151
|
Yamaji Y, Yasunaga H, Hirata Y, Yamada A, Yoshida S, Horiguchi H, Fushimi K, Koike K. Association Between Colorectal Cancer and Atherosclerotic Diseases: A Study Using a National Inpatient Database in Japan. Dig Dis Sci 2016; 61:1677-85. [PMID: 26831490 DOI: 10.1007/s10620-016-4041-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 01/16/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Both colorectal neoplasms and atherosclerosis can be induced by common visceral fat accumulation. However, the association between these diseases at the advanced stage, colorectal cancer and cardiovascular/cerebrovascular diseases, has not been elucidated. AIMS This study aimed to investigate the association between colorectal cancer and vascular diseases in relation to obesity and metabolic disorders, using a nationwide database of hospitalized patients in Japan. We used non-cardiac gastric cancer patients as a reference, because they were considered to be neutral for obesity or metabolic disorders. METHODS We identified 54,591 patients with colorectal cancer and 19,565 patients with gastric cancer from the Diagnosis Procedure Combination database. The data collected included: sex; age; body mass index (BMI); smoking status; comorbidity; and medication data, comprising hypertension, diabetes, hyperlipidemia, coronary artery disease, and stroke. We compared these data in a cross-sectional setting by multivariate analyses. RESULTS Multivariate logistic regression analyses showed that female sex, increased BMI, hypertension [odds ratio (OR) 1.11; 95 % confidence interval (CI) 1.07-1.15; P < 0.0001], and diabetes (OR 1.17; 95 % CI 1.12-1.23; P < 0.0001) were more associated with colorectal cancer than with gastric cancer. Smoking, aspirin use (OR 0.85; 95 % CI 0.79-0.92; P < 0.0001), and coronary artery disease (OR 0.90; 95 % CI 0.86-0.95; P = 0.0001) were inversely associated with colorectal cancer. CONCLUSIONS Obesity and metabolic disorders were more associated with colorectal cancer than with non-cardiac gastric cancer, while coronary artery disease (CAD) was inversely associated. Some mechanisms involving separate populations of colorectal cancer and CAD under visceral fat accumulation might be suggested.
Collapse
Affiliation(s)
- Yutaka Yamaji
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yoshihiro Hirata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Atsuo Yamada
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shuntaro Yoshida
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiromasa Horiguchi
- Department of Clinical Data Management and Research, Clinical Research Center, National Hospital Organization Headquarters, 2-5-21 Higashigaoka, Meguro-ku, Tokyo, 152-8621, Japan
| | - Kiyohide Fushimi
- Department of Health Policy and Informatics, Tokyo Medical and Dental University Graduate School of Medicine, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
152
|
Gao X, Liu W, Zhang X, Tang L, Wang L, Yan L, Yang H, Li T, Yang L, Ma N, Liu D. Genetic polymorphism of HLA-DQ confers susceptibility to hepatitis B virus-related hepatocellular carcinoma: a case-control study in Han population in China. Tumour Biol 2016; 37:12103-12111. [PMID: 27209411 DOI: 10.1007/s13277-016-5077-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 05/15/2016] [Indexed: 12/27/2022] Open
Abstract
Several recent genome-wide studies showed that the genetic polymorphisms in the HLA-DQ region (rs9275572 and rs2856718) were related to chronic hepatitis B virus (HBV) infection and chronic hepatitis C virus (HCV)-associated hepatocellular carcinoma (HCC). We analyzed the two single-nucleotide polymorphisms for major HBV outcomes in Han Chinese. A total of 1291 samples were involved and peripheral blood samples were collected in this study. Matrix-assisted laser desorption/ionization time of flight mass spectrometry were used to genotype the single-nucleotide polymorphisms in the HLA-DQ region. Our study indicated the clear relationship between the HLA-DQ rs9275572 and HBV-related HCC after control for the effects of sex, drinking, and smoking. Health subjects with the HLA-DQ rs9275572 GA genotype would have a 0.641 (95 % CI 0.416, 0.985; P = 0.043) times lower odds of having HCC, and those with the AA genotype would have a 0.256 (95 % CI 0.106, 0.618; P = 0.002) times lower odds of having HCC, whereas the values of the other covariates were fixed. Whereas there was no significant difference found for the HLA-DQ rs2856718 AG and GG genotype. Our study suggested that HLA-DQ loci (rs9275572) were associated with HBV-related HCC as a protective factor in Han Chinese.
Collapse
Affiliation(s)
- Xia Gao
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Wenxuan Liu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Xiaolin Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Longmei Tang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Liqin Wang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Lina Yan
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Haitao Yang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Tao Li
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Lei Yang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Ning Ma
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | - Dianwu Liu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China.
| |
Collapse
|
153
|
Sim S, Hibberd ML. Genomic approaches for understanding dengue: insights from the virus, vector, and host. Genome Biol 2016; 17:38. [PMID: 26931545 PMCID: PMC4774013 DOI: 10.1186/s13059-016-0907-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The incidence and geographic range of dengue have increased dramatically in recent decades. Climate change, rapid urbanization and increased global travel have facilitated the spread of both efficient mosquito vectors and the four dengue virus serotypes between population centers. At the same time, significant advances in genomics approaches have provided insights into host–pathogen interactions, immunogenetics, and viral evolution in both humans and mosquitoes. Here, we review these advances and the innovative treatment and control strategies that they are inspiring.
Collapse
Affiliation(s)
- Shuzhen Sim
- Infectious Diseases, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Martin L Hibberd
- Infectious Diseases, Genome Institute of Singapore, Singapore, 138672, Singapore. .,Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK.
| |
Collapse
|
154
|
Tatsumi T, Takehara T. Impact of natural killer cells on chronic hepatitis C and hepatocellular carcinoma. Hepatol Res 2016; 46:416-22. [PMID: 26574168 DOI: 10.1111/hepr.12619] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/01/2015] [Indexed: 01/20/2023]
Abstract
Natural killer (NK) cells are involved in the pathogenesis of hepatitis C viral (HCV) infection and hepatocellular carcinoma (HCC). Recent immunological progresses have revealed the molecular mechanisms of activation or inhibition of NK cells. In patients infected with HCV, the percentages of NK cells are decreased and the NK receptor expression and function of NK cells including cytotoxicity and cytokine production are altered. These alterations in NK cells are associated with persistent infection with HCV, liver injury, liver fibrosis and liver carcinogenesis. In HCV treatment, NK cells play a role in the eradication of HCV in both interferon (IFN)-based therapy and IFN-free therapy using direct-acting antivirals (DAA). In HCC patients, the exhaustion of NK cells that represents lower cytotoxicity and impaired cytokine production may contribute to the progression of HCC. Several immunotherapies targeting NK cells have been reported. NK cell transfer and NK-activating gene therapy have been demonstrated to be effective in mouse liver cancer models and several clinical trials are ongoing. Recently, the role of major histocompatibility complex class I-related chain A (MICA), a human ligand of NKG2D, has attracted attention in the development of HCC. The expression of MICA could be controlled by anti-HCC drugs including sorafenib. A new chemo-immunotherapy may be expected in the treatment of HCC. In this review, we summarize the impact of NK cells on chronic hepatitis C and HCC.
Collapse
Affiliation(s)
- Tomohide Tatsumi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
155
|
Niu J, Lin Y, Guo Z, Niu M, Su C. The Epidemiological Investigation on the Risk Factors of Hepatocellular Carcinoma: A Case-Control Study in Southeast China. Medicine (Baltimore) 2016; 95:e2758. [PMID: 26871825 PMCID: PMC4753921 DOI: 10.1097/md.0000000000002758] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Incidence of hepatocellular carcinoma (HCC) ranked the fifth in male and ninth in the female counterparts, and 50% of incidence HCC cases were occurred in China with high hepatitis B virus (HBV) prevalence. HCC has seriously compromised the health status of general population in China. A case-control study of 314 HCC cases and 346 controls was conducted in Xiamen, which is an epidemic area in China for both hepatitis B infection and HCC. Face-to-face interview was conducted to gather information on demographic characteristics as well as exposure of environmental factors. Commercial enzyme-linked immunosorbent assay kits were used to determine the status of serological markers of HBV infection. Odds ratios and 95% confidence intervals were estimated by using unconditional logistic regression. Multivariate unconditional logistic regression analysis was applied to evaluate the potential interactions of variables or confounders.As expected, HBV and alcohol intake still are the major risk factors of HCC. Liver disease history and passive smoking are also associated with elevated HCC risk. Indoor air pollution and pesticide exposure have newly identified as risk factors of HCC. Fruit and tea intake can significantly lower the HCC risk.The application of HBV vaccine and reduction on alcohol intake should be further promoted in high-risk population. Fruit and tea can be served as chemoprevention in daily life due to their high accessibility.
Collapse
Affiliation(s)
- Jianjun Niu
- From the Zhongshan Hospital, Xiamen University (JN); Xiamen Center for Disease Control and Prevention (YL, ZG, MN, CS); and School of Public Health, Xiamen University, Xiamen, Fujian Province, People's Republic of China (CS)
| | | | | | | | | |
Collapse
|
156
|
Burza MA, Motta BM, Mancina RM, Pingitore P, Pirazzi C, Lepore SM, Spagnuolo R, Doldo P, Russo C, Lazzaro V, Fischer J, Berg T, Aghemo A, Cheroni C, De Francesco R, Fargion S, Colombo M, Datz C, Stickel F, Valenti L, Romeo S. DEPDC5 variants increase fibrosis progression in Europeans with chronic hepatitis C virus infection. Hepatology 2016; 63:418-27. [PMID: 26517016 PMCID: PMC4737289 DOI: 10.1002/hep.28322] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 10/25/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Chronic hepatitis C virus (HCV) infection may progress to cirrhosis and hepatocellular carcinoma (HCC). Recently, two genetic variants, DEPDC5 rs1012068 and MICA rs2596542, were associated with the onset of HCC in Asian subjects with chronic HCV infection. The aim of the present study was to analyze whether DEPDC5 and MICA genetic variants were associated with liver disease progression in European subjects with chronic HCV infection. In a Northern Italian discovery cohort (n = 477), neither DEPDC5 rs1012068 nor MICA rs2596542 were associated with HCC (n = 150). However, DEPDC5 rs1012068 was independently associated with cirrhosis (n = 300; P = 0.049). The association of rs1012068 with moderate to severe fibrosis was confirmed in an independent cross-sectional German cohort (n = 415; P = 0.006). Furthermore, DEPDC5 rs1012068 predicted faster fibrosis progression in a prospective cohort (n = 247; P = 0.027). Next, we examined the distribution of nonsynonymous DEPDC5 variants in the overall cross-sectional cohort (n = 912). The presence of at least one variant increased the risk of moderate/severe fibrosis by 54% (P = 0.040). To understand the molecular mechanism underlying the genetic association of DEPDC5 variants with fibrosis progression, we performed in vitro studies on immortalized hepatic stellate cells (LX-2). In these cells, down-regulation of DEPDC5 resulted in increased expression of β-catenin and production of its target matrix metallopeptidase 2 (MMP2), a secreted enzyme involved in fibrosis progression. CONCLUSION DEPDC5 variants increase fibrosis progression in European subjects with chronic HCV infection. Our findings suggest that DEPDC5 down-regulation may contribute to HCV-related fibrosis by increasing MMP2 synthesis through the β-catenin pathway.
Collapse
Affiliation(s)
- Maria Antonella Burza
- Department of Molecular and Clinical Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Benedetta Maria Motta
- Department of Molecular and Clinical Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | | | - Piero Pingitore
- Department of Molecular and Clinical Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Carlo Pirazzi
- Department of Molecular and Clinical Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Saverio Massimo Lepore
- Clinical Nutrition Unit, Department of Medical and Surgical SciencesMagna Graecia UniversityCatanzaroItaly
| | - Rocco Spagnuolo
- Department of Experimental and Clinical MedicineMagna Graecia UniversityCatanzaroItaly
| | - Patrizia Doldo
- Department of Experimental and Clinical MedicineMagna Graecia UniversityCatanzaroItaly
| | - Cristina Russo
- Clinical Nutrition Unit, Department of Medical and Surgical SciencesMagna Graecia UniversityCatanzaroItaly
| | - Veronica Lazzaro
- Clinical Nutrition Unit, Department of Medical and Surgical SciencesMagna Graecia UniversityCatanzaroItaly
| | - Janett Fischer
- Department of Gastroenterology and Rheumatology, Section of HepatologyUniversity HospitalLeipzigGermany
| | - Thomas Berg
- Department of Gastroenterology and Rheumatology, Section of HepatologyUniversity HospitalLeipzigGermany
| | - Alessio Aghemo
- Department of GastroenterologyFondazione IRCCS Ca' Granda Ospedale Policlinico MilanoMilanItaly
| | - Cristina Cheroni
- Virology ProgramINGM‐Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Raffaele De Francesco
- Virology ProgramINGM‐Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Silvia Fargion
- Università degli Studi di Milano, Fondazione IRCCS Ca'Granda Ospedale Policlinico Milano, Department of Pathophysiology and TransplantationMilanItaly
| | - Massimo Colombo
- Department of GastroenterologyFondazione IRCCS Ca' Granda Ospedale Policlinico MilanoMilanItaly,Università degli Studi di Milano, Fondazione IRCCS Ca'Granda Ospedale Policlinico Milano, Department of Pathophysiology and TransplantationMilanItaly
| | - Christian Datz
- Department of Internal Medicine, Hospital OberndorfTeaching Hospital of the Paracelsus Private University of SalzburgOberndorfAustria
| | - Felix Stickel
- Department of Gastroenterology and HepatologyUniversity Hospital of Zürich, Rämistrasse 100, CH‐8091ZürichSwitzerland
| | - Luca Valenti
- Università degli Studi di Milano, Fondazione IRCCS Ca'Granda Ospedale Policlinico Milano, Department of Pathophysiology and TransplantationMilanItaly
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden,Clinical Nutrition Unit, Department of Medical and Surgical SciencesMagna Graecia UniversityCatanzaroItaly,Department of CardiologySahlgrenska University HospitalGothenburgSweden
| |
Collapse
|
157
|
Matsuura K, Tanaka Y. Host genetic variants influencing the clinical course of hepatitis C virus infection. J Med Virol 2016; 88:185-195. [PMID: 26211651 DOI: 10.1002/jmv.24334] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2015] [Indexed: 12/16/2022]
Abstract
The clinical course of hepatitis C virus (HCV) infection greatly differs in individuals. Various viral, host, and environmental factors influence the natural history of HCV infection. Recent genome-wide association studies identified several host genetic factors influencing treatment efficacy or clinical course in HCV infection. A landmark discovery was that IFNL3-IFNL4 variants are strongly associated with responses to interferon-based treatment. Genetic variants in IFNL3-IFNL4 as well as those in HLA class II loci influence the spontaneous clearance of acute HCV infection. Interestingly, these genetic variants also affect the activity of hepatitis, or disease progression in chronic hepatitis C. In addition, polymorphisms in apoptosis-related genes such as RNF7, TULP1, and MERTK are associated with fibrosis progression, and DEPDC5 and MICA variants are associated with HCV-related hepatocellular carcinoma. Understanding the genetic factors associated with the clinical course of HCV infection is essential for personalized treatment and surveillance of disease progression and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Kentaro Matsuura
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
- Department of Transfusion Medicine, Clinical CenterInfectious Disease and Immunogenetics Section, National Institutes of Health, Bethesda, Maryland
| | - Yasuhito Tanaka
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| |
Collapse
|
158
|
Hou SH, Hu J, Zhang Y, Li QL, Guo JJ. Effects of interaction between genetic variants in human leukocyte antigen DQ and granulysin genes in Chinese Han subjects infected with hepatitis B virus. Microbiol Immunol 2016; 59:209-18. [PMID: 25644528 DOI: 10.1111/1348-0421.12239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/25/2015] [Accepted: 01/28/2015] [Indexed: 12/13/2022]
Abstract
Single nucleotide polymorphisms (SNPs) of HLA-DQ and granulysin (GNLY) are reportedly associated with HBV infection. The aim of this study was to investigate the effects of interactions between SNPs in HLA-DQ and GNLY on the outcome of hepatitis B virus (HBV) infection in Chinese Han subjects. HLA-DQ (rs9275572) and GNLY (rs1866139 and rs11127) were genotyped in 310 subjects with HBV-related chronic liver disease, 295 in whom spontaneous clearance of HBV had occurred and 316 who had not been exposed to HBV. HLA-DQ rs9275572 was significantly correlated with HBV clearance (dominant genetic model: OR, 1.84; 95% CI, 1.30-2.61; adjusted P = 0.001). There was no statistical association of GNLY rs1866139 and rs11127with HBV infection outcomes. However, significant sex-specific associations with HBV susceptibility were observed in men who carried rs1866139 CG or rs11127 TC and in women who carried rs1866139 GG or rs11127 CC. The findings were the same in the validation cohort, which was composed of 829 subjects. Based on a multifactor dimensionality reduction test with permutation correction, a three-way interaction between SNPs in HLA-DQ and GNLY was identified in terms of HBV clearance. In conclusion, additional evidence for an association of HLA-DQ and GNLY SNPs with HBV infection outcomes has been identified and a SNP-SNP interaction between HLA-DQ and GNLY on HBV clearance observed.
Collapse
Affiliation(s)
- Si-hui Hou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Chongqing Medical University
| | | | | | | | | |
Collapse
|
159
|
Huang JF, Yeh ML, Yu ML, Dai CY, Huang CF, Huang CI, Tsai PC, Lin PC, Chen YL, Chang WT, Hou NJ, Lin ZY, Chen SC, Chuang WL. The tertiary prevention of hepatocellular carcinoma in chronic hepatitis C patients. J Gastroenterol Hepatol 2015; 30:1768-1774. [PMID: 26094738 DOI: 10.1111/jgh.13012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIM Pegylated interferon-alpha plus ribavirin combination (PegIFN/RBV) therapy possesses positive effect in the secondary prevention of hepatocellular carcinoma (HCC) in chronic hepatitis C (CHC) patients. The current study aimed to assess its efficacy in the tertiary prevention and to validate the performance of the MHC class I polypeptide-related chain A (MICA) level in the prediction of hepatocellular carcinoma (HCC) recurrence. METHODS A multi-center study enrolling 105 consecutive HCC patients post curative therapies were prospectively recruited. The primary outcome measurement was recurrence of HCC. RESULTS The mean observational period was 52.7 months (range = 3.9-121.5 months). Fifty-six (53.3%) patients achieved sustained virological response (SVR). After completion of treatment, 43 (41.0%) patients developed HCC recurrence, and 24 (55.8%) of them had their recurrence within 6 months after completion of therapy. Thirty-three (76.7%) of the patients with HCC recurrence were of de novo pattern. Those responders tended to have a lower cumulative incidence of recurrence than those non-responders (43.2 vs 84.8/100 person-month, log-rank P = 0.13). Those non-responders with a high MICA level (>100 pg/mL) carried the lowest cancer-free survival than those non-responders with a low MICA level and those responders (P = 0.002). Cox regression hazard analysis showed high baseline MICA level (Odds ratio [OR] = 4.8, 95% confidence interval [CI] = 1.1-20.8, P = 0.04) and a low platelet count (<100 000/mm(3) ) (OR = 5.4, 95% CI = 1.1-27.0, P = 0.04) predicted HCC recurrence. CONCLUSIONS PegIFN/RBV therapy carried a limited effect in the tertiary prevention of HCC. A high MICA level predicted HCC recurrence, particularly among those non-responders.
Collapse
Affiliation(s)
- Jee-Fu Huang
- Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lun Yeh
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yen Dai
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Feng Huang
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Ching-I Huang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Pei-Chien Tsai
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Pei-Chen Lin
- Center for Teaching and Research, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Li Chen
- Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wen-Tsan Chang
- Division of HBP Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Nai-Jen Hou
- Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zu-Yau Lin
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shinn-Cherng Chen
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Long Chuang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
160
|
Isernhagen A, Schilling D, Monecke S, Shah P, Elsner L, Walter L, Multhoff G, Dressel R. The MICA-129Met/Val dimorphism affects plasma membrane expression and shedding of the NKG2D ligand MICA. Immunogenetics 2015; 68:109-23. [PMID: 26585323 PMCID: PMC4728179 DOI: 10.1007/s00251-015-0884-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022]
Abstract
The MHC class I chain-related molecule A (MICA) is a ligand for the activating natural killer (NK) cell receptor NKG2D. A polymorphism causing a valine to methionine exchange at position 129 affects binding to NKG2D, cytotoxicity, interferon-γ release by NK cells and activation of CD8+ T cells. It is known that tumors can escape NKG2D-mediated immune surveillance by proteolytic shedding of MICA. Therefore, we investigated whether this polymorphism affects plasma membrane expression (pmMICA) and shedding of MICA. Expression of pmMICA was higher in a panel of tumor (n = 16, P = 0.0699) and melanoma cell lines (n = 13, P = 0.0429) carrying the MICA-129Val/Val genotype. MICA-129Val homozygous melanoma cell lines released more soluble MICA (sMICA) by shedding (P = 0.0015). MICA-129Met or MICA-129Val isoforms differing only in this amino acid were expressed in the MICA-negative melanoma cell line Malme, and clones with similar pmMICA expression intensity were selected. The MICA-129Met clones released more sMICA (P = 0.0006), and a higher proportion of the MICA-129Met than the MICA-129Val variant was retained in intracellular compartments (P = 0.0199). The MICA-129Met clones also expressed more MICA messenger RNA (P = 0.0047). The latter phenotype was also observed in mouse L cells transfected with the MICA expression constructs (P = 0.0212). In conclusion, the MICA-129Met/Val dimorphism affects the expression density of MICA on the plasma membrane. More of the MICA-129Met variants were retained intracellularly. If expressed at the cell surface, the MICA-129Met isoform was more susceptible to shedding. Both processes appear to limit the cell surface expression of MICA-129Met variants that have a high binding avidity to NKG2D.
Collapse
Affiliation(s)
- Antje Isernhagen
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Humboldtallee 34, 37073, Göttingen, Germany
| | - Daniela Schilling
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Institute of Innovative Radiotherapy (iRT), Radiation Immune Biology, Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Munich, Germany
| | - Sebastian Monecke
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Humboldtallee 34, 37073, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Göttingen, Germany
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Humboldtallee 34, 37073, Göttingen, Germany
| | - Leslie Elsner
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Humboldtallee 34, 37073, Göttingen, Germany
| | - Lutz Walter
- Primate Genetics Laboratory, German Primate Center, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Göttingen, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Institute of Innovative Radiotherapy (iRT), Radiation Immune Biology, Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Munich, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Humboldtallee 34, 37073, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
161
|
Jiang DK, Ma XP, Wu X, Peng L, Yin J, Dan Y, Huang HX, Ding DL, Zhang LY, Shi Z, Zhang P, Yu H, Sun J, Lilly Zheng S, Deng G, Xu J, Liu Y, Guo J, Cao G, Yu L. Genetic variations in STAT4,C2,HLA-DRB1 and HLA-DQ associated with risk of hepatitis B virus-related liver cirrhosis. Sci Rep 2015; 5:16278. [PMID: 26538132 PMCID: PMC4633722 DOI: 10.1038/srep16278] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 10/05/2015] [Indexed: 12/16/2022] Open
Abstract
Recent genome-wide associated studies (GWASs) have revealed several common loci associated with the risk of hepatitis B virus (HBV)- or hepatitis C virus (HCV)-related hepatocellular carcinoma (HCC). We selected 15 single nucleotide polymorphisms (SNPs) identified through GWASs on HBV- or HCV-related HCC, and genotyped them in two independent Chinese cohorts of chronic HBV carriers, including 712 LC cases and 2601 controls. The association of each SNP with the risk of HBV-related LC was assessed by meta-analysis of the two cohorts. Of the 12 SNPs reported in HBV-related HCC GWASs, five SNPs (rs7574865 in STAT4, rs9267673 near C2, rs2647073 and rs3997872 near HLA-DRB1 and rs9275319 near HLA-DQ), were found to be significantly associated with the risk of HBV-related LC (rs7574865: P = 1.79 × 10(-2), OR = 1.17, 95% CI = 1.03-1.34; rs9267673: P = 4.91 × 10(-4), OR = 1.37, 95% CI = 1.15-1.63; rs2647073: P = 3.53 × 10(-5), OR = 1.63, 95% CI = 1.29-2.06; rs3997872: P = 4.22 × 10(-4), OR = 1.86, 95% CI = 1.32-2.62; rs9275319: P = 1.30 × 10(-2), OR = 1.32, 95% CI = 1.06-1.64). However, among the three SNPs associated with the risk of HCV-related HCC in previous GWASs, none of them showed significant association with the risk of HBV-related LC. Our results suggested that genetic variants associated with HBV-related hepatocarcinogenesis may already play an important role in the progression from CHB to LC.
Collapse
Affiliation(s)
- De-Ke Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, Fudan University, Shanghai, China
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Program for Personalized Cancer Care, NorthShore University HealthSystem, the University of Chicago, IL, USA
| | - Xiao-Pin Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaopan Wu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Lijun Peng
- Division of Digestive Diseases, Zhongshan Hospital, Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianhua Yin
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Yunjie Dan
- Department of Infectious Diseases, Southwest Hospital, Institute of Immunology, Third Military Medical University, and Chongqing Key Laboratory of Infectious Diseases, Chongqing, China
| | - Hui-Xing Huang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Dong-Lin Ding
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Lu-Yao Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhuqing Shi
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, Fudan University, Shanghai, China
| | - Pengyin Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, Fudan University, Shanghai, China
| | - Hongjie Yu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, Fudan University, Shanghai, China
| | - Jielin Sun
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - S. Lilly Zheng
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Program for Personalized Cancer Care, NorthShore University HealthSystem, the University of Chicago, IL, USA
| | - Guohong Deng
- Department of Infectious Diseases, Southwest Hospital, Institute of Immunology, Third Military Medical University, and Chongqing Key Laboratory of Infectious Diseases, Chongqing, China
| | - Jianfeng Xu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- Center for Genetic Translational Medicine and Prevention, Fudan University, Shanghai, China
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Program for Personalized Cancer Care, NorthShore University HealthSystem, the University of Chicago, IL, USA
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jinsheng Guo
- Division of Digestive Diseases, Zhongshan Hospital, Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guangwen Cao
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Institute of Biomedical Science, Fudan University, Shanghai, China
| |
Collapse
|
162
|
Kishikawa T, Otsuka M, Ohno M, Yoshikawa T, Sato M, Koike K. Development of a screening method to identify regulators of MICA shedding. Biochem Biophys Res Commun 2015; 465:764-768. [PMID: 26299929 DOI: 10.1016/j.bbrc.2015.08.081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 08/19/2015] [Indexed: 12/27/2022]
Abstract
Immune cells, such as natural killer (NK) cells, recognize virally infected and transformed cells, and eliminate them through the interaction between NKG2D receptors on NK cells and NKG2D ligands on pathogenic cells. Shedding of NKG2D ligands is thought to be a type of counter-mechanism employed by pathogenic cells to evade from NKG2D-mediated immune surveillance. MHC class I polypeptide-related sequence A (MICA) is a prototypical NKG2D ligand. We previously reported that, in soluble form, MICA expression levels are significantly associated with hepatitis virus-induced hepatocellular carcinoma. Here, we report a MICA shedding assay that utilizes membrane-bound MICA tagged at its N-terminus with a nano-luciferase reporter to quantify MICA shedding into culture media. Using this method, we screened a compound library and identified putative regulators of MICA shedding that have the potential to enhance the immune reaction by simultaneously increasing cell surface MICA levels and decreasing soluble MICA levels. This shedding assay may be useful for screening regulators of cell surface molecule shedding.
Collapse
Affiliation(s)
- Takahiro Kishikawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Japan Science and Technology Agency, PRESTO, Kawaguchi, Saitama 332-0012, Japan.
| | - Motoko Ohno
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Takeshi Yoshikawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Masaya Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
163
|
Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM. Genetic Landscape and Biomarkers of Hepatocellular Carcinoma. Gastroenterology 2015; 149:1226-1239.e4. [PMID: 26099527 DOI: 10.1053/j.gastro.2015.05.061] [Citation(s) in RCA: 939] [Impact Index Per Article: 93.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/08/2015] [Accepted: 05/20/2015] [Indexed: 12/02/2022]
Abstract
Hepatocellular carcinoma (HCC) has emerged as a major cause of cancer-related death. Its mortality has increased in Western populations, with a minority of patients diagnosed at early stages, when curative treatments are feasible. Only the multikinase inhibitor sorafenib is available for the management of advanced cases. During the last 10 years, there has been a clear delineation of the landscape of genetic alterations in HCC, including high-level DNA amplifications in chromosome 6p21 (VEGFA) and 11q13 (FGF19/CNND1), as well as homozygous deletions in chromosome 9 (CDKN2A). The most frequent mutations affect TERT promoter (60%), associated with an increased telomerase expression. TERT promoter can also be affected by copy number variations and hepatitis B DNA insertions, and it can be found mutated in preneoplastic lesions. TP53 and CTNNB1 are the next most prevalent mutations, affecting 25%-30% of HCC patients, that, in addition to low-frequency mutated genes (eg, AXIN1, ARID2, ARID1A, TSC1/TSC2, RPS6KA3, KEAP1, MLL2), help define some of the core deregulated pathways in HCC. Conceptually, some of these changes behave as prototypic oncogenic addiction loops, being ideal biomarkers for specific therapeutic approaches. Data from genomic profiling enabled a proposal of HCC in 2 major molecular clusters (proliferation and nonproliferation), with differential enrichment in prognostic signatures, pathway activation and tumor phenotype. Translation of these discoveries into specific therapeutic decisions is an unmet medical need in this field.
Collapse
Affiliation(s)
- Jessica Zucman-Rossi
- Inserm, UMR-1162, Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue Contre le Cancer, Institut Universitaire d'Hematologie, Paris, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; Université Paris 13, Sorbonne Paris Cité, Unité de Formation et de Recherche Santé, Médecine, Biologie Humaine, Bobigny, France; Université Paris Diderot, Paris.
| | - Augusto Villanueva
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jean-Charles Nault
- Inserm, UMR-1162, Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue Contre le Cancer, Institut Universitaire d'Hematologie, Paris, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; Service d'hépatologie, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bondy, France
| | - Josep M Llovet
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Liver Cancer Translational Research Laboratory, Barcelona-Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Liver Unit, CIBEREHD, Hospital Clínic, Barcelona, Catalonia, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.
| |
Collapse
|
164
|
Mitchell JK, Lemon SM, McGivern DR. How do persistent infections with hepatitis C virus cause liver cancer? Curr Opin Virol 2015; 14:101-8. [PMID: 26426687 PMCID: PMC4628866 DOI: 10.1016/j.coviro.2015.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/02/2015] [Accepted: 09/08/2015] [Indexed: 12/21/2022]
Abstract
Persistent infection with hepatitis C virus (HCV) is associated with an increased risk of hepatocellular carcinoma (HCC). Cancer typically develops in a setting of chronic hepatic inflammation and advanced fibrosis or cirrhosis, and such tissue represents a pre-neoplastic 'cancer field'. However, not all persistent infections progress to HCC and a combination of viral and host immune factors likely contributes to carcinogenesis. HCV may disrupt cellular pathways involved in detecting and responding to DNA damage, potentially adding to the risk of cancer. Efforts to unravel how HCV promotes HCC are hindered by lack of a robust small animal model, but a better understanding of molecular mechanisms could identify novel biomarkers for early detection and allow for development of improved therapies.
Collapse
Affiliation(s)
- Jonathan K Mitchell
- Division of Infectious Diseases, Department of Medicine, and Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stanley M Lemon
- Division of Infectious Diseases, Department of Medicine, and Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - David R McGivern
- Division of Infectious Diseases, Department of Medicine, and Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
165
|
Sasaki R, Kanda T, Wu S, Nakamoto S, Haga Y, Jiang X, Nakamura M, Shirasawa H, Yokosuka O. Association between hepatitis B virus and MHC class I polypeptide-related chain A in human hepatocytes derived from human-mouse chimeric mouse liver. Biochem Biophys Res Commun 2015; 464:1192-1195. [PMID: 26212443 DOI: 10.1016/j.bbrc.2015.07.102] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 07/21/2015] [Indexed: 01/10/2023]
Abstract
Due to the lack of efficient hepatitis B virus (HBV) infection systems, progress in understanding the role of innate immunity in HBV infection has remained challenging. Here we used human hepatocytes from a humanized severe combined immunodeficiency albumin promoter/enhancer driven-urokinase-type plasminogen activator mouse model for HBV infection. HBV DNA levels in culture medium from these human hepatocytes were 4.8-5.7 log IU/mL between day 16 and day 66 post-infection by HBV genotype C inoculum. HBV surface antigen (HBsAg) was also detected by chemiluminescent immunoassay from day 7 to day 66 post-infection. Western blot analysis revealed that major histocompatibility complex class I-related chain A (MICA), which plays a role in the innate immune system, was induced in HBV-infected human hepatocytes 27 days after infection compared with the uninfected control. MICA was reduced at day 62 and undetectable at day 90. Of interest, MICA expression by human hepatocytes increased after HBV infection and decreased before HBsAg loss. Human hepatocytes derived from chimeric mice with hepatocyte-humanized liver could support HBV genome replication. Further studies of the association between HBV replication and MICA induction should be conducted.
Collapse
Affiliation(s)
- Reina Sasaki
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Tatsuo Kanda
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.
| | - Shuang Wu
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Shingo Nakamoto
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan; Department of Molecular Virology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yuki Haga
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Xia Jiang
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Masato Nakamura
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Hiroshi Shirasawa
- Department of Molecular Virology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Osamu Yokosuka
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
166
|
Chiba T, Suzuki E, Saito T, Ogasawara S, Ooka Y, Tawada A, Iwama A, Yokosuka O. Biological features and biomarkers in hepatocellular carcinoma. World J Hepatol 2015; 7:2020-2028. [PMID: 26261691 PMCID: PMC4528275 DOI: 10.4254/wjh.v7.i16.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/06/2015] [Accepted: 07/27/2015] [Indexed: 02/06/2023] Open
Abstract
Similar to other cancers, a multistep process of carcinogenesis is observed in hepatocellular carcinoma (HCC). Although the mechanisms underlying the development of HCC have been investigated in terms of oncology, virology, and stem cell biology, the whole picture of hepatocarcinogenesis remains to be elucidated. Recent progress in molecular biology has provided clues to the underlying cause of various diseases. In particular, sequencing technologies, such as whole genome and exome sequencing analyses, have made an impact on genomic research on a variety of cancers including HCC. Comprehensive genomic analyses have detected numerous abnormal genetic alterations, such as mutations and copy number alterations. Based on these findings, signaling pathways and cancer-related genes involved in hepatocarcinogenesis could be analyzed in detail. Simultaneously, a number of novel biomarkers, both from tissue and blood samples, have been recently reported. These biomarkers have been successfully applied to early diagnosis and prognostic prediction of patients with HCC. In this review, we focus on the recent developments in molecular cancer research on HCC and explain the biological features and novel biomarkers.
Collapse
|
167
|
Zhang Y, Qian J, Wu M, Liu M, Zhang K, Lin Y, Guo X, Zhou Z, Hu Z, Sha J. A susceptibility locus rs7099208 is associated with non-obstructive azoospermia via reduction in the expression of FAM160B1. J Biomed Res 2015; 29:491-500. [PMID: 26668583 PMCID: PMC4662211 DOI: 10.7555/jbr.29.20150034] [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: 02/28/2015] [Revised: 03/08/2015] [Accepted: 05/06/2015] [Indexed: 12/25/2022] Open
Abstract
Non-obstructive azoospermia (NOA) is a severe defect in male reproductive health that occurs in 1% of adult men. In a previous study, we identified that rs7099208 is located within the last intron of FAM160B1 at 10q25.3. In this study, we analysed expression Quantitative Trait Loci (eQTL) of FAM160B1, ABLIM1 and TRUB1, the three genes surrounding rs7099208. Only the expression level of FAM160B1 was reduced for the homozygous alternate genotype (GG) of rs7099208, but not for the homozygous reference or heterozygous genotypes. FAM160B1 is predominantly expressed in human testes, where it is found in spermatocytes and round spermatids. From 17 patients with NOA and five with obstructive azoospermia (OA), immunohistochemistry revealed that expression of FAM160B1 is reduced, or undetectable in NOA patients, but not in OA cases or normal men. We conclude that rs7099208 is associated with NOA via a reduction in the expression of FAM160B1.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory of Reproductive Medicine ; The Center for Reproductive Medicine, The Second Affiliated Hospital of Nanjing Medical University , Nanjing, Jiangsu 210011 , China
| | - Jing Qian
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Minghui Wu
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Mingxi Liu
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Kai Zhang
- State Key Laboratory of Reproductive Medicine ; Department of Epidemiology and Biostatistics and Key Laboratory of Modern Toxicology of Ministry of Education Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Yuan Lin
- State Key Laboratory of Reproductive Medicine ; Department of Epidemiology and Biostatistics and Key Laboratory of Modern Toxicology of Ministry of Education Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Zuomin Zhou
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine ; Department of Epidemiology and Biostatistics and Key Laboratory of Modern Toxicology of Ministry of Education Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| |
Collapse
|
168
|
Zhu DY, Jiang LF, Deng XZ, Xiao W, Pei JP, Li BJ, Wang CJ, Zhang JH, Zhang Q, Zhou ZX, Ding WL, Xu XD, Yue M. TBX21 polymorphisms are associated with virus persistence in hepatitis C virus infection patients from a high-risk Chinese population. Eur J Clin Microbiol Infect Dis 2015; 34:1309-1318. [PMID: 25759111 DOI: 10.1007/s10096-015-2337-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 01/22/2015] [Indexed: 01/29/2023]
Abstract
Hepatitis C virus (HCV) infection is a major cause of chronic liver disease and the varied outcomes of the infection depend on both viral and host factors. We have demonstrated that the HCV alternate reading frame protein (F protein) is related to Th1/Th2 bias which is involved in virus persistence in chronic hepatitis C (CHC) patients. The purpose of this study was to test the hypothesis that genetic variants of TBX21 (T cell specific T-box transcription factor) were associated with the outcomes of HCV infection and F protein generation. Three single nucleotide polymorphisms (SNPs) (rs17250932, rs2074190, rs4794067) in the TBX21 gene were genotyped in a case-control study in a cohort of a high-risk group, including 354 healthy controls and 747 CHC patients (190 anti-F protein antibody seronegative patients and 557 anti-F protein antibody seropositive patients). Results showed that the rs4794067 C allele in the TBX21 promoter was significantly more common in CHC patients (OR = 1.335, 95% CI = 1.058-1.684, P = 0.015), exceptionally in anti-F protein seropositive patients (OR = 1.547, 95% CI = 1.140-2.101, P = 0.005), compared with healthy controls. And the risk effect was also significantly high in patients with HCV 1b genotype and mild fibrosis (P = 0.021, P = 0.010, respectively). Compared with the most frequent haplotype TAT, haplotype analysis showed that the distribution of TAC was significantly different between the chronic HCV carrier group and the healthy group, and so was the anti-F antibody seronegativity group and the anti-F antibody seronegativity group (all P < 0.001). Our results suggested that TBX21 variants may be involved in the etiology of this disease.
Collapse
Affiliation(s)
- D Y Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Nanjing Medical University, No. 293, Zhongshan East Road, Nanjing, 210002, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Abstract
Hepatitis C virus (HCV) is a leading etiology of hepatocellular carcinoma (HCC). The interaction of HCV with its human host is complex and multilayered; stemming in part from the fact that HCV is a RNA virus with no ability to integrate in the host's genome. Direct and indirect mechanisms of HCV-induced HCC include activation of multiple host pathways such as liver fibrogenic pathways, cellular and survival pathways, interaction with the immune and metabolic systems. Host factors also play a major role in HCV-induced HCC as evidenced by genomic studies identifying polymorphisms in immune, metabolic, and growth signaling systems associated with increased risk of HCC. Despite highly effective direct-acting antiviral agents, the morbidity and incidence of liver-related complications of HCV, including HCC, is likely to persist in the near future. Clinical markers to selectively identify HCV subjects at higher risk of developing HCC have been reported however they require further validation, especially in subjects who have experienced sustained virological response. Molecular biomarkers allowing further refinement of HCC risk are starting to be implemented in clinical platforms, allowing objective stratification of risk and leading to individualized therapy and surveillance for HCV individuals. Another role for molecular biomarker-based stratification could be enrichment of HCC chemoprevention clinical trials leading to smaller sample size, shorter trial duration, and reduced costs.
Collapse
Affiliation(s)
- Nicolas Goossens
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Division of Gastroenterology and Hepatology, Geneva University Hospital, Geneva, Switzerland
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
170
|
Pollicino T, Koumbi L. Role natural killer group 2D-ligand interactions in hepatitis B infection. World J Hepatol 2015; 7:819-824. [PMID: 25937859 PMCID: PMC4411524 DOI: 10.4254/wjh.v7.i6.819] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 02/17/2015] [Accepted: 03/16/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) infection is the leading cause of liver disease and hepatocellular carcinoma (HCC) worldwide, in spite of prophylactic vaccination and antiviral treatment modalities. The immunopathogenesis of HBV infection has been intensively studied and is propelled by complex interactions between the virus and the host immune system. Natural killer group 2D (NKG2D) is a well-characterized activating receptor, expressed on natural killer (NK) cells, NK T cells and CD8(+) cytotoxic T cells. This receptor is present in both humans and mice and binds to a diverge family of ligands that resemble the MHC-class I molecules. Increasing evidence shows that NKG2D-ligand interactions are critical in the establishment of HBV persistence and the development of liver injury and HCC. The expression of NKG2D ligands depends on the presence of several polymorphisms and is also modulated post-transcriptionally by HBV. While it is known that HBV circumvents host's innate immunity via the NKG2D pathway but the exact mechanisms involved are still elusive. This letter discusses previous accomplishments on the role of NKG2D ligand regulation in the development of chronic HBV, liver injury and HCC.
Collapse
Affiliation(s)
- Teresa Pollicino
- Teresa Pollicino, Department of Pediatric, Gynecologic, Microbiologic, and Biomedical Sciences, University Hospital of Messina, 98124 Messina, Italy
| | - Lemonica Koumbi
- Teresa Pollicino, Department of Pediatric, Gynecologic, Microbiologic, and Biomedical Sciences, University Hospital of Messina, 98124 Messina, Italy
| |
Collapse
|
171
|
Replication the association of 2q32.2–q32.3 and 14q32.11 with hepatocellular carcinoma. Gene 2015; 561:63-7. [DOI: 10.1016/j.gene.2015.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 01/21/2015] [Accepted: 02/05/2015] [Indexed: 01/05/2023]
|
172
|
Eslam M, Hashem AM, Leung R, Romero-Gomez M, Berg T, Dore GJ, Chan HL, Irving WL, Sheridan D, Abate ML, Adams LA, Mangia A, Weltman M, Bugianesi E, Spengler U, Shaker O, Fischer J, Mollison L, Cheng W, Powell E, Nattermann J, Riordan S, McLeod D, Armstrong NJ, Douglas MW, Liddle C, Booth DR, George J, Ahlenstiel G. Interferon-λ rs12979860 genotype and liver fibrosis in viral and non-viral chronic liver disease. Nat Commun 2015; 6:6422. [PMID: 25740255 PMCID: PMC4366528 DOI: 10.1038/ncomms7422] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 01/28/2015] [Indexed: 12/11/2022] Open
Abstract
Tissue fibrosis is a core pathologic process that contributes to mortality in ~45% of the population and is likely to be influenced by the host genetic architecture. Here we demonstrate, using liver disease as a model, that a single-nucleotide polymorphism (rs12979860) in the intronic region of interferon-λ4 (IFNL4) is a strong predictor of fibrosis in an aetiology-independent manner. In a cohort of 4,172 patients, including 3,129 with chronic hepatitis C (CHC), 555 with chronic hepatitis B (CHB) and 488 with non-alcoholic fatty liver disease (NAFLD), those with rs12979860CC have greater hepatic inflammation and fibrosis. In CHC, those with rs12979860CC also have greater stage-constant and stage-specific fibrosis progression rates (P<0.0001 for all). The impact of rs12979860 genotypes on fibrosis is maximal in young females, especially those with HCV genotype 3. These findings establish rs12979860 genotype as a strong aetiology-independent predictor of tissue inflammation and fibrosis.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Ahmed M. Hashem
- Faculty of Engineering, Department of Systems and Biomedical Engineering, Minia University, Minia 6111, Egypt
| | - Reynold Leung
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Sydney, New South Wales 2145, Australia
- Institute of Immunology and Allergy Research, Westmead Hospital and Westmead Millennium Institute, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Manuel Romero-Gomez
- Unit for The Clinical Management of Digestive Diseases and CIBERehd, Hospital Universitario de Valme, Sevilla 41014, Spain
| | - Thomas Berg
- Medizinische Klinik m.S. Hepatologie und Gastroenterologie, Charite, Campus Virchow-Klinikum, Universitätsmedizin Berlin, Berlin 04103, Germany
- Department of Hepatology, Clinic for Gastroenterology and Rheumatology, University Clinic Leipzig, Leipzig 04103, Germany
| | - Gregory J. Dore
- Kirby Institute, The University of New South Wales, Sydney, New South Wales 2052, Australia
- St Vincent’s Hospital, Sydney, New South Wales 2052, Australia
| | - Henry L.K. Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - William L. Irving
- NIHR Biomedical Research Unit in Gastroenterology and the Liver, University of Nottingham, Nottingham NG7 2UH, UK
| | - David Sheridan
- Liver Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Institute of Translational and Stratified Medicine, Plymouth University, Plymouth PL4 8AA, UK
| | - Maria L. Abate
- Division of Gastroenterology and Hepatology, Department of Medical Science, University of Turin, Turin 10126, Italy
| | - Leon A. Adams
- School of Medicine and Pharmacology, Sir Charles Gairdner Hospital Unit, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Alessandra Mangia
- Division of Hepatology, Ospedale Casa Sollievo della Sofferenza, IRCCS, San Giovanni Rotondo 71013, Italy
| | - Martin Weltman
- Department of Gastroenterology and Hepatology, Nepean Hospital, Sydney, New South Wales 2747, Australia
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Science, University of Turin, Turin 10126, Italy
| | - Ulrich Spengler
- Department of Internal Medicine I, University of Bonn, Bonn 53105, Germany
| | - Olfat Shaker
- Faculty of Medicine, Medical Biochemistry and Molecular Biology Department, Cairo University, Cairo 11562, Egypt
| | - Janett Fischer
- Medizinische Klinik m.S. Hepatologie und Gastroenterologie, Charite, Campus Virchow-Klinikum, Universitätsmedizin Berlin, Berlin 04103, Germany
| | - Lindsay Mollison
- Department of Gastroenterology and Hepatology, Fremantle Hospital, Fremantle, Western Australia 6160, Australia
| | - Wendy Cheng
- Department of Gastroenterology and Hepatology, Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | - Elizabeth Powell
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
- The University of Queensland, School of Medicine, Princess Alexandra Hospital, Woolloongabba, Queensland 4072, Australia
| | - Jacob Nattermann
- Department of Internal Medicine I, University of Bonn, Bonn 53105, Germany
| | - Stephen Riordan
- Gastrointestinal and Liver Unit, Prince of Wales Hospital and University of New South Wales, Sydney, New South Wales 2031, Australia
| | - Duncan McLeod
- Department of Anatomical Pathology, Institute of Clinical Pathology and Medical Research (ICPMR), Westmead Hospital, Sydney, New South Wales 2145, Australia
| | - Nicola J. Armstrong
- School of Mathematics and Statistics, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Mark W. Douglas
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Christopher Liddle
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Sydney, New South Wales 2145, Australia
| | - David R. Booth
- Institute of Immunology and Allergy Research, Westmead Hospital and Westmead Millennium Institute, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Jacob George
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Golo Ahlenstiel
- Storr Liver Unit, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Sydney, New South Wales 2145, Australia
| |
Collapse
|
173
|
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent causes of cancer-related death globally. Above well-known risk factors for HCC development ranging from various toxins to diseases such as diabetes mellitus, chronic infection with hepatitis B virus and hepatitis C virus (HCV) poses the most serious threat, constituting the cause in more than 80 % of cases. In addition to the viral genes intensively investigated, the pathophysiological importance of host genetic factors has also been greatly and increasingly appreciated. Genome-wide association studies (GWAS) comprehensively search the host genome at the single-nucleotide level, and have successfully identified the genomic region associated with a whole variety of diseases. With respect to HCC, there have been reports from several groups on single nucleotide polymorphisms (SNPs) associated with hepatocarcinogenesis, among which was our GWAS discovering MHC class I polypeptide-related sequence A (MICA) as a susceptibility gene for HCV-induced HCC. MICA is a natural killer (NK) group 2D (NKG2D) ligand, whose interaction with NKG2D triggers NK cell-mediated cytotoxicity toward the target cells, and is a key molecule in tumor immune surveillance as its expression is induced on stressed cells such as transformed tumor cells for the detection by NK cells. In this review, the latest understanding of the MICA-NKG2D system in viral HCC, particularly focused on its antitumor properties and the involvement of MICA SNPs, is summarized, followed by a discussion of targets for state-of-the-art cancer immunotherapy with personalized medicine in view.
Collapse
|
174
|
Chaiteerakij R, Addissie BD, Roberts LR. Update on biomarkers of hepatocellular carcinoma. Clin Gastroenterol Hepatol 2015; 13:237-45. [PMID: 24275343 PMCID: PMC4032371 DOI: 10.1016/j.cgh.2013.10.038] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/31/2013] [Accepted: 10/31/2013] [Indexed: 02/07/2023]
Abstract
New biomarkers of hepatocellular carcinoma (HCC) have been identified using advanced genomic, proteomic, and metabolomics technologies. These are being developed not only for use in diagnosis of HCC, but also in prediction of patient and treatment outcomes and individualization of therapy. Some HCC biomarkers are currently used in surveillance to detect early stage HCCs and reduce mortality. Further studies are needed to determine whether the recently identified HCC biomarkers can be used in clinical practice; most are only in phase 1 or 2 studies. The diagnostic and predictive abilities of biomarkers are limited by the heterogeneous nature of HCCs; there is no perfect single biomarker of this tumor. To improve performance, combinations of biomarkers (panels), or combinations of biomarkers and clinical parameters or laboratory test results, might be required. We describe recently discovered biomarkers of HCC and discuss challenges to their development and application.
Collapse
Affiliation(s)
- Roongruedee Chaiteerakij
- Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN,Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Benyam D. Addissie
- Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic and Mayo Clinic Cancer Center, Rochester, MN
| |
Collapse
|
175
|
Cheng CY, Yamashiro K, Chen LJ, Ahn J, Huang L, Huang L, Cheung CMG, Miyake M, Cackett PD, Yeo IY, Laude A, Mathur R, Pang J, Sim KS, Koh AH, Chen P, Lee SY, Wong D, Chan CM, Loh BK, Sun Y, Davila S, Nakata I, Nakanishi H, Akagi-Kurashige Y, Gotoh N, Tsujikawa A, Matsuda F, Mori K, Yoneya S, Sakurada Y, Iijima H, Iida T, Honda S, Lai TYY, Tam POS, Chen H, Tang S, Ding X, Wen F, Lu F, Zhang X, Shi Y, Zhao P, Zhao B, Sang J, Gong B, Dorajoo R, Yuan JM, Koh WP, van Dam RM, Friedlander Y, Lin Y, Hibberd ML, Foo JN, Wang N, Wong CH, Tan GS, Park SJ, Bhargava M, Gopal L, Naing T, Liao J, Ong PG, Mitchell P, Zhou P, Xie X, Liang J, Mei J, Jin X, Saw SM, Ozaki M, Mizoguchi T, Kurimoto Y, Woo SJ, Chung H, Yu HG, Shin JY, Park DH, Kim IT, Chang W, Sagong M, Lee SJ, Kim HW, Lee JE, Li Y, Liu J, Teo YY, Heng CK, Lim TH, Yang SK, Song K, Vithana EN, Aung T, Bei JX, Zeng YX, Tai ES, Li XX, Yang Z, Park KH, et alCheng CY, Yamashiro K, Chen LJ, Ahn J, Huang L, Huang L, Cheung CMG, Miyake M, Cackett PD, Yeo IY, Laude A, Mathur R, Pang J, Sim KS, Koh AH, Chen P, Lee SY, Wong D, Chan CM, Loh BK, Sun Y, Davila S, Nakata I, Nakanishi H, Akagi-Kurashige Y, Gotoh N, Tsujikawa A, Matsuda F, Mori K, Yoneya S, Sakurada Y, Iijima H, Iida T, Honda S, Lai TYY, Tam POS, Chen H, Tang S, Ding X, Wen F, Lu F, Zhang X, Shi Y, Zhao P, Zhao B, Sang J, Gong B, Dorajoo R, Yuan JM, Koh WP, van Dam RM, Friedlander Y, Lin Y, Hibberd ML, Foo JN, Wang N, Wong CH, Tan GS, Park SJ, Bhargava M, Gopal L, Naing T, Liao J, Ong PG, Mitchell P, Zhou P, Xie X, Liang J, Mei J, Jin X, Saw SM, Ozaki M, Mizoguchi T, Kurimoto Y, Woo SJ, Chung H, Yu HG, Shin JY, Park DH, Kim IT, Chang W, Sagong M, Lee SJ, Kim HW, Lee JE, Li Y, Liu J, Teo YY, Heng CK, Lim TH, Yang SK, Song K, Vithana EN, Aung T, Bei JX, Zeng YX, Tai ES, Li XX, Yang Z, Park KH, Pang CP, Yoshimura N, Wong TY, Khor CC. New loci and coding variants confer risk for age-related macular degeneration in East Asians. Nat Commun 2015; 6:6063. [PMID: 25629512 PMCID: PMC4317498 DOI: 10.1038/ncomms7063] [Show More Authors] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 12/09/2014] [Indexed: 01/17/2023] Open
Abstract
Age-related macular degeneration (AMD) is a major cause of blindness, but presents differently in Europeans and Asians. Here, we perform a genome-wide and exome-wide association study on 2,119 patients with exudative AMD and 5,691 controls, with independent replication in 4,226 patients and 10,289 controls, all of East Asian descent, as part of The Genetics of AMD in Asians (GAMA) Consortium. We find a strong association between CETP Asp442Gly (rs2303790), an East Asian-specific mutation, and increased risk of AMD (odds ratio (OR)=1.70, P=5.60 × 10−22). The AMD risk allele (442Gly), known to protect from coronary heart disease, increases HDL cholesterol levels by 0.17 mmol l−1 (P=5.82 × 10−21) in East Asians (n=7,102). We also identify three novel AMD loci: C6orf223 Ala231Ala (OR=0.78, P=6.19 × 10−18), SLC44A4 Asp47Val (OR=1.27, P=1.08 × 10−11) and FGD6 Gln257Arg (OR=0.87, P=2.85 × 10−8). Our findings suggest that some of the genetic loci conferring AMD susceptibility in East Asians are shared with Europeans, yet AMD in East Asians may also have a distinct genetic signature. Age-related macular degeneration (AMD) is a major cause of blindness worldwide. Here, the authors carry out a two-stage genome-wide association study for AMD and identify three new AMD risk loci, highlighting the shared and distinct genetic basis of the disease in East Asians and Europeans.
Collapse
Affiliation(s)
- Ching-Yu Cheng
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [3] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [4] Singapore National Eye Center, Singapore 168751, Singapore
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeeyun Ahn
- Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul 156-707, Korea
| | - Lulin Huang
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Lvzhen Huang
- 1] Key Laboratory of Vision Loss and Restoration, Ministry of Education of China, Beijing 100044, China [2] Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100871, China [3] Department of Ophthalmology, People's Hospital, Peking University, Beijing 100871, China
| | - Chui Ming G Cheung
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Singapore National Eye Center, Singapore 168751, Singapore
| | - Masahiro Miyake
- 1] Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan [2] Center for Genomic Medicine/Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Peter D Cackett
- 1] Singapore National Eye Center, Singapore 168751, Singapore [2] Princess Alexandra Eye Pavilion, Edinburgh EH3 9HA, UK
| | - Ian Y Yeo
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Augustinus Laude
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Ranjana Mathur
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Junxiong Pang
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Kar Seng Sim
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Adrian H Koh
- 1] Singapore National Eye Center, Singapore 168751, Singapore [2] Eye and Retinal Surgeons, Camden Medical Centre, Singapore 248649, Singapore
| | - Peng Chen
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Shu Yen Lee
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Doric Wong
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Choi Mun Chan
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Boon Kwang Loh
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Yaoyao Sun
- 1] Key Laboratory of Vision Loss and Restoration, Ministry of Education of China, Beijing 100044, China [2] Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100871, China [3] Department of Ophthalmology, People's Hospital, Peking University, Beijing 100871, China
| | - Sonia Davila
- 1] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [2] Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Isao Nakata
- 1] Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan [2] Center for Genomic Medicine/Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Hideo Nakanishi
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Yumiko Akagi-Kurashige
- 1] Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan [2] Center for Genomic Medicine/Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Norimoto Gotoh
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Akitaka Tsujikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine/Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Keisuke Mori
- Department of Ophthalmology, Saitama Medical University, Iruma 3500495, Japan
| | - Shin Yoneya
- Department of Ophthalmology, Saitama Medical University, Iruma 3500495, Japan
| | - Yoichi Sakurada
- Department of Ophthalmology, Faculty of Medicine, University of Yamanashi, Yamanashi 4093898, Japan
| | - Hiroyuki Iijima
- Department of Ophthalmology, Faculty of Medicine, University of Yamanashi, Yamanashi 4093898, Japan
| | - Tomohiro Iida
- Department of Ophthalmology, Tokyo Women's Medical University Hospital, Tokyo 1628666, Japan
| | - Shigeru Honda
- Department of Surgery, Division of Ophthalmology, Kobe University Graduate School of Medicine, Kobe 6500017, Japan
| | - Timothy Yuk Yau Lai
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Pancy Oi Sin Tam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Haoyu Chen
- 1] Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China [2] Shantou University/Chinese University of Hong Kong Joint Shantou International Eye Center, Shantou 515041, China
| | - Shibo Tang
- 1] Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China [2] Aier School of Ophthalmology, Central South University, Changsha 410000, China
| | - Xiaoyan Ding
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Feng Wen
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Fang Lu
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xiongze Zhang
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Yi Shi
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Bowen Zhao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Ophthalmology, Beijing 100730, China
| | - Jinghong Sang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Ophthalmology, Beijing 100730, China
| | - Bo Gong
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Rajkumar Dorajoo
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Jian-Min Yuan
- 1] Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15260, USA [2] Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | - Woon-Puay Koh
- 1] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [2] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | | | - Ying Lin
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Martin L Hibberd
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Jia Nee Foo
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Ophthalmology, Beijing 100730, China
| | - Chang Hua Wong
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Gavin S Tan
- Singapore National Eye Center, Singapore 168751, Singapore
| | - Sang Jun Park
- Department of Ophthalmology, Seoul National University Bundang Hospital, Gyeonggi 463-707, Korea
| | - Mayuri Bhargava
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Lingam Gopal
- Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Thet Naing
- Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Jiemin Liao
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Peng Guan Ong
- Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Paul Mitchell
- Department of Ophthalmology, University of Sydney and Westmead Millennium Institute, Sydney 2145, Australia
| | - Peng Zhou
- Eye and ENT Hospital of Fudan University, Shanghai 200433, China
| | | | | | - Junpu Mei
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen 518083, China
| | - Seang-Mei Saw
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [3] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [4] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | | | | | - Yasuo Kurimoto
- Department of Ophthalmology, Kobe City General Hospital, Kobe 6500046, Japan
| | - Se Joon Woo
- 1] Department of Ophthalmology, Seoul National University Bundang Hospital, Gyeonggi 463-707, Korea [2] Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Hum Chung
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Hyeong-Gon Yu
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Joo Young Shin
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Dong Ho Park
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu 700-721, Korea
| | - In Taek Kim
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu 700-721, Korea
| | - Woohyok Chang
- Department of Ophthalmology, Yeungnam University College of Medicine, Daegu 705-802, Korea
| | - Min Sagong
- Department of Ophthalmology, Yeungnam University College of Medicine, Daegu 705-802, Korea
| | - Sang-Joon Lee
- Department of Ophthalmology, College of Medicine, Kosin University, Pusan 606-701, Korea
| | - Hyun Woong Kim
- Department of Ophthalmology, Pusan Paik Hospital, Inje University College of Medicine, Pusan 614-735, Korea
| | - Ji Eun Lee
- 1] Department of Ophthalmology, Pusan National University Hospital, Pusan 602-739, Korea [2] Medical Research Institute, Pusan National University, Pusan 602-739, Korea
| | - Yi Li
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Jianjun Liu
- 1] Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore [2] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Yik Ying Teo
- 1] Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore [2] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Chew Kiat Heng
- Department of Pediatrics, National University Health System and National University of Singapore, Singapore 119228, Singapore
| | - Tock Han Lim
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center and University of Ulsan College of Medicine, Seoul 138-736, Korea
| | - Kyuyoung Song
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 138-736, Korea
| | - Eranga N Vithana
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [3] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Tin Aung
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [3] Singapore National Eye Center, Singapore 168751, Singapore
| | - Jin Xin Bei
- 1] State Key Laboratory of Oncology in Southern China, Guangzhou 510060, China [2] Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510080, China
| | - Yi Xin Zeng
- 1] State Key Laboratory of Oncology in Southern China, Guangzhou 510060, China [2] Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510080, China [3] Peking Union Medical College, Chinese Academy of Medical Science, Beijing 100730, China
| | - E Shyong Tai
- 1] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [2] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore [3] Department of Medicine, National University Health System and National University of Singapore, Singapore 119228, Singapore
| | - Xiao Xin Li
- 1] Key Laboratory of Vision Loss and Restoration, Ministry of Education of China, Beijing 100044, China [2] Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100871, China [3] Department of Ophthalmology, People's Hospital, Peking University, Beijing 100871, China
| | - Zhenglin Yang
- 1] Sichuan Provincial Key Laboratory for Human Disease Gene Study, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu 610072, China [2] School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Kyu-Hyung Park
- Department of Ophthalmology, Seoul National University Bundang Hospital, Gyeonggi 463-707, Korea
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Nagahisa Yoshimura
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Tien Yin Wong
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Duke-NUS Graduate Medical School, National University of Singapore, Singapore 169857, Singapore [3] Department of Ophthalmology, National University of Singapore and National University Health System, Singapore 119228, Singapore [4] Singapore National Eye Center, Singapore 168751, Singapore
| | - Chiea Chuen Khor
- 1] Singapore Eye Research Institute, Singapore 169856, Singapore [2] Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore [3] Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| |
Collapse
|
176
|
Yin J, Hou P, Wu Z, Wang T, Nie Y. Circulating miR-375 and miR-199a-3p as potential biomarkers for the diagnosis of hepatocellular carcinoma. Tumour Biol 2015; 36:4501-7. [PMID: 25618599 DOI: 10.1007/s13277-015-3092-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 01/08/2015] [Indexed: 12/26/2022] Open
Abstract
Aiming to find novel non-invasive biomarkers with high accuracy for the detention of early-stage hepatocellular carcinoma (HCC), we examined the predictive power of two microRNAs (miR; miR-375 and miR-199a-3p) as potential biomarkers in early-stage HCC. A total of 234 serum samples (78 samples from HCC patients, 156 samples from healthy controls) were collected. We measured the levels of the two mature microRNAs (miRNAs) (miR-375 and miR-199a-3p) with probe-based stem-loop quantitative reverse-transcriptase PCR (RT-qPCR) in all subjects. In addition, the correlation between the expression levels of two miRs and clinicopathological factors was explored. Receiver operating characteristic curve (ROC) analyses revealed that the two serum miRs could be promising biomarkers for HCC, with relatively high area under the curve (AUC) values as follows: miR-375, 0. 637 with 95 % confidence interval (CI) of 0.560-0.741; miR-199a-3p, 0. 883 with 95 % CI of 0.827-0.938. Stratified analyses indicated that circulating miR-199a-3p showed better predictive value in patients with long-term drinking. Our data suggested that circulating miR-375 and miR-199a-3p could be a novel serum biomarker for HCC. Nevertheless, further validating and improving study with larger sample should be conducted to confirm our results.
Collapse
Affiliation(s)
- Jian Yin
- Department of Gastroenterology, The First Affiliated to Chinese PLA General Hospital, 51 Fucheng Road, Beijing, 100048, China,
| | | | | | | | | |
Collapse
|
177
|
Tong HV, Song LH, Hoan NX, Cuong BK, Sy BT, Son HA, Quyet D, Binh VQ, Kremsner PG, Bock CT, Velavan TP, Toan NL. Soluble MICB protein levels and platelet counts during hepatitis B virus infection and response to hepatocellular carcinoma treatment. BMC Infect Dis 2015; 15:25. [PMID: 25626490 PMCID: PMC4318451 DOI: 10.1186/s12879-015-0754-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 01/14/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The human major histocompatibility complex class I polypeptide-related sequence B (MICB) is a protein that modulates the NK and T cell activation through the NKG2D receptor and is related to several diseases including cancer. METHODS The study investigated the prognostic role of soluble MICB (sMICB) protein in the progression of HBV-related liver diseases and to HBV-related HCC treatment. The sMICB serum levels were measured in 266 chronic HBV-infected Vietnamese patients and in healthy controls, and correlated with clinical and laboratory parameters and with therapeutic interventions for HBV-related HCC. RESULTS Significant differences in both clinical and laboratory parameters were observed among the patient groups with different stages of hepatitis. The platelet counts were significantly decreased with disease progression (P < 0.001). The sMICB serum levels were significantly increased in HBV patients compared to healthy controls (P < 0.0001). Among the patients with different stages of hepatitis, asymptomatic individuals (ASYM) revealed higher sMICB serum levels while liver cirrhosis (LC) patients revealed lower sMICB serum levels (P < 0.0001) compared to other patient groups. Notably, the sMICB serum levels were decreased in treated HCC patient group compared to not-treated HCC patient group (P = 0.05). Additionally, the sMICB levels were significantly correlated with platelet counts in ASYM and HCC patients (r = -0.37, P = 0.009; and r = 0.22, P = 0.025, respectively). CONCLUSIONS Our results demonstrate a potential role of sMICB serum levels and platelet counts during immune response to the HBV infection, liver disease progression and response to the HCC treatment.
Collapse
Affiliation(s)
- Hoang Van Tong
- Vietnam Military Medical University, 160 Phung Hung Street, Ha Dong District, Ha Noi, Viet Nam.
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.
| | - Le Huu Song
- Tran Hung Dao Hospital, 108 Institute of Clinical Medical and Pharmaceutical Sciences, No 1 Tran Hung Dao Street, Hai Ba Trung District, Hanoi, Vietnam.
| | - Nghiem Xuan Hoan
- Tran Hung Dao Hospital, 108 Institute of Clinical Medical and Pharmaceutical Sciences, No 1 Tran Hung Dao Street, Hai Ba Trung District, Hanoi, Vietnam.
| | - Bui Khac Cuong
- Vietnam Military Medical University, 160 Phung Hung Street, Ha Dong District, Ha Noi, Viet Nam.
- Department of Pathophysiology, Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Vietnam.
| | - Bui Tien Sy
- Vietnam Military Medical University, 160 Phung Hung Street, Ha Dong District, Ha Noi, Viet Nam.
- Robert Koch Institute, Seestrasse 10, D-13353, Berlin, Germany.
| | - Ho Anh Son
- Vietnam Military Medical University, 160 Phung Hung Street, Ha Dong District, Ha Noi, Viet Nam.
- Department of Pathophysiology, Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Vietnam.
| | - Do Quyet
- Vietnam Military Medical University, 160 Phung Hung Street, Ha Dong District, Ha Noi, Viet Nam.
| | - Vu Quoc Binh
- Vietnam Military Medical Bureau, No 276 Nghi Tam, Tay Ho District, Hanoi, Vietnam.
| | - Peter G Kremsner
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.
| | | | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.
- Fondation Congolaise pour la Recherche Medicale, P.O Box 2672, Brazzaville, Republic of Congo.
| | - Nguyen Linh Toan
- Vietnam Military Medical University, 160 Phung Hung Street, Ha Dong District, Ha Noi, Viet Nam.
- Department of Pathophysiology, Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Vietnam.
| |
Collapse
|
178
|
Marquardt JU, Andersen JB. Liver cancer oncogenomics: opportunities and dilemmas for clinical applications. Hepat Oncol 2015; 2:79-93. [PMID: 26257864 DOI: 10.2217/hep.14.24] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Primary liver cancers are among the most rapidly evolving malignant tumors worldwide. An underlying chronic inflammatory liver disease, which precedes liver cancer development for several decades and frequently creates a pro-oncogenic microenvironment, impairs progress in therapeutic approaches. Molecular heterogeneity of liver cancer is potentiated by a crosstalk between epithelial tumor and stromal cells that complicate translational efforts to unravel molecular mechanisms of hepatocarcinogenesis with a drugable intend. Next-generation sequencing has greatly advanced our understanding of cancer development. With regards to liver cancer, the unprecedented coverage of next-generation sequencing has created a detailed map of genetic alterations and identified key somatic changes such as CTNNB1 and TP53 as well as several previously unrecognized recurrent disease-causing alterations that could contribute to new therapeutic approaches. Importantly, these investigations indicate that a classical oncogene addiction cannot be assumed for primary liver cancer. Therefore, hepatocarcinogenesis can be considered a paradigm suitable for individualized medicine.
Collapse
Affiliation(s)
- Jens U Marquardt
- Department of Medicine I, Universitätsmedizin Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Jesper B Andersen
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| |
Collapse
|
179
|
Su CH, Lin Y, Cai L. Genetic factors, viral infection, other factors and liver cancer: an update on current progress. Asian Pac J Cancer Prev 2014; 14:4953-60. [PMID: 24175758 DOI: 10.7314/apjcp.2013.14.9.4953] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Primary liver cancer is one of the most common cancers at the global level, accounting for half of all cancers in some undeveloped countries. This disease tends to occur in livers damaged through alcohol abuse, or chronic infection with hepatitis B and C, on a background of cirrhosis. Various cancer-causing substances are associated with primary liver cancer, including certain pesticides and such chemicals as vinyl chloride and arsenic. The strong association between HBV infection and liver cancer is well documented in epidemiological studies. It is generally acknowledged that the virus is involved through long term chronic infection, frequently associated with cirrhosis, suggesting a nonspecific mechanism triggered by the immune response. Chronic inflammation of liver, continuous cell death, abnormal cell growth, would increase the occurrence rate of genetic alterations and risk of disease. However, the statistics indicated that only about one fifth of HBV carries would develop HCC in lifetime, suggesting that individual variation in genome would also influence the susceptibility of HCC. The goal of this review is to highlight present level of knowledge on the role of viral infection and genetic variation in the development of liver cancer.
Collapse
Affiliation(s)
- Cheng-Hao Su
- Department of Emergency Countermeasure and Information Management, Xiamen Center for Disease Control and Prevention, Xiamen, China E-mail :
| | | | | |
Collapse
|
180
|
Doganay L, Katrinli S, Colak Y, Senates E, Zemheri E, Ozturk O, Enc FY, Tuncer I, Doganay GD. HLA DQB1 alleles are related with nonalcoholic fatty liver disease. Mol Biol Rep 2014; 41:7937-7943. [PMID: 25156535 DOI: 10.1007/s11033-014-3688-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 08/20/2014] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease. NAFLD is a complex disease and inflammation is a crucial component in the disease pathogenesis. Recent genome wide association studies in hepatology area highlighted significant relations with human leukocyte antigen (HLA) DQ region and certain liver diseases. The previous animal models also emphasized the involvement of adaptive immune system in the liver damage pathways. To investigate possible polymorphisms in the HLA region that can contribute to the immune response affecting the NAFLD, we enrolled 93 consecutive biopsy proven NAFLD patients and a control group consisted of 101 healthy people and genotyped HLA DQB1 alleles at high resolution by sequence specific primers-polymerase chain reaction. The mean NAFLD activity score (NAS) was 5.2 ± 1.2, fibrosis score was 0.9 ± 0.9, ALT was 77 ± 47.4 U/L, AST was 49.4 ± 26.3 U/L. Among 13 HLA DQB1 alleles analyzed in this study, DQB1*06:04 was observed significantly at a more frequent rate among the NAFLD patients compared to that of healthy controls (12.9 vs. 2 % χ(2) = 8.6, P = 0.003, P c = 0.039, OR: 7.3 95 % CI 1.6-33.7). In addition, the frequency of DQB1*03:02 was significantly higher in the healthy control group than the NAFLD patients (24.8 vs. 7.5 %, χ(2) = 10.4, P = 0.001, P c = 0.013, OR: 0.2, 95 % CI 0.1-0.6). NAFLD patients were grouped according to their fibrosis score and NAS. The distribution of DQB1 alleles over stratified NAFLD patients did not reveal any statistically significant relation. Taken together, immune repertoire of individuals may have an effect on NAFLD pathogenesis and therefore, in NAFLD, adaptive immunity pathways should be investigated.
Collapse
Affiliation(s)
- Levent Doganay
- Department of Gastroenterology, Goztepe Teaching and Research Hospital, Medeniyet University, Istanbul, Turkey,
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Association between polymorphisms in tumor suppressor genes and oncogenes and risk of hepatocellular carcinoma: a case-control study in an HCC epidemic area within the Han Chinese population. Med Oncol 2014; 31:356. [PMID: 25412941 DOI: 10.1007/s12032-014-0356-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/12/2014] [Indexed: 02/08/2023]
Abstract
Data concerning the risk of hepatocellular carcinoma (HCC) and specific single nucleotide polymorphisms (SNPs) in an HBV-free population are currently limited. Therefore, we performed a case-control study to investigate the association between SNPs and the risk of HCC in individuals without chronic HBV infection. A total of 160 Han Chinese patients with HCC and an identical number of healthy controls were enrolled in this study. rs1042522, rs10814325, rs17401966, and rs2279744 genotypes were determined using matrix-associated laser desorption ionization-time of flight-mass spectrometry (MALDI-TOF-MS). CG and GG genotypes in rs1042522 and heterozygote and homozygote in rs2279744 were significantly associated with an elevated risk of HCC. Homozygous mutation of rs1081432 conferred a 2.68-fold risk of HCC (95% CI 1.35-5.34); however heterozygosity was not statistically significant. rs17401966 heterozygosity or homozygosity was not significantly associated with a increased risk of HCC. Several polymorphisms associated with a significantly increased risk of HCC were identified. These may serve as biomarkers in evaluating HCC risk in the general population.
Collapse
|
182
|
Abstract
Hepatitis C virus (HCV) is one of the major etiologic agents of liver cancer. HCV is an RNA virus that, unlike hepatitis B virus, is unable to integrate into the host genome. Through complex interactions between viral and host proteins that induce host responses and promote inflammation, fibrosis, and ultimately cirrhosis, HCV infection can result in the development of hepatocellular carcinoma (HCC). The HCV oncogenic process involves genetic and epigenetic alterations and oncogenic effects mediated by viral proteins in the activation of cellular oncogenes, inactivation of tumor-suppressor genes, and dysregulation of multiple signal-transduction pathways. Advances in genetics and gene expression profiling have enhanced our current understanding of the pathways involved in HCV-associated liver cancer development. In this review, we summarize the current understanding of mechanisms of hepatocarcinogenesis induced by HCV infection.
Collapse
Affiliation(s)
- Ming V Lin
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114; , ,
| | | | | |
Collapse
|
183
|
Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat Genet 2014; 46:1267-73. [PMID: 25362482 DOI: 10.1038/ng.3126] [Citation(s) in RCA: 603] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 10/03/2014] [Indexed: 11/08/2022]
Abstract
Diverse epidemiological factors are associated with hepatocellular carcinoma (HCC) prevalence in different populations. However, the global landscape of the genetic changes in HCC genomes underpinning different epidemiological and ancestral backgrounds still remains uncharted. Here a collection of data from 503 liver cancer genomes from different populations uncovered 30 candidate driver genes and 11 core pathway modules. Furthermore, a collaboration of two large-scale cancer genome projects comparatively analyzed the trans-ancestry substitution signatures in 608 liver cancer cases and identified unique mutational signatures that predominantly contribute to Asian cases. This work elucidates previously unexplored ancestry-associated mutational processes in HCC development. A combination of hotspot TERT promoter mutation, TERT focal amplification and viral genome integration occurs in more than 68% of cases, implicating TERT as a central and ancestry-independent node of hepatocarcinogenesis. Newly identified alterations in genes encoding metabolic enzymes, chromatin remodelers and a high proportion of mTOR pathway activations offer potential therapeutic and diagnostic opportunities.
Collapse
|
184
|
Kiryluk K, Li Y, Scolari F, Sanna-Cherchi S, Choi M, Verbitsky M, Fasel D, Lata S, Prakash S, Shapiro S, Fischman C, Snyder HJ, Appel G, Izzi C, Viola BF, Dallera N, Vecchio LD, Barlassina C, Salvi E, Bertinetto FE, Amoroso A, Savoldi S, Rocchietti M, Amore A, Peruzzi L, Coppo R, Salvadori M, Ravani P, Magistroni R, Ghiggeri GM, Caridi G, Bodria M, Lugani F, Allegri L, Delsante M, Maiorana M, Magnano A, Frasca G, Boer E, Boscutti G, Ponticelli C, Mignani R, Marcantoni C, Di Landro D, Santoro D, Pani A, Polci R, Feriozzi S, Chicca S, Galliani M, Gigante M, Gesualdo L, Zamboli P, Maixnerová D, Tesar V, Eitner F, Rauen T, Floege J, Kovacs T, Nagy J, Mucha K, Pączek L, Zaniew M, Mizerska-Wasiak M, Roszkowska-Blaim M, Pawlaczyk K, Gale D, Barratt J, Thibaudin L, Berthoux F, Canaud G, Boland A, Metzger M, Panzer U, Suzuki H, Goto S, Narita I, Caliskan Y, Xie J, Hou P, Chen N, Zhang H, Wyatt RJ, Novak J, Julian BA, Feehally J, Stengel B, Cusi D, Lifton RP, Gharavi AG. Discovery of new risk loci for IgA nephropathy implicates genes involved in immunity against intestinal pathogens. Nat Genet 2014; 46:1187-1196. [PMID: 25305756 PMCID: PMC4213311 DOI: 10.1038/ng.3118] [Citation(s) in RCA: 463] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 09/23/2014] [Indexed: 12/12/2022]
Abstract
We performed a genome-wide association study (GWAS) of IgA nephropathy (IgAN), the most common form of glomerulonephritis, with discovery and follow-up in 20,612 individuals of European and East Asian ancestry. We identified six new genome-wide significant associations, four in ITGAM-ITGAX, VAV3 and CARD9 and two new independent signals at HLA-DQB1 and DEFA. We replicated the nine previously reported signals, including known SNPs in the HLA-DQB1 and DEFA loci. The cumulative burden of risk alleles is strongly associated with age at disease onset. Most loci are either directly associated with risk of inflammatory bowel disease (IBD) or maintenance of the intestinal epithelial barrier and response to mucosal pathogens. The geospatial distribution of risk alleles is highly suggestive of multi-locus adaptation, and genetic risk correlates strongly with variation in local pathogens, particularly helminth diversity, suggesting a possible role for host-intestinal pathogen interactions in shaping the genetic landscape of IgAN.
Collapse
Affiliation(s)
- Krzysztof Kiryluk
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Yifu Li
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Francesco Scolari
- Div. of Nephrology, Azienda Ospedaliera Spedali Civili of Brescia, Montichiari Hospital, Univ of Brescia, Brescia, Italy
- Dept. of Medical and Surgical Specialties, Radiological Sciences, University of Brescia, Brescia, Italy
| | - Simone Sanna-Cherchi
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Murim Choi
- Dept. of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- Dept. of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Miguel Verbitsky
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - David Fasel
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Sneh Lata
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Sindhuri Prakash
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Samantha Shapiro
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Clara Fischman
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Holly J. Snyder
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Gerald Appel
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Claudia Izzi
- Div. of Nephrology, Azienda Ospedaliera Spedali Civili of Brescia, Montichiari Hospital, Univ of Brescia, Brescia, Italy
- Prenatal Diagnosis Unit, Department of Obstetrics and Gynecology, University of Brescia, Brescia, Italy
| | - Battista Fabio Viola
- Div. of Nephrology, Azienda Ospedaliera Spedali Civili of Brescia, Spedali Civili Hospital, Univ of Brescia, Brescia, Italy
| | - Nadia Dallera
- Div. of Nephrology, Azienda Ospedaliera Spedali Civili of Brescia, Montichiari Hospital, Univ of Brescia, Brescia, Italy
- Dept. of Medical and Surgical Specialties, Radiological Sciences, University of Brescia, Brescia, Italy
| | - Lucia Del Vecchio
- Renal Div., DMCO, San Paolo Hospital, School of Medicine, University of Milan, Milan, Italy
| | - Cristina Barlassina
- Renal Div., DMCO, San Paolo Hospital, School of Medicine, University of Milan, Milan, Italy
| | - Erika Salvi
- Renal Div., DMCO, San Paolo Hospital, School of Medicine, University of Milan, Milan, Italy
| | - Francesca Eleonora Bertinetto
- Immunogenetics and Biology of Transplantation, Città della Salute e della Scienza, University Hospital of Turin, Italy
- Medical Genetics, Dept. of Medical Sciences, University of Torino, Torino, Italy
| | - Antonio Amoroso
- Immunogenetics and Biology of Transplantation, Città della Salute e della Scienza, University Hospital of Turin, Italy
- Medical Genetics, Dept. of Medical Sciences, University of Torino, Torino, Italy
| | - Silvana Savoldi
- Nephrology and Dialysis Unit, Ospedali di Cirié e Chivasso, Cirié, Torino, Italy
| | - Marcella Rocchietti
- Nephrology and Dialysis Unit, Ospedali di Cirié e Chivasso, Cirié, Torino, Italy
| | - Alessandro Amore
- Nephrology, Dialysis, and Transplantation Unit, Regina Margheritra Hospital, Torino, Italy
| | - Licia Peruzzi
- Nephrology, Dialysis, and Transplantation Unit, Regina Margheritra Hospital, Torino, Italy
| | - Rosanna Coppo
- Nephrology, Dialysis, and Transplantation Unit, Regina Margheritra Hospital, Torino, Italy
| | - Maurizio Salvadori
- Div. of Nephrology and Renal Transplantation, Carreggi Hospital, Florence, Italy
| | - Pietro Ravani
- Dept. of Medicine, University of Calgary, Calgary, Canada
- Dept. of Community Health Sciences, University of Calgary, Calgary, Canada
| | - Riccardo Magistroni
- Div. of Nephrology Dialysis and Transplantation, Azienda Ospedaliero Universitaria Policlinico di Modena, Università di Modena e Reggio Emilia, Italy
| | - Gian Marco Ghiggeri
- Div. of Nephrology, Dialysis and Transplantation, Giannina Gaslini Institute, Genova, Italy
| | - Gianluca Caridi
- Div. of Nephrology, Dialysis and Transplantation, Giannina Gaslini Institute, Genova, Italy
| | - Monica Bodria
- Div. of Nephrology, Dialysis and Transplantation, Giannina Gaslini Institute, Genova, Italy
| | - Francesca Lugani
- Div. of Nephrology, Dialysis and Transplantation, Giannina Gaslini Institute, Genova, Italy
| | - Landino Allegri
- Div. of Nephrology, Azienda Ospedaliero-Universitaria and Chair of Nephrology, University of Parma, Parma, Italy
| | - Marco Delsante
- Div. of Nephrology, Azienda Ospedaliero-Universitaria and Chair of Nephrology, University of Parma, Parma, Italy
| | - Mariarosa Maiorana
- Div. of Nephrology, Azienda Ospedaliero-Universitaria and Chair of Nephrology, University of Parma, Parma, Italy
| | - Andrea Magnano
- Div. of Nephrology, Azienda Ospedaliero-Universitaria and Chair of Nephrology, University of Parma, Parma, Italy
| | - Giovanni Frasca
- Div. of Nephrology, Dialysis and Renal Transpantation, Riuniti Hospital, Ancona, Italy
| | - Emanuela Boer
- Div. of Nephrology and Dialysis, Gorizia Hospital, Gorizia, Italy
| | - Giuliano Boscutti
- Div. of Nephrology, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Trieste, Trieste, Italy
| | | | - Renzo Mignani
- Div. of Nephrology and Dialysis, Infermi Hospital, Rimini, Italy
| | | | | | - Domenico Santoro
- Div. of Nephology and Dialysis, Chair of Nephrology, University of Messina, Azienda Ospedaliero-Universitaria Policlinico, Messina, Italy
| | - Antonello Pani
- Dept. of Nephrology and Dialysis, G. Brotzu Hospital, Cagliari, Italy
| | - Rosaria Polci
- Nephrology and Dialysis, Hospital of Viterbo, Viterbo, Italy
| | - Sandro Feriozzi
- Nephrology and Dialysis, Hospital of Viterbo, Viterbo, Italy
| | - Silvana Chicca
- Div. of Nephrology and Dialysis, Sandro Pertini Hospital, Rome, Italy
| | - Marco Galliani
- Div. of Nephrology and Dialysis, Sandro Pertini Hospital, Rome, Italy
| | - Maddalena Gigante
- Dept. of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Loreto Gesualdo
- Section of Nephrology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | | | - Dita Maixnerová
- Dept. of Nephrology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Vladimir Tesar
- Dept. of Nephrology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Frank Eitner
- Dept. of Nephrology, RWTH University of Aachen, Aachen, Germany
- Kidney Diseases Research, Bayer Pharma AG, Wuppertal, Germany
| | - Thomas Rauen
- Dept. of Nephrology, RWTH University of Aachen, Aachen, Germany
| | - Jürgen Floege
- Dept. of Nephrology, RWTH University of Aachen, Aachen, Germany
| | - Tibor Kovacs
- Nephrology Center, Medical Faculty, University of Pécs, Pécs, Hungary
- Second Dept. of Internal Medicine, Medical Faculty, University of Pécs, Pécs, Hungary
| | - Judit Nagy
- Nephrology Center, Medical Faculty, University of Pécs, Pécs, Hungary
- Second Dept. of Internal Medicine, Medical Faculty, University of Pécs, Pécs, Hungary
| | - Krzysztof Mucha
- Dept. of Immunology, Transplantology, and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Pączek
- Dept. of Immunology, Transplantology, and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Zaniew
- Children’s Hospital, Krysiewicza 7/8, Poznań, Poland
| | | | | | - Krzysztof Pawlaczyk
- Dept. of Nephrology, Transplantology, and Internal Medicine, Poznan Medical University, Poznan, Poland
| | - Daniel Gale
- University College London-Centre for Nephrology, Royal Free Hospital Pond Street, London
| | - Jonathan Barratt
- The John Walls Renal Unit, University Hospitals of Leicester, Leicester, United Kingdom
- Dept. of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Lise Thibaudin
- Nephrology, Dialysis, and Renal Transplantation Dept., University North Hospital, Saint Etienne, France
| | - Francois Berthoux
- Nephrology, Dialysis, and Renal Transplantation Dept., University North Hospital, Saint Etienne, France
| | - Guillaume Canaud
- Service de Néphrologie Transplantation Adultes, Hôpital Necker - Enfants Malades, Paris, France
- INSERM, Institut Necker Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Boland
- Centre National de Génotypage, CEA, Institut de Génomique, Evry, France
| | - Marie Metzger
- INSERM, Centre for Research in Epidemiology and Population Health, Villejuif, France and University Paris-Sud, Villejuif, France
| | - Ulf Panzer
- III Medizinische Klinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Hitoshi Suzuki
- Division of Nephrology, Dept. of Internal Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Shin Goto
- Division of Clinical Nephrology and Rheumatology, Niigata University, Niigata, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Niigata University, Niigata, Japan
| | - Yasar Caliskan
- Division of Nephrology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Jingyuan Xie
- Dept. of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Hou
- Renal Div., Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Nan Chen
- Dept. of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Zhang
- Renal Div., Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Robert J. Wyatt
- Div. of Pediatric Nephrology, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
- Children’s Foundation Research Center, Le Bonheur Children’s Hospital, Memphis, Tennessee, USA
| | - Jan Novak
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bruce A. Julian
- Dept. of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John Feehally
- The John Walls Renal Unit, University Hospitals of Leicester, Leicester, United Kingdom
- Dept. of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Benedicte Stengel
- INSERM, Centre for Research in Epidemiology and Population Health, Villejuif, France and University Paris-Sud, Villejuif, France
| | - Daniele Cusi
- Renal Div., DMCO, San Paolo Hospital, School of Medicine, University of Milan, Milan, Italy
| | - Richard P. Lifton
- Dept. of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ali G. Gharavi
- Dept. of Medicine, Div. of Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
185
|
Hoshida Y, Fuchs BC, Bardeesy N, Baumert TF, Chung RT. Pathogenesis and prevention of hepatitis C virus-induced hepatocellular carcinoma. J Hepatol 2014; 61:S79-90. [PMID: 25443348 PMCID: PMC4435677 DOI: 10.1016/j.jhep.2014.07.010] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 07/03/2014] [Accepted: 07/10/2014] [Indexed: 02/08/2023]
Abstract
Hepatitis C virus (HCV) is one of the major aetiologic agents that causes hepatocellular carcinoma (HCC) by generating an inflammatory, fibrogenic, and carcinogenic tissue microenvironment in the liver. HCV-induced HCC is a rational target for cancer preventive intervention because of the clear-cut high-risk condition, cirrhosis, associated with high cancer incidence (1% to 7% per year). Studies have elucidated direct and indirect carcinogenic effects of HCV, which have in turn led to the identification of candidate HCC chemoprevention targets. Selective molecular targeted agents may enable personalized strategies for HCC chemoprevention. In addition, multiple experimental and epidemiological studies suggest the potential value of generic drugs or dietary supplements targeting inflammation, oxidant stress, or metabolic derangements as possible HCC chemopreventive agents. While the successful use of highly effective direct-acting antiviral agents will make important inroads into reducing long-term HCC risk, there will remain an important role for HCC chemoprevention even after viral cure, given the persistence of HCC risk in persons with advanced HCV fibrosis, as shown in recent studies. The successful development of cancer preventive therapies will be more challenging compared to cancer therapeutics because of the requirement for larger and longer clinical trials and the need for a safer toxicity profile given its use as a preventive agent. Molecular biomarkers to selectively identify high-risk population could help mitigate these challenges. Genome-wide, unbiased molecular characterization, high-throughput drug/gene screening, experimental model-based functional analysis, and systems-level in silico modelling are expected to complement each other to facilitate discovery of new HCC chemoprevention targets and therapies.
Collapse
Affiliation(s)
- Yujin Hoshida
- Liver Cancer Program, Tisch Cancer Institute, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, United States.
| | - Bryan C Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, United States
| | - Nabeel Bardeesy
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, United States
| | - Thomas F Baumert
- INSERM Unité 1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, and Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, France; Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, United States
| | - Raymond T Chung
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, United States.
| |
Collapse
|
186
|
Abstract
The major histocompatibility complex class I polypeptide-related sequence A gene (MICA) encodes a membrane-bound protein acting as a ligand to stimulate an activating receptor, NKG2D, expressed on the surface of essentially all human natural killer (NK), γδ T and CD8(+) αβ T cells. MICA protein is absent from most cells but can be induced by infections and oncogenic transformation and is frequently expressed in epithelial tumors. Upon binding to MICA, NKG2D activates cytolytic responses of NK and γδ T cells against infected and tumor cells expressing MICA. Therefore, membrane-bound MICA acts as a signal during the early immune response against infection or spontaneously arising tumors. On the other hand, human tumor cells spontaneously release a soluble form of MICA, causing the downregulation of NKG2D and in turn severe impairment of the antitumor immune response of NK and CD8(+) T cells. This is considered to promote tumor immune evasion and also to compromise host resistance to infections. MICA is the most polymorphic non-classical class I gene. A possible association of MICA polymorphism with genetic predisposition to different cancer types has been investigated in candidate gene-based studies. Two genome-wide association studies have identified loci in MICA that influence susceptibility to cervical neoplasia and hepatitis C virus-induced hepatocellular carcinoma, respectively. Given the current level of interest in the field of MICA gene, we discuss the genetics and biology of the MICA gene and the role of its polymorphism in cancer. Gaps in our understanding and future research needs are also discussed.
Collapse
Affiliation(s)
- Dan Chen
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala, Uppsala University, Biomedical Center, Box 815, 75108 Uppsala, Sweden
| | - Ulf Gyllensten
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala, Uppsala University, Biomedical Center, Box 815, 75108 Uppsala, Sweden
| |
Collapse
|
187
|
Pulse-wave velocity, the ankle-brachial index, and the visceral fat area are highly associated with colorectal adenoma. Dig Liver Dis 2014; 46:943-9. [PMID: 24953207 DOI: 10.1016/j.dld.2014.05.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 05/06/2014] [Accepted: 05/17/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although both colorectal neoplasms and atherosclerosis may be induced by visceral fat accumulation, no association between these factors has yet been elucidated. METHODS Among Japanese participants in our colonoscopic screening and annual surveillance programs, asymptomatic subjects were investigated. Visceral fat area was measured by computed tomography, and the ankle-brachial index and pulse-wave velocity (markers of systemic atherosclerosis) were also estimated. Colorectal findings during screening were investigated in association with these parameters. RESULTS Nine hundred and seven subjects (males: 71.9%; mean age: 57.3±8.7 years) were enrolled. Colorectal adenomas were identified during colorectal screening of 407 subjects (44.9%). The prevalence of all colorectal adenoma and advanced neoplasia increased with an elevated pulse-wave velocity from 32.2% (advanced neoplasia, 2.6) to 62.1% (8.4%) in higher quartiles (p<0.001 and p=0.003). The risk of advanced neoplasia was higher in subjects with an abnormal ankle-brachial index (33% vs. 4.6%, p<0.001). Upon multivariate analysis, visceral fat area and pulse-wave velocity were associated with all adenoma, and the ankle-brachial index with advanced neoplasia. CONCLUSIONS The risk of colorectal adenoma was strongly associated with visceral fat area and markers of atherosclerosis. Colorectal adenoma and atherosclerosis may share a common pathogenesis and the same populations are at high-risk.
Collapse
|
188
|
Tong HV, Bock CT, Velavan TP. Genetic insights on host and hepatitis B virus in liver diseases. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2014; 762:65-75. [PMID: 25475418 DOI: 10.1016/j.mrrev.2014.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 02/08/2023]
Abstract
Hepatitis B virus (HBV) infection is a major global health problem and many studies have underlined the importance of inter individual variability and somatic mutations during the clinical course of HBV infection. In recent years, high-throughput technologies have provided new possibilities to study the genetic basis of many diseases. We reviewed all literature available on genome-wide association studies (GWASs), whole genome, exome and RNA sequencing studies as well as studies on HBV infection and the pathogenesis of related liver disease. Many GWASs conclude that the genetic variants in the HLA region (HLA-DP, HLA-DQ, HLA-DR and MICA), KIF1B, DEPDC5 and PNPLA3 influence HBV infection, its clinical course and the response to hepatitis B vaccination. The next generation sequencing approach provides important clues on the mutational landscape of genes involved in signaling pathways in particular JAK/STAT, Wnt/β-catenin, p53 pathways and multiple chromatin regulator genes that significantly promote hepatocarcinogenesis. In addition, the hotspots of recurrent integrations of HBV-DNA into host chromosomes such as hTERT, PDGF receptor, MLL are involved in pathogenesis of hepatocellular carcinoma (HCC). Additionally, the transitions T>C/A>G, C>T/G>A, C>A/G>T and T>A/A>T remain specific for HCC induced by viral infection and the DNA methylation in the CpG island is proposed as a biomarker for HCC. We have described common mutations in the HBV genome (G1896A, rtM204V, rtM204I) which modulate the pathogenesis and carcinogenesis of the liver. Further GWASs in different ethnic groups and additional functional studies are required to warrant the significance of such defined genetic factors. Such findings continue to shape our understanding of the genetic architecture of host-virus interactions and provide new clues and directions in determining genetic markers that modulate HBV infection and related liver diseases. The studies using high-throughput technologies help identifying potential genetic threats however the utility of mutational information can be complex in predicting prognostic significance and shall pose challenges to its clinical implementation.
Collapse
Affiliation(s)
- Hoang van Tong
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.
| | - C Thomas Bock
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | | |
Collapse
|
189
|
Sato M, Kato N, Tateishi R, Muroyama R, Kowatari N, Li W, Goto K, Otsuka M, Shiina S, Yoshida H, Omata M, Koike K. Impact of PNPLA3 polymorphisms on the development of hepatocellular carcinoma in patients with chronic hepatitis C virus infection. Hepatol Res 2014; 44:E137-E144. [PMID: 24125181 DOI: 10.1111/hepr.12258] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/02/2013] [Accepted: 10/04/2013] [Indexed: 02/06/2023]
Abstract
AIM The PNPLA3 rs738409 C>G polymorphism (encoding for I148M) has recently been identified as a susceptibility factor for steatosis-mediated liver damage. We evaluated the influence of this polymorphism on hepatocarcinogenesis in patients with chronic hepatitis C (CHC) virus infection. METHODS We genotyped the rs738409 single nucleotide polymorphism in 358 hepatitis C-associated hepatocellular carcinoma (HCC) patients and correlated the age at onset of HCC and the interval between hepatitis C virus (HCV) infection and the development of HCC in patients with each genotype. RESULTS The frequencies of CC, CG and GG genotypes were 27.9% (100/358), 49.2% (176/358) and 22.9% (82/358), respectively, and were in Hardy-Weinberg equilibrium. The median age at onset of HCC for the GG genotype was significantly younger compared to for non-GG genotypes (67.81 vs 69.87 years, P < 0.001), and the median interval between HCV infection and the development of HCC was significantly shorter in patients with the GG genotype (39.96 vs 40.85 years, P = 0.008). PNPLA3 GG genotype was also associated with a higher aspartate aminotransferase level (69.5 vs 59.0 IU/L, P = 0.02), lower prothrombin time (73.0% vs 78.0%, P = 0.008) and a higher prevalence of histological steatosis (40.0% vs. 22.2%, P = 0.01) at the time of HCC onset. CONCLUSION The PNPLA3 genotype GG may be associated with accelerated hepatocarcinogenesis in CHC patients through increased steatosis in the liver.
Collapse
Affiliation(s)
- Masaya Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Dongiovanni P, Romeo S, Valenti L. Hepatocellular carcinoma in nonalcoholic fatty liver: Role of environmental and genetic factors. World J Gastroenterol 2014; 20:12945-12955. [PMID: 25278690 PMCID: PMC4177475 DOI: 10.3748/wjg.v20.i36.12945] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/28/2014] [Accepted: 05/26/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth cause of cancer related mortality, and its incidence is rapidly increasing. Viral hepatitis, alcohol abuse, and exposure to hepatotoxins are major risk factors, but nonalcoholic fatty liver disease (NAFLD) associated with obesity, insulin resistance, and type 2 diabetes, is an increasingly recognized trigger, especially in developed countries. Older age, severity of insulin resistance and diabetes, and iron overload have been reported to predispose to HCC in this context. Remarkably, HCCs have been reported in non-cirrhotic livers in a higher proportion of cases in NAFLD patients than in other etiologies. Inherited factors have also been implicated to explain the different individual susceptibility to develop HCC, and their role seems magnified in fatty liver, where only a minority of affected subjects progresses to cancer. In particular, the common I148M variant of the PNPLA3 gene influencing hepatic lipid metabolism influences HCC risk independently of its effect on the progression of liver fibrosis. Recently, rare loss-of-function mutations in Apolipoprotein B resulting in very low density lipoproteins hepatic retention and in Telomerase reverse transcriptase influencing cellular senescence have also been linked to HCC in NAFLD. Indeed, hepatic stellate cells senescence has been suggested to bridge tissue aging with alterations of the intestinal microbiota in the pathogenesis of obesity-related HCC. A deeper understanding of the mechanisms mediating hepatic carcinogenesis during insulin resistance, and the identification of its genetic determinants will hopefully provide new diagnostic and therapeutic tools.
Collapse
|
191
|
Mitchell JK, McGivern DR. Mechanisms of hepatocarcinogenesis in chronic hepatitis C. Hepat Oncol 2014; 1:293-307. [PMID: 30190964 DOI: 10.2217/hep.14.7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Infection with hepatitis C virus (HCV) is a major risk factor for hepatocellular carcinoma. The genetic changes that drive cancer development are heterogeneous and how chronic hepatitis C promotes the initiation of hepatocellular carcinoma is incompletely understood. Cancer typically arises in the setting of advanced fibrosis and/or cirrhosis where chronic immune-mediated inflammation over decades promotes hepatocyte turnover providing selective pressure that favors the malignant phenotype. As well as contributions of unresolved inflammation to carcinogenesis, evidence from transgenic mice with liver-specific expression of viral sequences suggests that some HCV-encoded proteins may directly promote cancer. Numerous in vitro studies suggest roles for HCV proteins in subversion of cellular pathways that normally act to suppress tumorigenesis. Here, we review the mechanisms by which persistent HCV infection might promote cancer in addition to the procarcinogenic effects of inflammatory liver disease.
Collapse
Affiliation(s)
- Jonathan K Mitchell
- Lineberger Comprehensive Cancer Center & Division of Infectious Diseases, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599-7295, USA
| | - David R McGivern
- Lineberger Comprehensive Cancer Center & Division of Infectious Diseases, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599-7295, USA
| |
Collapse
|
192
|
Zhao X, Hu S, Wang L, Zhang Q, Zhu X, Zhao H, Wang C, Tao R, Guo S, Wang J, Xu J, He Y, Gao Y. Functional short tandem repeat polymorphism of PTPN11 and susceptibility to hepatocellular carcinoma in Chinese populations. PLoS One 2014; 9:e106841. [PMID: 25198338 PMCID: PMC4157842 DOI: 10.1371/journal.pone.0106841] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/01/2014] [Indexed: 02/07/2023] Open
Abstract
Background PTPN11, which encodes tyrosine phosphatase Shp2, is a critical gene mediating cellular responses to hormones and cytokines. Loss of Shp2 promotes hepatocellular carcinoma (HCC), suggesting that PTPN11 functions as a tumor suppressor in HCC tumorgenesis. The aim of this study was to evaluate the effects of the short tandem repeat (STR) polymorphism (rs199618935) within 3'UTR of PTPN11 on HCC susceptibility in Chinese populations. Methodology/Principal Findings We analyzed the associations in 400 patients from Jiangsu province of China, validating the findings in an additional 305 patients from Shanghai of China. Unconditional logistic regression was used to analyze the association between rs199618935 and HCC risk. Additional biochemical investigations and in-silico studies were used to evaluate the possible functional significance of this polymorphism. Logistic regression analysis showed that compared with individuals carrying shorter alleles (11 and 12 repeats), those subjects who carry longer alleles (13 and 14 repeats) had a significantly decreased risk of HCC [adjusted odds ratio (OR) = 0.63, 95% confidence interval (CI) = 0.53–0.76, P = 2.00×10−7], with the risk decreased even further in those carrying allele 15 and 16 (adjusted OR = 0.46, 95% CI = 0.34–0.62, P = 1.00×10−7). Biochemical investigations showed that longer alleles of rs199618935 conferred higher PTPN11 expression in vivo and in vitro. The altered luciferase activities in reporter gene system suggested that STR regulation of PTPN11 expression could be a transcriptional event. Finally, in-silico prediction revealed that different alleles of rs199618935 could alter the local structure of PTPN11 mRNA. Conclusions/Significance Taken together, our findings suggested that the STR polymorphism within PTPN11 contributes to hepatocarcinogenesis, possibly by affecting PTPN11 expression through a structure-dependent mechanism. The replication of our studies and further functional studies are needed to validate our findings.
Collapse
Affiliation(s)
- Xiankun Zhao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Shuxiang Hu
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Lu Wang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Qing Zhang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Xiaodan Zhu
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Hua Zhao
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Chaoqun Wang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Ruiyang Tao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Siping Guo
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jing Wang
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jiejie Xu
- Key Laboratory of Medical Molecular Virology, MOE & MOH, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan He
- Department of Epidemiology, Medical College of Soochow University, Suzhou, Jiangsu, P. R. China
- * E-mail: (YG); (YH)
| | - Yuzhen Gao
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, Jiangsu, P.R. China
- * E-mail: (YG); (YH)
| |
Collapse
|
193
|
Liu M, Jiang L, Guan XY. The genetic and epigenetic alterations in human hepatocellular carcinoma: a recent update. Protein Cell 2014; 5:673-91. [PMID: 24916440 PMCID: PMC4145080 DOI: 10.1007/s13238-014-0065-9] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 04/13/2014] [Indexed: 01/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent human malignancies worldwide with very poor prognosis. It is generally accepted that the progression of HCC is a long-term process with accumulation of multiple genetic and epigenetic alterations, which further lead to the activation of critical oncogenes or inactivation of tumor suppressor genes. HCC is characterized with multiple cancer hallmarks including their ability to proliferate, anti-apoptosis, invade, metastasis, as well as the emerging features such as stem cell properties and energy metabolic switch. The irreversible alterations at genetic level could be detected as early as in the pre-neoplastic stages and accumulate during cancer progression. Thus, they might account for the cancer initiating steps and further malignant transformation. In addition to genetic alterations, epigenetic alterations can affect the cancer transcriptome more extensively. Alterations in DNA methylation, histone modification, miRNAs, RNA editing, and lncRNAs might result in disrupted gene regulation networks and substantially contribute to HCC progression. In this review, the genetic and epigenetic alterations which significantly contribute to the malignant capabilities of HCC will be updated and summarized in detail. Further characterization of those critical molecular events might better elucidate the pathogenesis of HCC and provide novel therapeutic targets for treatment of this deadly disease.
Collapse
Affiliation(s)
- Ming Liu
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, China
| | - Lingxi Jiang
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, China
| |
Collapse
|
194
|
The association of toll-like receptor 4 polymorphism with hepatitis C virus infection in Saudi Arabian patients. BIOMED RESEARCH INTERNATIONAL 2014; 2014:357062. [PMID: 25177689 PMCID: PMC4142570 DOI: 10.1155/2014/357062] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 07/09/2014] [Indexed: 02/08/2023]
Abstract
Hepatitis C virus (HCV) is a single stranded RNA virus. It affects millions of people worldwide and is considered as a leading cause of liver diseases including cirrhosis and hepatocellular carcinoma. A recent study reported that TLR4 gene polymorphisms are good prognostic predictors and are associated with protection from liver fibrosis among Caucasians. This study aims to investigate the implication of genetic polymorphisms of TLR4 gene on the HCV infection in Saudi Arabian patients. Two SNPs in the TLR4 gene, rs4986790 (A/G) and rs4986791 (C/T), were genotyped in 450 HCV patients and 600 uninfected controls. The association analysis confirmed that both SNPs showed a significant difference in their distribution between HCV-infected patients and uninfected control subjects (P < 0.0001; OR = 0.404, 95% CI = 0.281-0.581) and (P < 0.0001; OR = 0.298, 95% CI = 0.201-0.443), respectively. More importantly, haplotype analysis revealed that four haplotypes, AC, GT, GC, and AT (rs4986790, rs4986791), were significantly associated with HCV infection when compared with control subjects. One haplotype AC was more prominently found when chronic HCV-infected patients were compared with cirrhosis/HCC patients (frequency = 94.7% and P = 0.04). Both TLR4 SNPs under investigation were found to be significantly implicated with HCV-infection among Saudi Arabian population.
Collapse
|
195
|
Abel L, Alcaïs A, Schurr E. The dissection of complex susceptibility to infectious disease: bacterial, viral and parasitic infections. Curr Opin Immunol 2014; 30:72-8. [PMID: 25083600 DOI: 10.1016/j.coi.2014.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/17/2014] [Accepted: 07/06/2014] [Indexed: 01/01/2023]
Abstract
Infectious diseases are the result of the exposure of susceptible hosts to pathogenic microbes. Genetic factors are important determinants of host susceptibility and efforts are being made to establish the molecular identity of such genetic susceptibility variants by genome-wide association studies. Results obtained to date partly confirm already known genetic vulnerabilities, but also point to new and unexpected mechanisms of susceptibility that extend from classical innate and acquired immunity to weaknesses in constitutional resistance. These studies also revealed an overlap in genetic control between infectious disease and other common immune and inflammatory disorders.
Collapse
Affiliation(s)
- Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U.980, University Paris Descartes, Necker Enfants-Malades Hospital, Paris 75015, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Alexandre Alcaïs
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U.980, University Paris Descartes, Necker Enfants-Malades Hospital, Paris 75015, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA; URC, CIC, Necker and Cochin Hospitals, Paris, France
| | - Erwin Schurr
- McGill International TB Centre & Departments of Human Genetics and Medicine, McGill University, Montreal, Quebec, Canada; Program in Immunology and Infectious Diseases in Global Health, The Research Institute of the McGill University Health Centre, Canada.
| |
Collapse
|
196
|
Ohno M, Otsuka M, Kishikawa T, Shibata C, Yoshikawa T, Takata A, Muroyama R, Kowatari N, Sato M, Kato N, Kuroda S, Koike K. Specific delivery of microRNA93 into HBV-replicating hepatocytes downregulates protein expression of liver cancer susceptible gene MICA. Oncotarget 2014; 5:5581-5590. [PMID: 25026299 PMCID: PMC4170619 DOI: 10.18632/oncotarget.2143] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 06/24/2014] [Indexed: 01/05/2023] Open
Abstract
Chronic hepatitis B virus (HBV) infection is a major cause of hepatocellular carcinoma (HCC). To date, the lack of efficient in vitro systems supporting HBV infection and replication has been a major limitation of HBV research. Although primary human hepatocytes support the complete HBV life cycle, their limited availability and difficulties with gene transduction remain problematic. Here, we used human primary hepatocytes isolated from humanized chimeric uPA/SCID mice as efficient sources. These hepatocytes supported HBV replication in vitro. Based on analyses of mRNA and microRNA (miRNA) expression levels in HBV-infected hepatocytes, miRNA93 was significantly downregulated during HBV infection. MiRNA93 is critical for regulating the expression levels of MICA protein, which is a determinant for HBV-induced HCC susceptibility. Exogenous addition of miRNA93 in HBV-infected hepatocytes using bionanocapsules consisted of HBV envelope L proteins restored MICA protein expression levels in the supernatant. These results suggest that the rescued suppression of soluble MICA protein levels by miRNA93 targeted to HBV-infected hepatocytes using bionanocapsules may be useful for the prevention of HBV-induced HCC by altering deregulated miRNA93 expression.
Collapse
Affiliation(s)
- Motoko Ohno
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Japan Science and Technology Agency, PRESTO, Kawaguchi, Saitama, Japan
| | - Takahiro Kishikawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Chikako Shibata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Yoshikawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akemi Takata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryosuke Muroyama
- Unit of Disease Control Genome Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Norie Kowatari
- Unit of Disease Control Genome Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaya Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoya Kato
- Unit of Disease Control Genome Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shun'ichi Kuroda
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
197
|
Kanda T, Jiang X, Yokosuka O. Androgen receptor signaling in hepatocellular carcinoma and pancreatic cancers. World J Gastroenterol 2014; 20:9229-9236. [PMID: 25071315 PMCID: PMC4110552 DOI: 10.3748/wjg.v20.i28.9229] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/07/2014] [Accepted: 04/30/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) and pancreatic cancer remain difficult to treat, and despite the ongoing development of new treatments, the overall survival rate has only modestly improved over the past decade. Liver and pancreatic progenitors commonly develop from endoderm cells in the embryonic foregut. A previous study showed that HCC and pancreatic cancer cell lines variably express androgen receptor (AR), and these cancers and the surrounding tissues also express AR. AR is a ligand-dependent transcription factor that belongs to the nuclear receptor superfamily. Androgen response element is present in regulatory elements on the AR-responsive target genes, such as transforming growth factor beta-1 (TGF beta-1) and vascular endothelial growth factor (VEGF). It is well known that the activation of AR is associated with human carcinogenesis in prostate cancer as well as HCC and pancreatic cancer and that GRP78, TGF beta, and VEGF all play important roles in carcinogenesis and cancer development in these cancers. HCC is a male-dominant cancer irrespective of its etiology. Previous work has reported that vertebrae forkhead box A 1/2 are involved in estrogen receptors and/or AR signaling pathways, which may contribute to the gender differences observed with HCC. Our recent work also showed that AR has a critical role in pancreatic cancer development, despite pancreatic cancer not being a male dominant cancer. Aryl hydrocarbon (or dioxin) receptor is also involved in both HCC and pancreatic cancer through the formation of complex with AR. It is possible that AR might be involved in their carcinogenesis through major histocompatibility complex class I chain-related gene A/B. This review article describes AR and its role in HCC and pancreatic cancer and suggests that more specific AR signaling-inhibitors may be useful in the treatment of these "difficult to treat" cancers.
Collapse
|
198
|
Senapati S, Gutierrez-Achury J, Sood A, Midha V, Szperl A, Romanos J, Zhernakova A, Franke L, Alonso S, Thelma BK, Wijmenga C, Trynka G. Evaluation of European coeliac disease risk variants in a north Indian population. Eur J Hum Genet 2014; 23:530-5. [PMID: 25052311 PMCID: PMC4666579 DOI: 10.1038/ejhg.2014.137] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 06/10/2014] [Accepted: 06/18/2014] [Indexed: 01/06/2023] Open
Abstract
Studies in European populations have contributed to a better understanding of the genetics of complex diseases, for example, in coeliac disease (CeD), studies of over 23 000 European samples have reported association to the HLA locus and another 39 loci. However, these associations have not been evaluated in detail in other ethnicities. We sought to better understand how disease-associated loci that have been mapped in Europeans translate to a disease risk for a population with a different ethnic background. We therefore performed a validation of European risk loci for CeD in 497 cases and 736 controls of north Indian origin. Using a dense-genotyping platform (Immunochip), we confirmed the strong association to the HLA region (rs2854275, P=8.2 × 10−49). Three loci showed suggestive association (rs4948256, P=9.3 × 10−7, rs4758538, P=8.6 × 10−5 and rs17080877, P=2.7 × 10−5). We directly replicated five previously reported European variants (P<0.05; mapping to loci harbouring FASLG/TNFSF18, SCHIP1/IL12A, PFKFB3/PRKCQ, ZMIZ1 and ICOSLG). Using a transferability test, we further confirmed association at PFKFB3/PRKCQ (rs2387397, P=2.8 × 10−4) and PTPRK/THEMIS (rs55743914, P=3.4 × 10−4). The north Indian population has a higher degree of consanguinity than Europeans and we therefore explored the role of recessively acting variants, which replicated the HLA locus (rs9271850, P=3.7 × 10−23) and suggested a role of additional four loci. To our knowledge, this is the first replication study of CeD variants in a non-European population.
Collapse
Affiliation(s)
| | - Javier Gutierrez-Achury
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Ajit Sood
- Department of Gastroenterology, Dayanand Medical College and Hospital, Ludhiana, India
| | - Vandana Midha
- Department of Medicine, Dayanand Medical College and Hospital, Ludhiana, India
| | - Agata Szperl
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Jihane Romanos
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Santos Alonso
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, Leioa, Spain
| | - B K Thelma
- Department of Genetics, University of Delhi South Campus, New Delhi, India
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| | - Gosia Trynka
- Department of Genetics, University of Groningen, University Medical Hospital Groningen, Groningen, The Netherlands
| |
Collapse
|
199
|
Elfn1 recruits presynaptic mGluR7 in trans and its loss results in seizures. Nat Commun 2014; 5:4501. [PMID: 25047565 DOI: 10.1038/ncomms5501] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 06/23/2014] [Indexed: 01/08/2023] Open
Abstract
GABAergic interneurons are highly heterogeneous, and much is unknown about the specification and functional roles of their neural circuits. Here we show that a transinteraction of Elfn1 and mGluR7 controls targeted interneuron synapse development and that loss of Elfn1 results in hyperactivity and sensory-triggered epileptic seizures in mice. Elfn1 protein increases during postnatal development and localizes to postsynaptic sites of somatostatin-containing interneurons (SOM-INs) in the hippocampal CA1 stratum oriens and dentate gyrus (DG) hilus. Elfn1 knockout (KO) mice have deficits in mGluR7 recruitment to synaptic sites on SOM-INs, and presynaptic plasticity is impaired at these synapses. In patients with epilepsy and attention deficit hyperactivity disorder (ADHD), we find damaging missense mutations of ELFN1 that are clustered in the carboxy-terminal region required for mGluR7 recruitment. These results reveal a novel mechanism for interneuron subtype-specific neural circuit establishment and define a common basis bridging neurological disorders.
Collapse
|
200
|
Zhang X, Jia J, Dong J, Yu F, Ma N, Li M, Liu X, Liu W, Li T, Liu D. HLA-DQ polymorphisms with HBV infection: different outcomes upon infection and prognosis to lamivudine therapy. J Viral Hepat 2014; 21:491-8. [PMID: 24750255 DOI: 10.1111/jvh.12159] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Two recent genome-wide studies showed that the single-nucleotide polymorphisms in the HLA-DQ region (rs2856718 and rs9275572) were associated with chronic hepatitis B virus infection and chronic hepatitis C virus-associated hepatocellular carcinoma in Japanese patients. We tested the effects of the two single-nucleotide polymorphisms for all major HBV outcomes and lamivudine treatment in Han Chinese. A total of 1649 samples were enrolled, and peripheral blood samples were collected in this study. The single-nucleotide polymorphisms in the HLA-DQ region were genotyped using matrix-assisted laser desorption/ionization time of flight mass spectrometry. Our study demonstrated the clear relevance of HLA-DQ rs2856718 and rs9275572 with HBV susceptibility, natural clearance and HBV-associated HCC. HLA-DQ rs2856718G and rs9275572A were strongly associated with decreased risk of chronic HBV infection (odds ratio = 0.641; P = 2.64 × 10(-4) ; odds ratio = 0.627, P = 7.22 × 10(-5) ) and HBV natural clearance (odds ratio = 0.610; P = 4.80 × 10(-4) ; odds ratio = 0.714, P = 0.013). Moreover, rs9275572A was also associated with development of cirrhosis and hepatocellular carcinoma (odds ratio = 0.632, P = 0.008). In addition, we showed for the first time to our knowledge that rs9275572 was a predictor for lamivudine therapy (viral response: odds ratio = 2.599, P = 4.43 × 10(-4) ; biochemical response: odds ratio = 2.279, P = 4.23 × 10(-4) ). Our study suggested that HLA-DQ loci were associated with both HBV clearance and HBV-related diseases and outcomes of lamivudine treatment in Han Chinese.
Collapse
Affiliation(s)
- X Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shi Jiazhuang, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|