51
|
Kaltenecker D, Themanns M, Mueller KM, Spirk K, Golob-Schwarzl N, Friedbichler K, Kenner L, Haybaeck J, Moriggl R. STAT5 deficiency in hepatocytes reduces diethylnitrosamine-induced liver tumorigenesis in mice. Cytokine 2018; 124:154573. [PMID: 30377054 DOI: 10.1016/j.cyto.2018.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/03/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
Abstract
Chronic liver diseases and the development of hepatocellular carcinoma are closely linked and pose a major medical challenge as treatment options are limited. Animal studies have shown that genetic deletion of the signal transducer and activator of transcription (STAT) 5 in liver is associated with higher susceptibility to fatty liver disease, fibrosis and cancer, indicating a protective role of hepatic STAT5 in mouse models of chronic liver disease. To investigate the role of STAT5 in the etiology of liver cancer in more detail, we applied the chemical carcinogen diethylnitrosamine (DEN) to mice harboring a hepatocyte-specific deletion of Stat5 (S5KO). At 8 months after DEN injections, tumor formation in S5KO was significantly reduced. This was associated with diminished tumor frequency and less aggressive liver cancer progression. Apoptosis and inflammation markers were not changed in S5KO livers suggesting that the reduced tumor burden was not due to impaired inflammatory response. Despite reduced mRNA expression of the DEN bio-activator cytochrome P450 2e1 (Cyp2e1) in S5KO livers, protein levels were similar. Yet, delayed tumor formation in S5KO mice coincided with decreased activation of c-Jun N-terminal Kinase (JNK). Taken together, while STAT5 has a protective role in fatty liver-associated liver cancer, it exerts oncogenic functions in DEN-induced liver cancer.
Collapse
Affiliation(s)
- Doris Kaltenecker
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Madeleine Themanns
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Kristina M Mueller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Katrin Spirk
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Nicole Golob-Schwarzl
- Center for Biomarker Research in Medicine, Graz, Austria; Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Lukas Kenner
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria; Department of Clinical Pathology, Medical University of Vienna, Vienna, Austria; Unit of Pathology of Laboratory Animals, University of Veterinary Medicine, Vienna, Austria; CBMed Core Lab2, Medical University of Vienna, Vienna, Austria
| | - Johannes Haybaeck
- Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, Graz, Austria; Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Department of Pathology, Medical University of Innsbruck, Innsbruck, Austria
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria; Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
52
|
Yan T, Wang H, Cao L, Wang Q, Takahashi S, Yagai T, Li G, Krausz KW, Wang G, Gonzalez FJ, Hao H. Glycyrrhizin Alleviates Nonalcoholic Steatohepatitis via Modulating Bile Acids and Meta-Inflammation. Drug Metab Dispos 2018; 46:1310-1319. [PMID: 29959134 PMCID: PMC6081736 DOI: 10.1124/dmd.118.082008] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/27/2018] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the progressive stage of nonalcoholic fatty liver disease that may ultimately lead to cirrhosis and liver cancer, and there are few therapeutic options for its treatment. Glycyrrhizin (GL), extracted from the traditional Chinese medicine liquorice, has potent hepatoprotective effects in both preclinical animal models and in humans. However, little is currently known about its effects and mechanisms in treating NASH. To explore the effects of GL on NASH, GL or its active metabolite glycyrrhetinic acid (GA) was administered to mice treated with a methionine- and choline-deficient (MCD) diet-induced NASH model, and histologic and biochemical analyses were used to measure the degree of lipid disruption, liver inflammation, and fibrosis. GL significantly improved MCD diet-induced hepatic steatosis, inflammation, and fibrosis and inhibited activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome. GL significantly attenuated serum bile acid accumulation in MCD diet-fed mice partially by restoring inflammation-mediated hepatic farnesoid X receptor inhibition. In Raw 264.7 macrophage cells, both GL and GA inhibited deoxycholic acid-induced NLRP3 inflammasome-associated inflammation. Notably, both intraperitoneal injection of GL's active metabolite GA and oral administration of GL prevented NASH in mice, indicating that GL may attenuate NASH via its active metabolite GA. These results reveal that GL, via restoration of bile acid homeostasis and inhibition of inflammatory injury, can be a therapeutic option for treatment of NASH.
Collapse
Affiliation(s)
- Tingting Yan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Hong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Lijuan Cao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Qiong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Shogo Takahashi
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Tomoki Yagai
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Guolin Li
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Kristopher W Krausz
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Frank J Gonzalez
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| |
Collapse
|
53
|
Schuette D, Moore LM, Robert ME, Taddei TH, Ehrlich BE. Hepatocellular Carcinoma Outcome Is Predicted by Expression of Neuronal Calcium Sensor 1. Cancer Epidemiol Biomarkers Prev 2018; 27:1091-1100. [PMID: 29789326 PMCID: PMC8465775 DOI: 10.1158/1055-9965.epi-18-0167] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/06/2018] [Accepted: 05/17/2018] [Indexed: 11/16/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide. There is an urgent demand for prognostic biomarkers that facilitate early tumor detection, as the incidence of HCC has tripled in the United States in the last three decades. Biomarkers to identify populations at risk would have significant impact on survival. We recently found that expression of Neuronal Calcium Sensor 1 (NCS1), a Ca2+-dependent signaling molecule, predicted disease outcome in breast cancer, but its predictive value in other cancer types is unknown. This protein is potentially useful because increased NCS1 regulates Ca2+ signaling and increased Ca2+ signaling is a hallmark of metastatic cancers, conferring cellular motility and an increasingly aggressive phenotype to tumors.Methods: We explored the relationship between NCS1 expression levels and patient survival in two publicly available liver cancer cohorts and a tumor microarray using data mining strategies.Results: High NCS1 expression levels are significantly associated with worse disease outcome in Asian patients within these cohorts. In addition, a variety of Ca2+-dependent and tumor growth-promoting genes are transcriptionally coregulated with NCS1 and many of them are involved in cytoskeleton organization, suggesting that NCS1 induced dysregulated Ca2+ signaling facilitates cellular motility and metastasis.Conclusions: We found NCS1 to be a novel biomarker in HCC. Furthermore, our study identified a pharmacologically targetable signaling complex that can influence tumor progression in HCC.Impact: These results lay the foundation for using NCS1 as a prognostic biomarker in prospective cohorts of HCC patients and for further functional assessment of the characterized signaling axis. Cancer Epidemiol Biomarkers Prev; 27(9); 1091-100. ©2018 AACR.
Collapse
Affiliation(s)
- Daniel Schuette
- Department of Pharmacology, Yale University, New Haven, Connecticut
| | - Lauren M Moore
- Department of Pharmacology, Yale University, New Haven, Connecticut
| | - Marie E Robert
- Department of Pathology, Yale University, New Haven, Connecticut
| | - Tamar H Taddei
- Department of Medicine (Digestive Diseases), Yale University, New Haven, Connecticut
| | - Barbara E Ehrlich
- Department of Pharmacology, Yale University, New Haven, Connecticut.
| |
Collapse
|
54
|
Huang B, Bao J, Cao YR, Gao HF, Jin Y. Cytochrome P450 1A1 (CYP1A1) Catalyzes Lipid Peroxidation of Oleic Acid-Induced HepG2 Cells. BIOCHEMISTRY (MOSCOW) 2018; 83:595-602. [PMID: 29738693 DOI: 10.1134/s0006297918050127] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic hepatic disease associated with excessive accumulation of lipids in hepatocytes. As the disease progresses, oxidative stress plays a pivotal role in the development of hepatic lipid peroxidation. Cytochrome P450 1A1 (CYP1A1), a subtype of the cytochrome P450 family, has been shown to be a vital modulator in production of reactive oxygen species. However, the exact role of CYP1A1 in NAFLD is still unclear. The aim of this study was to investigate the effects of CYP1A1 on lipid peroxidation in oleic acid (OA)-treated human hepatoma cells (HepG2). We found that the expression of CYP1A1 is elevated in OA-stimulated HepG2 cells. The results of siRNA transfection analysis indicated that CYP1A1-siRNA inhibited the lipid peroxidation in OA-treated HepG2 cells. Additionally, compared with siRNA-transfected and benzo[a]pyrene (BaP)-OA-induced HepG2 cells, overexpression of CYP1A1 by BaP further accelerated the lipid peroxidation in OA-treated HepG2 cells. These observations reveal a regulatory role of CYP1A1 in liver lipid peroxidation and imply CYP1A1 as a potential therapeutic target.
Collapse
Affiliation(s)
- B Huang
- Key Laboratory of Antiinflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - J Bao
- Key Laboratory of Antiinflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Y-R Cao
- Key Laboratory of Antiinflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - H-F Gao
- Key Laboratory of Antiinflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Y Jin
- Key Laboratory of Antiinflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
55
|
Iogna Prat L, Tsochatzis EA. The effect of antidiabetic medications on non-alcoholic fatty liver disease (NAFLD). Hormones (Athens) 2018; 17:219-229. [PMID: 29858843 DOI: 10.1007/s42000-018-0021-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 02/21/2018] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome and is prevalent in more than 50% of patients with type II diabetes. At present, there is no approved therapy for NASH. Until now, the only proven effective interventions in improving biochemical and histological features of NASH, including fibrosis, are weight loss and physical activity even without weight loss. Because of the common epidemiological and pathophysiological features between NAFLD and T2DM, many antidiabetics drugs have been tested in patients with NAFLD over the years. Among these, pioglitazone and liraglutide seem to improve some histological features of NASH but have no clear effect on fibrosis. Metformin has been largely studied in the past years without convincing evidence of improving NAFLD. Data on other compounds such as DDP-4 and SGLT-2 inhibitors are limited. The rational and results of such studies are discussed in the present review.
Collapse
Affiliation(s)
- Laura Iogna Prat
- UCL Institute for Liver and Digestive Health, Royal Free Hospital and UCL, London, UK
| | - Emmanuel A Tsochatzis
- UCL Institute for Liver and Digestive Health, Royal Free Hospital and UCL, London, UK.
| |
Collapse
|
56
|
Bucher S, Tête A, Podechard N, Liamin M, Le Guillou D, Chevanne M, Coulouarn C, Imran M, Gallais I, Fernier M, Hamdaoui Q, Robin MA, Sergent O, Fromenty B, Lagadic-Gossmann D. Co-exposure to benzo[a]pyrene and ethanol induces a pathological progression of liver steatosis in vitro and in vivo. Sci Rep 2018; 8:5963. [PMID: 29654281 PMCID: PMC5899096 DOI: 10.1038/s41598-018-24403-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023] Open
Abstract
Hepatic steatosis (i.e. lipid accumulation) and steatohepatitis have been related to diverse etiologic factors, including alcohol, obesity, environmental pollutants. However, no study has so far analyzed how these different factors might interplay regarding the progression of liver diseases. The impact of the co-exposure to the environmental carcinogen benzo[a]pyrene (B[a]P) and the lifestyle-related hepatotoxicant ethanol, was thus tested on in vitro models of steatosis (human HepaRG cell line; hybrid human/rat WIF-B9 cell line), and on an in vivo model (obese zebrafish larvae). Steatosis was induced prior to chronic treatments (14, 5 or 7 days for HepaRG, WIF-B9 or zebrafish, respectively). Toxicity and inflammation were analyzed in all models; the impact of steatosis and ethanol towards B[a]P metabolism was studied in HepaRG cells. Cytotoxicity and expression of inflammation markers upon co-exposure were increased in all steatotic models, compared to non steatotic counterparts. A change of B[a]P metabolism with a decrease in detoxification was detected in HepaRG cells under these conditions. A prior steatosis therefore enhanced the toxicity of B[a]P/ethanol co-exposure in vitro and in vivo; such a co-exposure might favor the appearance of a steatohepatitis-like state, with the development of inflammation. These deleterious effects could be partly explained by B[a]P metabolism alterations.
Collapse
Affiliation(s)
- Simon Bucher
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Arnaud Tête
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Marie Liamin
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Dounia Le Guillou
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Martine Chevanne
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Cédric Coulouarn
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Muhammad Imran
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Isabelle Gallais
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Morgane Fernier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Quentin Hamdaoui
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Marie-Anne Robin
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Odile Sergent
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Bernard Fromenty
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
57
|
Meng C, Bai C, Brown TD, Hood LE, Tian Q. Human Gut Microbiota and Gastrointestinal Cancer. GENOMICS PROTEOMICS & BIOINFORMATICS 2018. [PMID: 29474889 DOI: 10.1016/j.gpb.2017.06.002.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Human gut microbiota play an essential role in both healthy and diseased states of humans. In the past decade, the interactions between microorganisms and tumors have attracted much attention in the efforts to understand various features of the complex microbial communities, as well as the possible mechanisms through which the microbiota are involved in cancer prevention, carcinogenesis, and anti-cancer therapy. A large number of studies have indicated that microbial dysbiosis contributes to cancer susceptibility via multiple pathways. Further studies have suggested that the microbiota and their associated metabolites are not only closely related to carcinogenesis by inducing inflammation and immune dysregulation, which lead to genetic instability, but also interfere with the pharmacodynamics of anticancer agents. In this article, we mainly reviewed the influence of gut microbiota on cancers in the gastrointestinal (GI) tract (including esophageal, gastric, colorectal, liver, and pancreatic cancers) and the regulation of microbiota by diet, prebiotics, probiotics, synbiotics, antibiotics, or the Traditional Chinese Medicine. We also proposed some new strategies in the prevention and treatment of GI cancers that could be explored in the future. We hope that this review could provide a comprehensive overview of the studies on the interactions between the gut microbiota and GI cancers, which are likely to yield translational opportunities to reduce cancer morbidity and mortality by improving prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Changting Meng
- Institute for Systems Biology, Seattle, WA 98109, USA; Department of Oncology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Beijing 100730, China
| | | | - Leroy E Hood
- Institute for Systems Biology, Seattle, WA 98109, USA; Swedish Cancer Institute, Seattle, WA 98104, USA
| | - Qiang Tian
- Institute for Systems Biology, Seattle, WA 98109, USA; P4 Medicine Institute, Seattle, WA 98109, USA.
| |
Collapse
|
58
|
Porcu C, Antonucci L, Barbaro B, Illi B, Nasi S, Martini M, Licata A, Miele L, Grieco A, Balsano C. Copper/MYC/CTR1 interplay: a dangerous relationship in hepatocellular carcinoma. Oncotarget 2018; 9:9325-9343. [PMID: 29507693 PMCID: PMC5823635 DOI: 10.18632/oncotarget.24282] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023] Open
Abstract
Free serum copper correlates with tumor incidence and progression of human cancers, including hepatocellular carcinoma (HCC). Copper extracellular uptake is provided by the transporter CTR1, whose expression is regulated to avoid excessive intracellular copper entry. Inadequate copper serum concentration is involved in the pathogenesis of Non Alcoholic Fatty Liver Disease (NAFLD), which is becoming a major cause of liver damage progression and HCC incidence. Finally, MYC is over-expressed in most of HCCs and is a critical regulator of cellular growth, tumor invasion and metastasis. The purpose of our study was to understand if higher serum copper concentrations might be involved in the progression of NAFLD-cirrhosis toward-HCC. We investigated whether high exogenous copper levels sensitize liver cells to transformation and if it exists an interplay between copper-related proteins and MYC oncogene. NAFLD-cirrhotic patients were characterized by a statistical significant enhancement of serum copper levels, even more evident in HCC patients. We demonstrated that high extracellular copper concentrations increase cell growth, migration, and invasion of liver cancer cells by modulating MYC/CTR1 axis. We highlighted that MYC binds a specific region of the CTR1 promoter, regulating its transcription. Accordingly, CTR1 and MYC proteins expression were progressively up-regulated in liver tissues from NAFLD-cirrhotic to HCC patients. This work provides novel insights on the molecular mechanisms by which copper may favor the progression from cirrhosis to cancer. The Cu/MYC/CTR1 interplay opens a window to refine HCC diagnosis and design new combined therapies.
Collapse
Affiliation(s)
- Cristiana Porcu
- MESVA Department, University of L’Aquila, L’Aquila, Italy
- F. Balsano Foundation, Rome, Italy
| | - Laura Antonucci
- MESVA Department, University of L’Aquila, L’Aquila, Italy
- F. Balsano Foundation, Rome, Italy
| | - Barbara Barbaro
- MESVA Department, University of L’Aquila, L’Aquila, Italy
- F. Balsano Foundation, Rome, Italy
| | - Barbara Illi
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Sergio Nasi
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Maurizio Martini
- Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Licata
- DIBIMIS, University of Palermo, School of Medicine, Palermo, Italy
| | - Luca Miele
- Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Grieco
- Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Clara Balsano
- MESVA Department, University of L’Aquila, L’Aquila, Italy
- F. Balsano Foundation, Rome, Italy
| |
Collapse
|
59
|
Esterson YB, Grimaldi GM. Radiologic Imaging in Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Clin Liver Dis 2018; 22:93-108. [PMID: 29128063 DOI: 10.1016/j.cld.2017.08.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The article reviews the multimodality (ultrasound, computed tomography, and magnetic resonance [MR]) imaging appearance of nonalcoholic fatty liver disease (NAFLD) and discusses the radiologic diagnostic criteria as well as the sensitivity and specificity of these imaging methods. The authors review the role of both ultrasound and MR elastography for the diagnosis of fibrosis and for the longitudinal evaluation of patients following therapeutic intervention. Lastly, the authors briefly discuss the screening and diagnosis of hepatocellular carcinoma in patients with NAFLD, as there are special considerations in this population.
Collapse
Affiliation(s)
- Yonah B Esterson
- Department of Radiology, Northwell Health System, Hofstra Northwell School of Medicine, 300 Community Drive, Manhasset, NY 11030, USA.
| | - Gregory M Grimaldi
- Department of Radiology, Northwell Health System, Hofstra Northwell School of Medicine, 300 Community Drive, Manhasset, NY 11030, USA
| |
Collapse
|
60
|
Meng C, Bai C, Brown TD, Hood LE, Tian Q. Human Gut Microbiota and Gastrointestinal Cancer. GENOMICS, PROTEOMICS & BIOINFORMATICS 2018; 16:33-49. [PMID: 29474889 PMCID: PMC6000254 DOI: 10.1016/j.gpb.2017.06.002] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 06/08/2017] [Accepted: 07/04/2017] [Indexed: 02/06/2023]
Abstract
Human gut microbiota play an essential role in both healthy and diseased states of humans. In the past decade, the interactions between microorganisms and tumors have attracted much attention in the efforts to understand various features of the complex microbial communities, as well as the possible mechanisms through which the microbiota are involved in cancer prevention, carcinogenesis, and anti-cancer therapy. A large number of studies have indicated that microbial dysbiosis contributes to cancer susceptibility via multiple pathways. Further studies have suggested that the microbiota and their associated metabolites are not only closely related to carcinogenesis by inducing inflammation and immune dysregulation, which lead to genetic instability, but also interfere with the pharmacodynamics of anticancer agents. In this article, we mainly reviewed the influence of gut microbiota on cancers in the gastrointestinal (GI) tract (including esophageal, gastric, colorectal, liver, and pancreatic cancers) and the regulation of microbiota by diet, prebiotics, probiotics, synbiotics, antibiotics, or the Traditional Chinese Medicine. We also proposed some new strategies in the prevention and treatment of GI cancers that could be explored in the future. We hope that this review could provide a comprehensive overview of the studies on the interactions between the gut microbiota and GI cancers, which are likely to yield translational opportunities to reduce cancer morbidity and mortality by improving prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Changting Meng
- Institute for Systems Biology, Seattle, WA 98109, USA; Department of Oncology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Beijing 100730, China
| | | | - Leroy E Hood
- Institute for Systems Biology, Seattle, WA 98109, USA; Swedish Cancer Institute, Seattle, WA 98104, USA
| | - Qiang Tian
- Institute for Systems Biology, Seattle, WA 98109, USA; P4 Medicine Institute, Seattle, WA 98109, USA.
| |
Collapse
|
61
|
Rodriguez-Echevarria R, Macias-Barragan J, Parra-Vargas M, Davila-Rodriguez JR, Amezcua-Galvez E, Armendariz-Borunda J. Diet switch and omega-3 hydroxy-fatty acids display differential hepatoprotective effects in an obesity/nonalcoholic fatty liver disease model in mice. World J Gastroenterol 2018; 24:461-474. [PMID: 29398867 PMCID: PMC5787781 DOI: 10.3748/wjg.v24.i4.461] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To study the effect of 18-hydroxy-eicosapentaenoic acid (18-HEPE) and 17-hydroxy-docosahexaenoic acid (17-HDHA) in a murine model of obesity/nonalcoholic fatty liver disease.
METHODS C57BL/6 mice were fed with standard chow diet (CD) or high-fat, fructose-enriched diet (HFD) for 16 wk. Then, three groups were treated for 14 d with either, diet switch (HFD for CD), 18-HEPE, or 17-HDHA. Weight and fasting glucose were recorded on a weekly basis. Insulin tolerance test was performed at the end of treatment. Histological analysis (HE and Masson’s trichrome stain) and determination of serum insulin, glucagon, glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic polypeptide, adiponectin and resistin were carried out as well as liver proteins by western blot.
RESULTS Mice treated with hydroxy-fatty acids 18-HEPE and 17-HDHA displayed no weight loss or improved insulin sensitivity. However, these mice groups showed a significant amelioration on serum GLP-1, adiponectin and resistin levels. Also, a significant reduction on inflammatory infiltrate was observed at both portal and lobular zones. Furthermore, up-regulation of PPARα/γ protein levels was observed in liver tissue and it was associated with decreased levels of NF-κB also determined by western blot analysis. On the other hand, diet switch regimen resulted in a marked improvement in most parameters including: weight loss, increased insulin sensitivity, decreased steatosis, restored levels of insulin, glucagon, leptin, adiponectin and resistin. However, no significant changes were observed regarding inflammatory infiltrate in this last group.
CONCLUSION 18-HEPE and 17-HDHA differentially exert hepatoprotective effects through up-regulation of nuclear receptors PPARα/γ and amelioration of serum adipokines profile.
Collapse
Affiliation(s)
- Roberto Rodriguez-Echevarria
- Institute for Molecular Biology and Gene Therapy-CUCS, Department of Molecular Biology and Genomics, University of Guadalajara, Guadalajara 44340, Mexico
| | - Jose Macias-Barragan
- Department of Health Sciences-CUValles, University of Guadalajara, Guadalajara 46600, Mexico
| | - Marcela Parra-Vargas
- Institute for Molecular Biology and Gene Therapy-CUCS, Department of Molecular Biology and Genomics, University of Guadalajara, Guadalajara 44340, Mexico
| | | | | | - Juan Armendariz-Borunda
- Institute for Molecular Biology and Gene Therapy-CUCS, Department of Molecular Biology and Genomics, University of Guadalajara, Guadalajara 44340, Mexico
| |
Collapse
|
62
|
Nonalcoholic fatty liver disease: epidemiology, pathogenesis and therapeutic implications. J Med Life 2018; 11:20-23. [PMID: 29696060 PMCID: PMC5909941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The increase of the prevalence of nonalcoholic fatty liver disease in the context of the explosive epidemy of obesity worldwide over the last decades and the emergence of new effective therapies for viral hepatitis has brought this entity to the forefront of hepatologist concerns. Today is a certainty that fatty liver can complicate with cirrhosis and hepatocellular carcinoma; moreover, nonalcoholic fatty liver disease is the main cause of cryptogenic cirrhosis and the second cause of liver transplantation. This review revises the epidemiology of the disease, brings forward some progress in pathogenesis and outlines the directions to be followed in nonalcoholic fatty liver disease prevention and therapy. Today, nonalcoholic fatty liver disease is considered to be the liver manifestation of metabolic syndome, with its same prevalence (20-30%). If the patients do not die through cardiovascular disease, it can lead to serious liver complications.
Collapse
|
63
|
Perumpail BJ, Khan MA, Yoo ER, Cholankeril G, Kim D, Ahmed A. Clinical epidemiology and disease burden of nonalcoholic fatty liver disease. World J Gastroenterol 2017; 23:8263-8276. [PMID: 29307986 PMCID: PMC5743497 DOI: 10.3748/wjg.v23.i47.8263] [Citation(s) in RCA: 503] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/08/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is defined as the presence of hepatic fat accumulation after the exclusion of other causes of hepatic steatosis, including other causes of liver disease, excessive alcohol consumption, and other conditions that may lead to hepatic steatosis. NAFLD encompasses a broad clinical spectrum ranging from nonalcoholic fatty liver to nonalcoholic steatohepatitis (NASH), advanced fibrosis, cirrhosis, and finally hepatocellular carcinoma (HCC). NAFLD is the most common liver disease in the world and NASH may soon become the most common indication for liver transplantation. Ongoing persistence of obesity with increasing rate of diabetes will increase the prevalence of NAFLD, and as this population ages, many will develop cirrhosis and end-stage liver disease. There has been a general increase in the prevalence of NAFLD, with Asia leading the rise, yet the United States is following closely behind with a rising prevalence from 15% in 2005 to 25% within 5 years. NAFLD is commonly associated with metabolic comorbidities, including obesity, type II diabetes, dyslipidemia, and metabolic syndrome. Our understanding of the pathophysiology of NAFLD is constantly evolving. Based on NAFLD subtypes, it has the potential to progress into advanced fibrosis, end-stage liver disease and HCC. The increasing prevalence of NAFLD with advanced fibrosis, is concerning because patients appear to experience higher liver-related and non-liver-related mortality than the general population. The increased morbidity and mortality, healthcare costs and declining health related quality of life associated with NAFLD makes it a formidable disease, and one that requires more in-depth analysis.
Collapse
Affiliation(s)
- Brandon J Perumpail
- Department of Medicine, College of Medicine, Drexel University, Philadelphia, PA 19129, United States
| | - Muhammad Ali Khan
- Division of Gastroenterology and Hepatology, Health Science Center, University of Tennessee, Memphis, TN 38163, United States
| | - Eric R Yoo
- Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA 95128, United States
| | - George Cholankeril
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Palo Alto, CA 94304, United States
| | - Donghee Kim
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Palo Alto, CA 94304, United States
| | - Aijaz Ahmed
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Palo Alto, CA 94304, United States
| |
Collapse
|
64
|
Wong CR, Njei B, Nguyen MH, Nguyen A, Lim JK. Survival after treatment with curative intent for hepatocellular carcinoma among patients with vs without non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2017; 46:1061-1069. [PMID: 28960360 DOI: 10.1111/apt.14342] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/16/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is expected to become a leading aetiology of hepatocellular carcinoma (HCC)-related mortality in the United States. HCC treatments with curative intent (OLT, orthotopic liver transplantation; resection; RFA, radiofrequency ablation) can improve survival in carefully selected patients. AIM To compare survival after receipt of curative treatment for NAFLD and non-NAFLD-HCC aetiologies (HCV, chronic hepatitis C; HBV, chronic hepatitis B; ALD, alcoholic liver disease) and by treatment was performed. METHODS A cohort of 17 664 patients was assembled using linked Surveillance, Epidemiology, and End Results and Medicare data from 1991 to 2011 with confirmed diagnosis of HCC. RESULTS The cohort was mostly male, aged 70 (21-106) years, without cardiovascular disease, and had liver cirrhosis without decompensation, metastatic HCC or large tumour size (>5 cm). The NAFLD-HCC group was mostly female and older with more cardiovascular disease, metastatic HCC, and large tumour size and less cirrhosis and decompensated liver disease than the non-NAFLD-HCC groups. The NAFLD group was 47% less likely to receive any curative treatment as compared with non-NAFLD aetiologies (OR 0.53, P < .001). NAFLD-HCC had worse median survival after OLT (3.2, 0-12.9 years, P = .01) but had improved survival after resection (2.4, 0-12.0 years, P < .001) as compared with non-NAFLD-HCC. No significant survival differences existed for RFA by HCC aetiology. NAFLD was not an independent predictor of mortality after OLT, resection or RFA. CONCLUSION Patients with NAFLD-HCC had worse survival after OLT but favourable survival after resection, particularly in the absence of cirrhosis, as compared with non-NAFLD-HCC aetiologies.
Collapse
Affiliation(s)
- C R Wong
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - B Njei
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - M H Nguyen
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - A Nguyen
- Weill Cornell Medical College, New York, NY, USA
| | - J K Lim
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
65
|
Jaruvongvanich V, Wijarnpreecha K, Ungprasert P. The utility of NAFLD fibrosis score for prediction of mortality among patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis of cohort study. Clin Res Hepatol Gastroenterol 2017; 41:629-634. [PMID: 28716600 DOI: 10.1016/j.clinre.2017.03.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/04/2017] [Accepted: 03/28/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a common liver disorder worldwide. Several noninvasive diagnostic scoring systems have been developed to determine the severity of liver fibrosis and to predict long-term outcome of patients with NAFLD in lieu of liver biopsy. We conducted this systematic review and meta-analysis to investigate the role of NAFLD fibrosis score (NFS) for prediction of mortality from NAFLD. METHODS MEDLINE and EMBASE databases were searched through October 2016 for studies that investigated the association between high NFS and mortality. Pooled risk ratio (RR) and 95% confidence interval (CI) were calculated using a random-effects model, generic inverse variance method. The between-study heterogeneity of effect-size was quantified using the Q statistic and I2. RESULTS A total of five cohort studies with 5033 NAFLD patients were identified. High NFS (score of greater than 0.675) was significantly associated with increased mortality with the pooled RR of 4.54 (95%CI: 1.85-11.17). The statistical heterogeneity was high with I2 of 88% (Pheterogeneity<0.01). CONCLUSIONS High NFS is associated with increased risk of mortality among patients with NAFLD. This scoring system may be considered as an alternative to liver biopsy for prediction of mortality outcome.
Collapse
Affiliation(s)
- Veeravich Jaruvongvanich
- Department of Internal Medicine, University of Hawaii, Honolulu, HI, USA; Department of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand.
| | - Karn Wijarnpreecha
- Department of Internal Medicine, Bassett Medical Center and Columbia University College of Physicians and Surgeons, Cooperstown, NY, USA.
| | - Patompong Ungprasert
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA; Division of Rheumatology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
66
|
Royse KE, El-Serag HB, Chen L, White DL, Hale L, Sangi-Haghpeykar H, Jiao L. Sleep Duration and Risk of Liver Cancer in Postmenopausal Women: The Women's Health Initiative Study. J Womens Health (Larchmt) 2017; 26:1270-1277. [PMID: 28933583 PMCID: PMC6037184 DOI: 10.1089/jwh.2017.6412] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Sleep duration has been associated with nonalcoholic fatty liver disease, but its association with liver cancer remains unknown. MATERIAL AND METHODS In the prospective Women's Health Initiative Study, 139,368 postmenopausal women reported sleep habits at baseline (1993-1998). We ascertained 175 incident liver cancer cases during an average 13.8 years of follow-up through August 2014. We used multivariable Cox proportional hazard regression models to estimate a hazard ratio (HR) and its 95% confidence interval (95% CI) for risk of liver cancer in association with nocturnal sleep duration. RESULTS Compared to women reporting 6-8 hours of sleep, the HR for liver cancer was 1.94 (95% CI 1.07-3.53) for women reporting ≥9 hours of sleep. Among the obese women, the HR associated with ≥9 hours of sleep was 3.18 (95% CI 1.84-8.60). The HR was 0.93 (95% CI 0.34-2.53) among nonobese women (p value for interaction = 0.18). Short sleep duration (≤5 hours) was not associated with liver cancer risk. CONCLUSION Long sleep duration was associated with a moderate increase in liver cancer risk in obese postmenopausal women in the United States. Larger study is needed to confirm our observation on effect modification by adiposity status.
Collapse
Affiliation(s)
- Kathryn E. Royse
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
- Center for Innovations in Quality, Effectiveness and Safety (iQuEST), Michael E. DeBakey VA Medical Center, Houston, Texas
| | - Hashem B. El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
- Center for Innovations in Quality, Effectiveness and Safety (iQuEST), Michael E. DeBakey VA Medical Center, Houston, Texas
- Texas Medical Center Digestive Disease Center, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Liang Chen
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
- Center for Innovations in Quality, Effectiveness and Safety (iQuEST), Michael E. DeBakey VA Medical Center, Houston, Texas
| | - Donna L. White
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
- Center for Innovations in Quality, Effectiveness and Safety (iQuEST), Michael E. DeBakey VA Medical Center, Houston, Texas
- Texas Medical Center Digestive Disease Center, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas
| | - Lauren Hale
- Program in Public Health, School of Medicine, Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | | | - Li Jiao
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
- Center for Innovations in Quality, Effectiveness and Safety (iQuEST), Michael E. DeBakey VA Medical Center, Houston, Texas
- Texas Medical Center Digestive Disease Center, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas
| |
Collapse
|
67
|
Affiliation(s)
- Anna M Diehl
- From the Department of Medicine, Duke University, Durham, NC (A.M.D); and Newcastle University Medical School, Newcastle upon Tyne, United Kingdom (C.D.)
| | - Christopher Day
- From the Department of Medicine, Duke University, Durham, NC (A.M.D); and Newcastle University Medical School, Newcastle upon Tyne, United Kingdom (C.D.)
| |
Collapse
|
68
|
Wu WY, Tao SQ, Wang XN, Lobie PE, Wu ZS. XIAP 3'-untranslated region serves as a competitor for HMGA2 by arresting endogenous let-7a-5p in human hepatocellular carcinoma. Tumour Biol 2017; 39:1010428317719578. [PMID: 28691642 DOI: 10.1177/1010428317719578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
X-linked inhibitor of apoptosis protein functions as an intrinsic regulator of apoptosis by inhibition of caspase activity and possesses a pivotal role in human cancer development and progression. A growing body of literature has demonstrated that microRNAs lead to the degradation or translational repression of messenger RNAs by binding to the non-coding region of messenger RNA at the 3'-untranslated region. Here, we revealed that the expression of HMGA2 is upregulated with X-linked inhibitor of apoptosis protein after transfection of X-linked inhibitor of apoptosis protein 3'-untranslated region in hepatocellular carcinoma cells, suggesting that X-linked inhibitor of apoptosis protein 3'-untranslated region serves as a competitor for microRNAs and prevent the co-targeted messenger RNA, HMGA2, from being suppressed. We further identified that let-7a-5p could bind to both the X-linked inhibitor of apoptosis protein 3'-untranslated region and HMGA2 3'-untranslated region. Moreover, we demonstrated that the forced expression of X-linked inhibitor of apoptosis protein 3'-untranslated region increases the oncogenicity of hepatocellular carcinoma cells in vitro. Cell functional analyses were performed to examine the association of HMGA2 status and X-linked inhibitor of apoptosis protein 3'-untranslated region. We have also measured the functional readout of let-7a-5p and HMGA2, an assay often employed to provide substantial evidence for the effects of X-linked inhibitor of apoptosis protein 3'-untranslated region on hepatocellular carcinoma cells. In general, our findings suggest that X-linked inhibitor of apoptosis protein 3'-untranslated region serves as a competitive endogenous RNA for HMGA2 to activate hepatocellular carcinoma progression by arresting endogenous let-7a-5p.
Collapse
Affiliation(s)
- Wen-Yong Wu
- 1 Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Si-Qi Tao
- 2 Department of Pathology, Anhui Medical University, Hefei, China
| | - Xiao-Nan Wang
- 3 Laboratory of Pathogenic Microbiology and Immunology, Anhui Medical University, Hefei, China
| | - Peter E Lobie
- 4 Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,5 Tsinghua Berkeley Shenzhen Institute, Tsinghua University Graduate School at Shenzhen, Shenzhen, China
| | - Zheng-Sheng Wu
- 2 Department of Pathology, Anhui Medical University, Hefei, China
| |
Collapse
|
69
|
Gosalia AJ, Martin P, Jones PD. Advances and Future Directions in the Treatment of Hepatocellular Carcinoma. Gastroenterol Hepatol (N Y) 2017; 13:398-410. [PMID: 28867968 PMCID: PMC5572970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide. Liver transplant is considered the gold standard for curative therapy for HCC when patients are not candidates for surgical resection or ablation. Because a subset of patients with HCC have a survival rate with liver transplantation that is comparable to that of cirrhotic patients without tumors, the organ allocation system allows for increased priority for transplant in potential recipients within the Milan criteria. With the recent change in the Model for End-Stage Liver Disease exception point allocation, patients with HCC will now need to wait at least 6 months before being awarded extra points. This extension leads to increased time on the transplant waiting list and underscores the importance of locoregional therapy to contain the tumor burden. Fortunately, there has been significant progress in therapy for HCC in the past few decades, namely due to advances in interventional radiology, radiotherapy, and expanded surgical and transplant criteria. Recent advances in immunotherapy also provide promising options for patients who are not candidates for other therapies. This article highlights the major therapeutic options for HCC, including surgical resection, liver transplant, thermal and nonthermal ablation, chemoembolization, radiotherapy, and systemic chemotherapy, as well as discusses the evidence supporting these approaches.
Collapse
Affiliation(s)
- Ashil J Gosalia
- Dr Gosalia is a gastroenterology fellow in the Department of Medicine at the University of Miami Miller School of Medicine in Miami, Florida. Dr Martin is a professor and Dr Jones is an assistant professor in the Division of Hepatology at the University of Miami Miller School of Medicine. Dr Martin and Dr Jones are also affiliated with the Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine
| | - Paul Martin
- Dr Gosalia is a gastroenterology fellow in the Department of Medicine at the University of Miami Miller School of Medicine in Miami, Florida. Dr Martin is a professor and Dr Jones is an assistant professor in the Division of Hepatology at the University of Miami Miller School of Medicine. Dr Martin and Dr Jones are also affiliated with the Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine
| | - Patricia D Jones
- Dr Gosalia is a gastroenterology fellow in the Department of Medicine at the University of Miami Miller School of Medicine in Miami, Florida. Dr Martin is a professor and Dr Jones is an assistant professor in the Division of Hepatology at the University of Miami Miller School of Medicine. Dr Martin and Dr Jones are also affiliated with the Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine
| |
Collapse
|
70
|
Abbate V, Marcantoni M, Giuliante F, Vecchio FM, Gatto I, Mele C, Saviano A, Arciuolo D, Gaetani E, Ferrari MC, Giarretta I, Ardito F, Riccardi L, Nicoletti A, Ponziani FR, Gasbarrini A, Pompili M, Pola R. HepPar1-Positive Circulating Microparticles Are Increased in Subjects with Hepatocellular Carcinoma and Predict Early Recurrence after Liver Resection. Int J Mol Sci 2017; 18:E1043. [PMID: 28498353 PMCID: PMC5454955 DOI: 10.3390/ijms18051043] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/14/2017] [Accepted: 05/01/2017] [Indexed: 12/19/2022] Open
Abstract
Circulating microparticles (MPs) are novel potential biomarkers in cancer patients. Their role in hepatocellular carcinoma (HCC) is under intensive investigation. In this study, we tested the hypothesis that MPs expressing the antigen HepPar1 are increased in the blood of subjects with HCC and may serve as markers of early recurrence after liver resection (LR). We studied 15 patients affected by HCC undergoing LR, and used flow cytometry to assess the number of circulating HepPar1+ MPs. Ten subjects without HCC (five with liver cirrhosis and five with healthy livers) were used as controls. After LR, HCC patients underwent a follow-up to check for early recurrence, which occurred in seven cases. The number of circulating HepPar1+ MPs was significantly higher in subjects affected by HCC, compared to individuals without cancer (p < 0.01). We also found that, among HCC patients, the number of circulating HepPar1+ MPs, measured before LR, was significantly higher in those who displayed early recurrence compared to those without recurrence (p = 0.02). Of note, other types of circulating MPs, such as those derived from endothelial cells (CD144+) or those produced by the activated endothelium (CD144+/CD62+), were not associated with HCC, nor could they predict HCC recurrence. HepPar1+ MPs deserve further investigation as novel biomarkers of disease and prognosis in HCC patients.
Collapse
Affiliation(s)
- Valeria Abbate
- Division of Internal Medicine and Gastroenterology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Margherita Marcantoni
- Division of Vascular Medicine, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Felice Giuliante
- Hepatobiliary Surgery Unit, and Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Fabio M Vecchio
- Department of Pathology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Ilaria Gatto
- Division of Vascular Medicine, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Caterina Mele
- Hepatobiliary Surgery Unit, and Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Antonio Saviano
- Division of Internal Medicine and Gastroenterology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Damiano Arciuolo
- Department of Pathology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Eleonora Gaetani
- Division of Internal Medicine and Gastroenterology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Maria C Ferrari
- Division of Vascular Medicine, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Igor Giarretta
- Division of Vascular Medicine, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Francesco Ardito
- Hepatobiliary Surgery Unit, and Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Laura Riccardi
- Division of Internal Medicine and Gastroenterology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Alberto Nicoletti
- Division of Internal Medicine and Gastroenterology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Francesca R Ponziani
- Division of Internal Medicine and Gastroenterology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Antonio Gasbarrini
- Division of Internal Medicine and Gastroenterology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Maurizio Pompili
- Division of Internal Medicine and Gastroenterology, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| | - Roberto Pola
- Division of Vascular Medicine, Catholic University School of Medicine, A. Gemelli University Hospital, Rome 00168, Italy.
| |
Collapse
|
71
|
Pedersen M, Andersen ZJ, Stafoggia M, Weinmayr G, Galassi C, Sørensen M, Eriksen KT, Tjønneland A, Loft S, Jaensch A, Nagel G, Concin H, Tsai MY, Grioni S, Marcon A, Krogh V, Ricceri F, Sacerdote C, Ranzi A, Sokhi R, Vermeulen R, Hoogh KD, Wang M, Beelen R, Vineis P, Brunekreef B, Hoek G, Raaschou-Nielsen O. Ambient air pollution and primary liver cancer incidence in four European cohorts within the ESCAPE project. ENVIRONMENTAL RESEARCH 2017; 154:226-233. [PMID: 28107740 DOI: 10.1016/j.envres.2017.01.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/19/2016] [Accepted: 01/05/2017] [Indexed: 05/21/2023]
Abstract
BACKGROUND Tobacco smoke exposure increases the risk of cancer in the liver, but little is known about the possible risk associated with exposure to ambient air pollution. OBJECTIVES We evaluated the association between residential exposure to air pollution and primary liver cancer incidence. METHODS We obtained data from four cohorts with enrolment during 1985-2005 in Denmark, Austria and Italy. Exposure to nitrogen oxides (NO2 and NOX), particulate matter (PM) with diameter of less than 10µm (PM10), less than 2.5µm (PM2.5), between 2.5 and 10µm (PM2.5-10) and PM2.5 absorbance (soot) at baseline home addresses were estimated using land-use regression models from the ESCAPE project. We also investigated traffic density on the nearest road. We used Cox proportional-hazards models with adjustment for potential confounders for cohort-specific analyses and random-effects meta-analyses to estimate summary hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS Out of 174,770 included participants, 279 liver cancer cases were diagnosed during a mean follow-up of 17 years. In each cohort, HRs above one were observed for all exposures with exception of PM2.5 absorbance and traffic density. In the meta-analysis, all exposures were associated with elevated HRs, but none of the associations reached statistical significance. The summary HR associated with a 10-μg/m3 increase in NO2 was 1.10 (95% confidence interval (CI): 0.93, 1.30) and 1.34 (95% CI: 0.76, 2.35) for a 5-μg/m3 increase in PM2.5. CONCLUSIONS The results provide suggestive evidence that ambient air pollution may increase the risk of liver cancer. Confidence intervals for associations with NO2 and NOX were narrower than for the other exposures.
Collapse
Affiliation(s)
- Marie Pedersen
- The Danish Cancer Society Research Center, Copenhagen, Denmark; Centre for Epidemiology and Screening, Department of Public Health, University of Copenhagen, Copenhagen, Denmark.
| | - Zorana J Andersen
- Centre for Epidemiology and Screening, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Massimo Stafoggia
- Department of Epidemiology, Lazio Regional Health Service, Local Health Unit ASL RM1, Rome, Italy; Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gudrun Weinmayr
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Claudia Galassi
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Mette Sørensen
- The Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Anne Tjønneland
- The Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Steffen Loft
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Jaensch
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Gabriele Nagel
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany; Agency for Preventive and Social Medicine, Bregenz, Austria
| | - Hans Concin
- Agency for Preventive and Social Medicine, Bregenz, Austria
| | - Ming-Yi Tsai
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Sara Grioni
- Epidemiology and Prevention Unit, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandro Marcon
- Unit of Epidemiology & Medical Statistics, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Vittorio Krogh
- Epidemiology and Prevention Unit, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Fulvio Ricceri
- Unit of Epidemiology, Regional Health Service ASL TO3, Grugliasco, Italy; Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Andrea Ranzi
- Environmental Health Reference Centre, Regional Agency for Prevention, Environment and Energy of Emilia-Romagna, Modena, Italy
| | - Ranjeet Sokhi
- Centre for Atmospheric and Instrumentation Research, University of Hertfordshire, College Lane, Hatfield, United Kingdom
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands; School of Public Health, Imperial College, London, United Kingdom
| | - Kees de Hoogh
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Meng Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Rob Beelen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands; National Institute for Public Health (RIVM), Bilthoven, The Netherlands
| | - Paolo Vineis
- School of Public Health, Imperial College, London, United Kingdom; Molecular end Epidemiology Unit, HuGeF, Human Genetics Foundation, Torino, Italy
| | - Bert Brunekreef
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerard Hoek
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ole Raaschou-Nielsen
- The Danish Cancer Society Research Center, Copenhagen, Denmark; Department of Environmental Science, Aarhus University, Roskilde, Denmark
| |
Collapse
|
72
|
Abstract
PURPOSE OF REVIEW This review aims to assess the epidemiological evidence for a link between type 2 diabetes and hepatocellular carcinoma and to investigate possible pathophysiological mechanisms. RECENT FINDINGS The presence of type 2 diabetes significantly increases the risk of developing hepatocellular carcinoma, and treatment with metformin may be associated with a lower risk. Treatment with insulin and sulphonylureas may be associated with increased risk. The pathophysiology underlying development of hepatocellular carcinoma in this context is complex and is likely to involve increased proinflammatory mediators, oxidative stress, JNK-1 activation, increased IGF-1 activity, altered gut microbiota and immunomodulation. Hepatocellular carcinoma incidence is increasing and this is likely to be linked to the increasing incidence of type 2 diabetes, obesity and the metabolic syndrome. These conditions increase the risk of developing hepatocellular carcinoma, and a greater understanding of the underlying pathophysiology may help with the development of novel treatments.
Collapse
Affiliation(s)
- P Wainwright
- Clinical Biochemistry, University Hospital Southampton, Southampton, UK.
- Chemical Pathology and Metabolic Medicine, Department of Laboratory Medicine, D-Level Pathology Block, University Hospital Southampton, Tremona Road, Southampton, SO16 6YD, UK.
| | - E Scorletti
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health Research Southampton Biomedical Research Center (in Nutrition) and Respiratory Biomedical Research Unit, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK
| | - C D Byrne
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health Research Southampton Biomedical Research Center (in Nutrition) and Respiratory Biomedical Research Unit, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK
| |
Collapse
|
73
|
Metabolic Disorders and Cancer: Hepatocyte Store-Operated Ca2+ Channels in Nonalcoholic Fatty Liver Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:595-621. [DOI: 10.1007/978-3-319-57732-6_30] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|