51
|
Fang S, Zhang D, Weng W, Lv X, Zheng L, Chen M, Fan X, Mao J, Mao C, Ye Y, Xu M, Ji J. CPSF7 regulates liver cancer growth and metastasis by facilitating WWP2-FL and targeting the WWP2/PTEN/AKT signaling pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118624. [PMID: 31837982 DOI: 10.1016/j.bbamcr.2019.118624] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 11/15/2019] [Accepted: 12/10/2019] [Indexed: 01/01/2023]
Abstract
Alternative splicing within a gene can create different versions of an mRNA, called isoforms. CFIm, composed of a small subunit CFIm25 and two large subunits CFIm68 and CFIm59 (also known as CPSF7), has been proposed as an enhancer-dependent activator of mRNA 3' processing. In this study, we investigated the role of CPSF7 in hepatocellular carcinoma. Experimental evidence suggests that the expression level of CPSF7 is higher in liver cancer cells and tissues than in non-tumor hepatic cells and tissues. Furthermore, knockdown of CPSF7 effectively suppressed cell proliferation, migration and colony formation in liver cancer cells by inhibiting PTEN/AKT signaling. CPSF7 promoted WWP2-FL due to the presence of PTEN ubiquitination sites in this longer transcript. Taken together, we identified that CPSF7 regulates liver cancer growth by targeting WWP2-FL that in turn regulates AKT activation in a PTEN-dependent manner.
Collapse
Affiliation(s)
- Shiji Fang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China
| | - Dengke Zhang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China
| | - Wei Weng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China
| | - Xiuling Lv
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China
| | - Liyun Zheng
- Lishui Hospital of Zhejiang University, China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China
| | - Xiaoxi Fan
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China
| | - Jianting Mao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China
| | - Chenchen Mao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China
| | - Yani Ye
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China
| | - Min Xu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China.
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University/Affiliated Lishui Hospital of Zhejiang University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China; Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui 323000, China.
| |
Collapse
|
52
|
Ni HM, Chao X, Yang H, Deng F, Wang S, Bai Q, Qian H, Cui Y, Cui W, Shi Y, Zong WX, Wang Z, Yang L, Ding WX. Dual Roles of Mammalian Target of Rapamycin in Regulating Liver Injury and Tumorigenesis in Autophagy-Defective Mouse Liver. Hepatology 2019; 70:2142-2155. [PMID: 31095752 PMCID: PMC6858484 DOI: 10.1002/hep.30770] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022]
Abstract
Autophagy is a lysosomal degradation pathway that degrades cytoplasmic proteins and organelles. Absence of autophagy in hepatocytes has been linked to promoting liver injury and tumorigenesis; however, the mechanisms behind why a lack of autophagy induces these complications are not fully understood. The role of mammalian target of rapamycin (mTOR) in impaired autophagy-induced liver pathogenesis and tumorigenesis was investigated by using liver-specific autophagy related 5 knockout (L-ATG5 KO) mice, L-ATG5/mTOR, and L-ATG5/Raptor double knockout (DKO) mice. We found that deletion of mTOR or Raptor in L-ATG5 KO mice at 2 months of age attenuated hepatomegaly, cell death, and inflammation but not fibrosis. Surprisingly, at 6 months of age, L-ATG5/mTOR DKO and L-ATG5/Raptor DKO mice also had increased hepatic inflammation, fibrosis, and liver injury, similar to the L-ATG5 KO mice. Moreover, more than 50% of L-ATG5/mTOR DKO and L-ATG5/Raptor DKO mice already developed spontaneous tumors, but none of the L-ATG5 KO mice had developed any tumors at 6 months of age. At 9 months of age, all L-ATG5/mTOR DKO and L-ATG5/Raptor DKO had developed liver tumors. Mechanistically, L-ATG5/mTOR DKO and L-ATG5/Raptor DKO mice had decreased levels of hepatic ubiquitinated proteins and persistent nuclear erythroid 2 p45-related factor 2 activation but had increased Akt activation compared with L-ATG5 KO mice. Conclusion: Loss of mTOR signaling attenuates the liver pathogenesis in mice with impaired hepatic autophagy but paradoxically promotes tumorigenesis in mice at a relatively young age. Therefore, the balance of mTOR is critical in regulating the liver pathogenesis and tumorigenesis in mice with impaired hepatic autophagy.
Collapse
Affiliation(s)
- Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics,
The University of Kansas Medical Center, Kansas City, KS 66160;,Correspondence to: Hong-Min Ni, Department of
Pharmacology, Toxicology and Therapeutics; The University of Kansas Medical
Center; MS 1018; 3901 Rainbow Blvd.; Kansas City, Kansas 66160; Phone:
913-588-9813, Fax: 913-588-7501; ; or Wen-Xing Ding,
Department of Pharmacology, Toxicology and Therapeutics; The University of
Kansas Medical Center; MS 1018; 3901 Rainbow Blvd. Kansas City, Kansas 66160;
Phone: 913-588-9813, Fax: 913-588-7501;
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics,
The University of Kansas Medical Center, Kansas City, KS 66160
| | - Hua Yang
- Department of Pharmacology, Toxicology and Therapeutics,
The University of Kansas Medical Center, Kansas City, KS 66160;,Department of General Surgery, Shanghai Public Health
Clinical Center, Fudan University, Shanghai, China
| | - Fengyan Deng
- Department of Pharmacology, Toxicology and Therapeutics,
The University of Kansas Medical Center, Kansas City, KS 66160
| | - Shaogui Wang
- Department of Pharmacology, Toxicology and Therapeutics,
The University of Kansas Medical Center, Kansas City, KS 66160
| | - Qingyun Bai
- Department of Pharmacology, Toxicology and Therapeutics,
The University of Kansas Medical Center, Kansas City, KS 66160;,Institute of Chinese Materia Medica, Shanghai University
of Traditional Chinese Medicine, Shanghai, China;,School of chemical and biological engineering, Yichun
University, Jiangxi, China
| | - Hui Qian
- Department of Pharmacology, Toxicology and Therapeutics,
The University of Kansas Medical Center, Kansas City, KS 66160
| | - Yue Cui
- Department of Environmental and Occupational Health
Sciences, University of Washington; Seattle, WA 98159, USA
| | - Wei Cui
- Department of Pathology, The University of Kansas Medical
Center, Kansas City, KS 66160
| | - Yinghong Shi
- Department of Liver Surgery & Transplantation, Liver
Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei-Xing Zong
- Department of Chemical Biology, Ernest Mario School of
Pharmacy, Rutgers University, 164 Frelinghuysen Road, Piscataway, NJ 08854,
USA
| | - Zhengtao Wang
- Institute of Chinese Materia Medica, Shanghai University
of Traditional Chinese Medicine, Shanghai, China
| | - Li Yang
- Institute of Chinese Materia Medica, Shanghai University
of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics,
The University of Kansas Medical Center, Kansas City, KS 66160;,Correspondence to: Hong-Min Ni, Department of
Pharmacology, Toxicology and Therapeutics; The University of Kansas Medical
Center; MS 1018; 3901 Rainbow Blvd.; Kansas City, Kansas 66160; Phone:
913-588-9813, Fax: 913-588-7501; ; or Wen-Xing Ding,
Department of Pharmacology, Toxicology and Therapeutics; The University of
Kansas Medical Center; MS 1018; 3901 Rainbow Blvd. Kansas City, Kansas 66160;
Phone: 913-588-9813, Fax: 913-588-7501;
| |
Collapse
|
53
|
Li X, Wang H, Li T, Wang L, Wu X, Liu J, Xu Y, Wei W. Circulating tumor DNA/circulating tumor cells and the applicability in different causes induced hepatocellular carcinoma. Curr Probl Cancer 2019; 44:100516. [PMID: 31836136 DOI: 10.1016/j.currproblcancer.2019.100516] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/31/2019] [Accepted: 09/30/2019] [Indexed: 12/25/2022]
Abstract
In 2015, liquid biopsy was rated one of the top 10 breakthrough technologies of the year by MIT Technology Review. Liquid biopsy is a type of in vitro diagnostic method involving a noninvasive blood test. It is also a breakthrough technology used to detect tumors and cancers and assist in therapeutic strategies. The most widely used markers are circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). Primary carcinoma of the liver is a malignancy of hepatocytes or intrahepatic biliary epithelial cells. The most common type of liver cancer is hepatocellular carcinoma (HCC), the causes of which mainly include infection with hepatitis B virus (HBV) and/or hepatitis C virus (HCV), alcohol abuse, aflatoxicosis, and nonalcoholic fatty liver disease/ nonalcoholic steatohepatitis. As there are few typical clinical characteristics during the early stage of the disease, early diagnosis of HCC is very challenging. However, CTCs and ctDNA carry tumor-specific information. Therefore, the detection and analysis of CTCs and ctDNA can provide evidence for the early diagnosis of HCC and guide treatment. Furthermore, several studies have indicated that different inducers of HCC cause different DNA mutations, and accordingly, detection of specific mutations in ctDNA will facilitate the determination of the HCC type and help physicians provide distinctive therapies.
Collapse
Affiliation(s)
- Xuemei Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huihui Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tao Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Anti-inflammatory and Immune Medicine innovation team, Hefei, China.
| | - Lianzi Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xian Wu
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiaqing Liu
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanhong Xu
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Anti-inflammatory and Immune Medicine innovation team, Hefei, China.
| |
Collapse
|
54
|
Rahmani F, Ziaeemehr A, Shahidsales S, Gharib M, Khazaei M, Ferns GA, Ryzhikov M, Avan A, Hassanian SM. Role of regulatory miRNAs of the PI3K/AKT/mTOR signaling in the pathogenesis of hepatocellular carcinoma. J Cell Physiol 2019; 235:4146-4152. [PMID: 31663122 DOI: 10.1002/jcp.29333] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/30/2019] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the common malignant human tumors with high morbidity worldwide. Aberrant activation of the oncogenic phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling is related to clinicopathological features of HCC. Emerging data revealed that microRNAs (miRNAs) have prominent implications for regulating cellular proliferation, differentiation, apoptosis, and metabolism through targeting the PI3K/AKT/mTOR signaling axis. The recognition of the crucial role of miRNAs in hepatocarcinogenesis represents a promising area to identify novel anticancer therapeutics for HCC. The present study summarizes the major findings about the regulatory role of miRNAs in the PI3K/AKT/mTOR pathway in the pathogenesis of HCC.
Collapse
Affiliation(s)
- Farzad Rahmani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aghigh Ziaeemehr
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Masoumeh Gharib
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Brighton, UK
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Washington University, Saint Louis, Missouri
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed M Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
55
|
Pascale RM, Simile MM, Peitta G, Seddaiu MA, Feo F, Calvisi DF. Experimental Models to Define the Genetic Predisposition to Liver Cancer. Cancers (Basel) 2019; 11:cancers11101450. [PMID: 31569678 PMCID: PMC6826893 DOI: 10.3390/cancers11101450] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/24/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a frequent human cancer and the most frequent liver tumor. The study of genetic mechanisms of the inherited predisposition to HCC, implicating gene-gene and gene-environment interaction, led to the discovery of multiple gene loci regulating the growth and multiplicity of liver preneoplastic and neoplastic lesions, thus uncovering the action of multiple genes and epistatic interactions in the regulation of the individual susceptibility to HCC. The comparative evaluation of the molecular pathways involved in HCC development in mouse and rat strains differently predisposed to HCC indicates that the genes responsible for HCC susceptibility control the amplification and/or overexpression of c-Myc, the expression of cell cycle regulatory genes, and the activity of Ras/Erk, AKT/mTOR, and of the pro-apoptotic Rassf1A/Nore1A and Dab2IP/Ask1 pathways, the methionine cycle, and DNA repair pathways in mice and rats. Comparative functional genetic studies, in rats and mice differently susceptible to HCC, showed that preneoplastic and neoplastic lesions of resistant mouse and rat strains cluster with human HCC with better prognosis, while the lesions of susceptible mouse and rats cluster with HCC with poorer prognosis, confirming the validity of the studies on the influence of the genetic predisposition to hepatocarinogenesis on HCC prognosis in mouse and rat models. Recently, the hydrodynamic gene transfection in mice provided new opportunities for the recognition of genes implicated in the molecular mechanisms involved in HCC pathogenesis and prognosis. This method appears to be highly promising to further study the genetic background of the predisposition to this cancer.
Collapse
Affiliation(s)
- Rosa M Pascale
- Department of Medical, Surgical and Experimental Sciences, Via P. Manzella 4, 07100 Sassari, Italy.
| | - Maria M Simile
- Department of Medical, Surgical and Experimental Sciences, Via P. Manzella 4, 07100 Sassari, Italy.
| | - Graziella Peitta
- Department of Medical, Surgical and Experimental Sciences, Via P. Manzella 4, 07100 Sassari, Italy.
| | - Maria A Seddaiu
- Department of Medical, Surgical and Experimental Sciences, Via P. Manzella 4, 07100 Sassari, Italy.
| | - Francesco Feo
- Department of Medical, Surgical and Experimental Sciences, Via P. Manzella 4, 07100 Sassari, Italy.
| | - Diego F Calvisi
- Department of Medical, Surgical and Experimental Sciences, Via P. Manzella 4, 07100 Sassari, Italy.
| |
Collapse
|
56
|
Preclinical efficacy of a novel dual PI3K/mTOR inhibitor, CMG002, alone and in combination with sorafenib in hepatocellular carcinoma. Cancer Chemother Pharmacol 2019; 84:809-817. [PMID: 31385002 DOI: 10.1007/s00280-019-03918-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/29/2019] [Indexed: 12/23/2022]
Abstract
PURPOSE Sorafenib has been the only first systemic drug that improves survival of patients with advanced hepatocellular carcinoma (HCC). However, because the response rate of sorafenib is relatively low, novel therapeutic strategies are needed to improve survival in patients with HCC. This study investigated the effect of CMG002 alone and in combination with sorafenib on HCC in vitro and vivo. METHODS The effect of a newly developed dual PI3K/mTOR inhibitor, CMG002, on the proliferation of Huh-7 and HepG2 HCC cells was investigated using the MTT assay. Western blotting was performed to assess phosphorylation of the key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. HepG2 cells were inoculated into mice, which were treated with vehicle, sorafenib, CMG002, and their combinations. Tumor cell proliferation and tumor angiogenesis were evaluated by immunohistochemical analysis of Ki-67 and CD31, respectively. Tumor cell apoptosis was detected by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Levels of key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways were evaluated by western blot analysis. RESULTS The combination of sorafenib and CMG002 additively inhibited Huh-7 and HepG2 cell proliferation compared to single-agent treatment. Sorafenib and CMG002 as single agents differentially inhibited or activated key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. The combination of sorafenib and CMG002 inhibited all key enzymes in the two pathways. Treatment with CMG002 for 4 weeks alone and in combination with sorafenib strongly inhibited tumor growth. CMG002 inhibited HCC cell proliferation, induced apoptosis, and decreased tumor angiogenesis. Furthermore, these effects were enhanced when CMG002 was combined with sorafenib. CONCLUSIONS The combination of CMG002 and sorafenib significantly inhibited HCC cell proliferation and tumorigenesis by inhibiting the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. These findings suggest that CMG002 to be a potential novel candidate treatment for HCC.
Collapse
|
57
|
Genome-wide promoter DNA methylation profiling of hepatocellular carcinomas arising either spontaneously or due to chronic exposure to Ginkgo biloba extract (GBE) in B6C3F1/N mice. Arch Toxicol 2019; 93:2219-2235. [PMID: 31278416 DOI: 10.1007/s00204-019-02505-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 06/26/2019] [Indexed: 12/15/2022]
Abstract
Epigenetic modifications, such as DNA methylation, play an important role in carcinogenesis. In a recent NTP study, chronic exposure of B6C3F1/N mice to Ginkgo biloba extract (GBE) resulted in a high incidence of hepatocellular carcinomas (HCC). Genome-wide promoter methylation profiling on GBE-exposed HCC (2000 mg/kg group), spontaneous HCC (vehicle-control group), and age-matched vehicle control liver was performed to identify differentially methylated genes in GBE-exposed HCC and spontaneous HCC. DNA methylation alterations were correlated to the corresponding global gene expression changes. Compared to control liver, 1296 gene promoters (719 hypermethylated, 577 hypomethylated) in GBE-exposed HCC and 738 (427 hypermethylated, 311 hypomethylated) gene promoters in spontaneous HCC were significantly differentially methylated, suggesting an impact of methylation on GBE-exposed HCC. Differential methylation of promoter regions in relevant cancer genes (cMyc, Spry2, Dusp5) and their corresponding differential gene expression was validated by quantitative pyrosequencing and qRT-PCR, respectively. In conclusion, we have identified differentially methylated promoter regions of relevant cancer genes altered in GBE-exposed HCC compared to spontaneous HCC. Further study of unique sets of differentially methylated genes in chemical-exposed mouse HCC could potentially be used to differentiate treatment-related tumors from spontaneous-tumors in cancer bioassays and provide additional understanding of the underlying epigenetic mechanisms of chemical carcinogenesis.
Collapse
|
58
|
Miura K, Ohnishi H, Morimoto N, Minami S, Ishioka M, Watanabe S, Tsukui M, Takaoka Y, Nomoto H, Isoda N, Yamamoto H. Ezetimibe suppresses development of liver tumors by inhibiting angiogenesis in mice fed a high-fat diet. Cancer Sci 2019; 110:771-783. [PMID: 30520543 PMCID: PMC6361611 DOI: 10.1111/cas.13902] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a common cause of liver cirrhosis and hepatocellular carcinoma (HCC). However, effective therapeutic strategies for preventing and treating NASH‐mediated liver cirrhosis and HCC are lacking. Cholesterol is closely associated with vascular endothelial growth factor (VEGF), a key factor that promotes HCC. Recent reports have demonstrated that statins could prevent HCC development. In contrast, we have little information on ezetimibe, an inhibitor of cholesterol absorption, in regards to the prevention of NASH‐related liver cirrhosis and HCC. In the present study, a steatohepatitis‐related HCC model, hepatocyte‐specific phosphatase and tensin homolog (Pten)‐deficient (PtenΔhep) mice were fed a high‐fat (HF) diet with/without ezetimibe. In the standard‐diet group, ezetimibe did not reduce the development of liver tumors in PtenΔhep mice, in which the increase of serum cholesterol levels was mild. Feeding of a HF diet increased serum cholesterol levels markedly and subsequently increased serum levels of VEGF, a crucial component of angiogenesis. The HF diet increased the number of VEGF‐positive cells and vascular endothelial cells in the tumors of PtenΔhep mice. Kupffer cells, macrophages in the liver, increased VEGF expression in response to fat overload. Ezetimibe treatment lowered cholesterol levels and these angiogenetic processes. As a result, ezetimibe also suppressed inflammation, liver fibrosis and tumor growth in PtenΔhep mice on the HF diet. Tumor cells were highly proliferative with HF‐diet feeding, which was inhibited by ezetimibe. In conclusion, ezetimibe suppressed development of liver tumors by inhibiting angiogenesis in PtenΔhep mice with hypercholesterolemia.
Collapse
Affiliation(s)
- Kouichi Miura
- Division of Gastroenterology, Department of Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hirohide Ohnishi
- Department of Gastroenterology, Saitama Medical Center, Jichi Medical University, Saitama, Japan.,Japan Organization of Occupational Health and Safety, Kanagawa, Japan
| | - Naoki Morimoto
- Division of Gastroenterology, Department of Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Shinichiro Minami
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Mitsuaki Ishioka
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Shunji Watanabe
- Division of Gastroenterology, Department of Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Mamiko Tsukui
- Division of Gastroenterology, Department of Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yoshinari Takaoka
- Division of Gastroenterology, Department of Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hiroaki Nomoto
- Division of Gastroenterology, Department of Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Norio Isoda
- Division of Gastroenterology, Department of Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hironori Yamamoto
- Division of Gastroenterology, Department of Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
59
|
Bai J, Yao B, Wang L, Sun L, Chen T, Liu R, Yin G, Xu Q, Yang W. lncRNA A1BG-AS1 suppresses proliferation and invasion of hepatocellular carcinoma cells by targeting miR-216a-5p. J Cell Biochem 2018; 120:10310-10322. [PMID: 30556161 DOI: 10.1002/jcb.28315] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022]
Abstract
Extensive evidence indicate that long noncoding RNAs (lncRNAs) regulates the tumorigenesis and progression of hepatocellular carcinoma (HCC). However, the expression and biological function of lncRNA A1BG antisense RNA 1 (A1BG-AS1) were poorly known in HCC. Here, we found the underexpression of A1BG-AS1 in HCC via analysis of The Cancer Genome Atlas database. Further analyses confirmed that A1BG-AS1 expression in HCC was markedly lower than that in noncancerous tissues based on our HCC cohort. Clinical association analysis revealed that low A1BG-AS1 expression correlated with poor prognostic features, such as microvascular invasion, high tumor grade, and advanced tumor stage. Follow-up data indicated that low A1BG-AS1 level evidently correlated with poor clinical outcomes of HCC patients. Moreover, forced expression of A1BG-AS1 repressed proliferation, migration, and invasion of HCC cells in vitro. Conversely, A1BG-AS1 knockdown promoted these malignant behaviors in HepG2 cells. Mechanistically, A1BG-AS1 functioned as a competing endogenous RNA by directly sponging miR-216a-5p in HCC cells. Notably, miR-216a-5p restoration rescued A1BG-AS1 attenuated proliferation, migration and invasion of HCCLM3 cells. A1BG-AS1 positively regulated the levels of phosphatase and tensin homolog and SMAD family member 7, which were reduced by miR-216a-5p in HCC cells. Altogether, we conclude that A1BG-AS1 exerts a tumor suppressive role in HCC progression.
Collapse
Affiliation(s)
- Jigang Bai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bowen Yao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Liang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Liankang Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Tianxiang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Runkun Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guozhi Yin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qiuran Xu
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Wei Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
60
|
Sou FM, Wu CK, Chang KC, Lu SN, Wang JH, Hung CH, Chen CH, Kee KM, Yen YH, Lin MT, Tsai MC, Hu TH. Clinical characteristics and prognosis of HCC occurrence after antiviral therapy for HCV patients between sustained and non-sustained responders. J Formos Med Assoc 2018; 118:504-513. [PMID: 30527565 DOI: 10.1016/j.jfma.2018.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/16/2018] [Accepted: 10/25/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Hepatitis C virus (HCV)-infected patients who achieved sustained virologic response (SVR) may still develop hepatocellular carcinoma (HCC). The characteristic of HCC and the prognosis between SVR and non-SVR patients were not well known. METHODS Among 1884 HCV-infected patients who were treated with pegylated IFN plus ribavirin therapies, 122 patients developed HCC during follow-up were enrolled in this study. Laboratory data were collected before and at least 1 year after IFN-based therapy, as well as the latest follow-up. RESULTS Both SVR and non-SVR patients had similar risk factors to develop HCC, but with a little difference. Liver cirrhosis plays a key role in HCC occurrence in both groups. Among the patients who developed HCC, non-SVR patients had significantly higher total bilirubin, higher FIB-4, lower pre-treatment platelet count, higher pre-treatment AFP levels and higher proportion of cirrhosis than SVR patients before occurrence of HCC. After curative treatment, SVR patients had lower recurrence and longer overall survival than non-SVR patients by Kaplan-Meier analysis. Multivariate analysis revealed that APRI ≥0.7 was the independent risk factor for HCC recurrence; and AFP ≥20 ng/ml post IFN therapy, as well as HCC recurrence were the independent risk factors of mortality. CONCLUSION Liver cirrhosis plays a key role in HCC occurrence after antiviral therapies. SVR patients may have lower HCC recurrence and longer survival rates than non-SVR patients. Only APRI was associated with HCC recurrence; and post-IFN AFP and HCC recurrence were predictive of subsequent mortality independently.
Collapse
Affiliation(s)
- Fai-Meng Sou
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Cheng-Kun Wu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kuo-Chin Chang
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Sheng-Nan Lu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jing-Houng Wang
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chao-Hung Hung
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chien-Hung Chen
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kwong-Ming Kee
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Hao Yen
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ming-Tsung Lin
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ming-Chao Tsai
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tsung-Hui Hu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
61
|
Tu WL, You LR, Tsou AP, Chen CM. Pten Haplodeficiency Accelerates Liver Tumor Growth in miR-122a–Null Mice via Expansion of Periportal Hepatocyte-Like Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2688-2702. [DOI: 10.1016/j.ajpath.2018.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 07/05/2018] [Accepted: 07/26/2018] [Indexed: 01/07/2023]
|
62
|
Liu L, Long H, Wu Y, Li H, Dong L, Zhong JL, Liu Z, Yang X, Dai X, Shi L, Ren M, Lin Z. HRD1-mediated PTEN degradation promotes cell proliferation and hepatocellular carcinoma progression. Cell Signal 2018; 50:90-99. [DOI: 10.1016/j.cellsig.2018.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/01/2018] [Accepted: 06/20/2018] [Indexed: 10/28/2022]
|
63
|
Singh AK, Kumar R, Pandey AK. Hepatocellular Carcinoma: Causes, Mechanism of Progression and Biomarkers. Curr Chem Genom Transl Med 2018; 12:9-26. [PMID: 30069430 PMCID: PMC6047212 DOI: 10.2174/2213988501812010009] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/15/2018] [Accepted: 05/20/2018] [Indexed: 01/18/2023] Open
Abstract
Hepatocellular Carcinoma (HCC) is one of the most common malignant tumours in the world. It is a heterogeneous group of a tumour that vary in risk factor and genetic and epigenetic alteration event. Mortality due to HCC in last fifteen years has increased. Multiple factors including viruses, chemicals, and inborn and acquired metabolic diseases are responsible for its development. HCC is closely associated with hepatitis B virus, and at least in some regions of the world with hepatitis C virus. Liver injury caused by viral factor affects many cellular processes such as cell signalling, apoptosis, transcription, DNA repair which in turn induce important effects on cell survival, growth, transformation and maintenance. Molecular mechanisms of hepatocellular carcinogenesis may vary depending on different factors and this is probably why a large set of mechanisms have been associated with these tumours. Various biomarkers including α-fetoprotein, des-γ-carboxyprothrombin, glypican-3, golgi protein-73, squamous cell carcinoma antigen, circulating miRNAs and altered DNA methylation pattern have shown diagnostic significance. This review article covers up key molecular pathway alterations, biomarkers for diagnosis of HCC, anti-HCC drugs and relevance of key molecule/pathway/receptor as a drug target.
Collapse
Affiliation(s)
| | | | - Abhay K. Pandey
- Department of Biochemistry, University of Allahabad, Allahabad 211002, India
| |
Collapse
|
64
|
Chen J, Liang J, Liu S, Song S, Guo W, Shen F. Differential regulation of AKT1 contributes to survival and proliferation in hepatocellular carcinoma cells by mediating Notch1 expression. Oncol Lett 2018; 15:6857-6864. [PMID: 29725418 PMCID: PMC5920202 DOI: 10.3892/ol.2018.8193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/10/2017] [Indexed: 11/06/2022] Open
Abstract
The RAC serine/threonine-protein kinase (AKT) family of serine/threonine protein kinases, particularly the AKT1 isoform, has been identified abnormally expressed in hepatocellular carcinoma (HCC) cells, and is highly associated with cell behavior, including proliferation, survival, metabolism, and tumorigenesis. However, the specific mechanism by which AKT1 elicits these effects requires further study. The purpose of the present study was to reveal the effects of AKT1 on the survival and proliferation of HCC cells, and to investigate the mechanisms involved. Western blotting and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) to evaluate the expression levels of AKT1 in HCC SMMC-7721 cell line. Molecular mechanisms and the influences of different regulation the expression of AKT1 on HCC cell growth, proliferation were determined by western blotting, MTT and colony formation assays, cell cycle and apoptosis were investigated by flow cytometry. The activation of AKT1 suppressed the expression of phosphatase and tensin homolog and increased the activation of Notch1. The inhibition of AKT1 effectively suppressed the expression of Notch1. Furthermore, the data of the present study indicated that B-cell lymphoma 2 and cyclin D1 is involved in the regulation of AKT1 expression.
Collapse
Affiliation(s)
- Jing Chen
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jun Liang
- Department of Oncology, International Cancer Hospital of Beijing University, Beijing 100000, P.R. China
| | - Shihai Liu
- Central Laboratory, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Shanai Song
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Wenxuan Guo
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Fangzhen Shen
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
65
|
Tseng PL, Wu WH, Hu TH, Chen CW, Cheng HC, Li CF, Tsai WH, Tsai HJ, Hsieh MC, Chuang JH, Chang WT. Decreased succinate dehydrogenase B in human hepatocellular carcinoma accelerates tumor malignancy by inducing the Warburg effect. Sci Rep 2018; 8:3081. [PMID: 29449614 PMCID: PMC5814459 DOI: 10.1038/s41598-018-21361-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/02/2018] [Indexed: 12/17/2022] Open
Abstract
Changes in TCA cycle enzymes or respiratory activity are possible mechanisms of aerobic glycolysis that contributes to tumor progression. To clarify whether the decrease of succinate dehydrogenase B (SDHB) alters energy metabolism, induces the Warburg effect and results in tumor malignancy, SDHB expression was examined and modulated in hepatocellular carcinoma (HCC) tissues and cells, respectively. SDHB level was often decreased in malignant HCC cells and tissues. Furthermore, the reduced SDHB expression was associated with advanced tumor stage and poor survival rate. Moreover, silencing of SDHB altered energy metabolism switched from aerobic respiration to glycolysis, resulted in the Warburg effect, and enhanced cell proliferation and motility. In contrast, the SDHB overexpression deregulated bioenergetic metabolism and decreased cell growth and migration. In mouse xenograft models, subcutaneous implantation and tail vein injection with SDHB knockdown cells resulted in a larger tumor volume and accelerated cancer metastasis, respectively. A mutation or decrease in SDHB induced the switch from aerobic respiration to glycolysis. This metabolic alteration was associated with tumor cell dedifferentiation, proliferation, motility and overall patient survival in HCC.
Collapse
Affiliation(s)
- Po-Lin Tseng
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, 302, Taiwan
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, 833, Taiwan
| | - Wei-Hsuan Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Tsung-Hui Hu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, 833, Taiwan
| | - Chih-Wei Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Surgery, Chi Mei Foundation Medical Center, Tainan, 710, Taiwan
- Department of Occupational Safety and Health/Institute of Industrial Safety and Disaster Prevention, College of Sustainable Environment, Chia Nan University of Pharmacy and Science, Tainan, 717, Taiwan
| | - Hung-Chi Cheng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Chien-Feng Li
- Department of Pathology, Chi Mei Foundation Medical Center, Tainan, 710, Taiwan
| | - Wen-Hui Tsai
- Department of Pediatrics, Chi Mei Foundation Medical Center, Tainan, 710, Taiwan
| | - Hui-Ju Tsai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Meng-Che Hsieh
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Jiin-Haur Chuang
- Department of Pediatric Surgery, Kaohsiung Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, 833, Taiwan
| | - Wen-Tsan Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan.
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
66
|
Ao L, Song X, Li X, Tong M, Guo Y, Li J, Li H, Cai H, Li M, Guan Q, Yan H, Guo Z. An individualized prognostic signature and multi‑omics distinction for early stage hepatocellular carcinoma patients with surgical resection. Oncotarget 2018; 7:24097-110. [PMID: 27006471 PMCID: PMC5029687 DOI: 10.18632/oncotarget.8212] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 03/02/2016] [Indexed: 12/31/2022] Open
Abstract
Previously reported prognostic signatures for predicting the prognoses of postsurgical hepatocellular carcinoma (HCC) patients are commonly based on predefined risk scores, which are hardly applicable to samples measured by different laboratories. To solve this problem, using gene expression profiles of 170 stage I/II HCC samples, we identified a prognostic signature consisting of 20 gene pairs whose within-sample relative expression orderings (REOs) could robustly predict the disease-free survival and overall survival of HCC patients. This REOs-based prognostic signature was validated in two independent datasets. Functional enrichment analysis showed that the patients with high-risk of recurrence were characterized by the activations of pathways related to cell proliferation and tumor microenvironment, whereas the low-risk patients were characterized by the activations of various metabolism pathways. We further investigated the distinct epigenomic and genomic characteristics of the two prognostic groups using The Cancer Genome Atlas samples with multi-omics data. Epigenetic analysis showed that the transcriptional differences between the two prognostic groups were significantly concordant with DNA methylation alternations. The signaling network analysis identified several key genes (e.g. TP53, MYC) with epigenomic or genomic alternations driving poor prognoses of HCC patients. These results help us understand the multi-omics mechanisms determining the outcomes of HCC patients.
Collapse
Affiliation(s)
- Lu Ao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - Xuekun Song
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Xiangyu Li
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - Mengsha Tong
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - You Guo
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - Jing Li
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - Hongdong Li
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - Hao Cai
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - Mengyao Li
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - Qingzhou Guan
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - Haidan Yan
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| | - Zheng Guo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
67
|
Xu Z, Hu J, Cao H, Pilo MG, Cigliano A, Shao Z, Xu M, Ribback S, Dombrowski F, Calvisi DF, Chen X. Loss of Pten synergizes with c-Met to promote hepatocellular carcinoma development via mTORC2 pathway. Exp Mol Med 2018; 50:e417. [PMID: 29303510 PMCID: PMC5992985 DOI: 10.1038/emm.2017.158] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly malignancy with limited treatment options. Activation of the AKT/mTOR cascade is one of the most frequent events along hepatocarcinogenesis. mTOR is a serine/threonine kinase and presents in two distinct complexes: mTORC1 and mTORC2. While mTORC1 has been extensively studied in HCC, the functional contribution of mTORC2 during hepatocarcinogenesis has not been well characterized, especially in vivo. Pten expression is one of the major mechanisms leading to the aberrant activation of the AKT/mTOR signaling. Here, we show that concomitant downregulation of Pten and upregulation of c-Met occurs in a subset of human HCC, mainly characterized by poor prognosis. Using CRISPR-based gene editing in combination with hydrodynamic injection, Pten was deleted in a subset of mouse hepatocytes (sgPten). We found that loss of Pten synergizes with overexpression of c-Met to promote HCC development in mice (sgPten/c-Met). At the molecular level, sgPten/c-Met liver tumor tissues display increased AKT and mTOR signaling. Using Rictor conditional knockout mice, we demonstrate that sgPten/c-Met-driven HCC development strictly depends on an intact mTORC2 complex. Our findings therefore support the critical role of mTORC2 in hepatocarcinogenesis. sgPten/c-Met mouse model represents a novel valuable system that can be used for the development of targeted therapy against this deadly malignancy.
Collapse
Affiliation(s)
- Zhong Xu
- Department of Gastroenterology, Guizhou Provincial People’s Hospital, The Affiliated People’s Hospital of Guizhou Medical University, Guiyang, PR China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
| | - Junjie Hu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, PR China
| | - Hui Cao
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, PR China
| | - Maria G Pilo
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Antonio Cigliano
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Zixuan Shao
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
- Lowell High School, San Francisco, CA, USA
| | - Meng Xu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, PR China
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Frank Dombrowski
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Diego F Calvisi
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, PR China
| |
Collapse
|
68
|
Prenner S, Kulik L. Hepatocellular Carcinoma. ZAKIM AND BOYER'S HEPATOLOGY 2018:668-692.e9. [DOI: 10.1016/b978-0-323-37591-7.00046-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
69
|
Choi JI, Cho EH, Kim SB, Kim R, Kwon J, Park M, Shin HJ, Ryu HS, Park SH, Lee KH. Promoter methylation of cysteine dioxygenase type 1: gene silencing and tumorigenesis in hepatocellular carcinoma. Ann Hepatobiliary Pancreat Surg 2017; 21:181-187. [PMID: 29264579 PMCID: PMC5736736 DOI: 10.14701/ahbps.2017.21.4.181] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/11/2017] [Indexed: 12/31/2022] Open
Abstract
Backgrounds/Aims Cysteine dioxygenase type 1 (CDO1) acts as a tumor suppressor and is silenced by promoter methylation in various malignancies. The relationship between the CDO1 methylation status and hepatocellular carcinoma (HCC) tumorigenesis was evaluated. Methods Using a HCC cell line (SNU423), an in vitro demethylation study was performed to confirm whether promoter methylation causes CDO1 down-regulation. The SNU423 cells transfected with the CDO1 cell function was compared to that of naïve cells. An in vivo study using immunohistochemical staining of HCC specimens that were collected from patients who underwent curative liver resection was also performed. Results CDO1 was activated after demethylation treatment in the HCC specimens. Moreover, tumor cell proliferation, colony-forming, migration, and invasion activities significantly decreased after CDO1 transfection (p<0.05). The percentage of tumors that were larger than 5 cm was higher in patients who had a lower expression of CDO1 (p=0.030). Vascular invasion and histological grade were independent prognostic factors for poor overall and recurrence-free survival. The degree of CDO1 expression was not an independent prognostic factor in this study's population. Conclusions These results suggested that methylation down-regulated CDO1 expression in the HCC cells. CDO1 methylation may be a potentially valuable diagnostic biomarker for HCC.
Collapse
Affiliation(s)
- Jung-Il Choi
- Department of Surgery, Korea Cancer Center Hospital, Seoul, Korea
| | - Eung-Ho Cho
- Department of Surgery, Korea Cancer Center Hospital, Seoul, Korea
| | - Sang Bum Kim
- Department of Surgery, Korea Cancer Center Hospital, Seoul, Korea
| | - Ryounggo Kim
- Department of Surgery, Dongnam Institution of Radiological & Medical Sciences, Busan, Korea
| | - Junhye Kwon
- Department of Translational Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Misun Park
- Department of Translational Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hye-Jin Shin
- Department of Translational Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Sun-Hoo Park
- Department of Pathology, Korea Cancer Center Hospital, Seoul, Korea
| | - Kee-Ho Lee
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medication Sciences, Seoul, Korea
| |
Collapse
|
70
|
Awan FM, Naz A, Obaid A, Ikram A, Ali A, Ahmad J, Naveed AK, Janjua HA. MicroRNA pharmacogenomics based integrated model of miR-17-92 cluster in sorafenib resistant HCC cells reveals a strategy to forestall drug resistance. Sci Rep 2017; 7:11448. [PMID: 28904393 PMCID: PMC5597599 DOI: 10.1038/s41598-017-11943-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/31/2017] [Indexed: 12/27/2022] Open
Abstract
Among solid tumors, hepatocellular carcinoma (HCC) emerges as a prototypical therapy-resistant tumor. Considering the emerging sorafenib resistance crisis in HCC, future studies are urgently required to overcome resistance. Recently noncoding RNAs (ncRNAs) have emerged as significant regulators in signalling pathways involved in cancer drug resistance and pharmacologically targeting these ncRNAs might be a novel stratagem to reverse drug resistance. In the current study, using a hybrid Petri net based computational model, we have investigated the harmonious effect of miR-17-92 cluster inhibitors/mimics and circular RNAs on sorafenib resistant HCC cells in order to explore potential resistance mechanisms and to identify putative targets for sorafenib-resistant HCC cells. An integrated model was developed that incorporates seven miRNAs belonging to miR-17-92 cluster (hsa-miR-17-5p, hsa-miR-17-3p, hsa-miR-19a, hsa-miR-19b, hsa-miR-18a, hsa-miR-20a and hsa-miR-92) and crosstalk of two signaling pathways (EGFR and IL-6) that are differentially regulated by these miRNAs. The mechanistic connection was proposed by the correlation between members belonging to miR-17-92 cluster and corresponding changes in the protein levels of their targets in HCC, specifically those targets that have verified importance in sorafenib resistance. Current findings uncovered potential pathway features, underlining the significance of developing modulators of this cluster to combat drug resistance in HCC.
Collapse
Affiliation(s)
- Faryal Mehwish Awan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Anam Naz
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Ayesha Obaid
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Aqsa Ikram
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Amjad Ali
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Jamil Ahmad
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Abdul Khaliq Naveed
- Islamic International Medical College (IIMC), Riphah International University, Rawalpindi, Pakistan
| | - Hussnain Ahmed Janjua
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan.
| |
Collapse
|
71
|
Khemlina G, Ikeda S, Kurzrock R. The biology of Hepatocellular carcinoma: implications for genomic and immune therapies. Mol Cancer 2017; 16:149. [PMID: 28854942 PMCID: PMC5577674 DOI: 10.1186/s12943-017-0712-x] [Citation(s) in RCA: 310] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/15/2017] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the most common type of primary liver cancer, is a leading cause of cancer-related death worldwide. It is highly refractory to most systemic therapies. Recently, significant progress has been made in uncovering genomic alterations in HCC, including potentially targetable aberrations. The most common molecular anomalies in this malignancy are mutations in the TERT promoter, TP53, CTNNB1, AXIN1, ARID1A, CDKN2A and CCND1 genes. PTEN loss at the protein level is also frequent. Genomic portfolios stratify by risk factors as follows: (i) CTNNB1 with alcoholic cirrhosis; and (ii) TP53 with hepatitis B virus-induced cirrhosis. Activating mutations in CTNNB1 and inactivating mutations in AXIN1 both activate WNT signaling. Alterations in this pathway, as well as in TP53 and the cell cycle machinery, and in the PI3K/Akt/mTor axis (the latter activated in the presence of PTEN loss), as well as aberrant angiogenesis and epigenetic anomalies, appear to be major events in HCC. Many of these abnormalities may be pharmacologically tractable. Immunotherapy with checkpoint inhibitors is also emerging as an important treatment option. Indeed, 82% of patients express PD-L1 (immunohistochemistry) and response rates to anti-PD-1 treatment are about 19%, and include about 5% complete remissions as well as durable benefit in some patients. Biomarker-matched trials are still limited in this disease, and many of the genomic alterations in HCC remain challenging to target. Future studies may require combination regimens that include both immunotherapies and molecularly matched targeted treatments.
Collapse
Affiliation(s)
- Galina Khemlina
- Department of Geriatrics, University of California, UC San Diego, 9500 Gilman Drive, #9111, La Jolla, CA, 92093-9111, USA. .,Kaiser Permanente Southern California, San Diego, USA.
| | - Sadakatsu Ikeda
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, University of California, Moores Cancer Center, San Diego, USA.,Tokyo Medical and Dental University, Tokyo, Japan
| | - Razelle Kurzrock
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, University of California, Moores Cancer Center, San Diego, USA
| |
Collapse
|
72
|
Anti-invasive effects of CXCR4 and FAK inhibitors in non-small cell lung carcinomas with mutually inactivated p53 and PTEN tumor suppressors. Invest New Drugs 2017; 35:718-732. [PMID: 28733702 DOI: 10.1007/s10637-017-0494-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/13/2017] [Indexed: 01/10/2023]
Abstract
Non-small cell lung carcinoma (NSCLC) is the most common type of lung cancer. At the time of diagnosis, a large percentage of NSCLC patients have already developed metastasis, responsible for extremely high mortality rates. CXCR4 receptor and focal adhesion kinase (FAK) are known to regulate such invasive cancer behavior. Their expression is downregulated by p53 and PTEN tumor suppressors which are commonly co-inactivated in NSCLC patients and contribute to metastasis. Therefore, targeting CXCR4 or FAK seems to be a promising strategy in suppressing metastatic spread of p53/PTEN deficient NSCLCs. In this study, we first examined the invasive characteristics of NSCLC cells with suppressed p53 and PTEN activity using wound healing, gelatin degradation and invasion assays. Further, changes in the expression of CXCR4 and FAK were evaluated by RT-qPCR and Western Blot analysis. Finally, we tested the ability of CXCR4 and FAK inhibitors (WZ811 and PF-573228, respectively) to suppress the migratory and invasive potential of p53/PTEN deficient NSCLC cells, in vitro and in vivo using metastatic models of human NSCLC. Our results showed that cells with mutually inactive p53 and PTEN have significantly increased invasive potential associated with hyperactivation of CXCR4 and FAK signaling pathways. Treatments with WZ811 and PF-573228 inhibitors significantly reduced migratory and invasive capacity in vitro and showed a trend of improved survival in vivo. Accordingly, we demonstrated that p53/PTEN deficient NSCLCs have extremely invasive phenotype and provided a rationale for the use of CXCR4 or FAK inhibitors for the suppression of NSCLC dissemination.
Collapse
|
73
|
Khalid A, Hussain T, Manzoor S, Saalim M, Khaliq S. PTEN: A potential prognostic marker in virus-induced hepatocellular carcinoma. Tumour Biol 2017. [DOI: 10.1177/1010428317705754] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Ayesha Khalid
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied BioSciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Tabinda Hussain
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied BioSciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Sobia Manzoor
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied BioSciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Muhammad Saalim
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied BioSciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Saba Khaliq
- University of Health Sciences, Lahore, Pakistan
| |
Collapse
|
74
|
Liu Y, Qi X, Zeng Z, Wang L, Wang J, Zhang T, Xu Q, Shen C, Zhou G, Yang S, Chen X, Lu F. CRISPR/Cas9-mediated p53 and Pten dual mutation accelerates hepatocarcinogenesis in adult hepatitis B virus transgenic mice. Sci Rep 2017; 7:2796. [PMID: 28584302 PMCID: PMC5459841 DOI: 10.1038/s41598-017-03070-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/21/2017] [Indexed: 12/24/2022] Open
Abstract
The p53 mutation and altered Pten expression are two most common genetic events in Hepatitis B virus (HBV) infection related hepatocellular carcinoma (HCC). To confirm the causative role of p53 and Pten somatic mutation in HCC development, we established CRISPR/Cas9-mediated somatic gene disruption via hydrodynamic tail vein injection, allowing for in vivo targeting p53 and Pten simultaneously in adult HBV transgenic mice. Here we demonstrated that the utility of this approach resulted in macroscopic liver tumors as early as 4 months' post injection and most tumors harbored both p53 and Pten loss-of-function alterations. Immunohistochemical (IHC) and histopathology analysis demonstrated that the tumors were positive for Glutamine synthetase (GS), a marker of HCC and accompanied with prominent lipid accumulation. The study here indicated that CRISPR/Cas9-mediated p53 and Pten somatic mutation accelerated hepatocarcinogenesis in adult HBV transgenic mice. This method also provides a fast and convenient system for generating mouse model of HCC with HBV infection characteristics.
Collapse
Affiliation(s)
- Yongzhen Liu
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Xuewei Qi
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Zhenzhen Zeng
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Lu Wang
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Jie Wang
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Ting Zhang
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Qiang Xu
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Congle Shen
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Guangde Zhou
- Department of Pathology and Hepatology, Beijing 302 Hospital, Beijing, 100039, P.R. China
| | - Shaomin Yang
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Xiangmei Chen
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China.
| | - Fengmin Lu
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| |
Collapse
|
75
|
Szparecki G, Ilczuk T, Gabzdyl N, Górnicka B. Comparison of Subtypes of Hepatocellular Adenoma to Hepatocellular Carcinoma and Non-Neoplastic Liver Tissue in Terms of PTEN Expression. Folia Biol (Praha) 2017; 63:202-208. [PMID: 29687774 DOI: 10.14712/fb2017063050202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
PTEN is a tumour suppressor gene whose loss of function has been found to be present in a variety of neoplasms, both benign and malignant. In hepatocellular carcinoma (HCC), loss of PTEN is associated with poorly differentiated cancer, advanced clinical stage and tendency to recur. The extent and meaning of PTEN loss in hepatocellular adenoma (HA), one of the precursor lesions for HCC, has not yet been analysed. The aim of the present study was to evaluate the possible loss of PTEN expression in HA in the wider context of hepatocarcinogenesis. Immunohistochemical analysis of PTEN expression was performed in non-neoplastic liver tissue, HAs and HCCs. It has been found that the loss of PTEN was markedly present in poorly differentiated HCC, whereas well to moderately differentiated HCC showed similar levels of PTEN expression to nonneoplastic liver. HAs presented as a heterogeneous group, with loss of PTEN observed in the inflammatory and HNF1A-mutated subtype and relatively intact PTEN expression in HA with nuclear β-catenin overexpression. This suggests that the loss of PTEN might occur both in HA and HCC, constituting different outcomes of the same molecular lesion in the various contexts of malignant or benign neoplasms.
Collapse
Affiliation(s)
- G Szparecki
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland
| | - T Ilczuk
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland
| | - N Gabzdyl
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland
| | - B Górnicka
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
76
|
Augello G, Puleio R, Emma MR, Cusimano A, Loria GR, McCubrey JA, Montalto G, Cervello M. A PTEN inhibitor displays preclinical activity against hepatocarcinoma cells. Cell Cycle 2016; 15:573-83. [PMID: 26794644 DOI: 10.1080/15384101.2016.1138183] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Phosphatase and tensin homolog (PTEN) gene is considered a tumor suppressor gene. However, PTEN mutations rarely occur in hepatocellular carcinoma (HCC), whereas heterozygosity of PTEN, resulting in reduced PTEN expression, has been observed in 32-44% of HCC patients. In the present study, we investigated the effects of the small molecule PTEN inhibitor VO-OHpic in HCC cells. VO-OHpic inhibited cell viability, cell proliferation and colony formation, and induced senescence-associated β-galactosidase activity in Hep3B (low PTEN expression) and to a lesser extent in PLC/PRF/5 (high PTEN expression) cells, but not in PTEN-negative SNU475 cells. VO-OHpic synergistically inhibited cell viability when combined with PI3K/mTOR and RAF/MEK/ERK pathway inhibitors, but only in Hep3B cells, and significantly inhibited tumor growth in nude mice bearing xenografts of Hep3B cells. Therefore, we demonstrated for the first time that VO-OHpic inhibited cell growth and induced senescence in HCC cells with low PTEN expression, and that the combination of VO-OHpic with PI3K/mTOR and RAF/MEK/ERK inhibitors resulted in a more effective tumor cell kill. Our findings, hence, provide proof-of-principle evidence that pharmacological inhibition of PTEN may represent a promising approach for HCC therapy in a subclass of patients with a low PTEN expression.
Collapse
Affiliation(s)
- Giuseppa Augello
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy
| | - Roberto Puleio
- b Istituto Zooprofilattico Sperimentale della Sicilia "A Mirri,", Area Diagnostica Specialistica, Laboratorio di Istopatologia ed Immunoistochimica , Palermo , Italy
| | - Maria Rita Emma
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy
| | - Antonella Cusimano
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy
| | - Guido R Loria
- b Istituto Zooprofilattico Sperimentale della Sicilia "A Mirri,", Area Diagnostica Specialistica, Laboratorio di Istopatologia ed Immunoistochimica , Palermo , Italy
| | - James A McCubrey
- c Department of Microbiology and Immunology , Brody School of Medicine at East Carolina University , Greenville , NC , USA
| | - Giuseppe Montalto
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy.,d Biomedical Department of Internal Medicine and Specialties (DiBiMIS), University of Palermo , Palermo , Italy
| | - Melchiorre Cervello
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy
| |
Collapse
|
77
|
Mohamed NK, Hamad MA, Hafez MZ, Wooley KL, Elsabahy M. Nanomedicine in management of hepatocellular carcinoma: Challenges and opportunities. Int J Cancer 2016; 140:1475-1484. [DOI: 10.1002/ijc.30517] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/30/2016] [Accepted: 11/08/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Nourhan K. Mohamed
- Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University; Egypt
| | - Mostafa A. Hamad
- Department of Surgery; Faculty of Medicine, Assiut University; Egypt
| | - Mohamed Z.E. Hafez
- Department of Internal Medicine; Faculty of Medicine, Aswan University; Egypt
| | - Karen L. Wooley
- Departments of Chemistry; Chemical Engineering and Materials Science and Engineering, Texas A&M University; College Station TX
- Laboratory for Synthetic-Biologic Interactions; Department of Chemistry, Texas A&M University; College Station TX
| | - Mahmoud Elsabahy
- Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University; Egypt
- Laboratory for Synthetic-Biologic Interactions; Department of Chemistry, Texas A&M University; College Station TX
- Department of Pharmaceutics; Faculty of Pharmacy, Assiut University; Egypt
- Misr University for Science and Technology; 6th of October City Egypt
| |
Collapse
|
78
|
Singh AR, Joshi S, Burgoyne AM, Sicklick JK, Ikeda S, Kono Y, Garlich JR, Morales GA, Durden DL. Single Agent and Synergistic Activity of the "First-in-Class" Dual PI3K/BRD4 Inhibitor SF1126 with Sorafenib in Hepatocellular Carcinoma. Mol Cancer Ther 2016; 15:2553-2562. [PMID: 27496136 DOI: 10.1158/1535-7163.mct-15-0976] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 07/08/2016] [Indexed: 01/30/2023]
Abstract
Deregulated PI3K/AKT/mTOR, Ras/Raf/MAPK, and c-Myc signaling pathways are of prognostic significance in hepatocellular carcinoma (HCC). Sorafenib, the only drug clinically approved for patients with advanced HCC, blocks the Ras/Raf/MAPK pathway but it does not inhibit the PI3K/AKT/mTOR pathway or c-Myc activation. Hence, there is an unmet medical need to identify potent PI3K/BRD4 inhibitors, which can be used either alone or in combination with sorafenib to treat patients with advanced HCC. Herein, we show that SF1126 (pan PI3K/BRD4 inhibitor) as single agent or in combination with sorafenib inhibited proliferation, cell cycle, apoptosis, and multiple key enzymes in PI3K/AKT/mTOR and Ras/Raf/MAPK pathway in Hep3B, HepG2, SK-Hep1, and Huh7 HCC cell lines. We demonstrate that the active moiety of the SF1126 prodrug LY294002 binds to and blocks BRD4 interaction with the acetylated histone-H4 chromatin mark protein and displaced BRD4 coactivator protein from the transcriptional start site of MYC in Huh7 and SK-Hep1 HCC cell lines. Moreover, SF1126 blocked expression levels of c-Myc in HCC cells. Treatment of SF1126 either alone or in combination with sorafenib showed significant antitumor activity in vivo Our results establish that SF1126 is a dual PI3K/BRD4 inhibitor. This agent has completed a phase I clinical trial in humans with good safety profile. Our data support the potential future consideration of a phase II clinical trial of SF1126, a clinically relevant dual "first-in-class" PI3K/BRD4 inhibitor in advanced HCC, and a potential combination with sorafenib. Mol Cancer Ther; 15(11); 2553-62. ©2016 AACR.
Collapse
Affiliation(s)
- Alok R Singh
- Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Shweta Joshi
- Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Adam M Burgoyne
- Division of Hematology-Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Jason K Sicklick
- Division of Surgical Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Sadakatsu Ikeda
- Division of Hematology-Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Yuko Kono
- Division of Hepatology, Department of Medicine, University of California San Diego, La Jolla, California
| | | | | | - Donald L Durden
- Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, California.
- SignalRx Pharmaceuticals, San Diego, California
- Division of Pediatric Hematology-Oncology, UCSD Rady Children's Hospital, University of California San Diego Health System, La Jolla, California
| |
Collapse
|
79
|
Hu B, Yan X, Liu F, Zhu C, Zhou H, Chen Y, Liu J, Gu X, Ni R, Zhang T. Downregulated Expression of PTPN9 Contributes to Human Hepatocellular Carcinoma Growth and Progression. Pathol Oncol Res 2016; 22:555-565. [PMID: 26715439 DOI: 10.1007/s12253-015-0038-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 12/22/2015] [Indexed: 12/16/2022]
Abstract
Human hepatocellular carcinoma (HCC) is one of the most common malignant cancers, whose molecular mechanisms is remains largely. PTPN9 has recently been reported to play a critical role in breast cancer development. However, the role of PTPN9 in human HCC remains elusive. The present study aimed at investigating the potential role of PTPN9 in HCC. Western blot and immunohistochemistry were used to examine the expression of PTPN9 protein in HCC and adjacent non-tumorous tissues in 45 patients. Furthermore, Cell Counting Kit-8, flow cytometry and RNA interference experiments were performed to analyze the role of PTPN9 in the regulation of HCC cell proliferation. We showed that the expression level of PTPN9 was significantly reduced in HCC, compared with adjacent non-tumorous tissues. PTPN9 expression was inversely associated with Tumor size (P = 0.014), serum AFP level (P = 0.004) and Ki-67 expression. Low expression of PTPN9 predicted poor survival in HCC patients. Moreover, PTPN9 interference assay that PTPN9 inhibited cell proliferation in HepG2 cells. Cell apoptosis assay revealed that, silencing of PTPN9 expression significantly reduced cell apoptosis, compared with control ShRNA treatment group. Our results suggested that PTPN9 expression was down-regulated in HCC tumor tissues, and reduced PTPN9 expression was associated with worsened overall survival in HCC patients. Depletion of PTPN9 inhibits the apoptosis and promotes the proliferation of HCC cells.
Collapse
Affiliation(s)
- Baoying Hu
- Basic Medic Research Centre, Medical College, Nantong University, Nantong, 226001, People's Republic of China
| | - Xia Yan
- Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Fang Liu
- Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Changlai Zhu
- Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Huiling Zhou
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226019, Jiangsu, People's Republic of China
| | - Yuyan Chen
- Class 5, Grade 13, Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Jinxia Liu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226019, Jiangsu, People's Republic of China
| | - Xingxing Gu
- Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Runzhou Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Tianyi Zhang
- Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
80
|
LIU LIANG, JI PING, QU NING, PU WEILIN, JIANG DAOWEN, LIU WEIYAN, LI YAQI, SHI RONGLIANG. The impact of high co-expression of Sp1 and HIF1α on prognosis of patients with hepatocellular cancer. Oncol Lett 2016; 12:504-512. [PMID: 27347172 PMCID: PMC4906840 DOI: 10.3892/ol.2016.4634] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/12/2016] [Indexed: 12/12/2022] Open
Abstract
Transcription factor specificity protein 1 (Sp1) and hypoxia-inducible factor 1α (HIF1α) serve vital roles in tumor growth and metastasis. The present study aimed to evaluate the impact of co-expression of Sp1 and HIF1α on the prognosis of patients with hepatocellular cancer (HCC) using The Cancer Genome Atlas (TCGA) database and to validate the association between the expression levels of Sp1/HIF1α in HCC specimens and patient survival using immunohistochemical analysis. A total of 214 eligible patients with HCC from TCGA database were collected for the study. The expression profile of Sp1 and HIF1α were obtained from the TCGA RNAseq database. Clinicopathological characteristics, including age, height, weight, gender, race, ethnicity, family cancer history, serum α-fetoprotein (AFP), surgical procedures and TNM stage were collected. The Cox proportional hazards regression model and Kaplan-Meier curves were used to assess the relative factors. Receiver operating characteristic (ROC) curves for cancer-specific survival (CSS) prediction were plotted to compare the prediction ability of expression of Sp1 and HIF1α and their co-expression. The location and expression of Sp1 and HIF1α in the HCC tissues were detected by immunohistochemistry (IHC) to verify the association between these two genes and CSS. The results demonstrated that the expressions of Sp1 and HIF1α were significantly increased in the succumbed group (P=0.001), compared with the surviving group. The CSS rates were 60.1% at 3 years (1,067 days), 35.8% at 5 years (1,823 days) and 9.5% at 10 years (3,528 days). Multivariate Cox regression analysis demonstrated that only the high expression levels of Sp1 and HIF1α (≥2×103) were independent predictors for cancer mortality, with P=0.001 and P=0.029, respectively. The area under the curve for the ROC was found to be higher using the combination testing for two genes (0.751) in predicting cancer mortality, compared to a single gene (0.632 for Sp1 and 0.717 for HIF1α). Based on the cutoff points for gene expression, patients were divided into 3 groups: G1 (both genes <2×103), G2 (either gene ≥2×103) and G3 (both genes ≥2×103). The risk of cancer mortality increased with high expression of genes, and G3 exhibited a greater risk than G2 when compared with the G1 group (HR=5.420, 95% CI 2.767-10.616, P=0.001; HR=3.270, 95% CI 1.843-5.803, P=0.001). The IHC staining results indicated that patients who died of cancer presented with significantly higher expression levels of these genes compared with those that did not (P=0.001). In summary, high expression levels of Sp1 and HIF1α in HCC tissues were associated with poor prognosis; in particular, the co-expression of these two genes increased the risk of cancer mortality.
Collapse
Affiliation(s)
- LIANG LIU
- Department of Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, P.R. China
| | - PING JI
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, P.R. China
| | - NING QU
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, P.R. China
| | - WEI-LIN PU
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200433, P.R. China
| | - DAO-WEN JIANG
- Department of General Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - WEI-YAN LIU
- Department of General Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - YA-QI LI
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, P.R. China
| | - RONG-LIANG SHI
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, P.R. China
- Department of General Surgery, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| |
Collapse
|
81
|
Lamarca A, Mendiola M, Barriuso J. Hepatocellular carcinoma: Exploring the impact of ethnicity on molecular biology. Crit Rev Oncol Hematol 2016; 105:65-72. [PMID: 27372199 DOI: 10.1016/j.critrevonc.2016.06.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/15/2016] [Accepted: 06/14/2016] [Indexed: 01/17/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer in the world and the third leading cause of cancer-related death. The high rate of diagnosis in non-curable stages and the lack of novel active treatments make it necessary to review all the possible sources of misleading results in this scenario. The incidence of HCC shows clear geographical variation with higher annual incidence in Asia and Africa than in Western countries; we aimed to review the literature to find if there are different trends in the main activated molecular pathways. Hyperactivation of RAS/RAF/MEK/ERK and PI3K/AKT/mTOR signalling and epithelial to mesenchymal transition (EMT) process are more prevalent in the Western population; however, fibroblast growth factor (FGF), transforming growth factor β (TGFβ) and Notch pathways seems to be more relevant in Asian population. Whether these variations just reflect the distinct distribution of known causes of HCC or proper ethnical differences remain to be elucidated. Nevertheless, these clearly different patterns are relevant to regional or worldwide clinical trial design. If this information is neglected by sponsors and researchers the rate of failure in HCC trials will not improve.
Collapse
Affiliation(s)
- Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Marta Mendiola
- Cancer Molecular Pathology and Therapeutic Targets Research Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Jorge Barriuso
- Faculty of Life Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
82
|
Qiu ZX, Zhao S, Li L, Li WM. Loss of Expression of PTEN is Associated with Worse Prognosis in Patients with Cancer. Asian Pac J Cancer Prev 2016; 16:4691-8. [PMID: 26107225 DOI: 10.7314/apjcp.2015.16.11.4691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The tumor suppressor phosphatase and tensin homolog (PTEN) is an important negative regulator of cell-survival signaling. However, available results for the prognostic value of PTEN expression in patients with cancer remain controversial. Therefore, a meta-analysis of published studies investigating this issue was performed. MATERIALS AND METHODS A literature search via PubMed and EMBASE databases was conducted. Statistical analysis was performed by using the STATA 12.0 (STATA Corp., College, TX). Data from eligible studies were extracted and included into the meta-analysis using a random effects model. RESULTS A total of 3,810 patients from 27 studies were included in the meta-analysis, 22 investigating the relationship between PTEN expression and overall survival (OS) using univariate analysis, and nine with multivariate analysis. The pooled hazard ratio (HR) for OS was 1.64 (95% confidence interval (CI): 1.32-2.05) by univariate analysis and 1.56 (95% CI: 1.20-2.03) by multivariate analysis. In addition, eight papers including two disease-free-survival analyses (DFSs), four relapse-free-survival analyses (RFSs), three progression-free-survival analyses (PFSs) and one metastasis-free-survival analysis (MFS) reported the effect of PTEN on survival. The results showed that loss of PTEN expression was significant correlated with poor prognosis, with a combined HR of 1.74 (95% CI: 1.24-2.44). Furthermore, in the stratified analysis by the year of publication, ethnicity, cancer type, method, cut- off value, median follow-up time and neoadjuvant therapy in which the study was conducted, we found that the ethnicity, cancer type, method, median follow-up time and neoadjuvant therapy are associated with prognosis. CONCLUSIONS Our study shows that negative or loss of expression of PTEN is associated with worse prognosis in patients with cancer. However, adequately designed prospective studies need to be performed for confirmation.
Collapse
Affiliation(s)
- Zhi-Xin Qiu
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China E-mail :
| | | | | | | |
Collapse
|
83
|
Ju HL, Han KH, Lee JD, Ro SW. Transgenic mouse models generated by hydrodynamic transfection for genetic studies of liver cancer and preclinical testing of anti-cancer therapy. Int J Cancer 2016; 138:1601-1608. [PMID: 26220477 DOI: 10.1002/ijc.29703] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/15/2015] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal cancers worldwide; however, the genetic mechanisms underlying its pathogenesis are incompletely understood. Genetically engineered mouse (GEM) models of HCC have been developed to elucidate the role of individual cancer-related genes in hepatocarcinogenesis. However, the expensive and time-consuming processes related to generating a GEM model discourage the development of diverse genotype models. Recently, a simple and inexpensive liver-specific transgenic approach was developed, in which a hydrodynamics-based transfection (HT) method was coupled with the Sleeping Beauty transposase system. Various HT models in which different oncogenic pathways are activated and/or tumor-suppressing pathways inactivated have been developed in recent years. The applicability of HT models in liver cancer research is expected to broaden and ultimately elucidate the cooperation between oncogenic signaling pathways and aid in designing molecular therapy to target altered pathways.
Collapse
Affiliation(s)
- Hye-Lim Ju
- Liver Cirrhosis Clinical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Kwang-Hyub Han
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Doo Lee
- Department of Nuclear Medicine, Catholic Kwandong University, Seoul, Korea
| | - Simon Weonsang Ro
- Liver Cirrhosis Clinical Research Center, Yonsei University College of Medicine, Seoul, Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
84
|
Miura K, Ishioka M, Minami S, Horie Y, Ohshima S, Goto T, Ohnishi H. Toll-like Receptor 4 on Macrophage Promotes the Development of Steatohepatitis-related Hepatocellular Carcinoma in Mice. J Biol Chem 2016; 291:11504-17. [PMID: 27022031 DOI: 10.1074/jbc.m115.709048] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Indexed: 01/18/2023] Open
Abstract
The role of Toll-like receptor (TLR) signaling has attracted much attention in the development of hepatic inflammation and hepatocellular carcinoma (HCC). We herein sought to determine the role of TLRs and responsible cells in steatohepatitis-related HCC. We used hepatocyte-specific Pten-deficient (Pten(Δ) (hep)) mice, which exhibit steatohepatitis followed by liver tumor formation, including HCC. We then generated Pten(Δ) (hep)/Tlr4(-/-) and Pten(Δ) (hep)/Tlr2(-/-) double-mutant mice and investigated the role of macrophages using reconstitution of bone marrow (BM)-derived cells, chemical depletion of macrophages, and isolated macrophages. Tlr4 but not Tlr2 deficiency in the Pten(Δ) (hep) mice suppressed tumor growth as well as hepatic inflammation. Gut sterilization by an antibiotic mixture reduced the portal LPS levels as well as tumor growth in the Pten(Δ) (hep) mice. Tumor growth was also decreased by reconstitution of BM-derived cells to Tlr4(-/-) BM cells. In addition, chemical depletion of macrophages significantly reduced tumor size and numbers. Macrophages expressing Ly6C were increased in number, which was associated with inflammation and tumor progression in the Pten(Δ) (hep) mice. Hepatic macrophages isolated from the Pten(Δ) (hep) mice abundantly expressed the Ly6C gene and produced much more IL-6 and TNFα in response to LPS. These proinflammatory cytokines induced the proliferation of HCC cells as well as oval cells, putative cancer progenitor cells. Indeed, putative cancer progenitor cells emerged before the development of macroscopic liver tumors and then increased in number under sustained inflammation. TLR4 on macrophages contributes to the development of steatohepatitis-related HCC in mice.
Collapse
Affiliation(s)
- Kouichi Miura
- From the Department of Gastroenterology and Hepato-Biliary-Pancreatology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Mitsuaki Ishioka
- From the Department of Gastroenterology and Hepato-Biliary-Pancreatology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Shinichiro Minami
- From the Department of Gastroenterology and Hepato-Biliary-Pancreatology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Yasuo Horie
- From the Department of Gastroenterology and Hepato-Biliary-Pancreatology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Shigetoshi Ohshima
- From the Department of Gastroenterology and Hepato-Biliary-Pancreatology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Takashi Goto
- From the Department of Gastroenterology and Hepato-Biliary-Pancreatology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Hirohide Ohnishi
- From the Department of Gastroenterology and Hepato-Biliary-Pancreatology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| |
Collapse
|
85
|
Zhang T, Zhang X, Shi W, Xu J, Fan H, Zhang S, Ni R. The DNA damage repair protein Ku70 regulates tumor cell and hepatic carcinogenesis by interacting with FOXO4. Pathol Res Pract 2016; 212:153-61. [DOI: 10.1016/j.prp.2015.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 10/29/2015] [Accepted: 12/17/2015] [Indexed: 12/31/2022]
|
86
|
Teng YC, Shen ZQ, Kao CH, Tsai TF. Hepatocellular carcinoma mouse models: Hepatitis B virus-associated hepatocarcinogenesis and haploinsufficient tumor suppressor genes. World J Gastroenterol 2016; 22:300-325. [PMID: 26755878 PMCID: PMC4698494 DOI: 10.3748/wjg.v22.i1.300] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/14/2015] [Accepted: 11/24/2015] [Indexed: 02/06/2023] Open
Abstract
The multifactorial and multistage pathogenesis of hepatocellular carcinoma (HCC) has fascinated a wide spectrum of scientists for decades. While a number of major risk factors have been identified, their mechanistic roles in hepatocarcinogenesis still need to be elucidated. Many tumor suppressor genes (TSGs) have been identified as being involved in HCC. These TSGs can be classified into two groups depending on the situation with respect to allelic mutation/loss in the tumors: the recessive TSGs with two required mutated alleles and the haploinsufficient TSGs with one required mutated allele. Hepatitis B virus (HBV) is one of the most important risk factors associated with HCC. Although mice cannot be infected with HBV due to the narrow host range of HBV and the lack of a proper receptor, one advantage of mouse models for HBV/HCC research is the numerous and powerful genetic tools that help investigate the phenotypic effects of viral proteins and allow the dissection of the dose-dependent action of TSGs. Here, we mainly focus on the application of mouse models in relation to HBV-associated HCC and on TSGs that act either in a recessive or in a haploinsufficient manner. Discoveries obtained using mouse models will have a great impact on HCC translational medicine.
Collapse
|
87
|
Jasphin SSR, Desai D, Pandit S, Gonsalves NM, Nayak PB, Iype A. Immunohistochemical expression of phosphatase and tensin homolog in histologic gradings of oral squamous cell carcinoma. Contemp Clin Dent 2016; 7:524-528. [PMID: 27994422 PMCID: PMC5141669 DOI: 10.4103/0976-237x.194111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Context: Phosphatase and tensin homolog (PTEN) is a tumor suppressor gene located on chromosome 10q23. PTEN has its major function in the regulation of cell adhesion, cell cycle arrest, migration, apoptosis programming, and differentiation. This genomic region suffers loss of heterozygosity in many human cancers. Aims: The aim of this study was to compare the immunohistochemical expression of PTEN in normal oral mucosa and oral squamous cell carcinoma (OSCC) and to correlate the PTEN expression in gradings of OSCC. Materials and Methods: Thirty cases of paraffin tissue sections of previously diagnosed OSCC were taken. Of thirty cases, ten were well differentiated, ten were moderately differentiated, and ten were poorly differentiated. As a control, ten paraffin sections of oral normal mucosa tissue specimens were taken from patients undergoing extractions. The sections were stained for immunohistochemical expression of PTEN. The cells stained by PTEN antibody were counted, and an immunohistochemical score was obtained. Statistical Analysis Used: Statistical analysis was done using Mann–Whitney's test and Kruskal–Wallis test. Results: Statistical analysis revealed that there was a significant difference between normal mucosa and OSCC in immunohistochemistry staining. However, there was no significant difference in PTEN expression among gradings of OSCC. Conclusions: The study concluded that there was a decrease in PTEN expression in OSCC than normal mucosa. It also concluded that PTEN is a tumor suppressor gene which has a wide role in oral carcinogenesis.
Collapse
Affiliation(s)
- Shiny S R Jasphin
- National Post - Doctoral Fellow (SERB), Melaka Manipal Medical College, Manipal University, Manipal, Karnataka, India
| | - Dinkar Desai
- Department of Oral Pathology, A.J Institute of Dental Science, Mangaluru, Karnataka, India
| | - Siddharth Pandit
- Department of Oral Pathology, A.J Institute of Dental Science, Mangaluru, Karnataka, India
| | - Nithin M Gonsalves
- Department of Oral Pathology, A.J Institute of Dental Science, Mangaluru, Karnataka, India
| | - Preethi B Nayak
- Department of Oral Pathology, A.J Institute of Dental Science, Mangaluru, Karnataka, India
| | - Amal Iype
- Department of Oral Pathology, Malabar Dental College, Vattamkulam, Kerala, India
| |
Collapse
|
88
|
Intact PTEN Expression by Immunohistochemistry is Associated With Decreased Survival in Advanced Stage Ovarian/Primary Peritoneal High-grade Serous Carcinoma. Int J Gynecol Pathol 2015; 34:497-506. [DOI: 10.1097/pgp.0000000000000205] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
89
|
Shu G, Yue L, Zhao W, Xu C, Yang J, Wang S, Yang X. Isoliensinine, a Bioactive Alkaloid Derived from Embryos of Nelumbo nucifera, Induces Hepatocellular Carcinoma Cell Apoptosis through Suppression of NF-κB Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:8793-8803. [PMID: 26389520 DOI: 10.1021/acs.jafc.5b02993] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Isoliensinine (isolie) is an alkaloid produced by the edible plant Nelumbo nucifera. Here, we unveiled that isolie was able to provoke HepG2, Huh-7, and H22 hepatocellular carcinoma (HCC) cell apoptosis. Isolie decreased NF-κB activity and constitutive phosphorylation of NF-κB p65 subunit at Ser536 in HCC cells. Overexpression of p65 Ser536 phosphorylation mimics abrogated isolie-mediated HCC cell apoptosis. Furthermore, intraperitoneal injection of isolie inhibited the growth of Huh-7 xenografts in nude mice. Additionally, isolie given by both intraperitoneal injection and gavage diminished the proliferation of transplanted H22 cells in Kunming mice. Reduced tumor growth in vivo was associated with inhibited p65 phosphorylation at Ser536 and declined NF-κB activity in tumor tissues. Finally, we revealed that isolie was bioavailable in the blood of mice and exhibited no detectable toxic effects on tumor-bearing mice. Our data provided strong evidence for the anti-HCC effect of isolie.
Collapse
Affiliation(s)
- Guangwen Shu
- College of Pharmacy, South-Central University for Nationalities , Wuhan, P. R. China
| | - Ling Yue
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Military Command , Wuhan, P. R. China
| | - Wenhao Zhao
- College of Pharmacy, South-Central University for Nationalities , Wuhan, P. R. China
| | - Chan Xu
- College of Pharmacy, South-Central University for Nationalities , Wuhan, P. R. China
| | - Jing Yang
- College of Pharmacy, South-Central University for Nationalities , Wuhan, P. R. China
| | - Shaobing Wang
- College of Pharmacy, South-Central University for Nationalities , Wuhan, P. R. China
| | - Xinzhou Yang
- College of Pharmacy, South-Central University for Nationalities , Wuhan, P. R. China
| |
Collapse
|
90
|
Affiliation(s)
- Mahmoud Elsabahy
- Department of Chemistry, Department of Chemical Engineering, Department of Materials Science & Engineering, Laboratory for Synthetic-Biologic Interactions, Texas A&M University, P.O. Box 30012, 3255 TAMU, College Station, Texas 77842-3012, United States
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, 71515 Assiut, Egypt, and Misr University for Science and Technology, 6 of October City, Egypt
| | - Gyu Seong Heo
- Department of Chemistry, Department of Chemical Engineering, Department of Materials Science & Engineering, Laboratory for Synthetic-Biologic Interactions, Texas A&M University, P.O. Box 30012, 3255 TAMU, College Station, Texas 77842-3012, United States
| | - Soon-Mi Lim
- Department of Chemistry, Department of Chemical Engineering, Department of Materials Science & Engineering, Laboratory for Synthetic-Biologic Interactions, Texas A&M University, P.O. Box 30012, 3255 TAMU, College Station, Texas 77842-3012, United States
| | - Guorong Sun
- Department of Chemistry, Department of Chemical Engineering, Department of Materials Science & Engineering, Laboratory for Synthetic-Biologic Interactions, Texas A&M University, P.O. Box 30012, 3255 TAMU, College Station, Texas 77842-3012, United States
| | - Karen L. Wooley
- Department of Chemistry, Department of Chemical Engineering, Department of Materials Science & Engineering, Laboratory for Synthetic-Biologic Interactions, Texas A&M University, P.O. Box 30012, 3255 TAMU, College Station, Texas 77842-3012, United States
| |
Collapse
|
91
|
The RNA-binding protein Sam68 regulates tumor cell viability and hepatic carcinogenesis by inhibiting the transcriptional activity of FOXOs. J Mol Histol 2015; 46:485-97. [PMID: 26438629 DOI: 10.1007/s10735-015-9639-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 09/30/2015] [Indexed: 12/31/2022]
Abstract
Src associated in mitosis (Sam68; 68 kDa) is a KH domain RNA-binding protein that belongs to the signal transduction and activation of RNA family, and has been implicated in the oncogenesis and progression of several human cancers. Our study aimed to investigated the clinicopathologic significance of Sam68 expression and its role in cell proliferation and the underlying molecular mechanism in hepatocellular carcinoma (HCC). We demonstrated that Sam68 expression was significantly increased in HCC and high expression of Sam68 was significantly associated with Edmondson grade, tumor size, tumor nodule number, HBsAg status and Ki-67 expression. The Kaplan-Meier survival curves showed that increased expression of Sam68 was correlated with poor prognosis in HCC patients and served as an independent prognostic marker of overall survival in a multivariable analysis. In addition, through serum starvation and refeeding assay, we demonstrated that Sam68 was lowly expressed in serum-starved HCC cells, and was progressively increased after serum-additioning. Furthermore, siRNA knockdown of endogenous Sam68 inhibited cell proliferation and tumourigenicity of HCC cells in vitro, through blocking the G1 to S phase transition. Moreover, we reported that the anti-proliferative effect of silencing Sam68 was accompanied with up-regulated expression of cyclin-dependent kinase inhibitors, p21(Cip1) and p27(Kip1), enhanced transactivation of FOXO factors (FOXO4), and dysreuglation of Akt/GSK-3β signaling. Taken together, these findings provide a rational framework for the progression of HCC and thereby indicated that Sam68 might be a novel and useful prognostic marker and a potential target for human HCC treatment.
Collapse
|
92
|
Du Y, Zhang YW, Pu R, Han X, Hu JP, Zhang HW, Wang HY, Cao GW. Phosphatase and tensin homologue genetic polymorphisms and their interactions with viral mutations on the risk of hepatocellular carcinoma. Chin Med J (Engl) 2015; 128:1005-13. [PMID: 25881591 PMCID: PMC4832937 DOI: 10.4103/0366-6999.155057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: Chronic hepatitis B virus (HBV) infection is the major cause of hepatocellular carcinoma (HCC). Some HBV mutants and dysregulation of phosphatase and tensin homolog (PTEN) may promote the development of HCC synergistically. We aimed to test the effects of PTEN genetic polymorphisms and their interactions with important HBV mutations on the development of HCC in HBV-infected subjects. Methods: Quantitative polymerase chain reaction was applied to genotype PTEN polymorphisms (rs1234220, rs2299939, rs1234213) in 1012 healthy controls, 302 natural clearance subjects, and 2011 chronic HBV-infected subjects including 1021 HCC patients. HBV mutations were determined by sequencing. The associations of PTEN polymorphisms and their interactions with HBV mutations with HCC risk were assessed using multivariate logistic regression analysis. Results: Rs1234220 C allele was significantly associated with HCC risk compared to healthy controls (adjusted odds ratio [AOR] = 1.35, 95% confidence interval [CI] = 1.07–1.69) and HCC-free HBV-infected subjects (AOR = 1.27, 95% CI = 1.01–1.57). rs1234220 C allele was significantly associated with increased frequencies of HCC-risk A1652G, C1673T, and C1730G mutations in genotype B HBV-infected subjects. Rs2299939 GT genotype was inversely associated with HCC risk in HBV-infected patients (AOR = 0.75, 95% CI = 0.62–0.92). The interaction of rs2299939 variant genotypes (GT+TT) with A3054T mutation significantly increased HCC risk (AOR = 2.41, 95% CI = 1.08–5.35); whereas its interaction with C3116T mutation significantly reduced HCC risk (AOR = 0.34, 95% CI = 0.18–0.66). These significant effects were only evident in males after stratification. Conclusions: PTEN polymorphisms and their interactions with HBV mutations may contribute to hepatocarcinogenesis in males. The host-virus interactions are important in identifying HBV-infected subjects who are more likely to develop HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Guang-Wen Cao
- Department of Epidemiology, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
93
|
Mu X, Español-Suñer R, Mederacke I, Affò S, Manco R, Sempoux C, Lemaigre FP, Adili A, Yuan D, Weber A, Unger K, Heikenwälder M, Leclercq IA, Schwabe RF. Hepatocellular carcinoma originates from hepatocytes and not from the progenitor/biliary compartment. J Clin Invest 2015; 125:3891-903. [PMID: 26348897 DOI: 10.1172/jci77995] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 07/23/2015] [Indexed: 12/19/2022] Open
Abstract
In many organs, including the intestine and skin, cancers originate from cells of the stem or progenitor compartment. Despite its nomenclature, the cellular origin of hepatocellular carcinoma (HCC) remains elusive. In contrast to most organs, the liver lacks a defined stem cell population for organ maintenance. Previous studies suggest that both hepatocytes and facultative progenitor cells within the biliary compartment are capable of generating HCC. As HCCs with a progenitor signature carry a worse prognosis, understanding the origin of HCC is of clinical relevance. Here, we used complementary fate-tracing approaches to label the progenitor/biliary compartment and hepatocytes in murine hepatocarcinogenesis. In genotoxic and genetic models, HCCs arose exclusively from hepatocytes but never from the progenitor/biliary compartment. Cytokeratin 19-, A6- and α-fetoprotein-positive cells within tumors were hepatocyte derived. In summary, hepatocytes represent the cell of origin for HCC in mice, and a progenitor signature does not reflect progenitor origin, but dedifferentiation of hepatocyte-derived tumor cells.
Collapse
|
94
|
Borgas DL, Gao JS, Tong M, de la Monte SM. Potential Role of Phosphorylation as a Regulator of Aspartyl-(asparaginyl)-β-hydroxylase: Relevance to Infiltrative Spread of Human Hepatocellular Carcinoma. Liver Cancer 2015; 4:139-53. [PMID: 26675015 PMCID: PMC4608650 DOI: 10.1159/000367731] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Abundant expression of aspartyl-(asparaginyl)-β-hydroxylase (AAH) correlates with infiltrative growth of hepatocellular carcinoma (HCC). Herein, we examine the role of phosphorylation in relation to AAH's protein expression, hydroxylase activity, promotion of cell motility, and activation of Notch signaling in human Huh7 hepatoma cells. Predicted glycogen synthase kinase-3β (GSK-3β), protein kinase A (PKA), protein kinase C (PKC), and casein kinase 2 (CK2) phosphorylation sites encoded by human AAH cDNA were ablated by S/T→A site-directed mutagenesis using N-Myc-tagged constructs in which gene expression was controlled by a cytomegalovirus promoter. Functional consequences were assessed in transiently transfected Huh7 cells. Cells transfected with wildtype AAH had significantly increased AAH expression, catalytic activity, HES-1 expression, and directional motility relative to controls. Single phosphorylation site mutations in the C-terminus largely abrogated these effects and further inhibited catalytic activity relative to that in cells transfected with empty vector, whereas the effects of single point mutations within the N-terminus were more varied. In contrast, AAH cDNAs carrying multiple phosphorylation site mutations exhibited wildtype levels of AAH catalytic activity suggesting that the effects of AAH phosphorylation are complex and non-uniform. AAH expression and function can be modulated by direct phosphorylation of the protein. These findings suggest additional strategies for inhibiting infiltrative growth of HCC.
Collapse
Affiliation(s)
| | | | | | - Suzanne M. de la Monte
- *Suzanne M. de la Monte, MD, MPH, Liver Research Center, Divisions of Gastroenterology and, Neuropathology, and Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and the Warren Alpert Medical, School of Brown University, Pierre Galletti Research Building, Rhode Island Hospital, 55 Claverick Street, Room 419, Providence, RI 02903 (USA), Tel. +1 401 444 7364, E-Mail
| |
Collapse
|
95
|
Cheng D, Zhang L, Yang G, Zhao L, Peng F, Tian Y, Xiao X, Chung RT, Gong G. Hepatitis C virus NS5A drives a PTEN-PI3K/Akt feedback loop to support cell survival. Liver Int 2015; 35:1682-91. [PMID: 25388655 DOI: 10.1111/liv.12733] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/06/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Decreased levels of phosphatase and tensin homologue (PTEN) are associated with hepatocellular carcinoma (HCC) pathogenesis and poor prognosis in hepatitis C virus (HCV)-infected HCC patients. The molecular processes governing the reduction in PTEN and outcome of PTEN dysfunction in hepatocytes are poorly understood. METHODS The levels of proteins and mRNA were assessed by real time PCR and immunoblot. PTEN promoter activity was measured by reporter assay. Signalling pathways were perturbed using siRNAs or pharmacological inhibitors. RESULTS Here, we report that HCV down-regulates PTEN expression at the transcriptional level by decreasing its promoter activity, mRNA transcription, and protein levels. We further identify NS5A protein as a key determinant of PTEN reduction among HCV proteins. NS5A-mediated down-regulation of PTEN occurs through a cooperation of reactive oxygen species (ROS)-dependent Nuclear Factor- kappa B (NF-κB) and ROS-independent phosphoinositol-3-kinase (PI3K) pathways. Moreover, NS5A protects cells against apoptosis. In addition, we found that down-regulation of PTEN relieves its inhibitory effect on PI3K-Akt pathway and triggers cumulative activation of Akt. This PTEN-PI3K/Akt feedback network mediates the suppression of cell apoptosis caused by NS5A. CONCLUSIONS These data demonstrate that HCV NS5A down-regulates PTEN expression through a cooperation of ROS-dependent and -independent pathways that subsequently drives a PTEN-PI3K/Akt feedback loop to support cell survival. Our findings provide new insights suggesting that NS5A contributes to HCV-related hepatocarcinogenesis.
Collapse
Affiliation(s)
- Du Cheng
- Department of Gastroenterology, Renmin Hospital, Wuhan University, Wuhan, China.,Liver Center, Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Liver Diseases Center, Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Leiliang Zhang
- Liver Center, Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guangbo Yang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Zhao
- Liver Center, Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Feng Peng
- Liver Diseases Center, Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Tian
- Liver Diseases Center, Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xinqiang Xiao
- Liver Diseases Center, Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Raymond T Chung
- Liver Center, Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guozhong Gong
- Liver Diseases Center, Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
96
|
Grech G, Zhan X, Yoo BC, Bubnov R, Hagan S, Danesi R, Vittadini G, Desiderio DM. EPMA position paper in cancer: current overview and future perspectives. EPMA J 2015; 6:9. [PMID: 25908947 PMCID: PMC4407842 DOI: 10.1186/s13167-015-0030-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 02/26/2015] [Indexed: 12/31/2022]
Abstract
At present, a radical shift in cancer treatment is occurring in terms of predictive, preventive, and personalized medicine (PPPM). Individual patients will participate in more aspects of their healthcare. During the development of PPPM, many rapid, specific, and sensitive new methods for earlier detection of cancer will result in more efficient management of the patient and hence a better quality of life. Coordination of the various activities among different healthcare professionals in primary, secondary, and tertiary care requires well-defined competencies, implementation of training and educational programs, sharing of data, and harmonized guidelines. In this position paper, the current knowledge to understand cancer predisposition and risk factors, the cellular biology of cancer, predictive markers and treatment outcome, the improvement in technologies in screening and diagnosis, and provision of better drug development solutions are discussed in the context of a better implementation of personalized medicine. Recognition of the major risk factors for cancer initiation is the key for preventive strategies (EPMA J. 4(1):6, 2013). Of interest, cancer predisposing syndromes in particular the monogenic subtypes that lead to cancer progression are well defined and one should focus on implementation strategies to identify individuals at risk to allow preventive measures and early screening/diagnosis. Implementation of such measures is disturbed by improper use of the data, with breach of data protection as one of the risks to be heavily controlled. Population screening requires in depth cost-benefit analysis to justify healthcare costs, and the parameters screened should provide information that allow an actionable and deliverable solution, for better healthcare provision.
Collapse
Affiliation(s)
- Godfrey Grech
- />Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Xianquan Zhan
- />Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
| | - Byong Chul Yoo
- />Colorectal Cancer Branch, Division of Translational and Clinical Research I, Research Institute, National Cancer Center, Gyeonggi, 410-769 Republic of Korea
| | - Rostyslav Bubnov
- />Clinical Hospital ‘Pheophania’ of State Management of Affairs Department, Kyiv, Ukraine
- />Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Suzanne Hagan
- />Dept of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Romano Danesi
- />Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giorgio Vittadini
- />Bracco Imaging, Centro Ricerche Bracco, San Donato Milanese, Italy
| | - Dominic M Desiderio
- />Department of Neurology, University of Tennessee Center for Health Science, Memphis, USA
| |
Collapse
|
97
|
Zhou X, Zhu H, Lu J. PTEN and hTERT gene expression and the correlation with human hepatocellular carcinoma. Pathol Res Pract 2015; 211:316-9. [DOI: 10.1016/j.prp.2014.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 11/11/2014] [Indexed: 11/29/2022]
|
98
|
Huynh H, Hao HX, Chan SL, Chen D, Ong R, Soo KC, Pochanard P, Yang D, Ruddy D, Liu M, Derti A, Balak MN, Palmer MR, Wang Y, Lee BH, Sellami D, Zhu AX, Schlegel R, Huang A. Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus. Mol Cancer Ther 2015; 14:1224-35. [PMID: 25724664 DOI: 10.1158/1535-7163.mct-14-0768] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/17/2015] [Indexed: 11/16/2022]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer deaths worldwide and hyperactivation of mTOR signaling plays a pivotal role in HCC tumorigenesis. Tuberous sclerosis complex (TSC), a heterodimer of TSC1 and TSC2, functions as a negative regulator of mTOR signaling. In the current study, we discovered that TSC2 loss-of-function is common in HCC. TSC2 loss was found in 4 of 8 HCC cell lines and 8 of 28 (28.6%) patient-derived HCC xenografts. TSC2 mutations and deletions are likely to be the underlying cause of TSC2 loss in HCC cell lines, xenografts, and primary tumors for most cases. We further demonstrated that TSC2-null HCC cell lines and xenografts had elevated mTOR signaling and, more importantly, were significantly more sensitive to RAD001/everolimus, an mTORC1 inhibitor. These preclinical findings led to the analysis of TSC2 status in HCC samples collected in the EVOLVE-1 clinical trial of everolimus using an optimized immunohistochemistry assay and identified 15 of 139 (10.8%) samples with low to undetectable levels of TSC2. Although the sample size is too small for formal statistical analysis, TSC2-null/low tumor patients who received everolimus tended to have longer overall survival than those who received placebo. Finally, we performed an epidemiology survey of more than 239 Asian HCC tumors and found the frequency of TSC2 loss to be approximately 20% in Asian HBV(+) HCC. Taken together, our data strongly argue that TSC2 loss is a predictive biomarker for the response to everolimus in HCC patients.
Collapse
Affiliation(s)
- Hung Huynh
- Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore.
| | - Huai-Xiang Hao
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Stephen L Chan
- State Key Laboratory in Oncology in South China, Department of Clinical Oncology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, PR China
| | - David Chen
- Oncology Global Development, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Richard Ong
- Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore
| | - Khee Chee Soo
- Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore
| | - Panisa Pochanard
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - David Yang
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - David Ruddy
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Manway Liu
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Adnan Derti
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Marissa N Balak
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Michael R Palmer
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Yan Wang
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Benjamin H Lee
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Dalila Sellami
- Oncology Global Development, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Andrew X Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Robert Schlegel
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Alan Huang
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts.
| |
Collapse
|
99
|
Gene expression in hepatocellular carcinoma: pilot study of potential transarterial chemoembolization response biomarkers. J Vasc Interv Radiol 2015; 26:723-32. [PMID: 25724086 DOI: 10.1016/j.jvir.2014.12.610] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/16/2014] [Accepted: 12/20/2014] [Indexed: 02/06/2023] Open
Abstract
PURPOSE To perform a feasibility study to explore the relationship between hepatocellular carcinoma genetics and transarterial chemoembolization treatment response to identify potential biomarkers associated with enhanced treatment efficacy. MATERIALS AND METHODS In this single-institution study, pretreatment hepatocellular carcinoma biopsy specimens for tumors in 19 patients (14 men, five women; mean age, 59 y) treated with chemoembolization between 2007 and 2013 were analyzed for a panel of 60 chemotherapy-sensitivity, hypoxia, mitosis, and inflammatory genes with the QuantiGene Plex 2.0 mRNA detection assay. Demographic, disease, and procedure data and tumor response outcomes were collected. Quantitative mRNA levels were compared based on radiologic response between tumors exhibiting complete response (CR) versus partial response (PR). RESULTS The study sample included 19 biopsy specimens from tumors (mean size, 3.0 cm; grade 1, n = 6; grade 2, n = 9; grade 3, n = 4) in patients treated with a mean of two conventional chemoembolization sessions. Thirteen and six tumors exhibited CR and PR, respectively, at a mean of 116 days after treatment. Tumors with CR showed a significant increase in (P < .05) or trend toward (P < .1) greater (range, 1.49-3.50 fold) pretreatment chemotherapy-sensitivity and mitosis (ATF4, BAX, CCNE1, KIF11, NFX1, PPP3CA, SNX1, TOP2A, and TOP2B) gene mRNA expression compared with tumors with PR, in addition to lower CXCL10 levels (0.48-fold), and had significantly (P < .05) higher (1.65-fold) baseline VEGFA levels. CONCLUSIONS Genetic signatures may allow prechemoembolization stratification of tumor response probability, and gene analysis may therefore offer an opportunity to personalize locoregional therapy by enhancing treatment modality allocation. Further corroboration of identified markers and exploration of their respective predictive capacity thresholds is necessary.
Collapse
|
100
|
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide, and the third leading cause of cancer mortality. The great majority of patients are not eligible for curative therapies, and therapeutic approaches for advanced disease show only limited efficacy. Difficulties to treat HCC are due to the heterogenous genetic alterations of HCC, profound alterations in the hepatic microenvironment, and incomplete understanding of HCC biology. Mouse models of HCC will be helpful to improve our understanding of HCC biology, the contributions of the specific pathways and genetic alterations to carcinogenesis. In addition, mouse models of HCC may contribute to elucidate the role of the tumor microenvironment, and serve as models for preclinical studies. As no single mouse model is appropriate to study all of the above, we discuss key features and limitations of commonly used models. Furthermore, we provide detailed protocols for select models, in which HCC is induced genetically, chemically or by transplantation of tumor cells.
Collapse
Affiliation(s)
- Jorge Matias Caviglia
- Department of Medicine, Columbia University, Russ Berrie Pavilion, Room 415, 1150 St. Nicholas Ave, New York, NY, 10032, USA
| | | |
Collapse
|