51
|
Abstract
Metallothioneins (MTs) are small cysteine-rich proteins that play important roles in metal homeostasis and protection against heavy metal toxicity, DNA damage, and oxidative stress. In humans, MTs have four main isoforms (MT1, MT2, MT3, and MT4) that are encoded by genes located on chromosome 16q13. MT1 comprises eight known functional (sub)isoforms (MT1A, MT1B, MT1E, MT1F, MT1G, MT1H, MT1M, and MT1X). Emerging evidence shows that MTs play a pivotal role in tumor formation, progression, and drug resistance. However, the expression of MTs is not universal in all human tumors and may depend on the type and differentiation status of tumors, as well as other environmental stimuli or gene mutations. More importantly, the differential expression of particular MT isoforms can be utilized for tumor diagnosis and therapy. This review summarizes the recent knowledge on the functions and mechanisms of MTs in carcinogenesis and describes the differential expression and regulation of MT isoforms in various malignant tumors. The roles of MTs in tumor growth, differentiation, angiogenesis, metastasis, microenvironment remodeling, immune escape, and drug resistance are also discussed. Finally, this review highlights the potential of MTs as biomarkers for cancer diagnosis and prognosis and introduces some current applications of targeting MT isoforms in cancer therapy. The knowledge on the MTs may provide new insights for treating cancer and bring hope for the elimination of cancer.
Collapse
Affiliation(s)
- Manfei Si
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730 China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730 China
| |
Collapse
|
52
|
Zeng JD, Zhang N, Zhao GJ, Xu LX, Yang Y, Xu XY, Chen MK, Wang HY, Zheng SXF, Li XX. MT1G is Silenced by DNA Methylation and Contributes to the Pathogenesis of Hepatocellular Carcinoma. J Cancer 2018; 9:2807-2816. [PMID: 30123349 PMCID: PMC6096370 DOI: 10.7150/jca.25680] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/02/2018] [Indexed: 12/11/2022] Open
Abstract
Using genome-wide screening and TCGA-based data analysis, we identified a DNA methylation-related gene named metallothionein-1G (MT1G), which may play an important role in hepatocellular carcinoma (HCC). In this study, we found that MT1G expression was silenced in 4/6 HCC cell lines and negatively related to aberrant promoter hypermethylation. Its mRNA level was restored with demethylation treatment. Moreover, MT1G downregulation at both the transcriptional and protein level was also detected in 8 pairs of clinical HCC samples compared with its expression in adjacent normal tissues. Ectopic expression of MT1G in silenced HCC cell lines inhibited colony formation, suppressed cell migration and invasion, and repressed xenograft tumor growth in nude mice. In contrast, knockdown of MT1G by short hairpin RNA showed the opposite effect on cell proliferation and the malignant phenotype. Moreover, our data showed that MT1G suppressed tumor invasion and metastasis mainly through regulating the expression of proteins in the matrix metalloproteinase family (MMP) and modulating the epithelial-mesenchymal transition (EMT) process. To our surprise, the data from TCGA showed that hypermethylation of MT1G is associated with good survival of HCC patients. In conclusion, our study demonstrated that MT1G acts as a tumor suppressor gene in HCC development, but its clinical potential in HCC requires further evaluation.
Collapse
Affiliation(s)
- Ju-deng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ning Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Gui-jun Zhao
- Department of Gastroenterology and Hepatology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Li-xia Xu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yang Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xiao-yi Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Meng-ke Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Hui-yun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Steven Xiao-feng Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Rutgers Cancer Institute of New Jersey and Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Xiao-xing Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
53
|
Fu CL, Pan B, Pan JH, Gan MF. Metallothionein 1M suppresses tumorigenesis in hepatocellular carcinoma. Oncotarget 2018; 8:33037-33046. [PMID: 28380433 PMCID: PMC5464848 DOI: 10.18632/oncotarget.16521] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/10/2017] [Indexed: 12/29/2022] Open
Abstract
Members of the metallothionein (MT) family are involved in metal detoxifcation and in the protection of cells against certain electrophilic carcinogens. In present study, it was found that MT1M was downregulated in more than 77.1% (91/118) of hepatocellular carcinoma (HCC) tissues compared with adjacent non-tumor tissues. Furthermore, overexpression of MT1M inhibited cell viability, colony formation, cell migration and invasion in HCC cell lines and tumor cell growth in xenograft nude mice, and activated cell apoptosis in HCC cell lines. In addition, immunohistochemistry analysis showed MT1M was negative or weak staining in tumor tissues but moderate or strong staining in adjacent non-tumor tissues. The sensitivity and specificity of MT1M for HCC diagnosis were 76.27% and 89.83%, respectively. In conclusion, MT1M was identified as a potential tumor marker for HCC and may serve as a useful therapeutic agent for HCC gene therapy.
Collapse
Affiliation(s)
- Cheng-Lin Fu
- Department of Pathology, The First Hospital of Taizhou, Wenzhou Medical University, Taizhou 318020, China
| | - Bing Pan
- Department of Pathology, The First Hospital of Taizhou, Wenzhou Medical University, Taizhou 318020, China
| | - Ju-Hua Pan
- Department of Pathology, The First Hospital of Taizhou, Wenzhou Medical University, Taizhou 318020, China
| | - Mei-Fu Gan
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Taizhou Hospital of Zhejiang Province, Taizhou Enze Medical Center (Group), Linhai 317000, China
| |
Collapse
|
54
|
Shi G, Zhang J, Lu Z, Liu D, Wu Y, Wu P, Yin J, Yuan H, Zhu Q, Chen L, Fu Y, Peng Y, Wang Y, Jiang K, Miao Y. A novel messenger RNA signature as a prognostic biomarker for predicting relapse in pancreatic ductal adenocarcinoma. Oncotarget 2017; 8:110849-110860. [PMID: 29340021 PMCID: PMC5762289 DOI: 10.18632/oncotarget.22861] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/04/2017] [Indexed: 01/09/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) death rate and recurrence rate have remained obstinately high. Current methods can not satisfy the need of predicting cancer relapse accurately. Utilizing expression profiles of 10 GEO datasets (N = 774), we identified 154 differentially expressed genes (DEGs) between PDAC and normal pancreas tissue or paracancerous tissue. Next we built a 16-mRNA-based signature by means of the LASSO COX regression model. We also validated the prognostic value of the signature. Patients were classified into high-risk and low-risk group according to the signature risk score; 1 year RFS was 45% (95% CI: 31.6%–63.9%) for high-risk group in contrast to 92.5% (95% CI: 86.3%–99.1%) for low-risk group. Moreover, it could predict RFS well in cases with the receipt of different treatment modalities. The 16-mRNA-based signature was an independent and powerful prognostic biomarker for RFS for PDAC patients (HR = 7.74, 95% CI: 3.25–18.45, p < 0.0001).
Collapse
Affiliation(s)
- Guodong Shi
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Jingjing Zhang
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Zipeng Lu
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Dongfang Liu
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Yang Wu
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Pengfei Wu
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Jie Yin
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Hao Yuan
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Qicong Zhu
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Lei Chen
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Yue Fu
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Yunpeng Peng
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Yan Wang
- Endoscopy Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kuirong Jiang
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Yi Miao
- Pancreas Center, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
55
|
Krizkova S, Kepinska M, Emri G, Eckschlager T, Stiborova M, Pokorna P, Heger Z, Adam V. An insight into the complex roles of metallothioneins in malignant diseases with emphasis on (sub)isoforms/isoforms and epigenetics phenomena. Pharmacol Ther 2017; 183:90-117. [PMID: 28987322 DOI: 10.1016/j.pharmthera.2017.10.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metallothioneins (MTs) belong to a group of small cysteine-rich proteins that are ubiquitous throughout all kingdoms. The main function of MTs is scavenging of free radicals and detoxification and homeostating of heavy metals. In humans, 16 genes localized on chromosome 16 have been identified to encode four MT isoforms labelled by numbers (MT-1-MT-4). MT-2, MT-3 and MT-4 proteins are encoded by a single gene. MT-1 comprises many (sub)isoforms. The known active MT-1 genes are MT-1A, -1B, -1E, -1F, -1G, -1H, -1M and -1X. The rest of the MT-1 genes (MT-1C, -1D, -1I, -1J and -1L) are pseudogenes. The expression and localization of individual MT (sub)isoforms and pseudogenes vary at intra-cellular level and in individual tissues. Changes in MT expression are associated with the process of carcinogenesis of various types of human malignancies, or with a more aggressive phenotype and therapeutic resistance. Hence, MT (sub)isoform profiling status could be utilized for diagnostics and therapy of tumour diseases. This review aims on a comprehensive summary of methods for analysis of MTs at (sub)isoforms levels, their expression in single tumour diseases and strategies how this knowledge can be utilized in anticancer therapy.
Collapse
Affiliation(s)
- Sona Krizkova
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, CZ-616 00 Brno, Czech Republic; Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic
| | - Marta Kepinska
- Department of Biomedical and Environmental Analysis, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| | - Gabriella Emri
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei krt 98, H-4032 Debrecen, Hungary
| | - Tomas Eckschlager
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University, and University Hospital Motol, V Uvalu 84, CZ-150 06 Prague 5, Czech Republic
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, CZ-128 40 Prague 2, Czech Republic
| | - Petra Pokorna
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, CZ-128 40 Prague 2, Czech Republic; Department of Oncology, 2nd Faculty of Medicine, Charles University, and University Hospital Motol, V Uvalu 84, CZ-150 06 Prague 5, Czech Republic
| | - Zbynek Heger
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, CZ-616 00 Brno, Czech Republic; Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic
| | - Vojtech Adam
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, CZ-616 00 Brno, Czech Republic; Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic.
| |
Collapse
|
56
|
Okajima W, Komatsu S, Ichikawa D, Miyamae M, Ohashi T, Imamura T, Kiuchi J, Nishibeppu K, Arita T, Konishi H, Shiozaki A, Morimura R, Ikoma H, Okamoto K, Otsuji E. Liquid biopsy in patients with hepatocellular carcinoma: Circulating tumor cells and cell-free nucleic acids. World J Gastroenterol 2017; 23:5650-5668. [PMID: 28883691 PMCID: PMC5569280 DOI: 10.3748/wjg.v23.i31.5650] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/14/2017] [Accepted: 07/04/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC), with its high incidence and mortality rate, is one of the most common malignant tumors. Despite recent development of a diagnostic and treatment method, the prognosis of HCC remains poor. Therefore, to provide optimal treatment for each patient with HCC, more precise and effective biomarkers are urgently needed which could facilitate a more detailed individualized decision-making during HCC treatment, including the following; risk assessment, early cancer detection, prediction of treatment or prognostic outcome. In the blood of cancer patients, accumulating evidence about circulating tumor cells and cell-free nucleic acids has suggested their potent clinical utilities as novel biomarker. This concept, so-called "liquid biopsy" is widely known as an alternative approach to cancer tissue biopsy. This method might facilitate a more sensitive diagnosis and better decision-making by obtaining genetic and epigenetic aberrations that are closely associated with cancer initiation and progression. In this article, we review recent developments based on the available literature on both circulating tumor cells and cell-free nucleic acids in cancer patients, especially focusing on Hepatocellular carcinoma.
Collapse
Affiliation(s)
- Wataru Okajima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Daisuke Ichikawa
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Mahito Miyamae
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Taisuke Imamura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Keiji Nishibeppu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Ryo Morimura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hisashi Ikoma
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
57
|
Agarwal R, Narayan J, Bhattacharyya A, Saraswat M, Tomar AK. Gene expression profiling, pathway analysis and subtype classification reveal molecular heterogeneity in hepatocellular carcinoma and suggest subtype specific therapeutic targets. Cancer Genet 2017; 216-217:37-51. [PMID: 29025594 DOI: 10.1016/j.cancergen.2017.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/12/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023]
Abstract
A very low 5-year survival rate among hepatocellular carcinoma (HCC) patients is mainly due to lack of early stage diagnosis, distant metastasis and high risk of postoperative recurrence. Hence ascertaining novel biomarkers for early diagnosis and patient specific therapeutics is crucial and urgent. Here, we have performed a comprehensive analysis of the expression data of 423 HCC patients (373 tumors and 50 controls) downloaded from The Cancer Genome Atlas (TCGA) followed by pathway enrichment by gene ontology annotations, subtype classification and overall survival analysis. The differential gene expression analysis using non-parametric Wilcoxon test revealed a total of 479 up-regulated and 91 down-regulated genes in HCC compared to controls. The list of top differentially expressed genes mainly consists of tumor/cancer associated genes, such as AFP, THBS4, LCN2, GPC3, NUF2, etc. The genes over-expressed in HCC were mainly associated with cell cycle pathways. In total, 59 kinases associated genes were found over-expressed in HCC, including TTK, MELK, BUB1, NEK2, BUB1B, AURKB, PLK1, CDK1, PKMYT1, PBK, etc. Overall four distinct HCC subtypes were predicted using consensus clustering method. Each subtype was unique in terms of gene expression, pathway enrichment and median survival. Conclusively, this study has exposed a number of interesting genes which can be exploited in future as potential markers of HCC, diagnostic as well as prognostic and subtype classification may guide for improved and specific therapy.
Collapse
Affiliation(s)
- Rahul Agarwal
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Jitendra Narayan
- Unité de recherche en biologie environnementale et évolutive (URBE), University of Namur, Belgium
| | | | - Mayank Saraswat
- Transplantation Laboratory, Haartmaninkatu 3, University of Helsinki, Helsinki, Finland
| | - Anil Kumar Tomar
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.
| |
Collapse
|
58
|
Tian MM, Fan YC, Zhao J, Gao S, Zhao ZH, Chen LY, Wang K. Hepatocellular carcinoma suppressor 1 promoter hypermethylation in serum. A diagnostic and prognostic study in hepatitis B. Clin Res Hepatol Gastroenterol 2017; 41:171-180. [PMID: 28189396 DOI: 10.1016/j.clinre.2016.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/20/2016] [Accepted: 10/18/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND Liver cancer ranks as the second leading cause of cancer-related mortality in man worldwide, and hepatocellular carcinoma (HCC) is the most prevalent malignant neoplasm of the liver. The sensitivity of alpha-fetoprotein (AFP) as an HCC diagnostic marker for HCC diagnosis is 39-65%, and one-third patients with HCC are missed using AFP. New biomarkers are needed to diagnose HCC at an earlier stage and to individualize treatment strategies. Hepatocellular carcinoma suppressor 1 (HCCS1) is a newly identified liver tumor suppressor gene. OBJECTIVE Our study evaluated the diagnostic value of serum HCCS1 promoter methylation in patients with HCC associated with hepatitis B. METHODS We determined the methylation status of serum HCCS1 promoter in 120 patients with HCC, 146 patients with chronic hepatitis B (CHB) and 27 healthy controls (HCs) by methylation-specific polymerase chain reaction (MSP). Evaluation of a cohort with 63 patients with HCC and 44 patients with CHB was set as a validation dataset. RESULTS The frequency of HCCS1 promoter methylation in patients with HCC was significantly higher than that in patients with CHB (P<0.001) and HCs (P<0.001), and was associated with tumor node-metastasis (TNM) stage (P=0.01). The sensitivity of serum HCCS1 promoter methylation for discriminating patients with HCC from CHB was 62.5% and that of AFP alone was 55%. Notably, the sensitivity of serum HCCS1 promoter methylation plus AFP level was 81.7%. CONCLUSION HCCS1 has potential as a biomarker for diagnosis and prognosis of patients with HCC.
Collapse
Affiliation(s)
- Ming-Ming Tian
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China; Institute of Hepatology, Shandong University, Jinan 250012, China
| | - Jing Zhao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Shuai Gao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Ze-Hua Zhao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Long-Yan Chen
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China; Institute of Hepatology, Shandong University, Jinan 250012, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China; Institute of Hepatology, Shandong University, Jinan 250012, China.
| |
Collapse
|
59
|
Yang Z, Zhang T, Gao H. Genetic aspects of pituitary carcinoma: A systematic review. Medicine (Baltimore) 2016; 95:e5268. [PMID: 27893664 PMCID: PMC5134857 DOI: 10.1097/md.0000000000005268] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 09/27/2016] [Accepted: 10/08/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Pituitary carcinoma (PC) is a rare type of malignant intracranial neoplasm defined as distant metastasis of pituitary adenoma (PA). Although PC incidence is low because only 0.1% to 0.2% of PAs ultimately develop into PCs, the prognosis is poor and 66% of patients die within the first year. Existing therapeutic measures, including surgical removal, chemotherapy, and radiotherapy, have limited effectiveness. The lack of efficacy of current treatments is largely caused by the limited understanding of the molecular pathogenesis of PA and the malignant transformation to PC. Therefore, the aim of this systematic review was to summarize published research regarding gene and protein expression in PC to clarify the molecular mechanisms underlying PC genesis and development and identify new candidate diagnostic biomarkers and therapeutic targets for potential use in personalized treatment of PC. METHODS We followed the PRISMA guidelines to plan and conduct this systematic review. PubMed, Embase, and Web of Science databases were searched for relevant studies conducted before December 16, 2015 describing the association of PC with gene expression at the mRNA and protein levels. MeSH terms combined with free terms were used to retrieve the references. RESULTS In total, 207 records were obtained by primary search, and 32 were included in the systematic review. Compared with normal pituitary gland and/or PA, 30 and 18 genes were found to have higher or lower expression, respectively, in PCs using different analytical methods. Among them, we selected 9 upregulated and 7 downregulated genes for further analysis based on their identification as candidate treatment targets in other cancers, potential clinical application, or further research value. CONCLUSION Previous studies demonstrated that many genes promote PC malignant transformation, angiogenesis, invasion, metastasis, and recurrence. Although most of these genes and proteins have not been fully analyzed with regard to their downstream mechanisms or potential diagnostic and therapeutic application, they have the potential to become candidate PC biomarkers and/or molecular targets for guiding personalized treatment. Modern advanced technologies should be utilized in future research to identify more candidate genes for PC pathogenesis, as precisely targeted gene therapies against PC are urgently required.
Collapse
Affiliation(s)
- Zijiang Yang
- Department of Neurosurgery, Jiangyin People's Hospital Affiliated to Nantong University
| | - Ting Zhang
- Central Laboratory, Jiangyin People's Hospital Affiliated to Nantong University
| | - Heng Gao
- Department of Neurosurgery, Jiangyin people's Hospital Affiliated to Nantong University, Jiangyin, Wuxi, China
| |
Collapse
|
60
|
Dong X, Ding W, Ye J, Yan D, Xue F, Xu L, Yin J, Guo W. MiR-24-3p enhances cell growth in hepatocellular carcinoma by targeting metallothionein 1M. Cell Biochem Funct 2016; 34:491-496. [PMID: 27650047 DOI: 10.1002/cbf.3213] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 08/14/2016] [Accepted: 08/15/2016] [Indexed: 12/12/2022]
Abstract
Dysregulation of microRNAs has been demonstrated to contribute to malignant progression of cancers, including hepatocellular carcinoma (HCC). MiR-24-3p was previously reported to be significantly upregulated in HCC. However, the potential role and mechanism of action of miR-24-3p in the initiation and progression of HCC remain largely unknown. Quantitative reverse transcription polymerase chain reaction demonstrated that miR-24-3p was significantly upregulated in HCC tumor tissues compared with nontumor tissues. The cell viability, colony formation assay, and tumorigenicity assays in nude mice showed that miR-24-3p could enhance HCC cell growth in vitro and in vivo. Metallothionein 1M was verified as an miR-24-3p target gene by using dual-luciferase reporter assays, quantitative reverse transcription polymerase chain reaction, and Western blotting, which was involved in miR-24-3p regulated HCC cell growth. These results indicated that miR-24-3p plays an important role in the initiation and progression of HCC by targeting metallothionein 1M, and the miR-24-3p/metallothionein 1M pathway may contribute to the development of novel therapeutic strategies for HCC in the future.
Collapse
Affiliation(s)
- Xiaogang Dong
- Department of Hepatopancreatobiliary Surgery, Cancer Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Wei Ding
- Department of Hepatopancreatobiliary Surgery, Cancer Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jianwei Ye
- Department of Cancer Center, The first affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Dong Yan
- Department of Hepatopancreatobiliary Surgery, Cancer Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Feng Xue
- Department of Hepatopancreatobiliary Surgery, Cancer Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Lin Xu
- Department of Hepatopancreatobiliary Surgery, Cancer Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jiwei Yin
- Department of Hepatopancreatobiliary Surgery, Cancer Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Wenjia Guo
- Department of Cancer Research Institute, Cancer Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| |
Collapse
|
61
|
Microarray-based identification of genes associated with cancer progression and prognosis in hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:127. [PMID: 27567667 PMCID: PMC5002170 DOI: 10.1186/s13046-016-0403-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 08/09/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths. The average survival and 5-year survival rates of HCC patients still remains poor. Thus, there is an urgent need to better understand the mechanisms of cancer progression in HCC and to identify useful biomarkers to predict prognosis. METHODS Public data portals including Oncomine, The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) profiles were used to retrieve the HCC-related microarrays and to identify potential genes contributed to cancer progression. Bioinformatics analyses including pathway enrichment, protein/gene interaction and text mining were used to explain the potential roles of the identified genes in HCC. Quantitative real-time polymerase chain reaction analysis and Western blotting were used to measure the expression of the targets. The data were analysed by SPSS 20.0 software. RESULTS We identified 80 genes that were significantly dysregulated in HCC according to four independent microarrays covering 386 cases of HCC and 327 normal liver tissues. Twenty genes were consistently and stably dysregulated in the four microarrays by at least 2-fold and detection of gene expression by RT-qPCR and western blotting showed consistent expression profiles in 11 HCC tissues compared with corresponding paracancerous tissues. Eleven of these 20 genes were associated with disease-free survival (DFS) or overall survival (OS) in a cohort of 157 HCC patients, and eight genes were associated with tumour pathologic PT, tumour stage or vital status. Potential roles of those 20 genes in regulation of HCC progression were predicted, primarily in association with metastasis. INTS8 was specifically correlated with most clinical characteristics including DFS, OS, stage, metastasis, invasiveness, diagnosis, and age. CONCLUSION The significantly dysregulated genes identified in this study were associated with cancer progression and prognosis in HCC, and might be potential therapeutic targets for HCC treatment or potential biomarkers for diagnosis and prognosis.
Collapse
|
62
|
Houessinon A, François C, Sauzay C, Louandre C, Mongelard G, Godin C, Bodeau S, Takahashi S, Saidak Z, Gutierrez L, Régimbeau JM, Barget N, Barbare JC, Ganne N, Chauffert B, Coriat R, Galmiche A. Metallothionein-1 as a biomarker of altered redox metabolism in hepatocellular carcinoma cells exposed to sorafenib. Mol Cancer 2016; 15:38. [PMID: 27184800 PMCID: PMC4894370 DOI: 10.1186/s12943-016-0526-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/12/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Sorafenib, a kinase inhibitor active against various solid tumours, induces oxidative stress and ferroptosis, a new form of oxidative necrosis, in some cancer cells. Clinically-applicable biomarkers that reflect the impact of sorafenib on the redox metabolism of cancer cells are lacking. METHODS We used gene expression microarrays, real-time PCR, immunoblot, protein-specific ELISA, and gene reporter constructs encoding the enzyme luciferase to study the response of a panel of cancer cells to sorafenib. Tumour explants prepared from surgical hepatocellular carcinoma (HCC) samples and serum samples obtained from HCC patients receiving sorafenib were also used. RESULTS We observed that genes of the metallothionein-1 (MT1) family are induced in the HCC cell line Huh7 exposed to sorafenib. Sorafenib increased the expression of MT1G mRNA in a panel of human cancer cells, an effect that was not observed with eight other clinically-approved kinase inhibitors. We identified the minimal region of the MT1G promoter that confers inducibility by sorafenib to a 133 base pair region containing an Anti-oxidant Response Element (ARE) and showed the essential role of the transcription factor NRF2 (Nuclear factor erythroid 2-Related Factor 2). We examined the clinical relevance of our findings by analysing the regulation of MT1G in five tumour explants prepared from surgical HCC samples. Finally, we showed that the protein levels of MT1 increase in the serum of some HCC patients receiving sorafenib, and found an association with reduced overall survival. CONCLUSION These findings indicate that MT1 constitute a biomarker adapted for exploring the impact of sorafenib on the redox metabolism of cancer cells.
Collapse
Affiliation(s)
- Aline Houessinon
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Amiens Sud, Avenue Laennec, 80054, Amiens, Cedex, France.,EA4666, Université de Picardie Jules Verne (UPJV), Amiens, France
| | | | - Chloé Sauzay
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Amiens Sud, Avenue Laennec, 80054, Amiens, Cedex, France.,EA4666, Université de Picardie Jules Verne (UPJV), Amiens, France
| | - Christophe Louandre
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Amiens Sud, Avenue Laennec, 80054, Amiens, Cedex, France.,EA4666, Université de Picardie Jules Verne (UPJV), Amiens, France
| | - Gaelle Mongelard
- Centre de Ressources en Biologie Moléculaire, UPJV, Amiens, France
| | - Corinne Godin
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Amiens Sud, Avenue Laennec, 80054, Amiens, Cedex, France.,EA4666, Université de Picardie Jules Verne (UPJV), Amiens, France
| | | | - Shinichiro Takahashi
- Department of clinical laboratory, Tohoku Medical and Pharmaceutical University Hospital, Sendai, Japan
| | - Zuzana Saidak
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Amiens Sud, Avenue Laennec, 80054, Amiens, Cedex, France
| | | | | | - Nathalie Barget
- Centre de Ressources Biologiques, Hôpitaux Universitaires Paris Seine-Saint-Denis, APHP, Bondy, France
| | | | - Nathalie Ganne
- Service d'Hépatologie, Pôle d'Activités Cancérologiques Spécialisées, APHP, Hôpitaux Universitaires Paris Seine-Saint-Denis, Site Jean Verdier, Bondy, France.,Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Bobigny, France.,Inserm UMR-1162, Génomique Fonctionnelle des Tumeurs solides, Paris, France
| | - Bruno Chauffert
- EA4666, Université de Picardie Jules Verne (UPJV), Amiens, France
| | - Romain Coriat
- Service de Gastroentérologie et d'Endoscopie, Hôpital Cochin, APHP, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UFR de médecine, Paris, France
| | - Antoine Galmiche
- Laboratoire de Biochimie, Centre de Biologie Humaine (CBH), CHU Amiens Sud, Avenue Laennec, 80054, Amiens, Cedex, France. .,EA4666, Université de Picardie Jules Verne (UPJV), Amiens, France.
| |
Collapse
|
63
|
Liao W, Mao Y, Ge P, Yang H, Xu H, Lu X, Sang X, Zhong S. Value of quantitative and qualitative analyses of circulating cell-free DNA as diagnostic tools for hepatocellular carcinoma: a meta-analysis. Medicine (Baltimore) 2015; 94:e722. [PMID: 25860220 PMCID: PMC4554041 DOI: 10.1097/md.0000000000000722] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/08/2015] [Accepted: 03/09/2015] [Indexed: 12/24/2022] Open
Abstract
Qualitative and quantitative analyses of circulating cell-free DNA (cfDNA) are potential methods for the detection of hepatocellular carcinoma (HCC). Many studies have evaluated these approaches, but the results have been variable. This meta-analysis is the first to synthesize these published results and evaluate the use of circulating cfDNA values for HCC diagnosis. All articles that met our inclusion criteria were assessed using QUADAS guidelines after the literature research. We also investigated 3 subgroups in this meta-analysis: qualitative analysis of abnormal concentrations of circulating cfDNA; qualitative analysis of single-gene methylation alterations; and multiple analyses combined with alpha-fetoprotein (AFP). Statistical analyses were performed using the software Stata 12.0. We synthesized these published results and calculated accuracy measures (pooled sensitivity and specificity, positive/negative likelihood ratios [PLRs/NLRs], diagnostic odds ratios [DORs], and corresponding 95% confidence intervals [95% CIs]). Data were pooled using bivariate generalized linear mixed model. Furthermore, summary receiver operating characteristic curves and area under the curve (AUC) were used to summarize overall test performance. Heterogeneity and publication bias were also examined. A total of 2424 subjects included 1280 HCC patients in 22 studies were recruited in this meta-analysis. Pooled sensitivity and specificity, PLR, NLR, DOR, AUC, and CIs of quantitative analysis were 0.741 (95% CI: 0.610-0.840), 0.851 (95% CI: 0.718-0.927), 4.970 (95% CI: 2.694-9.169), 0.304 (95% CI: 0.205-0.451), 16.347 (95% CI: 8.250-32.388), and 0.86 (95% CI: 0.83-0.89), respectively. For qualitative analysis, the values were 0.538 (95% CI: 0.401-0.669), 0.944 (95% CI: 0.889-0.972), 9.545 (95% CI: 5.298-17.196), 0.490 (95% CI: 0.372-0.646), 19.491 (95% CI: 10.458-36.329), and 0.87 (95% CI: 0.84-0.90), respectively. After combining with AFP assay, the values were 0.818 (95% CI: 0.676-0.906), 0.960 (95% CI: 0.873-0.988), 20.195 (95% CI: 5.973-68.282), 0.190 (95% CI: 0.100-0.359), 106.270 (95% CI: 22.317-506.055), and 0.96 (95% CI: 0.94-0.97), respectively. The results in this meta-analysis suggest that circulating cfDNA have potential value for HCC diagnosis. However, it would not be recommended for using independently, which is based on the nonrobust results. After combining with AFP, the diagnostic performance will be improved. Further investigation with more data is needed.
Collapse
Affiliation(s)
- Wenjun Liao
- From the Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | | | | | | | | | | | | | | |
Collapse
|
64
|
Li DJ, Zhang GY, Jie ZG, Li ZR. MicroRNAs related to invasiveness and metastasis of gastric cancer. Shijie Huaren Xiaohua Zazhi 2014; 22:3219-3224. [DOI: 10.11569/wcjd.v22.i22.3219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Metastasis plays an important role in the prognosis of patients with cancer. It is known that several steps are necessary for clonal cells to disseminate from their primary tumor site and colonize distant tissues. It will provide useful insights for effective treatment of cancer to investigate the molecular actors regulating this process. MicroRNAs, 19-25 nt in length, are a class of non-coding RNA, and they can result in degradation of specific mRNAs or inhibit their translation. They have been known as negative regulators of gene expression and are involved in many biological processes, including cell growth, differentiation and apoptosis. The relationship between the abnormal expression of microRNAs and tumors has been widely studied. Some recent research has clarified the role of microRNAs in tumor invasion and metastases. This paper reviews the recent progress in research of microRNAs related to invasiveness and metastasis of gastric cancer.
Collapse
|
65
|
Gao S, Wang K. DNA methylation in liver diseases. World J Clin Infect Dis 2014; 4:41. [DOI: 10.5495/wjcid.v4.i4.41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 07/24/2014] [Accepted: 10/10/2014] [Indexed: 02/06/2023] Open
|