Skip to main content

REVIEW article

Front. Oncol., 15 March 2021
Sec. Molecular and Cellular Oncology
This article is part of the Research Topic Epigenetic Regulation in Microenvironment and Metabolism in Human Cancers View all 9 articles

The Epigenetic Regulation of Microenvironment in Hepatocellular Carcinoma

  • 1Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
  • 2Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States

Hepatocellular carcinoma (HCC) is a highly lethal and complex malignancy strongly influenced by the surrounding tumor microenvironment. The HCC microenvironment comprises hepatic stellate cells (HSCs), tumor-associated macrophages (TAMs), stromal and endothelial cells, and the underlying extracellular matrix (ECM). Emerging evidence demonstrates that epigenetic regulation plays a crucial role in altering numerous components of the HCC tumor microenvironment. In this review, we summarize the current understanding of the mechanisms of epigenetic regulation of the microenvironment in HCC. We review recent studies demonstrating how specific epigenetic mechanisms (DNA methylation, histone regulation, and non-coding RNAs mediated regulation) in HSCs, TAMs, and ECM, and how they contribute to HCC development, so as to gain new insights into the treatment of HCC via regulating epigenetic regulation in the tumor microenvironment.

Introduction

Hepatocellular carcinoma (HCC) is the fourth most common cause of cancer death worldwide (1, 2). HCC patients' prognosis is low, with a 5-year survival of just 18% (2). For many years, the only FDA-approved systemic treatment option for advanced HCC was the multi-kinase inhibitor sorafenib, which itself provided only a modest increase in overall survival of 2.8 months compared to placebo (3). More recently, other therapeutics have garnered first-line treatment approval, including lenvatinib and atezolizumab combined with bevacizumab (4, 5). Second-line treatment options for relapsed and refractory HCC have also expanded in recent years to include regorafenib, cabozantinib, and the PD-1 checkpoint inhibitors nivolumab and pembrolizumab (69). While the arsenal of therapies for advanced HCC has expanded, all aforementioned agents are marked by low response rates and limited improvements in overall survival (49). Understanding the molecular mechanisms underlying HCC development are critical to identifying new drug candidates capable of providing enhanced clinical benefit.

In recent years, researchers have started to decipher the complicated crosstalk between the tumor and the surrounding tumor microenvironment (TME). The TME is composed of numerous different elements depending on the tumor type but generally includes surrounding vasculature, immune cells, fibroblasts, and the underlying extracellular matrix (ECM). TME has been repeatedly shown to significantly contribute to tumor initiation, progression, invasiveness, metastases formation, and angiogenesis (1012). In the context of HCC, the TME primarily consists of hepatic stellate cells (HSCs), tumor-associated macrophages (TAMs), the ECM, mesenchymal stem/stromal cells (MSCs), myeloid-derived suppressor cells (MDSCs), and endothelial cells (ECs) with each element performing its unique role in HCC pathogenesis (1321).

Epigenetics is the heritable modification of gene function without changes in the DNA sequence, which is mediated by a number of different factors, including but not limited to, DNA methylation, histone modifications, and non-coding RNAs (22, 23). Epigenetic regulation in tumor cells is extensively studied and plays critical roles in HCC development (2426). Emerging evidence demonstrates that epigenetic alterations in TME also contribute to the initiation and progression of HCC (2733). As the epigenetic regulation of MSCs, MDSCs, and ECs in HCC remain poorly defined, this review will focus on the epigenetics of HSCs, TAMs, and the ECM (Table 1).

TABLE 1
www.frontiersin.org

Table 1. The epigenetic regulation of tumor microenvironment in hepatocellular carcinoma.

Hepatic Stellate Cells

HSCs are resident perisinusoidal cells that contribute to diverse aspects of liver physiology, including hepatic organogenesis, regeneration, vitamin A storage, and wound healing (34). Under non-pathologic conditions, HSCs remain quiescent in the liver and are only activated in response to liver injury (35). In addition to serving as the primary source of ECM proteins, activated HSCs secrete a multitude of cytokines and growth factors that are required for fibrogenesis and promote HCC tumorigenesis (3439). Namely, HSCs can modulate the ECM through secretion and upregulation of matrix metalloproteinases (MMPs), such as MMP2 and MMP9, both of which promote HCC tumor migration (4043). Moreover, activated HSCs promote FAK-MMP9 signaling and invasiveness in HCC, highlighting the crosstalk between tumor cells and act ivated HSCs in the hepatic TME (4446). The activation of HSCs is the result of extensive, but reversible, alterations in gene expression: activated HSCs can reclaim their quiescent state upon regression/resolution of liver damage in a dynamic process that is modulated extensively by epigenetic reprogramming (38, 4749).

DNA Methylation in HSC Activation

DNA methylation influences HSC activation and activity. Activation of HSCs in vitro results in significant changes in DNA methylation and treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine (5-azadC) block can block HSC transdifferentiation (50, 51). DNA methyltransferase 1 (DNMT1)-mediated hypermethylation of the Phosphatase and tensin homolog (PTEN) promoter leads to a loss of PTEN expression, subsequent activation of the PI3K/AKT and ERK pathways, and HSC activation (52). DNMT1 expression is increased in activated HSCs and rat liver fibrosis tissue, which leads to enhanced methylation of the lncRNA H19 promoter and elevates H19 expression and ERK activation. Treatment with 5'-aza-2'-deoxycytidine in activated HSCs model reduced fibrosis-associated gene expression as well as DNMT1 expression while simultaneously enhancing H19 expression and attenuated HSCs activation. Consistently, sennoside A can prevent liver fibrosis by binding DNMT1 and suppress DNMT1-mediated PTEN hypermethylation in HSC activation and proliferation (53).

Inhibition of DNA methyltransferase 3a (DNMT3a) causes activated HSCs to lose the fibrogenic phenotype in a carbon tetrachloride (CCl4)-induced liver fibrosis model (54). In addition to PTEN, DNA methylation of prostacyclin synthase (PTGIS) enhances HSC activation and liver fibrogenesis, with its methylation being induced by DNMT1 as well as DNMT3b, as determined by chromatin immunoprecipitation (ChIP) (55). DNMT3a has also been reported to regulate the methylation of Septin9 to promote hepatic stellate cell activation and liver fibrogenesis (56).

SAD1/UNC84 domain protein-2 (SUN2) gene hypermethylation at CpG sites has also been identified during liver fibrogenesis in mice with CCl4-induced hepatic fibrosis, accompanied by low expression of SUN2 (57). In vivo overexpression of SUN2 following adeno-associated virus-9 (AAV9) administration inhibited CCl4-induced liver injury and reduced fibrogenesis marker expression. Mechanistically, DNMT3b is the principal regulator of SUN2 expression, and inhibition of AKT phosphorylation may be a crucial pathway for SUN2-mediated HSC activation. S-adenosylmethionine suppresses the expression of Smad3/4 in activated human hepatic stellate cells via Rac1 promoter methylation (58). Methylation of RCAN1.4 is mediated by DNMT1 and DNMT3b and enhances HSC activation and liver fibrogenesis through Calcineurin/NFAT3 signaling (59).

Methionine adenosyltransferases (MATs) catalyze the biosynthesis of S-adenosylmethionine (SAMe), the principal methyl donor in DNA methylation. Methionine adenosyltransferase 2A and 2B (MAT2A and MAT2B), the sole regulators of the SAMe homeostasis in HSCs, are induced during in vitro and in vivo HSC activation. MATII enzyme activity and intracellular SAMe levels decline during HSC activation, accompanied by a global decrease in DNA methylation. MAT2A and MAT2B are induced during HSC activation and are essential for this process. The SAMe level falls, resulting in global DNA hypomethylation (60). Elevated nicotinamide N-methyltransferase (NNMT) expression induced by hepatic stellate cells promotes HCC metastasis by altering the histone H3 methylation and transcriptionally activating CD44 (61).

Histone Modification in HSC Activation

Histone modification also plays a critical in HSC activation. Acetylation and methylation are the two most extensively characterized histone modifications. Histone acetylation and methylation are carried out by two families of enzymes: histone acetyltransferases (HATs) and histone methyltransferases (HMTs). In opposition to these enzymes, histones are deacetylated and demethylated by histone deacetylases (HDACs) and histone demethylases (HDMTs), respectively. Both epigenetic marks can change quickly in response to intracellular and environmental cues and confer immense control of cellular responses (62). The p300 HAT is involved in stiffness-mediated HSC activation to promote liver tumor metastasis (63). Mechanistically, stiffness induces p300 nuclear accumulation, which, in turn, promotes the p300-dependent transcription of critical fibrogenic genes and several secreted profibrogenic and tumor-promoting factors. Furthermore, Sirtuin 6 (SIRT6), a NAD-dependent histone deacetylase which is a critical epigenetic regulator in alcoholic liver disease (64), deacetylates Smad family member 3 (Smad3) and attenuates its expression induced by transforming growth factor β (TGF-β) in the activated HSCs (65).

HSC activation and transdifferentiation are accompanied by gene expression changes in numerous HDACs. Notably, HDAC1, HDAC2, HDAC9, and HDAC10 were reported to be downregulated during HSC activation, while the expression of HDAC4, HDAC5, HDAC6, and HDAC8 is increased (66). MC1568, a class II selective HDAC4 and HDAC5 inhibitor, abrogates HSC activation and proliferation in vitro and in vivo (67). Inhibition of HDAC1, HDAC2, and HDAC4 by nilotinib is also known to induce HSC cell death (68). The mechanisms by which several of these HDACs regulate HSC activation and activity have been characterized in detail. For example, HDAC4 accumulates during HSC activation, and forced overexpression of HDAC4 in quiescent HSCs suppresses expression of the endopeptidases MMP9 and MMP13 resulting in ECM accumulation (66, 67, 69). Conversely, genetic knockdown of HDAC4 in activated HSCs promotes the expression of MMP9 and MMP13, thereby promoting ECM degradation. HDAC7 is also known to contribute to HSC activation by regulating the expression of hepatocyte growth factor (HGF), inhibiting HSC activation and liver fibrosis (70). The expression of HGF is reduced during the activation of HSCs, and knockdown of HDAC7 restores HGF levels to quiescent HSC levels (70). HDAC3 is required to activate HSCs by regulating TGF-β, which plays a crucial role in ECM formation and remodeling (71). Consistently, HDAC inhibitors such as SAHA suppress HSC activation and liver fibrosis by attenuating the TGF-β signaling pathway (72).

Besides HDACs, HMTs regulate gene expression during HSC activation. Ethanol exposure promotes HSC activation by inducing the expression of mixed lineage leukemia protein-1 (MLL1), a histone 3 lysine 4 (H3K4) methyltransferase. Following increased expression, MLL1 is recruited to the elastin gene promoter, where it is associated with increased H3K4me3 levels, increased ECM deposition, and HSC transdifferentiation (73). During HSC activation, several histone methyltransferases, such as ASH2, WDR5, and SET1, are recruited to the promoters of pro-fibrogenic genes in response to TGF-β treatment by myocardin-related transcription factor A (MRTF-A) (74). Enhancer of zeste homolog 2 (EZH2), the catalytic methyltransferase subunit of the polycomb repressive complex 2 (PCR2), is another HMT that regulates HSC activation and hepatic fibrosis. EZH2 expression is induced in HSCs upon TGF-β treatment in both in vitro and CCl4-treated mouse livers (75). Upregulation of EZH2 downregulates expression of peroxisome proliferator-activated receptor-gamma (PPARγ), a nuclear receptor essential for HSC activation (76, 77).

Histone demethylation by HDMTs is also a process critical to HSC activation. Jumonji domain-containing protein 1A (JMJD1A) is one of these HDMTs. JMJD1A knockdown in HSCs increases H3K9me2 levels on the PPARγ gene promoter and represses the expression of PPARγ (78). JMJD2D, a histone H3 demethylase, also contributes to HSC activation by demethylating H3K9 residues. The result of this demethylation is increased TLR4 transcription and activation of the TLR4/MyD88/NF-kB signaling pathway, which has a well-established role in liver fibrosis (79). JMJD2D expression is markedly increased in activated HSCs, and AAV9 shRNA-mediated knockdown of JMJD2D suppresses hepatic fibrosis in the CCl4 model of liver fibrosis (80).

Micro-RNA Involvement in HSC Activation

miRNAs are small, non-coding RNAs (20–25 nucleotides in length) that regulate gene expression by binding to target mRNA transcripts and subsequently facilitating the transcript's cleavage, destabilization, or inhibiting its translation (81). MiRNAs can also control the activation of HSCs by several different mechanisms (38). Namely, HCC cells secrete extracellular vesicles (EV) with oncogenic miRNAs (oncomiRs), specifically miR-21, miR-221, and miR-151, which can activate HSCs. This activation promotes HCC cell invasion, epithelial to mesenchymal transition (EMT), and activation of the AKT/ERK signaling pathway (82). miR-214 promotes HSC activation by inhibiting the expression of suppressor-of-fused homolog (Sufu), a negative regulator of Hedgehog signaling pathway (83). miR-542-3p is also capable of stimulating HSC activation and fibrosis by targeting bone morphogenetic protein 7 (BMP7), a potent surpressor of TGF-β signaling (84). miR-942, which is induced by both TGF-β and LPS, assists HSC activation through downregulation of BMP and activin membrane-bound inhibitor homolog (BAMBI) (85). miR-146b boosts HSC activation by targeting of Kruppel-like factor 4 (KLF4) (86). miR-17-5p targets Smad7 thus stimulating the activation of HSCs (87). Likewise, miR-34a facilitates HSC activation through decreasing the expression of acyl-CoA synthetase-1 (ACSL1) (88). miR-125a-5p advances activation of HSCs by targeting a negative regulator of hypoxia-inducible factor 1 (HIF1), the aptly named factor inhibiting HIF-1 or FIH1 (89).

Conversely, several miRNAs have been shown to suppress HSC activation. miR-96-5p suppresses the activation of HSCs by inhibiting ATG7 and autophagy (90). The miR-23b/27b cluster bind to 3′-UTR of gremlin 1, contributing to the reduction of TGF-β expression and HSC inactivation (91). Additionally, miR-338-3p inhibits HSC activation and proliferation through targeting cyclin-dependent kinase 4 (CDK4) (92). mi-378 represses HSC activation by targeting Gli3 and reducing the expression of Gli3 (93). miR-155 inhibits STAT5 and FOXO3a expression, thus silencing HSC activation (94). miR-146a-5p abrogates HSC activation by inhibiting TGF-β1/Smad and LPS/NF-κB signaling pathways (95, 96). Similarly, both miR-9-5p and miR-130-3p attenuate the activation of HSCs by targeting TGFβR1 and TGFβR2 (97). miR-30a impedes HSC activation through the inhibition of EMT via directly targeting SNAI1 (98).

Tumor-Associated Macrophages

Macrophages are essential innate immune cells in the tumor microenvironment and are also closely associated with cancer occurrence, metastases, and disease progression (99). Tumor-associated macrophages (TAMs) that reside in the surrounding TME are crucial components in the HCC immune microenvironment for the formation and development of HCC (100, 101). TAMs promote HCC growth, angiogenesis, invasion, and migration while simultaneously suppressing the antitumor immune response (20). The epigenetic regulation of TAMs has been shown to be critical to their differentiation and functional programming (102105).

DNA Methylation and Histone Modification in TAMs

DNA methylation and histone modification are critical regulatory mechanisms in TAMs for promoting tumor growth and progression (106109). However, the regulation of DNA methylation and histone modification of TAMs in HCC remains poorly understood. A recent study showed that colony-stimulating factor 1 receptor (CSF1R), which plays an essential role in forming TAMs (108, 110), is regulated by DNA methylation in HCC (111). DNA methylation leads to decreased expression of CSF1R in HCC tissues and associates with poor clinicopathological characteristics of HCC (111). SIRT1, a NAD-dependent histone deacetylase, suppresses HCC metastasis by facilitating M1 macrophage polarization via the stimulation of the NF-κB pathway (112). Downregulation of SIRT4, another sirtuin protein, in TAMs promotes macrophages activation and HCC growth via the FAO-PPARδ-STAT3 axis (113).

miRNAs and Long Non-coding RNAs in TAMs

The epigenetic remodeling by miRNA regulates macrophages' activation and functional programming (114117). For example, the down-regulation of miR-98 was accompanied by up-regulation of IL-10 in TAMs of HCC (118). miR-98 modulates macrophage polarization from M2 to M1 in HCC by targeting IL-10, thus stifling the effects of TAMs on advancing EMT and HCC metastasis (118, 119). miR-101 targets dual-specificity phosphatase 1 (DUSP1) to inhibit TAM-induced HCC growth through TGF-β secretion regulation (120). Decreased expression of miR-28-5p suppresses HCC growth and metastasis by directly targeting interleukin-34 (IL-34) and affecting IL-34-mediated TAM infiltration (121). Reduced expression of miR-125a and miR-125b secreted from TAMs promote HCC proliferation and cancer stem cell properties directly targeting CD90 (122). miR-15b secreted from TAMs promotes the aggressiveness of HCC by impeding the LATS1-mediated hippo pathway (123).

Long non-coding RNAs (lncRNAs) are a class of RNAs >200 nucleotides in length, which lack protein-coding capabilities and have gained increased attention in recent years due to their ability to function guides, signals, and decoys in a highly tissue-specific manner (124). Several lncRNAs have been shown to participate in tumorigenesis and cancer progression, a number of which do so by modulating elements of the TME (125, 126). In the context of HCC, lncRNA cox-2 reduces HCC immune evasion and metastasis formation by suppressing M2 macrophage polarization (127). lncRNA H19, induced by TAMs, promotes the progression of HCC via regulating the miR-193b/MAPK1 axis (128).

Epigenetics in the Extracellular Matrix

The ECM plays a critical role in physiological and pathological processes in HCC (129). MMPs, which digest the ECM, are key players in ECM remodeling and are associated with tumor growth and invasion through collagen and matrix degradation (130, 131). Metallopeptidase inhibitor proteins (TIMPs) are natural inhibitors of MMPs (132). Epigenetic modification is a critical mechanism for regulating MMP and TIMP expression, thus playing a pivotal role in HCC development (133).

DNA Methylation and Histone Modification in MMPs and TIMPs

Overexpression of MMPs, such as MMP2 and MMP9, is frequently observed in HCC patients and associated with cancer invasive potential (41, 42). While some studies have shown that DNA methylation can regulate the expression of MMPs in other cancers, it remains unknown if DNA methylation directly regulates the expression of MMPs in HCC (134). Emerging evidence, however, indicates that DNA methylation promotes the dysregulation of TIMPs in HCC. Specifically, TIMP3 suppresses HCC cell proliferation and survival, and TIMP3 expression is suppressed by promoter methylation in HCC cells (135, 136). Histone deacetylation by HDAC4 represses MMP promoter activities and the expression of MMP9 and MMP13 in HSCs (66, 137). Sodium butyrate, a histone deacetylase inhibitor, decreases MMP1 mRNA expression in human liver cancer cells (138). Further, overexpression of histone methyltransferase EZH2 decreases MMP9 expression in HSCs (139).

miRNAs in MMPs and TIMPs

miRNAs are also involved in the Regulation of MMP and TIMP expression (140). For instance, miR-29b directly targets MMP2 and suppresses HCC growth (141). The blocking of MMP2, either by neutralizing antibody or RNA interference, phenocopies the anti-invasion and antiangiogenesis effects of miR-29b, whereas, the introduction of MMP2 antagonizes the function of miR-29b in HCC (141). miR-107 has also been shown to downregulate MMP2 and MMP9 in HCC cells (142). miR-328-3p and miR-133a modulate the expression of MMP9 to inhibit HCC cell proliferation (143, 144). miR-191 promotes EMT and HCC tumor growth by directly targeting TIMP3 and inhibiting TIMP3 expression (145). MiR-181b and miR-221/222 can also target TIMP3 to promote HCC growth (146, 147). Moreover, miR-519d targets TIMP2 and promotes HCC cell proliferation and metastasis (148).

Conclusion and Future Perspectives

The tumor microenvironment plays a critical role in HCC initiation and progression. Epigenetic regulation is a crucial mechanism for the alteration of TMEs in HCC. Overall, epigenetic reprogramming extensively modulates gene expression alterations in HSCs, TAMs, and ECMs, thus promoting HCC development (Figure 1). The effect of metabolic factors (e.g., SAMe) on epigenetic alterations of TMEs in HCC is evident, suggesting dysregulation metabolites establish can affect the epigenome and subsequently impact HCC development. The mechanisms of epigenetic regulation in MSCs, MDSCs, and ECs remain poorly investigated, thus necessitating future study. Understanding the interactions between tumors and TMEs will further enhance our understanding of critical drivers or suppressors for tumor progression. Despite substantial advances in understanding the mechanisms of TMEs, developing therapeutics using this knowledge remains a challenge. Further examination and deciphering of the complex epigenetic landscape of the TME in HCC will help identify new targets and strategies for the treatment of HCC.

FIGURE 1
www.frontiersin.org

Figure 1. HCC microenvironment components and their epigenetic regulation.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Funding

This project was supported by RSG-18-107 (WQ) from the American Cancer Society and R01CA197128 (WQ) from the National Cancer Institute. The content is solely the authors' responsibility and does not necessarily represent the official views of the American Cancer Society or the National Institutes of Health.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Jacques F, Isabelle S, Rajesh D, Sultan E, Colin M, Marise R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. (2015) 136:E359–86. doi: 10.1002/ijc.29210

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Villanueva. Hepatocellular Carcinoma. N Engl J Med. (2019) 380:1450–62. doi: 10.1056/NEJMra1713263

CrossRef Full Text

3. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc F-J, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. (2008) 359:378–90. doi: 10.1056/NEJMoa0708857

CrossRef Full Text | Google Scholar

4. Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. The Lancet. (2018) 391:1163–73. doi: 10.1016/S0140-6736(18)30207-1

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med. (2020) 382:1894–905. doi: 10.1056/NEJMoa1915745

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Bruix J, Qin S, Merle P, Granito A, Huang Y-H, Bodoky G, et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet. (2017) 389:56–66. doi: 10.1016/S0140-6736(16)32453-9

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Abou-Alfa GK, Meyer T, Cheng A-L, El-Khoueiry AB, Rimassa L, Ryoo B-Y, et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N Engl J Med. (2018) 379:54–63. doi: 10.1056/NEJMoa1717002

PubMed Abstract | CrossRef Full Text | Google Scholar

8. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. The Lancet. (2017) 389:2492–502. doi: 10.1016/S0140-6736(17)31046-2

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Finn RS, Ryoo B-Y, Merle P, Kudo M, Bouattour M, Lim H-Y, et al. Investigators, Results of KEYNOTE-240: phase 3 study of pembrolizumab (Pembro) vs best supportive care (BSC) for second line therapy in advanced hepatocellular carcinoma (HCC). J Clin Oncol. (2019) 37:4004. doi: 10.1200/JCO.2019.37.15_suppl.4004

CrossRef Full Text | Google Scholar

10. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. (2011) 144:646–74. doi: 10.1016/j.cell.2011.02.013

CrossRef Full Text | Google Scholar

11. Hernandez-Gea V, Toffanin S, Friedman SL, Llovet JM. Role of the microenvironment in the pathogenesis and treatment of hepatocellular carcinoma. Gastroenterology. (2013) 144:512–27. doi: 10.1053/j.gastro.2013.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Zhou BY, Gong JH, Cai XY, Wang JX, Luo F, Jiang N, et al. An imbalance between stellate cells and gamma delta T cells contributes to hepatocellular carcinoma aggressiveness and recurrence. Hepatol Int. (2019) 13:631–40. doi: 10.1007/s12072-019-09969-w

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Fu JL, Xu DP, Liu ZW, Shi M, Zhao P, Fu BY, et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology. (2007) 132:2328–39. doi: 10.1053/j.gastro.2007.03.102

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Kruger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology. (2008) 135:234–43. doi: 10.1053/j.gastro.2008.03.020

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Budhu A, Wang XW. The role of cytokines in hepatocellular carcinoma. J Leukocyte Biol. (2006) 80:1197–213. doi: 10.1189/jlb.0506297

CrossRef Full Text | Google Scholar

16. Budhu A, Forgues M, Ye QH, Jia HL, He P, Zanetti KA, et al. Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell. (2006) 10:99–111. doi: 10.1016/j.ccr.2006.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Jeong WI, Park O, Suh YG, Byun JS, Park SY, Choi E, et al. Suppression of innate immunity (natural killer cell/interferon-gamma) in the advanced stages of liver fibrosis in mice. Hepatology. (2011) 53:1342–51. doi: 10.1002/hep.24190

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Seo HR. Roles of tumor microenvironment in hepatocelluar carcinoma. Curr Cancer Ther Rev. (2015) 11:82–93. doi: 10.2174/1573394711666151022203313

CrossRef Full Text | Google Scholar

19. Sevic I, Spinelli FM, Cantero MJ, Reszegi A, Kovalszky I, Garcia MG, et al. Chapter 2: The role of the tumor microenvironment in the development and progression of hepatocellular carcinoma. In: Tirnitz-Parker JEE, editor. Hepatocellular Carcinoma. Brisbane, QLD: Codon Publications (2019). doi: 10.15586/hepatocellularcarcinoma.2019.ch2

CrossRef Full Text | Google Scholar

20. Capece D, Fischietti M, Verzella D, Gaggiano A, Cicciarelli G, Tessitore A, et al. The inflammatory microenvironment in hepatocellular carcinoma: a pivotal role for tumor-associated macrophages. Biomed Res Int. (2013) 2013:187204. doi: 10.1155/2013/187204

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Bresnahan E, Lindblad KE, Ruiz de Galarreta M, Lujambio A. Mouse models of oncoimmunology in hepatocellular carcinoma. Clin Cancer Res. (2020) 26:5276–86. doi: 10.1158/1078-0432.CCR-19-2923

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Hardy T, Mann DA. Epigenetics in liver disease: from biology to therapeutics. Gut. (2016) 65:1895–905. doi: 10.1136/gutjnl-2015-311292

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Hanover JA, Krause MW, Love DC. Bittersweet memories: linking metabolism to epigenetics through O-GlcNAcylation. Nat Rev Mol Cell Biol. (2012) 13:312–21. doi: 10.1038/nrm3334

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Santhakumar C, Gane EJ, Liu K, McCaughan GW. Current perspectives on the tumor microenvironment in hepatocellular carcinoma. Hepatol Int. (2020) 14:947–57. doi: 10.1007/s12072-020-10104-3

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Wang G, Wang Q, Liang N, Xue H, Yang T, Chen X, et al. Oncogenic driver genes and tumor microenvironment determine the type of liver cancer. Cell Death Dis. (2020) 11:313. doi: 10.1038/s41419-020-2509-x

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Thomson JP, Ottaviano R, Unterberger EB, Lempiainen H, Muller A, Terranova R, et al. Loss of tet1-associated 5-hydroxymethylcytosine is concomitant with aberrant promoter hypermethylation in liver cancer. Cancer Res. (2016) 76:3097–108. doi: 10.1158/0008-5472.CAN-15-1910

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Wachowska M, Gabrysiak M, Golab J. Epigenetic remodeling combined with photodynamic therapy elicits anticancer immune responses. Oncoimmunology. (2014) 3:e28837. doi: 10.4161/onci.28837

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Setiadi AF, David MD, Seipp RP, Hartikainen JA, Gopaul R, Jefferies WA. Epigenetic control of the immune escape mechanisms in malignant carcinomas. Mol Cell Biol. (2007) 27:7886–94. doi: 10.1128/MCB.01547-07

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Liu M, Jiang L, Guan XY. The genetic and epigenetic alterations in human hepatocellular carcinoma: a recent update. Protein Cell. (2014) 5:673–91. doi: 10.1007/s13238-014-0065-9

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Sceusi EL, Loose DS, Wray CJ. Clinical implications of DNA methylation in hepatocellular carcinoma. HPB (Oxford). (2011) 13:369–76. doi: 10.1111/j.1477-2574.2011.00303.x

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Nakamura M, Chiba T, Kanayama K, Kanzaki H, Saito T, Kusakabe Y, et al. Epigenetic dysregulation in hepatocellular carcinoma: an up-to-date review. Hepatol Res. (2019) 49:3–13. doi: 10.1111/hepr.13250

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Toh TB, Lim JJ, Chow EK. Epigenetics of hepatocellular carcinoma. Clin Transl Med. (2019) 8:13. doi: 10.1186/s40169-019-0230-0

CrossRef Full Text | Google Scholar

33. Fernandez-Barrena MG, Arechederra M, Colyn L, Berasain C, Avila MA. Epigenetics in hepatocellular carcinoma development and therapy: the tip of the iceberg. JHEP Rep. (2020) 2:100167. doi: 10.1016/j.jhepr.2020.100167

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Friedman SL. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev. (2008) 88:125–72. doi: 10.1152/physrev.00013.2007

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Barry AE, Baldeosingh R, Lamm R, Patel K, Zhang K, Dominguez DA, et al. Hepatic stellate cells and hepatocarcinogenesis. Front Cell Dev Biol. (2020) 8:709. doi: 10.3389/fcell.2020.00709

CrossRef Full Text | Google Scholar

36. Amann T, Bataille F, Spruss T, Muhlbauer M, Gabele E, Scholmerich J, et al. Activated hepatic stellate cells promote tumorigenicity of hepatocellular carcinoma. Cancer Sci. (2009) 100:646–53. doi: 10.1111/j.1349-7006.2009.01087.x

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Santamato A, Fransvea E, Dituri F, Caligiuri A, Quaranta M, Niimi T, et al. Hepatic stellate cells stimulate HCC cell migration via laminin-5 production. Clin Sci (Lond). (2011) 121:159–68. doi: 10.1042/CS20110002

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastro Hepat. (2017) 14:397–411. doi: 10.1038/nrgastro.2017.38

CrossRef Full Text | Google Scholar

39. Coulouarn C, Clement B. Stellate cells and the development of liver cancer: therapeutic potential of targeting the stroma. J Hepatol. (2014) 60:1306–9. doi: 10.1016/j.jhep.2014.02.003

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Lachowski D, Cortes E, Rice A, Pinato D, Rombouts K, Hernandez AD. Matrix stiffness modulates the activity of MMP-9 and TIMP-1 in hepatic stellate cells to perpetuate fibrosis. Sci Rep-Uk. (2019) 9:7299: doi: 10.1038/s41598-019-43759-6

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Giannelli G, Bergamini C, Marinosci F, Fransvea E, Quaranta M, Lupo L, et al. Clinical role of MMP-2/TIMP-2 imbalance in hepatocellular carcinoma. Int J Cancer. (2002) 97:425–31. doi: 10.1002/ijc.1635

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Arii S, Mise M, Harada T, Furutani M, Ishigami S, Niwano M, et al. Overexpression of matrix metalloproteinase 9 gene in hepatocellular carcinoma with invasive potential. Hepatology. (1996) 24:316–22. doi: 10.1002/hep.510240206

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Scheau C, Badarau IA, Costache R, Caruntu C, Mihai GL, Didilescu AC, et al. The role of matrix metalloproteinases in the epithelial-mesenchymal transition of hepatocellular carcinoma. Anal Cell Pathol. (2019) 2019:9423907. doi: 10.1155/2019/9423907

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Chen JS, Huang XH, Wang Q, Chen XL, Fu XH, Tan HX, et al. FAK is involved in invasion and metastasis of hepatocellular carcinoma. Clin Exp Metastas. (2010) 27:71–82. doi: 10.1007/s10585-010-9306-3

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Jia YL, Shi L, Zhou JN, Fu CJ, Chen L, Yuan HF, et al. Epimorphin promotes human hepatocellular carcinoma invasion and metastasis through activation of focal adhesion kinase/extracellular signal-regulated kinase/matrix metalloproteinase-9 axis. Hepatology. (2011) 54:1808–18. doi: 10.1002/hep.24562

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Han SS, Han L, Yao YM, Sun H, Zan XF, Liu QG. Activated hepatic stellate cells promote hepatocellular carcinoma cell migration and invasion via the activation of FAK-MMP9 signaling. Oncol Rep. (2014) 31:641–8. doi: 10.3892/or.2013.2872

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Yin C, Evason KJ, Asahina K, Stainier DY. Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Investig. (2013) 123:1902–10. doi: 10.1172/JCI66369

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Kisseleva T, Cong M, Paik Y, Scholten D, Jiang C, Benner C, et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci U S A. (2012) 109:9448–53. doi: 10.1073/pnas.1201840109

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Troeger JS, Mederacke I, Gwak GY, Dapito DH, Mu X, Hsu CC, et al. Deactivation of hepatic stellate cells during liver fibrosis resolution in mice. Gastroenterology. (2012) 143:1073–83 e22. doi: 10.1053/j.gastro.2012.06.036

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Mann J, Oakley F, Akiboye F, Elsharkawy A, Thorne AW, Mann DA. Regulation of myofibroblast transdifferentiation by DNA methylation and MeCP2: implications for wound healing and fibrogenesis. Cell Death Differ. (2007) 14:275–85. doi: 10.1038/sj.cdd.4401979

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Gotze S, Schumacher EC, Kordes C, Haussinger D. epigenetic changes during hepatic stellate cell activation. PLoS ONE. (2015) 10:e0128745. doi: 10.1371/journal.pone.0128745

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Bian EB, Huang C, Ma TT, Tao H, Zhang H, Cheng C, et al. DNMT1-mediated PTEN hypermethylation confers hepatic stellate cell activation and liver fibrogenesis in rats. Toxicol Appl Pharm. (2012) 264:13–22. doi: 10.1016/j.taap.2012.06.022

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Zhu H, He C, Zhao H, Jiang W, Xu S, Li J, et al. Sennoside A prevents liver fibrosis by binding DNMT1 and suppressing DNMT1-mediated PTEN hypermethylation in HSC activation and proliferation. FASEB J. (2020) 34:14558–71. doi: 10.1096/fj.202000494RR

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Page A, Paoli P, Salvador EM, White S, French J, Mann J. Hepatic stellate cell transdifferentiation involves genome-wide remodeling of the DNA methylation landscape. J Hepatol. (2016) 64:661–73. doi: 10.1016/j.jhep.2015.11.024

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Pan XY, Yang Y, Meng HW, Li HD, Chen X, Huang HM, et al. DNA methylation of PTGIS enhances hepatic stellate cells activation and liver fibrogenesis. Fron Pharmacol. (2018) 9:553. doi: 10.3389/fphar.2018.00553

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Wu Y, Bu F, Yu H, Li W, Huang C, Meng X, et al. Methylation of Septin9 mediated by DNMT3a enhances hepatic stellate cells activation and liver fibrogenesis. Toxicol Appl Pharmacol. (2017) 315:35–49. doi: 10.1016/j.taap.2016.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Chen X, Li WX, Chen Y, Li XF, Li HD, Huang HM, et al. Suppression of SUN2 by DNA methylation is associated with HSCs activation and hepatic fibrosis. Cell Death Dis. (2018) 9:1021. doi: 10.1038/s41419-018-1032-9

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Bian K, Zhang F, Wang T, Zou X, Duan X, Chen G, et al. S-Adenosylmethionine suppresses the expression of Smad3/4 in activated human hepatic stellate cells via Rac1 promoter methylation. Mol Med Rep. (2016) 13:3867–73. doi: 10.3892/mmr.2016.4997

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Pan X-Y, You HM, Wang L, Bi YH, Yang Y, Meng HW, et al. Methylation of RCAN1.4 mediated by DNMT1 and DNMT3b enhances hepatic stellate cell activation and liver fibrogenesis through Calcineurin/NFAT3 signaling. Theranostics. (2019) 9:4308–23. doi: 10.7150/thno.32710

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Ramani K, Yang H, Kuhlenkamp J, Tomasi L, Tsukamoto H, Mato JM, et al. Changes in the expression of methionine adenosyltransferase genes and S-adenosylmethionine homeostasis during hepatic stellate cell activation. Hepatology. (2010) 51:986–95. doi: 10.1002/hep.23411

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Li J, You S, Zhang S, Hu Q, Wang F, Chi X, et al. Elevated N-methyltransferase expression induced by hepatic stellate cells contributes to the metastasis of hepatocellular carcinoma via regulation of the CD44v3 isoform. Mol Oncol. (2019) 13:1993–2009. doi: 10.1002/1878-0261.12544

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Zhang T, Cooper S, Brockdorff N. The interplay of histone modifications - writers that read. EMBO Rep. (2015) 16:1467–81. doi: 10.15252/embr.201540945

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Dou C, Liu Z, Tu K, Zhang H, Chen C, Yaqoob U, et al. P300 acetyltransferase mediates stiffness-induced activation of hepatic stellate cells into tumor-promoting myofibroblasts. Gastroenterology. (2018) 154:2209–21 e14. doi: 10.1053/j.gastro.2018.02.015

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Kim HG, Huang M, Xin Y, Zhang Y, Zhang X, Wang G, et al. The epigenetic regulator SIRT6 protects the liver from alcohol-induced tissue injury by reducing oxidative stress in mice. J Hepatol. (2019) 71:960–9. doi: 10.1016/j.jhep.2019.06.019

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Zhong X, Huang M, Kim HG, Zhang Y, Chowdhury K, Cai W, et al. SIRT6 protects against liver fibrosis by deacetylation and suppression of SMAD3 in hepatic stellate cells. Cell Mol Gastroenterol Hepatol. (2020) 10:341–64. doi: 10.1016/j.jcmgh.2020.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Qin L, Han YP. Epigenetic repression of matrix metalloproteinases in myofibroblastic hepatic stellate cells through histone deacetylases 4: implication in tissue fibrosis. Am J Pathol. (2010) 177:1915–28. doi: 10.2353/ajpath.2010.100011

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Mannaerts I, Eysackers N, Onyema OO, Van Beneden K, Valente S, Mai A, et al. Class II HDAC inhibition hampers hepatic stellate cell activation by induction of microRNA-29. PloS ONE. (2013) 8:e55786. doi: 10.1371/journal.pone.0055786

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Shaker ME, Ghani A, Shiha GE, Ibrahim TM, Mehal WZ. Nilotinib induces apoptosis and autophagic cell death of activated hepatic stellate cells via inhibition of histone deacetylases. Biochim Biophysica Acta. (2013) 1833:1992–2003. doi: 10.1016/j.bbamcr.2013.02.033

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Duarte S, Baber J, Fujii T, Coito AJ. Matrix metalloproteinases in liver injury, repair and fibrosis. Matrix Biol. (2015) 44–46:147–56. doi: 10.1016/j.matbio.2015.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Pannem RR, Dorn C, Hellerbrand C, Massoumi R. Cylindromatosis gene CYLD regulates hepatocyte growth factor expression in hepatic stellate cells through interaction with histone deacetylase 7. Hepatology. (2014) 60:1066–81. doi: 10.1002/hep.27209

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Barter MJ, Pybus L, Litherland GJ, Rowan AD, Clark IM, Edwards DR, et al. HDAC-mediated control of ERK- and PI3K-dependent TGF-beta-induced extracellular matrix-regulating genes. Matrix Biol. (2010) 29:602–12. doi: 10.1016/j.matbio.2010.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Wang Y, Zhao L, Jiao FZ, Zhang WB, Chen Q, Gong ZJ. Histone deacetylase inhibitor suberoylanilide hydroxamic acid alleviates liver fibrosis by suppressing the transforming growth factor-beta1 signal pathway. Hepatobiliary Pancreat Dis Int. (2018) 17:423–9. doi: 10.1016/j.hbpd.2018.09.013

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Page A, Paoli PP, Hill SJ, Howarth R, Wu R, Kweon SM, et al. Alcohol directly stimulates epigenetic modifications in hepatic stellate cells. J Hepatol. (2015) 62:388–97. doi: 10.1016/j.jhep.2014.09.033

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Tian W, Fan Z, Li J, Hao C, Li M, Xu H, et al. Myocardin-related transcription factor A (MRTF-A) plays an essential role in hepatic stellate cell activation by epigenetically modulating TGF-beta signaling. Int J Biochem Cell Biol. (2016) 71:35–43. doi: 10.1016/j.biocel.2015.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Martin-Mateos R, De Assuncao TM, Arab JP, Jalan-Sakrikar N, Yaqoob U, Greuter T, et al. Enhancer of zeste homologue 2 inhibition attenuates TGF-beta dependent hepatic stellate cell activation and liver fibrosis. Cell Mol Gastroenterol Hepatol. (2019) 7:197–209. doi: 10.1016/j.jcmgh.2018.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Mann J, Chu DC, Maxwell A, Oakley F, Zhu NL, Tsukamoto H, et al. MeCP2 controls an epigenetic pathway that promotes myofibroblast transdifferentiation and fibrosis. Gastroenterology. (2010) 138:705–14. doi: 10.1053/j.gastro.2009.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Panebianco C, Oben JA, Vinciguerra M, Pazienza V. Senescence in hepatic stellate cells as a mechanism of liver fibrosis reversal: a putative synergy between retinoic acid and PPAR-gamma signalings. Clin Exp Med. (2017) 17:269–80. doi: 10.1007/s10238-016-0438-x

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Jiang Y, Wang S, Zhao Y, Lin C, Zhong F, Jin L, et al. Histone H3K9 demethylase JMJD1A modulates hepatic stellate cells activation and liver fibrosis by epigenetically regulating peroxisome proliferator-activated receptor gamma. FASEB J. (2015) 29:1830–41. doi: 10.1096/fj.14-251751

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA, et al. TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med. (2007) 13:1324–32. doi: 10.1038/nm1663

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Dong F, Jiang S, Li J, Wang Y, Zhu L, Huang Y, et al. The histone demethylase KDM4D promotes hepatic fibrogenesis by modulating Toll-like receptor 4 signaling pathway. EBioMedicine. (2019) 39:472–83. doi: 10.1016/j.ebiom.2018.11.055

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. (2004) 116:281–97. doi: 10.1016/S0092-8674(04)00045-5

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Li J, Yan Y, Ang L, Li X, Liu C, Sun B, et al. Extracellular vesicles-derived OncomiRs mediate communication between cancer cells and cancer-associated hepatic stellate cells in hepatocellular carcinoma microenvironment. Carcinogenesis. (2020) 41:223–34. doi: 10.1093/carcin/bgz096

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Ma L, Yang X, Wei R, Ye T, Zhou JK, Wen M, et al. MicroRNA-214 promotes hepatic stellate cell activation and liver fibrosis by suppressing Sufu expression. Cell Death Dis. (2018) 9:718. doi: 10.1038/s41419-018-0752-1

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Ji F, Wang K, Zhang Y, Mao XL, Huang Q, Wang J, et al. MiR-542-3p controls hepatic stellate cell activation and fibrosis via targeting BMP-7. J Cell Biochem. (2019) 120:4573–81. doi: 10.1002/jcb.27746

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Tao L, Xue D, Shen D, Ma W, Zhang J, Wang X, et al. MicroRNA-942 mediates hepatic stellate cell activation by regulating BAMBI expression in human liver fibrosis. Arch Toxicol. (2018) 92:2935–46 doi: 10.1007/s00204-018-2278-9

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Ge S, Zhang L, Xie J, Liu F, He J, He J, et al. MicroRNA-146b regulates hepatic stellate cell activation via targeting of KLF4. Ann Hepatol. (2016) 15:918–28. doi: 10.5604/16652681.1222111

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Yu F, Guo Y, Chen B, Dong P, Zheng J. MicroRNA-17-5p activates hepatic stellate cells through targeting of Smad7. Lab Invest. (2015) 95:781–9. doi: 10.1038/labinvest.2015.58

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Yan G, Li B, Xin X, Xu M, Ji G, Yu H. MicroRNA-34a promotes hepatic stellate cell activation via targeting ACSL1. Med Sci Monit. (2015) 21:3008–15. doi: 10.12659/MSM.894000

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Li G, Li J, Li C, Qi H, Dong P, Zheng J, et al. MicroRNA-125a-5p contributes to hepatic stellate cell activation through targeting FIH1. Cell Physiol Biochem. (2016) 38:1544–52. doi: 10.1159/000443095

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Yu K, Li N, Cheng Q, Zheng J, Zhu M, Bao S, et al. miR-96-5p prevents hepatic stellate cell activation by inhibiting autophagy via ATG7. J Mol Med (Berl). (2018) 96:65–74. doi: 10.1007/s00109-017-1593-6

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Zeng XY, Zhang YQ, He XM, Wan LY, Wang H, Ni YR, et al. Suppression of hepatic stellate cell activation through downregulation of gremlin1 expression by the miR-23b/27b cluster. Oncotarget. (2016) 7:86198–210. doi: 10.18632/oncotarget.13365

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Duan B, Hu J, Zhang T, Luo X, Zhou Y, Liu S, et al. miRNA-338-3p/CDK4 signaling pathway suppressed hepatic stellate cell activation and proliferation. BMC Gastroenterol. (2017) 17:12. doi: 10.1186/s12876-017-0571-3

CrossRef Full Text | Google Scholar

93. Hyun J, Wang S, Kim J, Rao KM, Park SY, Chung I, et al. MicroRNA-378 limits activation of hepatic stellate cells and liver fibrosis by suppressing Gli3 expression. Nat Commun. (2016) 7:10993. doi: 10.1038/ncomms10993

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Zhu D, Yang C, Shen P, Chen L, Chen J, Sun X, et al. rSjP40 suppresses hepatic stellate cell activation by promoting microRNA-155 expression and inhibiting STAT5 and FOXO3a expression. J Cell Mol Med. (2018) 22:5486–93. doi: 10.1111/jcmm.13819

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Zou Y, Cai Y, Lu D, Zhou Y, Yao Q, Zhang S. MicroRNA-146a-5p attenuates liver fibrosis by suppressing profibrogenic effects of TGFbeta1 and lipopolysaccharide. Cell Signal. (2017) 39:1–8. doi: 10.1016/j.cellsig.2017.07.016

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Chen Y, Wu Z, Yuan B, Dong Y, Zhang L, Zeng Z. MicroRNA-146a-5p attenuates irradiation-induced and LPS-induced hepatic stellate cell activation and hepatocyte apoptosis through inhibition of TLR4 pathway. Cell Death Dis. (2018) 9:22. doi: 10.1038/s41419-017-0038-z

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Yu F, Chen B, Fan X, Li G, Dong P, Zheng J. Epigenetically-regulated MicroRNA-9-5p suppresses the activation of hepatic stellate cells via TGFBR1 and TGFBR2. Cell Physiol Biochem. (2017) 43:2242–52. doi: 10.1159/000484303

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Zheng J, Wang W, Yu F, Dong P, Chen B, Zhou MT. MicroRNA-30a Suppresses the activation of hepatic stellate cells by inhibiting epithelial-to-mesenchymal transition. Cell Physiol Biochem. (2018) 46:82–92. doi: 10.1159/000488411

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Larionova E, Kazakova E, Patysheva M, Kzhyshkowska J. Transcriptional, epigenetic and metabolic programming of tumor-associated macrophages. Cancers (Basel). (2020) 12:1411: doi: 10.3390/cancers12061411

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Singh S, Mehta N, Lilan J, Budhthoki MB, Chao F, Yong L. Initiative action of tumor-associated macrophage during tumor metastasis. Biochim Open. (2017) 4:8–18. doi: 10.1016/j.biopen.2016.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Zhou D, Luan J, Huang C, Li J. Tumor-associated macrophages in hepatocellular carcinoma: friend or foe? Gut Liver. (2020). doi: 10.5009/gnl20223

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Hoeksema MA, de Winther MP. Epigenetic regulation of monocyte and macrophage function. Antioxid Redox Signal. (2016) 25:758–74. doi: 10.1089/ars.2016.6695

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Jain N, Shahal T, Gabrieli T, Gilat N, Torchinsky D, Michaeli Y, et al. Global modulation in DNA epigenetics during pro-inflammatory macrophage activation. Epigenetics. (2019) 14:1183–93. doi: 10.1080/15592294.2019.1638700

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Nakamura K, Smyth MJ. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell Mol Immunol. (2020) 17:1–12. doi: 10.1038/s41423-019-0306-1

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Van den Bossche J, Neele AE, Hoeksema MA, de Winther MP. Macrophage polarization: the epigenetic point of view. Curr Opin Lipidol. (2014) 25:367–73. doi: 10.1097/MOL.0000000000000109

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Dekkers KF, Neele AE, Jukema JW, Heijmans BTM, de Winther PJ. Human monocyte-to-macrophage differentiation involves highly localized gain and loss of DNA methylation at transcription factor binding sites. Epigenetics Chromatin. (2019) 12:34. doi: 10.1186/s13072-019-0279-4

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Wang X, Cao Q, Yu L, Shi H, Xue B, Shi H. Epigenetic regulation of macrophage polarization and inflammation by DNA methylation in obesity. JCI Insight. (2016) 1:e87748. doi: 10.1172/jci.insight.87748

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Ishii M, Wen H, Corsa CA, Liu T, Coelho AL, Allen RM, et al. Epigenetic regulation of the alternatively activated macrophage phenotype. Blood. (2009) 114:3244–54. doi: 10.1182/blood-2009-04-217620

PubMed Abstract | CrossRef Full Text | Google Scholar

109. de Groot AE, Pienta KJ. Epigenetic control of macrophage polarization: implications for targeting tumor-associated macrophages. Oncotarget. (2018) 9:20908–27. doi: 10.18632/oncotarget.24556

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Zhu Y, Yang J, Xu D, Gao XM, Zhang Z, Hsu JL, et al. Disruption of tumour-associated macrophage trafficking by the osteopontin-induced colony-stimulating factor-1 signalling sensitises hepatocellular carcinoma to anti-PD-L1 blockade. Gut. (2019) 68:1653–66. doi: 10.1136/gutjnl-2019-318419

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Cui B, Fan XX, Zhou DZ, He LF, Li YR, Li DD, et al. CSF1R methylation is a key regulatory mechanism of tumor-associated macrophages in hepatocellular carcinoma. Oncol Lett. (2020) 20:1835–45. doi: 10.3892/ol.2020.11726

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Zhou B, Yang Y, Li C. SIRT1 inhibits hepatocellular carcinoma metastasis by promoting M1 macrophage polarization via NF-kappaB pathway. OncoTargets Ther. (2019) 12:2519–29. doi: 10.2147/OTT.S195234

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Li Z, Li H, Zhao ZB, Zhu W, Feng PP, Zhu XW, et al. SIRT4 silencing in tumor-associated macrophages promotes HCC development via PPARdelta signalling-mediated alternative activation of macrophages. J Exp Clin Cancer Res. (2019) 38:469. doi: 10.1186/s13046-019-1456-9

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Zhang Y, Zhang M, Zhong M, Suo Q, Lv K. Expression profiles of miRNAs in polarized macrophages. Int J Mol Med. (2013) 31:797–802. doi: 10.3892/ijmm.2013.1260

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Liu G, Abraham E. MicroRNAs in immune response and macrophage polarization. Arterioscler Thromb Vasc Biol. (2013) 33:170–7. doi: 10.1161/ATVBAHA.112.300068

CrossRef Full Text | Google Scholar

116. Cai X, Yin Y, Li N, Zhu D, Zhang J, Zhang CY, et al. Re-polarization of tumor-associated macrophages to pro-inflammatory M1 macrophages by microRNA-155. J Mol Cell Biol. (2012) 4:341–3. doi: 10.1093/jmcb/mjs044

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Liu M, Zhou J, Chen Z, Cheng AS. Understanding the epigenetic regulation of tumours and their microenvironments: opportunities and problems for epigenetic therapy. J Pathol. (2017) 241:10–24. doi: 10.1002/path.4832

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Li L, Sun P, Zhang C, Li Z, Zhou W. MiR-98 suppresses the effects of tumor-associated macrophages on promoting migration and invasion of hepatocellular carcinoma cells by regulating IL-10. Biochimie. (2018) 150:23–30. doi: 10.1016/j.biochi.2018.04.016

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Li L, Sun P, Zhang C, Li Z, Cui K, Zhou W. MiR-98 modulates macrophage polarization and suppresses the effects of tumor-associated macrophages on promoting invasion and epithelial-mesenchymal transition of hepatocellular carcinoma. Cancer Cell Int. (2018) 18:95. doi: 10.1186/s12935-018-0590-3

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Wei X, Tang C, Lu X, Liu R, Zhou M, He D, et al. MiR-101 targets DUSP1 to regulate the TGF-beta secretion in sorafenib inhibits macrophage-induced growth of hepatocarcinoma. Oncotarget. (2015) 6:18389–405. doi: 10.18632/oncotarget.4089

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Zhou SL, Hu ZQ, Zhou ZJ, Dai Z, Wang Z, Cao Y, et al. miR-28-5p-IL-34-macrophage feedback loop modulates hepatocellular carcinoma metastasis. Hepatology. (2016) 63:1560–75. doi: 10.1002/hep.28445

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Wang Y, Wang B, Xiao S, Li Y, Chen Q. miR-125a/b inhibits tumor-associated macrophages mediated in cancer stem cells of hepatocellular carcinoma by targeting CD90. J Cell Biochem. (2019) 120:3046–55. doi: 10.1002/jcb.27436

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Li J, Xue J, Ling M, Sun J, Xiao T, Dai X, et al. MicroRNA-15b in extracellular vesicles from arsenite-treated macrophages promotes the progression of hepatocellular carcinomas by blocking the LATS1-mediated Hippo pathway. Cancer Lett. (2021) 497:137–53. doi: 10.1016/j.canlet.2020.10.023

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Mol Cell. (2011) 43:904–14. doi: 10.1016/j.molcel.2011.08.018

CrossRef Full Text | Google Scholar

125. Han D, Fang Y, Guo Y, Hong W, Tu J, Wei W. The emerging role of long non-coding RNAs in tumor-associated macrophages. J Cancer. (2019) 10:6738–46. doi: 10.7150/jca.35770

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, et al. The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. (2015) 47:199–208. doi: 10.1038/ng.3192

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Ye Y, Xu Y, Lai Y, He W, Li Y, Wang R, et al. Long non-coding RNA cox-2 prevents immune evasion and metastasis of hepatocellular carcinoma by altering M1/M2 macrophage polarization. J Cell Biochem. (2018) 119:2951–63. doi: 10.1002/jcb.26509

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Ye Y, Guo J, Xiao P, Ning J, Zhang R, Liu P, et al. Macrophages-induced long noncoding RNA H19 up-regulation triggers and activates the miR-193b/MAPK1 axis and promotes cell aggressiveness in hepatocellular carcinoma. Cancer Lett. (2020) 469:310–22. doi: 10.1016/j.canlet.2019.11.001

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Carloni V, Luong TV, Rombouts K. Hepatic stellate cells and extracellular matrix in hepatocellular carcinoma: more complicated than ever. Liver Int. (2014) 34:834–43. doi: 10.1111/liv.12465

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Hadler-Olsen E, Fadnes B, Sylte I, Uhlin-Hansen L, Winberg JO. Regulation of matrix metalloproteinase activity in health and disease. FEBS J. (2011) 278:28–45. doi: 10.1111/j.1742-4658.2010.07920.x

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Stamenkovic I. Matrix metalloproteinases in tumor invasion and metastasis. Semin Cancer Biol. (2000) 10:415–33. doi: 10.1006/scbi.2000.0379

CrossRef Full Text | Google Scholar

132. Jackson HW, Defamie V, Waterhouse P, Khokha R. TIMPs: versatile extracellular regulators in cancer. Nat Rev Cancer. (2017) 17:38–53. doi: 10.1038/nrc.2016.115

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Han TS, Ban HS, Hur K, Cho HS. The epigenetic regulation of HCC metastasis. Int J Mol Sci. (2018) 19:3978: doi: 10.3390/ijms19123978

CrossRef Full Text | Google Scholar

134. Chernov AV, Strongin AY. Epigenetic regulation of matrix metalloproteinases and their collagen substrates in cancer. Biomol Concepts. (2011) 2:135–147. doi: 10.1515/bmc.2011.017

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Shen B, Jiang Y, Chen YR, Zheng HC, Zeng W, Li YY, et al. Expression and inhibitory role of TIMP-3 in hepatocellular carcinoma. Oncol Rep. (2016) 36:494–502. doi: 10.3892/or.2016.4818

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Yuan Y, Wang J, Li J, Wang L, Li M, Yang Z, et al. Frequent epigenetic inactivation of spleen tyrosine kinase gene in human hepatocellular carcinoma. Clin Cancer Res. (2006) 12:6687–95. doi: 10.1158/1078-0432.CCR-06-0921

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Yang Z, Liu Y, Qin L, Wu P, Xia Z, Luo M, et al. Cathepsin H-mediated degradation of HDAC4 for matrix metalloproteinase expression in hepatic stellate cells: implications of epigenetic suppression of matrix metalloproteinases in fibrosis through stabilization of class IIa histone deacetylases. Am J Pathol. (2017) 187:781–97. doi: 10.1016/j.ajpath.2016.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Kaneko F, Saito H, Saito Y, Wakabayashi K, Nakamoto N, Tada S, et al. Down-regulation of matrix-invasive potential of human liver cancer cells by type I interferon and a histone deacetylase inhibitor sodium butyrate. Int J Oncol. (2004) 24:837–45. doi: 10.3892/ijo.24.4.837

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Atta H, El-Rehany M, Hammam O, Abdel-Ghany H, Ramzy M, Roderfeld M, et al. Mutant MMP-9 and HGF gene transfer enhance resolution of CCl4-induced liver fibrosis in rats: role of ASH1 and EZH2 methyltransferases repression. PloS ONE. (2014) 9:e112384. doi: 10.1371/journal.pone.0112384

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Wentz-Hunter KK, Potashkin JA. The role of miRNAs as key regulators in the neoplastic microenvironment. Mol Biol Int. (2011) 2011:839872. doi: 10.4061/2011/839872

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Fang JH, Zhou HC, Zeng C, Yang J, Liu Y, Huang X, et al. MicroRNA-29b suppresses tumor angiogenesis, invasion, and metastasis by regulating matrix metalloproteinase 2 expression. Hepatology. (2011) 54:1729–40. doi: 10.1002/hep.24577

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Xiao D, Gao HX. Mechanism of miR-107-targeting of regulator of G-protein signaling 4 in hepatocellular carcinoma. Oncol Lett. (2019) 18:5145–54. doi: 10.3892/ol.2019.10857

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Li JZ, Li J, Liu BZ. MicroRNA-328-3p inhibits malignant progression of hepatocellular carcinoma by regulating MMP-9 level. Eur Rev Med Pharmacol Sci. (2019) 23:9331–40. doi: 10.26355/eurrev_201911_19426

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Chen X, Bo L, Zhao X, Chen Q. MicroRNA-133a inhibits cell proliferation, colony formation ability, migration and invasion by targeting matrix metallopeptidase 9 in hepatocellular carcinoma. Mol Med Rep. (2015) 11:3900–7. doi: 10.3892/mmr.2015.3232

PubMed Abstract | CrossRef Full Text | Google Scholar

145. He Y, Cui Y, Wang W, Gu J, Guo S, Ma K, et al. Hypomethylation of the hsa-miR-191 locus causes high expression of hsa-mir-191 and promotes the epithelial-to-mesenchymal transition in hepatocellular carcinoma. Neoplasia. (2011) 13:841–53. doi: 10.1593/neo.11698

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Wang B, Hsu SH, Majumder S, Kutay H, Huang W, Jacob ST, et al. TGFbeta-mediated upregulation of hepatic miR-181b promotes hepatocarcinogenesis by targeting TIMP3. Oncogene. (2010) 29:1787–97. doi: 10.1038/onc.2009.468

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Garofalo M, Di Leva G, Romano G, Nuovo G, Suh SS, Ngankeu A, et al. miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell. (2009) 16:498–509. doi: 10.1016/j.ccr.2009.10.014

CrossRef Full Text | Google Scholar

148. Fornari F, Milazzo M, Chieco P, Negrini M, Marasco E, Capranico G, et al. In hepatocellular carcinoma miR-519d is up-regulated by p53 and DNA hypomethylation and targets CDKN1A/p21, PTEN, AKT3 and TIMP2. J Pathol. (2012) 227:275–85. doi: 10.1002/path.3995

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: tumor microenvironment, DNA methylation, histone methyltransferases, histone deacetylases, microRNAs, hepatic stellate cells, tumor-associated macrophages, extracellular matrix

Citation: Wang F, Malnassy G and Qiu W (2021) The Epigenetic Regulation of Microenvironment in Hepatocellular Carcinoma. Front. Oncol. 11:653037. doi: 10.3389/fonc.2021.653037

Received: 13 January 2021; Accepted: 22 February 2021;
Published: 15 March 2021.

Edited by:

Dongshi Chen, University of Pittsburgh, United States

Reviewed by:

Chiung-Kuei Huang, Indiana University School of Medicine-Lafayette, United States
Alessandro Carrer, Veneto Institute of Molecular Medicine (VIMM), Italy

Copyright © 2021 Wang, Malnassy and Qiu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Wei Qiu, wqiu@luc.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.