Genetics, Immunity and Nutrition Boost the Switching from NASH to HCC
Abstract
:1. Introduction
2. Common Genetic Variations Promote the Switch from NASH to HCC
3. Genetic Variants in Immunoregulatory Genes Modulate the Risk of HCC in NAFLD Patients
4. The Pathogenic Role of Rare Genetic Variants in NAFLD-HCC Development
5. Epigenetic Variations Driving NAFLD-HCC
6. Inflammation
7. Gut Microbiota
8. Nutrition and HCC
8.1. Alcohol Drinking Accelerates NASH-HCC Onset
8.2. The Role of Aflatoxin B1 in Hepatocarcinogenesis
8.3. Iron Overload Increases the Risk of HCC
8.4. Dietary Cholesterol: The Main Lipid Driver of the Switching from Simple Steatosis to NASH-HCC
8.5. Protective Compounds against Hepatic Damage
8.6. Dietary Fibers
8.7. Branched-Chain Amino Acids
8.8. Omega-3 Polyunsaturated Fatty Acids
9. Preclinical Models to Induce NASH-HCC: From Dietary Supplementation to Genetics
10. Concluding Remarks
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
α-SMA | alpha-smooth muscle actin |
ABL | abetalipoproteinemia |
ACVR2A | Activin A Receptor Type 2A |
ADH | Alcohol Dehydrogenase |
AF-B1 | Aflatoxin B1 |
AGER | advanced glycosylation end product-specific receptor |
ALDH | Aldehyde Dehydrogenase |
ALIOS | American Lifestyle-Induced Obesity Syndrome |
APOB | apolipoprotein B |
Ath+HF | atherogenic and high-fat |
BCAA | Branched-Chain Amino Acids |
BMI | body mass index |
CAR | chimeric antigen receptor |
CD-HFD | choline-deficient high-fat diet |
CDKI1A | cyclin dependent kinase inhibitor 1A |
c-Jun | c-Jun N-terminal kinase |
CRISPR/Cas9 | clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 |
CTNNB1 | Catenin beta 1 |
CYP2E1 | Cytochrome P450 2E1 |
DAMPs | damage-associated molecular patterns |
DEN | diethylnitrosamine |
DIAMOND | diet-induced animal model of non-alcoholic fatty liver disease |
DNMT | DNA methyltransferases |
EPIC | European Prospective Investigation into Cancer and Nutrition |
ER | endoplasmic reticulum |
EZH2 | polycomb protein enhancer of zeste homolog 2 |
FFAs | free fatty acids |
GWAS | genome-wide association study |
HBV | hepatitis B infection |
HCC | hepatocellular carcinoma |
HDAC8 | histone deacetylase 8 |
HFD | high-fat diet |
HFHC | high fat high cholesterol |
Ho-FHBL | hypobetalipoproteinemia |
HPCs | hepatic progenitor cells |
HR | hazard ratio |
HSCs | hepatic stellate cells |
HSD17B13 | hydroxysteroid 17-β dehydrogenase 13 |
ILs | interleukins |
IgA | Immunoglobulin A |
IR | insulin resistance |
KO | knock-out |
LD | lipid droplet |
LDL | low-density lipoprotein |
Lyso-PI | lyso-phosphatidylinositol |
LOX-1 | lectin-like oxidized LDL receptor-1 |
LPIAT1 | lyso-phosphatidylinositol (Lyso-PI) acyl-transferase1 |
LPS | lipopolysaccharides |
MAT | methionine adenosyltransferase |
MBOAT7 | membrane bound o-acyltransferase domain-containing 7 |
MC4R | melanocortin 4 receptor-deficient mice |
miRNA | microRNA |
mtDNA | mitochondrial DNA |
MT-ND6 | mitochondrially encoded NADH dehydrogenase 6 |
MTTP | microsomal triglyceride transfer protein large subunit gene |
NAFLD | nonalcoholic fatty liver disease |
NASH | nonalcoholic steatohepatitis |
NF-κB | nuclear factor kappa-light-chain-enhancer of activated B-cells |
NLRs | intracellular nucleotide-binding and oligomerization domain (NOD)-like receptors |
OR | odd ratio |
OS | overall survival |
PAMPs | pathogen-associated molecular patterns |
PD-1 | death protein 1 |
PGC-1α | peroxisome proliferator-activated receptor γ coactivator-1α |
PI | phosphatidylinositol |
PNPLA3 | patatin-like phospholipase domain-containing 3 |
PPARs | peroxisome proliferator activating receptors |
PRSs | polygenic risk scores |
PTEN | phosphatase and tensin homolog |
PUFAs | polyunsaturated fatty acids |
ROC | receiver operating characteristics |
ROS | reactive oxygen species |
RR | relative risk |
SAM | S-adenyl methionine |
SASP | senescence-associated secretory phenotype |
SERCA | sarco/ER calcium ATPase |
SIRTs | sirtuins |
SNP | single nucleotide polymorphism |
SR-A | scavenger receptor |
SREBP1 | Sterol Regulatory Element Binding Transcription Factor 1 |
STZ | streptozotocin |
STAT3 | Signal Transducer and Activator of Transcription 3 |
T2D | type 2 diabetes |
TERT | telomerase reverse transcriptase |
TG | triglyceride |
TGF-β | transforming growth factor β |
TLRs | toll like receptors |
TM6SF2 | transmembrane 6 superfamily member 2 |
TMC4 | transmembrane channel like 4 |
TNF-α | tumor necrosis factor α |
uPA | urokinase plasminogen activator |
UPR | unfolded protein response |
VLDL | very-low density lipoproteins |
WD | Western diet |
WNT | wingless |
References
- Younossi, Z.M.; Henry, L. Epidemiology of non-alcoholic fatty liver disease and hepatocellular carcinoma. JHEP Rep. 2021, 3, 100305. [Google Scholar] [CrossRef] [PubMed]
- Huang, D.Q.; El-Serag, H.B.; Loomba, R. Global epidemiology of NAFLD-related HCC: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 223–238. [Google Scholar] [CrossRef]
- Golabi, P.; Rhea, L.; Henry, L.; Younossi, Z.M. Hepatocellular carcinoma and non-alcoholic fatty liver disease. Hepatol. Int. 2019, 13, 688–694. [Google Scholar] [CrossRef]
- Younossi, Z.M.; Stepanova, M.; Ong, J.; Trimble, G.; AlQahtani, S.; Younossi, I.; Ahmed, A.; Racila, A.; Henry, L. Nonalcoholic Steatohepatitis Is the Most Rapidly Increasing Indication for Liver Transplantation in the United States. Clin. Gastroenterol. Hepatol. 2021, 19, 580–589.e585. [Google Scholar] [CrossRef] [PubMed]
- Yasui, K.; Hashimoto, E.; Komorizono, Y.; Koike, K.; Arii, S.; Imai, Y.; Shima, T.; Kanbara, Y.; Saibara, T.; Mori, T.; et al. Characteristics of patients with nonalcoholic steatohepatitis who develop hepatocellular carcinoma. Clin. Gastroenterol. Hepatol. 2011, 9, 428–433. [Google Scholar] [CrossRef] [PubMed]
- Meroni, M.; Longo, M.; Paolini, E.; Lombardi, R.; Piciotti, R.; Francione, P.; Badiali, S.; Maggioni, M.; Fracanzani, A.L. MAFLD definition underestimates the risk to develop HCC in genetically predisposed patients. J. Intern. Med. 2021. [Google Scholar] [CrossRef]
- Longo, M.; Paolini, E.; Meroni, M.; Dongiovanni, P. Remodeling of Mitochondrial Plasticity: The Key Switch from NAFLD/NASH to HCC. Int. J. Mol. Sci. 2021, 22, 4173. [Google Scholar] [CrossRef]
- Meroni, M.; Longo, M.; Tria, G.; Dongiovanni, P. Genetics Is of the Essence to Face NAFLD. Biomedicines 2021, 9, 1359. [Google Scholar] [CrossRef]
- Valenti, L.; Alisi, A.; Galmozzi, E.; Bartuli, A.; Del Menico, B.; Alterio, A.; Dongiovanni, P.; Fargion, S.; Nobili, V. I148M patatin-like phospholipase domain-containing 3 gene variant and severity of pediatric nonalcoholic fatty liver disease. Hepatology 2010, 52, 1274–1280. [Google Scholar] [CrossRef]
- Valenti, L.; Dongiovanni, P. Mutant PNPLA3 I148M protein as pharmacological target for liver disease. Hepatology 2017, 66, 1026–1028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meroni, M.; Longo, M.; Rustichelli, A.; Dongiovanni, P. Nutrition and Genetics in NAFLD: The Perfect Binomium. Int. J. Mol. Sci. 2020, 21, 2986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Kory, N.; BasuRay, S.; Cohen, J.C.; Hobbs, H.H. PNPLA3, CGI-58, and Inhibition of Hepatic Triglyceride Hydrolysis in Mice. Hepatology 2019, 69, 2427–2441. [Google Scholar] [CrossRef] [Green Version]
- Bruschi, F.V.; Claudel, T.; Tardelli, M.; Caligiuri, A.; Stulnig, T.M.; Marra, F.; Trauner, M. The PNPLA3 I148M variant modulates the fibrogenic phenotype of human hepatic stellate cells. Hepatology 2017, 65, 1875–1890. [Google Scholar] [CrossRef] [Green Version]
- Dongiovanni, P.; Donati, B.; Fares, R.; Lombardi, R.; Mancina, R.M.; Romeo, S.; Valenti, L. PNPLA3 I148M polymorphism and progressive liver disease. World J. Gastroenterol. 2013, 19, 6969–6978. [Google Scholar] [CrossRef]
- Pirazzi, C.; Valenti, L.; Motta, B.M.; Pingitore, P.; Hedfalk, K.; Mancina, R.M.; Burza, M.A.; Indiveri, C.; Ferro, Y.; Montalcini, T.; et al. PNPLA3 has retinyl-palmitate lipase activity in human hepatic stellate cells. Hum. Mol. Genet. 2014, 23, 4077–4085. [Google Scholar] [CrossRef] [Green Version]
- Trépo, E.; Nahon, P.; Bontempi, G.; Valenti, L.; Falleti, E.; Nischalke, H.D.; Hamza, S.; Corradini, S.G.; Burza, M.A.; Guyot, E.; et al. Association between the PNPLA3 (rs738409 C>G) variant and hepatocellular carcinoma: Evidence from a meta-analysis of individual participant data. Hepatology 2014, 59, 2170–2177. [Google Scholar] [CrossRef]
- Carpino, G.; Pastori, D.; Baratta, F.; Overi, D.; Labbadia, G.; Polimeni, L.; Di Costanzo, A.; Pannitteri, G.; Carnevale, R.; Del Ben, M.; et al. PNPLA3 variant and portal/periportal histological pattern in patients with biopsy-proven non-alcoholic fatty liver disease: A possible role for oxidative stress. Sci. Rep. 2017, 7, 15756. [Google Scholar] [CrossRef] [PubMed]
- Bruschi, F.V.; Tardelli, M.; Herac, M.; Claudel, T.; Trauner, M. Metabolic regulation of hepatic PNPLA3 expression and severity of liver fibrosis in patients with NASH. Liver Int. 2020, 40, 1098–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Min, H.K.; Sookoian, S.; Pirola, C.J.; Cheng, J.; Mirshahi, F.; Sanyal, A.J. Metabolic profiling reveals that PNPLA3 induces widespread effects on metabolism beyond triacylglycerol remodeling in Huh-7 hepatoma cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 307, G66–G76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Banini, B.A.; Kumar, D.P.; Cazanave, S.; Seneshaw, M.; Mirshahi, F.; Santhekadur, P.K.; Wang, L.; Guan, H.P.; Oseini, A.M.; Alonso, C.; et al. Identification of a Metabolic, Transcriptomic, and Molecular Signature of Patatin-Like Phospholipase Domain Containing 3-Mediated Acceleration of Steatohepatitis. Hepatology 2021, 73, 1290–1306. [Google Scholar] [CrossRef]
- Bruschi, F.V.; Tardelli, M.; Einwallner, E.; Claudel, T.; Trauner, M. PNPLA3 I148M Up-Regulates Hedgehog and Yap Signaling in Human Hepatic Stellate Cells. Int. J. Mol. Sci. 2020, 21, 8711. [Google Scholar] [CrossRef] [PubMed]
- Pinyol, R.; Torrecilla, S.; Wang, H.; Montironi, C.; Piqué-Gili, M.; Torres-Martin, M.; Wei-Qiang, L.; Willoughby, C.E.; Ramadori, P.; Andreu-Oller, C.; et al. Molecular characterisation of hepatocellular carcinoma in patients with non-alcoholic steatohepatitis. J. Hepatol. 2021, 75, 865–878. [Google Scholar] [CrossRef] [PubMed]
- Kozlitina, J.; Smagris, E.; Stender, S.; Nordestgaard, B.G.; Zhou, H.H.; Tybjærg-Hansen, A.; Vogt, T.F.; Hobbs, H.H.; Cohen, J.C. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nat. Genet. 2014, 46, 352–356. [Google Scholar] [CrossRef] [Green Version]
- Ruhanen, H.; Nidhina Haridas, P.A.; Eskelinen, E.L.; Eriksson, O.; Olkkonen, V.M.; Käkelä, R. Depletion of TM6SF2 disturbs membrane lipid composition and dynamics in HuH7 hepatoma cells. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 2017, 1862, 676–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Hare, E.A.; Yang, R.; Yerges-Armstrong, L.M.; Sreenivasan, U.; McFarland, R.; Leitch, C.C.; Wilson, M.H.; Narina, S.; Gorden, A.; Ryan, K.A.; et al. TM6SF2 rs58542926 impacts lipid processing in liver and small intestine. Hepatology 2017, 65, 1526–1542. [Google Scholar] [CrossRef] [Green Version]
- Dongiovanni, P.; Petta, S.; Maglio, C.; Fracanzani, A.L.; Pipitone, R.; Mozzi, E.; Motta, B.M.; Kaminska, D.; Rametta, R.; Grimaudo, S.; et al. Transmembrane 6 superfamily member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease. Hepatology 2015, 61, 506–514. [Google Scholar] [CrossRef]
- Goffredo, M.; Caprio, S.; Feldstein, A.E.; D’Adamo, E.; Shaw, M.M.; Pierpont, B.; Savoye, M.; Zhao, H.; Bale, A.E.; Santoro, N. Role of TM6SF2 rs58542926 in the pathogenesis of nonalcoholic pediatric fatty liver disease: A multiethnic study. Hepatology 2016, 63, 117–125. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Que, S.; Zhou, L.; Zheng, S.; Romeo, S.; Mardinoglu, A.; Valenti, L. The effect of the TM6SF2 E167K variant on liver steatosis and fibrosis in patients with chronic hepatitis C: A meta-analysis. Sci. Rep. 2017, 7, 9273. [Google Scholar] [CrossRef]
- Eslam, M.; Mangia, A.; Berg, T.; Chan, H.L.; Irving, W.L.; Dore, G.J.; Abate, M.L.; Bugianesi, E.; Adams, L.A.; Najim, M.A.; et al. Diverse impacts of the rs58542926 E167K variant in TM6SF2 on viral and metabolic liver disease phenotypes. Hepatology 2016, 64, 34–46. [Google Scholar] [CrossRef] [Green Version]
- Li, T.T.; Li, T.H.; Peng, J.; He, B.; Liu, L.S.; Wei, D.H.; Jiang, Z.S.; Zheng, X.L.; Tang, Z.H. TM6SF2: A novel target for plasma lipid regulation. Atherosclerosis 2018, 268, 170–176. [Google Scholar] [CrossRef]
- Sookoian, S.; Castaño, G.O.; Scian, R.; Mallardi, P.; Fernández Gianotti, T.; Burgueño, A.L.; San Martino, J.; Pirola, C.J. Genetic variation in transmembrane 6 superfamily member 2 and the risk of nonalcoholic fatty liver disease and histological disease severity. Hepatology 2015, 61, 515–525. [Google Scholar] [CrossRef]
- Liu, Y.-L.; Reeves, H.L.; Burt, A.D.; Tiniakos, D.; McPherson, S.; Leathart, J.B.S.; Allison, M.E.D.; Alexander, G.J.; Piguet, A.-C.; Anty, R.; et al. TM6SF2 rs58542926 influences hepatic fibrosis progression in patients with non-alcoholic fatty liver disease. Nat. Commun. 2014, 5, 4309. [Google Scholar] [CrossRef] [Green Version]
- Raksayot, M.; Chuaypen, N.; Khlaiphuengsin, A.; Pinjaroen, N.; Treeprasertsuk, S.; Poovorawan, Y.; Tanaka, Y.; Tangkijvanich, P. Independent and additive effects of PNPLA3 and TM6SF2 polymorphisms on the development of non-B, non-C hepatocellular carcinoma. J. Gastroenterol. 2019, 54, 427–436. [Google Scholar] [CrossRef]
- Chen, X.; Zhou, P.; De, L.; Li, B.; Su, S. The roles of transmembrane 6 superfamily member 2 rs58542926 polymorphism in chronic liver disease: A meta-analysis of 24,147 subjects. Mol. Genet. Genom. Med. 2019, 7, e824. [Google Scholar] [CrossRef]
- Stickel, F.; Buch, S.; Nischalke, H.D.; Weiss, K.H.; Gotthardt, D.; Fischer, J.; Rosendahl, J.; Marot, A.; Elamly, M.; Casper, M.; et al. Genetic variants in PNPLA3 and TM6SF2 predispose to the development of hepatocellular carcinoma in individuals with alcohol-related cirrhosis. Am. J. Gastroenterol. 2018, 113, 1475–1483. [Google Scholar] [CrossRef]
- Longo, M.; Meroni, M.; Paolini, E.; Macchi, C.; Dongiovanni, P. Mitochondrial dynamics and nonalcoholic fatty liver disease (NAFLD): New perspectives for a fairy-tale ending? Metab. Clin. Exp. 2021, 117, 154708. [Google Scholar] [CrossRef] [PubMed]
- Longo, M.; Meroni, M.; Erconi, V.; Carli, F.; Macchi, C.; Fortunato, F.; Ronchi, D.; Sabatini, S.; Paolini, E.; De Caro, E.R.; et al. TM6SF2/PNPLA3/MBOAT7 loss-of-function genetic variants impact on NAFLD development and progression both in patients and in in vitro models. bioRxiv 2020. [Google Scholar] [CrossRef]
- Meroni, M.; Longo, M.; Fracanzani, A.L.; Dongiovanni, P. MBOAT7 down-regulation by genetic and environmental factors predisposes to MAFLD. EBioMedicine 2020, 57, 102866. [Google Scholar] [CrossRef]
- Stickel, F.; Moreno, C.; Hampe, J.; Morgan, M.Y. The genetics of alcohol dependence and alcohol-related liver disease. J. Hepatol. 2017, 66, 195–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buch, S.; Stickel, F.; Trepo, E.; Way, M.M.; Herrmann, A.; Nischalke, H.D.; Brosch, M.M.; Rosendahl, J.J.; Berg, T.; Ridinger, M.M.; et al. A genome-wide association study confirms PNPLA3 and identifies TM6SF2 and MBOAT7 as risk loci for alcohol-related cirrhosis. Nat. Genet. 2015, 47, 1443–1448. [Google Scholar] [CrossRef]
- Donati, B.; Dongiovanni, P.; Romeo, S.; Meroni, M.; McCain, M.; Miele, L.; Petta, S.; Maier, S.; Rosso, C.; De Luca, L.; et al. MBOAT7 rs641738 variant and hepatocellular carcinoma in non-cirrhotic individuals. Sci. Rep. 2017, 7, 4492. [Google Scholar] [CrossRef] [Green Version]
- Freund, C.; Wahlers, A.; Begli, N.H.; Leopold, Y.; Kloters-Plachky, P.; Mehrabi, A.; Mohr, I.; Sander, J.; Rupp, C.; Gotthardt, D.N.; et al. The MBOAT7 rs641738 variant is associated with an improved outcome in primary sclerosing cholangitis. Clin. Res. Hepatol. Gastroenterol. 2020, 44, 646–652. [Google Scholar] [CrossRef] [PubMed]
- Meroni, M.; Dongiovanni, P.; Longo, M.; Carli, F.; Baselli, G.; Rametta, R.; Pelusi, S.; Badiali, S.; Maggioni, M.; Gaggini, M.; et al. Mboat7 down-regulation by hyper-insulinemia induces fat accumulation in hepatocytes. EBioMedicine 2020, 52, 102658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mancina, R.M.; Dongiovanni, P.; Petta, S.; Pingitore, P.; Meroni, M.; Rametta, R.; Borén, J.; Montalcini, T.; Pujia, A.; Wiklund, O.; et al. The MBOAT7-TMC4 Variant rs641738 Increases Risk of Nonalcoholic Fatty Liver Disease in Individuals of European Descent. Gastroenterology 2016, 150, 1219–1230.e1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luukkonen, P.K.; Zhou, Y.; Hyötyläinen, T.; Leivonen, M.; Arola, J.; Orho-Melander, M.; Orešič, M.; Yki-Järvinen, H. The MBOAT7 variant rs641738 alters hepatic phosphatidylinositols and increases severity of non-alcoholic fatty liver disease in humans. J. Hepatol. 2016, 65, 1263–1265. [Google Scholar] [CrossRef] [Green Version]
- Tanaka, Y.; Shimanaka, Y.; Caddeo, A.; Kubo, T.; Mao, Y.; Kubota, T.; Kubota, N.; Yamauchi, T.; Mancina, R.M.; Baselli, G.; et al. LPIAT1/MBOAT7 depletion increases triglyceride synthesis fueled by high phosphatidylinositol turnover. Gut 2021, 70, 180–193. [Google Scholar] [CrossRef]
- Helsley, R.N.; Varadharajan, V.; Brown, A.L.; Gromovsky, A.D.; Schugar, R.C.; Ramachandiran, I.; Fung, K.; Kabbany, M.N.; Banerjee, R.; Neumann, C.K.; et al. Obesity-linked suppression of membrane-bound O-acyltransferase 7 (MBOAT7) drives non-alcoholic fatty liver disease. eLife 2019, 8, e49882. [Google Scholar] [CrossRef]
- Dongiovanni, P.; Stender, S.; Pietrelli, A.; Mancina, R.M.; Cespiati, A.; Petta, S.; Pelusi, S.; Pingitore, P.; Badiali, S.; Maggioni, M.; et al. Causal relationship of hepatic fat with liver damage and insulin resistance in nonalcoholic fatty liver. J. Intern. Med. 2018, 283, 356–370. [Google Scholar] [CrossRef]
- Bianco, C.; Jamialahmadi, O.; Pelusi, S.; Baselli, G.; Dongiovanni, P.; Zanoni, I.; Santoro, L.; Maier, S.; Liguori, A.; Meroni, M.; et al. Non-invasive stratification of hepatocellular carcinoma risk in non-alcoholic fatty liver using polygenic risk scores. J. Hepatol. 2021, 74, 775–782. [Google Scholar] [CrossRef] [PubMed]
- Pfister, D.; Núñez, N.G.; Pinyol, R.; Govaere, O.; Pinter, M.; Szydlowska, M.; Gupta, R.; Qiu, M.; Deczkowska, A.; Weiner, A.; et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 2021, 592, 450–456. [Google Scholar] [CrossRef]
- Anstee, Q.M.; Reeves, H.L.; Kotsiliti, E.; Govaere, O. From NASH to HCC: Current concepts and future challenges. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 411–428. [Google Scholar] [CrossRef]
- Zhu, A.X.; Finn, R.S.; Edeline, J.; Cattan, S.; Ogasawara, S.; Palmer, D.; Verslype, C.; Zagonel, V.; Fartoux, L.; Vogel, A.; et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): A non-randomised, open-label phase 2 trial. Lancet Oncol. 2018, 19, 940–952. [Google Scholar] [CrossRef]
- El-Khoueiry, A.B.; Sangro, B.; Yau, T.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Kim, T.Y.; Choo, S.P.; Trojan, J.; Welling, T.H.R.; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017, 389, 2492–2502. [Google Scholar] [CrossRef]
- Finn, R.S.; Ryoo, B.Y.; Merle, P.; Kudo, M.; Bouattour, M.; Lim, H.Y.; Breder, V.; Edeline, J.; Chao, Y.; Ogasawara, S.; et al. Pembrolizumab as Second-Line Therapy in Patients with Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. 2020, 38, 193–202. [Google Scholar] [CrossRef] [PubMed]
- Salmaninejad, A.; Khoramshahi, V.; Azani, A.; Soltaninejad, E.; Aslani, S.; Zamani, M.R.; Zal, M.; Nesaei, A.; Hosseini, S.M. PD-1 and cancer: Molecular mechanisms and polymorphisms. Immunogenetics 2018, 70, 73–86. [Google Scholar] [CrossRef] [PubMed]
- Boussiotis, V.A. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. N. Engl. J. Med. 2016, 375, 1767–1778. [Google Scholar] [CrossRef] [Green Version]
- Chihab, H.; Jadid, F.Z.; Foka, P.; Zaidane, I.; El Fihry, R.; Georgopoulou, U.; Marchio, A.; Elhabazi, A.; Chair, M.; Pineau, P.; et al. Programmed cell death-1 3′-untranslated region polymorphism is associated with spontaneous clearance of hepatitis B virus infection. J. Med. Virol. 2018, 90, 1730–1738. [Google Scholar] [CrossRef]
- Zang, B.; Chen, C.; Zhao, J.Q. PD-1 gene rs10204525 and rs7421861 polymorphisms are associated with increased risk and clinical features of esophageal cancer in a Chinese Han population. Aging 2020, 12, 3771–3790. [Google Scholar] [CrossRef]
- Eldafashi, N.; Darlay, R.; Shukla, R.; McCain, M.V.; Watson, R.; Liu, Y.L.; McStraw, N.; Fathy, M.; Fawzy, M.A.; Zaki, M.Y.W.; et al. A PDCD1 Role in the Genetic Predisposition to NAFLD-HCC? Cancers 2021, 13, 1412. [Google Scholar] [CrossRef]
- Di Filippo, M.; Moulin, P.; Roy, P.; Samson-Bouma, M.E.; Collardeau-Frachon, S.; Chebel-Dumont, S.; Peretti, N.; Dumortier, J.; Zoulim, F.; Fontanges, T.; et al. Homozygous MTTP and APOB mutations may lead to hepatic steatosis and fibrosis despite metabolic differences in congenital hypocholesterolemia. J. Hepatol. 2014, 61, 891–902. [Google Scholar] [CrossRef]
- Stein, E.A.; Dufour, R.; Gagne, C.; Gaudet, D.; East, C.; Donovan, J.M.; Chin, W.; Tribble, D.L.; McGowan, M. Apolipoprotein B synthesis inhibition with mipomersen in heterozygous familial hypercholesterolemia: Results of a randomized, double-blind, placebo-controlled trial to assess efficacy and safety as add-on therapy in patients with coronary artery disease. Circulation 2012, 126, 2283–2292. [Google Scholar] [CrossRef]
- Jansson-Knodell, C.L.; Gawrieh, S.; McIntyre, A.D.; Liang, T.; Hegele, R.A.; Chalasani, N. Apolipoprotein B and PNPLA3 Double Heterozygosity in a Father-Son Pair With Advanced Nonalcoholic Fatty Liver Disease. Hepatology 2020, 71, 383–385. [Google Scholar] [CrossRef]
- Pelusi, S.; Baselli, G.; Pietrelli, A.; Dongiovanni, P.; Donati, B.; McCain, M.V.; Meroni, M.; Fracanzani, A.L.; Romagnoli, R.; Petta, S.; et al. Rare Pathogenic Variants Predispose to Hepatocellular Carcinoma in Nonalcoholic Fatty Liver Disease. Sci. Rep. 2019, 9, 3682. [Google Scholar] [CrossRef]
- The Cancer Genome Atlas Research Network. Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell 2017, 169, 1327–1341.e1323. [Google Scholar] [CrossRef] [Green Version]
- Ki Kim, S.; Ueda, Y.; Hatano, E.; Kakiuchi, N.; Takeda, H.; Goto, T.; Shimizu, T.; Yoshida, K.; Ikura, Y.; Shiraishi, Y.; et al. TERT promoter mutations and chromosome 8p loss are characteristic of nonalcoholic fatty liver disease-related hepatocellular carcinoma. Int. J. Cancer 2016, 139, 2512–2518. [Google Scholar] [CrossRef]
- Donati, B.; Pietrelli, A.; Pingitore, P.; Dongiovanni, P.; Caddeo, A.; Walker, L.; Baselli, G.; Pelusi, S.; Rosso, C.; Vanni, E.; et al. Telomerase reverse transcriptase germline mutations and hepatocellular carcinoma in patients with nonalcoholic fatty liver disease. Cancer Med. 2017, 6, 1930–1940. [Google Scholar] [CrossRef]
- Aravinthan, A.; Mells, G.; Allison, M.; Leathart, J.; Kotronen, A.; Yki-Jarvinen, H.; Daly, A.K.; Day, C.P.; Anstee, Q.M.; Alexander, G. Gene polymorphisms of cellular senescence marker p21 and disease progression in non-alcohol-related fatty liver disease. Cell Cycle 2014, 13, 1489–1494. [Google Scholar] [CrossRef] [Green Version]
- Meroni, M.; Longo, M.; Paolini, E.; Alisi, A.; Miele, L.; De Caro, E.R.; Pisano, G.; Maggioni, M.; Soardo, G.; Valenti, L.V.; et al. The rs599839 A>G Variant Disentangles Cardiovascular Risk and Hepatocellular Carcinoma in NAFLD Patients. Cancers 2021, 13, 1783. [Google Scholar] [CrossRef]
- Dongiovanni, P.; Meroni, M.; Petta, S.; Longo, M.; Alisi, A.; Soardo, G.; Valenti, L.; Miele, L.; Grimaudo, S.; Pennisi, G.; et al. Neurotensin up-regulation is associated with advanced fibrosis and hepatocellular carcinoma in patients with MAFLD. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158765. [Google Scholar] [CrossRef]
- Jung, B.; Doctolero, R.T.; Tajima, A.; Nguyen, A.K.; Keku, T.; Sandler, R.S.; Carethers, J.M. Loss of activin receptor type 2 protein expression in microsatellite unstable colon cancers. Gastroenterology 2004, 126, 654–659. [Google Scholar] [CrossRef]
- Dongiovanni, P.; Meroni, M.; Longo, M.; Fargion, S.; Fracanzani, A.L. miRNA Signature in NAFLD: A Turning Point for a Non-Invasive Diagnosis. Int. J. Mol. Sci. 2018, 19, 3966. [Google Scholar] [CrossRef] [Green Version]
- Dongiovanni, P.; Valenti, L. A Nutrigenomic Approach to Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2017, 18, 1534. [Google Scholar] [CrossRef] [Green Version]
- Murphy, S.K.; Yang, H.; Moylan, C.A.; Pang, H.; Dellinger, A.; Abdelmalek, M.F.; Garrett, M.E.; Ashley-Koch, A.; Suzuki, A.; Tillmann, H.L.; et al. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology 2013, 145, 1076–1087. [Google Scholar] [CrossRef] [Green Version]
- Pirola, C.J.; Gianotti, T.F.; Burgueno, A.L.; Rey-Funes, M.; Loidl, C.F.; Mallardi, P.; Martino, J.S.; Castano, G.O.; Sookoian, S. Epigenetic modification of liver mitochondrial DNA is associated with histological severity of nonalcoholic fatty liver disease. Gut 2013, 62, 1356–1363. [Google Scholar] [CrossRef]
- Kuramoto, J.; Arai, E.; Tian, Y.; Funahashi, N.; Hiramoto, M.; Nammo, T.; Nozaki, Y.; Takahashi, Y.; Ito, N.; Shibuya, A.; et al. Genome-wide DNA methylation analysis during non-alcoholic steatohepatitis-related multistage hepatocarcinogenesis: Comparison with hepatitis virus-related carcinogenesis. Carcinogenesis 2017, 38, 261–270. [Google Scholar] [CrossRef]
- Pogribny, I.P.; Shpyleva, S.I.; Muskhelishvili, L.; Bagnyukova, T.V.; James, S.J.; Beland, F.A. Role of DNA damage and alterations in cytosine DNA methylation in rat liver carcinogenesis induced by a methyl-deficient diet. Mutat. Res. 2009, 669, 56–62. [Google Scholar] [CrossRef]
- Tian, Y.; Wong, V.W.S.; Wong, G.L.H.; Yang, W.; Sun, H.; Shen, J.; Tong, J.H.M.; Go, M.Y.Y.; Cheung, Y.S.; Lai, P.B.S.; et al. Histone Deacetylase HDAC8 Promotes Insulin Resistance and β-Catenin Activation in NAFLD-Associated Hepatocellular Carcinoma. Cancer Res. 2015, 75, 4803–4816. [Google Scholar] [CrossRef] [Green Version]
- de Conti, A.; Dreval, K.; Tryndyak, V.; Orisakwe, O.E.; Ross, S.A.; Beland, F.A.; Pogribny, I.P. Inhibition of the Cell Death Pathway in Nonalcoholic Steatohepatitis (NASH)-Related Hepatocarcinogenesis Is Associated with Histone H4 lysine 16 Deacetylation. Mol. Cancer Res. 2017, 15, 1163–1172. [Google Scholar] [CrossRef] [Green Version]
- Panera, N.; Gnani, D.; Crudele, A.; Ceccarelli, S.; Nobili, V.; Alisi, A. MicroRNAs as controlled systems and controllers in non-alcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 15079–15086. [Google Scholar] [CrossRef]
- Meroni, M.; Longo, M.; Erconi, V.; Valenti, L.; Gatti, S.; Fracanzani, A.L.; Dongiovanni, P. mir-101-3p Downregulation Promotes Fibrogenesis by Facilitating Hepatic Stellate Cell Transdifferentiation During Insulin Resistance. Nutrients 2019, 11, 2597. [Google Scholar] [CrossRef] [Green Version]
- Takaki, Y.; Saito, Y.; Takasugi, A.; Toshimitsu, K.; Yamada, S.; Muramatsu, T.; Kimura, M.; Sugiyama, K.; Suzuki, H.; Arai, E.; et al. Silencing of microRNA-122 is an early event during hepatocarcinogenesis from non-alcoholic steatohepatitis. Cancer Sci. 2014, 105, 1254–1260. [Google Scholar] [CrossRef]
- Liu, B.; Sun, T.; Wu, G.; Shang-Guan, H.; Jiang, Z.J.; Zhang, J.R.; Zheng, Y.F. MiR-15a suppresses hepatocarcinoma cell migration and invasion by directly targeting cMyb. Am. J. Transl. Res. 2017, 9, 520–532. [Google Scholar]
- Aqeilan, R.I.; Calin, G.A.; Croce, C.M. miR-15a and miR-16-1 in cancer: Discovery, function and future perspectives. Cell Death Differ. 2010, 17, 215–220. [Google Scholar] [CrossRef] [Green Version]
- Wu, W.L.; Wang, W.Y.; Yao, W.Q.; Li, G.D. Suppressive effects of microRNA-16 on the proliferation, invasion and metastasis of hepatocellular carcinoma cells. Int. J. Mol. Med. 2015, 36, 1713–1719. [Google Scholar] [CrossRef] [Green Version]
- Sun, T.Y.; Xie, H.J.; Li, Z.; Kong, L.F.; Gou, X.N.; Li, D.J.; Shi, Y.J.; Ding, Y.Z. miR-34a regulates HDAC1 expression to affect the proliferation and apoptosis of hepatocellular carcinoma. Am. J. Transl. Res. 2017, 9, 103–114. [Google Scholar]
- Xia, C.; Shui, L.; Lou, G.; Ye, B.; Zhu, W.; Wang, J.; Wu, S.; Xu, X.; Mao, L.; Xu, W.; et al. 0404 inhibits hepatocellular carcinoma through a p53/miR-34a/SIRT1 positive feedback loop. Sci. Rep. 2017, 7, 4396. [Google Scholar] [CrossRef] [Green Version]
- Gramantieri, L.; Fornari, F.; Ferracin, M.; Veronese, A.; Sabbioni, S.; Calin, G.A.; Grazi, G.L.; Croce, C.M.; Bolondi, L.; Negrini, M. MicroRNA-221 targets Bmf in hepatocellular carcinoma and correlates with tumor multifocality. Clin. Cancer Res. 2009, 15, 5073–5081. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Wang, C.; Jiao, X.; Zhao, S.; Liu, X.; Wang, Y.; Zhang, J. miR-221 promotes growth and invasion of hepatocellular carcinoma cells by constitutive activation of NFκB. Am. J. Transl. Res. 2016, 8, 4764–4777. [Google Scholar]
- Fornari, F.; Pollutri, D.; Patrizi, C.; La Bella, T.; Marinelli, S.; Casadei Gardini, A.; Marisi, G.; Baron Toaldo, M.; Baglioni, M.; Salvatore, V.; et al. In Hepatocellular Carcinoma miR-221 Modulates Sorafenib Resistance through Inhibition of Caspase-3-Mediated Apoptosis. Clin. Cancer Res. 2017, 23, 3953–3965. [Google Scholar] [CrossRef] [Green Version]
- Dongiovanni, P.; Meroni, M.; Baselli, G.A.; Bassani, G.A.; Rametta, R.; Pietrelli, A.; Maggioni, M.; Facciotti, F.; Trunzo, V.; Badiali, S.; et al. Insulin resistance promotes Lysyl Oxidase Like 2 induction and fibrosis accumulation in non-alcoholic fatty liver disease. Clin. Sci. 2017, 131, 1301–1315. [Google Scholar] [CrossRef] [Green Version]
- Park, J.; Morley, T.S.; Kim, M.; Clegg, D.J.; Scherer, P.E. Obesity and cancer--mechanisms underlying tumour progression and recurrence. Nat. Rev. Endocrinol. 2014, 10, 455–465. [Google Scholar] [CrossRef] [Green Version]
- Donadon, V.; Balbi, M.; Zanette, G. Hyperinsulinemia and risk for hepatocellular carcinoma in patients with chronic liver diseases and Type 2 diabetes mellitus. Expert. Rev. Gastroenterol. Hepatol. 2009, 3, 465–467. [Google Scholar] [CrossRef]
- Kubes, P.; Mehal, W.Z. Sterile inflammation in the liver. Gastroenterology 2012, 143, 1158–1172. [Google Scholar] [CrossRef] [PubMed]
- Afonso, M.B.; Rodrigues, P.M.; Carvalho, T.; Caridade, M.; Borralho, P.; Cortez-Pinto, H.; Castro, R.E.; Rodrigues, C.M. Necroptosis is a key pathogenic event in human and experimental murine models of non-alcoholic steatohepatitis. Clin. Sci. 2015, 129, 721–739. [Google Scholar] [CrossRef] [PubMed]
- Park, E.J.; Lee, J.H.; Yu, G.Y.; He, G.; Ali, S.R.; Holzer, R.G.; Osterreicher, C.H.; Takahashi, H.; Karin, M. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 2010, 140, 197–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stefanou, N.; Papanikolaou, V.; Furukawa, Y.; Nakamura, Y.; Tsezou, A. Leptin as a critical regulator of hepatocellular carcinoma development through modulation of human telomerase reverse transcriptase. BMC Cancer 2010, 10, 442. [Google Scholar] [CrossRef] [Green Version]
- He, G.; Karin, M. NF-κB and STAT3—Key players in liver inflammation and cancer. Cell Res. 2011, 21, 159–168. [Google Scholar] [CrossRef] [Green Version]
- Bozaykut, P.; Sahin, A.; Karademir, B.; Ozer, N.K. Endoplasmic reticulum stress related molecular mechanisms in nonalcoholic steatohepatitis. Mech. Ageing Dev. 2016, 157, 17–29. [Google Scholar] [CrossRef]
- Meroni, M.; Longo, M.; Dongiovanni, P. Alcohol or Gut Microbiota: Who Is the Guilty? Int. J. Mol. Sci. 2019, 20, 4568. [Google Scholar] [CrossRef] [Green Version]
- Lindstedt, G.; Lindstedt, S.; Gustafsson, B.E. Mucus in Intestinal Contents of Germfree Rats. J. Exp. Med. 1965, 121, 201–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Fouts, D.E.; Starkel, P.; Hartmann, P.; Chen, P.; Llorente, C.; DePew, J.; Moncera, K.; Ho, S.B.; Brenner, D.A.; et al. Intestinal REG3 Lectins Protect against Alcoholic Steatohepatitis by Reducing Mucosa-Associated Microbiota and Preventing Bacterial Translocation. Cell Host Microbe 2016, 19, 227–239. [Google Scholar] [CrossRef] [Green Version]
- Cani, P.D.; Osto, M.; Geurts, L.; Everard, A. Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes 2012, 3, 279–288. [Google Scholar] [CrossRef] [Green Version]
- Boulange, C.L.; Neves, A.L.; Chilloux, J.; Nicholson, J.K.; Dumas, M.E. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 2016, 8, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giorgio, V.; Miele, L.; Principessa, L.; Ferretti, F.; Villa, M.P.; Negro, V.; Grieco, A.; Alisi, A.; Nobili, V. Intestinal permeability is increased in children with non-alcoholic fatty liver disease, and correlates with liver disease severity. Dig. Liver Dis. 2014, 46, 556–560. [Google Scholar] [CrossRef] [PubMed]
- Miele, L.; Valenza, V.; La Torre, G.; Montalto, M.; Cammarota, G.; Ricci, R.; Masciana, R.; Forgione, A.; Gabrieli, M.L.; Perotti, G.; et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 2009, 49, 1877–1887. [Google Scholar] [CrossRef] [PubMed]
- Bajaj, J.S. Alcohol, liver disease and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 235–246. [Google Scholar] [CrossRef]
- Martinez-Medina, M.; Denizot, J.; Dreux, N.; Robin, F.; Billard, E.; Bonnet, R.; Darfeuille-Michaud, A.; Barnich, N. Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation. Gut 2014, 63, 116–124. [Google Scholar] [CrossRef]
- Agus, A.; Denizot, J.; Thevenot, J.; Martinez-Medina, M.; Massier, S.; Sauvanet, P.; Bernalier-Donadille, A.; Denis, S.; Hofman, P.; Bonnet, R.; et al. Western diet induces a shift in microbiota composition enhancing susceptibility to Adherent-Invasive E. coli infection and intestinal inflammation. Sci. Rep. 2016, 6, 19032. [Google Scholar] [CrossRef] [Green Version]
- Singh, R.K.; Chang, H.W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [Green Version]
- Hills, R.D., Jr.; Pontefract, B.A.; Mishcon, H.R.; Black, C.A.; Sutton, S.C. Gut Microbiome: Profound Implications for Diet and Disease. Nutrients 2019, 11, 1613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Del Chierico, F.; Nobili, V.; Vernocchi, P.; Russo, A.; De Stefanis, C.; Gnani, D.; Furlanello, C.; Zandona, A.; Paci, P.; Capuani, G.; et al. Gut microbiota profiling of pediatric nonalcoholic fatty liver disease and obese patients unveiled by an integrated meta-omics-based approach. Hepatology 2017, 65, 451–464. [Google Scholar] [CrossRef] [PubMed]
- Delarue, J.; Lalles, J.P. Nonalcoholic fatty liver disease: Roles of the gut and the liver and metabolic modulation by some dietary factors and especially long-chain n-3 PUFA. Mol. Nutr. Food Res. 2016, 60, 147–159. [Google Scholar] [CrossRef]
- Bibbo, S.; Ianiro, G.; Dore, M.P.; Simonelli, C.; Newton, E.E.; Cammarota, G. Gut Microbiota as a Driver of Inflammation in Nonalcoholic Fatty Liver Disease. Mediat. Inflamm. 2018, 2018, 9321643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Puri, P.; Sanyal, A.J. The Intestinal Microbiome in Nonalcoholic Fatty Liver Disease. Clin. Liver Dis. 2018, 22, 121–132. [Google Scholar] [CrossRef] [PubMed]
- Borrelli, A.; Bonelli, P.; Tuccillo, F.M.; Goldfine, I.D.; Evans, J.L.; Buonaguro, F.M.; Mancini, A. Role of gut microbiota and oxidative stress in the progression of non-alcoholic fatty liver disease to hepatocarcinoma: Current and innovative therapeutic approaches. Redox Biol. 2018, 15, 467–479. [Google Scholar] [CrossRef] [PubMed]
- Zhu, L.; Baker, S.S.; Gill, C.; Liu, W.; Alkhouri, R.; Baker, R.D.; Gill, S.R. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH. Hepatology 2013, 57, 601–609. [Google Scholar] [CrossRef]
- Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metab. Clin. Exp. 2016, 65, 1038–1048. [Google Scholar] [CrossRef]
- Schnabl, B.; Brenner, D.A. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 2014, 146, 1513–1524. [Google Scholar] [CrossRef] [Green Version]
- Kolodziejczyk, A.A.; Zheng, D.; Shibolet, O.; Elinav, E. The role of the microbiome in NAFLD and NASH. EMBO Mol. Med. 2019, 11, e9302. [Google Scholar] [CrossRef]
- Ivanov, I.I.; Frutos Rde, L.; Manel, N.; Yoshinaga, K.; Rifkin, D.B.; Sartor, R.B.; Finlay, B.B.; Littman, D.R. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 2008, 4, 337–349. [Google Scholar] [CrossRef] [Green Version]
- Boursier, J.; Diehl, A.M. Implication of gut microbiota in nonalcoholic fatty liver disease. PLoS Pathog. 2015, 11, e1004559. [Google Scholar] [CrossRef] [Green Version]
- Nolan, J.P. The role of intestinal endotoxin in liver injury: A long and evolving history. Hepatology 2010, 52, 1829–1835. [Google Scholar] [CrossRef]
- Ren, Z.; Li, A.; Jiang, J.; Zhou, L.; Yu, Z.; Lu, H.; Xie, H.; Chen, X.; Shao, L.; Zhang, R.; et al. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut 2019, 68, 1014–1023. [Google Scholar] [CrossRef] [PubMed]
- Yu, L.X.; Yan, H.X.; Liu, Q.; Yang, W.; Wu, H.P.; Dong, W.; Tang, L.; Lin, Y.; He, Y.Q.; Zou, S.S.; et al. Endotoxin accumulation prevents carcinogen-induced apoptosis and promotes liver tumorigenesis in rodents. Hepatology 2010, 52, 1322–1333. [Google Scholar] [CrossRef] [PubMed]
- Dapito, D.H.; Mencin, A.; Gwak, G.Y.; Pradere, J.P.; Jang, M.K.; Mederacke, I.; Caviglia, J.M.; Khiabanian, H.; Adeyemi, A.; Bataller, R.; et al. Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell 2012, 21, 504–516. [Google Scholar] [CrossRef] [Green Version]
- Schwabe, R.F.; Jobin, C. The microbiome and cancer. Nat. Rev. Cancer 2013, 13, 800–812. [Google Scholar] [CrossRef] [Green Version]
- Ray, K. Gut microbiota: Obesity-induced microbial metabolite promotes HCC. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 442. [Google Scholar] [CrossRef] [PubMed]
- Moussa, I.; Day, R.S.; Li, R.; Du, X.L.; Kaseb, A.O.; Jalal, P.K.; Daniel-MacDougall, C.; Hatia, R.I.; Abdelhakeem, A.; Rashid, A.; et al. Dietary Patterns and Hepatocellular Carcinoma Risk among US Adults. Nutrients 2021, 13, 2011. [Google Scholar] [CrossRef]
- Yang, Y.; Zhang, D.; Feng, N.; Chen, G.; Liu, J.; Chen, G.; Zhu, Y. Increased intake of vegetables, but not fruit, reduces risk for hepatocellular carcinoma: A meta-analysis. Gastroenterology 2014, 147, 1031–1042. [Google Scholar] [CrossRef] [Green Version]
- Turati, F.; Trichopoulos, D.; Polesel, J.; Bravi, F.; Rossi, M.; Talamini, R.; Franceschi, S.; Montella, M.; Trichopoulou, A.; La Vecchia, C.; et al. Mediterranean diet and hepatocellular carcinoma. J. Hepatol. 2014, 60, 606–611. [Google Scholar] [CrossRef]
- Duarte-Salles, T.; Fedirko, V.; Stepien, M.; Aleksandrova, K.; Bamia, C.; Lagiou, P.; Laursen, A.S.; Hansen, L.; Overvad, K.; Tjønneland, A.; et al. Dietary fat, fat subtypes and hepatocellular carcinoma in a large European cohort. Int. J. Cancer 2015, 137, 2715–2728. [Google Scholar] [CrossRef] [Green Version]
- Fedirko, V.; Trichopolou, A.; Bamia, C.; Duarte-Salles, T.; Trepo, E.; Aleksandrova, K.; Nöthlings, U.; Lukanova, A.; Lagiou, P.; Boffetta, P.; et al. Consumption of fish and meats and risk of hepatocellular carcinoma: The European Prospective Investigation into Cancer and Nutrition (EPIC). Ann. Oncol. 2013, 24, 2166–2173. [Google Scholar] [CrossRef] [PubMed]
- Fedirko, V.; Lukanova, A.; Bamia, C.; Trichopolou, A.; Trepo, E.; Nöthlings, U.; Schlesinger, S.; Aleksandrova, K.; Boffetta, P.; Tjønneland, A.; et al. Glycemic index, glycemic load, dietary carbohydrate, and dietary fiber intake and risk of liver and biliary tract cancers in Western Europeans. Ann. Oncol. 2013, 24, 543–553. [Google Scholar] [CrossRef] [PubMed]
- Luo, J.; Yang, Y.; Liu, J.; Lu, K.; Tang, Z.; Liu, P.; Liu, L.; Zhu, Y. Systematic review with meta-analysis: Meat consumption and the risk of hepatocellular carcinoma. Aliment. Pharmacol. Ther. 2014, 39, 913–922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bagnardi, V.; Blangiardo, M.; La Vecchia, C.; Corrao, G. A meta-analysis of alcohol drinking and cancer risk. Br. J. Cancer 2001, 85, 1700–1705. [Google Scholar] [CrossRef]
- Matsushita, H.; Takaki, A. Alcohol and hepatocellular carcinoma. BMJ Open Gastroenterol. 2019, 6, e000260. [Google Scholar] [CrossRef]
- Meroni, M.; Longo, M.; Rametta, R.; Dongiovanni, P. Genetic and Epigenetic Modifiers of Alcoholic Liver Disease. Int. J. Mol. Sci. 2018, 19, 3857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, P.; Kang, D.; Cao, W.; Wang, Y.; Liu, Z. Diabetes mellitus and risk of hepatocellular carcinoma: A systematic review and meta-analysis. Diabetes/Metab. Res. Rev. 2012, 28, 109–122. [Google Scholar] [CrossRef]
- Loomba, R.; Yang, H.I.; Su, J.; Brenner, D.; Barrett-Connor, E.; Iloeje, U.; Chen, C.J. Synergism between obesity and alcohol in increasing the risk of hepatocellular carcinoma: A prospective cohort study. Am. J. Epidemiol. 2013, 177, 333–342. [Google Scholar] [CrossRef] [Green Version]
- Khan, F.Z.; Perumpail, R.B.; Wong, R.J.; Ahmed, A. Advances in hepatocellular carcinoma: Nonalcoholic steatohepatitis-related hepatocellular carcinoma. World J. Hepatol. 2015, 7, 2155–2161. [Google Scholar] [CrossRef] [Green Version]
- Morgan, T.R.; Mandayam, S.; Jamal, M.M. Alcohol and hepatocellular carcinoma. Gastroenterology 2004, 127, S87–S96. [Google Scholar] [CrossRef]
- Donohue, T.M., Jr. Alcohol-induced steatosis in liver cells. World J. Gastroenterol. 2007, 13, 4974–4978. [Google Scholar] [CrossRef]
- Svegliati-Baroni, G.; Inagaki, Y.; Rincon-Sanchez, A.R.; Else, C.; Saccomanno, S.; Benedetti, A.; Ramirez, F.; Rojkind, M. Early response of α2(I) collagen to acetaldehyde in human hepatic stellate cells is TGF-β independent. Hepatology 2005, 42, 343–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, B.; Bataller, R. Alcoholic liver disease: Pathogenesis and new therapeutic targets. Gastroenterology 2011, 141, 1572–1585. [Google Scholar] [CrossRef] [Green Version]
- Plaz Torres, M.C.; Bodini, G.; Furnari, M.; Marabotto, E.; Zentilin, P.; Giannini, E.G. Nuts and Non-Alcoholic Fatty Liver Disease: Are Nuts Safe for Patients with Fatty Liver Disease? Nutrients 2020, 12, 3363. [Google Scholar] [CrossRef]
- Nault, J.C.; Cheng, A.L.; Sangro, B.; Llovet, J.M. Milestones in the pathogenesis and management of primary liver cancer. J. Hepatol. 2020, 72, 209–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Plaz Torres, M.C.; Bodini, G.; Furnari, M.; Marabotto, E.; Zentilin, P.; Strazzabosco, M.; Giannini, E.G. Surveillance for Hepatocellular Carcinoma in Patients with Non-Alcoholic Fatty Liver Disease: Universal or Selective? Cancers 2020, 12, 1422. [Google Scholar] [CrossRef] [PubMed]
- Kanwal, F.; Kramer, J.R.; Mapakshi, S.; Natarajan, Y.; Chayanupatkul, M.; Richardson, P.A.; Li, L.; Desiderio, R.; Thrift, A.P.; Asch, S.M.; et al. Risk of Hepatocellular Cancer in Patients with Non-Alcoholic Fatty Liver Disease. Gastroenterology 2018, 155, 1828–1837.e1822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luyendyk, J.P.; Shores, K.C.; Ganey, P.E.; Roth, R.A. Bacterial lipopolysaccharide exposure alters aflatoxin B(1) hepatotoxicity: Benchmark dose analysis for markers of liver injury. Toxicol. Sci. 2002, 68, 220–225. [Google Scholar] [CrossRef] [Green Version]
- Barton, C.C.; Barton, E.X.; Ganey, P.E.; Kunkel, S.L.; Roth, R.A. Bacterial lipopolysaccharide enhances aflatoxin B1 hepatotoxicity in rats by a mechanism that depends on tumor necrosis factor alpha. Hepatology 2001, 33, 66–73. [Google Scholar] [CrossRef] [PubMed]
- Gao, Y.; Meng, L.; Liu, H.; Wang, J.; Zheng, N. The Compromised Intestinal Barrier Induced by Mycotoxins. Toxins 2020, 12, 619. [Google Scholar] [CrossRef] [PubMed]
- Wogan, G.N.; Kensler, T.W.; Groopman, J.D. Present and future directions of translational research on aflatoxin and hepatocellular carcinoma. A review. Food Addit. Contam. Part A 2012, 29, 249–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kensler, T.W.; Chen, J.G.; Egner, P.A.; Fahey, J.W.; Jacobson, L.P.; Stephenson, K.K.; Ye, L.; Coady, J.L.; Wang, J.B.; Wu, Y.; et al. Effects of glucosinolate-rich broccoli sprouts on urinary levels of aflatoxin-DNA adducts and phenanthrene tetraols in a randomized clinical trial in He Zuo township, Qidong, People’s Republic of China. Cancer Epidemiol. Biomark. Prev. 2005, 14, 2605–2613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soni, K.B.; Lahiri, M.; Chackradeo, P.; Bhide, S.V.; Kuttan, R. Protective effect of food additives on aflatoxin-induced mutagenicity and hepatocarcinogenicity. Cancer Lett. 1997, 115, 129–133. [Google Scholar] [CrossRef]
- Bishayee, A.; Politis, T.; Darvesh, A.S. Resveratrol in the chemoprevention and treatment of hepatocellular carcinoma. Cancer Treat. Rev. 2010, 36, 43–53. [Google Scholar] [CrossRef] [PubMed]
- Rametta, R.; Meroni, M.; Dongiovanni, P. From Environment to Genome and Back: A Lesson from HFE Mutations. Int. J. Mol. Sci. 2020, 21, 3505. [Google Scholar] [CrossRef]
- Sorrentino, P.; D’Angelo, S.; Ferbo, U.; Micheli, P.; Bracigliano, A.; Vecchione, R. Liver iron excess in patients with hepatocellular carcinoma developed on non-alcoholic steato-hepatitis. J. Hepatol. 2009, 50, 351–357. [Google Scholar] [CrossRef]
- George, D.K.; Goldwurm, S.; MacDonald, G.A.; Cowley, L.L.; Walker, N.I.; Ward, P.J.; Jazwinska, E.C.; Powell, L.W. Increased hepatic iron concentration in nonalcoholic steatohepatitis is associated with increased fibrosis. Gastroenterology 1998, 114, 311–318. [Google Scholar] [CrossRef]
- Abe, N.; Tsuchida, T.; Yasuda, S.I.; Oka, K. Dietary iron restriction leads to a reduction in hepatic fibrosis in a rat model of non-alcoholic steatohepatitis. Biol. Open 2019, 8, bio040519. [Google Scholar] [CrossRef] [Green Version]
- Qiao, Y.; Zhang, X.; Zhang, Y.; Wang, Y.; Xu, Y.; Liu, X.; Sun, F.; Wang, J. High Glucose Stimulates Tumorigenesis in Hepatocellular Carcinoma Cells Through AGER-Dependent O-GlcNAcylation of c-Jun. Diabetes 2016, 65, 619–632. [Google Scholar] [CrossRef] [Green Version]
- Van Rooyen, D.M.; Larter, C.Z.; Haigh, W.G.; Yeh, M.M.; Ioannou, G.; Kuver, R.; Lee, S.P.; Teoh, N.C.; Farrell, G.C. Hepatic free cholesterol accumulates in obese, diabetic mice and causes nonalcoholic steatohepatitis. Gastroenterology 2011, 141, 1393–1403.E5. [Google Scholar] [CrossRef] [Green Version]
- Ioannou, G.N.; Morrow, O.B.; Connole, M.L.; Lee, S.P. Association between dietary nutrient composition and the incidence of cirrhosis or liver cancer in the United States population. Hepatology 2009, 50, 175–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matsuzawa, N.; Takamura, T.; Kurita, S.; Misu, H.; Ota, T.; Ando, H.; Yokoyama, M.; Honda, M.; Zen, Y.; Nakanuma, Y.; et al. Lipid-induced oxidative stress causes steatohepatitis in mice fed an atherogenic diet. Hepatology 2007, 46, 1392–1403. [Google Scholar] [CrossRef]
- He, F.; Ru, X.; Wen, T. NRF2, a Transcription Factor for Stress Response and Beyond. Int. J. Mol. Sci. 2020, 21, 4777. [Google Scholar] [CrossRef] [PubMed]
- Marí, M.; Morales, A.; Colell, A.; García-Ruiz, C.; Fernández-Checa, J.C. Mitochondrial cholesterol accumulation in alcoholic liver disease: Role of ASMase and endoplasmic reticulum stress. Redox Biol. 2014, 3, 100–108. [Google Scholar] [CrossRef] [Green Version]
- Leroux, A.; Ferrere, G.; Godie, V.; Cailleux, F.; Renoud, M.L.; Gaudin, F.; Naveau, S.; Prévot, S.; Makhzami, S.; Perlemuter, G.; et al. Toxic lipids stored by Kupffer cells correlates with their pro-inflammatory phenotype at an early stage of steatohepatitis. J. Hepatol. 2012, 57, 141–149. [Google Scholar] [CrossRef]
- Caballero, F.; Fernández, A.; De Lacy, A.M.; Fernández-Checa, J.C.; Caballería, J.; García-Ruiz, C. Enhanced free cholesterol, SREBP-2 and StAR expression in human NASH. J. Hepatol. 2009, 50, 789–796. [Google Scholar] [CrossRef]
- Armstrong, L.E.; Guo, G.L. Role of FXR in Liver Inflammation during Nonalcoholic Steatohepatitis. Curr. Pharmacol. Rep. 2017, 3, 92–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carr, B.I.; Giannelli, G.; Guerra, V.; Giannini, E.G.; Farinati, F.; Rapaccini, G.L.; Marco, M.D.; Zoli, M.; Caturelli, E.; Masotto, A.; et al. Plasma cholesterol and lipoprotein levels in relation to tumor aggressiveness and survival in HCC patients. Int. J. Biol. Markers 2018, 33, 423–431. [Google Scholar] [CrossRef] [Green Version]
- Liang, J.Q.; Teoh, N.; Xu, L.; Pok, S.; Li, X.; Chu, E.S.H.; Chiu, J.; Dong, L.; Arfianti, E. Dietary cholesterol promotes steatohepatitis related hepatocellular carcinoma through dysregulated metabolism and calcium signaling. Nat. Commun. 2018, 9, 4490. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Coker, O.O.; Chu, E.S.; Fu, K.; Lau, H.C.H.; Wang, Y.-X.; Chan, A.W.H.; Wei, H.; Yang, X.; Sung, J.J.Y.; et al. Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut 2021, 70, 761–774. [Google Scholar] [CrossRef]
- Tran, K.T.; McMenamin, Ú.C.; Coleman, H.G.; Cardwell, C.R.; Murchie, P.; Iversen, L.; Lee, A.J.; Thrift, A.P. Statin use and risk of liver cancer: Evidence from two population-based studies. Int. J. Cancer 2020, 146, 1250–1260. [Google Scholar] [CrossRef] [PubMed]
- Cruz, P.M.; Mo, H.; McConathy, W.J.; Sabnis, N.; Lacko, A.G. The role of cholesterol metabolism and cholesterol transport in carcinogenesis: A review of scientific findings, relevant to future cancer therapeutics. Front. Pharmacol. 2013, 4, 119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nelson, E.R. The significance of cholesterol and its metabolite, 27-hydroxycholesterol in breast cancer. Mol. Cell. Endocrinol. 2018, 466, 73–80. [Google Scholar] [CrossRef]
- Bhurwal, A.; Rattan, P.; Yoshitake, S.; Pioppo, L.; Reja, D.; Dellatore, P.; Rustgi, V. Inverse Association of Coffee with Liver Cancer Development: An Updated Systematic Review and Meta-analysis. J. Gastrointest. Liver Dis. 2020, 29, 421–428. [Google Scholar] [CrossRef] [PubMed]
- Kennedy, O.J.; Fallowfield, J.A.; Poole, R.; Hayes, P.C.; Parkes, J.; Roderick, P.J. All coffee types decrease the risk of adverse clinical outcomes in chronic liver disease: A UK Biobank study. BMC Public Health 2021, 21, 970. [Google Scholar] [CrossRef] [PubMed]
- Bravi, F.; Bosetti, C.; Tavani, A.; La Vecchia, C. Coffee drinking and hepatocellular carcinoma: An update. Hepatology 2009, 50, 1317–1318. [Google Scholar] [CrossRef]
- Tao, K.S.; Wang, W.; Wang, L.; Cao, D.Y.; Li, Y.Q.; Wu, S.X.; Dou, K.F. The multifaceted mechanisms for coffee’s anti-tumorigenic effect on liver. Med. Hypotheses 2008, 71, 730–736. [Google Scholar] [CrossRef]
- Darvesh, A.S.; Bishayee, A. Chemopreventive and therapeutic potential of tea polyphenols in hepatocellular cancer. Nutr. Cancer 2013, 65, 329–344. [Google Scholar] [CrossRef]
- Johnson, L.J.; Meacham, S.L.; Kruskall, L.J. The antioxidants—Vitamin C, vitamin E, selenium, and carotenoids. J. Agromed. 2003, 9, 65–82. [Google Scholar] [CrossRef]
- Glauert, H.P.; Calfee-Mason, K.; Stemm, D.N.; Tharappel, J.C.; Spear, B.T. Dietary antioxidants in the prevention of hepatocarcinogenesis: A review. Mol. Nutr. Food Res. 2010, 54, 875–896. [Google Scholar] [CrossRef]
- Ko, W.S.; Yang, Y.P.; Shen, F.P.; Wu, M.C.; Shih, C.J.; Lu, M.C.; Yan, Y.H.; Chiou, Y.L. The Study of Correlation Between Serum Vitamin D(3) Concentrations and HBV DNA Levels and Immune Response in Chronic Hepatitis Patients. Nutrients 2020, 12, 1114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adelani, I.B.; Rotimi, O.A.; Maduagwu, E.N.; Rotimi, S.O. Vitamin D: Possible Therapeutic Roles in Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 642653. [Google Scholar] [CrossRef]
- Yi, Z.; Wang, L.; Tu, X. Effect of Vitamin D Deficiency on Liver Cancer Risk: A Systematic Review and Meta-Analysis. Asian Pac. J. Cancer Prev. 2021, 22, 991–997. [Google Scholar] [CrossRef] [PubMed]
- Yoshimoto, S.; Loo, T.M.; Atarashi, K.; Kanda, H.; Sato, S.; Oyadomari, S.; Iwakura, Y.; Oshima, K.; Morita, H.; Hattori, M.; et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013, 499, 97–101. [Google Scholar] [CrossRef] [PubMed]
- Devkota, S.; Wang, Y.; Musch, M.W.; Leone, V.; Fehlner-Peach, H.; Nadimpalli, A.; Antonopoulos, D.A.; Jabri, B.; Chang, E.B. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice. Nature 2012, 487, 104–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meroni, M.; Longo, M.; Dongiovanni, P. The Role of Probiotics in Nonalcoholic Fatty Liver Disease: A New Insight into Therapeutic Strategies. Nutrients 2019, 11, 2642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2014, 7, 17–44. [Google Scholar] [CrossRef]
- Chassard, C.; Lacroix, C. Carbohydrates and the human gut microbiota. Curr. Opin. Clin. Nutr. Metab. Care 2013, 16, 453–460. [Google Scholar] [CrossRef]
- Rivière, A.; Selak, M.; Lantin, D.; Leroy, F.; De Vuyst, L. Bifidobacteria and Butyrate-Producing Colon Bacteria: Importance and Strategies for Their Stimulation in the Human Gut. Front. Microbiol. 2016, 7, 979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Behary, J.; Amorim, N.; Jiang, X.T.; Raposo, A.; Gong, L.; McGovern, E.; Ibrahim, R.; Chu, F.; Stephens, C.; Jebeili, H.; et al. Gut microbiota impact on the peripheral immune response in non-alcoholic fatty liver disease related hepatocellular carcinoma. Nat. Commun. 2021, 12, 187. [Google Scholar] [CrossRef]
- Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y.M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Asarat, M.; Apostolopoulos, V.; Vasiljevic, T.; Donkor, O. Short-Chain Fatty Acids Regulate Cytokines and Th17/Treg Cells in Human Peripheral Blood Mononuclear Cells in vitro. Immunol. Investig. 2016, 45, 205–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, M.; Zhou, Q.; Dorfman, R.G.; Huang, X.; Fan, T.; Zhang, H.; Zhang, J.; Yu, C. Butyrate inhibits interleukin-17 and generates Tregs to ameliorate colorectal colitis in rats. BMC Gastroenterol. 2016, 16, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef] [PubMed]
- Singh, V.; Yeoh, B.S.; Chassaing, B.; Xiao, X.; Saha, P.; Aguilera Olvera, R.; Lapek, J.D., Jr.; Zhang, L.; Wang, W.B.; Hao, S.; et al. Dysregulated Microbial Fermentation of Soluble Fiber Induces Cholestatic Liver Cancer. Cell 2018, 175, 679–694.e622. [Google Scholar] [CrossRef] [Green Version]
- Qin, Y.; Wade, P.A. Crosstalk between the microbiome and epigenome: Messages from bugs. J. Biochem. 2018, 163, 105–112. [Google Scholar] [CrossRef] [Green Version]
- Krautkramer, K.A.; Kreznar, J.H.; Romano, K.A.; Vivas, E.I.; Barrett-Wilt, G.A.; Rabaglia, M.E.; Keller, M.P.; Attie, A.D.; Rey, F.E.; Denu, J.M. Diet-Microbiota Interactions Mediate Global Epigenetic Programming in Multiple Host Tissues. Mol. Cell 2016, 64, 982–992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, Q.; Ouyang, J.; Sun, F.; Yang, J. Short-Chain Fatty Acids: A Soldier Fighting Against Inflammation and Protecting from Tumorigenesis in People with Diabetes. Front. Immunol. 2020, 11, 590685. [Google Scholar] [CrossRef]
- Mirzaei, R.; Afaghi, A.; Babakhani, S.; Sohrabi, M.R.; Hosseini-Fard, S.R.; Babolhavaeji, K.; Khani Ali Akbari, S.; Yousefimashouf, R.; Karampoor, S. Role of microbiota-derived short-chain fatty acids in cancer development and prevention. Biomed. Pharmacother. Biomed. Pharmacother. 2021, 139, 111619. [Google Scholar] [CrossRef]
- Tajiri, K.; Shimizu, Y. Branched-chain amino acids in liver diseases. World J. Gastroenterol. 2013, 19, 7620–7629. [Google Scholar] [CrossRef]
- Iwasa, J.; Shimizu, M.; Shiraki, M.; Shirakami, Y.; Sakai, H.; Terakura, Y.; Takai, K.; Tsurumi, H.; Tanaka, T.; Moriwaki, H. Dietary supplementation with branched-chain amino acids suppresses diethylnitrosamine-induced liver tumorigenesis in obese and diabetic C57BL/KsJ-db/db mice. Cancer Sci. 2010, 101, 460–467. [Google Scholar] [CrossRef] [PubMed]
- Takegoshi, K.; Honda, M.; Okada, H.; Takabatake, R.; Matsuzawa-Nagata, N.; Campbell, J.S.; Nishikawa, M.; Shimakami, T.; Shirasaki, T.; Sakai, Y.; et al. Branched-chain amino acids prevent hepatic fibrosis and development of hepatocellular carcinoma in a non-alcoholic steatohepatitis mouse model. Oncotarget 2017, 8, 18191–18205. [Google Scholar] [CrossRef]
- D’Antona, G.; Ragni, M.; Cardile, A.; Tedesco, L.; Dossena, M.; Bruttini, F.; Caliaro, F.; Corsetti, G.; Bottinelli, R.; Carruba, M.O.; et al. Branched-chain amino acid supplementation promotes survival and supports cardiac and skeletal muscle mitochondrial biogenesis in middle-aged mice. Cell Metab. 2010, 12, 362–372. [Google Scholar] [CrossRef] [Green Version]
- Kawaguchi, T.; Shiraishi, K.; Ito, T.; Suzuki, K.; Koreeda, C.; Ohtake, T.; Iwasa, M.; Tokumoto, Y.; Endo, R.; Kawamura, N.H.; et al. Branched-chain amino acids prevent hepatocarcinogenesis and prolong survival of patients with cirrhosis. Clin. Gastroenterol. Hepatol. 2014, 12, 1012–1018.e1011. [Google Scholar] [CrossRef] [PubMed]
- Hayaishi, S.; Chung, H.; Kudo, M.; Ishikawa, E.; Takita, M.; Ueda, T.; Kitai, S.; Inoue, T.; Yada, N.; Hagiwara, S.; et al. Oral branched-chain amino acid granules reduce the incidence of hepatocellular carcinoma and improve event-free survival in patients with liver cirrhosis. Dig. Dis. 2011, 29, 326–332. [Google Scholar] [CrossRef]
- Sawada, N.; Inoue, M.; Iwasaki, M.; Sasazuki, S.; Shimazu, T.; Yamaji, T.; Takachi, R.; Tanaka, Y.; Mizokami, M.; Tsugane, S. Consumption of n-3 fatty acids and fish reduces risk of hepatocellular carcinoma. Gastroenterology 2012, 142, 1468–1475. [Google Scholar] [CrossRef] [PubMed]
- Lim, K.; Han, C.; Dai, Y.; Shen, M.; Wu, T. Omega-3 polyunsaturated fatty acids inhibit hepatocellular carcinoma cell growth through blocking beta-catenin and cyclooxygenase-2. Mol. Cancer Ther. 2009, 8, 3046–3055. [Google Scholar] [CrossRef] [Green Version]
- Da Silva-Santi, L.G.; Antunes, M.M.; Caparroz-Assef, S.M.; Carbonera, F.; Masi, L.N.; Curi, R.; Visentainer, J.V.; Bazotte, R.B. Liver Fatty Acid Composition and Inflammation in Mice Fed with High-Carbohydrate Diet or High-Fat Diet. Nutrients 2016, 8, 682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patterson, E.; Wall, R.; Fitzgerald, G.F.; Ross, R.P.; Stanton, C. Health implications of high dietary omega-6 polyunsaturated Fatty acids. J. Nutr. Metab. 2012, 2012, 539426. [Google Scholar] [CrossRef]
- Pikarsky, E.; Porat, R.M.; Stein, I.; Abramovitch, R.; Amit, S.; Kasem, S.; Gutkovich-Pyest, E.; Urieli-Shoval, S.; Galun, E.; Ben-Neriah, Y. NF-κB functions as a tumour promoter in inflammation-associated cancer. Nature 2004, 431, 461–466. [Google Scholar] [CrossRef]
- Dowman, J.K.; Hopkins, L.J.; Reynolds, G.M.; Nikolaou, N.; Armstrong, M.J.; Shaw, J.C.; Houlihan, D.D.; Lalor, P.F.; Tomlinson, J.W.; Hübscher, S.G.; et al. Development of hepatocellular carcinoma in a murine model of nonalcoholic steatohepatitis induced by use of a high-fat/fructose diet and sedentary lifestyle. Am. J. Pathol. 2014, 184, 1550–1561. [Google Scholar] [CrossRef] [Green Version]
- Wolf, M.J.; Adili, A.; Piotrowitz, K.; Abdullah, Z.; Boege, Y.; Stemmer, K.; Ringelhan, M.; Simonavicius, N.; Egger, M.; Wohlleber, D.; et al. Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell 2014, 26, 549–564. [Google Scholar] [CrossRef] [Green Version]
- Kishida, N.; Matsuda, S.; Itano, O.; Shinoda, M.; Kitago, M.; Yagi, H.; Abe, Y.; Hibi, T.; Masugi, Y.; Aiura, K.; et al. Development of a novel mouse model of hepatocellular carcinoma with nonalcoholic steatohepatitis using a high-fat, choline-deficient diet and intraperitoneal injection of diethylnitrosamine. BMC Gastroenterol. 2016, 16, 61. [Google Scholar] [CrossRef] [Green Version]
- Tsuchida, T.; Lee, Y.A.; Fujiwara, N.; Ybanez, M.; Allen, B.; Martins, S.; Fiel, M.I.; Goossens, N.; Chou, H.I.; Hoshida, Y.; et al. A simple diet- and chemical-induced murine NASH model with rapid progression of steatohepatitis, fibrosis and liver cancer. J. Hepatol. 2018, 69, 385–395. [Google Scholar] [CrossRef] [PubMed]
- Fujii, M.; Shibazaki, Y.; Wakamatsu, K.; Honda, Y.; Kawauchi, Y.; Suzuki, K.; Arumugam, S.; Watanabe, K.; Ichida, T.; Asakura, H.; et al. A murine model for non-alcoholic steatohepatitis showing evidence of association between diabetes and hepatocellular carcinoma. Med. Mol. Morphol. 2013, 46, 141–152. [Google Scholar] [CrossRef]
- Asgharpour, A.; Cazanave, S.C.; Pacana, T.; Seneshaw, M.; Vincent, R.; Banini, B.A.; Kumar, D.P.; Daita, K.; Min, H.K.; Mirshahi, F.; et al. A diet-induced animal model of non-alcoholic fatty liver disease and hepatocellular cancer. J. Hepatol. 2016, 65, 579–588. [Google Scholar] [CrossRef] [Green Version]
- Nakagawa, H.; Umemura, A.; Taniguchi, K.; Font-Burgada, J.; Dhar, D.; Ogata, H.; Zhong, Z.; Valasek, M.A.; Seki, E.; Hidalgo, J.; et al. ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development. Cancer Cell 2014, 26, 331–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Horie, Y.; Suzuki, A.; Kataoka, E.; Sasaki, T.; Hamada, K.; Sasaki, J.; Mizuno, K.; Hasegawa, G.; Kishimoto, H.; Iizuka, M.; et al. Hepatocyte-specific Pten deficiency results in steatohepatitis and hepatocellular carcinomas. J. Clin. Investig. 2004, 113, 1774–1783. [Google Scholar] [CrossRef] [Green Version]
- Mueller, K.M.; Kornfeld, J.W.; Friedbichler, K.; Blaas, L.; Egger, G.; Esterbauer, H.; Hasselblatt, P.; Schlederer, M.; Haindl, S.; Wagner, K.U.; et al. Impairment of hepatic growth hormone and glucocorticoid receptor signaling causes steatosis and hepatocellular carcinoma in mice. Hepatology 2011, 54, 1398–1409. [Google Scholar] [CrossRef] [PubMed]
- Martínez-López, N.; Varela-Rey, M.; Fernández-Ramos, D.; Woodhoo, A.; Vázquez-Chantada, M.; Embade, N.; Espinosa-Hevia, L.; Bustamante, F.J.; Parada, L.A.; Rodriguez, M.S.; et al. Activation of LKB1-Akt pathway independent of phosphoinositide 3-kinase plays a critical role in the proliferation of hepatocellular carcinoma from nonalcoholic steatohepatitis. Hepatology 2010, 52, 1621–1631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Itoh, M.; Suganami, T.; Nakagawa, N.; Tanaka, M.; Yamamoto, Y.; Kamei, Y.; Terai, S.; Sakaida, I.; Ogawa, Y. Melanocortin 4 receptor-deficient mice as a novel mouse model of nonalcoholic steatohepatitis. Am. J. Pathol. 2011, 179, 2454–2463. [Google Scholar] [CrossRef]
- Hirsova, P. Modeling NASH and NASH-Induced Hepatocellular Carcinoma: Faster and Better. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 1149–1150. [Google Scholar] [CrossRef]
- Ganguly, S.; Muench, G.A.; Shang, L.; Rosenthal, S.B.; Rahman, G.; Wang, R.; Wang, Y.; Kwon, H.C.; Diomino, A.M.; Kisseleva, T.; et al. Nonalcoholic Steatohepatitis and HCC in a Hyperphagic Mouse Accelerated by Western Diet. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 891–920. [Google Scholar] [CrossRef] [PubMed]
- Llovet, J.M.; Kelley, R.K.; Villanueva, A.; Singal, A.G.; Pikarsky, E.; Roayaie, S.; Lencioni, R.; Koike, K.; Zucman-Rossi, J.; Finn, R.S. Hepatocellular carcinoma. Nat. Rev. Dis. Prim. 2021, 7, 6. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Dongiovanni, P.; Meroni, M.; Longo, M.; Fargion, S.; Fracanzani, A.L. Genetics, Immunity and Nutrition Boost the Switching from NASH to HCC. Biomedicines 2021, 9, 1524. https://doi.org/10.3390/biomedicines9111524
Dongiovanni P, Meroni M, Longo M, Fargion S, Fracanzani AL. Genetics, Immunity and Nutrition Boost the Switching from NASH to HCC. Biomedicines. 2021; 9(11):1524. https://doi.org/10.3390/biomedicines9111524
Chicago/Turabian StyleDongiovanni, Paola, Marica Meroni, Miriam Longo, Silvia Fargion, and Anna Ludovica Fracanzani. 2021. "Genetics, Immunity and Nutrition Boost the Switching from NASH to HCC" Biomedicines 9, no. 11: 1524. https://doi.org/10.3390/biomedicines9111524
APA StyleDongiovanni, P., Meroni, M., Longo, M., Fargion, S., & Fracanzani, A. L. (2021). Genetics, Immunity and Nutrition Boost the Switching from NASH to HCC. Biomedicines, 9(11), 1524. https://doi.org/10.3390/biomedicines9111524