui Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Wenbo Dou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Huakang Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhonglin Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shuo Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Ma YH, Maekawa S, Takano S, Yamaguchi T, Ishida T, Takaoka S, Muraoka M, Komiyama Y, Takada H, Suzuki Y, Sato M, Fan J, Enomoto N. Elevated nuclear expression of ZHX1 correlates with poor prognosis in hepatocellular carcinoma (HCC): Comparison of nuclear and cytoplasmic distribution of the ZHX family in HCC cells. Hepatol Res 2024. [PMID: 39207766 DOI: 10.1111/hepr.14100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
AIM The role of the zinc fingers and homeoboxes family (ZHX1-3), transcriptional repressors, through their subcellular localization in hepatocellular carcinoma (HCC), is not fully understood. The present study aimed to examine the differential nuclear and cytoplasmic expression of ZHXs in HCC tissues. METHODS Immunohistochemistry was utilized to detect the expression of ZHXs in 54 liver tissues from HCC (n = 33), hepatitis C (n = 16), and the normal liver tissue surrounding hepatic metastasis of colorectal cancer (n = 5). Next-generation sequencing and digital polymerase chain reaction identified gene mutations associated with HCC. Kaplan-Meier curves were constructed to evaluate the relationship between ZHX expression and survival. The results were validated using data from The Cancer Genome Atlas. Univariate and multivariate Cox regression analyses were undertaken to identify independent prognostic factors. RESULTS High nuclear expression of ZHX1 was associated with poor overall survival (OS), while high nuclear expression of ZHX2 correlated with higher recurrence. Conversely, patients with high cytoplasmic expression of ZHX3 had lower recurrence and better OS. Hepatitis B virus-associated HCC was related to high cytoplasmic expression of ZHX1, which was marginally related to telomerase reverse transcriptase (TERT) promoter mutation-negative HCC. In contrast, low nuclear expression of ZHX3 was associated with TERT promoter mutation-positive HCC and HCC patients over 70 years old. CONCLUSIONS These results suggest that the expression and localization of different ZHXs may be related to HCC progression, potentially inferring genetic backgrounds such as TERT promoter mutation. Further studies on the relationship between HCC and ZHXs will enhance our understanding and control of HCC.
Collapse
Affiliation(s)
- Yu-Hong Ma
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
- Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Shinya Maekawa
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Shinichi Takano
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Tatsuya Yamaguchi
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Takeshi Ishida
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Shinya Takaoka
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Masaru Muraoka
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Yasuyuki Komiyama
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Hitomi Takada
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Yuichiro Suzuki
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Mitsuaki Sato
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Jianglin Fan
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Nobuyuki Enomoto
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| |
Collapse
|
3
|
Zhang Y, Fan Y, Hu H, Zhang X, Wang Z, Wu Z, Wang L, Yu X, Song X, Xiang P, Zhang X, Wang T, Tan S, Li C, Gao L, Liang X, Li S, Li N, Yue X, Ma C. ZHX2 emerges as a negative regulator of mitochondrial oxidative phosphorylation during acute liver injury. Nat Commun 2023; 14:7527. [PMID: 37980429 PMCID: PMC10657347 DOI: 10.1038/s41467-023-43439-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023] Open
Abstract
Mitochondria dysfunction contributes to acute liver injuries, and mitochondrial regulators, such as PGC-1α and MCJ, affect liver regeneration. Therefore, identification of mitochondrial modulators may pave the way for developing therapeutic strategies. Here, ZHX2 is identified as a mitochondrial regulator during acute liver injury. ZHX2 both transcriptionally inhibits expression of several mitochondrial electron transport chain genes and decreases PGC-1α stability, leading to reduction of mitochondrial mass and OXPHOS. Loss of Zhx2 promotes liver recovery by increasing mitochondrial OXPHOS in mice with partial hepatectomy or CCl4-induced liver injury, and inhibition of PGC-1α or electron transport chain abolishes these effects. Notably, ZHX2 expression is higher in liver tissues from patients with drug-induced liver injury and is negatively correlated with mitochondrial mass marker TOM20. Delivery of shRNA targeting Zhx2 effectively protects mice from CCl4-induced liver injury. Together, our data clarify ZHX2 as a negative regulator of mitochondrial OXPHOS and a potential target for developing strategies for improving liver recovery after acute injuries.
Collapse
Affiliation(s)
- Yankun Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Yuchen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Huili Hu
- Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaohui Zhang
- Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Zehua Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Liyuan Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Xiangguo Yu
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Xiaojia Song
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Peng Xiang
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Xiaodong Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Tixiao Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Siyu Tan
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Chunyang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China
| | - Shuijie Li
- College of Pharmacy, Harbin Medical University, Harbin, China
| | - Nailin Li
- Department of Medicine-Solna, Cardiovascular Medicine Unit, Karolinska Institute, Stockholm, Sweden
| | - Xuetian Yue
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China.
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College of Shandong University, Jinan, China.
| |
Collapse
|
4
|
Li D, Wang W, Liu B, Jin D, Wang Y, He G, Guo L, Liu W, Li Y. Characterization of circSEC11A as a novel regulator of Iodine-125 radioactive seed-induced anticancer effects in hepatocellular carcinoma via targeting ZHX2/GADD34 axis. Cell Death Discov 2023; 9:294. [PMID: 37563132 PMCID: PMC10415397 DOI: 10.1038/s41420-023-01593-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/19/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023] Open
Abstract
Iodine-125 (I-125) radioactive seed implantation is used for the local treatment of hepatocellular carcinoma (HCC), but the molecular mechanisms regulating its anticancer effects remain incompletely understood. In this study, we report that hsa_circ_0000647 (circSEC11A) is highly expressed after I-125 treatment in HCC cell lines and tissues and is a key regulator of I-125-induced anticancer effects. CircSEC11A acts as a competing endogenous RNA (ceRNA) to sponge miR-3529-3p, promoting the expression of zinc fingers and homeoboxes 2 (ZHX2) and enhancing I-125-induced anticancer effects. Dual-luciferase reporter assay, RNA pull-down, RNA immunoprecipitation, and fluorescence in situ hybridization were thereafter performed to verify the interaction among the molecules. Anticancer effects were detected using CCK-8, flow cytometry, TUNEL, EdU, transwell, and wound healing assays. Furthermore, ZHX2 transcriptionally inhibits GADD34, a negative regulator of endoplasmic reticulum stress (ERS), to enhance I-125- induced anticancer effects in vivo and in vitro. In conclusion, we characterized circSEC11A as a novel regulator of I-125-induced anticancer effects in HCC via miR-3529-3p/ZHX2/GADD34 axis-mediated ERS. Thus, circSEC11A may act as a potential therapeutic target for I-125 implantation in the clinic.
Collapse
Affiliation(s)
- Dong Li
- Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
- Institute of Interventional Oncology, Shandong University, Jinan, China
| | - Wujie Wang
- Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
- Institute of Interventional Oncology, Shandong University, Jinan, China
| | - Bin Liu
- Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
- Institute of Interventional Oncology, Shandong University, Jinan, China
| | - Die Jin
- Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
- Institute of Interventional Oncology, Shandong University, Jinan, China
| | - Yang Wang
- Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China
- Institute of Interventional Oncology, Shandong University, Jinan, China
| | - Guanghui He
- Department of Interventional Medicine, Weifang Second People's Hospital, Weifang, China
| | - Lei Guo
- Department of Vascular Anomalies and Interventional Radiology, Children's Hospital Affiliated to Shandong University, Jinan, China
| | - Wen Liu
- Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China.
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| | - Yuliang Li
- Department of Interventional Medicine, The Second Hospital of Shandong University, Jinan, China.
- Institute of Interventional Oncology, Shandong University, Jinan, China.
| |
Collapse
|
5
|
Abstract
As a transcriptional factor and the negative regulator of alpha fetal protein (AFP), Zinc fingers and homeoboxes 2 (ZHX2) has a well-established role in protection against hepatocellular carcinoma (HCC). However, recent studies have suggested ZHX2 as an oncogene in clear cell renal cell carcinoma (ccRCC) and triple-negative breast cancer (TNBC). Moreover, mounting evidence has illustrated a much broader role of ZHX2 in multiple cellular processes, including cell proliferation, cell differentiation, lipid metabolism, and immunoregulation. This comprehensive review emphasizes the role of ZHX2 in health and diseases which have been more recently uncovered.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Dept. Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education and Dept. Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Dept. Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University, Jinan, Shandong, China
| |
Collapse
|
6
|
Bao Y, Zhang H, Han Z, Guo Y, Yang W. Zinc Fingers and Homeobox Family in Cancer: A Double-Edged Sword. Int J Mol Sci 2022; 23:ijms231911167. [PMID: 36232466 PMCID: PMC9570228 DOI: 10.3390/ijms231911167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
The zinc fingers and homeobox (ZHX) family includes ZHX1, ZHX2, and ZHX3, and their proteins have similar unique structures, containing two C2H2-type zinc finger motifs and four or five HOX-like homeodomains. The members of the ZHX family can form homodimers or heterodimers with each other or with a subunit of nuclear factor Y. Previous studies have suggested that ZHXs can function as positive or negative transcriptional regulators. Recent studies have further revealed their biological functions and underlying mechanisms in cancers. This review summarized the advances of ZHX-mediated functions, including tumor-suppressive and oncogenic functions in cancer formation and progression, the molecular mechanisms, and regulatory functions, such as cancer cell proliferation, migration, invasion, and metastasis. Moreover, the differential expression levels and their association with good or poor outcomes in patients with various malignancies and differential responses to chemotherapy exert opposite functions of oncogene or tumor suppressors. Therefore, the ZHXs act as a double-edged sword in cancers.
Collapse
Affiliation(s)
- Yonghua Bao
- Department of Pathology, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Haifeng Zhang
- Department of Pathology, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Zhixue Han
- Department of Pathology, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Yongchen Guo
- Department of Immunology, Mudanjiang Medical University, Mudanjiang 157011, China
- Correspondence: (Y.G.); (W.Y.)
| | - Wancai Yang
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (Y.G.); (W.Y.)
| |
Collapse
|
7
|
You Y, Hu F, Hu S. Attenuated ZHX3 expression is predictive of poor outcome for liver cancer: Indication for personalized therapy. Oncol Lett 2022; 24:224. [PMID: 35720472 PMCID: PMC9185145 DOI: 10.3892/ol.2022.13345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/09/2022] [Indexed: 11/05/2022] Open
Abstract
The zinc-fingers and homeoboxes (ZHX) family members have been characterized as master regulators in cancer initiation and development. The present study performed in silico data-mining with publicly available datasets and immunohistochemistry to assess the expression status of ZHX factors and the corresponding prognostic implications in liver cancer. Increased ZHX3 mRNA expression was associated with favorable overall survival in patients with liver cancer. Subgroups analyses revealed a significant association between the expression of ZHX factors and outcomes in select patient cohorts. Immunohistochemical analysis supported that ZHX3 expression was an independent prognostic indicator for patient survival. These results suggested that dysregulation of ZHX factors is involved in disease progression and ZHX3 expression may serve as a prognostic biomarker for liver cancer.
Collapse
Affiliation(s)
- Yanjie You
- Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750002, P.R. China
| | - Fangrui Hu
- Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750002, P.R. China
| | - Shengjuan Hu
- Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750002, P.R. China
| |
Collapse
|
8
|
Zhao Y, Gao L, Jiang C, Chen J, Qin Z, Zhong F, Yan Y, Tong R, Zhou M, Yuan A, Pu J. The transcription factor zinc fingers and homeoboxes 2 alleviates NASH by transcriptional activation of phosphatase and tensin homolog. Hepatology 2022; 75:939-954. [PMID: 34545586 DOI: 10.1002/hep.32165] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/25/2021] [Accepted: 08/08/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS NASH, which is a common clinical condition predisposing to advanced liver diseases, has become a worldwide epidemic. A large and growing unmet therapeutic need for this condition reflects incomplete understanding of its pathogenesis. In the current study, we identified a transcription factor, zinc fingers and homeoboxes 2 (ZHX2), in hepatocytes as a protective factor against steatohepatitis. APPROACH AND RESULTS We found that hepatic ZHX2 was significantly suppressed in NASH models and steatotic hepatic cells. Hepatocyte-specific ablation of ZHX2 exacerbated NASH-related phenotypes in mice, including lipid accumulation, enhanced inflammation, and hepatic fibrosis. Conversely, hepatocyte-specific overexpression of ZHX2 significantly alleviated the progression of NASH in an experimental setting. Integrated analysis of transcriptomic profiling and chromatin immunoprecipitation sequencing data demonstrated that the phosphatase and tensin homolog (PTEN) was a target gene of ZHX2 in hepatocyte. ZHX2 bound to the promoter of PTEN gene and subsequently promoted the transcription of PTEN, which mediated the beneficial role of ZHX2 against NASH. CONCLUSIONS The current findings demonstrate a protective role of ZHX2 against NASH progression by transcriptionally activating PTEN. These findings shed light on the therapeutic potential of targeting ZHX2 for treating NASH and related metabolic disorders.
Collapse
Affiliation(s)
- Yichao Zhao
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai Cancer InstituteShanghaiChina
| | - Lingchen Gao
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai Cancer InstituteShanghaiChina
| | - Chenglin Jiang
- Graduate School of Bengbu Medical CollegeBengbuAnhuiChina
| | - Jianqing Chen
- Graduate School of Bengbu Medical CollegeBengbuAnhuiChina
| | - Zihan Qin
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai Cancer InstituteShanghaiChina
| | - Fangyuan Zhong
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai Cancer InstituteShanghaiChina
| | - Yang Yan
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai Cancer InstituteShanghaiChina
| | - Renyang Tong
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai Cancer InstituteShanghaiChina
| | - Meng Zhou
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai Cancer InstituteShanghaiChina
| | - Ancai Yuan
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai Cancer InstituteShanghaiChina
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related GenesDivision of CardiologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai Cancer InstituteShanghaiChina
| |
Collapse
|
9
|
Zheng X, Liu X, Lei Y, Wang G, Liu M. Glypican-3: A Novel and Promising Target for the Treatment of Hepatocellular Carcinoma. Front Oncol 2022; 12:824208. [PMID: 35251989 PMCID: PMC8889910 DOI: 10.3389/fonc.2022.824208] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
Glypican-3 (GPC3) is a membrane-associated proteoglycan that is specifically up-regulated in hepatocellular carcinoma (HCC) although rarely or not expressed in normal liver tissues, making it a perfect diagnostic and treatment target for HCC. Several GPC3-based clinical trials are ongoing and recently several innovative GPC3-targeted therapeutic methods have emerged with exciting results, including GPC3 vaccine, anti-GPC3 immunotoxin, combined therapy with immune checkpoint blockades (ICBs), and chimeric antigen receptor (CAR) T or NK cells. Here, we review the value of GPC3 in the diagnosis and prognosis of HCC, together with its signaling pathways, with a specific focus on GPC3-targeted treatments of HCC and some prospects for the future GPC3-based therapeutic strategies in HCC.
Collapse
Affiliation(s)
- Xiufeng Zheng
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xun Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Yanna Lei
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Ming Liu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Zhang Y, Sun M, Gao L, Liang X, Ma C, Lu J, Yue X. ZHX2 inhibits thyroid cancer metastasis through transcriptional inhibition of S100A14. Cancer Cell Int 2022; 22:76. [PMID: 35151335 PMCID: PMC8840030 DOI: 10.1186/s12935-022-02499-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/30/2022] [Indexed: 12/21/2022] Open
Abstract
Background Thyroid cancer is the most common malignant endocrine tumour, and metastasis has become the main reason for treatment failure. However, the underlying molecular mechanism of thyroid cancer metastasis remains poorly understood. We investigated the role of the tumour suppressor zinc fingers and homeoboxes 2 (ZHX2) in the metastasis of thyroid cancer. Methods To study the role of ZHX2 in thyroid cancer metastasis, we evaluated the EMT process using cell migration, wound healing and lung metastatic tumour formation in vitro and in vivo models. Results ZHX2 expression was significantly decreased in thyroid cancer tissues, which correlated with poor prognosis of thyroid cancer patients. ZHX2 knockdown significantly promoted the migration of thyroid cancer cells. Mechanistically, ZHX2 associated with the S100 calcium binding protein A14 (S100A14) promoter to decrease the transcription of S100A14. Moreover, S100A14 was highly expressed in human thyroid cancer samples, and its expression negatively correlated with ZHX2 expression. Conclusions Inhibition of S100A14 attenuated the ZHX2 knockdown-induced enhanced metastasis of thyroid cancer cells both in vitro and in vivo. The evidence presented here suggests that ZHX2 inhibits the progression of thyroid cancer by blocking S100A14-mediated metastasis. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02499-w.
Collapse
|
11
|
Yu X, Lin Q, Wu Z, Zhang Y, Wang T, Zhao S, Song X, Chen C, Wang Z, Xu L, Li C, Gao L, Liang X, Yue X, Ma C. ZHX2 inhibits SREBP1c-mediated de novo lipogenesis in hepatocellular carcinoma via miR-24-3p. J Pathol 2020; 252:358-370. [PMID: 32770671 DOI: 10.1002/path.5530] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. Lipogenesis has been considered as a critical player in HCC initiation and progression. However, the underlying mechanism is still not fully understood. Here, we identified zinc fingers and homeoboxes 2 (ZHX2), an HCC-associated tumor suppressor, as an important repressor of de novo lipogenesis. Ectopic expression of ZHX2 significantly inhibited de novo lipogenesis in HCC cells and decreased expression of FASN, ACL, ACC1, and SCD1. In accordance with this, ZHX2 was negatively associated with SREBP1c, the master regulator of de novo lipogenesis, in HCC cell lines and human specimens. Results from silencing and overexpression demonstrated that ZHX2 inhibited de novo lipogenesis and consequent HCC progression via repression of SREBP1c. Furthermore, treatment with the SREBP1c inhibitor fatostatin dampened the spontaneous formation of tumors in liver-specific Zhx2 knockout mice. Mechanistically, ZHX2 increased expression of miR-24-3p transcriptionally, which targeted SREBP1c and led to its degradation. In conclusion, our data suggest a novel mechanism through which ZHX2 suppresses HCC progression, which may provide a new strategy for the treatment of HCC. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Xiangguo Yu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Qinghai Lin
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Yankun Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Tixiao Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Songbai Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xiaojia Song
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Chaojia Chen
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Zehua Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Leiqi Xu
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Chunyang Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xuetian Yue
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Cell Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, PR China.,Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| |
Collapse
|
12
|
Jiang J, Sun Y, Xu J, Xu T, Xu Z, Liu P. ZHX2 mediates proteasome inhibitor resistance via regulating nuclear translocation of NF-κB in multiple myeloma. Cancer Med 2020; 9:7244-7252. [PMID: 32780537 PMCID: PMC7541163 DOI: 10.1002/cam4.3347] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/09/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Background Multiple myeloma (MM) is an incurable hematological malignancy. Although proteasome inhibitors and immunomodulators have significantly improved patient outcomes, some patients respond poorly to treatment and almost all patients will relapse. Mechanisms of proteasome inhibitor resistance in multiple myeloma have not been fully elucidated. ZHX2 is a transcription regulator degraded via proteasome and presents both oncogenic or tumor suppressive effect in different cancers, however, it is still unknown that the role of ZHX2 in myeloma. In this study, we aim to demonstrate the effect and mechanism of ZHX2 on proteasome inhibitor resistance in MM. Methods GSE24080 gene expression profile datasets from Gene Expression Omnibus (GEO) were analyzed to evaluate the relationship between ZHX2 expression level and survival in MM. Expression of ZHX2 in human MM cell lines at baseline and after bortezomib (BTZ) treatment was determined by Western blotting (WB). The proliferation and apoptosis rate of MM cells treated with BTZ after the knockdown of ZHX2 were analyzed by flow cytometry. Nuclear translocation of NF‐κB after the knockdown of ZHX2 was evaluated by WB and immunofluorescence, and the expression of NF‐κB target genes was measured by real‐time quantitative PCR. Co‐immunoprecipitation (Co‐IP) and WB were used to detect the interaction of ZHX2 with NF‐κB. Results We found that higher ZHX2 expression was correlated with poorer clinical outcomes of patients. In addition, ZHX2 expression was relatively higher in RPMI‐8226 and MM.1S cell lines and the level of ZHX2 protein was upregulated after BTZ treatment. Knockdown of ZHX2 significantly enhanced the sensitivity of MM cells to BTZ, inhibited nuclear translocation of NF‐κB, and reduced mRNA expression of NF‐κB target genes. It was also revealed that ZHX2 directly binds to NF‐κB. Conclusion Our study showed that ZHX2 can promote proteasome inhibitor resistance in MM cells by regulating the nuclear translocation of NF‐κB.
Collapse
Affiliation(s)
- Jifeng Jiang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifeng Sun
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiadai Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianhong Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhao Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Wang Z, Kong L, Tan S, Zhang Y, Song X, Wang T, Lin Q, Wu Z, Xiang P, Li C, Gao L, Liang X, Ma C. Zhx2 Accelerates Sepsis by Promoting Macrophage Glycolysis via Pfkfb3. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2232-2241. [PMID: 32179636 DOI: 10.4049/jimmunol.1901246] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/12/2020] [Indexed: 01/03/2025]
Abstract
Sepsis is a life-threatening condition with limited therapeutic options, characterized as excessive systemic inflammation and multiple organ failure. Macrophages play critical roles in sepsis pathogenesis. Metabolism orchestrates homeostasis of macrophages. However, the precise mechanism of macrophage metabolism during sepsis remains poorly elucidated. In this study, we identified the key role of zinc fingers and homeoboxes (Zhx2), a ubiquitous transcription factor, in macrophage glycolysis and sepsis by enhancing 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (Pfkfb3) expression. Mice with myeloid Zhx2-specific deletion (abbreviated as MKO) showed more resistance to cecal ligation and puncture and LPS-induced sepsis, exhibiting as prolonged survival, attenuated pulmonary injury, and reduced level of proinflammatory cytokines, such as TNF-α, IL-6, and IL-1β. Interestingly, Zhx2 deletion conferred macrophage tolerance to LPS-induced glycolysis, accompanied by reduced proinflammatory cytokines and lactate. Consistently, treatment of glycolytic inhibitor 2-deoxyglucose almost completely abrogated the protection of mice from LPS-induced sepsis initiated by Zhx2 deletion in macrophages. RNA sequencing and chromatin immunoprecipitation assays confirmed that Zhx2 enhanced transcription of Pfkfb3, the glycolysis rate-limiting enzyme, via binding with Pfkfb3 promoter. Furthermore, Pfkfb3 overexpression not only rescued the reduction of macrophage glycolysis caused by Zhx2 deficiency, displaying as extracellular acidification rates and lactate production but also destroyed the resistance of mice to LPS-induced sepsis initiated by transfer of bone marrow-derived macrophages from MKO mice. These findings highlight the novel role of transcription factor Zhx2 in sepsis via regulating Pfkfb3 expression and reprogramming macrophage metabolism, which would shed new insights into the potential strategy to intervene sepsis.
Collapse
Affiliation(s)
- Zehua Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Liang Kong
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250012, People's Republic of China
| | - Siyu Tan
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Yankun Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Xiaojia Song
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Tixiao Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Qinghai Lin
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Peng Xiang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Chunyang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology and Embryology, Shandong University School of Basic Medical Science, Jinan, Shandong 250012, People's Republic of China; and
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China;
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| |
Collapse
|
14
|
Lin Q, Wu Z, Yue X, Yu X, Wang Z, Song X, Xu L, He Y, Ge Y, Tan S, Wang T, Song H, Yuan D, Gong Y, Gao L, Liang X, Ma C. ZHX2 restricts hepatocellular carcinoma by suppressing stem cell-like traits through KDM2A-mediated H3K36 demethylation. EBioMedicine 2020; 53:102676. [PMID: 32114388 PMCID: PMC7047184 DOI: 10.1016/j.ebiom.2020.102676] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/16/2020] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
Background Liver cancer stem cells (CSCs) are critical determinants of HCC relapse and therapeutic resistance, but the mechanisms underlying the maintenance of CSCs are poorly understood. We aimed to explore the role of tumor repressor Zinc-fingers and homeoboxes 2 (ZHX2) in liver CSCs. Methods CD133+ or EPCAM+ stem-like liver cancer cells were sorted from tumor tissues of HCC patients and HCC cell lines by flow cytometry. In addition, sorafenib-resistant cells, tumor-sphere forming cells and side population (SP) cells were respectively cultured and isolated as hepatic CSCs. The tumor-initiating and chemoresistance properties of ZHX2-overexpressing and ZHX2-knockdown cells were analyzed in vivo and in vitro. Microarray, luciferase reporter assay, chromatin immunoprecipitation (ChIP) and ChIP-on-chip analyses were performed to explore ZHX2 target genes. The expression of ZHX2 and its target gene were determined by quantitative RT-PCR, western blot, immunofluorescence and immunohistochemical staining in hepatoma cells and tumor and adjacent tissues from HCC patients. Results ZHX2 expression was significantly reduced in liver CSCs from different origins. ZHX2 deficiency led to enhanced liver tumor progression and expansion of CSC populations in vitro and in vivo. Re-expression of ZHX2 restricted capabilities of hepatic CSCs in supporting tumor initiation, self-renewal and sorafenib-resistance. Mechanically, ZHX2 suppressed liver CSCs via inhibiting KDM2A-mediated demethylation of histone H3 lysine 36 (H3K36) at the promoter regions of stemness-associated transcription factors, such as NANOG, SOX4 and OCT4. Moreover, patients with lower expression of ZHX2 and higher expression of KDM2A in tumor tissues showed significantly poorer survival. Conclusion ZHX2 counteracts stem cell traits through transcriptionally repressing KDM2A in HCC. Our data will aid in a better understanding of molecular mechanisms underlying HCC relapse and drug resistance.
Collapse
Affiliation(s)
- Qinghai Lin
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Xuetian Yue
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Xiangguo Yu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Zehua Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Xiaojia Song
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Leiqi Xu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China; Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | - Ying He
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China; Institute of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | - Yutong Ge
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Siyu Tan
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Tixiao Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Hui Song
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Detian Yuan
- Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, PR China
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, Shandong University School of Basic Medical Sciences, Jinan, Shandong, PR China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, 44# Wenhua Xi Road, Jinan, Shandong 250012, PR China; Advanced Medical Research Institute, Shandong University, Jinan, Shandong, PR China.
| |
Collapse
|
15
|
Wu Z, Ma H, Wang L, Song X, Zhang J, Liu W, Ge Y, Sun Y, Yu X, Wang Z, Wang J, Zhang Y, Li C, Li N, Gao L, Liang X, Yue X, Ma C. Tumor suppressor ZHX2 inhibits NAFLD-HCC progression via blocking LPL-mediated lipid uptake. Cell Death Differ 2019; 27:1693-1708. [PMID: 31740790 PMCID: PMC7206072 DOI: 10.1038/s41418-019-0453-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/27/2019] [Accepted: 10/29/2019] [Indexed: 11/21/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) leads to hepatocellular carcinoma (HCC). However, the underlying mechanism remains largely unclear. Here, we investigated the role of the tumor suppressor Zinc fingers and homeoboxes 2 (ZHX2) in the progression of NAFLD to HCC. ZHX2 expression was significantly decreased in fatty liver tissues, especially in the liver with NAFLD–HCC. ZHX2 overexpression disturbed lipid homeostasis of cultured HCC cells, and inhibited lipid deposition in hepatocytes both in vitro and in vivo. Moreover, ZHX2 inhibited uptake of exogenous lipids through transcriptional suppression of lipid lipase (LPL), leading to retarded proliferation of HCC cells. Importantly, LPL overexpression significantly reversed ZHX2-mediated inhibition of HCC cell proliferation, xenograft tumor growth, lipid deposition, and spontaneous liver tumor formation. Consistently, IHC staining demonstrated a negative correlation of ZHX2 with LPL in an HCC cohort. Collectively, ZHX2 protects hepatocytes from abnormal lipid deposition in NAFLD through transcriptional repression of LPL, which subsequently retards cell growth and NAFLD–HCC progression. These findings illustrate a novel mechanism of NAFLD progression into HCC.
Collapse
Affiliation(s)
- Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Hongxin Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China.,Clinical Laboratory, Shandong Cancer Hospital & Institute Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250012, PR China
| | - Liyuan Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaojia Song
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Jie Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Wen Liu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yutong Ge
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yang Sun
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiangguo Yu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Zehua Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Jianping Wang
- Department of General Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250012, PR China
| | - Yankun Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Chunyang Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Nailin Li
- Karolinska Institutet, Department of Medicine-Solna, Clinical Pharmacology Group, 171 76, Stockholm, Sweden
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xuetian Yue
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Cell Biology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
16
|
Intratumoral reciprocal expression of monocarboxylate transporter 4 and glypican-3 in hepatocellular carcinomas. BMC Res Notes 2019; 12:741. [PMID: 31706332 PMCID: PMC6842510 DOI: 10.1186/s13104-019-4778-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/01/2019] [Indexed: 12/26/2022] Open
Abstract
Objective We previously reported the identification of monocarboxylate transporter 4 (MCT4) and glypican-3 (GPC3) as prognostic factors for hepatocellular carcinoma (HCC), which are now considered significant poor prognostic factors for the disease. This study aimed to clarify the detailed interaction of these two factors in HCC to improve our understanding of aggressive HCC phenotypes. A total of 225 Japanese patients with HCC from our previous study were subjected to immunohistochemical analyses. Results The number of MCT4-positive (MCT4+) HCC cases was 47 (21%), and most MCT4+ HCC showed high GPC3 expression (94%, 44/47 cases). In 44 MCT4+/GPC3+ HCC cases, intratumoral heterogeneity of GPC3 or MCT4 expression was further evaluated. We observed reciprocal (inverse), synergistic, mixed reciprocal and synergistic, or irrelevant interaction of MCT4 and GPC3 expression in 29 (66%), 5 (11%), 1 (2%), and 9 cases (21%), respectively. The cases exhibiting reciprocal expression of both markers tended to have cirrhosis without a history of neoadjuvant therapy. In summary, although MCT4+ HCC cases are mostly GPC3+, intratumoral expression patterns of MCT4 and GPC3 are frequently reciprocal each other, suggesting that dual targeting of MCT4 and GPC3 may achieve a better antitumor effect for MCT4+ HCC cases.
Collapse
|
17
|
Tyrosine Kinase Inhibitor Imatinib Mesylate Alters DMBA-Induced Early Onco/Suppressor Gene Expression with Tissue-Specificity in Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8670398. [PMID: 30882001 PMCID: PMC6383434 DOI: 10.1155/2019/8670398] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/28/2018] [Accepted: 01/13/2019] [Indexed: 12/30/2022]
Abstract
Tyrosine kinases play crucial roles in cellular development and tumorigenesis. Tyrosine kinase inhibitors (TKIs) are effective and widely used drug molecules in targeted cancer therapies. Altered expressions of protooncogenes and tumor suppressor genes after DMBA (7,12-dimethylbenz[a]anthracene) treatment have been described as early markers of tumor induction; however their tissue-specific effects remain still unclear. Our study was aimed at examining the short-term possible antineoplastic and chemopreventive effects of a TKI compound (imatinib mesylate) on a DMBA-induced mouse tumor model. In addition, we also investigated the tissue-specific expressions of Hras, Kras, Myc, and Trp53 genes in the brain, bone marrow, spleen, liver, abdominal lymph nodes, thymus, lungs, and kidneys, respectively. 24 hours after the imatinib mesylate injection, we observed significant Kras downregulation in the bone marrow and lung of the DMBA-treated mice. Moreover, the mRNA expression of Myc was also found to be decreased significantly in the spleen. Interestingly, while Trp53 expression was significantly increased in the lung, it was decreased in the other tissues. However, there was also a tendency in the decreased Myc level in the bone marrow, brain, kidneys, lungs, and lymph nodes and in the decreased Hras level in the bone marrow, kidneys, and lungs, although no significant differences were observed. Our findings indicate rapid tissue-specific impact of imatinib mesylate on DMBA-induced gene expression in vivo, supporting the chemopreventive potential of imatinib mesylate in cancer.
Collapse
|
18
|
You Y, Ma Y, Wang Q, Ye Z, Deng Y, Bai F. Attenuated ZHX3 expression serves as a potential biomarker that predicts poor clinical outcomes in breast cancer patients. Cancer Manag Res 2019; 11:1199-1210. [PMID: 30787639 PMCID: PMC6368119 DOI: 10.2147/cmar.s184340] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background The ZHX family has recently been in the spotlight as an integrator and an indispensable node in carcinogenesis, whose expression is frequently dysregulated in multiple cancers. The current study provides a novel investigation of the expression profiles of ZHX factors in breast cancer. Materials and methods The mRNA levels of ZHXs and follow-up periods in breast cancer patients were mined through the Oncomine, Cancer Cell Line Encyclopedia, bc-GenExMiner, cBioPortal and Kaplan–Meier plotter databases. In addition, ZHX3 protein expression was examined in 98 primary tumor samples by immunohistochemistry to investigate its association with clinicopathological parameters and patient outcomes. Results We found that the transcriptional levels of ZHX1, ZHX2 and ZHX3 were not significantly altered in tumor tissues compared with those in nontumor tissues. ZHX2 and ZHX3 mRNA levels were observed to be positively correlated with estrogen receptor and progesterone receptor expression, while ZHX2 mRNA levels were negatively associated with HER2 expression. Survival analyses revealed that high mRNA levels of ZHX2 and ZHX3 correlated with better overall survival in patients with breast cancer. Immunohistochemical analysis revealed that patients with decreased ZHX3 protein levels had poorer outcomes. Multivariate analysis exhibited that ZHX3 expression may serve as an independent high-risk prognostic predictor. Conclusion Dysregulated expression of ZHXs may be involved in the progression of breast cancer and could serve as a novel biomarker and potential target for breast cancer.
Collapse
Affiliation(s)
- Yanjie You
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, China, ;
| | - Yuhong Ma
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, China, ;
| | - Qiang Wang
- Department of Science and Education, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, China
| | - Zhengcai Ye
- Endoscopy Center, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, China
| | - Yanhong Deng
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, China, ;
| | - Feihu Bai
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, China, ;
| |
Collapse
|
19
|
Song X, Tan S, Wu Z, Xu L, Wang Z, Xu Y, Wang T, Gao C, Gong Y, Liang X, Gao L, Spear BT, Ma C. HBV suppresses ZHX2 expression to promote proliferation of HCC through miR-155 activation. Int J Cancer 2018; 143:3120-3130. [PMID: 29752719 DOI: 10.1002/ijc.31595] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/29/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Abstract
Initiation of hepatocellular carcinoma (HCC) by chronic hepatitis B virus (HBV) infection is a complex process that includes both oncogene activation and tumor suppressor inhibition. The HBV X (HBx) protein has an important and complex role in processes leading to HCC. We previously identified the mammalian Zinc fingers and homeoboxes 2 (ZHX2) gene as an HCC-associated tumor suppressor gene. In the present study, we investigated whether the oncogenic properties of HBV and, more specifically, HBx, involved ZHX2 silencing. Our data indicates that ZHX2 expression is significantly decreased in tumor tissues from HBV-positive HCC patients and livers from HBV transgenic mice. In vitro and in vivo studies confirmed that HBV-encoded proteins, particularly HBx, inhibits both the expression and tumor suppression properties of ZHX2. Further analyses identified miR-155, a well-known oncomiR in various cancers, as an important link between HBx and ZHX2 inhibition. Increased miR-155 levels were found in HBV-positive tumors, livers of HBV transgenic mice and HBx-overexpressing hepatoma cell lines. MiR-155 overexpression reduced ZHX2 levels via miR-155 seed sites in the ZHX2 3'UTR, whereas blocking miR-155 levels led to increased ZHX2 levels. Taken together, our data indicate that HCC-promoting properties of HBV may include ZHX2 silencing via a miR-155 dependent pathway and suggests a novel therapy for HBV-related HCC.
Collapse
Affiliation(s)
- Xiaojia Song
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Siyu Tan
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Leiqi Xu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Zehua Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Yong Xu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Tixiao Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, People's Republic of China
| | - Yaoqin Gong
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Genetics, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China
| | - Brett T Spear
- Department of Microbiology, Immunology and Molecular Genetics, Lexington, KY.,Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, 250012, People's Republic of China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, People's Republic of China
| |
Collapse
|
20
|
Xu L, Wu Z, Tan S, Wang Z, Lin Q, Li X, Song X, Liu Y, Song Y, Zhang J, Peng J, Gao L, Gong Y, Liang X, Zuo X, Ma C. Tumor suppressor ZHX2 restricts hepatitis B virus replication via epigenetic and non-epigenetic manners. Antiviral Res 2018; 153:114-123. [DOI: 10.1016/j.antiviral.2018.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/14/2018] [Accepted: 03/22/2018] [Indexed: 01/10/2023]
|
21
|
The Transcriptional Network Structure of a Myeloid Cell: A Computational Approach. Int J Genomics 2017; 2017:4858173. [PMID: 29119102 PMCID: PMC5651161 DOI: 10.1155/2017/4858173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/28/2017] [Accepted: 08/09/2017] [Indexed: 01/24/2023] Open
Abstract
Understanding the general principles underlying genetic regulation in eukaryotes is an incomplete and challenging endeavor. The lack of experimental information regarding the regulation of the whole set of transcription factors and their targets in different cell types is one of the main reasons to this incompleteness. So far, there is a small set of curated known interactions between transcription factors and their downstream genes. Here, we built a transcription factor network for human monocytic THP-1 myeloid cells based on the experimentally curated FANTOM4 database where nodes are genes and the experimental interactions correspond to links. We present the topological parameters which define the network as well as some global structural features and introduce a relative inuence parameter to quantify the relevance of a transcription factor in the context of induction of a phenotype. Genes like ZHX2, ADNP, or SMAD6 seem to be highly regulated to avoid an avalanche transcription event. We compare these results with those of RegulonDB, a highly curated transcriptional network for the prokaryotic organism E. coli, finding similarities between general hallmarks on both transcriptional programs. We believe that an approach, such as the one shown here, could help to understand the one regulation of transcription in eukaryotic cells.
Collapse
|
22
|
Luan F, Liu B, Zhang J, Cheng S, Zhang B, Wang Y. Correlation between HBV protein preS2 and tumor markers of hepatocellular carcinoma. Pathol Res Pract 2017; 213:1037-1042. [PMID: 28869105 DOI: 10.1016/j.prp.2017.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/21/2017] [Accepted: 08/17/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Alpha-fetoprotein (AFP) and Glypican 3 (GPC3) are both oncogenes and reactivated in hepatocellular carcinoma (HCC). PreS2 has been proved to be an important transactivator in HCC. In this study, we aim to provide evidence that HBV protein preS2 is responsible for AFP and GPC3's reactivation in HCC. METHODS Totally Sixty-three cases of HCC, aged 34-79, who were surgically treated and pathologically confirmed were enrolled. The levels of AFP in peripheral serum were detected with electrochemical luminescence method before surgery. Levels of GPC3 in HCC samples were evaluated by immunohistochemistry. Luciferase reporter assays were used to measure the effect of preS2 on AFP and GPC3 promoters. RESULTS AFP level and GPC3 but not albumin were significantly higher in preS2-positive HCC samples than preS2-negative HCC samples. And the preS2 protein expression was positively related with serum AFP level and GPC3 expression. Furtherly, dual luciferase assay showed that preS2 activated AFP and GPC3 promoter activity. CONCLUSION The expression of preS2 protein relates closely to HCC markers AFP and GPC3.
Collapse
Affiliation(s)
- Fang Luan
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, PR China
| | - Bin Liu
- Department of Biomedical Engineering, Shandong Provincial Hospital affiliated to Shandong University, PR China
| | - Junguo Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, PR China
| | - Shiqing Cheng
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, PR China
| | - Bingchang Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, PR China
| | - Yong Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, PR China.
| |
Collapse
|
23
|
Zhou F, Shang W, Yu X, Tian J. Glypican-3: A promising biomarker for hepatocellular carcinoma diagnosis and treatment. Med Res Rev 2017. [PMID: 28621802 DOI: 10.1002/med.21455] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Liver cancer is the second leading cause of cancer-related deaths, and hepatocellular carcinoma (HCC) is the most common type. Therefore, molecular targets are urgently required for the early detection of HCC and the development of novel therapeutic approaches. Glypican-3 (GPC3), an oncofetal proteoglycan anchored to the cell membrane, is normally detected in the fetal liver but not in the healthy adult liver. However, in HCC patients, GPC3 is overexpressed at both the gene and protein levels, and its expression predicts a poor prognosis. Mechanistic studies have revealed that GPC3 functions in HCC progression by binding to molecules such as Wnt signaling proteins and growth factors. Moreover, GPC3 has been used as a target for molecular imaging and therapeutic intervention in HCC. To date, GPC3-targeted magnetic resonance imaging, positron emission tomography, and near-infrared imaging have been investigated for early HCC detection, and various immunotherapeutic protocols targeting GPC3 have been developed, including the use of humanized anti-GPC3 cytotoxic antibodies, treatment with peptide/DNA vaccines, immunotoxin therapies, and genetic therapies. In this review, we summarize the current knowledge regarding the structure, function, and biology of GPC3 with a focus on its clinical potential as a diagnostic molecule and a therapeutic target in HCC immunotherapy.
Collapse
Affiliation(s)
- Fubo Zhou
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wenting Shang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiaoling Yu
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
24
|
Tracking Cancer Genetic Evolution using OncoTrack. Sci Rep 2016; 6:29647. [PMID: 27412732 PMCID: PMC4944131 DOI: 10.1038/srep29647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 06/20/2016] [Indexed: 02/07/2023] Open
Abstract
It is difficult for existing methods to quantify, and track the constant evolution of cancers due to high heterogeneity of mutations. However, structural variations associated with nucleotide number changes show repeatable patterns in localized regions of the genome. Here we introduce SPKMG, which generalizes nucleotide number based properties of genes, in statistical terms, at the genome-wide scale. It is measured from the normalized amount of aligned NGS reads in exonic regions of a gene. SPKMG values are calculated within OncoTrack. SPKMG values being continuous numeric variables provide a statistical metric to track DNA level changes. We show that SPKMG measures of cancer DNA show a normative pattern at the genome-wide scale. The analysis leads to the discovery of core cancer genes and also provides novel dynamic insights into the stage of cancer, including cancer development, progression, and metastasis. This technique will allow exome data to also be used for quantitative LOH/CNV analysis for tracking tumour progression and evolution with a higher efficiency.
Collapse
|
25
|
Proliferation and osteo/odontogenic differentiation of stem cells from apical papilla regulated by Zinc fingers and homeoboxes 2: An in vitro study. Biochem Biophys Res Commun 2015; 469:599-605. [PMID: 26679602 DOI: 10.1016/j.bbrc.2015.11.135] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 11/25/2015] [Accepted: 11/30/2015] [Indexed: 11/24/2022]
Abstract
In the process of tooth root development, stem cells from the apical papilla (SCAPs) can differentiate into odontoblasts and form root dentin, however, molecules regulating SCAPs differentiation have not been elucidated. Zinc fingers and homeoboxes 2 (ZHX2) is a novel transcriptional inhibitor. It is reported to modulate the development of nerve cells, liver cells, B cells, red blood cells, and so on. However, the role of ZHX2 in tooth root development remains unclear. In this study, we explored the potential role of ZHX2 in the process of SCAPs differentiation. The results showed that overexpression of ZHX2 upregulated the expression of osteo/odontogenic related genes and ALP activity, inhibited the proliferation of SCAPs. Consistently, ZHX2 knockdown reduced SCAPs mineralization and promoted SCAPs proliferation. These results indicated that ZHX2 plays a critical role in the proliferation and osteo/odontogenic differentiation of SCAPs.
Collapse
|
26
|
Berasain C, Avila MA. Regulation of hepatocyte identity and quiescence. Cell Mol Life Sci 2015; 72:3831-51. [PMID: 26089250 PMCID: PMC11114060 DOI: 10.1007/s00018-015-1970-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/23/2015] [Accepted: 06/12/2015] [Indexed: 12/11/2022]
Abstract
The liver is a highly differentiated organ with a central role in metabolism, detoxification and systemic homeostasis. To perform its multiple tasks, liver parenchymal cells, the hepatocytes, express a large complement of enabling genes defining their complex phenotype. This phenotype is progressively acquired during fetal development and needs to be maintained in adulthood to guarantee the individual's survival. Upon injury or loss of functional mass, the liver displays an extraordinary regenerative response, mainly based on the proliferation of hepatocytes which otherwise are long-lived quiescent cells. Increasing observations suggest that loss of hepatocellular differentiation and quiescence underlie liver malfunction in chronic liver disease and pave the way for hepatocellular carcinoma development. Here, we briefly review the essential mechanisms leading to the acquisition of liver maturity. We also identify the key molecular factors involved in the preservation of hepatocellular homeostasis and finally discuss potential strategies to preserve liver identity and function.
Collapse
Affiliation(s)
- Carmen Berasain
- Division of Hepatology, CIMA, University of Navarra, CIBEREHD, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Avda. Pio XII, n55, 31008, Pamplona, Spain.
| | - Matías A Avila
- Division of Hepatology, CIMA, University of Navarra, CIBEREHD, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Avda. Pio XII, n55, 31008, Pamplona, Spain.
| |
Collapse
|
27
|
Abstract
BACKGROUND The carcinogenesis of hepatocellular carcinoma (HCC) is a multi-factorial, multi-step and complex process. Early diagnosis and effective treatments are of utmost importance. This review summarized the recent studies of oncofetal glypican-3 (GPC-3), a membrane-associated heparan sulfate proteoglycan, in the diagnosis and treatment of HCC. DATA SOURCES English-language reports published from June 2001 to September 2014 were searched from MEDLINE. The key words searched included: GPC-3, biomarker, target and HCC. The sensitivity, specificity, positive and negative predictive values were extracted, and the effect of GPC-3 targeted therapy on HCC was also evaluated. RESULTS GPC-3 plays a crucial role in HCC cell proliferation and metastasis. It mediates oncogenesis involving signaling pathways during hepatocyte malignant transformation. GPC-3 expression is increased in atypical hyperplasia and cancerous tissues. GPC-3 levels in HCC patients are related to HBV infection, TNM stage, periportal cancerous embolus, and extrahepatic metastasis. The diagnostic accuracy of the combination of serum GPC-3 and alpha-fetoprotein in HCC is up to 94.3%. Down-regulation of GPC-3 with specific siRNA or anti-GPC-3 antibody alters cell migration, metastasis and invasion behaviors. The nude mice xenograft tumor growth is inhibited by silencing GPC-3 gene transcription. CONCLUSION Oncofetal GPC-3 is a highly specific biomarker for the diagnosis of HCC and a promising target molecule for HCC gene therapy.
Collapse
|