601
|
Guzzardi MA, Iozzo P, Salonen MK, Kajantie E, Airaksinen R, Kiviranta H, Rantakokko P, Eriksson JG. Exposure to Persistent Organic Pollutants Predicts Telomere Length in Older Age: Results from the Helsinki Birth Cohort Study. Aging Dis 2016; 7:540-552. [PMID: 27699078 PMCID: PMC5036950 DOI: 10.14336/ad.2016.0209] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 02/09/2016] [Indexed: 12/14/2022] Open
Abstract
As the population ages, the occurrence of chronic pathologies becomes more common. Leukocyte telomere shortening associates to ageing and age-related diseases. Recent studies suggest that environmental chemicals can affect telomere length. Persistent organic pollutants (POPs) are most relevant, since they are ingested with foods, and accumulate in the body for a long time. This longitudinal study was undertaken to test if circulating POPs predict telomere length and shortening in elderly people. We studied 1082 subjects belonging to the Helsinki Birth Cohort Study (born 1934-1944), undergoing two visits (2001-2004 and 2011-2014). POPs (oxychlordane, trans-nonachlor, p, p’-DDE, PCB 153, BDE 47, BDE 153) were analysed at baseline. Relative telomere length was measured twice, ’10 years apart, by quantitative real-time PCR. Oxychlordane, trans-nonachlor and PCB-153 levels were significant predictors of telomere length and shortening. In men, we did not find a linear relationship between POPs exposure and telomere shortening. In women, a significant reduction across quartiles categories of oxychlordane and trans-nonachlor exposure was observed. Baseline characteristics of subjects in the highest POPs categories included higher levels of C-reactive protein and fasting glucose, and lower body fat percentage. This is one of few studies combining POPs and telomere length. Our results indicate that exposure to oxychlordane, trans-nonachlor and PCB 153 predicts telomere attrition. This finding is important because concentrations of POPs observed here occur in contemporary younger people, and may contribute to an accelerated ageing.
Collapse
Affiliation(s)
| | - Patricia Iozzo
- 1Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
| | - Minna K Salonen
- 2National Institute for Health and Welfare, Chronic Disease Prevention Unit, Helsinki, Finland; 6Folkhälsan Research Centre, Helsinki, Helsingfors Universitet, Helsinki, Finland
| | - Eero Kajantie
- 2National Institute for Health and Welfare, Chronic Disease Prevention Unit, Helsinki, Finland; 3Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; 4PEDEGO Research Group, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Riikka Airaksinen
- 8National Institute for Health and Welfare, Department of Health Protection, Chemicals and Health Unit, Finland
| | - Hannu Kiviranta
- 8National Institute for Health and Welfare, Department of Health Protection, Chemicals and Health Unit, Finland
| | - Panu Rantakokko
- 8National Institute for Health and Welfare, Department of Health Protection, Chemicals and Health Unit, Finland
| | - Johan Gunnar Eriksson
- 2National Institute for Health and Welfare, Chronic Disease Prevention Unit, Helsinki, Finland; 5Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland; 6Folkhälsan Research Centre, Helsinki, Helsingfors Universitet, Helsinki, Finland; 7Unit of General Practice, Helsinki University Hospital, Finland
| |
Collapse
|
602
|
Salameh H, Hanayneh MA, Masadeh M, Naseemuddin M, Matin T, Erwin A, Singal AK. PNPLA3 as a Genetic Determinant of Risk for and Severity of Non-alcoholic Fatty Liver Disease Spectrum. J Clin Transl Hepatol 2016; 4:175-191. [PMID: 27777887 PMCID: PMC5075002 DOI: 10.14218/jcth.2016.00009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/10/2016] [Accepted: 05/23/2016] [Indexed: 12/15/2022] Open
Abstract
Background and Aims:Patatin-like phospholipase domain protein 3 (PNPLA3) polymorphisms (rs738409 C>G) are associated with non-alcoholic fatty liver disease (NAFLD). We performed a systematic review and meta-analysis to examine the association of PNPLA3 polymorphisms with the spectrum and severity of this disease. Methods: Studies evaluating the association between the PNPLA3 polymorphism spectrum (fatty liver, steatohepatitis, cirrhosis, and hepatocellular carcinoma) and NAFLD were included. Pooled data are reported as odds ratios (ORs) with 95% confidence intervals. Results: Of 393 potentially relevant studies, 35 on NAFLD were included in the analysis. Compared to healthy controls, the pooled ORs for rs738409 CG and GG compared to CC among patients with non-alcoholic fatty liver (NAFL) were 1.46 (1.16-1.85) and 2.76 (2.30-3.13), and were 1.75 (1.24-2.46) and 4.44 (2.92-6.76) among patients with non-alcoholic steatohepatitis respectively. The respective ORs for CG and GG compared to the CC genotype were 2.35 (0.90-6.13) and 5.05 (1.47-17.29) when comparing non-alcoholic hepatocellular carcinoma to NAFL patients. Among the NAFLD patients, the ORs for G allele frequency when comparing steatosis grade 2-3 to grade 0-1 NAFL, when comparing the NAFLD activity score of ≥ 4 to score ≤ 3, when comparing NASH to NAFLD, when comparing the presence of lobular inflammation to absence, and when comparing the presence of hepatocyte ballooning to absence were 2.33 (1.43-3.80), 1.80 (1.36-2.37), 1.66 (1.42-1.94), 1.58 (1.19-2.10), and 2.63 (1.87-3.69) respectively. Subgroup analysis based on ethnicity showed similar results. Conclusions:PNPLA3 polymorphisms have strong association with the risk for and severity of NAFLDs. PNPLA3 polymorphism plays an evolving role in diagnosis and treatment decisions in patients with NAFLD.
Collapse
Affiliation(s)
- Habeeb Salameh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Muhannad Al Hanayneh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Maen Masadeh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Tasnia Matin
- Department of Internal Medicine, University of Alabama, Birmingham, AL, USA
| | - Angelika Erwin
- Genomic Medicine Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Ashwani K. Singal
- Division of Gastroenterology and Hepatology, University of Alabama, Birmingham, AL, USA
- *Correspondence to: Ashwani Kumar Singal, Division of Gastroenterology and Hepatology, University of Alabama, 1808 7 Ave S BDB 351, Birmingham, AL 35294-0012, USA. Tel: +1-205-934-5623, Fax: +1-205-975-0961, E-mail:
| |
Collapse
|
603
|
Laloglu E, Aksoy H, Aksoy Y, Ozkaya F, Akcay F. The determination of serum and urinary endocan concentrations in patients with bladder cancer. Ann Clin Biochem 2016; 53:647-653. [PMID: 26748103 DOI: 10.1177/0004563216629169] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background Endocan (endothelial cell-specific molecule-1) is a proteoglycan and plays an important role in angiogenesis and inflammation. The aim of this study was to evaluate of serum and urinary concentrations of endothelial cell-specific molecule-1 in bladder cancer. Methods The study included 50 bladder cancer patients, 50 with urinary tract infection and 51 healthy volunteers. Serum and urinary endothelial cell-specific molecule-1 concentrations were measured with enzyme linked immunosorbent assay. Results In bladder cancer group, serum and urinary endothelial cell-specific molecule-1 concentrations were significantly higher than in the healthy subjects ( P = 0.003 and P < 0.0001). Urinary endothelial cell-specific molecule-1 concentrations in cases with urinary tract infection were higher than in healthy volunteers ( P = 0.002). There were no significant differences between bladder cancer and urinary tract infection groups in terms of serum and urinary endothelial cell-specific molecule-1 concentrations. Urinary endothelial cell-specific molecule-1 concentrations were higher than those of corresponding serum endothelial cell-specific molecule-1 concentrations ( P < 0.0001 for bladder cancer and urinary tract infection groups, P = 0.002 for healthy subjects). In bladder cancer group, there was a positive correlation between serum endothelial cell-specific molecule-1 and urinary endothelial cell-specific molecule-1 concentrations ( r = 0.32, P = 0.002). For serum endothelial cell-specific molecule-1, sensitivity and specificity were 50%, and 77%, and for urinary endothelial cell-specific molecule-1, 62%, and 71%, respectively. Conclusion Serum and urinary endothelial cell-specific molecule-1 concentrations increase in bladder cancer. This parameter also increases in serum and urine of cases with urinary tract infection. That urinary endothelial cell-specific molecule-1 values were higher than serum endothelial cell-specific molecule-1 values in all groups may be attributed to direct exfoliation of epithelial cells in bladder to urine.
Collapse
Affiliation(s)
- Esra Laloglu
- 1 Department of Medical Biochemistry, Medical School of Ataturk University, Erzurum, Turkey
| | - Hulya Aksoy
- 1 Department of Medical Biochemistry, Medical School of Ataturk University, Erzurum, Turkey
| | - Yılmaz Aksoy
- 2 Department of Urology, Medical School of Ataturk University, Erzurum, Turkey
| | - Fatih Ozkaya
- 2 Department of Urology, Medical School of Ataturk University, Erzurum, Turkey
| | - Fatih Akcay
- 1 Department of Medical Biochemistry, Medical School of Ataturk University, Erzurum, Turkey
| |
Collapse
|
604
|
Dioxin-like rather than non-dioxin-like PCBs promote the development of endometriosis through stimulation of endocrine–inflammation interactions. Arch Toxicol 2016; 91:1915-1924. [DOI: 10.1007/s00204-016-1854-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 09/15/2016] [Indexed: 02/06/2023]
|
605
|
Cheikh Rouhou M, Karelis A, St-Pierre D, Lamontagne L. Adverse effects of weight loss: Are persistent organic pollutants a potential culprit? DIABETES & METABOLISM 2016; 42:215-23. [DOI: 10.1016/j.diabet.2016.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/09/2016] [Accepted: 05/24/2016] [Indexed: 02/05/2023]
|
606
|
Xia MF, Yki-Järvinen H, Bian H, Lin HD, Yan HM, Chang XX, Zhou Y, Gao X. Influence of Ethnicity on the Accuracy of Non-Invasive Scores Predicting Non-Alcoholic Fatty Liver Disease. PLoS One 2016; 11:e0160526. [PMID: 27579785 PMCID: PMC5007035 DOI: 10.1371/journal.pone.0160526] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Presence of non-alcoholic fatty liver disease (NAFLD) can predict risks for diabetes, cardiovascular disease and advanced liver disease in the general population. We aimed to establish a non-invasive score for prediction of NAFLD in Han Chinese, the largest ethnic group in the world, and detect whether ethnicity influences the accuracy of such a score. METHODS Liver fat content (LFAT) was measured by quantitative ultrasound in 3548 subjects in the Shanghai Changfeng Community and a Chinese score was created using multivariate logistic regression analyses. This new score was internally validated in Chinese and externally in Finns. Its diagnostic performance was compared to the NAFLD liver fat score, fatty liver index (FLI) and hepatic steatosis index (HSI) developed in Finns, Italians and Koreans. We also analyzed how obesity related to LFAT measured by 1H-MRS in 79 Finns and 118 Chinese with type 2 diabetes (T2D). RESULTS The metabolic syndrome and T2D, fasting serum insulin, body mass index (BMI) and AST/ALT ratio were independent predictors of NAFLD in Chinese. The AUROC in the Chinese validation cohort was 0.76 (0.73-0.78) and in Finns 0.73 (0.68-0.78) (p<0.0001). 43%, 27%, 32% and 42% of Chinese had NAFLD when determined by the Chinese score, NAFLD liver fat score (p<0.001 vs. Chinese score), FLI (p<0.001) and HSI (NS). For any given BMI and waist circumference, the Chinese had a markedly higher LFAT than the Finns. CONCLUSION The predictors of NAFLD in Han Chinese are as in Europids but the Chinese have more LFAT for any given degree of obesity than Europids. Ethnicity needs to be considered when NAFLD is predicted using risk scores.
Collapse
Affiliation(s)
- Ming-Feng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Chronic Metabolic Diseases, Fudan Unversity, Shanghai, China
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Hua Bian
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Chronic Metabolic Diseases, Fudan Unversity, Shanghai, China
| | - Huan-Dong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Chronic Metabolic Diseases, Fudan Unversity, Shanghai, China
| | - Hong-Mei Yan
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Chronic Metabolic Diseases, Fudan Unversity, Shanghai, China
| | - Xin-Xia Chang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Chronic Metabolic Diseases, Fudan Unversity, Shanghai, China
| | - You Zhou
- Department of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Chronic Metabolic Diseases, Fudan Unversity, Shanghai, China
| |
Collapse
|
607
|
Abstract
Malnutrition is a frequent feature in Crohn's disease (CD), affects patient outcome and must be recognised. For chronic inflammatory diseases, recent guidelines recommend the development of combined malnutrition and inflammation risk scores. We aimed to design and evaluate a new screening tool that combines both malnutrition and inflammation parameters that might help predict clinical outcome. In a prospective cohort study, we examined fifty-five patients with CD in remission (Crohn's disease activity index (CDAI) <200) at 0 and 6 months. We assessed disease activity (CDAI, Harvey-Bradshaw index), inflammation (C-reactive protein (CRP), faecal calprotectin (FC)), malnutrition (BMI, subjective global assessment (SGA), serum albumin, handgrip strength), body composition (bioelectrical impedance analysis) and administered the newly developed 'Malnutrition Inflammation Risk Tool' (MIRT; containing BMI, unintentional weight loss over 3 months and CRP). All parameters were evaluated regarding their ability to predict disease outcome prospectively at 6 months. At baseline, more than one-third of patients showed elevated inflammatory markers despite clinical remission (36·4 % CRP ≥5 mg/l, 41·5 % FC ≥100 µg/g). Prevalence of malnutrition at baseline according to BMI, SGA and serum albumin was 2-16 %. At 6 months, MIRT significantly predicted outcome in numerous nutritional and clinical parameters (SGA, CD-related flares, hospitalisations and surgeries). In contrast, SGA, handgrip strength, BMI, albumin and body composition had no influence on the clinical course. The newly developed MIRT was found to reliably predict clinical outcome in CD patients. This screening tool might be used to facilitate clinical decision making, including treatment of both inflammation and malnutrition in order to prevent complications.
Collapse
|
608
|
Jaeger C, Tischkau SA. Role of Aryl Hydrocarbon Receptor in Circadian Clock Disruption and Metabolic Dysfunction. ENVIRONMENTAL HEALTH INSIGHTS 2016; 10:133-141. [PMID: 27559298 PMCID: PMC4990151 DOI: 10.4137/ehi.s38343] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 06/01/2023]
Abstract
The prevalence of metabolic syndrome, a clustering of three or more risk factors that include abdominal obesity, increased blood pressure, and high levels of glucose, triglycerides, and high-density lipoproteins, has reached dangerous and costly levels worldwide. Increases in morbidity and mortality result from a combination of factors that promote altered glucose metabolism, insulin resistance, and metabolic dysfunction. Although diet and exercise are commonly touted as important determinants in the development of metabolic dysfunction, other environmental factors, including circadian clock disruption and activation of the aryl hydrocarbon receptor (AhR) by dietary or other environmental sources, must also be considered. AhR binds a range of ligands, which prompts protein-protein interactions with other Per-Arnt-Sim (PAS)-domain-containing proteins and subsequent transcriptional activity. This review focuses on the reciprocal crosstalk between the activated AhR and the molecular circadian clock. AhR exhibits a rhythmic expression and time-dependent sensitivity to activation by AhR agonists. Conversely, AhR activation influences the amplitude and phase of expression of circadian clock genes, hormones, and the behavioral responses of the clock system to changes in environmental illumination. Both the clock and AhR status and activation play significant and underappreciated roles in metabolic homeostasis. This review highlights the state of knowledge regarding how AhR may act together with the circadian clock to influence energy metabolism. Understanding the variety of AhR-dependent mechanisms, including its interactions with the circadian timing system that promote metabolic dysfunction, reveals new targets of interest for maintenance of healthy metabolism.
Collapse
|
609
|
Jary M, Lecomte T, Bouché O, Kim S, Dobi E, Queiroz L, Ghiringhelli F, Etienne H, Léger J, Godet Y, Balland J, Lakkis Z, Adotevi O, Bonnetain F, Borg C, Vernerey D. Prognostic value of baseline seric Syndecan-1 in initially unresectable metastatic colorectal cancer patients: a simple biological score. Int J Cancer 2016; 139:2325-35. [PMID: 27472156 DOI: 10.1002/ijc.30367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 06/24/2016] [Accepted: 07/01/2016] [Indexed: 01/17/2023]
Abstract
In first-line metastatic colorectal cancer (mCRC), baseline prognostic factors allowing death risk and treatment strategy stratification are lacking. Syndecan-1 (CD138) soluble form was never described as a prognostic biomarker in mCRC. We investigated its additional prognostic value for overall survival (OS). mCRC patients with unresectable disease at diagnosis were treated with bevacizumab-based chemotherapy in two independent prospective clinical trials (development set: n = 126, validation set: n = 51, study NCT00489697 and study NCT00544011, respectively). Serums were collected at baseline for CD138 measurement. OS determinants were assessed and, based on the final multivariate model, a prognostic score was proposed. Two independent OS prognostic factors were identified: Lactate Dehydrogenase (LDH) high level (p = 0.0066) and log-CD138 high level (p = 0.0190). The determination of CD138 binary information (cutoff: 75 ng/mL) allowed the assessment of a biological prognostic score with CD138 and LDH values, identifying three risk groups for death (median OS= 38.9, 30.1 and 19.8 months for the low, intermediate and high risk groups, respectively; p < 0.0001). This score had a good discrimination ability (C-index = 0.63). These results were externally confirmed in the validation set. Our study provides robust evidence in favor of the additional baseline soluble CD138 prognostic value for OS, in mCRC patients. A simple biological scoring system is proposed including LDH and CD138 binary status values.
Collapse
Affiliation(s)
- Marine Jary
- Department of Medical Oncology, University Hospital, Besançon, France. .,INSERM, Unit 1098, University of Bourgogne- Franche Comté, Besançon, France. .,Clinical Investigation Center 1431, EFS Bourgogne-Franche Comté, Besançon, France.
| | - Thierry Lecomte
- CNRS, Unit 7292, University François-Rabelais, Tours, France.,Department of HepatoGastroenterology and Digestive Oncology, University Hospital, Tours, France
| | - Olivier Bouché
- Department of HepatoGastroenterology and Digestive Oncology, University Hospital Robert Debré, Reims, France
| | - Stefano Kim
- Department of Medical Oncology, University Hospital, Besançon, France.,Clinical Investigation Center 1431, EFS Bourgogne-Franche Comté, Besançon, France
| | - Erion Dobi
- Department of Medical Oncology, University Hospital, Besançon, France
| | - Lise Queiroz
- INSERM, Unit 1098, University of Bourgogne- Franche Comté, Besançon, France
| | | | - Hélène Etienne
- Department of Medical Oncology, University Hospital, Besançon, France.,Department of Gastroenterology, University Hospital, Besançon, France
| | - Julie Léger
- INSERM, Clinical Investigational Center CIC 1415, Tours, France
| | - Yann Godet
- INSERM, Unit 1098, University of Bourgogne- Franche Comté, Besançon, France
| | - Jérémy Balland
- INSERM, Unit 1098, University of Bourgogne- Franche Comté, Besançon, France
| | - Zaher Lakkis
- Department of Digestive Surgery and Liver Transplantation, University Hospital, Besançon, France
| | - Olivier Adotevi
- Department of Medical Oncology, University Hospital, Besançon, France.,INSERM, Unit 1098, University of Bourgogne- Franche Comté, Besançon, France
| | - Franck Bonnetain
- INSERM, Unit 1098, University of Bourgogne- Franche Comté, Besançon, France.,Clinical Investigation Center 1431, EFS Bourgogne-Franche Comté, Besançon, France.,Methodological and Quality of Life in Oncology Unit, EA 3181, University Hospital, Besançon, France
| | - Christophe Borg
- Department of Medical Oncology, University Hospital, Besançon, France.,INSERM, Unit 1098, University of Bourgogne- Franche Comté, Besançon, France.,Clinical Investigation Center 1431, EFS Bourgogne-Franche Comté, Besançon, France
| | - Dewi Vernerey
- INSERM, Unit 1098, University of Bourgogne- Franche Comté, Besançon, France.,Clinical Investigation Center 1431, EFS Bourgogne-Franche Comté, Besançon, France.,Methodological and Quality of Life in Oncology Unit, EA 3181, University Hospital, Besançon, France
| |
Collapse
|
610
|
Severson TJ, Besur S, Bonkovsky HL. Genetic factors that affect nonalcoholic fatty liver disease: A systematic clinical review. World J Gastroenterol 2016; 22:6742-6756. [PMID: 27547017 PMCID: PMC4970479 DOI: 10.3748/wjg.v22.i29.6742] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 03/28/2016] [Accepted: 05/23/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate roles of genetic polymorphisms in non-alcoholic fatty liver disease (NAFLD) onset, severity, and outcome through systematic literature review.
METHODS: The authors conducted both systematic and specific searches of PubMed through December 2015 with special emphasis on more recent data (from 2012 onward) while still drawing from more historical data for background. We identified several specific genetic polymorphisms that have been most researched and, at this time, appear to have the greatest clinical significance on NAFLD and similar hepatic diseases. These were further investigated to assess their specific effects on disease onset and progression and the mechanisms by which these effects occur.
RESULTS: We focus particularly on genetic polymorphisms of the following genes: PNPLA3, particularly the p. I148M variant, TM6SF2, particularly the p. E167K variant, and on variants in FTO, LIPA, IFNλ4, and iron metabolism, specifically focusing on HFE, and HMOX-1. We discuss the effect of these genetic variations and their resultant protein variants on the onset of fatty liver disease and its severity, including the effect on likelihood of progression to cirrhosis and hepatocellular carcinoma. While our principal focus is on NAFLD, we also discuss briefly effects of some of the variants on development and severity of other hepatic diseases, including hepatitis C and alcoholic liver disease. These results are briefly discussed in terms of clinical application and future potential for personalized medicine.
CONCLUSION: Polymorphisms and genetic factors of several genes contribute to NAFLD and its end results. These genes hold keys to future improvements in diagnosis and management.
Collapse
|
611
|
Dongiovanni P, Valenti L. Genetics of nonalcoholic fatty liver disease. Metabolism 2016; 65:1026-37. [PMID: 26409295 DOI: 10.1016/j.metabol.2015.08.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/23/2015] [Accepted: 08/26/2015] [Indexed: 02/07/2023]
Abstract
UNLABELLED Epidemiological, familial, and twin studies indicate that non-alcoholic fatty liver disease, now the leading cause of liver damage in developed countries, has a strong heritability. The common I148M variant of PNPLA3 impairing hepatocellular lipid droplets remodeling is the major genetic determinant of hepatic fat content. The I148M variant has a strong impact on the full spectrum of liver damage related to fatty liver, encompassing non-alcoholic steatohepatitis, advanced fibrosis, and hepatocellular carcinoma, and influences the response to therapeutic approaches. Common variants in GCKR enhance de novo hepatic lipogenesis in response to glucose and liver inflammation. Furthermore, the low-frequency E167K variant of TM6SF2 and rare mutations in APOB, which impair very low-density lipoproteins secretion, predispose to progressive fatty liver. CONCLUSIONS These and other recent findings reviewed here indicate that impaired lipid handling by hepatocytes has a major role in the pathogenesis of non-alcoholic fatty liver disease by triggering inflammation, fibrogenesis, and carcinogenesis. These discoveries have provided potential novel biomarkers for clinical use and have revealed intriguing therapeutic targets.
Collapse
Affiliation(s)
- Paola Dongiovanni
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico
| | - Luca Valenti
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Department of Pathophysiology and Transplantation, Università degli Studi Milano, Milan, Italy.
| |
Collapse
|
612
|
Joshi K, Kohli A, Manch R, Gish R. Alcoholic Liver Disease: High Risk or Low Risk for Developing Hepatocellular Carcinoma? Clin Liver Dis 2016; 20:563-80. [PMID: 27373617 DOI: 10.1016/j.cld.2016.02.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In this review we critically assess the literature to evaluate the level of risk posed by alcohol as both a primary etiology of hepatocellular carcinoma (HCC) and as a cofactor in its development. Although there have been conflicting findings, based on the body of evidence to date, it appears that the linkage between compensated alcoholic liver disease-associated cirrhosis and HCC is best characterized as medium-high risk, with the risk increasing with age and with quantity and duration of alcohol consumption and is more pronounced in females. While abstinence is the most effective way to reduce HCC risk, its effect seems largely dependent on the severity of liver damage at the point of cessation. Alcohol clearly interacts with other etiologies and conditions including viral hepatitis B and C, hereditary hemochromatosis, diabetes, and obesity to increase the risk for developing HCC, either synergistically or additively. Continued progress in genetics, especially through mechanistic-based and genome-wide association studies may ultimately identify which single nucleotide polymorphisms are risk factors for the onset of alcoholic liver disease and its progression to HCC and lead to the development of targeted therapeutics which may help providers better manage at-risk patients.
Collapse
Affiliation(s)
- Kartik Joshi
- Division of Hepatology, St. Joseph's Hospital and Medical Center, Creighton University School of Medicine, 500 West Thomas Road, Suite 900, Phoenix, AZ 85013, USA
| | - Anita Kohli
- Division of Hepatology, St. Joseph's Hospital and Medical Center, Creighton University School of Medicine, 500 West Thomas Road, Suite 900, Phoenix, AZ 85013, USA; Division of Infectious Disease, St. Joseph's Hospital and Medical Center, Creighton University School of Medicine, 500 West Thomas Road, Suite 900, Phoenix, AZ 85013, USA
| | - Richard Manch
- Division of Hepatology, St. Joseph's Hospital and Medical Center, Creighton University School of Medicine, 500 West Thomas Road, Suite 900, Phoenix, AZ 85013, USA
| | - Robert Gish
- Division of Hepatology, St. Joseph's Hospital and Medical Center, Creighton University School of Medicine, 500 West Thomas Road, Suite 900, Phoenix, AZ 85013, USA; Division of Hepatology and Gastroenterology, Stanford University Hospitals and Clinics, 300 Pasteur Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|
613
|
Iida M, Fujii S, Uchida M, Nakamura H, Kagami Y, Agusa T, Hirano M, Bak SM, Kim EY, Iwata H. Identification of aryl hydrocarbon receptor signaling pathways altered in TCDD-treated red seabream embryos by transcriptome analysis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2016; 177:156-170. [PMID: 27288597 DOI: 10.1016/j.aquatox.2016.05.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 05/16/2016] [Accepted: 05/20/2016] [Indexed: 06/06/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces a broad spectrum of toxic effects including craniofacial malformation and neural damage in fish embryos. These effects are mainly mediated by the aryl hydrocarbon receptor (AHR). However, the mode of action between TCDD-induced AHR activation and adverse outcomes is not yet understood. To provide a comprehensive picture of the AHR signaling pathway in fish embryos exposed to TCDD, red seabream (Pagrus major) embryos were treated with graded concentrations of TCDD (0.3-37nM) in seawater, or with a mixture of TCDD and 500nM CH223191, an AHR-specific antagonist. The transcriptome of red seabream embryos was analyzed using a custom-made microarray with 6000 probes specifically prepared for this species. A Jonckheere-Terpstra test was performed to screen for genes that demonstrated altered mRNA expression levels following TCDD exposure. The signals of 1217 genes (as human homologs) were significantly altered in a TCDD concentration-dependent manner (q-value<0.2). Notably, the TCDD-induced alteration in mRNA expression was alleviated by co-exposure to CH223191, suggesting that the mRNA expression level of these genes was regulated by AHR. To identify TCDD-activated pathways, the microarray data were further subjected to gene set enrichment analysis (GSEA) and functional protein-protein interaction (PPI) network analysis. GSEA demonstrated that the effects of TCDD on sets of genes involved calcium, mitogen-activated protein kinase (MAPK), actin cytoskeleton, chemokine, T cell receptor, melanoma, vascular endothelial growth factor (VEGF), axon guidance, and renal cell carcinoma signaling pathways. These results suggest the hypotheses that TCDD induces immunosuppression via the calcium, MAPK, chemokine, and T cell receptor signaling pathways, neurotoxicity via VEGF signaling, and axon guidance alterations and teratogenicity via the dysregulation of the actin cytoskeleton and melanoma and renal cell carcinoma signaling pathways. Furthermore, the PPI network analysis indicated that the adverse outcome pathways of TCDD in the embryos might be propagated through several hub genes such as cell division control protein 42, phosphoinositide-3-kinase regulatory subunit 1, and guanine nucleotide-binding proteins. Understanding these pathways potentially allows for exploring the adverse outcome pathway of the effects of TCDD on the red seabream embryos.
Collapse
Affiliation(s)
- Midori Iida
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, 790-8577, Japan; Computer Science and Systems Engineering, Kyusyu Institute of Technology, Iizuka, 820-0067, Japan.
| | - Satoshi Fujii
- Computer Science and Systems Engineering, Kyusyu Institute of Technology, Iizuka, 820-0067, Japan.
| | | | | | | | - Tetsuro Agusa
- Graduate School of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, Kumamoto, 862-0920, Japan.
| | - Masashi Hirano
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, 790-8577, Japan.
| | - Su-Min Bak
- Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Seoul, 130-701, Korea.
| | - Eun-Young Kim
- Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Seoul, 130-701, Korea.
| | - Hisato Iwata
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama, 790-8577, Japan.
| |
Collapse
|
614
|
Abstract
Alcoholic liver disease includes a broad clinical-histological spectrum from simple steatosis, cirrhosis, acute alcoholic hepatitis with or without cirrhosis to hepatocellular carcinoma as a complication of cirrhosis. The pathogenesis of alcoholic liver disease can be conceptually divided into (1) ethanol-mediated liver injury, (2) inflammatory immune response to injury, (3) intestinal permeability and microbiome changes. Corticosteroids may improve outcomes, but this is controversial and probably only impacts short-term survival. New pathophysiology-based therapies are under study, including antibiotics, caspase inhibition, interleukin-22, anakinra, FXR agonist and others. These studies provide hope for better future outcomes for this difficult disease.
Collapse
Affiliation(s)
- Winston Dunn
- Gastroenterology & Hepatology, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS USA
| | - Vijay H. Shah
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
615
|
Abstract
Genome-wide association studies (GWAS) in the field of liver diseases have revealed previously unknown pathogenic loci and generated new biological hypotheses. In 2008, a GWAS performed in a population-based sample study, where hepatic liver fat content was measured by magnetic spectroscopy, showed a strong association between a variant (rs738409 C>G p.I148M) in the patatin-like phospholipase domain containing 3 (PNPLA3) gene and nonalcoholic fatty liver disease. Further replication studies have shown robust associations between PNPLA3 and steatosis, fibrosis/cirrhosis, and hepatocellular carcinoma on a background of metabolic, alcoholic, and viral insults. The PNPLA3 protein has lipase activity towards triglycerides in hepatocytes and retinyl esters in hepatic stellate cells. The I148M substitution leads to a loss of function promoting triglyceride accumulation in hepatocytes. Although PNPLA3 function has been extensively studied, the molecular mechanisms leading to hepatic fibrosis and carcinogenesis remain unclear. This unsuspected association has highlighted the fact that liver fat metabolism may have a major impact on the pathophysiology of liver diseases. Conversely, alone, this locus may have limited predictive value with regard to liver disease outcomes in clinical practice. Additional studies at the genome-wide level will be required to identify new variants associated with liver damage and cancer to explain a greater proportion of the heritability of these phenotypes. Thus, incorporating PNPLA3 and other genetic variants in combination with clinical data will allow for the development of tailored predictive models. This attractive approach should be evaluated in prospective cohorts.
Collapse
|
616
|
Chollet L, Saboural P, Chauvierre C, Villemin JN, Letourneur D, Chaubet F. Fucoidans in Nanomedicine. Mar Drugs 2016; 14:E145. [PMID: 27483292 PMCID: PMC4999906 DOI: 10.3390/md14080145] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 12/19/2022] Open
Abstract
Fucoidans are widespread cost-effective sulfated marine polysaccharides which have raised interest in the scientific community over last decades for their wide spectrum of bioactivities. Unsurprisingly, nanomedicine has grasped these compounds to develop innovative therapeutic and diagnostic nanosystems. The applications of fucoidans in nanomedicine as imaging agents, drug carriers or for their intrinsic properties are reviewed here after a short presentation of the main structural data and biological properties of fucoidans. The origin and the physicochemical specifications of fucoidans are summarized in order to discuss the strategy of fucoidan-containing nanosystems in Human health. Currently, there is a need for reproducible, well characterized fucoidan fractions to ensure significant progress.
Collapse
Affiliation(s)
- Lucas Chollet
- Inserm, U1148, LVTS, University Paris Diderot, X Bichat Hospital, F-75877 Paris, France.
- Galilée Institute, University Paris 13, Sorbonne Paris Cité, F-93430 Villetaneuse, France.
- Algues & Mer, Kernigou, F-29242 Ouessant, France.
| | - Pierre Saboural
- Inserm, U1148, LVTS, University Paris Diderot, X Bichat Hospital, F-75877 Paris, France.
- Galilée Institute, University Paris 13, Sorbonne Paris Cité, F-93430 Villetaneuse, France.
| | - Cédric Chauvierre
- Inserm, U1148, LVTS, University Paris Diderot, X Bichat Hospital, F-75877 Paris, France.
- Galilée Institute, University Paris 13, Sorbonne Paris Cité, F-93430 Villetaneuse, France.
| | | | - Didier Letourneur
- Inserm, U1148, LVTS, University Paris Diderot, X Bichat Hospital, F-75877 Paris, France.
- Galilée Institute, University Paris 13, Sorbonne Paris Cité, F-93430 Villetaneuse, France.
| | - Frédéric Chaubet
- Inserm, U1148, LVTS, University Paris Diderot, X Bichat Hospital, F-75877 Paris, France.
- Galilée Institute, University Paris 13, Sorbonne Paris Cité, F-93430 Villetaneuse, France.
| |
Collapse
|
617
|
Molecular mechanisms of hepatitis C virus-induced hepatocellular carcinoma. Clin Microbiol Infect 2016; 22:853-861. [PMID: 27476823 DOI: 10.1016/j.cmi.2016.07.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/09/2016] [Accepted: 07/16/2016] [Indexed: 02/06/2023]
Abstract
Hepatitis C virus (HCV) is a major leading cause of hepatocellular carcinoma (HCC). HCV-induced hepatocarcinogenesis is a multistep process resulting from a combination of pathway alterations that are either caused directly by viral factors or immune mediated as a consequence of a chronic state of inflammation. Host genetic variation is now emerging as an additional element that contribute to increase the risk of developing HCC. The advent of direct-acting antiviral agents foresees a rapid decline of HCC rate in HCV patients. However, a full understanding of the HCV-mediated tumourigenic process is required to elucidate if pro-oncogenic signatures may persist after virus clearance, and to identify novel tools for HCC prevention and therapy. In this review, we summarize the current knowledge of the molecular mechanisms responsible for HCV-induced hepatocarcinogenesis.
Collapse
|
618
|
Abraham JA, Golubnitschaja O. Time for paradigm change in management of hepatocellular carcinoma: is a personalized approach on the horizon? Per Med 2016; 13:455-467. [PMID: 29767598 DOI: 10.2217/pme-2016-0013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most frequent cancer form but the second leading cause of all cancer-related deaths. There are several reasons for high mortality in the HCC cohort: lack of effective screening programs and consequently late diagnosis, multifactorial origin with cumulative risk factors, complex carcinogenesis, tumor heterogeneity, unpredictable impacts of individual microenvironment on tumor development and progression, and, as the consequence, frequently untargeted therapy and cancer resistance toward currently applied treatment approaches. The currently applied 'treat and wait' approach is inappropriate in the overall HCC management. Urgent need in paradigm change toward predictive, preventive and personalized medicine is discussed in this review article. Innovative strategies for an advanced predictive, preventive and personalized medicine approach in the overall HCC management benefiting the patient are presented.
Collapse
Affiliation(s)
- Jella-Andrea Abraham
- Department of Radiology, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Olga Golubnitschaja
- Department of Radiology, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| |
Collapse
|
619
|
Abella V, Pérez T, Scotece M, Conde J, Pirozzi C, Pino J, Lago F, González-Gay MÁ, Mera A, Gómez R, Gualillo O. Pollutants make rheumatic diseases worse: Facts on polychlorinated biphenyls (PCBs) exposure and rheumatic diseases. Life Sci 2016; 157:140-144. [PMID: 27312420 DOI: 10.1016/j.lfs.2016.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Polychlorinated biphenyls (PCBs) are persistent organic pollutants that bioaccumulate in adipose tissue, disturbing its metabolism and the balance of adipokines, related to obesity. The altering secretion pattern of adipokines from the adipose tissue and the increasing mechanical load in weight-bearing joints presented in obesity condition, are risk factors for osteoarthritis development. The most prevalent rheumatic diseases, osteoarthritis and rheumatoid arthritis, are chronic conditions that target the whole joints, leading to increasing disability and health care cost. The goal of this focused review is to summarize the current knowledge on the role of PCBs in osteoarthritis and rheumatoid arthritis pathogenesis. SEARCH STRATEGY A PubMed search was managed using keywords as "rheumatic diseases", "polychlorinated biphenyls", "obesity" and "endocrine disruption". MAIN RESULTS OF THE REVIEW The incidence of rheumatoid arthritis has been reported to be increased especially in urban areas in industrialized countries, emphasizing the importance of environment in the pathogenesis of rheumatic diseases. Analysis of two cohorts exposed to PCBs food contamination showed high incidence of arthritis. In addition, PCBs in serum correlated positively with the prevalence of self-reported arthritis. Few studies support the hypothesis that osteoarthritis development could be related to PCBs induction of chondrocytes apoptosis. CONCLUSION Evidences have emerged for a relationship between PCBs and development of several types of arthritis. Further research is encouraged to determine the correlation between PCBs exposure and the development of rheumatic diseases.
Collapse
Affiliation(s)
- Vanessa Abella
- SERGAS, Research Laboratory 9, NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Institute of Medical Research (IDIS), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain; Departamento de Bioloxía Celular e Molecular, Facultade de Ciencias, Universidade da Coruña (UDC), Campus de A Coruña, 15071 A Coruña, Spain
| | - Tamara Pérez
- SERGAS, Research Laboratory 9, NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Institute of Medical Research (IDIS), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Morena Scotece
- SERGAS, Research Laboratory 9, NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Institute of Medical Research (IDIS), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Javier Conde
- SERGAS, Research Laboratory 9, NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Institute of Medical Research (IDIS), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Claudio Pirozzi
- SERGAS, Research Laboratory 9, NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Institute of Medical Research (IDIS), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Jesús Pino
- SERGAS, Research Laboratory 9, NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Institute of Medical Research (IDIS), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Francisca Lago
- SERGAS, Research Laboratory 7, Cellular and Molecular Cardiology Laboratory, Institute of Medical Research (IDIS), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Miguel Ángel González-Gay
- SERGAS, Division of Rheumatology, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Antonio Mera
- SERGAS, Division of Rheumatology, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Rodolfo Gómez
- SERGAS, Research Laboratory 9, NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Institute of Medical Research (IDIS), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Oreste Gualillo
- SERGAS, Research Laboratory 9, NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Institute of Medical Research (IDIS), Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain.
| |
Collapse
|
620
|
Whole Genome Pathway Analysis Identifies an Association of Cadmium Response Gene Loss with Copy Number Variation in Mutant p53 Bearing Uterine Endometrial Carcinomas. PLoS One 2016; 11:e0159114. [PMID: 27391266 PMCID: PMC4938382 DOI: 10.1371/journal.pone.0159114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/27/2016] [Indexed: 12/02/2022] Open
Abstract
Background Massive chromosomal aberrations are a signature of advanced cancer, although the factors promoting the pervasive incidence of these copy number alterations (CNAs) are poorly understood. Gatekeeper mutations, such as p53, contribute to aneuploidy, yet p53 mutant tumors do not always display CNAs. Uterine Corpus Endometrial Carcinoma (UCEC) offers a unique system to begin to evaluate why some cancers acquire high CNAs while others evolve another route to oncogenesis, since about half of p53 mutant UCEC tumors have a relatively flat CNA landscape and half have 20–90% of their genome altered in copy number. Methods We extracted copy number information from 68 UCEC genomes mutant in p53 by the GISTIC2 algorithm. GO term pathway analysis, via GOrilla, was used to identify suppressed pathways. Genes within these pathways were mapped for focal or wide distribution. Deletion hotspots were evaluated for temporal incidence. Results Multiple pathways contributed to the development of pervasive CNAs, including developmental, metabolic, immunological, cell adhesion and cadmium response pathways. Surprisingly, cadmium response pathway genes are predicted as the earliest loss events within these tumors: in particular, the metallothionein genes involved in heavy metal sequestration. Loss of cadmium response genes were associated with copy number changes and poorer prognosis, contrasting with 'copy number flat' tumors which instead exhibited substantive mutation. Conclusion Metallothioneins are lost early in the development of high CNA endometrial cancer, providing a potential mechanism and biological rationale for increased incidence of endometrial cancer with cadmium exposure. Developmental and metabolic pathways are altered later in tumor progression.
Collapse
|
621
|
Zeng F, Sherry JP, Bols NC. Evaluating the toxic potential of benzothiazoles with the rainbow trout cell lines, RTgill-W1 and RTL-W1. CHEMOSPHERE 2016; 155:308-318. [PMID: 27131451 DOI: 10.1016/j.chemosphere.2016.04.079] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/07/2016] [Accepted: 04/20/2016] [Indexed: 06/05/2023]
Abstract
Benzothiazole (BTHs) are environmental contaminants of emerging concern for which little toxicological information is available. Therefore the toxic potential of twelve BTHs was evaluated with two rainbow trout epithelial cell lines, RTgill-W1 and RTL-W1. The BTHs were benzothiazole (BTH), 3,3'-diethylthia dicarbocyanine iodide (DTDC), C.I. sulphur orange 1 (SO), 2-mercaptobenzothiazole (2MBTH), zinc 2-mercaptobenzothiazole (ZnMBTH), sodium 2-mercaptobenzothiazole (NaMBTH), 2-hydroxy-benzothiazole (OHBTH), 2- aminobenzothiazole (2ABTH), C.I. vat yellow 2 (VY), N,N-dicyclohexyl-2-benzothiazolsulfene amide (NNA), 2,2'-dithiobis (benzothiazole) (DBTH) and 2-(p-aminophenyl)-6-methylbenzothiazole-7-sulfonic acid (MBTHS). All BTHs, except for NNA, DBTH, and MBTHS, caused both cytotoxicity and a transitory elevation in reactive oxygen species (ROS) levels. Yet, neither N-acetyl cysteine (NAC) nor IM-54 inhibited cytotoxicity, suggesting that ROS imbalance did not contribute to cell death. Cell death was not blocked by Necrostatin-1 nor accompanied by DNA laddering, suggesting that neither necroptosis nor apoptosis took place. The comet assay revealed DNA strand breaks after exposures to 2ABTH and OHBTH for 1 day and to BTH for 12 days. In RTL-W1, cytochrome P4501A was induced noticeably by 2ABTH, OHBTH, and MBTHS and weakly by NaMBTH, ZnMBTH, SO, VY, and NNA, suggesting that these BTHs have the potential to alter xenobiotic metabolism and activate the aryl hydrocarbon receptor. In summary, several toxic actions were initiated in vitro by some but not all BTHs, warranting further study of these BTHs in vivo.
Collapse
Affiliation(s)
- Fanxing Zeng
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - James P Sherry
- Aquatic Contaminants Research Division, Environment Canada, Burlington, ON L7R 4A6, Canada
| | - Niels C Bols
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada.
| |
Collapse
|
622
|
Ruiz P, Perlina A, Mumtaz M, Fowler BA. A Systems Biology Approach Reveals Converging Molecular Mechanisms that Link Different POPs to Common Metabolic Diseases. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1034-41. [PMID: 26685285 PMCID: PMC4937870 DOI: 10.1289/ehp.1510308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 12/10/2015] [Indexed: 05/11/2023]
Abstract
BACKGROUND A number of epidemiological studies have identified statistical associations between persistent organic pollutants (POPs) and metabolic diseases, but testable hypotheses regarding underlying molecular mechanisms to explain these linkages have not been published. OBJECTIVES We assessed the underlying mechanisms of POPs that have been associated with metabolic diseases; three well-known POPs [2,3,7,8-tetrachlorodibenzodioxin (TCDD), 2,2´,4,4´,5,5´-hexachlorobiphenyl (PCB 153), and 4,4´-dichlorodiphenyldichloroethylene (p,p´-DDE)] were studied. We used advanced database search tools to delineate testable hypotheses and to guide laboratory-based research studies into underlying mechanisms by which this POP mixture could produce or exacerbate metabolic diseases. METHODS For our searches, we used proprietary systems biology software (MetaCore™/MetaDrug™) to conduct advanced search queries for the underlying interactions database, followed by directional network construction to identify common mechanisms for these POPs within two or fewer interaction steps downstream of their primary targets. These common downstream pathways belong to various cytokine and chemokine families with experimentally well-documented causal associations with type 2 diabetes. CONCLUSIONS Our systems biology approach allowed identification of converging pathways leading to activation of common downstream targets. To our knowledge, this is the first study to propose an integrated global set of step-by-step molecular mechanisms for a combination of three common POPs using a systems biology approach, which may link POP exposure to diseases. Experimental evaluation of the proposed pathways may lead to development of predictive biomarkers of the effects of POPs, which could translate into disease prevention and effective clinical treatment strategies. CITATION Ruiz P, Perlina A, Mumtaz M, Fowler BA. 2016. A systems biology approach reveals converging molecular mechanisms that link different POPs to common metabolic diseases. Environ Health Perspect 124:1034-1041; http://dx.doi.org/10.1289/ehp.1510308.
Collapse
Affiliation(s)
- Patricia Ruiz
- Computational Toxicology and Methods Development Laboratory, Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, Atlanta, Georgia, USA
- Address correspondence to P. Ruiz, Division of Toxicology and Human Health Sciences, Computational Toxicology and Methods Development Lab, Agency for Toxic Substances and Disease Registry, 1600 Clifton Rd., MS-F57, Atlanta, GA 30333. Telephone: (770) 488-3348. E-mail:
| | - Ally Perlina
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Moiz Mumtaz
- Computational Toxicology and Methods Development Laboratory, Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, Atlanta, Georgia, USA
| | - Bruce A. Fowler
- Emory University Rollins School of Public Health, Atlanta, Georgia, USA
| |
Collapse
|
623
|
Dissociation Dynamics of XPC-RAD23B from Damaged DNA Is a Determining Factor of NER Efficiency. PLoS One 2016; 11:e0157784. [PMID: 27327897 PMCID: PMC4915676 DOI: 10.1371/journal.pone.0157784] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/03/2016] [Indexed: 12/18/2022] Open
Abstract
XPC-RAD23B (XPC) plays a critical role in human nucleotide excision repair (hNER) as this complex recognizes DNA adducts to initiate NER. To determine the mutagenic potential of structurally different bulky DNA damages, various studies have been conducted to define the correlation of XPC-DNA damage equilibrium binding affinity with NER efficiency. However, little is known about the effects of XPC-DNA damage recognition kinetics on hNER. Although association of XPC is important, our current work shows that the XPC-DNA dissociation rate also plays a pivotal role in achieving NER efficiency. We characterized for the first time the binding of XPC to mono- versus di-AAF-modified sequences by using the real time monitoring surface plasmon resonance technique. Strikingly, the half-life (t1/2 or the retention time of XPC in association with damaged DNA) shares an inverse relationship with NER efficiency. This is particularly true when XPC remained bound to clustered adducts for a much longer period of time as compared to mono-adducts. Our results suggest that XPC dissociation from the damage site could become a rate-limiting step in NER of certain types of DNA adducts, leading to repression of NER.
Collapse
|
624
|
Long-term in vivo polychlorinated biphenyl 126 exposure induces oxidative stress and alters proteomic profile on islets of Langerhans. Sci Rep 2016; 6:27882. [PMID: 27292372 PMCID: PMC4904407 DOI: 10.1038/srep27882] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/24/2016] [Indexed: 12/15/2022] Open
Abstract
It has been recently proposed that exposure to polychlorinated biphenyls (PCBs) is a risk factor to type 2 diabetes mellitus (DM2). We investigated this hypothesis using long-term in vivo PCB126 exposure to rats addressing metabolic, cellular and proteomic parameters. Male Wistar rats were exposed to PCB126 (0.1, 1 or 10 μg/kg of body weight/day; for 15 days) or vehicle by intranasal instillation. Systemic alterations were quantified by body weight, insulin and glucose tolerance, and blood biochemical profile. Pancreatic toxicity was measured by inflammatory parameters, cell viability and cycle, free radical generation, and proteomic profile on islets of Langerhans. In vivo PCB126 exposure enhanced the body weight gain, impaired insulin sensitivity, reduced adipose tissue deposit, and elevated serum triglycerides, cholesterol, and insulin levels. Inflammatory parameters in the pancreas and cell morphology, viability and cycle were not altered in islets of Langerhans. Nevertheless, in vivo PCB126 exposure increased free radical generation and modified the expression of proteins related to oxidative stress on islets of Langerhans, which are indicative of early β-cell failure. Data herein obtained show that long-term in vivo PCB126 exposure through intranasal route induced alterations on islets of Langerhans related to early end points of DM2.
Collapse
|
625
|
Anstee QM, Seth D, Day CP. Genetic Factors That Affect Risk of Alcoholic and Nonalcoholic Fatty Liver Disease. Gastroenterology 2016; 150:1728-1744.e7. [PMID: 26873399 DOI: 10.1053/j.gastro.2016.01.037] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 01/17/2016] [Accepted: 01/20/2016] [Indexed: 02/07/2023]
Abstract
Genome-wide association studies and candidate gene studies have informed our understanding of factors contributing to the well-recognized interindividual variation in the progression and outcomes of alcoholic liver disease and nonalcoholic fatty liver disease. We discuss the mounting evidence for shared modifiers and common pathophysiological processes that contribute to development of both diseases. We discuss the functions of proteins encoded by risk variants of genes including patatin-like phospholipase domain-containing 3 and transmembrane 6 superfamily member 2, as well as epigenetic factors that contribute to the pathogenesis of alcoholic liver disease and nonalcoholic fatty liver disease. We also discuss important areas of future genetic research and their potential to affect clinical management of patients.
Collapse
Affiliation(s)
- Quentin M Anstee
- Liver Research Group, Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom.
| | - Devanshi Seth
- Centenary Institute of Cancer Medicine, Royal Prince Alfred Hospital, Camperdown, Australia; Drug Health Services, Royal Prince Alfred Hospital, Camperdown, Australia; Central Clinical School, The University of Sydney, Camperdown, Australia
| | - Christopher P Day
- Liver Research Group, Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
626
|
Goossens N, Hoshida Y. Is Hepatocellular Cancer the Same Disease in Alcoholic and Nonalcoholic Fatty Liver Diseases? Gastroenterology 2016; 150:1710-7. [PMID: 26784140 PMCID: PMC5120760 DOI: 10.1053/j.gastro.2016.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 01/03/2016] [Accepted: 01/07/2016] [Indexed: 12/14/2022]
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, New York.
| |
Collapse
|
627
|
Zhang L, Liu C, Xu K, Chen J. Association between PNPLA3 rs738409 polymorphism and hepatocellular carcinoma risk: an updated meta-analysis. Genes Genomics 2016. [DOI: 10.1007/s13258-016-0428-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
628
|
Yu J, Marsh S, Hu J, Feng W, Wu C. The Pathogenesis of Nonalcoholic Fatty Liver Disease: Interplay between Diet, Gut Microbiota, and Genetic Background. Gastroenterol Res Pract 2016; 2016:2862173. [PMID: 27247565 PMCID: PMC4876215 DOI: 10.1155/2016/2862173] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/14/2016] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the world, and it comprises a spectrum of hepatic abnormalities from simple hepatic steatosis to steatohepatitis, fibrosis, cirrhosis, and liver cancer. While the pathogenesis of NAFLD remains incompletely understood, a multihit model has been proposed that accommodates causal factors from a variety of sources, including intestinal and adipose proinflammatory stimuli acting on the liver simultaneously. Prior cellular and molecular studies of patient and animal models have characterized several common pathogenic mechanisms of NAFLD, including proinflammation cytokines, lipotoxicity, oxidative stress, and endoplasmic reticulum stress. In recent years, gut microbiota has gained much attention, and dysbiosis is recognized as a crucial factor in NAFLD. Moreover, several genetic variants have been identified through genome-wide association studies, particularly rs738409 (Ile748Met) in PNPLA3 and rs58542926 (Glu167Lys) in TM6SF2, which are critical risk alleles of the disease. Although a high-fat diet and inactive lifestyles are typical risk factors for NAFLD, the interplay between diet, gut microbiota, and genetic background is believed to be more important in the development and progression of NAFLD. This review summarizes the common pathogenic mechanisms, the gut microbiota relevant mechanisms, and the major genetic variants leading to NAFLD and its progression.
Collapse
Affiliation(s)
- Jinsheng Yu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sharon Marsh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada T6G 2H7
| | - Junbo Hu
- Department of General Surgery, Tongji Hospital, Huazhong Science & Technology University, Wuhan, Hubei 430030, China
| | - Wenke Feng
- Department of Medicine, University of Louisville, Louisville, KY 40208, USA
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, Houston, TX 77843, USA
| |
Collapse
|
629
|
Rosen HR, Golden-Mason L, Daly AK, Yang I, Day CP. Variants in the LGALS9 Gene Are Associated With Development of Liver Disease in Heavy Consumers of Alcohol. Clin Gastroenterol Hepatol 2016; 14:762-8.e1. [PMID: 26598225 DOI: 10.1016/j.cgh.2015.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/13/2015] [Accepted: 11/09/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Alcohol consumption is a major cause of chronic liver disease and contributes to a large proportion of cirrhosis-related deaths worldwide. However, only a fraction of heavy consumers of alcohol develop advanced alcoholic liver disease (ALD), so there are likely to be other risk factors. We investigated whether polymorphisms in the gene encoding galectin-9 (LGALS9), previously shown to mediate liver injury, were associated with the development of ALD. METHODS We isolated DNA from peripheral blood mononuclear cells (PBMCs) of 575 individuals with at-risk alcohol consumption but no other risk factors for chronic liver disease; all subjects were white Europeans who had consumed more than 80 grams ethanol per day. Of the subjects, 388 had ALD (including, 268 with cirrhosis and 74 with alcoholic hepatitis; mean age, 49 y; 72% male) and 187 had normal liver function with no biochemical or clinical evidence of liver disease (controls; mean age, 42 y; 73% male). Select LGALS9 polymorphisms were genotyped using allelic discrimination. We also genotyped and measured expression of LGALS9 messenger RNA in PBMCs from individuals who were not heavy consumers of alcohol. RESULTS We used data from the HapMap project to identify 5 single-nucleotide polymorphisms (SNPs) that tag all the common haplotypes. When we looked for these SNPs in individuals with vs without liver disease, 4 (rs3751093, rs4239242, rs732222, and rs4794976) were associated with an increased risk of developing ALD. We found that levels of LGALS9 messenger RNA and protein expressed were associated with an allele carried by PBMCs. Multivariate analysis confirmed that rs4239242 and rs4794976 were associated with an increased risk of ALD. CONCLUSIONS In a genetic analysis of heavy consumers of alcohol, we associated 2 SNPS in LGALS9 with the development of ALD. Although larger studies are required, this information could be used to determine the risk of individuals developing ALD or to develop therapeutic agents.
Collapse
Affiliation(s)
- Hugo R Rosen
- Division of Gastroenterology/Hepatology, University of Colorado, Aurora, Colorado.
| | - Lucy Golden-Mason
- Division of Gastroenterology/Hepatology, University of Colorado, Aurora, Colorado
| | - Ann K Daly
- Institute of Cellular Medicine, Newcastle University Medical School, Newcastle upon Tyne, United Kingdom
| | - Ivana Yang
- Division of Pulmonary Medicine, University of Colorado, Aurora, Colorado
| | - Christopher P Day
- Institute of Cellular Medicine, Newcastle University Medical School, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
630
|
Definitions of Normal Liver Fat and the Association of Insulin Sensitivity with Acquired and Genetic NAFLD-A Systematic Review. Int J Mol Sci 2016; 17:ijms17050633. [PMID: 27128911 PMCID: PMC4881459 DOI: 10.3390/ijms17050633] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/13/2016] [Accepted: 04/20/2016] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) covers a spectrum of disease ranging from simple steatosis (NAFL) to non-alcoholic steatohepatitis (NASH) and fibrosis. “Obese/Metabolic NAFLD” is closely associated with obesity and insulin resistance and therefore predisposes to type 2 diabetes and cardiovascular disease. NAFLD can also be caused by common genetic variants, the patatin-like phospholipase domain-containing 3 (PNPLA3) or the transmembrane 6 superfamily member 2 (TM6SF2). Since NAFL, irrespective of its cause, can progress to NASH and liver fibrosis, its definition is of interest. We reviewed the literature to identify data on definition of normal liver fat using liver histology and different imaging tools, and analyzed whether NAFLD caused by the gene variants is associated with insulin resistance. Histologically, normal liver fat content in liver biopsies is most commonly defined as macroscopic steatosis in less than 5% of hepatocytes. In the population-based Dallas Heart Study, the upper 95th percentile of liver fat measured by proton magnetic spectroscopy (1H-MRS) in healthy subjects was 5.6%, which corresponds to approximately 15% histological liver fat. When measured by magnetic resonance imaging (MRI)-based techniques such as the proton density fat fraction (PDFF), 5% macroscopic steatosis corresponds to a PDFF of 6% to 6.4%. In contrast to “Obese/metabolic NAFLD”, NAFLD caused by genetic variants is not associated with insulin resistance. This implies that NAFLD is heterogeneous and that “Obese/Metabolic NAFLD” but not NAFLD due to the PNPLA3 or TM6SF2 genetic variants predisposes to type 2 diabetes and cardiovascular disease.
Collapse
|
631
|
Whitfield JB, Rahman K, Haber PS, Day CP, Masson S, Daly AK, Cordell HJ, Mueller S, Seitz HK, Liangpunsakul S, Westerhold C, Liang T, Lumeng L, Foroud T, Nalpas B, Mathurin P, Stickel F, Soyka M, Botwin GJ, Morgan TR, Seth D. Brief report: genetics of alcoholic cirrhosis-GenomALC multinational study. Alcohol Clin Exp Res 2016; 39:836-42. [PMID: 25872595 PMCID: PMC4398999 DOI: 10.1111/acer.12693] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 01/29/2015] [Indexed: 12/20/2022]
Abstract
Background The risk of alcohol‐related liver cirrhosis increases with increasing alcohol consumption, but many people with very high intake escape from liver disease. We postulate that susceptibility to alcoholic cirrhosis has a complex genetic component and propose that this can be dissected through a large and sufficiently powered genomewide association study (GWAS). Methods The GenomALC Consortium comprises researchers from Australia, France, Germany, Switzerland, United Kingdom, and United States, with a joint aim of exploring the genetic and genomic basis of alcoholic cirrhosis. For this National Institutes of Health/National Institute on Alcohol Abuse and Alcoholism funded study, we are recruiting high‐risk drinkers who are either cases (with alcoholic cirrhosis) or controls (drinking comparable amounts over similar time, but free of significant liver disease). Extensive phenotypic data are obtained using semistructured interviews and patient records, and blood samples are collected. Results We have successfully recruited 859 participants including 538 matched case–control samples as of September 2014, using study‐specific inclusion–exclusion criteria and data collection protocols. Of these, 580 are cases (442 men and 138 women) and 279 are controls (205 men and 74 women). Duration of excessive drinking was slightly greater in cases than controls and was significantly less in women than men. Cases had significantly lower lifetime alcohol intake than controls. Both cases and controls had a high prevalence of reported parental alcohol problems, but cases were significantly more likely to report that a father with alcohol problems had died from liver disease (odds ratio 2.53, 95% confidence interval 1.31 to 4.87, p = 0.0055). Conclusions Recruitment of participants for a GWAS of alcoholic cirrhosis has proved feasible across countries with multiple sites. Affected patients often consume less alcohol than unaffected ones, emphasizing the existence of individual vulnerability factors. Cases are more likely to report liver disease in a father with alcohol problems than controls, consistent with a potential genetic component to the risk of alcoholic cirrhosis.
Collapse
Affiliation(s)
- John B Whitfield
- Department of Genetic Epidemiology , QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
632
|
Attignon EA, Leblanc AF, Le-Grand B, Duval C, Aggerbeck M, Rouach H, Blanc EB. Novel roles for AhR and ARNT in the regulation of alcohol dehydrogenases in human hepatic cells. Arch Toxicol 2016; 91:313-324. [PMID: 27055685 DOI: 10.1007/s00204-016-1700-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/21/2016] [Indexed: 12/13/2022]
Abstract
The mechanisms by which pollutants participate in the development of diverse pathologies are not completely understood. The pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) activates the AhR (aryl hydrocarbon receptor) signaling pathway. We previously showed that TCDD (25 nM, 30 h) decreased the expression of several alcohol metabolism enzymes (cytochrome P450 2E1, alcohol dehydrogenases ADH1, 4 and 6) in differentiated human hepatic cells (HepaRG). Here, we show that, as rapidly as 8 h after treatment (25 nM TCDD) ADH expression decreased 40 % (p < 0.05). ADH1 and 4 protein levels decreased 40 and 27 %, respectively (p < 0.05), after 72 h (25 nM TCDD). The protein half-lives were not modified by TCDD which suggests transcriptional regulation of expression. The AhR antagonist CH-223191 or AhR siRNA reduced the inhibitory effect of 25 nM TCDD on ADH1A, 4 and 6 expression 50-100 % (p < 0.05). The genomic pathway (via the AhR/ARNT complex) and not the non-genomic pathway involving c-SRC mediated these effects. Other AhR ligands (3-methylcholanthrene and PCB 126) decreased ADH1B, 4 and 6 mRNAs by more than 78 and 55 %, respectively (p < 0.01). TCDD also regulated the expression of ADH4 in the HepG2 human hepatic cell line, in primary human hepatocytes and in C57BL/6J mouse liver. In conclusion, activation of the AhR/ARNT signaling pathway by AhR ligands represents a novel mechanism for regulating the expression of ADHs. These effects may be implicated in the toxicity of AhR ligands as well as in the alteration of ethanol or retinol metabolism and may be associated further with higher risk of liver diseases or/and alcohol abuse disorders.
Collapse
Affiliation(s)
- Eléonore A Attignon
- INSERM, UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints Pères, F-75006, Paris, France.,ComUE Sorbonne Paris Cité, Université Paris Descartes, CICB-Paris, 45 rue des Saints Pères, F-75006, Paris, France
| | - Alix F Leblanc
- INSERM, UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints Pères, F-75006, Paris, France.,ComUE Sorbonne Paris Cité, Université Paris Descartes, CICB-Paris, 45 rue des Saints Pères, F-75006, Paris, France
| | - Béatrice Le-Grand
- INSERM, UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints Pères, F-75006, Paris, France.,ComUE Sorbonne Paris Cité, Université Paris Descartes, CICB-Paris, 45 rue des Saints Pères, F-75006, Paris, France
| | - Caroline Duval
- INSERM, UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints Pères, F-75006, Paris, France.,ComUE Sorbonne Paris Cité, Université Paris Descartes, CICB-Paris, 45 rue des Saints Pères, F-75006, Paris, France
| | - Martine Aggerbeck
- INSERM, UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints Pères, F-75006, Paris, France.,ComUE Sorbonne Paris Cité, Université Paris Descartes, CICB-Paris, 45 rue des Saints Pères, F-75006, Paris, France
| | - Hélène Rouach
- INSERM, UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints Pères, F-75006, Paris, France.,ComUE Sorbonne Paris Cité, Université Paris Descartes, CICB-Paris, 45 rue des Saints Pères, F-75006, Paris, France
| | - Etienne B Blanc
- INSERM, UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints Pères, F-75006, Paris, France. .,ComUE Sorbonne Paris Cité, Université Paris Descartes, CICB-Paris, 45 rue des Saints Pères, F-75006, Paris, France.
| |
Collapse
|
633
|
Ueyama M, Nishida N, Korenaga M, Korenaga K, Kumagai E, Yanai H, Adachi H, Katsuyama H, Moriyama S, Hamasaki H, Sako A, Sugiyama M, Aoki Y, Imamura M, Murata K, Masaki N, Kawaguchi T, Torimura T, Hyogo H, Aikata H, Ito K, Sumida Y, Kanazawa A, Watada H, Okamoto K, Honda K, Kon K, Kanto T, Mizokami M, Watanabe S. The impact of PNPLA3 and JAZF1 on hepatocellular carcinoma in non-viral hepatitis patients with type 2 diabetes mellitus. J Gastroenterol 2016; 51:370-379. [PMID: 26337813 DOI: 10.1007/s00535-015-1116-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/14/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is an established independent risk factor for hepatocellular carcinoma (HCC). T2DM is associated with non-alcoholic steatohepatitis (NASH), which is a major cause of non-HBV and non-HCV-related HCC; nevertheless, it has been difficult to identify those patients with T2DM who have a high risk of developing HCC. The aim of this study was to identify genetic determinants that predispose T2DM patients to HCC by genotyping T2DM susceptibility loci and PNPLA3. METHODS We recruited 389 patients with T2DM who satisfied the following three criteria: negative for HBs-Ag and anti-HCV Ab, alcohol intake <60 g/day, and history of T2DM >10 years. These patients were divided into two groups: T2DM patients with HCC (DM-HCC, n = 59) or those without HCC (DM-non-HCC, n = 330). We genotyped 51 single-nucleotide polymorphisms (SNPs) previously reported as T2DM or NASH susceptibility loci (PNPLA3) compared between the DM-HCC and DM-non-HCC groups with regard to allele frequencies at each SNP. RESULTS The SNP rs738409 located in PNPLA3 was the greatest risk factor associated with HCC. The frequency of the PNPLA3 G allele was significantly higher among DM-HCC individuals than DM-non-HCC individuals (OR 2.53, p = 1.05 × 10(-5)). Among individuals homozygous for the PNPLA3 G allele (n = 115), the frequency of the JAZF1 rs864745 G allele was significantly higher among DM-HCC individuals than DM-non-HCC individuals (OR 3.44, p = 0.0002). CONCLUSIONS PNPLA3 and JAZF1 were associated with non-HBV and non-HCV-related HCC development among Japanese patients with T2DM.
Collapse
Affiliation(s)
- Misuzu Ueyama
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
- Department of Gastroenterology, Juntendo University School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Nao Nishida
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Masaaki Korenaga
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan.
| | - Keiko Korenaga
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Erina Kumagai
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
- Department of Gastroenterology, Juntendo University School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Hidekatsu Yanai
- Department of Internal Medicine, National Center for Global Health and Medicine Kohnodai Hospital, Ichikawa, Chiba, Japan
| | - Hiroki Adachi
- Department of Internal Medicine, National Center for Global Health and Medicine Kohnodai Hospital, Ichikawa, Chiba, Japan
| | - Hisayuki Katsuyama
- Department of Internal Medicine, National Center for Global Health and Medicine Kohnodai Hospital, Ichikawa, Chiba, Japan
| | - Sumie Moriyama
- Department of Internal Medicine, National Center for Global Health and Medicine Kohnodai Hospital, Ichikawa, Chiba, Japan
| | - Hidetaka Hamasaki
- Department of Internal Medicine, National Center for Global Health and Medicine Kohnodai Hospital, Ichikawa, Chiba, Japan
| | - Akahito Sako
- Department of Internal Medicine, National Center for Global Health and Medicine Kohnodai Hospital, Ichikawa, Chiba, Japan
| | - Masaya Sugiyama
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Yoshihiko Aoki
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Masatoshi Imamura
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Kazumoto Murata
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Naohiko Masaki
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hideyuki Hyogo
- Department of Gastroenterology and Metabolism, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Hiroshima University, Hiroshima, Japan
| | - Kiyoaki Ito
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yoshio Sumida
- Center for Digestive and Liver Diseases, Nara City Hospital, Nara, Japan
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan
| | - Akio Kanazawa
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Koji Okamoto
- Department of Nephrology and Endocrinology, Department of Hemodialysis and Apheresis, University Hospital, The University of Tokyo, Tokyo, Japan
| | - Kenjiro Honda
- Department of Nephrology and Endocrinology, Department of Hemodialysis and Apheresis, University Hospital, The University of Tokyo, Tokyo, Japan
| | - Kazuyoshi Kon
- Department of Gastroenterology, Juntendo University School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Tatsuya Kanto
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Masashi Mizokami
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Sumio Watanabe
- Department of Gastroenterology, Juntendo University School of Medicine, Bunkyo-Ku, Tokyo, Japan
| |
Collapse
|
634
|
Mueller JL, King LY, Johnson KB, Gao T, Nephew LD, Kothari D, Simpson MA, Zheng H, Wei L, Corey KE, Misdraji J, Lee JH, Lin MV, Gogela NA, Fuchs BC, Tanabe KK, Gordon FD, Curry MP, Chung RT. Impact of EGF, IL28B, and PNPLA3 polymorphisms on the outcome of allograft hepatitis C: a multicenter study. Clin Transplant 2016; 30:452-460. [PMID: 26854475 PMCID: PMC4868041 DOI: 10.1111/ctr.12710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2016] [Indexed: 01/03/2023]
Abstract
Hepatitis C virus (HCV) infection is accelerated following liver transplantation (LT). Single nucleotide polymorphisms (SNPs) near the epidermal growth factor (EGF) (rs4444903), IL28B (rs12979860), and PNPLA3 (rs738409) loci are associated with treatment response, fibrosis, and hepatocellular carcinoma in non-transplant hepatitis C, but allograft population data are limited. We sought to determine the role of these SNPs in 264 patients with HCV who underwent LT between 1990 and 2008. Genotypes were determined from donor wedge/allograft biopsies and recipient explants. Cox proportional hazards model was used to assess time to cirrhosis, liver-related death, and retransplantation, adjusting for donor age and sustained virological response (SVR). Over a median follow-up of 6.3 yr, a trend toward increased progression to graft cirrhosis was observed among recipients of an EGF non-AA vs. AA donor liver (adjusted HR 2.01; 95% CI 0.93-4.34; p = 0.08). No other genotypes predicted cirrhosis development or graft survival. The CC IL28B variant in both recipients and donors was associated with increased rate of SVR (R-CC/D-CC 8/12[67%], R-non-CC/D-CC or R-CC/D-non-CC 23/52[44%], R-non-CC/D-non-CC 12/45[27%], p linear trend = 0.009). Recipient EGF, IL28B, and PNPLA3, and donor IL28B and PNPLA3 genotypes do not predict adverse outcomes in HCV LT recipients. A potential association exists between donor EGF genotype and cirrhosis.
Collapse
Affiliation(s)
- Jessica L. Mueller
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Lindsay Y. King
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Kara B. Johnson
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Tian Gao
- Division of Gastroenterology, Department of Medicine, Boston Medical Center, Boston, MA
| | - Lauren D. Nephew
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Health System, Philadelphia, PA
| | - Darshan Kothari
- Harvard Medical School, Boston, MA
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconness Medical Center, Boston
| | - Mary Ann Simpson
- Division of Gastroenterology, Department of Medicine, Lahey Hospital & Medical Center, Burlington
| | - Hui Zheng
- Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA
| | - Lan Wei
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Kathleen E. Corey
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Joseph Misdraji
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Joon Hyoek Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - M. Valerie Lin
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Neliswa A. Gogela
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Bryan C. Fuchs
- Harvard Medical School, Boston, MA
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Kenneth K. Tanabe
- Harvard Medical School, Boston, MA
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Fredric D. Gordon
- Harvard Medical School, Boston, MA
- Division of Gastroenterology, Department of Medicine, Lahey Hospital & Medical Center, Burlington
| | - Michael P. Curry
- Harvard Medical School, Boston, MA
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconness Medical Center, Boston
| | - Raymond T. Chung
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
635
|
Inhibition of the aryl hydrocarbon receptor prevents Western diet-induced obesity. Model for AHR activation by kynurenine via oxidized-LDL, TLR2/4, TGFβ, and IDO1. Toxicol Appl Pharmacol 2016; 300:13-24. [PMID: 27020609 DOI: 10.1016/j.taap.2016.03.011] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/11/2016] [Accepted: 03/21/2016] [Indexed: 12/25/2022]
Abstract
Obesity is an increasingly urgent global problem, yet, little is known about its causes and less is known how obesity can be effectively treated. We showed previously that the aryl hydrocarbon receptor (AHR) plays a role in the regulation of body mass in mice fed Western diet. The AHR is a ligand-activated nuclear receptor that regulates genes involved in a number of biological pathways, including xenobiotic metabolism and T cell polarization. This study was an investigation into whether inhibition of the AHR prevents Western diet-based obesity. Male C57Bl/6J mice were fed control and Western diets with and without the AHR antagonist α-naphthoflavone or CH-223191, and a mouse hepatocyte cell line was used to delineate relevant cellular pathways. Studies are presented showing that the AHR antagonists α-naphthoflavone and CH-223191 significantly reduce obesity and adiposity and ameliorates liver steatosis in male C57Bl/6J mice fed a Western diet. Mice deficient in the tryptophan metabolizing enzyme indoleamine 2,3-dioxygenase 1 (IDO1) were also resistant to obesity. Using an AHR-directed, luciferase-expressing mouse hepatocyte cell line, we show that the transforming growth factor β1 (TGFβ1) signaling pathway via PI3K and NF-κB and the toll-like receptor 2/4 (TLR2/4) signaling pathway stimulated by oxidized low-density lipoproteins via NF-κB, each induce luciferase expression; however, TLR2/4 signaling was significantly reduced by inhibition of IDO1. At physiological levels, kynurenine but not kynurenic acid (both tryptophan metabolites and known AHR agonists) activated AHR-directed luciferase expression. We propose a hepatocyte-based model, in which kynurenine production is increased by enhanced IDO1 activity stimulated by TGFβ1 and TLR2/4 signaling, via PI3K and NF-κB, to perpetuate a cycle of AHR activation to cause obesity; and inhibition of the AHR, in turn, blocks the cycle's output to prevent obesity. The AHR with its broad ligand binding specificity is a promising candidate for a potentially simple therapeutic approach for the prevention and treatment of obesity and associated complications.
Collapse
|
636
|
Syndecan-4 shedding impairs macrovascular angiogenesis in diabetes mellitus. Biochem Biophys Res Commun 2016; 474:15-21. [PMID: 27018253 DOI: 10.1016/j.bbrc.2016.03.112] [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: 03/16/2016] [Accepted: 03/23/2016] [Indexed: 12/21/2022]
Abstract
PURPOSE Syndecan-4 (synd4) is a ubiquitous heparan sulfate proteoglycan cell surface receptor that modulates cell proliferation, migration, mechanotransduction, and endocytosis. The extracellular domain of synd4 sheds heavily in acute inflammation, but the shedding of synd4 in chronic inflammation, such as diabetes mellitus (DM), is still undefined. We investigated the alterations of synd4 endothelial expression in DM and the influence of impaired synd4 signaling on angiogenesis in human umbilical vein endothelial cells (HUVECs), diabetic rats, synd4 null mice, and db/db mice. MATERIAL AND METHODS HUVECs were incubated with advanced glycation end products (AGEs). Western blot analysis was used to determine synd4 protein expression and ELISA was used to detect soluble synd4 fragments. The concentration of synd4 in the aortic endothelia of diabetic rats was detected by immunohistochemical staining. Aortic ring assays were performed to study the process of angiogenesis in the diabetic rats and in synd4 null and db/db mice. Recombinant adenoviruses containing the synd4 gene or null were constructed to enhance synd4 aortic expression in db/db mice. RESULTS Western blot analysis showed decreased expression of the synd4 extracellular domain in HUVECs, and ELISA detected increased soluble fragments of synd4 in the media. Synd4 endothelial expression in the aortas of diabetic rats was decreased. Aortic ring assay indicated impaired angiogenesis in synd4 null and db/db mice, which was partially reversed by synd4 overexpression in db/db mice. CONCLUSION Synd4 shedding from vascular endothelial cells played an important role in the diabetes-related impairment of angiogenesis.
Collapse
|
637
|
Smagris E, Gilyard S, BasuRay S, Cohen JC, Hobbs HH. Inactivation of Tm6sf2, a Gene Defective in Fatty Liver Disease, Impairs Lipidation but Not Secretion of Very Low Density Lipoproteins. J Biol Chem 2016; 291:10659-76. [PMID: 27013658 PMCID: PMC4865914 DOI: 10.1074/jbc.m116.719955] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Indexed: 12/18/2022] Open
Abstract
A missense mutation (E167K) in TM6SF2 (transmembrane 6 superfamily member 2), a polytopic protein of unknown function, is associated with the full spectrum of fatty liver disease. To investigate the role of TM6SF2 in hepatic triglyceride (TG) metabolism, we inactivated the gene in mice. Chronic inactivation of Tm6sf2 in mice is associated with hepatic steatosis, hypocholesterolemia, and transaminitis, thus recapitulating the phenotype observed in humans. No dietary challenge was required to elicit the phenotype. Immunocytochemical and cell fractionation studies revealed that TM6SF2 was present in the endoplasmic reticulum and Golgi complex, whereas the excess neutral lipids in the Tm6sf2(-/-) mice were located in lipid droplets. Plasma VLDL-TG levels were reduced in the KO animals due to a 3-fold decrease in VLDL-TG secretion rate without any associated reduction in hepatic apoB secretion. Both VLDL particle size and plasma cholesterol levels were significantly reduced in KO mice. Despite levels of TM6SF2 protein being 10-fold higher in the small intestine than in the liver, dietary lipid absorption was only modestly reduced in the KO mice. Our data, taken together, reveal that TM6SF2 is required to mobilize neutral lipids for VLDL assembly but is not required for secretion of apoB-containing lipoproteins. Despite TM6SF2 being located in the endoplasmic reticulum and Golgi complex, the lipids that accumulate in its absence reside in lipid droplets.
Collapse
Affiliation(s)
- Eriks Smagris
- From the Departments of Molecular Genetics and Internal Medicine and
| | - Shenise Gilyard
- From the Departments of Molecular Genetics and Internal Medicine and
| | | | | | - Helen H Hobbs
- From the Departments of Molecular Genetics and Internal Medicine and the Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
638
|
Lu Y, Li N, Gao L, Xu YJ, Huang C, Yu K, Ling Q, Cheng Q, Chen S, Zhu M, Fang J, Chen M, Ong CN. Acetylcarnitine Is a Candidate Diagnostic and Prognostic Biomarker of Hepatocellular Carcinoma. Cancer Res 2016; 76:2912-20. [PMID: 26976432 DOI: 10.1158/0008-5472.can-15-3199] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/03/2016] [Indexed: 01/02/2023]
Abstract
The identification of serum biomarkers to improve the diagnosis and prognosis of hepatocellular carcinoma has been elusive to date. In this study, we took a mass spectroscopic approach to characterize metabolic features of the liver in hepatocellular carcinoma patients to discover more sensitive and specific biomarkers for diagnosis and progression. Global metabolic profiling of 50 pairs of matched liver tissue samples from hepatocellular carcinoma patients was performed. A series of 62 metabolites were found to be altered significantly in liver tumors; however, levels of acetylcarnitine correlated most strongly with tumor grade and could discriminate between hepatocellular carcinoma tumors and matched normal tissues. Post hoc analysis to evaluate serum diagnosis and progression potential further confirmed the diagnostic capability of serum acetylcarnitine. Finally, an external validation in an independent batch of 58 serum samples (18 hepatocellular carcinoma patients, 20 liver cirrhosis patients, and 20 healthy individuals) verified that serum acetylcarnitine was a meaningful biomarker reflecting hepatocellular carcinoma diagnosis and progression. These findings present a strong new candidate biomarker for hepatocellular carcinoma with potentially significant diagnostic and prognostic capabilities. Cancer Res; 76(10); 2912-20. ©2016 AACR.
Collapse
Affiliation(s)
- Yonghai Lu
- School of Public Health, National University of Singapore, Singapore
| | - Ning Li
- Department of Infectious Diseases and Hepatology of Huashan Hospital, Fudan University, Shanghai, China
| | - Liang Gao
- NUS Environmental Research Institute, National University of Singapore, Singapore
| | - Yong-Jiang Xu
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chong Huang
- Department of Infectious Diseases and Hepatology of Huashan Hospital, Fudan University, Shanghai, China
| | - Kangkang Yu
- Department of Infectious Diseases and Hepatology of Huashan Hospital, Fudan University, Shanghai, China
| | - Qingxia Ling
- Department of Infectious Diseases and Hepatology of Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Cheng
- Department of Infectious Diseases and Hepatology of Huashan Hospital, Fudan University, Shanghai, China
| | - Shengsen Chen
- Department of Infectious Diseases and Hepatology of Huashan Hospital, Fudan University, Shanghai, China
| | - Mengqi Zhu
- Department of Infectious Diseases and Hepatology of Huashan Hospital, Fudan University, Shanghai, China
| | - Jinling Fang
- School of Public Health, National University of Singapore, Singapore
| | - Mingquan Chen
- Department of Infectious Diseases and Hepatology of Huashan Hospital, Fudan University, Shanghai, China.
| | - Choon Nam Ong
- School of Public Health, National University of Singapore, Singapore. NUS Environmental Research Institute, National University of Singapore, Singapore.
| |
Collapse
|
639
|
Forner A, Reig M, Varela M, Burrel M, Feliu J, Briceño J, Sastre J, Martí-Bonmati L, Llovet JM, Bilbao JI, Sangro B, Pardo F, Ayuso C, Bru C, Tabernero J, Bruix J. [Diagnosis and treatment of hepatocellular carcinoma. Update consensus document from the AEEH, SEOM, SERAM, SERVEI and SETH]. Med Clin (Barc) 2016; 146:511.e1-511.e22. [PMID: 26971984 DOI: 10.1016/j.medcli.2016.01.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/22/2016] [Accepted: 01/28/2016] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma is the most common primary malignancy of the liver and one of the most frequent causes of death in patients with liver cirrhosis. Simultaneously with the recognition of the clinical relevance of this neoplasm, in recent years there have been important developments in the diagnosis, staging and treatment of HCC. Consequently, the Asociación Española para el Estudio del Hígado has driven the need to update clinical practice guidelines, continuing to invite all the societies involved in the diagnosis and treatment of this disease to participate in the drafting and approval of the document (Sociedad Española de Trasplante Hepático, Sociedad Española de Radiología Médica, Sociedad Española de Radiología Vascular e Intervencionista y Sociedad Española de Oncología Médica). The clinical practice guidelines published in 2009 accepted as Clinical Practice Guidelines of the National Health System has been taken as reference document, incorporating the most important advances that have been made in recent years. The scientific evidence for the treatment of HCC has been evaluated according to the recommendations of the National Cancer Institute (www.cancer.gov) and the strength of recommendation is based on the GRADE system.
Collapse
Affiliation(s)
- Alejandro Forner
- Unidad de Oncología Hepática (Barcelona Clinic Liver Cancer), Servicio de Hepatología, Hospital Clínic, Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, España; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), España
| | - María Reig
- Unidad de Oncología Hepática (Barcelona Clinic Liver Cancer), Servicio de Hepatología, Hospital Clínic, Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, España; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), España
| | - María Varela
- Sección de Hepatología, Servicio de Aparato Digestivo, Hospital Universitario Central de Asturias (HUCA), Universidad de Oviedo, Oviedo, España
| | - Marta Burrel
- Unidad de Oncología Hepática (Barcelona Clinic Liver Cancer), Servicio de Radiodiagnóstico, Hospital Clínic, Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, España
| | - Jaime Feliu
- Servicio de Oncología Médica, Hospital Universitario La Paz, Universidad Autónoma de Madrid, Sociedad Española de Oncología Médica, Madrid, España
| | - Javier Briceño
- Unidad de Trasplante Hepático, Servicio de Cirugía General y del Aparato Digestivo, Hospital Universitario Reina Sofía, Córdoba, España
| | - Javier Sastre
- Servicio de Oncología Médica, Hospital Clínico San Carlos, Madrid, España
| | - Luis Martí-Bonmati
- Departamento de Radiología, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - Josep María Llovet
- Unidad de Oncología Hepática (Barcelona Clinic Liver Cancer), Servicio de Hepatología, Hospital Clínic, Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, España; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), España; Mount Sinai Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, Estados Unidos
| | - José Ignacio Bilbao
- Unidad de Radiología Vascular e Intervencionista, Departamento de Radiodiagnóstico, Clínica Universidad de Navarra, Pamplona, España
| | - Bruno Sangro
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), España; Unidad de Hepatología, Departamento de Medicina Interna, Clínica Universidad de Navarra, Pamplona, España
| | - Fernando Pardo
- Servicio de Cirugía Hepatobliopancreática y Trasplante, Clínica Universidad de Navarra, Pamplona, España
| | - Carmen Ayuso
- Unidad de Oncología Hepática (Barcelona Clinic Liver Cancer), Servicio de Radiodiagnóstico, Hospital Clínic, Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, España
| | - Concepció Bru
- Unidad de Oncología Hepática (Barcelona Clinic Liver Cancer), Servicio de Radiodiagnóstico, Hospital Clínic, Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, España
| | - Josep Tabernero
- Servicio de Oncología Médica, Hospital Universitario Vall d'Hebrón, Barcelona, Universidad Autónoma de Barcelona, Barcelona, España
| | - Jordi Bruix
- Unidad de Oncología Hepática (Barcelona Clinic Liver Cancer), Servicio de Hepatología, Hospital Clínic, Barcelona, IDIBAPS, Universidad de Barcelona, Barcelona, España; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), España.
| |
Collapse
|
640
|
Jiao J, Watt GP, Lee M, Rahbar MH, Vatcheva KP, Pan JJ, McCormick JB, Fisher-Hoch SP, Fallon MB, Beretta L. Cirrhosis and Advanced Fibrosis in Hispanics in Texas: The Dominant Contribution of Central Obesity. PLoS One 2016; 11:e0150978. [PMID: 26950933 PMCID: PMC4780836 DOI: 10.1371/journal.pone.0150978] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/22/2016] [Indexed: 02/07/2023] Open
Abstract
Liver cirrhosis is a leading cause of death in Hispanics and Hispanics who live in South Texas have the highest incidence of liver cancer in the United States. We aimed at determining the prevalence and associated risk factors of cirrhosis in this population. Clinical and demographic variables were extracted for 2466 participants in the community-based Cameron County Hispanic Cohort in South Texas. Aspartate transaminase to Platelet Ratio Index (APRI) was used to predict cirrhosis in Cameron County Hispanic Cohort. The prevalence of cirrhosis using APRI≥2 was 0.94%, which is nearly 4-fold higher than the national prevalence. Using APRI≥1, the overall prevalence of cirrhosis/advanced fibrosis was 3.54%. In both analyses, highest prevalence was observed in males, specifically in the 25-34 age group. Risk factors independently associated with APRI≥2 and APRI≥1 included hepatitis C, diabetes and central obesity with a remarkable population attributable fraction of 52.5% and 65.3% from central obesity, respectively. Excess alcohol consumption was also independently associated with APRI≥2. The presence of patatin-like phospholipase domain-containing-3 gene variants was independently associated with APRI≥1 in participants >50 years old. Males with both central obesity and excess alcohol consumption presented with cirrhosis/advanced fibrosis at a young age. Alarmingly high prevalence of cirrhosis and advanced fibrosis was identified in Hispanics in South Texas, affecting young males in particular. Central obesity was identified as the major risk factor. Public health efforts are urgently needed to increase awareness and diagnosis of advanced liver fibrosis in Hispanics.
Collapse
Affiliation(s)
- Jingjing Jiao
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Gordon P. Watt
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, Texas, United States of America
| | - MinJae Lee
- Department of Internal Medicine, Division of Clinical and Translational Sciences, The University of Texas Medical School at Houston, Houston, Texas, United States of America
- Biostatistics/Epidemiology/Research Design (BERD) Core, Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Mohammad H. Rahbar
- Department of Internal Medicine, Division of Clinical and Translational Sciences, The University of Texas Medical School at Houston, Houston, Texas, United States of America
- Biostatistics/Epidemiology/Research Design (BERD) Core, Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- The University of Texas School of Public Health at Houston, Houston, Texas, United States of America
| | - Kristina P. Vatcheva
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, Texas, United States of America
| | - Jen-Jung Pan
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, The University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Joseph B. McCormick
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, Texas, United States of America
| | - Susan P. Fisher-Hoch
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, Texas, United States of America
| | - Michael B. Fallon
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, The University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
641
|
Petta S, Valenti L, Bugianesi E, Targher G, Bellentani S, Bonino F. A "systems medicine" approach to the study of non-alcoholic fatty liver disease. Dig Liver Dis 2016; 48:333-342. [PMID: 26698409 DOI: 10.1016/j.dld.2015.10.027] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 10/18/2015] [Accepted: 10/31/2015] [Indexed: 02/07/2023]
Abstract
The prevalence of fatty liver (steatosis) in the general population is rapidly increasing worldwide. The progress of knowledge in the physiopathology of fatty liver is based on the systems biology approach to studying the complex interactions among different physiological systems. Similarly, translational and clinical research should address the complex interplay between these systems impacting on fatty liver. The clinical needs drive the applications of systems medicine to re-define clinical phenotypes, assessing the multiple nature of disease susceptibility and progression (e.g. the definition of risk, prognosis, diagnosis criteria, and new endpoints of clinical trials). Based on this premise and in light of recent findings, the complex mechanisms involved in the pathology of fatty liver and their impact on the short- and long-term clinical outcomes of cardiovascular, metabolic liver diseases associated with steatosis are presented in this review using a new "systems medicine" approach. A new data set is proposed for studying the impairments of different physiological systems that have an impact on fatty liver in different subsets of subjects and patients.
Collapse
Affiliation(s)
- Salvatore Petta
- Section of Gastroenterology, Di.Bi.M.I.S Policlinico Paolo Giaccone Hospital, University of Palermo, Italy
| | - Luca Valenti
- Internal Medicine, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, University of Milan, Italy
| | - Elisabetta Bugianesi
- Gastroenterology and Hepatology, Department of Medical Sciences, Città della Salute e della Scienza di Torino Hospital, University of Turin, Italy
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University Hospital of Verona, Italy
| | - Stefano Bellentani
- Shrewsbury and Telford NHS Trust, Department of Gastroenterology, Shrewsbury, UK; Fondazione Italiana Fegato, Bassovizza, Trieste, Italy
| | - Ferruccio Bonino
- General Medicine 2, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Italy.
| |
Collapse
|
642
|
Donati B, Motta BM, Pingitore P, Meroni M, Pietrelli A, Alisi A, Petta S, Xing C, Dongiovanni P, del Menico B, Rametta R, Mancina RM, Badiali S, Fracanzani AL, Craxì A, Fargion S, Nobili V, Romeo S, Valenti L. The rs2294918 E434K variant modulates patatin-like phospholipase domain-containing 3 expression and liver damage. Hepatology 2016; 63:787-98. [PMID: 26605757 DOI: 10.1002/hep.28370] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 11/23/2015] [Indexed: 12/23/2022]
Abstract
UNLABELLED The patatin-like phosholipase domain-containing 3 (PNPLA3) rs738409 polymorphism (I148M) is a major determinant of hepatic fat and predisposes to the full spectrum of liver damage in nonalcoholic fatty liver disease (NAFLD). The aim of this study was to evaluate whether additional PNPLA3 coding variants contribute to NAFLD susceptibility, first in individuals with contrasting phenotypes (with early-onset NAFLD vs. very low aminotransferases) and then in a large validation cohort. Rare PNPLA3 variants were not detected by sequencing coding regions and intron-exon boundaries either in 142 patients with early-onset NAFLD nor in 100 healthy individuals with alanine aminotransferase <22/20 IU/mL. Besides rs738409 I148M, the rs2294918 G>A polymorphism (E434K sequence variant) was over-represented in NAFLD (adjusted P = 0.01). In 1,447 subjects with and without NAFLD, the 148M-434E (P < 0.0001), but not the 148M-434K, haplotype (P > 0.9), was associated with histological NAFLD and steatohepatitis. Both the I148M (P = 0.0002) and E434K variants (P = 0.044) were associated with serum ALT levels, by interacting with each other, in that the 434K hampered the association with liver damage of the 148M allele (P = 0.006). The E434K variant did not affect PNPLA3 enzymatic activity, but carriers of the rs2294918 A allele (434K) displayed lower hepatic PNPLA3 messenger RNA and protein levels (P < 0.05). CONCLUSIONS Rare loss-of-function PNPLA3 variants were not detected in early-onset NAFLD. However, PNPLA3 rs2294918 E434K decreased PNPLA3 expression, lessening the effect of the I148M variant on the predisposition to steatosis and liver damage. This suggests that the PNPLA3 I148M variant has a codominant negative effect on triglycerides mobilization from lipid droplets, mediated by inhibition of other lipases.
Collapse
Affiliation(s)
- Benedetta Donati
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Benedetta Maria Motta
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy.,Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Piero Pingitore
- Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marica Meroni
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Alessandro Pietrelli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy.,Istituto Nazionale Genetica Molecolare (INGM), "Romeo ed Enrica Invernizzi", Bioinformatic Group, Milano, Italy
| | - Anna Alisi
- Hepato-Metabolic Unit, Ospedale Bambin Gesù, Roma, Italy
| | - Salvatore Petta
- Department of Gastroenterology, Università di Palermo, Palermo, Italy
| | - Chao Xing
- UT Southwestern Medical Center, Dallas, TX
| | - Paola Dongiovanni
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Benedetta del Menico
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Raffaela Rametta
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Rosellina Margherita Mancina
- Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Sara Badiali
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Anna Ludovica Fracanzani
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Antonio Craxì
- Department of Gastroenterology, Università di Palermo, Palermo, Italy
| | - Silvia Fargion
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Valerio Nobili
- Hepato-Metabolic Unit, Ospedale Bambin Gesù, Roma, Italy
| | - Stefano Romeo
- Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.,Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.,Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| |
Collapse
|
643
|
Lee HA, Park SH, Hong YS, Ha EH, Park H. The Effect of Exposure to Persistent Organic Pollutants on Metabolic Health among KOREAN Children during a 1-Year Follow-Up. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13030270. [PMID: 26938545 PMCID: PMC4808933 DOI: 10.3390/ijerph13030270] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/14/2016] [Accepted: 02/25/2016] [Indexed: 12/25/2022]
Abstract
Previous evidence suggests the potential for adverse effects of persistent organic pollutants (POPs) on metabolic health even at low-dose exposure levels common among the general population, but there is less evidence of these associations among children. Therefore, as part of a prospective cohort study, 214 children were measured for POPs exposure. After the 1-year follow-up, we assessed the effect of circulating POPs exposure among 158 children aged 7-9 years (at baseline) on the change of metabolic components of metabolic syndrome using multiple regression analysis. In addition, we calculated the continuous metabolic syndrome (cMetS) score and assessed the variation among individuals by POPs exposure. The concentrations of marker polychlorinated biphenyls (PCBs) were significantly associated with increased change in diastolic blood pressure (BP) and triglyceride levels during a 1-year follow-up, after controlling for sex, age, household income, and change in body mass index. Total PCBs also showed a marginal association with increasing cMetS score from the baseline. Of the metabolic components, change in diastolic BP over time showed a notable association with specific PCBs, but no association with organochlorine pesticides. Here, we found that low-dose exposures to PCBs among children in the general population could negatively influence metabolic health, particularly diastolic BP. Increased disease sensitivity during childhood can continue to adulthood, thus, these results support the need for continuous assessment of the health impact of POPs.
Collapse
Affiliation(s)
- Hye Ah Lee
- Department of Preventive Medicine, School of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-ku, Seoul 158-710, Korea.
| | - Su Hyun Park
- Department of Preventive Medicine, School of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-ku, Seoul 158-710, Korea.
| | - Young Sun Hong
- Department of Internal Medicine, School of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-ku, Seoul 158-710, Korea.
| | - Eun Hee Ha
- Department of Preventive Medicine, School of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-ku, Seoul 158-710, Korea.
| | - Hyesook Park
- Department of Preventive Medicine, School of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-ku, Seoul 158-710, Korea.
| |
Collapse
|
644
|
Charrez B, Qiao L, Hebbard L. Hepatocellular carcinoma and non-alcoholic steatohepatitis: The state of play. World J Gastroenterol 2016; 22:2494-2502. [PMID: 26937137 PMCID: PMC4768195 DOI: 10.3748/wjg.v22.i8.2494] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/18/2015] [Accepted: 12/30/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is now the fifth cancer of greatest frequency and the second leading cause of cancer related deaths worldwide. Chief amongst the risks of HCC are hepatitis B and C infection, aflatoxin B1 ingestion, alcoholism and obesity. The latter can promote non-alcoholic fatty liver disease (NAFLD), that can lead to the inflammatory form non-alcoholic steatohepatitis (NASH), and can in turn promote HCC. The mechanisms by which NASH promotes HCC are only beginning to be characterized. Here in this review, we give a summary of the recent findings that describe and associate NAFLD and NASH with the subsequent HCC progression. We will focus our discussion on clinical and genomic associations that describe new risks for NAFLD and NASH promoted HCC. In addition, we will consider novel murine models that clarify some of the mechanisms that drive NASH HCC formation.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Disease Models, Animal
- Humans
- Liver/metabolism
- Liver/pathology
- Liver Neoplasms/etiology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Non-alcoholic Fatty Liver Disease/complications
- Non-alcoholic Fatty Liver Disease/genetics
- Non-alcoholic Fatty Liver Disease/metabolism
- Non-alcoholic Fatty Liver Disease/pathology
- Risk Factors
- Signal Transduction
Collapse
|
645
|
Korkina L. Metabolic and redox barriers in the skin exposed to drugs and xenobiotics. Expert Opin Drug Metab Toxicol 2016; 12:377-88. [PMID: 26854731 DOI: 10.1517/17425255.2016.1149569] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
INTRODUCTION Growing exposure of human skin to environmental and occupational hazards, to numerous skin care/beauty products, and to topical drugs led to a biomedical concern regarding sustainability of cutaneous chemical defence that is essential for protection against intoxication. Since skin is the largest extra-hepatic drug/xenobiotic metabolising organ where redox-dependent metabolic pathways prevail, in this review, publications on metabolic processes leading to redox imbalance (oxidative stress) and its autocrine/endocrine impact to cutaneous drug/xenobiotic metabolism were scrutinised. AREAS COVERED Chemical and photo-chemical skin barriers contain metabolic and redox compartments: their protective and homeostatic functions. The review will examine the striking similarity of adaptive responses to exogenous chemical/photo-chemical stressors and endogenous toxins in cutaneous metabolic and redox system; the role(s) of xenobiotics/drugs and phase II enzymes in the endogenous antioxidant defence and maintenance of redox balance; redox regulation of interactions between metabolic and inflammatory responses in skin cells; skin diseases sharing metabolic and redox problems (contact dermatitis, lupus erythematosus, and vitiligo) EXPERT OPINION Due to exceptional the redox dependence of cutaneous metabolic pathways and interaction of redox active metabolites/exogenous antioxidants with drug/xenobiotic metabolism, metabolic tests of topical xenobiotics/drugs should be combined with appropriate redox analyses and performed on 3D human skin models.
Collapse
Affiliation(s)
- Liudmila Korkina
- a Scientific Direction, Centre for Innovative Biotechnological Investigations 'NANOLAB' , Moscow , Russia
| |
Collapse
|
646
|
Köpp J, Fleßa S, Lieb W, Markus MRP, Teumer A, Homuth G, Wallaschofski H, Marschall P, Völzke H, Baumeister SE. Association of PNPLA3 rs738409 and TM6SF2 rs58542926 with health services utilization in a population-based study. BMC Health Serv Res 2016; 16:41. [PMID: 26847197 PMCID: PMC4741011 DOI: 10.1186/s12913-016-1289-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 01/29/2016] [Indexed: 01/04/2023] Open
Abstract
Background Hepatic steatosis confers an increased risk of metabolic and cardiovascular disease and higher health services use. Associations of the single nucleotide polymorphisms (SNP) PNPLA3 rs738409 and TM6SF2 rs58542926 with hepatic steatosis have recently been established. This study investigates the association between rs738409 and rs58542926 with health services utilization in a general population. Methods Data of 3759 participants from Study of Health in Pomerania (SHIP), a population-based study in Germany, were obtained. The annual number of outpatient visits, hospitalization and length of hospital stay was regressed on rs738409 and rs58542926 and adjusted for socio-economic factors, lifestyle habits, clinical factors, and health status. Results Minor allele homozygous subjects of rs738409 had an increased odds of hospitalization as compared to major allele homozygous subjects (odds ratio [OR] 1.51; 95 % confidence interval [CI], 1.02 to 2.15). Heterozygous subjects did not differ from major allele homozygous subjects with respect to their odds of hospitalization. The three genotype groups of rs738409 were similar with respect to the number of outpatient visits and inpatient days. Minor allele homozygous and heterozygous subjects of rs58542926 had higher outpatient utilization (+53.04 % and +67.56 %, p < 0.05, respectively) and inpatient days than major allele homozygous subjects. Conclusions After adjustment for several confounding factors, PNPLA3 rs738409 and TM6SF2 rs58542926 were associated with the number of outpatient visits, hospitalization, and inpatient days. Further studies are warranted to replicate our findings and to evaluate whether genetic data can be used to identify subjects with excess health services utilization.
Collapse
Affiliation(s)
- Julia Köpp
- Department of Community Medicine, University Medicine Greifswald, Walther-Rathenau-Straße 48, 17475, Greifswald, Germany
| | - Steffen Fleßa
- Department of Health Care Management, Ernst Moritz Arndt University of Greifswald, Friedrich-Loeffler-Straße 70, 17489, Greifswald, Germany
| | - Wolfgang Lieb
- Institute for Epidemiology, Christian Albrechts University, Niemannsweg 11, 24105, Kiel Kiel, Germany
| | | | - Alexander Teumer
- Department of Community Medicine, University Medicine Greifswald, Walther-Rathenau-Straße 48, 17475, Greifswald, Germany
| | - Georg Homuth
- Department of Functional Genomics, Ernst Moritz Arndt University of Greifswald, Greifswald, Germany
| | - Henri Wallaschofski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Paul Marschall
- Department of Health Care Management, Ernst Moritz Arndt University of Greifswald, Friedrich-Loeffler-Straße 70, 17489, Greifswald, Germany
| | - Henry Völzke
- Department of Community Medicine, University Medicine Greifswald, Walther-Rathenau-Straße 48, 17475, Greifswald, Germany
| | - Sebastian Edgar Baumeister
- Department of Community Medicine, University Medicine Greifswald, Walther-Rathenau-Straße 48, 17475, Greifswald, Germany. .,Institute for Epidemiology and Preventive Medicine, University of Regensburg, Franz-Josef-Strauß-Allee 11, D-93053, Regensburg, Germany.
| |
Collapse
|
647
|
Perkins JT, Petriello MC, Newsome BJ, Hennig B. Polychlorinated biphenyls and links to cardiovascular disease. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:2160-72. [PMID: 25877901 PMCID: PMC4609220 DOI: 10.1007/s11356-015-4479-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/31/2015] [Indexed: 05/19/2023]
Abstract
The pathology of cardiovascular disease is multi-faceted, with links to many modifiable and non-modifiable risk factors. Epidemiological evidence now implicates exposure to persistent organic pollutants, such as polychlorinated biphenyls (PCBs), with an increased risk of developing diabetes, hypertension, and obesity; all of which are clinically relevant to the onset and progression of cardiovascular disease. PCBs exert their cardiovascular toxicity either directly or indirectly via multiple mechanisms, which are highly dependent on the type and concentration of PCBs present. However, many PCBs may modulate cellular signaling pathways leading to common detrimental outcomes including induction of chronic oxidative stress, inflammation, and endocrine disruption. With the abundance of potential toxic pollutants increasing globally, it is critical to identify sensible means of decreasing associated disease risks. Emerging evidence now implicates a protective role of lifestyle modifications such as increased exercise and/or nutritional modulation via anti-inflammatory foods, which may help to decrease the vascular toxicity of PCBs. This review will outline the current state of knowledge linking coplanar and non-coplanar PCBs to cardiovascular disease and describe the possible molecular mechanism of this association.
Collapse
Affiliation(s)
- Jordan T Perkins
- Superfund Research Center, University of Kentucky, 900 S. Limestone Street, Lexington, KY, 40536, USA
- Department of Animal and Food Sciences, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY, 40536, USA
| | - Michael C Petriello
- Superfund Research Center, University of Kentucky, 900 S. Limestone Street, Lexington, KY, 40536, USA
- Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Bradley J Newsome
- Superfund Research Center, University of Kentucky, 900 S. Limestone Street, Lexington, KY, 40536, USA
- Department of Animal and Food Sciences, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY, 40536, USA
| | - Bernhard Hennig
- Superfund Research Center, University of Kentucky, 900 S. Limestone Street, Lexington, KY, 40536, USA.
- Department of Animal and Food Sciences, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY, 40536, USA.
- Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
648
|
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
|
649
|
Patel J, Yopp A, Waljee A, Singal AG. Development and Internal Validation of a Model for Early Detection of Hepatocellular Carcinoma in Patients With Cirrhosis. J Clin Gastroenterol 2016; 50:175-9. [PMID: 26125462 PMCID: PMC4691224 DOI: 10.1097/mcg.0000000000000377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Surveillance for hepatocellular carcinoma (HCC) is recommended in patients with cirrhosis; however, early detection efforts are limited by suboptimal effectiveness. AIM To derive and validate a model to accurately distinguish cirrhotic patients with and without HCC and compare the accuracy of the model to that of α-fetoprotein (AFP) alone. METHODS We conducted a case-control study of cirrhotic patients with and without HCC seen at a large urban hospital system between January 2005 and June 2012. We derived multivariate logistic regression models for the presence of HCC and early-stage HCC. Discriminatory power was evaluated using receiver operating characteristic curve analysis in derivation and validation cohorts using a 10-fold cross-validation approach. RESULTS We identified 1356 patients with cirrhosis, with (n=455, 147 early stage) and without (n=901) HCC. We found that AFP>20 ng/mL and FIB-4, a noninvasive marker of fibrosis, were significantly associated with the presence of HCC (OR=10.5; 95% CI, 7.9-13.9 and OR=1.05; 95% CI, 1.03-1.07, respectively) and early-stage HCC (OR=4.4; 95% CI, 2.9-6.5 and OR=1.06; 95% CI, 1.03-1.09, respectively). Models incorporating AFP and FIB-4 had good discriminatory power, with c-statistics of approximately 0.80, in both derivation and validation cohorts. The model for early-stage HCC had higher discriminatory power than AFP alone (c-statistic 0.73; 95% CI, 0.69-0.78) in derivation and validation cohorts (P=0.02 and 0.15, respectively). CONCLUSIONS Models including AFP and FIB-4 can accurately discriminate cirrhotic patients with early-stage HCC from those without HCC.
Collapse
Affiliation(s)
- Jaimin Patel
- Department of Internal Medicine, UT Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX
| | - Adam Yopp
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX
- Harold C. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, TX
| | - Akbar Waljee
- VA Center for Clinical Management Research, Ann Arbor, MI
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI
| | - Amit G. Singal
- Department of Internal Medicine, UT Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX
- Harold C. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
650
|
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
|