Next Article in Journal
Microneurotrophin BNN27 Reduces Astrogliosis and Increases Density of Neurons and Implanted Neural Stem Cell-Derived Cells after Spinal Cord Injury
Previous Article in Journal
Estetrol Increases Progesterone Genetic Response without Triggering Common Estrogenic Effects in Endometriotic Cell Lines and Primary Cultures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathogenesis of Hepatocellular Carcinoma: The Interplay of Apoptosis and Autophagy

by
Elias Kouroumalis
1,2,*,
Ioannis Tsomidis
2,3 and
Argyro Voumvouraki
3
1
Department of Gastroenterology, PAGNI University Hospital, University of Crete School of Medicine, 71500 Heraklion, Crete, Greece
2
Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Crete, Greece
3
1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Central Macedonia, Greece
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(4), 1166; https://doi.org/10.3390/biomedicines11041166
Submission received: 7 March 2023 / Revised: 9 April 2023 / Accepted: 12 April 2023 / Published: 13 April 2023

Abstract

:
The pathogenesis of hepatocellular carcinoma (HCC) is a multifactorial process that has not yet been fully investigated. Autophagy and apoptosis are two important cellular pathways that are critical for cell survival or death. The balance between apoptosis and autophagy regulates liver cell turnover and maintains intracellular homeostasis. However, the balance is often dysregulated in many cancers, including HCC. Autophagy and apoptosis pathways may be either independent or parallel or one may influence the other. Autophagy may either inhibit or promote apoptosis, thus regulating the fate of the liver cancer cells. In this review, a concise overview of the pathogenesis of HCC is presented, with emphasis on new developments, including the role of endoplasmic reticulum stress, the implication of microRNAs and the role of gut microbiota. The characteristics of HCC associated with a specific liver disease are also described and a brief description of autophagy and apoptosis is provided. The role of autophagy and apoptosis in the initiation, progress and metastatic potential is reviewed and the experimental evidence indicating an interplay between the two is extensively analyzed. The role of ferroptosis, a recently described specific pathway of regulated cell death, is presented. Finally, the potential therapeutic implications of autophagy and apoptosis in drug resistance are examined.

1. Introduction

Hepatocellular carcinoma (HCC) is a very complex world health problem. Approximately 905,677 new cases and 830,180 HCC-related deaths were reported in 2020 [1]. The estimation of more than 1 million deaths caused by HCC by 2030 has been predicted [2]. HCC-associated liver diseases are chronic viral hepatitis B (HBV) and hepatitis C virus (HCV) infection [3], non-alcoholic steatohepatitis (NASH) [4] and alcoholic liver disease (ALD). The etiological risk for HCC varies according to the geographical location [5]. Thus, in the United States, 54.9%, 16.4%, 14.1% and 9.5% of HCC cases are associated with the four commonest liver diseases HCV, HBV, NAFLD/NASH and ALD [6]. In Asia, NAFLD-associated HCC is lower compared to the West [7]. However, the etiology of HCC has changed over the last 20 years with a progressive increase in non-viral cases, such as metabolic HCC and a concomitant decline in viral etiology [8]. This was true in a study from Crete, where the initial high hepatitis C virus association decreased, and alcohol ranked first among risk factors for HCC. Non-alcoholic fatty liver disease was also continually increased as an important risk of HCC [9].
The development of HCC is associated with some form of cellular death, which may be either programmed (PCD) (such as apoptosis, necroptosis and autophagy-dependent cell death) or non-programmable (such as pyroptosis and necrosis) [10,11,12,13]. Apoptosis is probably the commonest cause of PCD. Characteristically, apoptosis does not elicit inflammation because apoptotic bodies are engulfed by macrophages and are degraded by lysosomes in autophagy [14,15]. Autophagy is an important degradation process of cellular contents, leading to the recirculation of structural components of the cell and improved survival. Autophagy-dependent cell death is a rare kind of PCD [16,17,18]. Lysosomes are the most important subcellular organelles involved in the autophagic degradation of protein aggregates [19,20]. Ferroptosis is a different form of PCD as it depends on excessive iron and lipid peroxidation [21,22] and is closely related to a specific form of autophagy called ferritinophagy, which is a critical part of the turnover of cellular iron through the autophagic degradation of ferritin [23]. In addition, other forms of autophagy, such as lipophagy, and heat shock protein 90 (HSP90)-mediated chaperone-mediated autophagy (CMA) may induce ferroptosis by promoting lipid peroxidation. The purpose of this review is to present the current views on HCC pathogenesis and the complex interplay of autophagy, apoptosis and ferroptosis in the pathophysiology and treatment of HCC.

2. Pathogenesis of HCC

The cells of origin of HCC are not clear. Experimental evidence supports the implication of transformed mature hepatocytes as the cell of origin, but also the possibility that the source is liver stem cells [24,25].
HCC is the result of either mutations, such as those in the TERT promoter or p53 suppressor gene [26,27], or epigenetic modifications. Some of them are directly involved or activate important signaling pathways leading to HCC [28]. Genes coding for several signaling pathways, such as Wnt/β-catenin, oxidative stress, AKT/mTOR and MAP kinase, are often mutated in HCC and are used for the molecular classification of HCC [29]. All these molecular abnormalities are triggered by external factors, such as alcohol consumption, viral infection and abnormal nutrition. In general, three mechanisms are implicated in the initiation and progress of HCC, namely, persistent liver inflammation, endoplasmic reticulum (ER) stress and abnormalities of cell signaling pathways [5]. Inflammation is an important pathogenetic factor irrespective of the etiology of the liver disease that leads to HCC [30]. Inflammation starts when hepatocytes undergoing programmed or accidental death liberate factors, such as HMGB1 and HDGF, to initiate an inflammatory response [31]. Different inflammasomes, particularly the nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain containing 3 (NLRP3), are activated and lead to the release of the pro-inflammatory cytokine IL-1β. NLRP3 activation is mostly triggered by the production of ATP from the mitochondria of the damaged cells [32] and lysosomal disruption [33]. NLRP3 inflammasome activation in hepatocytes is a two-step process. Priming is the first step when damage-associated molecular patterns (DAMPs) from damaged cells and pathogen-associated molecular patterns (PAMPs) stimulate TLR receptors, followed by translocation of NF-kB to the nucleus and increase in pro-IL-1β and pro-IL-18 expression. The second step is triggered when extracellular ATP or active lysosomal enzymes finally lead to the activation of caspase-1. Cleavage by caspase-1 turns pro-cytokines into mature IL-1β and IL-18, while the cleavage of gasdermin D leads to a programmed cell death called pyroptosis. In this canonical activation of pyroptosis, fragments of gasdermin D form pores in the plasma membrane, killing the cell and releasing IL-1β and IL-18, which aggravates inflammation [34,35].
In the non-canonical pyroptosis, lipopolysaccharides (LPSs), from Gram-negative bacteria, turn the pro-caspases 4, 5 and 11 into active enzymes that cleave gasdermin D. The pores in the plasma membrane are formed, but without maturation, and release IL-1β and IL-18. This is not always the case, as caspase-11 may activate the NLPR3-dependent caspase-1 inflammasome and indirectly stimulate the release of intracellular cytokines [36,37].
Interestingly, inflammasome-mediated pyroptosis also occurs in non-parenchymal liver cells, implicating the gut microbiota. DAMPS and gut-derived PAMPs activate Kupffer cells that produce IL-1β and TNF. NLRP3 activation in hepatic stellate cells (HSCs) promotes the production of the profibrogenic cytokine and induces the expression of the profibrogenic molecule TGF-β. These events in concert lead to liver inflammation and fibrosis, being the link between liver damage and hepatocellular carcinoma [38,39].
It was suggested that cathepsin B from lysosomes is the triggering stimulus of the NLRP3 inflammasome, which is mostly mediated by the release of Cathepsin B [40,41,42]. However, mice macrophages deficient in cathepsin B showed comparable NLRP3 inflammasome activation with wild-type animals [43], suggesting that other products contribute to NLRP3 inflammasome activation. Cathepsins B, L, C, S and X are probable candidates in NLRP3 inflammasome activation by silica particles [44]. In an adenovirus infection, cathepsin B release is also a mediator of inflammation, but reactive oxygen species (ROS) inhibition reduces IL-1β secretion, indicating that ROS production might be the mechanism of the induction of inflammasome activation by cathepsin B [45].

2.1. Endoplasmic Reticulum (ER) and Oxidative Stress

ER stress is caused by the accumulation of unfolded or misfolded proteins in the ER lumen. Pathogens, mutations and an increased metabolic rate led to an increase in the protein secretory load of the hepatocyte. The accurate monitoring of protein folding is not maintained in ER, inducing the unfolding protein response (UPR) either to normalize protein synthesis or to induce cell death in severe ER stress [46,47]. ER stress in murine hepatocytes activates inflammatory pathways, such as NF-kB and TNF, leading to HCC induction [48]. Chronic ER stress and increased UPR activity have been implicated in the development of HCC and are present in HCC tumors irrespective of grade or stage [49,50,51,52].

2.2. Abnormalities of Signaling Pathways

  • mTOR pathway
The abnormal activation of the oncogenic phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling is associated with HCC. This is not unexpected, as this pathway is involved in various cellular functions, such as cellular proliferation, differentiation, apoptosis and metabolism [53]. The AKT/mTOR pathway is known to interfere with aerobic glycolysis regulating three limiting enzymes in the glycolytic pathway (hexokinase 2, phosphofructokinase 1, and pyruvate kinases type M2), a fact that is crucial for HCC progression [54,55,56,57]. The mTOR pathway is analyzed later, as it is implicated in the interplay between autophagy and apoptosis.
  • Wnt/β-catenin pathway
The deregulation of Wnt/β-catenin signaling is critical in human HCC [58,59]. A total of 35% of human HCC tumors had a gain-of-function mutation of CTNNB1 encoding β-catenin and loss-of-function mutation of AXIN1 [60,61]. Resistance to sorafenib and regorafenib treatment was attributed to activated Wnt/β-catenin in HCC patients [62,63]. There is evidence that a second signal is required because Wnt/β-catenin alone is not sufficient to induce hepatocarcinogenesis. Oncogenic mutations of β-catenin cooperate with other oncogenes, such as c-Met [64,65,66] and K-RasV12 [67]. After activation, β-catenin induces several downstream targets that are implicated in HCC induction. c-MYC is one of the best-studied down-stream effectors of β-catenin [68,69].
  • miRNAs
miRNAs are regulators of several tumor-related genes in carcinogenesis, acting either as oncogenes or tumor suppressor genes. miRNAs are classified according to their implication in the main molecular pathways leading to HCC tumorigenesis [70]. miR-30a, miR-365, miR-526a, miR-377, miR-199a-5p and miR-330 all were implicated in apoptosis regulation and were either upregulated or downregulated in HCC [71]. Other mRNAs are involved in the repression of the PI3K/AKT/mTOR pathway or the Wnt/β-catenin pathway [53]. Tumor suppressor miRNAs are associated with either HCC initiation and progression [72] or with metastasis and recurrence [73]. Similarly, some pro-oncogenic miRNAs are associated with HCC initiation and progression [74], or are involved in HCC recurrence and metastasis [75].

2.3. Additional Factors Are Involved in HCC Pathogenesis

  • Exosomes
Exosomes transfer proteins, DNAs and RNAs, such as miRNAs, long non-coding RNAs (lncRNAs) and messenger RNAs (mRNAs), between HCC and normal cells. Exosomes initiate either local or systemic reactions, participating to the initiation and progression of HCC. Exosomes are used as biomarkers and therapeutic tools in HCC [76,77].
  • Ferroptosis
Ferroptosis is an iron-dependent process of regulated cell death, with accumulation of lipid peroxides that causes damage to liver cells and is associated with the development of HCC. It is analyzed later in the paper.
  • Microbiota
Microbial products in the bowel are deeply involved in HCC pathogenesis. Bacterial products from the gut microbiota can directly or indirectly damage DNA through the pro-duction of ROS [78,79]. The altered composition of the gut microbiota may be one of the mechanisms underlying the action of aflatoxin and other mycotoxins as powerful inducers of HCC [80]. In addition to gut microbial products, several studies have incriminated specific gut microbiota in association with HCC. Enterococcus faecalis is increased in HCV-induced HCC development [81]. A similar study showed a significant increase in Enterococcus species in patients with viral and alcoholic cirrhosis leading to HCC [82]. Microbiotas also interfere with bile acid metabolism, contributing to HCC development. Bile acid metabolites, produced by the gut microbiota, can cause inflammation, ROS overproduction and a reduction in apoptosis in the liver, finally leading to the development of HCC. Moreover, they can modulate the function of liver immune cells, affecting HCC progression. They can also indirectly contribute to the activation of the TLR4 receptor in hepatocytes and Kupffer cells. Bile acids increase gut permeability, acting on the tight junctions, and allow for an increased transportation of LPS to the liver, thus promoting angiogenesis and the downregulation of tumor suppressor miRNAs [83].
  • Calcium
Ca2+ is present in various cell compartments transported among them by transporters and exchangers, collectively known as the Ca2+ transportome. The impairment of the Ca2+ transportome contributes to HCC initiation, the formation of metastatic cells and reduction in cell death [84].
  • Autophagy and Apoptosis
These two critical parameters in the initiation and progress of HCC are analyzed separately. Two very informative reviews on the pathogenesis of HCC have been recently published [5,38].
Detailed pathophysiological factors implicated in HCC pathogenesis, including gene mutation and epigenetic changes, have been recently reviewed [85].

3. HCC Related to Specific Diseases

The pathogenesis of HCC has some discrete characteristics associated with the etiology of the liver disease.

3.1. HBV

HBV, as many other oncogenic viruses, does not directly lead to the development of cancer. It is the interaction with host factors that first initiates pre-neoplasia and then carcinoma [3]. Chronic inflammation from virally induced immune reactions is due to inflammasome activation, increased secretion of pro-inflammatory cytokines and increased levels of ROS within the liver microenvironment, which finally determines the development of cancer [86]. A chronic HBV infection leads to long-lasting hepatic inflammation, inducing cirrhosis and HCC progression due to increased hepatocyte turnover rates and the accumulation of mutations [87]. The alterations of platelets could also play a role in hepatocarcinogenesis [88,89].
In addition to the critical role of inflammation, HBV directly affects hepatocarcinogenesis, unlike HCV. This is because HBV integrates its genome into the DNA of the host, leading to genomic instability and mutagenesis in both proto-oncogenes and tumor suppressor genes [90]. HBV integration alters the tumor suppressor gene p53 or the combined p53–Rb pathway. Overall, most HCC cases harbor mutations in the component genes of either the p53 or the Rb pathway alone or of the combined p53–Rb pathway [91]. These alterations are associated with the inhibition of apoptosis [92]. This is the main mechanism through which HBV induces HCC in the absence of cirrhosis. In the presence of cirrhosis, hepatocarcinogenesis is multifactorial as there are several target genes impaired by HBV genome integration [93,94,95]. The integration of HBV DNA is constantly detected in 80% to 90% of tumor tissues and in 30% of non-HCC tissues next to HCC [93], even prior to the induction of HCC [96]. It is conceivable, therefore, that a hidden HBV infection may exist in HBsAg-negative patients and induce hepatocarcinogenesis [97]. The increased expression of truncated HBsAg, HBcAg and HBx proteins favors HCC development through the endoplasmic reticulum and mitochondrial stress [98,99].

3.1.1. The Important Role of HBx

HBx plays its role through several mechanisms [100,101]. HBx expression might induce hepatocarcinogenesis by interfering with telomerase activity during hepatocyte proliferation [96,102], upregulating the activation of human TERT [103,104,105]. In addition, the X protein interacts with several nuclear transcription factors and signal transduction pathways [106,107]. Among the most important deregulated pathways are the Wnt/β-catenin, the PI3K/Akt/mTOR and the Ras/Raf/mitogen-activated protein kinases (MAPK) pathways [108]. HBx and pre-S proteins activate mTOR signaling during an HBV infection and increase cell proliferation and angiogenesis [109,110]. Moreover, HBx has either anti-apoptotic [111,112,113] or pro-apoptotic activity [114]. These effects collectively lead to uncontrolled malignant transformation.
Epigenetic changes refer to chromatin changes without interference with the DNA sequence and include DNA methylation, histone modification and RNA-related silencing [115]. HBx causes epigenetic hyper- or hypo-methylation of the DNA and the tumor suppressor genes, inducing chromosomal instability [116,117]. HBx also promotes the acetylation of the histones H3 and H4, contributing to the pathogenesis of HCC [118,119,120,121].

3.1.2. The Role of RNAs

Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) also contribute to the initiation and progression of HBV-related HCC [122,123,124]. Several microRNAs can be regulated by HBV infection and promote hepatocarcinogenesis [125,126]. They modulate the Wnt/β-catenin signaling pathway, leading to the development of HCC [127].
The role of superinfection with HDV in the development of HCC is not clear. A direct oncogenic effect of HDV has not been unequivocally demonstrated [128]. A molecular signature of HDV–HCC different from HBV–HCC in malignant and non-malignant hepatocytes has been reported [129]. The pathogenesis of HBV-associated HCC has been recently reviewed [100,130].

3.2. HCV

The pathogenesis of HCC in HCV has certain similarities and differences compared to that in HBV. Thus, DNA damage also occurs during HCV replication, causing genomic instability and leading to hepatocarcinogenesis [131]. The role of chronic inflammation is as important in HCV as it is in HBV in relation to carcinogenesis. However, it has certain discrete characteristics, as inflammation is used for the immune escape of the virus. Macrophages are activated by the core and NS3 proteins of HCV, triggering the NLRP3 inflammasome and inducing the secretion of pro-inflammatory cytokines. IL-18 secretion induces NK cell activation. In addition, IL-1β and IL-6 production by macrophages support the activation of HSCs, increasing collagen deposition and fibrosis. IL-1β, IL-6 and TNF-α secretion may lead to malignant induction [132,133]. The chronic inflammatory environment in combination with certain viral proteins leads to a continuous activation of signaling pathways associated with hepatocyte survival, such as STAT3 and NF-kB. STAT3 is also involved in the development of myeloid-derived suppressor cells (MDSCs), which produce IL-10 favoring the expansion of regulatory T cells. Tregs impair immune response, and this is further accentuated by the increased expression of programmed cell death protein 1 (PD-1) and Fas ligand (FasL), driving the HCV-specific cytotoxic T lymphocytes to their apoptosis [134]. Epigenetic abnormalities similar to HBV also favor HCV-induced hepatocarcinogenesis. Thus, DNA methylation, histone modifications and microRNAs are also involved in the development of HCC [135]. LncRNAs and microRNAs similarly contribute to the induction and progression of HCV-associated HCC. miR-373 forms a complex with LINC00657, promoting uncontrolled cell growth [136]. The hypermethylation of certain promoter regions suppress mRNA expression, favoring the progression of HCV-associated HCC [137].
The major difference between HBV- and HCV-induced HCC is the fact that the RNA hepatitis C virus cannot integrate into the host genome in a way similar to HBV. On the contrary, most of the carcinogenic effects of HCV are mediated through the action of its viral proteins, which deregulate host cellular cycle checkpoints, resulting in DNA mutations in liver cells. Some effects of HCV proteins are similar to those produced by the HBx protein of HBV [100,134,138,139].
Earlier reports showed that the HCV core and NS5A proteins play a critical role in HCC development [140,141,142]. Thus, the association of NS5A and p53 allows the transcriptional repression of the p21/waf1, a downstream effector gene of p53, and may contribute to HCV-mediated HCC [143]. HCV core and NS3 proteins can also activate TERT, the enzyme responsible for the length of the telomere. Short telomeres lead hepatocytes to apoptosis. Therefore, active TERT reduces apoptosis, favoring HCC. Indeed, increased TERT activity was associated with the aggressiveness and a poor prognosis of HCC [144]. HCV infection activates the Wnt/β-catenin pathway, leading to the subsequent activation of cell survival genes. The core protein reduces the expression of the Wnt antagonists [145,146]. The NS5A protein activates PI3K/Akt signaling pathway, reducing the degradation of β-catenin [147,148] and blocking apoptosis [149,150]. The c-Myc oncogene is also activated through the Wnt/β-catenin pathway in a murine mode [151]. The activation of the PI3K/Akt/mTOR pathway by NS4A is similar to the activation of the same pathway by the HBx protein of the HBV, both inducing HCC [152]. Moreover, the activation of the mTOR pathway was related to tumor differentiation and vascular invasion in HCC patients [153]. Lysosomes degrade the tumor suppressor p53 protein through the CMA autophagy (CMA). CMA is activated as a result of chronic ER stress and increased unfolded protein response related to HCV infection [154,155].
The viral proteins core and NS5A also affect lipogenesis [156]. HCV-infected cells had increased levels of polyunsaturated fatty acids (PUFAs) [157]. The accumulation of long-chain fatty acids in the infected hepatocytes finally leads to the activation of the NF-kB pathway, which leads to increased cellular survival and the development of HCC [158].
Finally, HCV proteins are implicated in an interplay of four signaling pathways, all of which are implicated in the induction and progression of HCC. EGFR is phosphorylated after the virus binds to its entry receptor, CLDN1/CD81. EGF pathway activation is sustained by the action of NS3/4A and maintained by the reduction in EGF degradation mediated by NS5A.The STAT3 pathway is activated through the direct action of the core protein and indirectly by the NS5A protein. The activation of the TGF-β pathway is mediated by intermediary of the UPR, and via the core protein. The VEGF pathway is activated by the active HIF-1a, which is activated by the core protein. The HCV core protein can trigger angiogenesis through a crosstalk between TGF-β2 and VEGF expression, thus favoring the progress of HCC [159]. Details on the four pathways interplay can be found in a recent detailed review [134].
The NS5B protein is also implicated in the induction and progression of HCC. HCV infection negatively regulates the retinoblastoma tumor suppressor protein (Rb). This is mediated by the NS5B protein, which is complexed with Rb, targeting it for degradation via the proteasome. The disruption of the Rb pathway in cells infected with HCV inhibits apoptosis and promotes chromosomal instability, factors that favor the development of HCC [160]. Some of these mechanisms may persist even after HCV eradication, and thus the risk of HCC development is not abolished. This is still a hotly discussed subject [154,161].
The mechanisms of HCC induction and progression in HCV infection have been recently reviewed in detail [3,162].

3.3. NAFLD

NAFLD is the third commonest risk factor of HCC in the United States. Initial events include genetic, metabolic, immunologic and endocrine pathways, which in turn activate oncogenic mechanisms [4,163,164,165].
TERT, CTNNB1, TP53 and ACVR2A are frequently mutated genes in NASH-associated HCC. Interestingly, ACVR2A tumor suppressor gene mutations were more common in NASH–HCC than in the HCC of other etiologies [166]. The patatin-like phospholipase-3 (PNPLA3) I148M sequence variant is the best genetic association of NAFLD/NASH to date and a strong risk factor for HCC [167]. The cycle-related kinase (CCRK) androgen-driven oncogene interacts with pro-inflammatory signals induced by obesity to promote NASH-related HCC [168]. However, the molecular profiles of NAFLD-associated HCC are heterogeneous and no discrete mutation profile was identified as it is the case in hemochromatosis and HCV-related HCC. By contrast, in HBV- and ALD-related HCC, distinct mutational signatures are usually identified [27,169]. However, a novel mutational signature was recently associated with NASH–HCC, but this finding requires further validation [166].
Epigenetic alterations similar to viral HCCs, such as DNA methylation, histone modifications and the silencing of microRNAs, were identified in NAFLD–HCC as well [170,171,172]. Some DNA methylation changes during NASH–HCC are different from those of viral-hepatitis-associated HCC. MAML3 is one among the DNA hypomethylated genes. MAML3 is a co-activator of β-catenin-mediated transcription, increasing the transcriptional activity of β-catenin [173]. Yet, the only epigenetic alteration that has clearly been linked to NASH-related HCC is the gene encoding chromodomain helicase DNA-binding protein 1 (CHD1) [174].
Specific microRNAs may also participate in NAFLD progression into HCC. MiR-301a upregulation and miR-375 downregulation were reported as HCC progresses from the early to late stages [175].
Inflammation and metabolic disturbances, such as diabetes, obesity and iron deposition, provide a favorable tumor microenvironment for the progression of malignant lesions. Over 90% of HCC occurs in association with liver inflammation [30]. Inflammation in NASH is similar to HBV and HCV, leading to the upregulated release of pro-inflammatory cytokines, such as IL-18, IL-1β, TNF-α and IL-6. TNF-α is probably the best studied pro-tumor cytokine in HCC. It activates the NF-kB and JNK signaling pathways to promote cell survival and inhibit apoptosis [176,177,178]. IL-6-mediated STAT3 activation is also a major driver of hepatocyte repair and replication, favoring HCC development [176]. Inflammation and hyperinsulinism in NASH are constant proliferative signaling mechanisms, which cause rapid HCC growth [179]. However, the sequence fatty liver-inflammation–NASH–fibrosis–HCC is not always linear, and some patients may progress from fatty liver to advanced fibrosis and HCC in the absence of significant inflammation [180,181,182].
Insulin resistance and hyperinsulinemia activate the PI3K-Akt and MAPK pathways to induce cell proliferation and inhibit apoptosis [165,183]. Immune and endocrine mediators originating from the gut microbiome provide an additional molecular mechanism that is implicated in HCC progression [184].
Oxidative stress is another important factor in HCC development in NASH. Hepatocytes overloaded with fatty acids generate ROS and ER stress as a result of mitochondrial dysfunction to cause cell damage predisposing to HCC [185,186,187]. An early event in NAFLD, associated with oxidative stress, is the promotion of pathological polyploidization and may participate in HCC development [188]. Iron deposition in the liver is an important inducer of oxidative stress in NASH. Elevated levels of iron are observed in NASH patients and are associated with HCC development [189]. Interestingly, oxidative stress, associated with iron overload in NASH, activates Wnt/β-catenin signaling and triggers carcinogenesis [190].
It should be noted that HCV and NASH have common metabolic abnormalities, such as hepatic steatosis, insulin resistance and oxidative stress. However, the underlying mechanism is different. The metabolic deregulation of HCV is induced by the core protein, in contrast to the complicated metabolic abnormalities of NASH [191]. The important contribution of apoptosis and autophagy in NASH-related HCC is examined later.

3.4. Diabetes

Type 2 diabetes mellitus (T2DM) predisposes to HCC even after adjustment for the presence of alcoholism, obesity and chronic viral hepatitis [192,193,194]. Moreover, the HCC recurrence rate is 2.5–4-fold higher in patients with T2DM, independently of the presence of cirrhosis or of the etiology of the liver disease [195,196]. Only in patients with chronic hepatitis B or primary biliary cholangitis, diabetes did not increase the risk of HCC [197]. However, in a nationwide Japanese study, the annual incidence of HCC in diabetes increased from 0.11% to 1.0% when advanced fibrosis was present [198]. Mechanistically, insulin resistance (IR) results in hyperinsulinemia as well as the activation of the insulin receptor and insulin-like growth factor 1 (IGF-1) signaling pathways, which are important initiators and supporters of hepatocarcinogenesis. HCC cells overexpress IGF-1 and insulin receptor substrate-1 (IRS-1). IGF-1 inhibits apoptosis and favors, therefore, HCC cell proliferation [4]. IRS-1 increased activity results in the activation of several cytokine pathways, including PI3K/AKT/mTOR, which modify cell cycle and favor cellular proliferation. IRS-1 also seems to prevent TGF-β-mediated apoptosis. In addition, alterations in lipid and glucose metabolism stimulate the production of ROS and cause mutations in the p53 onco-suppressor gene [199,200]. Moreover, data show that LINC01572 is upregulated in HCC tissues from patients with diabetes. The overexpression of LINC01572 increased HCC cell proliferation through sponging miR-195-5p, leading to an increase in glycolysis and the activation of the PI3K-AKT signaling pathway [201].

3.5. ALD

In most European countries, ethanol participates in the development of HCC be-tween 30 and 50% [202]. In France, the geographical distribution of HCC is not uniform, but the most affected regions are areas with a high wine production or those with excessive alcohol consumption [203]. On the contrary, in Crete, Greece, hepatocellular carcinoma is associated with the dispersion of HCV and HBV. ALD-related HCC was not very common in the past, but an increasing trend has been identified [204].
Hepatocarcinogenesis in excessive alcohol consumption is mostly due to the metabolic mechanisms associated with ethanol metabolism into acetaldehyde by alcohol dehydrogenase (ADH) and the microsomal CYP2E1. Acetaldehyde enters the mitochondria and oxidizes to acetate by mitochondrial aldehyde dehydrogenase (ALDH) [205]. There are two main mechanisms of cellular damage caused by acetaldehyde. The first is the formation of DNA and protein adducts. The second is the production of increased amounts of ROS by mitochondria, causing oxidative stress through lipid peroxidation that further deteriorates DNA mutagenesis. Oxidative stress is further aggravated by the iron deposition and lipid accumulation associated with excess ethanol [206]. ROS accumulation damages cellular macromolecules and is a critical factor in the progression of hepatocarcinogenesis through the formation of lipid peroxides, such as 4-hydroxy-nonenal [207].
An additional important factor, participating in liver carcinogenesis, is the increased gut permeability and the bacterial overgrowth caused by ethanol metabolism in the gut [208]. Endotoxins from the gut enter the portal vein and activate Kupffer cells, interacting with the receptor TLR4, which leads to the secretion of pro-inflammatory cytokines, such as IL-1, IL-6 and TNF-a, and to the initiation of liver inflammation [209,210]. NF-kB, one of the regulators of the inflammatory response, is also activated by TNF-a [211,212]. Moreover, the IL-6/STAT3 and TNF-a/NF-kB pathways have been implicated in hepatocarcinogenesis [213]. The mitogenic activity of hepatocyte is increased and hepatocyte apoptosis is inhibited, resulting in the induction of HCC [211].
The role of inflammasomal activation has been investigated in mice with ALD. IL-1β signaling is mandatory for the development of alcohol-induced liver steatosis, inflammation and injury. The upregulation of caspase-1 activity and inflammasome activation are the mediators of increased IL-1β [214].
Acetaldehyde production in the hepatocyte is influenced by the genetic variants of ADH and ALDH. The alleles ADH1C*and ALDH2*2 are associated with an increased probability of HCC [215,216,217]. Other genetic determinants are implicated in the severity of ALD, such as the patatin-like phospholipase domain-containing protein 3 (PNPLA3), the transmembrane 6 superfamily member 2 (TM6SF2) and the membrane-bound O-acyltransferase domain-containing protein 7 (MBOAT7) [218,219,220].
Moreover, acetaldehyde interferes with methyl group transference, leading to DNA hypomethylation and modifications of both oncogenes and tumor suppressor genes [221,222].

3.6. Hemochromatosis

Iron overload is the characteristic of hereditary hemochromatosis (HH), and increased iron produces increased ROS through the Fenton reaction, leading to DNA damage and HCC. Studies on the association of hemochromatosis and ferroptosis as a risk factor of HCC are limited, possibly because these studies were conducted before ferroptosis was described [223].
A recent study demonstrated that ferric citrate triggers ferroptosis in cells, suggesting the involvement of ferroptosis in HH [27]. This study also showed that SLC7A11 is a candidate gene of ferroptosis in HH and indicated that the Nrf2 activation may be a compensatory mechanism to protect against iron-overload-induced ferroptosis in HH [224].
Nonetheless, the risk of HCC in HHH was clearly overestimated in the past. More recent studies have indicated that this risk is lower and mostly occurs in patients with cirrhosis at the time of diagnosis. The true incidence of HCC in HH is better derived from population-based studies [225].
In a population study, the overall standardized incidence ratio of HCC was 1, which increases among first-degree relatives of the patients [226].
However, a very recent study suggested that HH without cirrhosis is an independent risk factor for HCC after adjustment for all known risk factors. The aOR was 28.8 higher than any other disease risk factor for HCC [227].

4. Apoptosis

Apoptosis is one of the forms of programmed cell death, in which characteristic cellular contents are not liberated into the surrounding environment. Apoptosis is mediated by a sequential activation of a series of caspases. The initiator 8 and 9 caspases are activated from pro-caspases upon sensing the initial signal via intracellular sensors, and activate the executioner 3, 6 and 7 caspases. The intrinsic pathway initiates apoptosis by an internal cell damage, while an external signal initiates the extrinsic pathway of apoptosis [228]. External stimuli, such as TNF-α, Fas ligand and TNF-related apoptosis-inducing ligand (TRAIL), operate through surface death receptors, while intrinsic stimuli operate by the mitochondrial signaling pathway [229,230]. The intrinsic pathway is associated with mitochondrial outer membrane permeabilization (MOMP), followed by the release of cytochrome c and leading to apoptosome formation. The effector caspases cleave hundreds of cellular proteins causing DNA fragmentation and actin reorganization, leading to membrane blebbing. Phosphatidylserine (PS) molecules exposed on the plasma membrane act as “eat me” signals for macrophages [11].
MOMP is regulated by the BCL-2 family. The pro-apoptotic BCL-2 proteins, BCL-2-associated X protein (BAX) and BCL-2 antagonist killer 1 (BAK) are activated by the pro-apoptotic proteins BAD and BID, leading to the activation of the caspase cascade and cell apoptosis. Protection from apoptosis is provided by pro-survival BCL-2 proteins [231].
This intrinsic apoptotic pathway initially increases the activity of pro-apoptotic BH3-only proteins that bind and neutralize the members of the pro-survival BCL-2 family [232,233]. BAK and BAX are then free to assemble into structures that cause MOMP [234,235]. Details of the apoptotic pathway have been recently reviewed [236,237,238].

Apoptosis and HCC

In human HCC, the activation of the anti-apoptotic BCL-xL is usually associated with a parallel downregulation of BAX [239]. Moreover, the inhibition of caspases is also common in HCC, associated with TGF-β signaling. All these contribute to liver cancer initiation and progression [240]. As mentioned above, a significant number of HCC patients have alterations of the NF-kB pathway, particularly patients with NASH-induced HCC [241,242]. NF-kB activation via TNF promotes HCC by inhibiting apoptosis. The effects of NF-kB may promote HCC development by either activation or inhibition. The opposing different effect of NF-kB can be explained. The activation of NF-kB is linked to several pathways inhibiting apoptosis, such as the Bcl-2 family members and FLIP, but also to other pro-survival pathways [231,243]. On the other hand, increased hepatocyte apoptosis is associated with increased compensatory proliferation, leading to an increased incidence of oncogenic mutations. Murine experiments with reduced hepatocyte NF-kB activation increase hepatocyte apoptosis and compensatory proliferation, followed by increased predisposition to HCC [30,244]. Similar to NF-kB, c-Jun N-terminal kinase (JNK) can promote HCC development by inducing inflammation and hepatocyte proliferation, but it may also have an anti-tumorigenic function. Taken together, these findings indicate that the TNF-α, NF-kB and JNK pathways may have either pro-survival or cell death effects, both leading to HCC development [178,245,246,247].
The activation of caspases and other apoptosis-related molecules is a common finding in the liver of NASH patients [244]. Apoptotic hepatocytes stimulate immune cells and hepatic stellate cells, contributing to the progression of fibrosis. Inflammasomes, oxidative stress and ER stress also contribute to the progression of NASH and development of HCC by the induction of apoptosis [248], but the exact interplay is still debated.
Apoptosis may also be involved in the development of ALD-related HCC. JNK modulation has a dual role. Experiments in hepatocytes indicate that JNK activation by ethanol or acetaldehyde can be both pro- and anti-apoptotic. The activation of p42/44 MAPK, on the other hand, is anti-apoptotic, for both ethanol and acetaldehyde [249].

5. Autophagy

The understanding of the mechanism of autophagy ( a Greek word, meaning self-eating) is based on the pioneer works of Christian De Duve and Yoshinori Oshumi [250,251].
The sequential stages of autophagy include induction, phagophore, autophagosome and autolysosome formation and finally degradation [252,253,254].
The first step in the induction of autophagy is the formation of the ULK1 complex from the assembly of the ULK1, ATG13, FIP200 and ATG101 proteins. The ULK1 complex induces the formation of the PI3KC3 complex containing the proteins Beclin1, Atg14, VPS15 and VPS34. Both complexes are necessary for the formation of the autophagosome. Beclin1 regulates the effects of the complex. When the anti-apoptotic protein Bcl-2 binds to Beclin1, it reduces the affinity of Beclin-1 for VPS34 and inhibits autophagy. Beclin1 is released from Bcl-2 by BNIP3, another member of the Bcl-2 family with a BH3 domain, and autophagy is initiated. The protein Rubicon also binds to Beclin1 and inhibits the PIK3C3 activity. The transformation of phagophores into autophagosomes requires the Atg12–Atg5–Atg16 complex and the phosphatidylethanolamine (PE)-conjugated LC3II (Atg8) system. Finally, the autophagosome fuses with the lysosome for the degradation of the contents, which thereby degrades the autophagosomal contents (Figure 1) [255,256].
AMPK, a sensor of the cellular energy, is an important regulator of autophagy. Upon energy starvation, the activated AMPK initiates autophagy by increasing ULK1 activity through the serine phosphorylation of ULK1. Autophagy is inhibited by the PI3K/AKT/mTORC1 pathway, when enough cellular energy is available. AMPK can negatively regulate mTORC1, either directly through the phosphorylation of mTORC1 activity or indirectly by activating TSC2, which is a strong inhibitor of mTORC1 [257]. Recently, an additional mechanism for mTORC1 activation under energy-rich conditions was described. mTORC1 phosphorylates the protein Pacer, causing the disruption of the Pacer, Stx17 and HOPS complex, thus abolishing the autophagosome maturation mediated by this complex [258].
p38 also upregulates autophagy by inhibiting mTOR, while JNK and BNIP3 disrupt the Bcl-2–Beclin11 interaction, thereby initiating autophagy [259,260,261].
Two additional autophagy regulators have been described. The lncRNA NBR2 inhibits Beclin 1-dependent autophagy and suppresses autophagy-induced cell proliferation in HCC [262], while Forkhead box O3 (FOXO3), a member of the FOXO subfamily of transcription factors, upregulates autophagy, acting on ULK1, Beclin-1 and LC3 [263].
A detailed overview of the autophagy mechanisms involved in HCC development and progression have been recently published [264,265].
Mitophagy is a special form of autophagy that clears damaged mitochondria and is mediated by two molecular pathways. The first pathway is activated by the HIF1A/HIF-1a hypoxia-inducible factor 1 subunit alpha (HIF1A/HIF-1a). The second pathway is the PINK1 (PTEN-induced kinase 1)-PRKN (parkin RBR E3 ubiquitin protein ligase) pathway, activated by membrane depolarization. An important regulator of mitophagy is the TP53/p53, which facilitates mitochondrial dysfunction and disturbs the clearance of damaged mitochondria by mitophagy [266].

Autophagy and HCC

Autophagy is implicated in the initiation and progression of HCC in many ways. It is closely associated with inflammation, which is a critical factor in HCC development. Autophagy and inflammasomes are interconnected as the same mechanisms regulate them, but through different pathways. The NLRP3 inflammasome activated by DAMPS induces caspase-1, leading to pyroptosis, as mentioned above [34,35].
Caspase-1 is also a mediator of autophagy activation. Autophagy eliminates inflammasomes and also damaged cellular organelles that would otherwise act as DAMPS [267,268]. However, this negative correlation between autophagy and inflammasomes is not always operative, as both can move towards the same direction in cases of NF-kB activation [269]. Moreover, the behavior of autophagy depends on the involvement of liver cells. Autophagy is protective in NAFLD and ALD, reducing lipid accumulation and oxidative stress. Autophagy activation in Kupffer cells also inhibits inflammation and liver fibrosis, but favors fibrosis if activated in HSCs [270,271].
In the early stages of cancer, autophagy behaves as a tumor suppressor, eliminating damaged mitochondria and unfolded proteins. It also decreases lipid accumulation in liver cells, reducing inflammation. Autophagy, however, acts as a tumor promoter after HCC induction, maintaining oxygen homeostasis to help the survival of malignant cells. In addition, it favors the appearance of resistance to treatment [272,273,274,275]. Both macroautophagy and CMA act as a double-edged sword in liver hepatocarcinogenesis, as shown by experimental and clinical studies. Mice with defective autophagy do not develop HCC, irrespective of any challenge, due to the activation of tumor suppressors, such as p53. However, after the induction of HCC, autophagy is necessary to degrade tumor suppressors, thus promoting the progression of HCC [276,277].
Increased levels of the autophagy marker LC3-II are correlated with lymph node metastasis, high vascular invasion and, most importantly, the reduced 5-year survival of HCC patients [278,279].
The macroautophagy flux is impaired in the final stages of HCC. However, during the later stages of HCC, more than 95% of tumors have an expression of LAMP-2A that is consistent with the induction of CMA in HCC [280]. CMA is probably upregulated under continuous, severe stressful stimuli and functions as a potential compensatory mechanism to reduce macroautophagy after the induction and establishment of HCC [281].
The activation of the Wnt/β-catenin pathway favors the development of HCC, as previously mentioned. Experimental evidence indicated that Wnt/β-catenin inhibitors repress the proliferation of HCC cells by regulating autophagy [282]. However, another report suggested that other mechanisms not related to autophagy led to an interference with Wnt secretion and a reduction in tumor growth through alternative, not-yet-identified pathways [283].
A special form of autophagy that removes damaged mitochondria is also a double-edged sword in HCC growth. Increased mitophagy was reported to suppress HCC cell survival [284,285]. The opposite has also been suggested, as increased mitophagy may promote hepatoma cell survival either through an increased production of ROS or through the attenuation of p53 activity [286,287].
Certain points should be noted in connection to the HCC of specific etiology. In HBV, the HBx protein increased autophagosome formation and reduced lysosomal acidification and the accumulation of immature cathepsin D [288,289]. This repression of lysosomal acidification is important for the development of HBV-associated HCC [290]. The inhibition of lysosomal degradative function by hydroxychloroquine induced p53 and increased apoptosis, but the activation of autophagy using the Torin-1 inhibitor of mTOR increased HCC growth [280]. Moreover, Arrestin beta 1 (ARBB1) promoted HCC formation through the interaction of HBx with LC3 and the promotion of autophagy [291]. HCV generates cellular stress and activates CMA autophagy to promote cell survival. CMA activation leads to HCC induction due to the repression of hepatic innate immunity and the degradation of several tumor suppressors [292].
Lipolysis and autophagy are interconnected. Autophagy reduces lipid accumulation, oxidative stress and inflammation in the liver, but autophagy also regulates adipogenesis and differentiation in the adipose tissue [293]. Similar to HBV, autophagic flux and the level of mature cathepsin D were reduced in three murine models of NAFLD, suggesting defective lysosome acidification under endoplasmic reticulum stress [294]. In NASH and NASH–HCC, autophagy has a dual role. On the one hand, autophagy reduces intracellular lipid droplets, attenuating lipotoxicity and inflammation. On the other hand, autophagy also affects adipogenesis and adipocyte differentiation. Basal autophagy, therefore, behaves as a tumor suppressor. After the induction of HCC, unbalanced autophagy contributes to carcinoma cell survival [295,296]. However, the contention of whether autophagy favors or inhibits NASH progression has not been settled. Defective autophagy is also linked to NASH–HCC through the induction of pro-inflammatory NF-kB activity, while defective mitochondria are retained, producing ROS to damage cellular DNA [297]. Interestingly, under conditions of reduced autophagy, hepatocytes were found to release the high-mobility group box 1 (HMGB1) protein, driving the proliferation of isolated hepatic progenitor cells. This could be an additional mechanism for the development of NAFLD–HCC [298].
ALD-associated HCC is also related to autophagy. Mitochondrial aldehyde dehydrogenase (ALDH2) is a critical enzyme further metabolizing the acetaldehyde produced by ethanol metabolism. Experiments in ALDH2 transgenic mice demonstrated that ALDH2 mitigates alcohol-induced liver steatosis and inflammation through the regulation of autophagy [299]. Moreover, TNF-α-induced protein 8 (TNFAIP8) is involved in the progression of HCC. TNFAIP8 induces autophagy by inhibiting the AKT/mTOR pathway in HCC cells. In addition, a direct interaction with ATtg3–Atg7 proteins was also reported. This mechanism is operative in the ALD of mice and humans, but not in NASH [300].
An important aspect of HCC development is the effect that autophagy exerts in the tumor microenvironment and particularly in TAMs. The increased autophagy of TAMs leads to the anti-tumoral M1 polarization, while the inhibition of autophagy leads to M2 polarization that favors hepatocarcinogenesis. The activation of the mTOR pathway, which is a negative regulator of autophagy, leads to M2 phenotype polarization and the promotion of HCC. The coagulants tissue factor (TF) and factor VII (FVII), locally produced in tumor microenvironment, promote HCC growth by the repression of autophagy mediated by mTOR activation and Atg7 [301,302].

6. Interplay between Apoptosis and Autophagy

Autophagy and apoptosis are normally tumor suppressor pathways. The degradation of oncogenic molecules by autophagy prevents cancer initiation, while apoptosis eliminates cancer cells. Under conditions of stress, autophagy may facilitate the survival of tumor cells [303]. Similar external or internal signals can induce either apoptosis or autophagy. They usually exhibit mutual inhibition, although single-cell experiments indicated that, in many instances, they are both operational. Apoptosis and autophagy may act in concert to kill or, alternatively, the activity of one mechanism can exclude that of the other. The result is important for the effectiveness of chemotherapy in several cancers, including HCC [304]. Usually, autophagy precedes apoptosis [305]. The initial activation of autophagy is an effort towards survival. The initiation of apoptosis will eventually kill the cell if autophagy fails. The induction of autophagy inhibits apoptosis, while apoptosis suppresses autophagy initiation [306]. Bcl-2 is an important regulator of the interplay. Bcl-2 inhibits the pro-apoptotic Bax and interacts with the PI3K complex of the autophagy pathway, promoting survival. However, the phosphorylation of Bcl-2 inhibits its binding to Bax, leading to apoptosis [307]. A pro-apoptotic role of autophagy has also been reported [308].
An example of the concerted action of the two pathways to inhibit the replication of hepatocellular carcinoma cells was recently published. Solamargine, a traditional Chinese herb medicine, induced both apoptosis and autophagy to repress the replication of hepatoma cell lines [309]. Similarly, Jujuboside B, an ingredient of the traditional Chinese medicine Zizyphi Spinosi Semen, induced both autophagy and apoptosis in breast cancer cells [310]. A synergy between autophagy and apoptosis was also described in the anti-fibrotic activity of curcumol. It can induce both the autophagy and apoptosis of hepatic stellate cells. Since fibrosis is an important factor for the initiation of HCC, this dual action of curcumol may favorably influence HCC initiation [311]. However, a detrimental outcome may be the result of the concerted action of apoptosis and autophagy. This was reported in human kidney mesangial cells incubated with homocysteine, which induced ER stress. Both autophagy and apoptosis were activated, and the viability of cells was significantly reduced [312].
On the other hand, the ER stress and UPR that follows is an example of the mutual inhibition of apoptosis and autophagy. The accumulation of unfolded proteins in the ER lumen induces ER stress and the activation of three major UPR pathways (PERK, IRE1α and ATF6) leading to UPR. The final result is the inhibition of apoptosis and the activation of autophagy. This mechanism may be related to the proliferation of HCC cells and the resistance of HCC to chemotherapy [47]. The impact of autophagy on cell survival during ER stress varies according to the tissue type. ER-induced autophagy protects against cell death in colon and prostate cancer cells. However, in normal human colonocytes, autophagy does not counteract ER stress but facilitates ER-induced apoptosis [313]. A mutual exclusion is not operative only in the liver. It is operative in secondary hyperparathyroidism cells, where the autophagy inhibitor chloroquine enhances experimentally induced apoptosis [314]. In hepatocellular carcinoma, autophagy may either support apoptosis or antagonize apoptosis. The activation of autophagy may lead to the induction of apoptosis and the inhibition of the growth of hepatoma cells [315,316]. Experimental evidence indicates that lipophagy, a special form of autophagy, can also act in both ways. It can either supply tumor cells with energy important for their proliferation or suppress tumor development through the direct inhibitory effect of acid lipase [317,318]. Lipophagy can also induce apoptosis via the induction of mitochondrial stress [319].
Experimental evidence has also indicated that an interplay between autophagy and apoptosis may be implicated in the pathogenesis of NASH and ALD. JNK1 increases palmitate-induced lipoapoptosis, whereas JNK2 activates autophagy and inhibits palmitic acid lipotoxicity, improving the survival of hepatoma cells [320,321]. The promotion of autophagy by the mitochondrial uncoupling protein 2 (UCP2) also inhibits apoptosis [322]. The inhibition of autophagy by the tumor protein p53-binding protein 2 (TP53BP2) may be involved in NASH [323]. The overexpression of Rubicon, a Beclin-1-interacting negative regulator for autophagosome–lysosome fusion, causes the suppression of the late stage of autophagy. Its blockade mitigated autophagy suppression and reduced palmitate-induced ER stress and apoptosis [324]. Parkin-mediated mitophagy may attenuate apoptosis, improve the quality of mitochondria and suppress hepatocyte steatosis in models of ALD due to Parkin translocation into mitochondria [325]. Sirtuin 3 (SIRT3) is a nicotinamide adenine dinucleotide-dependent deacetylase located within the mitochondria. SIRT3 is a negative regulator of autophagy. SIRT3 overexpression causes AMPK inhibition, mTOR activation and finally autophagy suppression, promoting the hepatocyte lipotoxicity induced by saturated fatty acids [326].
An important field of research is the identification of pathways where autophagy and apoptosis meet (Figure 2). Several pathways that mediate the interplay between autophagy and apoptosis have been identified and are analyzed in this paper [327].

6.1. Beclin-1

The Beclin-1/BCL-2 interaction was the first described molecular connection between autophagy and apoptosis. Beclin-1, the mammalian homolog of the yeast Atg6, participates in autophagosome formation as a component of the PI3K complex [328,329]. The interplay between autophagy and apoptosis is mediated, in part, by the interaction between Beclin-1 and the anti-apoptotic proteins BCL-2 and BCL-XL [330,331], as previously mentioned. This inhibits the pro-autophagic function of Beclin-1, but does not interfere with the anti-apoptotic activity of the BCL-2 family proteins. In addition, the inactivation of Beclin-1 triggers apoptosis [331]. Several BH3-only proteins can activate both autophagy and apoptosis. To induce apoptosis, BH3-only proteins directly neutralize anti-apoptotic proteins from the BCL-2 family, and stimulate those with pro-apoptotic functions. Beclin-1 is such a protein, as it possesses a BH3 region [332]. inhibiting these anti-apoptotic proteins, or, alternatively, activating the pro-apoptotic BCL-2 family members, such as BAX and BAK [333]. On the other hand, BH3-only proteins disrupt this interaction and permit Beclin-1 to increase autophagic activity. Only BIM, a unique BH3-only protein, has an opposite effect on autophagy. BIM interacts with Beclin-1 and prevents autophagy. NIX, another BH3-only protein, localized in the mitochondria, favors mitophagy. JUN N-terminal kinase (JNK) is also associated with autophagy regulation. JNK induces autophagy or apoptosis through the phosphorylation and inactivation of BCL-2, leading to apoptosis or through the phosphorylation of BIM that disrupts the inhibitory interaction with Beclin-1, leading to autophagy [306].

6.2. Beclin-1 in HCC

Autophagy is significantly reduced in the most aggressive HCC cell lines and tissues, particularly when the Bcl-xL protein is overexpressed. These findings were corroborated in curative resection specimens from HCC patients where the reduced expression of Beclin-1 was negatively correlated with survival only in the Bcl-xL+ patients, indicating that an increased expression of the anti-apoptotic gene Bcl-xL was associated with decreased expression of Beclin-1 and a poor prognosis [334].
These results were verified in two additional studies. The first was performed in material from 103 HCC patients, where Beclin-1 was negatively correlated with the anti-apoptosis protein Bcl-2 and positively correlated with the pro-apoptosis protein Bax. The 5-year survival rates were considerably higher among patients with strong Beclin-1 positivity compared to those with weak or negative expression [335]. The second study of 35 HCC patients reported similar results [336].
Interestingly, it was found that nitric oxide (NO) may influence the autophagy–apoptosis balance in HCC through Beclin-1. The levels of NO were significantly increased in HBV-related HCC compared to cirrhosis. Further experiments with human hepatoma cells showed that NO induced apoptosis and inhibited autophagy, whereas the induction of autophagy could attenuate NO-induced apoptosis. NO controls the switch between apoptosis and autophagy, disrupting the Beclin-1/Vps34 complex and increasing the Bcl-2/Beclin-1 connection [337].
The actions of sorafenib, a drug used for the treatment of advanced HCC, are additional evidence for the significance of Beclin-1 in regulating autophagy and apoptosis. Sorafenib induces autophagic cell death in HCC through Beclin-1 and apoptosis [338]. The induction of apoptosis by sorafenib is probably a more important mechanism for hepatoma cell death as the inhibition of autophagy augments the effect of sorafenib, increasing apoptosis [339,340].
A very recent report used a different approach that showed the interplay between autophagy and apoptosis. Vaccinia-related kinase 2 (VRK2) increases sorafenib resistance in HCC cells. This is obtained by the phosphorylation of Bcl-2, thus enhancing the dissociation of Bcl-2 from Beclin-1, followed by the formation of the Beclin-1/Vps34 complex, which facilitates autophagy. Furthermore, VRK2 phosphorylated Bcl-2, promoting the interaction of Bcl-2 with BAX, thereby reducing apoptosis [341].
In addition, Beclin-1 is involved in the regulation of apoptosis through the action of caspase. Growth factor depletion leads to the caspase-mediated cleavage of Beclin-1, impairing autophagy. A fragment of Beclin-1 is then generated and localized to mitochondria, leading cells to apoptosis through the release of pro-apoptotic factors, such as BAX [342]. The pro-apoptotic protein BAX reduces autophagy, promoting the caspase-mediated cleavage of Beclin-1. This is an indication that apoptosis can suppress autophagy [343]. The link between autophagy and apoptosis is further supported by evidence that other autophagy-related proteins, such as ATG5, are also substrates for caspase cleavage and the induction of apoptosis. The cleaved ATG5 translocates into the mitochondria, inducing the mitochondrial apoptotic pathway [344]. Therefore, the caspase-mediated cleavage of ATG5 and Beclin-1 switches autophagy to apoptosis. The involvement of caspase-3 constitutes a switch between autophagic or apoptotic cell death [345].

6.3. mTOR Interaction with Autophagy–Apoptosis and the Regulation of mTOR in HCC

mTOR is implicated in several signaling pathways regulating cell proliferation, autophagy and apoptosis [346]. There are two main mTOR signaling pathways: the classical PI3K/Akt/mTOR and the LKB1/AMPK/mTOR signaling pathways. The glycogen synthase kinase 3 beta (GSK3B)-mediated phosphorylation of ULK1 is important in autophagy induction, suppressing the mTOR pathway and potentially inducing tumorigenesis [347,348].
mTOR has also several effects on apoptosis depending on the cells involved and its effect on the activation of downstream targets, such as p53 and BCL-2 proteins [349].
The anti-apoptotic BCL-2 homolog MCL1 controls autophagy and apoptosis. The interplay between BAX and Beclin-1 downstream of MCL1 degradation finally determines if autophagy or apoptosis will prevail. It should be noted that mTOR inhibition, following nutrient deprivation, causes MCL1 degradation [350]. On the contrary, both autophagy and apoptosis may be controlled through the activation of the mTOR pathway. Thus, β-carotene attenuated both the apoptosis and autophagy of enterocolitis IEC-6 cells stimulated with LPS, activating the PI3K/AKT/mTOR signaling pathway [351]. There is strong experimental evidence that the mTOR pathway regulates autophagy and apoptosis in HCC. The importance of the PI3K/Akt/mTOR signaling pathway in HCC induction and progression has been established. It is implicated in every etiology of HCC (viral, ALD and NASH). The mTOR pathway is overexpressed in almost 50% of HCC and the impaired activation of this pathway affects cell proliferation, differentiation, autophagy and the epithelial–mesenchymal transition (EMT) [352,353].
Apigenin, a dietary flavonoid, induced apoptosis and autophagy in HCC cells by inhibiting the PI3K/Akt/mTOR axis. Although autophagy protected cells from death, the end result was the inhibition of cellular proliferation [354]. Brusatol, a traditional Chinese herbal medicine, inhibited proliferation and induced apoptosis in liver cancer lines. The autophagy inhibitor chloroquine attenuated Brusatol-induced apoptosis, indicating that Brusatol promoted autophagy-induced apoptosis in HCC through the inhibition of the PI3K/Akt/mTOR axis [355].
The upregulation or downregulation of mTOR-related oncogenic lncRNAs contributes to the aberrant expression of oncoproteins, leading to the disturbed regulation of the mTOR axis [356,357]. The aberrant expression of lncRNAs is associated with the metastasis, recurrence and chemoresistance of HCC [358]. In particular, the inhibition of the lncRNA HIF1A-AS1 increases apoptosis by reducing HIF-1α/mTOR-induced autophagy, while its overexpression is related to the TNM stage and lymph node metastasis [359]. A synergistic effect of PI3K/AKT/mTOR pathway-induced autophagy and apoptosis was recently reported. The concomitant incubation of hepatoma cell lines with aloin and metformin inhibited cellular proliferation, increasing both autophagy and apoptosis [360]. The regulation of mTOR in HCC has been recently reviewed [361].

6.4. p27kip1

p27 kip1 is a cyclin-dependent kinase inhibitor and a tumor suppressor. p27Kip1 is a critical mediator of autophagy and apoptosis. Unlike other tumor suppressors, such as p53, the loss of p27 expression, frequently found in tumors, occurs via proteasomal degradation or re-localization, and not through genetic or epigenetic modifications [362]. The cellular location of p27Kip1 is partially controlled by phosphorylation from several kinases, such as Akt and AMPK. Thus, the cytoplasmic location of p27Kip1 has been found to promote cellular survival through autophagy (Figure 3).
Nuclear p27Kip1, however, increases cell susceptibility to apoptosis or senescence [363]. A reduction in energy metabolism activates the LKB1-AMPK energy-sensing pathway, leading to the phosphorylation and stabilization of p27kip1. Autophagy is induced and cell survival is increased. A reduction in p27kip1 under these conditions activates apoptosis [364,365]. Recently, the DNAJC5 protein was reported to be associated with the regulation of p27. DNAJC5 expression is frequently increased in human HCC and is strongly related to poor prognosis. DNAJC5 enhances the degradation of p27, while DNAJC5 knockdown reverses the decrease in p27 levels, indicating that the oncogenic function of this protein is p27-mediated [366]. A recent meta-analysis indicated that there was a significant correlation between low p27kip1 expression and aggressive progression, leading to a shorter overall survival in HCC patients [367].

6.5. The Anti-Apoptotic FLIP

The cellular FLICE inhibitory protein (c-FLIP) and the viral FLIP (vFLIP) are important anti-apoptotic proteins against death-receptor-mediated apoptosis and necroptosis [368]. There are three isoforms of c-FLIP: c-FLIPL (long form), c-FLIPS (short form) and c-FLIPR (Raji form). They all share the DED1 and DED2 domains [369].
Apoptosis is inhibited by FLIP through the interruption of the cell death machinery [370]. FLIP binds to procaspase 8, one of the molecules that is involved in apoptosis induction, and stops its maturation, inactivating thus the downstream apoptosis cascade [371]. However, the end result of FLIP implication depends on the level or type of c-FLIP isoforms involved. The c-FLIPL negatively regulates necroptosis, but the c-FLIPS promotes RIP3-mediated necroptosis [372]. The c-FLIP isoforms determine whether cell death follow either through the caspase-dependent apoptosis or through the RIP3-mediated necroptosis. Additionally, c-FLIP redresses autophagy, inhibiting Atg3-binding LC3, which is an essential component for autophagosome formation [373]. Therefore, FLIPs act not only as anti-apoptotic factors, but also as suppressors of autophagy. Moreover, a DED1 peptide or a DED2 peptide of FLIP effectively suppress the Atg3–FLIP interaction without affecting the Atg3–LC3 interaction, resulting in cell death. These FLIP-derived short peptides, therefore, induce growth suppression and cell death by autophagy [373].
FLIP is implicated in the many actions of the HBx protein. The pro-apoptotic function of HBx is mediated through its interaction with c-FLIP variants [374], thus being anti-viral. On the other hand, c-FLIP may be also pro-viral because it stabilizes HBx [375].
Associations between HCV viral proteins and c-FLIP were also described. The HCV core protein maintains the expression of c-FLIP, ultimately blocking TNFα-mediated apoptosis [376], but the opposite results were also reported as HCV core, NS4B and NS5B proteins enhance TNF-induced apoptosis. HCV proteins also reduced the expression of NF-kB-dependent anti-apoptotic proteins, such as Bcl-xL, and c-FLIPL [377]. The hedgehog proteins are implicated in the action of FLIP in HCC. The abnormal activation of the hedgehog pathway is associated with the occurrence of HCC. The protein Gli2 is a terminal transcription factor in this pathway. Gli2 downregulation enhanced TRAIL-induced apoptosis through the reduction in c-FLIP and Bcl-2, indicating the importance of Gli2 in the activation of c-FLIP. On the other hand, the increased expression of c-FLIP alleviated TRAIL-induced apoptosis via the suppression of caspase-8 [378].

6.6. The Role of the ATG12, ATG5 and ATG3 Proteins in Autophagy

ATG12 is an important mediator of the direction of the balance between autophagy and apoptosis. ATG12, in association with ATG3, inhibits the anti-apoptotic Bcl-2 and promotes apoptosis. When ATG12 is associated with ATG5, autophagy is increased. The calpain cleavage of ATG5 switches autophagy to apoptosis [305,344,379,380].

6.7. The Death-Associated Protein Kinase (DAPK) Family in Apoptosis and Autophagy

Death-associated protein kinases (DAPK) are members of a family of five related kinases that mediate several cellular pathways, including apoptosis, autophagy and tumor suppression. The three better-studied family members are DAPK1/DAPK, DAPK2 and DAPK3/ZIPK, which share a high degree of homology but different cellular localization [381]. Initial studies demonstrated that DAPK can induce apoptosis by several pathways, such as p53- and mitochondrion-dependent apoptosis in hepatoma cells [382,383]. However, the effect of DAPK2 in apoptosis is debatable. It seems that the overexpression of DAPK2 causes significant apoptosis but only in cancer cells detached from the extracellular matrix [381,384,385]. DAPK2 was shown to promote the initiation step of autophagy by decreasing mTORC1 activity [386]. DAPK2 is subsequently involved in the additional steps of autophagy. Beclin-1 is a target of DAPK. The DAPK-mediated phosphorylation of Beclin-1 promotes the dissociation of Beclin-1 from its inhibitor BCL-2 to induce autophagy [387,388]. SB203580 is an inhibitor of the p38 mitogen-activated protein kinase (MAPK) but also reduces cell proliferation in a p38/MAPK-independent way. This is achieved through the induction of autophagy in HCC cells associated with the activation of both AMPK and DAPK, which facilitates the phosphorylation of p53 and enhances Beclin-1 expression. The induction of autophagic death may, therefore, account for the antiproliferative effect of SB203580 in HCC cells [389]. Recently, the DEAD-box helicase 20 (DDX20) protein was identified as a downstream target of DAPK that leads to the tumor suppressor function of DAPK in HCC. DAPK1 ameliorated the proteasomal degradation of DDX20. DAPK also suppressed hepatoma cell migration and invasion, but not proliferation [390]. It should be noted that, in other cancers, an opposite effect may be observed. In human placental micro-vascular endothelial cells, DAPK2 overexpression led to a decrease in both autophagy and apoptosis connected to a decrease in Beclin-1 and BAX, along with an increase in Bcl-2 [391]. DAPK1 attenuated oxidative stress and reduced autophagy and inflammation by inhibiting the p38MAPK/NF-kB pathway in a mice model of acute lung injury [392]. In addition to autophagy and apoptosis, the precise role of DAPKs in HCC biology is not known. In a DAPK1 knockout model, hundreds of upregulated genes and downregulated genes were identified. The tissue metalloproteinase inhibitor 1 (TIMP1) and Alpha-2-HS-glycoprotein (AHSG) exhibited the strongest associations with DAPK1 elimination [393].

6.8. p53

The tumor suppressor p53 is encoded by the TP53 gene and is a critical regulator of autophagy and apoptosis in HCC. It is a sensor of cellular stress and responds to a variety of stimulants, such as DNA damage and oxidative stress [394]. It controls apoptosis by inducing the association of components of the extrinsic death receptor system [395] with several various mitochondrial pathways, such as PUMA and BAX, which in turn promote cell death [305,396,397,398,399]. Under stressful conditions, the cytoplasmic p53 translocates to the mitochondrial surface, promoting either the inhibition of the anti-apoptotic Bcl-2 family members or the activation of the pro-apoptotic members leading to the formation of pores in the mitochondrial outer membrane, cytochrome C release and apoptosis [400,401].
In contrast to apoptosis, the upregulation of cytoplasmic p53 or nuclear p53 has different effects in the regulation of autophagy. p53 exerts both pro- and anti-autophagic functions. This is dependent on its subcellular localization. The cytoplasmic p53 inhibits autophagy, acting on the UNC-51-like kinase 1 (ULK1) complex. Under stressful conditions, p53 translocates to the nucleus where it can promote autophagy by inhibiting mTOR through the activation of the AMP kinase [402,403] or the transactivation of the damage-regulated autophagy modulator (DRAM), which promotes the formation of autophagolysosomes [404]. The induction of autophagy via DRAM leads also to apoptotic cell death. Therefore, DRAM is an important element of the mechanism that controls p53-mediated apoptosis and autophagy [380,404]. In addition, nuclear p53 promotes the phosphorylation of Bcl-2. Phosphorylated Bcl-2 does not bind to Beclin-1, allowing the promotion of autophagy [380,405,406].
A different mechanism of the implication of p53 in autophagy and apoptosis regulation has been described. The high mobility group box 1 (HMGB1) and p53 form a complex that controls the balance between autophagy and apoptosis. The loss of p53 increased cytosolic HMGB1 expression and induced autophagy. On the other hand, the loss of HMGB1 increased cytosolic p53 and decreased autophagy. The effects on apoptosis were opposite. Therefore, p53 seems to be a negative regulator of the HMGB1/Beclin-1 complex, up- or downregulating autophagy and apoptosis [407].
The role of Krüppel-associated box (KRAB)-type zinc-finger protein ZNF498 in p53-induced apoptosis was recently reported in HCC. This protein suppressed apoptosis and ferroptosis by decreasing p53 phosphorylation in HCC development [408]. However, convincing evidence that p53 triggers apoptosis is available only for the wild-type. For instance, one study has shown that, in estrogen-positive breast cancer cells, the expression of a truncated p53 mutant increased BCL-2, thus decreasing their apoptosis in breast cancer cells [409]. Moreover, evidence has suggested that certain gain-of-function or loss-of-function mutations of the TP53 gene, as found in many cancers, turn p53 into an oncogene [410]. In this context, it should be considered that TP53 mutations are very common in hepatocellular carcinoma, and their interplay in the regulation of apoptosis and autophagy has not been investigated [411].

6.9. Tumor-Associated Macrophages (TAM) and the Tumor Microenvironment (TEM)

As previously mentioned, HCC, as most other cancers, have inflammation as a basic pathogenetic factor. TAMs play an important role in the maintenance of inflammation by producing several pro-inflammatory cytokines and chemokines [412]. The function of TAMs is regulated by autophagy [413]. Kupffer cells with autophagy deficiency promote liver inflammation and hepatocarcinogenesis via the production of ROS by the mitochondria [414]. TLR2 activation by hepatoma factors results in autophagy augmentation and the M2 immunosuppressive differentiation of TAMs [415]. TLR2-deficient mice had an unexpected increase in HCC induction and progression because TRL2 deficiency resulted in a decrease in macrophage infiltration and suppressed autophagy and apoptosis [416]. The natural compound baicalin shifted the differentiation of TAMs into the M1 anti-tumor phenotype and decreased hepatoma cell proliferation by increasing autophagy [417]. An interesting interplay of autophagy and a form of apoptosis called anoikis has been described. Cancer cell detachment from ECM induces cell death via anoikis. In the interplay between anoikis and autophagy, the ECM-integrin-activated dual tyrosine kinase complex of SRC is involved. SRC was demonstrated to regulate AMPK autophagy. When cells are detached, SRC is inactive, and AMPK is activated to induce protective autophagy against anoikis. When cells are attached again, SRC activation, reduces AMPK activity and downregulates autophagy, allowing cells to proliferate. Whether this mechanism is operative in HCC is not known at present [418].
TAMs are part of the tumor microenvironment that contains other immune cells, such as CD8+ T cells, T regulatory cells, myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), B cells and natural killer (NK) cells. These immune cells are regulated by similar signals and metabolic pathways with HCC cells. Therefore, this overlap makes them prone to similar vulnerabilities, with HCC cells making it difficult to attack only tumor cells without reducing antitumor immunity. Current research has produced conflicting results, but this is a very promising field that may exploit ferroptosis with immunotherapy in HCC treatment [419,420,421].

6.10. The Role of Mitochondria

Autophagy generates metabolic products, such as glutamine, to replenish TCA cycle intermediates that are used to sustain the mitochondrial metabolism of tumor cells, thereby sustaining mitochondrial metabolism in tumor cells [422]. In this context, chloroquine, a small-molecule inhibitor of autophagy, was shown to damage mitochondrial metabolism and diminish tumor growth [423,424,425].
Mitochondrial dysfunction promotes the accumulation of ROS, mtDNA damage and proto-oncogene activation, which are associated with the induction and progression of HCC [426,427]. A reduction in the mitochondrial membrane permeability (MMP) inhibits the apoptosis of HCC cells [428]. MicroRNAs targeting mitochondria showed that miR-518d-5p inhibits c-Jun/PUMA-induced apoptosis and increases sorafenib resistance in HCC [429]. On the contrary, the natural compound dehydrocrenatidine (DEC) reduced ATP production and disrupted the MMP of mitochondria in hepatoma cell lines. DEC induced mitochondrial impairment, increased apoptosis and exerted anti-tumor effects [430]. Interestingly, the inhibition of mitochondrial autophagy (mitophagy) induced the accumulation of damaged mitochondria in HepG2 cells and reduced both the proliferation of HCC cells and the resistance of HCC to sorafenib [431].
The role of mitochondria in tumor biology through the onset, maintenance and counteraction of apoptosis and autophagy has been recently reviewed [432,433].

6.11. Other Factors

There is evidence that Ca2+ regulates both autophagy and apoptosis, but the exact mechanisms are still unknown. An increase in Ca2+ induces autophagy but inhibits apoptosis, resulting in increased cell survival and proliferation. In theory, this is detrimental for HCC [14]. Activated ribosomes are associated with HCC. The RNA-binding protein PNO1 is an important ribosome in tumorigenesis. PNO1 was reported to be overexpressed in HCC, leading to autophagy promotion and apoptosis inhibition through the MAPK signaling pathway [434]. The interplay of autophagy and apoptosis in cancers has been recently reviewed [406].

6.12. Ferroptosis

Ferroptosis is an iron-dependent regulated cell death characterized by iron overload, lipid peroxidation and the overproduction of ROS [435,436]. The word derives from the Greek word “ptosis”, meaning a fall, and the Latin “ferrum”, for iron. Biochemically, ferroptosis is characterized by the consumption of glutathione (GSH) and the decreased activity of GPX4.
There are three main mechanisms regulating ferroptosis:
(1)
The glutathione/glutathione peroxidase 4 (GSH/GPX4) pathway, involving the system Xc−, which imports cystine and exports glutamate. A central role in this system is that of the cystine/glutamate exchanger solute carrier family 7 member 11 (SLC7A11) and the SLC3A2 exchanger [437,438,439].
(2)
Ferritinophagy and other iron metabolism pathways, particularly the p62-Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 regulatory pathways.
(3)
The lipid metabolism pathways, implicating the tumor suppressor p53. p53 promotes the sensitivity to ferroptosis via the suppression of SLC7A11 [435,438].
Experimental evidence showed that ferroptosis is controlled by a variety of external inhibitors and activators [23]. Ferroptosis is initiated by a special form of autophagy called “ferritinophagy”, leading to the degradation of ferritin [440]. Several proteins involved in autophagy are also involved in ferroptosis. The elimination of Atg 5 and Atg7 reduced ferroptosis, induced by the ferroptosis activator erastin. The nuclear receptor coactivator 4 (NCOA4) is the selective carrier of ferritin to ferritinophagy. The genetic inhibition of NCOA4 reduces ferritin degradation and represses ferroptosis, while the overexpression of NCOA4 increases ferritin degradation and ferroptosis.
Ferroptosis is also induced by lipid peroxidation. The overexpression of ACSL4 is responsible for the synthesis of increased levels of polyunsaturated fatty acids (PUFAs), mainly from cell membranes rich in phospholipids, which promote ferroptosis. On the other hand, ACSL3 is responsible for the synthesis of monounsaturated fatty acids (MUFA) that induce ferroptosis resistance. Three mechanisms, namely, the cystatin–GSH–GPX4, the CoQ10–FSP1 and the GCH1–BH4–DHFR axes, all fueled by NADPH, can counteract ferroptosis by inhibiting lipid peroxidation [438].
Inducers of ferroptosis, such as erastin and sorafenib, act by two mechanisms. They inhibit the Xc--mediated cystine antiporter, reducing GSH and GPX4 and leading, therefore, to ROS accumulation, and ferroptosis induction. Another mechanism is related to the p62-Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor E2-related factor 2 (NRF2) pathway. Nrf2 is a transcription factor that protects HCC cells from oxidative damage. p62 inhibits Keap1 and favors Nrf2 accumulation. Nrf2 activates retinoblastoma (Rb) and metallothionein (MT-1G) and induces ferritin heavy chain 1 (FTH1), quinone oxidoreductase 1 (NQO1) and HO-1. The administration of erastin or sorafenib leads to the upregulation of MT-1G and p62 and the downregulation of Rb (Figure 4) [223,435,441,442,443].
Beclin-1 was reported to increase ferroptosis by binding to SLC7A11. The elimination of Beclin-1 inhibits ferroptosis and is initiated by the system Xc- inhibitors, such as erastin, sulfasalazine and sorafenib. On the contrary, the activation of Beclin-1 promotes cancer cell death by ferroptosis, but not by apoptosis or necroptosis [444]. Autophagy can coincide with ferroptosis [445]. Ferroptosis was initially described as a separate type of regulated cell death, distinct from apoptosis and autophagy. However, it is now evident that autophagy, at least under certain conditions, contributes to ferroptotic cell death. Moreover, ferroptosis may share common signals or regulators with apoptosis [23,446,447].

6.13. Ferroptosis and HCC

Liver iron overload and ferroptosis have been conclusively linked to HCC initiation and progress [448,449,450]. As mentioned above, p53 is involved in the regulation of ferroptosis. A single-nucleotide polymorphism at codon 47 of TP53 leads to the disruption of p53 functions and resistance to ferroptosis, probably via the transcriptional regulation of SLC7A11 expression [451]. In general, genes may act as negative regulators of ferroptosis, increasing the resistance of HCC to drugs, such as sorafenib [452]. An increase in the expression of metallothionein-1G (MT-1G), which is a negative regulator of ferroptosis, increases resistance to sorafenib [453]. Ceruloplasmin also inhibits ferroptosis in HCC and increases the deposition of iron and ROS production [454]. In contrast, the synthetase long-chain family member 4 (ACSL4) is an essential mediator of ferroptosis execution and promotes ferroptosis in HCC (Figure 4) [455]. An upregulation of the ACSL4 protein in HCC tissues from responders to sorafenib has been demonstrated [456,457].
HBx causes the upregulation of ACSL4 by targeting miR-205, leading to the accumulation of cholesterol, and the development of HCC [458,459].
ACSL4 promotes the progression of HCC cells. The blocking of hexokinase H2 (HK2) activates ACSL4 effectively and leads to HCC progression [460,461].
Natural omega-3 PUFAs are important substrates in the induction of ferroptosis and the inhibition of tumor progression [462], a fact that can be exploited in HCC [463,464]. LncRNAs are also regulators of ferroptosis in HCC, but their action has not been clarified [465,466]. Recently, signature models using lncRNAs and ferroptosis were established, classifying HCC patients into two groups. The high-risk group had enhanced hepatocarcinogenesis and poor prognosis [467,468]. Equally, non-coding circular RNAs (circRNAs) are associated with the development of HCC through ferroptosis. Circ0097009 endogenous RNA controls the expression of SLC7A11 [469]. Novel ferroptosis-associated genes have been proposed for prognostic use in HCC [470,471]. Despite the all increasing importance of ferroptosis, there are no data on a possible interplay between ferroptosis, autophagy and apoptosis in HCC. The role of ferroptosis in HCC initiation and progression has been extensively reviewed [472,473].

7. Implications of Autophagy, Ferroptosis and Apoptosis in the Drug Treatment of HCC

Most patients with HCC are only candidates for drug treatments by the time they are diagnosed, as the tumor is unresectable or not suitable for loco-regional treatment [474].
Despite the introduction of several new drugs, the outcome is still unsatisfactory because resistance is rapidly developed. Interestingly a commonly used class of drugs may reduce the appearance of HCC. A meta-analysis demonstrated that statins may decrease HCC occurrence. This protection was more evident in HBV patients. Lipophilic statins, such as Atorvastatin, showed a greater effect. This effect was also dose-dependent [475].
The multi-kinase inhibitors sorafenib and lenvatinib are considered as first-line treatment. A combination of atezolizumab and bevacizumab has been recently proposed as a first-line treatment, but results are not impressive and many additional drugs have been tested. A recent meta-analysis suggested that regorafenib and cabozantinib may be the best candidates as second-line treatments in HCC [476].
However, there is extensive evidence that autophagy and ferroptosis are involved in the resistance of HCC to drugs, and their manipulation may improve the efficacy of treatments [477].
Autophagy inhibition may also be used as the treatment of HCC. GNS561, a new autophagy inhibitor, specifically inhibits the enzyme palmitoyl-protein thioesterase 1, (PPT1), leading to lysosomal membrane permeabilization, caspase activation and cell death [478].
Furthermore, the activation of the CD8+ T cells can induce ferroptosis by the suppression of the two components of the Xc- system [479]. RSL3, another ferroptosis inducer, also inhibits the proliferation of HCC cells [480]. Sorafenib resistance has been the most extensively investigated. However, investigations offered conflicting results as autophagy induced either increased resistance or increased efficacy in HCC sorafenib administration [481]. Sorafenib induces the ferroptosis of HCC cells due to the inhibition of the X−C system, followed by glutathione depletion. Ferroptosis inhibitors, such as ferrostatin-1, blocked the cellular death induced by sorafenib [482]. Sorafenib, combined with an aspirin treatment, synergistically induces apoptosis by blocking ACSL4 expression in HCC cells [456]. It was also found that the suppression of MT-1G leads to increased lipid peroxidation and sorafenib-induced ferroptosis in HCC cells [453]. Recent studies have shown the implication of the Yes-associated protein (YAP) in sorafenib resistance. The YAP/TAZ and ATF4 proteins are localized in the cytoplasm and antioxidant genes, such as SLC7A11, are not induced in sorafenib-sensitive cells and ferroptosis is increased. In sorafenib-resistant cells, however, YAP/TAZ and ATF4 are translocated to the nucleus and induce the SLC7A11 gene that represses ferroptosis [483]. Other factors associated with sorafenib resistance are hypercholesterolemia and the overexpression of the cholesterol sensor SCAP [484], and the high expression of the long non-coding RNA SNHG16 in association with low miR-23b-3p expression, leading to increased autophagy and apoptosis inhibition [485]. By contrast, the overexpression of miR-23a-3p directly targets ACSL4, leading to the suppression of ferroptosis and sorafenib resistance [486], while the dysregulation of miR-541 favors autophagy and increases sorafenib resistance [487]. FOXO3 upregulation increased autophagy and sorafenib resistance. Interestingly, the second-line drug regorafenib abolished this protective mechanism [488].
Recently, the variant 1 (tv1) of proliferating cell nuclear antigen clamp-associated factor (PCLAF) was found to reduce ferroptosis in HBV associated by decreasing Fe2+ accumulation [489]. On the other hand, the modulation of autophagy and/or ferroptosis may lead to an increased efficacy of sorafenib. Thus, quiescin sulfhydryl oxidase 1 increases ferroptosis and improves sorafenib efficacy [490].
Cholesterol reduces the degradation of the Golgi membrane protein 1 (GOLM1) and suppresses the GOLM1-dependent autophagy of receptor tyrosine kinases (RTKs), thus promoting HCC metastasis. Statins may, therefore, improve the efficacy of multiple tyrosine kinase inhibitors in HCC treatment [491].
CDGSH iron sulfur domain 2 (CISD2) is an iron-sulfur protein. The inhibition of CISD2 increased sorafenib-induced ferroptosis in resistant cells through either ferritinophagy or the inhibition of the p62–Keap1–NRF2 pathway [492]. The downregulation of complexin II (CPLX2) and haloperidol (a sigma receptor 1 antagonist) promotes the ferroptosis and cell death induced by sorafenib [493,494].
Autophagy inhibition improves sorafenib efficacy [495], while mitophagy induction increases sorafenib and lenvatinib resistance [431,496,497]. On the other hand, the downregulation of COX-2 by ketoconazole leads to mitophagy induction through the PINK–Parkin pathway and apoptosis stimulation [498].
Regorafenib resistance is due to reduction in the drug-induced apoptosis by topoisomerase IIα (TOP2A)-upregulated gene, which is involved in the resistance to regorafenib [499]. Interestingly, many natural products are effective, inhibiting protective autophagy or inducing autophagic death and the apoptosis of HCC cells [256].
Thus, heteronemin, a marine terpenoid, can induce ferroptosis in HCC cells [500].
The inhibition of the PI3K/AKT/mTOR pathway and the induction of autophagy and apoptosis is the mechanism of HCC anti-tumor effect of compounds, such as aloin (in combination with metformin), pueraria flavonoids, apigenin and Shikonin [354,360,501,502].
Solamargine has been shown to induce autophagy and apoptosis and inhibit HCC proliferation [309]. However, it should be noted that the stimulation of both apoptosis and autophagy may be detrimental, as autophagy supports HCC proliferation. Therefore, the combination with an autophagy inhibitor may be necessary as in the case of myricetin, which is a natural flavonoid [503].
This was not the case with sarmentosin, which induced caspase-mediated apoptosis in HCC cells blocked by the autophagy inhibitor chloroquine or the inhibition of Atg7, indicating that autophagy was important for sarmentosin efficacy. Mechanistically, sarmentosin inhibited mTOR and activated Nfr2 [504]. A detailed description of the mechanisms of drug resistance in HCC was recently published [505]. It should be stressed, however, that the above findings are based on experimental evidence and have not been tested in real life clinical trials.

8. Conclusions

Autophagy and apoptosis are two forms of regulated cell death. They are critically implicated in the regulation of HCC biology. Autophagy is interrelated with apoptosis and chemotherapy in HCC. Generally, the induction of autophagy inhibits caspase-dependent apoptosis, and the induction of apoptosis-associated caspase activation blocks the autophagic process. However, autophagy may also induce apoptosis. During HCC induction, autophagy acts as a tumor suppressor, but after induction, it behaves as a tumor promoter. Recently, ferroptosis, a separate form of regulated cell death, was identified. Despite its extensive implication in HCC, its interplay with autophagy and apoptosis, described in other conditions, has not been fully exploited in HCC. There are several switches that control the way in which the balance between autophagy and apoptosis turns. However, the initial cellular sensors that decide the direction of these two pathways have not yet been identified. A better clarification of the mechanisms involved may have clinical implications. The manipulation of either autophagy or apoptosis will improve the treatment outcomes of a difficult-to-treat tumor. There is a need to test, in clinical trials, substances that have been effective in experimental animals.

Author Contributions

Conceptualization and final draft: E.K. Search of the literature and original draft preparation: I.T. and A.V. All authors participated in the design, literature search and review of the manuscript and contributed equally to the discussion of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Villanueva, A. Hepatocellular carcinoma. N. Engl. J. Med. 2019, 380, 1450–1462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kanda, T.; Goto, T.; Hirotsu, Y.; Moriyama, M.; Omata, M. Molecular mechanisms driving progression of liver cirrhosis towards hepatocellular carcinoma in chronic hepatitis B and C infections: A review. Int. J. Mol. Sci. 2019, 20, 1358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kanda, T.; Goto, T.; Hirotsu, Y.; Masuzaki, R.; Moriyama, M.; Omata, M. Molecular mechanisms: Connections between nonalcoholic fatty liver disease, steatohepatitis and hepatocellular carcinoma. Int. J. Mol. Sci. 2020, 21, 1525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. 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]
  6. Younossi, Z.M.; Otgonsuren, M.; Henry, L.; Venkatesan, C.; Mishra, A.; Erario, M.; Hunt, S. Association of nonalcoholic fatty liver disease (NAFLD) with hepatocellular carcinoma (HCC) in the United States from 2004 to 2009. Hepatology 2015, 62, 1723–1730. [Google Scholar] [CrossRef] [PubMed]
  7. Wong, V.W.; Chan, W.K.; Chitturi, S.; Chawla, Y.; Dan, Y.Y.; Duseja, A.; Fan, J.; Goh, K.L.; Hamaguchi, M.; Hashimoto, E.; et al. Asia-Pacific working party on non-alcoholic fatty liver disease guidelines 2017-part 1: Definition, risk factors and assessment. J. Gastroenterol. Hepatol. 2018, 33, 70–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Garuti, F.; Neri, A.; Avanzato, F.; Gramenzi, A.; Rampoldi, D.; Rucci, P.; Farinati, F.; Giannini, E.G.; Piscaglia, F.; Rapaccini, G.L.; et al. The changing scenario of hepatocellular carcinoma in Italy: An update. Liver Int. 2021, 41, 585–597. [Google Scholar] [CrossRef]
  9. Karageorgos, S.A.; Stratakou, S.; Koulentaki, M.; Voumvouraki, A.; Mantaka, A.; Samonakis, D.; Notas, G.; Kouroumalis, E.A. Long-term change in incidence and risk factors of cirrhosis and hepatocellular carcinoma in Crete, Greece: A 25-year study. Ann. Gastroenterol. 2017, 30, 357–363. [Google Scholar] [CrossRef]
  10. Obeng, E. Apoptosis (programmed cell death) and its signals-a review. Braz. J. Biol. 2021, 81, 1133–1143. [Google Scholar] [CrossRef]
  11. Bedoui, S.; Herold, M.J.; Strasser, A. Emerging connectivity of programmed cell death pathways and its physiological implications. Nat. Rev. Mol. Cell Biol. 2020, 21, 678–695. [Google Scholar] [CrossRef]
  12. Wu, J.; Ye, J.; Kong, W.; Zhang, S.; Zheng, Y. Programmed cell death pathways in hearing loss: A review of apoptosis, autophagy and programmed necrosis. Cell Prolif. 2020, 53, 12915. [Google Scholar] [CrossRef] [PubMed]
  13. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef]
  14. Sukumaran, P.; Nascimento Da Conceicao, V.; Sun, Y.; Ahamad, N.; Saraiva, L.R.; Selvaraj, S.; Singh, B.B. Calcium signaling regulates autophagy and apoptosis. Cells 2021, 10, 2125. [Google Scholar] [CrossRef] [PubMed]
  15. Huang, R.; Hui, Z.; Wei, S.; Li, D.; Li, W.; Daping, W.; Alahdal, M. IRE1 signaling regulates chondrocyte apoptosis and death fate in the osteoarthritis. J. Cell. Physiol. 2022, 237, 118–127. [Google Scholar] [CrossRef] [PubMed]
  16. Faruk, M.O.; Ichimura, Y.; Komatsu, M. Selective autophagy. Cancer Sci. 2021, 112, 3972–3978. [Google Scholar] [CrossRef]
  17. Guo, R.; Wang, H.; Cui, N. Autophagy regulation on pyroptosis: Mechanism and medical implication in sepsis. Mediat. Inflamm. 2021, 2021, 9925059. [Google Scholar] [CrossRef]
  18. Patra, S.; Praharaj, P.P.; Klionsky, D.J.; Bhutia, S.K. Vorinostat in autophagic cell death: A critical insight into autophagy-mediated, -associated and -dependent cell death for cancer prevention. Drug Discov. Today 2022, 27, 269–279. [Google Scholar] [CrossRef]
  19. Buratta, S.; Tancini, B.; Sagini, K.; Delo, F.; Chiaradia, E.; Urbanelli, L.; Emiliani, C. Lysosomal exocytosis, exosome release and secretory autophagy: The autophagic-and endo-lysosomal systems go extracellular. Int. J. Mol. Sci. 2020, 21, 2576. [Google Scholar] [CrossRef] [Green Version]
  20. Schulze, R.J.; Krueger, E.W.; Weller, S.G.; Johnson, K.M.; Casey, C.A.; Schott, M.B.; McNiven, M.A. Direct lysosome-based autophagy of lipid droplets in hepatocytes. Proc. Natl. Acad. Sci. USA 2020, 117, 32443–32452. [Google Scholar] [CrossRef]
  21. Djulbegovic, M.B.; Uversky, V.N. Ferroptosis-an iron- and disorder-dependent programmed cell death. Int. J. Biol. Macromol. 2019, 135, 1052–1069. [Google Scholar] [CrossRef]
  22. Zhou, S.Y.; Cui, G.Z.; Yan, X.L.; Wang, X.; Qu, Y.; Guo, Z.N.; Jin, H. Mechanism of ferroptosis and its relationships with other types of programmed cell death: Insights for potential interventions after intracerebral hemorrhage. Front. Neurosci. 2020, 14, 589042. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, J.; Kuang, F.; Kroemer, G.; Klionsky, D.J.; Kang, R.; Tang, D. Autophagy-dependent ferroptosis: Machinery and regulation. Cell Chem. Biol. 2020, 27, 420–435. [Google Scholar] [CrossRef] [PubMed]
  24. Sia, D.; Villanueva, A.; Friedman, S.L.; Llovet, J.M. Liver cancer cell of origin, molecular class, and effects on patient prognosis. Gastroenterology 2017, 152, 745–761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Mu, X.; Español-Suñer, R.; Mederacke, I.; Affò, S.; Manco, R.; Sempoux, C.; Lemaigre, F.P.; Adili, A.; Yuan, D.; Weber, A.; et al. Hepatocellular carcinoma originates from hepatocytes and not from the progenitor/biliary compartment. J. Clin. Investig. 2015, 125, 3891–3903. [Google Scholar] [CrossRef] [Green Version]
  26. Schulze, K.; Nault, J.C.; Villanueva, A. Genetic profiling of hepatocellular carcinoma using next-generation sequencing. J. Hepatol. 2016, 65, 1031–1042. [Google Scholar] [CrossRef] [Green Version]
  27. Schulze, K.; Imbeaud, S.; Letouzé, E.; Alexandrov, L.B.; Calderaro, J.; Rebouissou, S.; Couchy, G.; Meiller, C.; Shinde, J.; Soysouvanh, F.; et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat. Genet. 2015, 47, 505–511. [Google Scholar] [CrossRef] [Green Version]
  28. Llovet, J.M.; Montal, R.; Sia, D.; Finn, R.S. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat. Rev. Clin. Oncol. 2018, 15, 599–616. [Google Scholar] [CrossRef]
  29. Rebouissou, S.; Nault, J.C. Advances in molecular classification and precision oncology in hepatocellular carcinoma. J. Hepatol. 2020, 72, 215–229. [Google Scholar] [CrossRef] [Green Version]
  30. Yang, Y.M.; Kim, S.Y.; Seki, E. Inflammation and liver cancer: Molecular mechanisms and therapeutic targets. Semin. Liver Dis. 2019, 39, 26–42. [Google Scholar] [CrossRef]
  31. Zong, W.X.; Thompson, C.B. Necrotic death as a cell fate. Genes Dev. 2006, 20, 1–15. [Google Scholar] [CrossRef] [Green Version]
  32. Iyer, S.S.; Pulskens, W.P.; Sadler, J.J.; Butter, L.M.; Teske, G.J.; Ulland, T.K.; Eisenbarth, S.C.; Florquin, S.; Flavell, R.A.; Leemans, J.C.; et al. Necrotic cells trigger a sterile inflammatory response through the Nlrp3 inflammasome. Proc. Natl. Acad. Sci. USA 2009, 106, 20388–20393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Hornung, V.; Bauernfeind, F.; Halle, A.; Samstad, E.O.; Kono, H.; Rock, K.L.; Fitzgerald, K.A.; Latz, E. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat. Immunol. 2008, 9, 847–856. [Google Scholar] [CrossRef] [PubMed]
  34. Hurtado-Navarro, L.; Angosto-Bazarra, D.; Pelegrín, P.; Baroja-Mazo, A.; Cuevas, S. NLRP3 inflammasome and pyroptosis in liver pathophysiology: The emerging relevance of Nrf2 inducers. Antioxidants 2022, 11, 870. [Google Scholar] [CrossRef] [PubMed]
  35. Papadakos, S.P.; Dedes, N.; Kouroumalis, E.; Theocharis, S. The role of the NLRP3 inflammasome in HCC carcinogenesis and treatment: Harnessing innate immunity. Cancers 2022, 14, 3150. [Google Scholar] [CrossRef]
  36. Kayagaki, N.; Stowe, I.B.; Lee, B.L.; O’Rourke, K.; Anderson, K.; Warming, S.; Cuellar, T.; Haley, B.; Roose-Girma, M.; Phung, Q.T.; et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 2015, 526, 666–671. [Google Scholar] [CrossRef]
  37. Shi, J.; Zhao, Y.; Wang, K.; Shi, X.; Wang, Y.; Huang, H.; Zhuang, Y.; Cai, T.; Wang, F.; Shao, F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015, 526, 660–665. [Google Scholar] [CrossRef]
  38. García-Pras, E.; Fernández-Iglesias, A.; Gracia-Sancho, J.; Pérez-Del-Pulgar, S. Cell death in hepatocellular carcinoma: Pathogenesis and therapeutic opportunities. Cancers 2021, 14, 48. [Google Scholar] [CrossRef]
  39. Gufler, S.; Seeboeck, R.; Schatz, C.; Haybaeck, J. The translational bridge between inflammation and hepatocarcinogenesis. Cells 2022, 11, 533. [Google Scholar] [CrossRef]
  40. Weber, K.; Schilling, J.D. Lysosomes integrate metabolic-inflammatory cross-talk in primary macrophage inflammasome activation. J. Biol. Chem. 2014, 289, 9158–9171. [Google Scholar] [CrossRef] [Green Version]
  41. Codolo, G.; Plotegher, N.; Pozzobon, T.; Brucale, M.; Tessari, I.; Bubacco, L.; de Bernard, M. Triggering of inflammasome by aggregated α-synuclein, an inflammatory response in synucleinopathies. PLoS ONE 2013, 8, 55375. [Google Scholar] [CrossRef] [Green Version]
  42. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 inflammasome: An overview of mechanisms of activation and regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [Green Version]
  43. Dostert, C.; Guarda, G.; Romero, J.F.; Menu, P.; Gross, O.; Tardivel, A.; Suva, M.L.; Stehle, J.C.; Kopf, M.; Stamenkovic, I.; et al. Malarial hemozoin is a Nalp3 inflammasome activating danger signal. PLoS ONE 2009, 4, 6510. [Google Scholar] [CrossRef] [Green Version]
  44. Orlowski, G.M.; Colbert, J.D.; Sharma, S.; Bogyo, M.; Robertson, S.A.; Rock, K.L. Multiple cathepsins promote pro-IL-1β synthesis and NLRP3-mediated IL-1β activation. J. Immunol. 2015, 195, 1685–1697. [Google Scholar] [CrossRef] [Green Version]
  45. Barlan, A.U.; Griffin, T.M.; McGuire, K.A.; Wiethoff, C.M. Adenovirus membrane penetration activates the NLRP3 inflammasome. J. Virol. 2011, 85, 146–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Walter, P.; Ron, D. The unfolded protein response: From stress pathway to homeostatic regulation. Science 2011, 334, 1081–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Khaled, J.; Kopsida, M.; Lennernäs, H.; Heindryckx, F. Drug resistance and endoplasmic reticulum stress in hepatocellular carcinoma. Cells 2022, 11, 632. [Google Scholar] [CrossRef] [PubMed]
  48. 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] [Green Version]
  49. Pavlović, N.; Heindryckx, F. Exploring the role of endoplasmic reticulum stress in hepatocellular carcinoma through mining of the human protein atlas. Biology 2021, 10, 640. [Google Scholar] [CrossRef]
  50. Wei, J.; Fang, D. Endoplasmic reticulum stress signaling and the pathogenesis of hepatocarcinoma. Int. J. Mol. Sci. 2021, 22, 1799. [Google Scholar] [CrossRef]
  51. Wu, J.; Qiao, S.; Xiang, Y.; Cui, M.; Yao, X.; Lin, R.; Zhang, X. Endoplasmic reticulum stress: Multiple regulatory roles in hepatocellular carcinoma. Biomed. Pharmacother 2021, 142, 112005. [Google Scholar] [CrossRef]
  52. Al-Rawashdeh, F.Y.; Scriven, P.; Cameron, I.C.; Vergani, P.V.; Wyld, L. Unfolded protein response activation contributes to chemoresistance in hepatocellular carcinoma. Eur. J. Gastroenterol. Hepatol. 2010, 22, 1099–1105. [Google Scholar] [CrossRef] [PubMed]
  53. Rahmani, F.; Ziaeemehr, A.; Shahidsales, S.; Gharib, M.; Khazaei, M.; Ferns, G.A.; Ryzhikov, M.; Avan, A.; Hassanian, S.M. Role of regulatory miRNAs of the PI3K/AKT/mTOR signaling in the pathogenesis of hepatocellular carcinoma. J. Cell. Physiol. 2020, 235, 4146–4152. [Google Scholar] [CrossRef] [PubMed]
  54. Cassim, S.; Raymond, V.A.; Lacoste, B.; Lapierre, P.; Bilodeau, M. Metabolite profiling identifies a signature of tumorigenicity in hepatocellular carcinoma. Oncotarget 2018, 9, 26868–26883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Cassim, S.; Raymond, V.A.; Dehbidi-Assadzadeh, L.; Lapierre, P.; Bilodeau, M. Metabolic reprogramming enables hepatocarcinoma cells to efficiently adapt and survive to a nutrient-restricted microenvironment. Cell Cycle 2018, 17, 903–916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Li, J.; Hu, Z.Q.; Yu, S.Y.; Mao, L.; Zhou, Z.J.; Wang, P.C.; Gong, Y.; Su, S.; Zhou, J.; Fan, J.; et al. CircRPN2 inhibits aerobic glycolysis and metastasis in hepatocellular carcinoma. Cancer Res. 2022, 82, 1055–1069. [Google Scholar] [CrossRef]
  57. Feng, J.; Li, J.; Wu, L.; Yu, Q.; Ji, J.; Wu, J.; Dai, W.; Guo, C. Emerging roles and the regulation of aerobic glycolysis in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2020, 39, 126. [Google Scholar] [CrossRef]
  58. Suzuki, T.; Yano, H.; Nakashima, Y.; Nakashima, O.; Kojiro, M. Beta-catenin expression in hepatocellular carcinoma: A possible participation of beta-catenin in the dedifferentiation process. J. Gastroenterol. Hepatol. 2002, 17, 994–1000. [Google Scholar] [CrossRef]
  59. Leung, H.W.; Leung, C.O.N.; Lau, E.Y.; Chung, K.P.S.; Mok, E.H.; Lei, M.M.L.; Leung, R.W.H.; Tong, M.; Keng, V.W.; Ma, C.; et al. EPHB2 activates β-catenin to enhance cancer stem cell properties and drive sorafenib resistance in hepatocellular carcinoma. Cancer Res. 2021, 81, 3229–3240. [Google Scholar] [CrossRef]
  60. Audard, V.; Grimber, G.; Elie, C.; Radenen, B.; Audebourg, A.; Letourneur, F.; Soubrane, O.; Vacher-Lavenu, M.C.; Perret, C.; Cavard, C.; et al. Cholestasis is a marker for hepatocellular carcinomas displaying beta-catenin mutations. J. Pathol. 2007, 212, 345–352. [Google Scholar] [CrossRef] [PubMed]
  61. Xu, C.; Xu, Z.; Zhang, Y.; Evert, M.; Calvisi, D.F.; Chen, X. β-Catenin signaling in hepatocellular carcinoma. J. Clin. Investig. 2022, 132, 154515. [Google Scholar] [CrossRef]
  62. Fan, Z.; Duan, J.; Wang, L.; Xiao, S.; Li, L.; Yan, X.; Yao, W.; Wu, L.; Zhang, S.; Zhang, Y.; et al. PTK2 promotes cancer stem cell traits in hepatocellular carcinoma by activating Wnt/β-catenin signaling. Cancer Lett. 2019, 450, 132–143. [Google Scholar] [CrossRef] [PubMed]
  63. Karabicici, M.; Azbazdar, Y.; Ozhan, G.; Senturk, S.; Firtina Karagonlar, Z.; Erdal, E. Changes in Wnt and TGF-β signaling mediate the development of regorafenib resistance in hepatocellular carcinoma cell line HuH7. Front. Cell Dev. Biol. 2021, 9, 639779. [Google Scholar] [CrossRef] [PubMed]
  64. Tao, J.; Xu, E.; Zhao, Y.; Singh, S.; Li, X.; Couchy, G.; Chen, X.; Zucman-Rossi, J.; Chikina, M.; Monga, S.P. Modeling a human hepatocellular carcinoma subset in mice through coexpression of met and point-mutant β-catenin. Hepatology 2016, 64, 1587–1605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Patil, M.A.; Lee, S.A.; Macias, E.; Lam, E.T.; Xu, C.; Jones, K.D.; Ho, C.; Rodriguez-Puebla, M.; Chen, X. Role of cyclin D1 as a mediator of c-Met- and beta-catenin-induced hepatocarcinogenesis. Cancer Res. 2009, 69, 253–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Qiao, Y.; Wang, J.; Karagoz, E.; Liang, B.; Song, X.; Shang, R.; Evert, K.; Xu, M.; Che, L.; Evert, M.; et al. Axis inhibition protein 1 (Axin1) deletion-induced hepatocarcinogenesis requires intact β-catenin but not Notch cascade in mice. Hepatology 2019, 70, 2003–2017. [Google Scholar] [CrossRef]
  67. Tao, J.; Zhang, R.; Singh, S.; Poddar, M.; Xu, E.; Oertel, M.; Chen, X.; Ganesh, S.; Abrams, M.; Monga, S.P. Targeting β-catenin in hepatocellular cancers induced by coexpression of mutant β-catenin and K-Ras in mice. Hepatology 2017, 65, 1581–1599. [Google Scholar] [CrossRef] [Green Version]
  68. Shang, X.Z.; Zhu, H.; Lin, K.; Tu, Z.; Chen, J.; Nelson, D.R.; Liu, C. Stabilized beta-catenin promotes hepatocyte proliferation and inhibits TNFalpha-induced apoptosis. Lab. Investig. 2004, 84, 332–341. [Google Scholar] [CrossRef] [Green Version]
  69. Tong, Z.; Li, M.; Wang, W.; Mo, P.; Yu, L.; Liu, K.; Ren, W.; Li, W.; Zhang, H.; Xu, J.; et al. Steroid receptor coactivator 1 promotes human hepatocellular carcinoma progression by enhancing Wnt/β-catenin signaling. J. Biol. Chem. 2015, 290, 18596–18608. [Google Scholar] [CrossRef] [Green Version]
  70. Vasuri, F.; Visani, M.; Acquaviva, G.; Brand, T.; Fiorentino, M.; Pession, A.; Tallini, G.; D’Errico, A.; de Biase, D. Role of microRNAs in the main molecular pathways of hepatocellular carcinoma. World J. Gastroenterol. 2018, 24, 2647–2660. [Google Scholar] [CrossRef]
  71. Ruiz-Manriquez, L.M.; Carrasco-Morales, O.; Sanchez, Z.E.A.; Osorio-Perez, S.M.; Estrada-Meza, C.; Pathak, S.; Banerjee, A.; Bandyopadhyay, A.; Duttaroy, A.K.; Paul, S. MicroRNA-mediated regulation of key signaling pathways in hepatocellular carcinoma: A mechanistic insight. Front. Genet. 2022, 13, 910733. [Google Scholar] [CrossRef] [PubMed]
  72. Ahsani, Z.; Mohammadi-Yeganeh, S.; Kia, V.; Karimkhanloo, H.; Zarghami, N.; Paryan, M. WNT1 Gene from WNT signaling pathway is a direct target of miR-122 in hepatocellular carcinoma. Appl. Biochem. Biotechnol. 2017, 181, 884–897. [Google Scholar] [CrossRef] [PubMed]
  73. Xu, X.; Tao, Y.; Shan, L.; Chen, R.; Jiang, H.; Qian, Z.; Cai, F.; Ma, L.; Yu, Y. The role of microRNAs in hepatocellular carcinoma. J. Cancer 2018, 9, 3557–3569. [Google Scholar] [CrossRef] [PubMed]
  74. Du, H.; Xu, Q.; Xiao, S.; Wu, Z.; Gong, J.; Liu, C.; Ren, G.; Wu, H. MicroRNA-424-5p acts as a potential biomarker and inhibits proliferation and invasion in hepatocellular carcinoma by targeting TRIM29. Life Sci. 2019, 224, 1–11. [Google Scholar] [CrossRef] [PubMed]
  75. Khare, S.; Khare, T.; Ramanathan, R.; Ibdah, J.A. Hepatocellular carcinoma: The role of microRNAs. Biomolecules 2022, 12, 645. [Google Scholar] [CrossRef]
  76. Sasaki, R.; Kanda, T.; Yokosuka, O.; Kato, N.; Matsuoka, S.; Moriyama, M. Exosomes and hepatocellular carcinoma: From bench to bedside. Int. J. Mol. Sci. 2019, 20, 1406. [Google Scholar] [CrossRef] [Green Version]
  77. Li, S.; Chen, L. Exosomes in pathogenesis, diagnosis, and treatment of hepatocellular carcinoma. Front. Oncol. 2022, 12, 793432. [Google Scholar] [CrossRef]
  78. Garrett, W.S. Cancer and the microbiota. Science 2015, 348, 80–86. [Google Scholar] [CrossRef] [Green Version]
  79. Kamiya, T.; Ohtani, N. The role of immune cells in the liver tumor microenvironment: An involvement of gut microbiota-derived factors. Int. Immunol. 2022, 34, 467–474. [Google Scholar] [CrossRef]
  80. Liew, W.P.; Mohd-Redzwan, S. Mycotoxin: Its impact on gut health and microbiota. Front. Cell. Infect. Microbiol. 2018, 8, 60. [Google Scholar] [CrossRef] [Green Version]
  81. Iida, N.; Mizukoshi, E.; Yamashita, T.; Yutani, M.; Seishima, J.; Wang, Z.; Arai, K.; Okada, H.; Yamashita, T.; Sakai, Y.; et al. Chronic liver disease enables gut Enterococcus faecalis colonization to promote liver carcinogenesis. Nat. Cancer 2021, 2, 1039–1054. [Google Scholar] [CrossRef] [PubMed]
  82. Zheng, R.; Wang, G.; Pang, Z.; Ran, N.; Gu, Y.; Guan, X.; Yuan, Y.; Zuo, X.; Pan, H.; Zheng, J.; et al. Liver cirrhosis contributes to the disorder of gut microbiota in patients with hepatocellular carcinoma. Cancer Med. 2020, 9, 4232–4250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Luo, W.; Guo, S.; Zhou, Y.; Zhao, J.; Wang, M.; Sang, L.; Chang, B.; Wang, B. Hepatocellular carcinoma: How the gut microbiota contributes to pathogenesis, diagnosis, and therapy. Front. Microbiol. 2022, 13, 873160. [Google Scholar] [CrossRef] [PubMed]
  84. Lai, H.T.; Canoy, R.J.; Campanella, M.; Vassetzky, Y.; Brenner, C. Ca2+ transportome and the interorganelle communication in hepatocellular carcinoma. Cells 2022, 11, 815. [Google Scholar] [CrossRef] [PubMed]
  85. Tümen, D.; Heumann, P.; Gülow, K.; Demirci, C.N.; Cosma, L.S.; Müller, M.; Kandulski, A. Pathogenesis and current treatment strategies of hepatocellular carcinoma. Biomedicines 2022, 10, 3202. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, P.; Markowitz, G.J.; Wang, X.F. The hepatitis B virus-associated tumor microenvironment in hepatocellular carcinoma. Natl. Sci. Rev. 2014, 1, 396–412. [Google Scholar] [CrossRef]
  87. Xie, Y. Hepatitis B virus-associated hepatocellular carcinoma. Adv. Exp. Med. Biol. 2017, 1018, 11–21. [Google Scholar] [CrossRef]
  88. Zanetto, A.; Campello, E.; Bulato, C.; Gavasso, S.; Saggiorato, G.; Shalaby, S.; Spiezia, L.; Cillo, U.; Farinati, F.; Russo, F.P.; et al. More pronounced hypercoagulable state and hypofibrinolysis in patients with cirrhosis with versus without HCC. Hepatol. Commun. 2021, 5, 1987–2000. [Google Scholar] [CrossRef]
  89. Zanetto, A.; Senzolo, M.; Campello, E.; Bulato, C.; Gavasso, S.; Shalaby, S.; Gambato, M.; Vitale, A.; Cillo, U.; Farinati, F.; et al. Influence of hepatocellular carcinoma on platelet aggregation in cirrhosis. Cancers 2021, 13, 1150. [Google Scholar] [CrossRef]
  90. Tu, T.; Budzinska, M.A.; Shackel, N.A.; Urban, S. HBV DNA integration: Molecular mechanisms and clinical implications. Viruses 2017, 9, 75. [Google Scholar] [CrossRef]
  91. Ringelhan, M.; O′Connor, T.; Protzer, U.; Heikenwalder, M. The direct and indirect roles of HBV in liver cancer: Prospective markers for HCC screening and potential therapeutic targets. J. Pathol. 2015, 235, 355–367. [Google Scholar] [CrossRef] [PubMed]
  92. Totoki, Y.; Tatsuno, K.; Covington, K.R.; Ueda, H.; Creighton, C.J.; Kato, M.; Tsuji, S.; Donehower, L.A.; Slagle, B.L.; Nakamura, H.; et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat. Genet. 2014, 46, 1267–1273. [Google Scholar] [CrossRef] [PubMed]
  93. Sung, W.K.; Zheng, H.; Li, S.; Chen, R.; Liu, X.; Li, Y.; Lee, N.P.; Lee, W.H.; Ariyaratne, P.N.; Tennakoon, C.; et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat. Genet. 2012, 44, 765–769. [Google Scholar] [CrossRef] [PubMed]
  94. Tamori, A.; Yamanishi, Y.; Kawashima, S.; Kanehisa, M.; Enomoto, M.; Tanaka, H.; Kubo, S.; Shiomi, S.; Nishiguchi, S. Alteration of gene expression in human hepatocellular carcinoma with integrated hepatitis B virus DNA. Clin. Cancer Res. 2005, 11, 5821–5826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Fujimoto, A.; Totoki, Y.; Abe, T.; Boroevich, K.A.; Hosoda, F.; Nguyen, H.H.; Aoki, M.; Hosono, N.; Kubo, M.; Miya, F.; et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat. Genet. 2012, 44, 760–764. [Google Scholar] [CrossRef]
  96. Wang, M.; Xi, D.; Ning, Q. Virus-induced hepatocellular carcinoma with special emphasis on HBV. Hepatol. Int. 2017, 11, 171–180. [Google Scholar] [CrossRef]
  97. Nakano, M.; Kawaguchi, T.; Nakamoto, S.; Kawaguchi, A.; Kanda, T.; Imazeki, F.; Kuromatsu, R.; Sumie, S.; Satani, M.; Yamada, S.; et al. Effect of occult hepatitis B virus infection on the early-onset of hepatocellular carcinoma in patients with hepatitis C virus infection. Oncol. Rep. 2013, 30, 2049–2055. [Google Scholar] [CrossRef] [Green Version]
  98. Wang, H.C.; Huang, W.; Lai, M.D.; Su, I.J. Hepatitis B virus pre-S mutants, endoplasmic reticulum stress and hepatocarcinogenesis. Cancer Sci. 2006, 97, 683–688. [Google Scholar] [CrossRef]
  99. Wang, H.C.; Wu, H.C.; Chen, C.F.; Fausto, N.; Lei, H.Y.; Su, I.J. Different types of ground glass hepatocytes in chronic hepatitis B virus infection contain specific pre-S mutants that may induce endoplasmic reticulum stress. Am. J. Pathol. 2003, 163, 2441–2449. [Google Scholar] [CrossRef] [Green Version]
  100. D′souza, S.; Lau, K.C.; Coffin, C.S.; Patel, T.R. Molecular mechanisms of viral hepatitis induced hepatocellular carcinoma. World J. Gastroenterol. 2020, 26, 5759–5783. [Google Scholar] [CrossRef]
  101. Kanda, T.; Yokosuka, O.; Imazeki, F.; Yamada, Y.; Imamura, T.; Fukai, K.; Nagao, K.; Saisho, H. Hepatitis B virus X protein (HBx)-induced apoptosis in HuH-7 cells: Influence of HBV genotype and basal core promoter mutations. Scand. J. Gastroenterol. 2004, 39, 478–485. [Google Scholar] [CrossRef]
  102. Koike, K.; Shirakata, Y.; Yaginuma, K.; Arii, M.; Takada, S.; Nakamura, I.; Hayashi, Y.; Kawada, M.; Kobayashi, M. Oncogenic potential of hepatitis B virus. Mol. Biol. Med. 1989, 6, 151–160. [Google Scholar] [PubMed]
  103. Liu, H.; Shi, W.; Luan, F.; Xu, S.; Yang, F.; Sun, W.; Liu, J.; Ma, C. Hepatitis B virus X protein upregulates transcriptional activation of human telomerase reverse transcriptase. Virus Genes 2010, 40, 174–182. [Google Scholar] [CrossRef] [PubMed]
  104. Zou, S.Q.; Qu, Z.L.; Li, Z.F.; Wang, X. Hepatitis B virus X gene induces human telomerase reverse transcriptase mRNA expression in cultured normal human cholangiocytes. World J. Gastroenterol. 2004, 10, 2259–2262. [Google Scholar] [CrossRef] [PubMed]
  105. Kojima, H.; Kaita, K.D.; Xu, Z.; Ou, J.H.; Gong, Y.; Zhang, M.; Minuk, G.Y. The absence of up-regulation of telomerase activity during regeneration after partial hepatectomy in hepatitis B virus X gene transgenic mice. J. Hepatol. 2003, 39, 262–268. [Google Scholar] [CrossRef]
  106. Miller, R.H.; Robinson, W.S. Common evolutionary origin of hepatitis B virus and retroviruses. Proc. Natl. Acad. Sci. USA 1986, 83, 2531–2535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Murakami, S. Hepatitis B virus X protein: A multifunctional viral regulator. J. Gastroenterol. 2001, 36, 651–660. [Google Scholar] [CrossRef]
  108. Torresi, J.; Tran, B.M.; Christiansen, D.; Earnest-Silveira, L.; Schwab, R.H.M.; Vincan, E. HBV-related hepatocarcinogenesis: The role of signalling pathways and innovative ex vivo research models. BMC Cancer 2019, 19, 707. [Google Scholar] [CrossRef] [Green Version]
  109. Yen, C.J.; Lin, Y.J.; Yen, C.S.; Tsai, H.W.; Tsai, T.F.; Chang, K.Y.; Huang, W.C.; Lin, P.W.; Chiang, C.W.; Chang, T.T. Hepatitis B virus X protein upregulates mTOR signaling through IKKβ to increase cell proliferation and VEGF production in hepatocellular carcinoma. PLoS ONE 2012, 7, 41931. [Google Scholar] [CrossRef]
  110. Teng, C.F.; Wu, H.C.; Shyu, W.C.; Jeng, L.B.; Su, I.J. Pre-S2 mutant-induced mammalian target of rapamycin signal pathways as potential therapeutic targets for hepatitis B virus-associated hepatocellular carcinoma. Cell. Transplant. 2017, 26, 429–438. [Google Scholar] [CrossRef] [Green Version]
  111. Wang, X.; Huo, B.; Liu, J.; Huang, X.; Zhang, S.; Feng, T. Hepatitis B virus X reduces hepatocyte apoptosis and promotes cell cycle progression through the Akt/mTOR pathway in vivo. Gene 2019, 691, 87–95. [Google Scholar] [CrossRef] [PubMed]
  112. Huo, T.I.; Wang, X.W.; Forgues, M.; Wu, C.G.; Spillare, E.A.; Giannini, C.; Brechot, C.; Harris, C.C. Hepatitis B virus X mutants derived from human hepatocellular carcinoma retain the ability to abrogate p53-induced apoptosis. Oncogene 2001, 20, 3620–3628. [Google Scholar] [CrossRef] [Green Version]
  113. Wang, X.W.; Gibson, M.K.; Vermeulen, W.; Yeh, H.; Forrester, K.; Stürzbecher, H.W.; Hoeijmakers, J.H.; Harris, C.C. Abrogation of p53-induced apoptosis by the hepatitis B virus X gene. Cancer Res. 1995, 55, 6012–6016. [Google Scholar]
  114. Kim, H.; Lee, H.; Yun, Y. X-gene product of hepatitis B virus induces apoptosis in liver cells. J. Biol. Chem. 1998, 273, 381–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Tian, Y.; Ou, J.H. Genetic and epigenetic alterations in hepatitis B virus-associated hepatocellular carcinoma. Virol. Sin. 2015, 30, 85–91. [Google Scholar] [CrossRef] [Green Version]
  116. Rongrui, L.; Na, H.; Zongfang, L.; Fanpu, J.; Shiwen, J. Epigenetic mechanism involved in the HBV/HCV-related hepatocellular carcinoma tumorigenesis. Curr. Pharm. Des. 2014, 20, 1715–1725. [Google Scholar] [CrossRef]
  117. Zhang, D.; Guo, S.; Schrodi, S.J. Mechanisms of DNA methylation in virus-host interaction in hepatitis B infection: Pathogenesis and oncogenetic properties. Int. J. Mol. Sci. 2021, 22, 9858. [Google Scholar] [CrossRef]
  118. Liu, X.Y.; Tang, S.H.; Wu, S.L.; Luo, Y.H.; Cao, M.R.; Zhou, H.K.; Jiang, X.W.; Shu, J.C.; Bie, C.Q.; Huang, S.M.; et al. Epigenetic modulation of insulin-like growth factor-II overexpression by hepatitis B virus X protein in hepatocellular carcinoma. Am. J. Cancer Res. 2015, 5, 956–978. [Google Scholar] [PubMed]
  119. Sarris, M.E.; Moulos, P.; Haroniti, A.; Giakountis, A.; Talianidis, I. Smyd3 is a transcriptional potentiator of multiple cancer-promoting genes and required for liver and colon cancer development. Cancer Cell 2016, 29, 354–366. [Google Scholar] [CrossRef] [Green Version]
  120. Wang, Y.; Xie, B.H.; Lin, W.H.; Huang, Y.H.; Ni, J.Y.; Hu, J.; Cui, W.; Zhou, J.; Shen, L.; Xu, L.F.; et al. Amplification of SMYD3 promotes tumorigenicity and intrahepatic metastasis of hepatocellular carcinoma via upregulation of CDK2 and MMP2. Oncogene 2019, 38, 4948–4961. [Google Scholar] [CrossRef]
  121. Yang, L.; He, J.; Chen, L.; Wang, G. Hepatitis B virus X protein upregulates expression of SMYD3 and C-MYC in HepG2 cells. Med. Oncol. 2009, 26, 445–451. [Google Scholar] [CrossRef] [PubMed]
  122. Qiu, L.; Wang, T.; Xu, X.; Wu, Y.; Tang, Q.; Chen, K. Long non-coding RNAs in hepatitis B virus-related hepatocellular carcinoma: Regulation, functions, and underlying mechanisms. Int. J. Mol. Sci. 2017, 18, 2505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Cui, S.; Qian, Z.; Chen, Y.; Li, L.; Li, P.; Ding, H. Screening of up- and downregulation of circRNAs in HBV-related hepatocellular carcinoma by microarray. Oncol. Lett. 2018, 15, 423–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wang, S.; Cui, S.; Zhao, W.; Qian, Z.; Liu, H.; Chen, Y.; Lv, F.; Ding, H.G. Screening and bioinformatics analysis of circular RNA expression profiles in hepatitis B-related hepatocellular carcinoma. Cancer Biomark. 2018, 22, 631–640. [Google Scholar] [CrossRef] [PubMed]
  125. Xu, J.; An, P.; Winkler, C.A.; Yu, Y. Dysregulated microRNAs in hepatitis B virus-related hepatocellular carcinoma: Potential as biomarkers and therapeutic targets. Front. Oncol. 2020, 10, 1271. [Google Scholar] [CrossRef]
  126. Zhu, H.T.; Liu, R.B.; Liang, Y.Y.; Hasan, A.M.E.; Wang, H.Y.; Shao, Q.; Zhang, Z.C.; Wang, J.; He, C.Y.; Wang, F.; et al. Serum microRNA profiles as diagnostic biomarkers for HBV-positive hepatocellular carcinoma. Liver Int. 2017, 37, 888–896. [Google Scholar] [CrossRef]
  127. Rana, M.A.; Ijaz, B.; Daud, M.; Tariq, S.; Nadeem, T.; Husnain, T. Interplay of Wnt β-catenin pathway and miRNAs in HBV pathogenesis leading to HCC. Clin. Res. Hepatol. Gastroenterol. 2019, 43, 373–386. [Google Scholar] [CrossRef]
  128. Baskiran, A.; Atay, A.; Baskiran, D.Y.; Akbulut, S. Hepatitis B/D-related hepatocellular carcinoma. A clinical literature review. J. Gastrointest. Cancer 2021, 52, 1192–1197. [Google Scholar] [CrossRef]
  129. Diaz, G.; Engle, R.E.; Tice, A.; Melis, M.; Montenegro, S.; Rodriguez-Canales, J.; Hanson, J.; Emmert-Buck, M.R.; Bock, K.W.; Moore, I.N.; et al. Molecular signature and mechanisms of hepatitis D virus-associated hepatocellular carcinoma. Mol. Cancer Res. 2018, 16, 1406–1419. [Google Scholar] [CrossRef] [Green Version]
  130. Rizzo, G.E.M.; Cabibbo, G.; Craxì, A. Hepatitis B virus-associated hepatocellular carcinoma. Viruses 2022, 14, 986. [Google Scholar] [CrossRef]
  131. Goossens, N.; Hoshida, Y. Hepatitis C virus-induced hepatocellular carcinoma. Clin. Mol. Hepatol. 2015, 21, 105–114. [Google Scholar] [CrossRef] [PubMed]
  132. Chang, S.; Dolganiuc, A.; Szabo, G. Toll-like receptors 1 and 6 are involved in TLR2-mediated macrophage activation by hepatitis C virus core and NS3 proteins. J. Leukoc. Biol. 2007, 82, 479–487. [Google Scholar] [CrossRef] [PubMed]
  133. Hosomura, N.; Kono, H.; Tsuchiya, M.; Ishii, K.; Ogiku, M.; Matsuda, M.; Fujii, H. HCV-related proteins activate Kupffer cells isolated from human liver tissues. Dig. Dis. Sci. 2011, 56, 1057–1064. [Google Scholar] [CrossRef] [PubMed]
  134. Goto, K.; Roca Suarez, A.A.; Wrensch, F.; Baumert, T.F.; Lupberger, J. Hepatitis C virus and hepatocellular carcinoma: When the host loses its grip. Int. J. Mol. Sci. 2020, 21, 3057. [Google Scholar] [CrossRef]
  135. Tian, Z.; Xu, C.; Yang, P.; Lin, Z.; Wu, W.; Zhang, W.; Ding, J.; Ding, R.; Zhang, X.; Dou, K. Molecular pathogenesis: Connections between viral hepatitis-induced and non-alcoholic steatohepatitis-induced hepatocellular carcinoma. Front. Immunol. 2022, 13, 984728. [Google Scholar] [CrossRef]
  136. Sur, S.; Sasaki, R.; Devhare, P.; Steele, R.; Ray, R.; Ray, R.B. Association between microRNA-373 and long noncoding RNA NORAD in hepatitis C virus-infected hepatocytes impairs Wee1 expression for growth promotion. J. Virol. 2018, 92, 01215–01218. [Google Scholar] [CrossRef] [Green Version]
  137. Kanda, T.; Tada, M.; Imazeki, F.; Yokosuka, O.; Nagao, K.; Saisho, H. 5-aza-2′-deoxycytidine sensitizes hepatoma and pancreatic cancer cell lines. Oncol. Rep. 2005, 14, 975–979. [Google Scholar] [CrossRef] [PubMed]
  138. Kanda, T.; Yokosuka, O.; Omata, M. Hepatitis C virus and hepatocellular carcinoma. Biology 2013, 2, 304–316. [Google Scholar] [CrossRef]
  139. Wirth, T.C.; Manns, M.P. The impact of the revolution in hepatitis C treatment on hepatocellular carcinoma. Ann. Oncol. 2016, 27, 1467–1474. [Google Scholar] [CrossRef]
  140. Moriya, K.; Fujie, H.; Shintani, Y.; Yotsuyanagi, H.; Tsutsumi, T.; Ishibashi, K.; Matsuura, Y.; Kimura, S.; Miyamura, T.; Koike, K. The core protein of hepatitis C virus induces hepatocellular carcinoma in transgenic mice. Nat. Med. 1998, 4, 1065–1067. [Google Scholar] [CrossRef]
  141. Kanda, T.; Steele, R.; Ray, R.; Ray, R.B. Hepatitis C virus core protein augments androgen receptor-mediated signaling. J. Virol. 2008, 82, 11066–11072. [Google Scholar] [CrossRef] [Green Version]
  142. Ghosh, A.K.; Majumder, M.; Steele, R.; Meyer, K.; Ray, R.; Ray, R.B. Hepatitis C virus NS5A protein protects against TNF-alpha mediated apoptotic cell death. Virus Res. 2000, 67, 173–178. [Google Scholar] [CrossRef]
  143. Majumder, M.; Ghosh, A.K.; Steele, R.; Ray, R.; Ray, R.B. Hepatitis C virus NS5A physically associates with p53 and regulates p21/waf1 gene expression in a p53-dependent manner. J. Virol. 2001, 75, 1401–1407. [Google Scholar] [CrossRef] [Green Version]
  144. Zhu, Z.; Tran, H.; Mathahs, M.M.; Moninger, T.O.; Schmidt, W.N. HCV induces telomerase reverse transcriptase, increases its catalytic activity, and promotes caspase degradation in infected human hepatocytes. PLoS ONE 2017, 12, 0166853. [Google Scholar] [CrossRef] [Green Version]
  145. Wang, W.; Pan, Q.; Fuhler, G.M.; Smits, R.; Peppelenbosch, M.P. Action and function of Wnt/β-catenin signaling in the progression from chronic hepatitis C to hepatocellular carcinoma. J. Gastroenterol. 2017, 52, 419–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Liu, J.; Ding, X.; Tang, J.; Cao, Y.; Hu, P.; Zhou, F.; Shan, X.; Cai, X.; Chen, Q.; Ling, N.; et al. Enhancement of canonical Wnt/β-catenin signaling activity by HCV core protein promotes cell growth of hepatocellular carcinoma cells. PLoS ONE 2011, 6, 27496. [Google Scholar] [CrossRef]
  147. Park, C.Y.; Choi, S.H.; Kang, S.M.; Kang, J.I.; Ahn, B.Y.; Kim, H.; Jung, G.; Choi, K.Y.; Hwang, S.B. Nonstructural 5A protein activates beta-catenin signaling cascades: Implication of hepatitis C virus-induced liver pathogenesis. J. Hepatol. 2009, 51, 853–864. [Google Scholar] [CrossRef]
  148. Street, A.; Macdonald, A.; Crowder, K.; Harris, M. The hepatitis C virus NS5A protein activates a phosphoinositide 3-kinase-dependent survival signaling cascade. J. Biol. Chem. 2004, 279, 12232–12241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. He, Y.; Nakao, H.; Tan, S.L.; Polyak, S.J.; Neddermann, P.; Vijaysri, S.; Jacobs, B.L.; Katze, M.G. Subversion of cell signaling pathways by hepatitis C virus nonstructural 5A protein via interaction with Grb2 and P85 phosphatidylinositol 3-kinase. J. Virol. 2002, 76, 9207–9217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Wang, J.; Tong, W.; Zhang, X.; Chen, L.; Yi, Z.; Pan, T.; Hu, Y.; Xiang, L.; Yuan, Z. Hepatitis C virus non-structural protein NS5A interacts with FKBP38 and inhibits apoptosis in Huh7 hepatoma cells. FEBS Lett. 2006, 580, 4392–4400. [Google Scholar] [CrossRef] [Green Version]
  151. Higgs, M.R.; Lerat, H.; Pawlotsky, J.M. Hepatitis C virus-induced activation of β-catenin promotes c-Myc expression and a cascade of pro-carcinogenetic events. Oncogene 2013, 32, 4683–4693. [Google Scholar] [CrossRef] [Green Version]
  152. Cotler, S.J.; Hay, N.; Xie, H.; Chen, M.L.; Xu, P.Z.; Layden, T.J.; Guzman, G. Immunohistochemical expression of components of the Akt-mTORC1 pathway is associated with hepatocellular carcinoma in patients with chronic liver disease. Dig. Dis. Sci. 2008, 53, 844–849. [Google Scholar] [CrossRef]
  153. Zhou, L.; Huang, Y.; Li, J.; Wang, Z. The mTOR pathway is associated with the poor prognosis of human hepatocellular carcinoma. Med. Oncol. 2010, 27, 255–261. [Google Scholar] [CrossRef]
  154. Aydin, Y.; Chatterjee, A.; Chandra, P.K.; Chava, S.; Chen, W.; Tandon, A.; Dash, A.; Chedid, M.; Moehlen, M.W.; Regenstein, F.; et al. Interferon-alpha-induced hepatitis C virus clearance restores p53 tumor suppressor more than direct-acting antivirals. Hepatol. Commun. 2017, 1, 256–269. [Google Scholar] [CrossRef] [PubMed]
  155. Dash, S.; Aydin, Y.; Wu, T. Integrated stress response in hepatitis C promotes Nrf2-related chaperone-mediated autophagy: A novel mechanism for host-microbe survival and HCC development in liver cirrhosis. Semin. Cell. Dev. Biol. 2020, 101, 20–35. [Google Scholar] [CrossRef] [PubMed]
  156. Chang, M.L. Metabolic alterations and hepatitis C: From bench to bedside. World J. Gastroenterol. 2016, 22, 1461–1476. [Google Scholar] [CrossRef] [PubMed]
  157. Hofmann, S.; Krajewski, M.; Scherer, C.; Scholz, V.; Mordhorst, V.; Truschow, P.; Schöbel, A.; Reimer, R.; Schwudke, D.; Herker, E. Complex lipid metabolic remodeling is required for efficient hepatitis C virus replication. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 2018, 1863, 1041–1056. [Google Scholar] [CrossRef]
  158. Luedde, T.; Schwabe, R.F. NF-κB in the liver--linking injury, fibrosis and hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 108–118. [Google Scholar] [CrossRef] [Green Version]
  159. Hassan, M.; Selimovic, D.; Ghozlan, H.; Abdel-kader, O. Hepatitis C virus core protein triggers hepatic angiogenesis by a mechanism including multiple pathways. Hepatology 2009, 49, 1469–1482. [Google Scholar] [CrossRef]
  160. Munakata, T.; Liang, Y.; Kim, S.; McGivern, D.R.; Huibregtse, J.; Nomoto, A.; Lemon, S.M. Hepatitis C virus induces E6AP-dependent degradation of the retinoblastoma protein. PLoS Pathog. 2007, 3, 1335–1347. [Google Scholar] [CrossRef] [Green Version]
  161. Luna-Cuadros, M.A.; Chen, H.W.; Hanif, H.; Ali, M.J.; Khan, M.M.; Lau, D.T. Risk of hepatocellular carcinoma after hepatitis C virus cure. World J. Gastroenterol. 2022, 28, 96–107. [Google Scholar] [CrossRef] [PubMed]
  162. Dash, S.; Aydin, Y.; Widmer, K.E.; Nayak, L. Hepatocellular carcinoma mechanisms associated with chronic HCV infection and the impact of direct-acting antiviral treatment. J. Hepatocell. Carcinoma 2020, 7, 45–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Margini, C.; Dufour, J.F. The story of HCC in NAFLD: From epidemiology, across pathogenesis, to prevention and treatment. Liver Int. 2016, 36, 317–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Kutlu, O.; Kaleli, H.N.; Ozer, E. Molecular pathogenesis of nonalcoholic steatohepatitis-(NASH-)related hepatocellular carcinoma. Can. J. Gastroenterol. Hepatol. 2018, 2018, 8543763. [Google Scholar] [CrossRef] [Green Version]
  166. 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]
  167. Singal, A.G.; Manjunath, H.; Yopp, A.C.; Beg, M.S.; Marrero, J.A.; Gopal, P.; Waljee, A.K. The effect of PNPLA3 on fibrosis progression and development of hepatocellular carcinoma: A meta-analysis. Am. J. Gastroenterol. 2014, 109, 325–334. [Google Scholar] [CrossRef] [Green Version]
  168. Sun, H.; Yang, W.; Tian, Y.; Zeng, X.; Zhou, J.; Mok, M.T.S.; Tang, W.; Feng, Y.; Xu, L.; Chan, A.W.H.; et al. An inflammatory-CCRK circuitry drives mTORC1-dependent metabolic and immunosuppressive reprogramming in obesity-associated hepatocellular carcinoma. Nat. Commun. 2018, 9, 5214. [Google Scholar] [CrossRef] [Green Version]
  169. Pei, Y.; Zhang, T.; Renault, V.; Zhang, X. An overview of hepatocellular carcinoma study by omics-based methods. Acta Biochim. Biophys. Sin. 2009, 41, 1–15. [Google Scholar] [CrossRef] [Green Version]
  170. Tian, Y.; Arai, E.; Makiuchi, S.; Tsuda, N.; Kuramoto, J.; Ohara, K.; Takahashi, Y.; Ito, N.; Ojima, H.; Hiraoka, N.; et al. Aberrant DNA methylation results in altered gene expression in non-alcoholic steatohepatitis-related hepatocellular carcinomas. J. Cancer Res. Clin. Oncol. 2020, 146, 2461–2477. [Google Scholar] [CrossRef]
  171. 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] [PubMed] [Green Version]
  172. 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] [Green Version]
  173. 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] [Green Version]
  174. Liu, F.; Li, H.; Chang, H.; Wang, J.; Lu, J. Identification of hepatocellular carcinoma-associated hub genes and pathways by integrated microarray analysis. Tumori J. 2015, 101, 206–214. [Google Scholar] [CrossRef] [PubMed]
  175. Guo, Y.; Xiong, Y.; Sheng, Q.; Zhao, S.; Wattacheril, J.; Flynn, C.R. A micro-RNA expression signature for human NAFLD progression. J. Gastroenterol. 2016, 51, 1022–1030. [Google Scholar] [CrossRef] [Green Version]
  176. Ringelhan, M.; Pfister, D.; O′Connor, T.; Pikarsky, E.; Heikenwalder, M. The immunology of hepatocellular carcinoma. Nat. Immunol. 2018, 19, 222–232. [Google Scholar] [CrossRef]
  177. Taniguchi, K.; Karin, M. NF-κB, inflammation, immunity and cancer: Coming of age. Nat. Rev. Immunol. 2018, 18, 309–324. [Google Scholar] [CrossRef] [PubMed]
  178. Das, M.; Garlick, D.S.; Greiner, D.L.; Davis, R.J. The role of JNK in the development of hepatocellular carcinoma. Genes Dev. 2011, 25, 634–645. [Google Scholar] [CrossRef] [Green Version]
  179. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  180. Wong, V.W.; Wong, G.L.; Choi, P.C.; Chan, A.W.; Li, M.K.; Chan, H.Y.; Chim, A.M.; Yu, J.; Sung, J.J.; Chan, H.L. Disease progression of non-alcoholic fatty liver disease: A prospective study with paired liver biopsies at 3 years. Gut 2010, 59, 969–974. [Google Scholar] [CrossRef]
  181. Pais, R.; Charlotte, F.; Fedchuk, L.; Bedossa, P.; Lebray, P.; Poynard, T.; Ratziu, V. LIDO Study Group. A systematic review of follow-up biopsies reveals disease progression in patients with non-alcoholic fatty liver. J. Hepatol. 2013, 59, 550–556. [Google Scholar] [CrossRef] [PubMed]
  182. Singh, S.; Allen, A.M.; Wang, Z.; Prokop, L.J.; Murad, M.H.; Loomba, R. Fibrosis progression in nonalcoholic fatty liver vs nonalcoholic steatohepatitis: A systematic review and meta-analysis of paired-biopsy studies. Clin. Gastroenterol. Hepatol. 2015, 13, 643–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Chettouh, H.; Lequoy, M.; Fartoux, L.; Vigouroux, C.; Desbois-Mouthon, C. Hyperinsulinaemia and insulin signalling in the pathogenesis and the clinical course of hepatocellular carcinoma. Liver Int. 2015, 35, 2203–2217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Zhang, Y.; Wang, H.; Xiao, H. Metformin actions on the liver: Protection mechanisms emerging in hepatocytes and immune cells against NASH-related HCC. Int. J. Mol. Sci. 2021, 22, 5016. [Google Scholar] [CrossRef]
  185. Anstee, Q.M.; Reeves, H.L.; Kotsiliti, E.; Govaere, O.; Heikenwalder, M. From NASH to HCC: Current concepts and future challenges. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 411–428. [Google Scholar] [CrossRef]
  186. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef] [PubMed]
  187. Nishida, N.; Yada, N.; Hagiwara, S.; Sakurai, T.; Kitano, M.; Kudo, M. Unique features associated with hepatic oxidative DNA damage and DNA methylation in non-alcoholic fatty liver disease. J. Gastroenterol. Hepatol. 2016, 31, 1646–1653. [Google Scholar] [CrossRef]
  188. Gentric, G.; Maillet, V.; Paradis, V.; Couton, D.; L’Hermitte, A.; Panasyuk, G.; Fromenty, B.; Celton-Morizur, S.; Desdouets, C. Oxidative stress promotes pathologic polyploidization in nonalcoholic fatty liver disease. J. Clin. Investig. 2015, 125, 981–992. [Google Scholar] [CrossRef] [Green Version]
  189. Nelson, J.E.; Wilson, L.; Brunt, E.M.; Yeh, M.M.; Kleiner, D.E.; Unalp-Arida, A.; Kowdley, K.V. Nonalcoholic Steatohepatitis Clinical Research Network. Relationship between the pattern of hepatic iron deposition and histological severity in nonalcoholic fatty liver disease. Hepatology 2011, 53, 448–457. [Google Scholar] [CrossRef] [Green Version]
  190. Hamaguchi, K.; Miyanishi, K.; Osuga, T.; Tanaka, S.; Ito, R.; Sakamoto, H.; Kubo, T.; Ohnuma, H.; Murase, K.; Takada, K.; et al. Association between hepatic oxidative stress related factors and activation of Wnt/β-catenin signaling in NAFLD-induced hepatocellular carcinoma. Cancers 2022, 14, 2066. [Google Scholar] [CrossRef]
  191. Koike, K.; Moriya, K. Metabolic aspects of hepatitis C viral infection: Steatohepatitis resembling but distinct from NASH. J. Gastroenterol. 2005, 40, 329–336. [Google Scholar] [CrossRef] [PubMed]
  192. Simon, T.G.; King, L.Y.; Chong, D.Q.; Nguyen, L.H.; Ma, Y.; VoPham, T.; Giovannucci, E.L.; Fuchs, C.S.; Meyerhardt, J.A.; Corey, K.E.; et al. Diabetes, metabolic comorbidities, and risk of hepatocellular carcinoma: Results from two prospective cohort studies. Hepatology 2018, 67, 1797–1806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Kramer, J.R.; Natarajan, Y.; Dai, J.; Yu, X.; Li, L.; El-Serag, H.B.; Kanwal, F. Effect of diabetes medications and glycemic control on risk of hepatocellular cancer in patients with nonalcoholic fatty liver disease. Hepatology 2022, 75, 1420–1428. [Google Scholar] [CrossRef] [PubMed]
  194. Chen, J.; Song, S.; Li, X.; Bian, D.; Wu, X. Association of metabolic traits with occurrence of nonalcoholic fatty liver disease-related hepatocellular carcinoma: A systematic review and meta-analysis of longitudinal cohort studies. Saudi J. Gastroenterol. 2022, 28, 92–100. [Google Scholar] [CrossRef]
  195. Davila, J.A.; Morgan, R.O.; Shaib, Y.; McGlynn, K.A.; El-Serag, H.B. Diabetes increases the risk of hepatocellular carcinoma in the United States: A population based case control study. Gut 2005, 54, 533–539. [Google Scholar] [CrossRef] [Green Version]
  196. El-Serag, H.B.; Hampel, H.; Javadi, F. The association between diabetes and hepatocellular carcinoma: A systematic review of epidemiologic evidence. Clin. Gastroenterol. Hepatol. 2006, 4, 369–380. [Google Scholar] [CrossRef]
  197. Doycheva, I.; Zhang, T.; Amjad, W.; Thuluvath, P.J. Diabetes and hepatocellular carcinoma: Incidence trends and impact of liver disease etiology. J. Clin. Exp. Hepatol. 2020, 10, 296–303. [Google Scholar] [CrossRef]
  198. Tateishi, R.; Matsumura, T.; Okanoue, T.; Shima, T.; Uchino, K.; Fujiwara, N.; Senokuchi, T.; Kon, K.; Sasako, T.; Taniai, M.; et al. Hepatocellular carcinoma development in diabetic patients: A nationwide survey in Japan. J. Gastroenterol. 2021, 56, 261–273. [Google Scholar] [CrossRef]
  199. Vetrano, E.; Rinaldi, L.; Mormone, A.; Giorgione, C.; Galiero, R.; Caturano, A.; Nevola, R.; Marfella, R.; Sasso, F.C. Non-alcoholic fatty liver disease (NAFLD), type 2 diabetes, and non-viral hepatocarcinoma: Pathophysiological mechanisms and new therapeutic strategies. Biomedicines 2023, 11, 468. [Google Scholar] [CrossRef]
  200. Ngo, M.T.; Jeng, H.Y.; Kuo, Y.C.; Diony Nanda, J.; Brahmadhi, A.; Ling, T.Y.; Chang, T.S.; Huang, Y.H. The role of IGF/IGF-1R signaling in hepatocellular carcinomas: Stemness-related properties and drug resistance. Int. J. Mol. Sci. 2021, 22, 1931. [Google Scholar] [CrossRef]
  201. Lai, S.; Quan, Z.; Hao, Y.; Liu, J.; Wang, Z.; Dai, L.; Dai, H.; He, S.; Tang, B. Long non-coding RNA LINC01572 promotes hepatocellular carcinoma progression via sponging miR-195-5p to enhance PFKFB4-mediated glycolysis and PI3K/AKT activation. Front. Cell Dev. Biol. 2021, 9, 783088. [Google Scholar] [CrossRef] [PubMed]
  202. Global Burden of Disease Liver Cancer Collaboration; Akinyemiju, T.; Abera, S.; Ahmed, M.; Alam, N.; Alemayohu, M.A.; Allen, C.; Al-Raddadi, R.; Alvis-Guzman, N.; Amoako, Y.; et al. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: Results from the Global Burden of Disease Study 2015. JAMA Oncol. 2017, 3, 1683–1691. [Google Scholar] [CrossRef]
  203. Goutté, N.; Sogni, P.; Bendersky, N.; Barbare, J.C.; Falissard, B.; Farges, O. Geographical variations in incidence, management and survival of hepatocellular carcinoma in a Western country. J. Hepatol. 2017, 66, 537–544. [Google Scholar] [CrossRef] [Green Version]
  204. Sifaki-Pistolla, D.; Karageorgos, S.A.; Koulentaki, M.; Samonakis, D.; Stratakou, S.; Digenakis, E.; Kouroumalis, E. Geoepidemiology of hepatocellular carcinoma in the island of Crete, Greece. A possible role of pesticides. Liver Int. 2016, 36, 588–594. [Google Scholar] [CrossRef] [PubMed]
  205. Seitz, H.K.; Bataller, R.; Cortez-Pinto, H.; Gao, B.; Gual, A.; Lackner, C.; Mathurin, P.; Mueller, S.; Szabo, G.; Tsukamoto, H. Alcoholic liver disease. Nat. Rev. Dis. Prim. 2018, 4, 16. [Google Scholar] [CrossRef]
  206. Sasaki-Tanaka, R.; Ray, R.; Moriyama, M.; Ray, R.B.; Kanda, T. Molecular changes in relation to alcohol consumption and hepatocellular carcinoma. Int. J. Mol. Sci. 2022, 23, 9679. [Google Scholar] [CrossRef] [PubMed]
  207. Sarsour, E.H.; Kumar, M.G.; Chaudhuri, L.; Kalen, A.L.; Goswami, P.C. Redox control of the cell cycle in health and disease. Antioxid. Redox Signal. 2009, 11, 2985–3011. [Google Scholar] [CrossRef]
  208. Parlesak, A.; Schäfer, C.; Schütz, T.; Bode, J.C.; Bode, C. Increased intestinal permeability to macromolecules and endotoxemia in patients with chronic alcohol abuse in different stages of alcohol-induced liver disease. J. Hepatol. 2000, 32, 742–747. [Google Scholar] [CrossRef]
  209. Wheeler, M.D. Endotoxin and Kupffer cell activation in alcoholic liver disease. Alcohol. Res. Health 2003, 27, 300–306. [Google Scholar]
  210. Fukui, H. Relation of endotoxin, endotoxin binding proteins and macrophages to severe alcoholic liver injury and multiple organ failure. Alcohol. Clin. Exp. Res. 2005, 29, 172–179. [Google Scholar] [CrossRef]
  211. Méndez-Sánchez, N.; Valencia-Rodriguez, A.; Vera-Barajas, A.; Abenavoli, L.; Scarpellini, E.; Ponciano-Rodriguez, G.; Wang, D.Q. The mechanism of dysbiosis in alcoholic liver disease leading to liver cancer. Hepatoma Res. 2020, 6, 5. [Google Scholar] [CrossRef]
  212. Mandrekar, P.; Szabo, G. Signalling pathways in alcohol-induced liver inflammation. J. Hepatol. 2009, 50, 1258–1266. [Google Scholar] [CrossRef] [Green Version]
  213. 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]
  214. Petrasek, J.; Bala, S.; Csak, T.; Lippai, D.; Kodys, K.; Menashy, V.; Barrieau, M.; Min, S.Y.; Kurt-Jones, E.A.; Szabo, G. IL-1 receptor antagonist ameliorates inflammasome-dependent alcoholic steatohepatitis in mice. J. Clin. Investig. 2012, 122, 3476–3489. [Google Scholar] [CrossRef] [Green Version]
  215. Homann, N.; Stickel, F.; König, I.R.; Jacobs, A.; Junghanns, K.; Benesova, M.; Schuppan, D.; Himsel, S.; Zuber-Jerger, I.; Hellerbrand, C.; et al. Alcohol dehydrogenase 1C*1 allele is a genetic marker for alcohol-associated cancer in heavy drinkers. Int. J. Cancer 2006, 118, 1998–2002. [Google Scholar] [CrossRef] [PubMed]
  216. Munaka, M.; Kohshi, K.; Kawamoto, T.; Takasawa, S.; Nagata, N.; Itoh, H.; Oda, S.; Katoh, T. Genetic polymorphisms of tobacco- and alcohol-related metabolizing enzymes and the risk of hepatocellular carcinoma. J. Cancer Res. Clin. Oncol. 2003, 129, 355–360. [Google Scholar] [CrossRef]
  217. Sakamoto, T.; Hara, M.; Higaki, Y.; Ichiba, M.; Horita, M.; Mizuta, T.; Eguchi, Y.; Yasutake, T.; Ozaki, I.; Yamamoto, K.; et al. Influence of alcohol consumption and gene polymorphisms of ADH2 and ALDH2 on hepatocellular carcinoma in a Japanese population. Int. J. Cancer 2006, 118, 1501–1507. [Google Scholar] [CrossRef] [PubMed]
  218. Salameh, H.; Raff, E.; Erwin, A.; Seth, D.; Nischalke, H.D.; Falleti, E.; Burza, M.A.; Leathert, J.; Romeo, S.; Molinaro, A.; et al. PNPLA3 gene polymorphism is associated with predisposition to and severity of alcoholic liver disease. Am. J. Gastroenterol. 2015, 110, 846–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Stickel, F.; Buch, S.; Lau, K.; Meyer zu Schwabedissen, H.; Berg, T.; Ridinger, M.; Rietschel, M.; Schafmayer, C.; Braun, F.; Hinrichsen, H.; et al. Genetic variation in the PNPLA3 gene is associated with alcoholic liver injury in caucasians. Hepatology 2011, 53, 86–95. [Google Scholar] [CrossRef] [PubMed]
  220. Buch, S.; Stickel, F.; Trépo, E.; Way, M.; Herrmann, A.; Nischalke, H.D.; Brosch, M.; Rosendahl, J.; Berg, T.; Ridinger, 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] [PubMed]
  221. Ganne-Carrié, N.; Nahon, P. Hepatocellular carcinoma in the setting of alcohol-related liver disease. J. Hepatol. 2019, 70, 284–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Seitz, H.K.; Stickel, F. Molecular mechanisms of alcohol-mediated carcinogenesis. Nat. Rev. Cancer 2007, 7, 599–612. [Google Scholar] [CrossRef]
  223. Wu, J.; Wang, Y.; Jiang, R.; Xue, R.; Yin, X.; Wu, M.; Meng, Q. Ferroptosis in liver disease: New insights into disease mechanisms. Cell Death Discov. 2021, 7, 276. [Google Scholar] [CrossRef] [PubMed]
  224. Wang, H.; An, P.; Xie, E.; Wu, Q.; Fang, X.; Gao, H.; Zhang, Z.; Li, Y.; Wang, X.; Zhang, J.; et al. Characterization of ferroptosis in murine models of hemochromatosis. Hepatology 2017, 66, 449–465. [Google Scholar] [CrossRef] [Green Version]
  225. Kowdley, K.V. Iron, hemochromatosis, and hepatocellular carcinoma. Gastroenterology 2004, 127, S79–S86. [Google Scholar] [CrossRef]
  226. Elmberg, M.; Hultcrantz, R.; Ekbom, A.; Brandt, L.; Olsson, S.; Olsson, R.; Lindgren, S.; Lööf, L.; Stål, P.; Wallerstedt, S.; et al. Cancer risk in patients with hereditary hemochromatosis and in their first-degree relatives. Gastroenterology 2003, 125, 1733–1741. [Google Scholar] [CrossRef]
  227. Haider, M.B.; Al Sbihi, A.; Chaudhary, A.J.; Haider, S.M.; Edhi, A.I. Hereditary hemochromatosis: Temporal trends, sociodemographic characteristics, and independent risk factor of hepatocellular cancer-nationwide population-based study. World J. Hepatol. 2022, 14, 1804–1816. [Google Scholar] [CrossRef] [PubMed]
  228. D′Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell. Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef]
  229. Adams, J.M. Ways of dying: Multiple pathways to apoptosis. Genes Dev. 2003, 17, 2481–2495. [Google Scholar] [CrossRef] [Green Version]
  230. Kroemer, G.; Galluzzi, L.; Brenner, C. Mitochondrial membrane permeabilization in cell death. Physiol. Rev. 2007, 87, 99–163. [Google Scholar] [CrossRef]
  231. Marquardt, J.U.; Edlich, F. Predisposition to apoptosis in hepatocellular carcinoma: From mechanistic insights to therapeutic strategies. Front. Oncol. 2019, 9, 1421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell. Biol. 2014, 15, 49–63. [Google Scholar] [CrossRef] [PubMed]
  233. Singh, R.; Letai, A.; Sarosiek, K. Regulation of apoptosis in health and disease: The balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell. Biol. 2019, 20, 175–193. [Google Scholar] [CrossRef]
  234. Cosentino, K.; García-Sáez, A.J. Bax and Bak pores: Are we closing the circle? Trends Cell. Biol. 2017, 27, 266–275. [Google Scholar] [CrossRef] [PubMed]
  235. O’Neill, K.L.; Huang, K.; Zhang, J.; Chen, Y.; Luo, X. Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane. Genes Dev. 2016, 30, 973–988. [Google Scholar] [CrossRef] [Green Version]
  236. Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell. Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef]
  237. Wang, Y.; Kanneganti, T.D. From pyroptosis, apoptosis and necroptosis to PANoptosis: A mechanistic compendium of programmed cell death pathways. Comput. Struct. Biotechnol. J. 2021, 19, 4641–4657. [Google Scholar] [CrossRef]
  238. Ketelut-Carneiro, N.; Fitzgerald, K.A. Apoptosis, pyroptosis, and necroptosis-oh my! The many ways a cell can die. J. Mol. Biol. 2022, 434, 167378. [Google Scholar] [CrossRef]
  239. Farazi, P.A.; DePinho, R.A. Hepatocellular carcinoma pathogenesis: From genes to environment. Nat. Rev. Cancer 2006, 6, 674–687. [Google Scholar] [CrossRef]
  240. Moreno-Càceres, J.; Fabregat, I. Apoptosis in liver carcinogenesis and chemotherapy. Hepat. Oncol. 2015, 2, 381–397. [Google Scholar] [CrossRef]
  241. Locatelli, I.; Sutti, S.; Vacchiano, M.; Bozzola, C.; Albano, E. NF-κB1 deficiency stimulates the progression of non-alcoholic steatohepatitis (NASH) in mice by promoting NKT-cell-mediated responses. Clin. Sci. 2013, 124, 279–287. [Google Scholar] [CrossRef]
  242. Grohmann, M.; Wiede, F.; Dodd, G.T.; Gurzov, E.N.; Ooi, G.J.; Butt, T.; Rasmiena, A.A.; Kaur, S.; Gulati, T.; Goh, P.K.; et al. Obesity drives STAT-1-dependent NASH and STAT-3-dependent HCC. Cell 2018, 175, 1289–1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Tang, G.; Minemoto, Y.; Dibling, B.; Purcell, N.H.; Li, Z.; Karin, M.; Lin, A. Inhibition of JNK activation through NF-kappaB target genes. Nature 2001, 414, 313–317. [Google Scholar] [CrossRef]
  244. Maeda, S.; Kamata, H.; Luo, J.L.; Leffert, H.; Karin, M. IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 2005, 121, 977–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Vucur, M.; Reisinger, F.; Gautheron, J.; Janssen, J.; Roderburg, C.; Cardenas, D.V.; Kreggenwinkel, K.; Koppe, C.; Hammerich, L.; Hakem, R.; et al. RIP3 inhibits inflammatory hepatocarcinogenesis but promotes cholestasis by controlling caspase-8- and JNK-dependent compensatory cell proliferation. Cell. Rep. 2013, 4, 776–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Schneider, A.T.; Gautheron, J.; Feoktistova, M.; Roderburg, C.; Loosen, S.H.; Roy, S.; Benz, F.; Schemmer, P.; Büchler, M.W.; Nachbur, U.; et al. RIPK1 suppresses a TRAF2-dependent pathway to liver cancer. Cancer Cell 2017, 31, 94–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Cubero, F.J.; Zhao, G.; Nevzorova, Y.A.; Hatting, M.; Al Masaoudi, M.; Verdier, J.; Peng, J.; Schaefer, F.M.; Hermanns, N.; Boekschoten, M.V.; et al. Haematopoietic cell-derived Jnk1 is crucial for chronic inflammation and carcinogenesis in an experimental model of liver injury. J. Hepatol. 2015, 62, 140–149. [Google Scholar] [CrossRef]
  248. Kanda, T.; Matsuoka, S.; Yamazaki, M.; Shibata, T.; Nirei, K.; Takahashi, H.; Kaneko, T.; Fujisawa, M.; Higuchi, T.; Nakamura, H.; et al. Apoptosis and non-alcoholic fatty liver diseases. World J. Gastroenterol. 2018, 24, 2661–2672. [Google Scholar] [CrossRef]
  249. Lee, Y.J.; Shukla, S.D. Pro- and anti-apoptotic roles of c-Jun N-terminal kinase (JNK) in ethanol and acetaldehyde exposed rat hepatocytes. Eur. J. Pharmacol. 2005, 508, 31–45. [Google Scholar] [CrossRef]
  250. Ohsumi, Y. Historical landmarks of autophagy research. Cell. Res. 2014, 24, 9–23. [Google Scholar] [CrossRef] [Green Version]
  251. Mizushima, N. A brief history of autophagy from cell biology to physiology and disease. Nat. Cell. Biol. 2018, 20, 521–527. [Google Scholar] [CrossRef] [PubMed]
  252. Tooze, S.A.; Yoshimori, T. The origin of the autophagosomal membrane. Nat. Cell. Biol. 2010, 12, 831–835. [Google Scholar] [CrossRef] [PubMed]
  253. Li, Y.; Ding, W.X. Adipose tissue autophagy and homeostasis in alcohol-induced liver injury. Liver Res. 2017, 1, 54–62. [Google Scholar] [CrossRef]
  254. Yu, L.; Chen, Y.; Tooze, S.A. Autophagy pathway: Cellular and molecular mechanisms. Autophagy 2018, 14, 207–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Saha, S.; Panigrahi, D.P.; Patil, S.; Bhutia, S.K. Autophagy in health and disease: A comprehensive review. Biomed. Pharmacother. 2018, 104, 485–495. [Google Scholar] [CrossRef]
  256. Fan, G.; Li, F.; Wang, P.; Jin, X.; Liu, R. Natural-product-mediated autophagy in the treatment of various liver diseases. Int. J. Mol. Sci. 2022, 23, 15109. [Google Scholar] [CrossRef]
  257. Dikic, I.; Elazar, Z. Mechanism and medical implications of mammalian autophagy. Nat. Rev. Mol. Cell. Biol. 2018, 19, 349–364. [Google Scholar] [CrossRef]
  258. Cheng, X.; Ma, X.; Zhu, Q.; Song, D.; Ding, X.; Li, L.; Jiang, X.; Wang, X.; Tian, R.; Su, H.; et al. Pacer is a mediator of mTORC1 and GSK3-TIP60 signaling in regulation of autophagosome maturation and lipid metabolism. Mol Cell. 2019, 73, 788–802. [Google Scholar] [CrossRef] [Green Version]
  259. Ren, H.; Zhao, F.; Zhang, Q.; Huang, X.; Wang, Z. Autophagy and skin wound healing. Burn. Trauma. 2022, 10, tkac003. [Google Scholar] [CrossRef]
  260. Tamargo-Gómez, I.; Mariño, G. AMPK: Regulation of metabolic dynamics in the context of autophagy. Int. J. Mol. Sci. 2018, 19, 3812. [Google Scholar] [CrossRef] [Green Version]
  261. Birgisdottir, Å.B.; Johansen, T. Autophagy and endocytosis-interconnections and interdependencies. J. Cell. Sci. 2020, 133, jcs228114. [Google Scholar] [CrossRef] [PubMed]
  262. Sheng, J.Q.; Wang, M.R.; Fang, D.; Liu, L.; Huang, W.J.; Tian, D.A.; He, X.X.; Li, P.Y. LncRNA NBR2 inhibits tumorigenesis by regulating autophagy in hepatocellular carcinoma. Biomed. Pharmacother. 2021, 133, 111023. [Google Scholar] [CrossRef] [PubMed]
  263. Di Malta, C.; Cinque, L.; Settembre, C. Transcriptional regulation of autophagy: Mechanisms and diseases. Front. Cell Dev. Biol. 2019, 7, 114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Hashemi, M.; Nadafzadeh, N.; Imani, M.H.; Rajabi, R.; Ziaolhagh, S.; Bayanzadeh, S.D.; Norouzi, R.; Rafiei, R.; Koohpar, Z.K.; Raei, B.; et al. Targeting and regulation of autophagy in hepatocellular carcinoma: Revisiting the molecular interactions and mechanisms for new therapy approaches. Cell Commun. Signal. 2023, 21, 32. [Google Scholar] [CrossRef] [PubMed]
  265. Dong, L.; He, J.; Luo, L.; Wang, K. Targeting the interplay of autophagy and ROS for cancer therapy: An updated overview on phytochemicals. Pharmaceuticals 2023, 16, 92. [Google Scholar] [CrossRef] [PubMed]
  266. Onishi, M.; Yamano, K.; Sato, M.; Matsuda, N.; Okamoto, K. Molecular mechanisms and physiological functions of mitophagy. EMBO J. 2021, 40, e104705. [Google Scholar] [CrossRef]
  267. Shibutani, S.T.; Saitoh, T.; Nowag, H.; Münz, C.; Yoshimori, T. Autophagy and autophagy-related proteins in the immune system. Nat. Immunol. 2015, 16, 1014–1024. [Google Scholar] [CrossRef]
  268. de Lavera, I.; Pavon, A.D.; Paz, M.V.; Oropesa-Avila, M.; de la Mata, M.; Alcocer-Gomez, E.; Garrido-Maraver, J.; Cotan, D.; Alvarez-Cordoba, M.; Sanchez-Alcazar, J.A. The connections among autophagy, inflammasome and mitochondria. Curr. Drug Targets 2017, 18, 1030–1038. [Google Scholar] [CrossRef]
  269. Wang, Z.; Zhang, S.; Xiao, Y.; Zhang, W.; Wu, S.; Qin, T.; Yue, Y.; Qian, W.; Li, L. NLRP3 inflammasome and inflammatory diseases. Oxid. Med. Cell. Longev. 2020, 2020, 4063562. [Google Scholar] [CrossRef]
  270. Codogno, P.; Meijer, A.J. Autophagy in the liver. J. Hepatol. 2013, 59, 389–391. [Google Scholar] [CrossRef] [Green Version]
  271. Gual, P.; Gilgenkrantz, H.; Lotersztajn, S. Autophagy in chronic liver diseases: The two faces of Janus. Am. J. Physiol. Cell. Physiol. 2017, 312, 263–273. [Google Scholar] [CrossRef] [Green Version]
  272. Sun, K.; Guo, X.L.; Zhao, Q.D.; Jing, Y.Y.; Kou, X.R.; Xie, X.Q.; Zhou, Y.; Cai, N.; Gao, L.; Zhao, X.; et al. Paradoxical role of autophagy in the dysplastic and tumor-forming stages of hepatocarcinoma development in rats. Cell. Death Dis. 2013, 4, 501. [Google Scholar] [CrossRef] [Green Version]
  273. Yazdani, H.O.; Huang, H.; Tsung, A. Autophagy: Dual response in the development of hepatocellular carcinoma. Cells 2019, 8, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Allaire, M.; Rautou, P.E.; Codogno, P.; Lotersztajn, S. Autophagy in liver diseases: Time for translation? J. Hepatol. 2019, 70, 985–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Wong, M.M.; Chan, H.Y.; Aziz, N.A.; Ramasamy, T.S.; Bong, J.J.; Ch′ng, E.S.; Armon, S.; Peh, S.C.; Teow, S.Y. Interplay of autophagy and cancer stem cells in hepatocellular carcinoma. Mol. Biol. Rep. 2021, 48, 3695–3717. [Google Scholar] [CrossRef] [PubMed]
  276. Tian, Y.; Kuo, C.F.; Sir, D.; Wang, L.; Govindarajan, S.; Petrovic, L.M.; Ou, J.H. Autophagy inhibits oxidative stress and tumor suppressors to exert its dual effect on hepatocarcinogenesis. Cell Death Differ. 2015, 22, 1025–1034. [Google Scholar] [CrossRef] [PubMed]
  277. Ni, H.M.; Chao, X.; Yang, H.; Deng, F.; Wang, S.; Bai, Q.; Qian, H.; Cui, Y.; Cui, W.; Shi, Y.; et al. Dual roles of mammalian target of rapamycin in regulating liver injury and tumorigenesis in autophagy-defective mouse liver. Hepatology 2019, 70, 2142–2155. [Google Scholar] [CrossRef]
  278. Lazova, R.; Camp, R.L.; Klump, V.; Siddiqui, S.F.; Amaravadi, R.K.; Pawelek, J.M. Punctate LC3B expression is a common feature of solid tumors and associated with proliferation, metastasis, and poor outcome. Clin. Cancer Res. 2012, 18, 370–379. [Google Scholar] [CrossRef] [Green Version]
  279. Wu, D.H.; Jia, C.C.; Chen, J.; Lin, Z.X.; Ruan, D.Y.; Li, X.; Lin, Q.; Dong, M.; Ma, X.K.; Wan, X.B.; et al. Autophagic LC3B overexpression correlates with malignant progression and predicts a poor prognosis in hepatocellular carcinoma. Tumour Biol. 2014, 35, 12225–12233. [Google Scholar] [CrossRef]
  280. Chava, S.; Lee, C.; Aydin, Y.; Chandra, P.K.; Dash, A.; Chedid, M.; Thung, S.N.; Moroz, K.; Wu, T.; Nayak, N.C.; et al. Chaperone-mediated autophagy compensates for impaired macroautophagy in the cirrhotic liver to promote hepatocellular carcinoma. Oncotarget 2017, 8, 40019–40036. [Google Scholar] [CrossRef] [Green Version]
  281. Karampa, A.D.; Goussia, A.C.; Glantzounis, G.K.; Mastoridou, E.M.; Anastasopoulos, N.T.; Charchanti, A.V. The role of macroautophagy and chaperone-mediated autophagy in the pathogenesis and management of hepatocellular carcinoma. Cancers 2022, 14, 760. [Google Scholar] [CrossRef]
  282. Turcios, L.; Chacon, E.; Garcia, C.; Eman, P.; Cornea, V.; Jiang, J.; Spear, B.; Liu, C.; Watt, D.S.; Marti, F.; et al. Autophagic flux modulation by Wnt/β-catenin pathway inhibition in hepatocellular carcinoma. PLoS ONE 2019, 14, 0212538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Wang, W.; Xu, L.; Liu, P.; Jairam, K.; Yin, Y.; Chen, K.; Sprengers, D.; Peppelenbosch, M.P.; Pan, Q.; Smits, R. Blocking Wnt secretion reduces growth of hepatocellular carcinoma cell lines mostly independent of β-catenin signaling. Neoplasia 2016, 18, 711–723. [Google Scholar] [CrossRef] [Green Version]
  284. Qian, H.; Yang, Y.; Wang, X. Curcumin enhanced adriamycin-induced human liver-derived Hepatoma G2 cell death through activation of mitochondria-mediated apoptosis and autophagy. Eur. J. Pharm. Sci. 2011, 43, 125–131. [Google Scholar] [CrossRef] [PubMed]
  285. Li, W.; Li, Y.; Siraj, S.; Jin, H.; Fan, Y.; Yang, X.; Huang, X.; Wang, X.; Wang, J.; Liu, L.; et al. FUN14 domain-containing 1-mediated mitophagy suppresses hepatocarcinogenesis by inhibition of inflammasome activation in mice. Hepatology 2019, 69, 604–621. [Google Scholar] [CrossRef]
  286. Huang, Q.; Zhan, L.; Cao, H.; Li, J.; Lyu, Y.; Guo, X.; Zhang, J.; Ji, L.; Ren, T.; An, J.; et al. Increased mitochondrial fission promotes autophagy and hepatocellular carcinoma cell survival through the ROS-modulated coordinated regulation of the NFKB and TP53 pathways. Autophagy 2016, 12, 999–1014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Liu, K.; Lee, J.; Kim, J.Y.; Wang, L.; Tian, Y.; Chan, S.T.; Cho, C.; Machida, K.; Chen, D.; Ou, J.J. Mitophagy controls the activities of tumor suppressor p53 to regulate hepatic cancer stem cells. Mol. Cell 2017, 68, 281–292. [Google Scholar] [CrossRef] [Green Version]
  288. Tang, H.; Da, L.; Mao, Y.; Li, Y.; Li, D.; Xu, Z.; Li, F.; Wang, Y.; Tiollais, P.; Li, T.; et al. Hepatitis B virus X protein sensitizes cells to starvation-induced autophagy via up-regulation of beclin 1 expression. Hepatology 2009, 49, 60–71. [Google Scholar] [CrossRef]
  289. Sir, D.; Tian, Y.; Chen, W.L.; Ann, D.K.; Yen, T.S.; Ou, J.H. The early autophagic pathway is activated by hepatitis B virus and required for viral DNA replication. Proc. Natl. Acad. Sci. USA 2010, 107, 4383–4388. [Google Scholar] [CrossRef] [Green Version]
  290. Liu, B.; Fang, M.; Hu, Y.; Huang, B.; Li, N.; Chang, C.; Huang, R.; Xu, X.; Yang, Z.; Chen, Z.; et al. Hepatitis B virus X protein inhibits autophagic degradation by impairing lysosomal maturation. Autophagy 2014, 10, 416–430. [Google Scholar] [CrossRef]
  291. Lei, Y.; Xu, X.; Liu, H.; Chen, L.; Zhou, H.; Jiang, J.; Yang, Y.; Wu, B. HBx induces hepatocellular carcinogenesis through ARRB1-mediated autophagy to drive the G1/S cycle. Autophagy 2021, 17, 4423–4441. [Google Scholar] [CrossRef] [PubMed]
  292. Dash, S.; Aydin, Y.; Moroz, K. Chaperone-mediated autophagy in the liver: Good or bad? Cells 2019, 8, 1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Onal, G.; Kutlu, O.; Gozuacik, D.; Dokmeci Emre, S. Lipid droplets in health and disease. Lipids Health Dis. 2017, 16, 128. [Google Scholar] [CrossRef] [Green Version]
  294. Wang, X.; Zhang, X.; Chu, E.S.H.; Chen, X.; Kang, W.; Wu, F.; To, K.F.; Wong, V.W.S.; Chan, H.L.Y.; Chan, M.T.V.; et al. Defective lysosomal clearance of autophagosomes and its clinical implications in nonalcoholic steatohepatitis. FASEB J. 2018, 32, 37–51. [Google Scholar] [CrossRef] [Green Version]
  295. Liu, L.; Liao, J.Z.; He, X.X.; Li, P.Y. The role of autophagy in hepatocellular carcinoma: Friend or foe. Oncotarget 2017, 8, 57707–57722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Mao, Y.; Yu, F.; Wang, J.; Guo, C.; Fan, X. Autophagy: A new target for nonalcoholic fatty liver disease therapy. Hepat. Med. 2016, 8, 27–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  297. Wu, W.K.K.; Zhang, L.; Chan, M.T.V. Autophagy, NAFLD and NAFLD-related HCC. Adv. Exp. Med. Biol. 2018, 1061, 127–138. [Google Scholar] [CrossRef]
  298. Khambu, B.; Huda, N.; Chen, X.; Antoine, D.J.; Li, Y.; Dai, G.; Köhler, U.A.; Zong, W.X.; Waguri, S.; Werner, S.; et al. HMGB1 promotes ductular reaction and tumorigenesis in autophagy-deficient livers. J. Clin. Investig. 2018, 128, 2419–2435. [Google Scholar] [CrossRef]
  299. Guo, R.; Xu, X.; Babcock, S.A.; Zhang, Y.; Ren, J. Aldehyde dedydrogenase-2 plays a beneficial role in ameliorating chronic alcohol-induced hepatic steatosis and inflammation through regulation of autophagy. J. Hepatol. 2015, 62, 647–656. [Google Scholar] [CrossRef] [Green Version]
  300. Niture, S.; Gyamfi, M.A.; Lin, M.; Chimeh, U.; Dong, X.; Zheng, W.; Moore, J.; Kumar, D. TNFAIP8 regulates autophagy, cell steatosis, and promotes hepatocellular carcinoma cell proliferation. Cell Death Dis. 2020, 11, 178. [Google Scholar] [CrossRef] [Green Version]
  301. Chen, K.D.; Lin, C.C.; Tsai, M.C.; Huang, K.T.; Chiu, K.W. Tumor microenvironment mediated by suppression of autophagic flux drives liver malignancy. Biomed. J. 2018, 41, 163–168. [Google Scholar] [CrossRef]
  302. Chen, W.; Ma, T.; Shen, X.N.; Xia, X.F.; Xu, G.D.; Bai, X.L.; Liang, T.B. Macrophage-induced tumor angiogenesis is regulated by the TSC2-mTOR pathway. Cancer Res. 2012, 72, 1363–1372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Degenhardt, K.; Mathew, R.; Beaudoin, B.; Bray, K.; Anderson, D.; Chen, G.; Mukherjee, C.; Shi, Y.; Gélinas, C.; Fan, Y.; et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 2006, 10, 51–64. [Google Scholar] [CrossRef] [Green Version]
  304. Booth, L.A.; Roberts, J.L.; Dent, P. The role of cell signaling in the crosstalk between autophagy and apoptosis in the regulation of tumor cell survival in response to sorafenib and neratinib. Semin. Cancer Biol. 2020, 66, 129–139. [Google Scholar] [CrossRef] [PubMed]
  305. Maiuri, M.C.; Zalckvar, E.; Kimchi, A.; Kroemer, G. Self-eating and self-killing: Crosstalk between autophagy and apoptosis. Nat. Rev. Mol. Cell. Biol. 2007, 8, 741–752. [Google Scholar] [CrossRef]
  306. Mariño, G.; Niso-Santano, M.; Baehrecke, E.H.; Kroemer, G. Self-consumption: The interplay of autophagy and apoptosis. Nat. Rev. Mol. Cell. Biol. 2014, 15, 81–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Ravanan, P.; Srikumar, I.F.; Talwar, P. Autophagy: The spotlight for cellular stress responses. Life Sci. 2017, 188, 53–67. [Google Scholar] [CrossRef]
  308. Chen, Z.H.; Lam, H.C.; Jin, Y.; Kim, H.P.; Cao, J.; Lee, S.J.; Ifedigbo, E.; Parameswaran, H.; Ryter, S.W.; Choi, A.M. Autophagy protein microtubule-associated protein 1 light chain-3B (LC3B) activates extrinsic apoptosis during cigarette smoke-induced emphysema. Proc. Natl. Acad. Sci. USA 2010, 107, 18880–18885. [Google Scholar] [CrossRef] [Green Version]
  309. Yin, S.; Jin, W.; Qiu, Y.; Fu, L.; Wang, T.; Yu, H. Solamargine induces hepatocellular carcinoma cell apoptosis and autophagy via inhibiting LIF/miR-192-5p/CYR61/Akt signaling pathways and eliciting immunostimulatory tumor microenvironment. J. Hematol. Oncol. 2022, 15, 32. [Google Scholar] [CrossRef]
  310. Guo, L.; Liang, Y.; Wang, S.; Li, L.; Cai, L.; Heng, Y.; Yang, J.; Jin, X.; Zhang, J.; Yuan, S.; et al. Jujuboside B inhibits the proliferation of breast cancer cell lines by inducing apoptosis and autophagy. Front. Pharmacol. 2021, 12, 668887. [Google Scholar] [CrossRef] [PubMed]
  311. Zheng, Y.; Xu, C.L.; Lu, N.Y.; Qiu, F.F.; Zhao, Y.J.; Chang, Y.X.; Wang, J.H.; Zhao, T.J.; Yuan, X.L. Study on mechanism of curcumol against liver fibrosis based on autophagy and apoptosis of hepatic stellate cells. Zhongguo Zhong Yao Za Zhi 2022, 47, 730–736. [Google Scholar] [CrossRef] [PubMed]
  312. Liang, S.; Liu, H.; Liu, S.; Wei, M.; Gao, F.; Xue, J.; Sun, L.; Wang, M.; Jiang, H.; Chen, L. Homocysteine induces human mesangial cell apoptosis via the involvement of autophagy and endoplasmic reticulum stress. RSC Adv. 2019, 9, 31720–31727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  313. Ding, W.X.; Ni, H.M.; Gao, W.; Hou, Y.F.; Melan, M.A.; Chen, X.; Stolz, D.B.; Shao, Z.M.; Yin, X.M. Differential effects of endoplasmic reticulum stress-induced autophagy on cell survival. J. Biol. Chem. 2007, 282, 4702–4710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Zeng, L.; Zou, Q.; Huang, P.; Xiong, L.; Cheng, Y.; Chen, Q.; Li, Y.; He, H.; Yi, W.; Wei, W. Inhibition of autophagy with chloroquine enhanced apoptosis induced by 5-aminolevulinic acid-photodynamic therapy in secondary hyperparathyroidism primary cells and organoids. Biomed. Pharmacother. 2021, 142, 111994. [Google Scholar] [CrossRef] [PubMed]
  315. Wang, K. Autophagy and apoptosis in liver injury. Cell Cycle 2015, 14, 1631–1642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  316. Kouroumalis, E.; Voumvouraki, A.; Augoustaki, A.; Samonakis, D.N. Autophagy in liver diseases. World J. Hepatol. 2021, 13, 6–65. [Google Scholar] [CrossRef]
  317. Gómez de Cedrón, M.; Ramírez de Molina, A. Microtargeting cancer metabolism: Opening new therapeutic windows based on lipid metabolism. J. Lipid Res. 2016, 57, 193–206. [Google Scholar] [CrossRef] [Green Version]
  318. Zhao, T.; Du, H.; Ding, X.; Walls, K.; Yan, C. Activation of mTOR pathway in myeloid-derived suppressor cells stimulates cancer cell proliferation and metastasis in lal(-/-) mice. Oncogene 2015, 34, 1938–1948. [Google Scholar] [CrossRef] [Green Version]
  319. Mukhopadhyay, S.; Schlaepfer, I.R.; Bergman, B.C.; Panda, P.K.; Praharaj, P.P.; Naik, P.P.; Agarwal, R.; Bhutia, S.K. ATG14 facilitated lipophagy in cancer cells induce ER stress mediated mitoptosis through a ROS dependent pathway. Free. Radic. Biol. Med. 2017, 104, 199–213. [Google Scholar] [CrossRef]
  320. Tu, Q.Q.; Zheng, R.Y.; Li, J.; Hu, L.; Chang, Y.X.; Li, L.; Li, M.H.; Wang, R.Y.; Huang, D.D.; Wu, M.C.; et al. Palmitic acid induces autophagy in hepatocytes via JNK2 activation. Acta Pharmacol. Sin. 2014, 35, 504–512. [Google Scholar] [CrossRef] [Green Version]
  321. Cai, N.; Zhao, X.; Jing, Y.; Sun, K.; Jiao, S.; Chen, X.; Yang, H.; Zhou, Y.; Wei, L. Autophagy protects against palmitate-induced apoptosis in hepatocytes. Cell Biosci. 2014, 4, 28. [Google Scholar] [CrossRef] [Green Version]
  322. Lou, J.; Wang, Y.; Wang, X.; Jiang, Y. Uncoupling protein 2 regulates palmitic acid-induced hepatoma cell autophagy. Biomed. Res. Int. 2014, 2014, 810401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  323. Xie, F.; Jia, L.; Lin, M.; Shi, Y.; Yin, J.; Liu, Y.; Chen, D.; Meng, Q. ASPP2 attenuates triglycerides to protect against hepatocyte injury by reducing autophagy in a cell and mouse model of non-alcoholic fatty liver disease. J. Cell. Mol. Med. 2015, 19, 155–164. [Google Scholar] [CrossRef] [PubMed]
  324. Tanaka, S.; Hikita, H.; Tatsumi, T.; Sakamori, R.; Nozaki, Y.; Sakane, S.; Shiode, Y.; Nakabori, T.; Saito, Y.; Hiramatsu, N.; et al. Rubicon inhibits autophagy and accelerates hepatocyte apoptosis and lipid accumulation in nonalcoholic fatty liver disease in mice. Hepatology 2016, 64, 1994–2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Eid, N.; Ito, Y.; Otsuki, Y. Triggering of Parkin mitochondrial translocation in mitophagy: Implications for liver diseases. Front. Pharmacol. 2016, 7, 100. [Google Scholar] [CrossRef] [Green Version]
  326. Li, S.; Dou, X.; Ning, H.; Song, Q.; Wei, W.; Zhang, X.; Shen, C.; Li, J.; Sun, C.; Song, Z. Sirtuin 3 acts as a negative regulator of autophagy dictating hepatocyte susceptibility to lipotoxicity. Hepatology 2017, 66, 936–952. [Google Scholar] [CrossRef] [Green Version]
  327. Nikoletopoulou, V.; Markaki, M.; Palikaras, K.; Tavernarakis, N. Crosstalk between apoptosis, necrosis and autophagy. Biochim. Biophys. Acta 2013, 1833, 3448–3459. [Google Scholar] [CrossRef] [Green Version]
  328. Xie, Z.; Klionsky, D.J. Autophagosome formation: Core machinery and adaptations. Nat. Cell. Biol. 2007, 9, 1102–1109. [Google Scholar] [CrossRef]
  329. Zeng, X.; Overmeyer, J.H.; Maltese, W.A. Functional specificity of the mammalian Beclin-Vps34 PI 3-kinase complex in macroautophagy versus endocytosis and lysosomal enzyme trafficking. J. Cell. Sci. 2006, 119, 259–270. [Google Scholar] [CrossRef] [Green Version]
  330. Pattingre, S.; Tassa, A.; Qu, X.; Garuti, R.; Liang, X.H.; Mizushima, N.; Packer, M.; Schneider, M.D.; Levine, B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 2005, 122, 927–939. [Google Scholar] [CrossRef] [Green Version]
  331. Takacs-Vellai, K.; Vellai, T.; Puoti, A.; Passannante, M.; Wicky, C.; Streit, A.; Kovacs, A.L.; Müller, F. Inactivation of the autophagy gene bec-1 triggers apoptotic cell death in C. elegans. Curr. Biol. 2005, 15, 1513–1517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  332. Maiuri, M.C.; Le Toumelin, G.; Criollo, A.; Rain, J.C.; Gautier, F.; Juin, P.; Tasdemir, E.; Pierron, G.; Troulinaki, K.; Tavernarakis, N.; et al. Functional and physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J. 2007, 26, 2527–2539. [Google Scholar] [CrossRef] [PubMed]
  333. Galonek, H.L.; Hardwick, J.M. Upgrading the BCL-2 network. Nat. Cell Biol. 2006, 8, 1317–1319. [Google Scholar] [CrossRef] [PubMed]
  334. Ding, Z.B.; Shi, Y.H.; Zhou, J.; Qiu, S.J.; Xu, Y.; Dai, Z.; Shi, G.M.; Wang, X.Y.; Ke, A.W.; Wu, B.; et al. Association of autophagy defect with a malignant phenotype and poor prognosis of hepatocellular carcinoma. Cancer Res. 2008, 68, 9167–9175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Qiu, D.M.; Wang, G.L.; Chen, L.; Xu, Y.Y.; He, S.; Cao, X.L.; Qin, J.; Zhou, J.M.; Zhang, Y.X.; Qun, E. The expression of beclin-1, an autophagic gene, in hepatocellular carcinoma associated with clinical pathological and prognostic significance. BMC Cancer 2014, 14, 327. [Google Scholar] [CrossRef] [Green Version]
  336. Al-Shenawy, H.A. Expression of Beclin-1, an autophagy-related marker, in chronic hepatitis and hepatocellular carcinoma and its relation with apoptotic markers. APMIS 2016, 124, 229–237. [Google Scholar] [CrossRef]
  337. Zhang, X.; Jin, L.; Tian, Z.; Wang, J.; Yang, Y.; Liu, J.; Chen, Y.; Hu, C.; Chen, T.; Zhao, Y.; et al. Nitric oxide inhibits autophagy and promotes apoptosis in hepatocellular carcinoma. Cancer Sci. 2019, 110, 1054–1063. [Google Scholar] [CrossRef] [Green Version]
  338. Tai, W.T.; Shiau, C.W.; Chen, H.L.; Liu, C.Y.; Lin, C.S.; Cheng, A.L.; Chen, P.J.; Chen, K.F. Mcl-1-dependent activation of Beclin 1 mediates autophagic cell death induced by sorafenib and SC-59 in hepatocellular carcinoma cells. Cell. Death. Dis. 2013, 4, 485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  339. Shi, Y.H.; Ding, Z.B.; Zhou, J.; Hui, B.; Shi, G.M.; Ke, A.W.; Wang, X.Y.; Dai, Z.; Peng, Y.F.; Gu, C.Y.; et al. Targeting autophagy enhances sorafenib lethality for hepatocellular carcinoma via ER stress-related apoptosis. Autophagy 2011, 7, 1159–1172. [Google Scholar] [CrossRef]
  340. Shimizu, S.; Takehara, T.; Hikita, H.; Kodama, T.; Tsunematsu, H.; Miyagi, T.; Hosui, A.; Ishida, H.; Tatsumi, T.; Kanto, T.; et al. Inhibition of autophagy potentiates the antitumor effect of the multikinase inhibitor sorafenib in hepatocellular carcinoma. Int. J. Cancer 2012, 131, 548–557. [Google Scholar] [CrossRef]
  341. Chen, S.; Du, Y.; Xu, B.; Li, Q.; Yang, L.; Jiang, Z.; Zeng, Z.; Chen, L. Vaccinia-related kinase 2 blunts sorafenib’s efficacy against hepatocellular carcinoma by disturbing the apoptosis-autophagy balance. Oncogene 2021, 40, 3378–3393. [Google Scholar] [CrossRef] [PubMed]
  342. Wirawan, E.; Vande Walle, L.; Kersse, K.; Cornelis, S.; Claerhout, S.; Vanoverberghe, I.; Roelandt, R.; De Rycke, R.; Verspurten, J.; Declercq, W.; et al. Caspase-mediated cleavage of Beclin-1 inactivates Beclin-1-induced autophagy and enhances apoptosis by promoting the release of proapoptotic factors from mitochondria. Cell. Death Dis. 2010, 1, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  343. Luo, S.; Rubinsztein, D.C. Apoptosis blocks Beclin 1-dependent autophagosome synthesis: An effect rescued by Bcl-xL. Cell Death Differ. 2010, 17, 268–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. Yousefi, S.; Perozzo, R.; Schmid, I.; Ziemiecki, A.; Schaffner, T.; Scapozza, L.; Brunner, T.; Simon, H.U. Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nat. Cell Biol. 2006, 8, 1124–1132. [Google Scholar] [CrossRef]
  345. Bursch, W.; Karwan, A.; Mayer, M.; Dornetshuber, J.; Fröhwein, U.; Schulte-Hermann, R.; Fazi, B.; Di Sano, F.; Piredda, L.; Piacentini, M.; et al. Cell death and autophagy: Cytokines, drugs, and nutritional factors. Toxicology 2008, 254, 147–157. [Google Scholar] [CrossRef] [Green Version]
  346. Zou, Z.; Tao, T.; Li, H.; Zhu, X. mTOR signaling pathway and mTOR inhibitors in cancer: Progress and challenges. Cell Biosci. 2020, 10, 31. [Google Scholar] [CrossRef] [Green Version]
  347. Nie, T.; Yang, S.; Ma, H.; Zhang, L.; Lu, F.; Tao, K.; Wang, R.; Yang, R.; Huang, L.; Mao, Z.; et al. Regulation of ER stress-induced autophagy by GSK3β-TIP60-ULK1 pathway. Cell Death Dis. 2016, 7, 2563. [Google Scholar] [CrossRef] [Green Version]
  348. Ryu, H.Y.; Kim, L.E.; Jeong, H.; Yeo, B.K.; Lee, J.W.; Nam, H.; Ha, S.; An, H.K.; Park, H.; Jung, S.; et al. GSK3B induces autophagy by phosphorylating ULK1. Exp. Mol. Med. 2021, 53, 369–383. [Google Scholar] [CrossRef]
  349. Castedo, M.; Ferri, K.F.; Kroemer, G. Mammalian target of rapamycin (mTOR): Pro- and anti-apoptotic. Cell. Death Differ. 2002, 9, 99–100. [Google Scholar] [CrossRef]
  350. Germain, M.; Nguyen, A.P.; Le Grand, J.N.; Arbour, N.; Vanderluit, J.L.; Park, D.S.; Opferman, J.T.; Slack, R.S. MCL-1 is a stress sensor that regulates autophagy in a developmentally regulated manner. EMBO J. 2011, 30, 395–407. [Google Scholar] [CrossRef] [Green Version]
  351. Xu, G.; Ma, T.; Zhou, C.; Zhao, F.; Peng, K.; Li, B. β-Carotene attenuates apoptosis and autophagy via PI3K/AKT/mTOR signaling pathway in necrotizing enterocolitis model cells IEC-6. Evid. Based Complement Alternat. Med. 2022, 2022, 2502263. [Google Scholar] [CrossRef] [PubMed]
  352. Porta, C.; Paglino, C.; Mosca, A. Targeting PI3K/Akt/mTOR signaling in cancer. Front. Oncol. 2014, 4, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  353. Matter, M.S.; Decaens, T.; Andersen, J.B.; Thorgeirsson, S.S. Targeting the mTOR pathway in hepatocellular carcinoma: Current state and future trends. J. Hepatol. 2014, 60, 855–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  354. Yang, J.; Pi, C.; Wang, G. Inhibition of PI3K/Akt/mTOR pathway by apigenin induces apoptosis and autophagy in hepatocellular carcinoma cells. Biomed. Pharmacother. 2018, 103, 699–707. [Google Scholar] [CrossRef] [PubMed]
  355. Ye, R.; Dai, N.; He, Q.; Guo, P.; Xiang, Y.; Zhang, Q.; Hong, Z.; Zhang, Q. Comprehensive anti-tumor effect of Brusatol through inhibition of cell viability and promotion of apoptosis caused by autophagy via the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Biomed. Pharmacother. 2018, 105, 962–973. [Google Scholar] [CrossRef]
  356. Wu, Y.; Zhang, Y.; Qin, X.; Geng, H.; Zuo, D.; Zhao, Q. PI3K/AKT/mTOR pathway-related long non-coding RNAs: Roles and mechanisms in hepatocellular carcinoma. Pharmacol. Res. 2020, 160, 105195. [Google Scholar] [CrossRef]
  357. Guo, M.; Li, N.; Zheng, J.; Wang, W.; Wu, Y.; Han, X.; Guo, J.; Chen, W.; Bai, Z.; Bai, W.; et al. Epigenetic regulation of hepatocellular carcinoma progression through the mTOR signaling pathway. Can. J. Gastroenterol. Hepatol. 2021, 2021, 5596712. [Google Scholar] [CrossRef]
  358. Wei, L.; Wang, X.; Lv, L.; Liu, J.; Xing, H.; Song, Y.; Xie, M.; Lei, T.; Zhang, N.; Yang, M. The emerging role of microRNAs and long noncoding RNAs in drug resistance of hepatocellular carcinoma. Mol. Cancer 2019, 18, 147. [Google Scholar] [CrossRef] [PubMed]
  359. Hong, F.; Gao, Y.; Li, Y.; Zheng, L.; Xu, F.; Li, X. Inhibition of HIF1A-AS1 promoted starvation-induced hepatocellular carcinoma cell apoptosis by reducing HIF-1α/mTOR-mediated autophagy. World J. Surg. Oncol. 2020, 18, 113. [Google Scholar] [CrossRef]
  360. Sun, R.; Zhai, R.; Ma, C.; Miao, W. Combination of aloin and metformin enhances the antitumor effect by inhibiting the growth and invasion and inducing apoptosis and autophagy in hepatocellular carcinoma through PI3K/AKT/mTOR pathway. Cancer Med. 2020, 9, 1141–1151. [Google Scholar] [CrossRef] [Green Version]
  361. Ferrín, G.; Guerrero, M.; Amado, V.; Rodríguez-Perálvarez, M.; De la Mata, M. Activation of mTOR signaling pathway in hepatocellular carcinoma. Int. J. Mol. Sci. 2020, 21, 1266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  362. Besson, A.; Dowdy, S.F.; Roberts, J.M. CDK inhibitors: Cell cycle regulators and beyond. Dev. Cell 2008, 14, 159–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  363. McKay, L.K.; White, J.P. The AMPK/p27Kip1 pathway as a novel target to promote autophagy and resilience in aged cells. Cells 2021, 10, 1430. [Google Scholar] [CrossRef] [PubMed]
  364. Liang, J.; Shao, S.H.; Xu, Z.X.; Hennessy, B.; Ding, Z.; Larrea, M.; Kondo, S.; Dumont, D.J.; Gutterman, J.U.; Walker, C.L.; et al. The energy sensing LKB1-AMPK pathway regulates p27(kip1) phosphorylation mediating the decision to enter autophagy or apoptosis. Nat. Cell. Biol. 2007, 9, 218–224. [Google Scholar] [CrossRef]
  365. White, J.P.; Billin, A.N.; Campbell, M.E.; Russell, A.J.; Huffman, K.M.; Kraus, W.E. The AMPK/p27Kip1 axis regulates autophagy/apoptosis decisions in aged skeletal muscle stem cells. Stem Cell Rep. 2018, 11, 425–439. [Google Scholar] [CrossRef] [Green Version]
  366. Wang, H.; Luo, J.; Tian, X.; Xu, L.; Zhai, Z.; Cheng, M.; Chen, L.; Luo, S. DNAJC5 promotes hepatocellular carcinoma cells proliferation though regulating SKP2 mediated p27 degradation. Biochim. Biophys. Acta Mol. Cell. Res. 2021, 1868, 118994. [Google Scholar] [CrossRef]
  367. Luo, Y.; Fu, Z.; Wu, P.; Zheng, D.; Zhang, X. The clinicopathological and prognostic significance of P27kip in hepatocellular carcinoma patients: A systemic review and meta-analysis. Gene 2020, 734, 144351. [Google Scholar] [CrossRef]
  368. Krueger, A.; Baumann, S.; Krammer, P.H.; Kirchhoff, S. FLICE-inhibitory proteins: Regulators of death receptor-mediated apoptosis. Mol. Cell. Biol. 2001, 21, 8247–8254. [Google Scholar] [CrossRef] [Green Version]
  369. Lee, A.R.; Park, Y.K.; Dezhbord, M.; Kim, K.H. Interaction between the hepatitis B virus and cellular FLIP variants in viral replication and the innate immune system. Viruses 2022, 14, 373. [Google Scholar] [CrossRef] [PubMed]
  370. Nakagiri, S.; Murakami, A.; Takada, S.; Akiyama, T.; Yonehara, S. Viral FLIP enhances Wnt signaling downstream of stabilized beta-catenin, leading to control of cell growth. Mol. Cell. Biol. 2005, 25, 9249–9258. [Google Scholar] [CrossRef] [Green Version]
  371. Bélanger, C.; Gravel, A.; Tomoiu, A.; Janelle, M.E.; Gosselin, J.; Tremblay, M.J.; Flamand, L. Human herpesvirus 8 viral FLICE-inhibitory protein inhibits Fas-mediated apoptosis through binding and prevention of procaspase-8 maturation. J. Hum. Virol. 2001, 4, 62–73. [Google Scholar]
  372. Feoktistova, M.; Geserick, P.; Kellert, B.; Dimitrova, D.P.; Langlais, C.; Hupe, M.; Cain, K.; MacFarlane, M.; Häcker, G.; Leverkus, M. cIAPs block ripoptosome formation, a RIP1/caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Mol. Cell 2011, 43, 449–463. [Google Scholar] [CrossRef] [Green Version]
  373. Lee, J.S.; Li, Q.; Lee, J.Y.; Lee, S.H.; Jeong, J.H.; Lee, H.R.; Chang, H.; Zhou, F.C.; Gao, S.J.; Liang, C.; et al. FLIP-mediated autophagy regulation in cell death control. Nat. Cell. Biol. 2009, 11, 1355–1362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  374. Kim, K.H.; Seong, B.L. Pro-apoptotic function of HBV X protein is mediated by interaction with c-FLIP and enhancement of death-inducing signal. EMBO J. 2003, 22, 2104–2116. [Google Scholar] [CrossRef] [PubMed]
  375. Lee, A.R.; Lim, K.H.; Park, E.S.; Kim, D.H.; Park, Y.K.; Park, S.; Kim, D.S.; Shin, G.C.; Kang, H.S.; Won, J.; et al. Multiple functions of cellular FLIP are essential for replication of hepatitis B virus. J. Virol. 2018, 92, 18. [Google Scholar] [CrossRef] [Green Version]
  376. Saito, K.; Meyer, K.; Warner, R.; Basu, A.; Ray, R.B.; Ray, R. Hepatitis C virus core protein inhibits tumor necrosis factor alpha-mediated apoptosis by a protective effect involving cellular FLICE inhibitory protein. J. Virol. 2006, 80, 4372–4379. [Google Scholar] [CrossRef] [Green Version]
  377. Park, J.; Kang, W.; Ryu, S.W.; Kim, W.I.; Chang, D.Y.; Lee, D.H.; Park, D.Y.; Choi, Y.H.; Choi, K.; Shin, E.C.; et al. Hepatitis C virus infection enhances TNFα-induced cell death via suppression of NF-κB. Hepatology 2012, 56, 831–840. [Google Scholar] [CrossRef] [PubMed]
  378. Zhang, D.W.; Li, H.Y.; Lau, W.Y.; Cao, L.Q.; Li, Y.; Jiang, X.F.; Yang, X.W.; Xue, P. Gli2 silencing enhances TRAIL-induced apoptosis and reduces tumor growth in human hepatoma cells in vivo. Cancer Biol. Ther. 2014, 15, 1667–1676. [Google Scholar] [CrossRef] [Green Version]
  379. Rubinstein, A.D.; Eisenstein, M.; Ber, Y.; Bialik, S.; Kimchi, A. The autophagy protein Atg12 associates with antiapoptotic Bcl-2 family members to promote mitochondrial apoptosis. Mol. Cell 2011, 44, 698–709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  380. Su, M.; Mei, Y.; Sinha, S. Role of the crosstalk between autophagy and apoptosis in cancer. J. Oncol. 2013, 2013, 102735. [Google Scholar] [CrossRef] [Green Version]
  381. Geering, B. Death-associated protein kinase 2: Regulator of apoptosis, autophagy and inflammation. Int. J. Biochem. Cell. Biol. 2015, 65, 151–154. [Google Scholar] [CrossRef]
  382. Raveh, T.; Droguett, G.; Horwitz, M.S.; DePinho, R.A.; Kimchi, A. DAP kinase activates a p19ARF/p53-mediated apoptotic checkpoint to suppress oncogenic transformation. Nat. Cell. Biol. 2001, 3, 1–7. [Google Scholar] [CrossRef]
  383. Jang, C.W.; Chen, C.H.; Chen, C.C.; Chen, J.Y.; Su, Y.H.; Chen, R.H. TGF-beta induces apoptosis through Smad-mediated expression of DAP-kinase. Nat. Cell. Biol. 2002, 4, 51–58. [Google Scholar] [CrossRef]
  384. Li, H.; Ray, G.; Yoo, B.H.; Erdogan, M.; Rosen, K.V. Down-regulation of death-associated protein kinase-2 is required for beta-catenin-induced anoikis resistance of malignant epithelial cells. J. Biol. Chem. 2009, 284, 2012–2022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  385. Schlegel, C.R.; Fonseca, A.V.; Stöcker, S.; Georgiou, M.L.; Misterek, M.B.; Munro, C.E.; Carmo, C.R.; Seckl, M.J.; Costa-Pereira, A.P. DAPK2 is a novel modulator of TRAIL-induced apoptosis. Cell. Death Differ. 2014, 21, 1780–1791. [Google Scholar] [CrossRef] [Green Version]
  386. Ber, Y.; Shiloh, R.; Gilad, Y.; Degani, N.; Bialik, S.; Kimchi, A. DAPK2 is a novel regulator of mTORC1 activity and autophagy. Cell. Death Differ. 2015, 22, 465–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  387. Zalckvar, E.; Berissi, H.; Mizrachy, L.; Idelchuk, Y.; Koren, I.; Eisenstein, M.; Sabanay, H.; Pinkas-Kramarski, R.; Kimchi, A. DAP-kinase-mediated phosphorylation on the BH3 domain of beclin 1 promotes dissociation of beclin 1 from Bcl-XL and induction of autophagy. EMBO Rep. 2009, 10, 285–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  388. Gilad, Y.; Shiloh, R.; Ber, Y.; Bialik, S.; Kimchi, A. Discovering protein-protein interactions within the programmed cell death network using a protein-fragment complementation screen. Cell Rep. 2014, 8, 909–921. [Google Scholar] [CrossRef] [Green Version]
  389. Zhang, H.; Chen, G.G.; Zhang, Z.; Chun, S.; Leung, B.C.; Lai, P.B. Induction of autophagy in hepatocellular carcinoma cells by SB203580 requires activation of AMPK and DAPK but not p38 MAPK. Apoptosis 2012, 17, 325–334. [Google Scholar] [CrossRef]
  390. Huang, Y.; Wang, C.; Li, K.; Ye, Y.; Shen, A.; Guo, L.; Chen, P.; Meng, C.; Wang, Q.; Yang, X.; et al. Death-associated protein kinase 1 suppresses hepatocellular carcinoma cell migration and invasion by upregulation of DEAD-box helicase 20. Cancer Sci. 2020, 111, 2803–2813. [Google Scholar] [CrossRef]
  391. Wang, Y.; Liu, L.L.; Tian, Y.; Chen, Y.; Zha, W.H.; Li, Y.; Wu, F.J. Upregulation of DAPK2 ameliorates oxidative damage and apoptosis of placental cells in hypertensive disorder complicating pregnancy by suppressing human placental microvascular endothelial cell autophagy through the mTOR signaling pathway. Int. J. Biol. Macromol. 2019, 121, 488–497. [Google Scholar] [CrossRef]
  392. Li, T.; Wu, Y.N.; Wang, H.; Ma, J.Y.; Zhai, S.S.; Duan, J. Dapk1 improves inflammation, oxidative stress and autophagy in LPS-induced acute lung injury via p38MAPK/NF-κB signaling pathway. Mol. Immunol. 2020, 120, 13–22. [Google Scholar] [CrossRef]
  393. Li, Y.; Huang, H.; Yu, H.; Mo, T.; Wei, T.; Li, G.; Jia, Y.; Huang, X.; Tu, M.; Yan, X.; et al. Differential gene expression analysis after DAPK1 knockout in hepatocellular carcinoma cells. PeerJ 2022, 10, 13711. [Google Scholar] [CrossRef] [PubMed]
  394. Levine, A.J. p53, the cellular gatekeeper for growth and division. Cell 1997, 88, 323–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  395. Kuribayashi, K.; El-Deiry, W.S. Regulation of programmed cell death by the p53 pathway. Adv. Exp. Med. Biol. 2008, 615, 201–221. [Google Scholar] [CrossRef] [PubMed]
  396. Brady, C.A.; Jiang, D.; Mello, S.S.; Johnson, T.M.; Jarvis, L.A.; Kozak, M.M.; Kenzelmann Broz, D.; Basak, S.; Park, E.J.; McLaughlin, M.E.; et al. Distinct p53 transcriptional programs dictate acute DNA-damage responses and tumor suppression. Cell 2011, 145, 571–583. [Google Scholar] [CrossRef] [Green Version]
  397. Riley, T.; Sontag, E.; Chen, P.; Levine, A. Transcriptional control of human p53-regulated genes. Nat. Rev. Mol. Cell. Biol. 2008, 9, 402–412. [Google Scholar] [CrossRef] [PubMed]
  398. Green, D.R.; Kroemer, G. Cytoplasmic functions of the tumour suppressor p53. Nature 2009, 458, 1127–1130. [Google Scholar] [CrossRef] [Green Version]
  399. Vaseva, A.V.; Marchenko, N.D.; Ji, K.; Tsirka, S.E.; Holzmann, S.; Moll, U.M. p53 opens the mitochondrial permeability transition pore to trigger necrosis. Cell 2012, 149, 1536–1548. [Google Scholar] [CrossRef] [Green Version]
  400. Vaseva, A.V.; Moll, U.M. The mitochondrial p53 pathway. Biochim. Biophys. Acta 2009, 1787, 414–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  401. Aubrey, B.J.; Kelly, G.L.; Janic, A.; Herold, M.J.; Strasser, A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018, 25, 104–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  402. Feng, Z.; Zhang, H.; Levine, A.J.; Jin, S. The coordinate regulation of the p53 and mTOR pathways in cells. Proc. Natl. Acad. Sci. USA 2005, 102, 8204–8209. [Google Scholar] [CrossRef] [Green Version]
  403. Tasdemir, E.; Maiuri, M.C.; Galluzzi, L.; Vitale, I.; Djavaheri-Mergny, M.; D’Amelio, M.; Criollo, A.; Morselli, E.; Zhu, C.; Harper, F.; et al. Regulation of autophagy by cytoplasmic p53. Nat. Cell. Biol. 2008, 10, 676–687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  404. Crighton, D.; Wilkinson, S.; O′Prey, J.; Syed, N.; Smith, P.; Harrison, P.R.; Gasco, M.; Garrone, O.; Crook, T.; Ryan, K.M. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 2006, 126, 121–134. [Google Scholar] [CrossRef] [Green Version]
  405. Thomas, A.; Giesler, T.; White, E. p53 mediates bcl-2 phosphorylation and apoptosis via activation of the Cdc42/JNK1 pathway. Oncogene 2000, 19, 5259–5269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  406. Das, S.; Shukla, N.; Singh, S.S.; Kushwaha, S.; Shrivastava, R. Mechanism of interaction between autophagy and apoptosis in cancer. Apoptosis 2021, 26, 512–533. [Google Scholar] [CrossRef] [PubMed]
  407. Livesey, K.M.; Kang, R.; Vernon, P.; Buchser, W.; Loughran, P.; Watkins, S.C.; Zhang, L.; Manfredi, J.J.; Zeh, H.J., III; Li, L.; et al. p53/HMGB1 complexes regulate autophagy and apoptosis. Cancer Res. 2012, 72, 1996–2005. [Google Scholar] [CrossRef] [Green Version]
  408. Zhang, X.; Zheng, Q.; Yue, X.; Yuan, Z.; Ling, J.; Yuan, Y.; Liang, Y.; Sun, A.; Liu, Y.; Li, H.; et al. ZNF498 promotes hepatocellular carcinogenesis by suppressing p53-mediated apoptosis and ferroptosis via the attenuation of p53 Ser46 phosphorylation. J. Exp. Clin. Cancer Res. 2022, 41, 79. [Google Scholar] [CrossRef]
  409. Pratt, M.A.; White, D.; Kushwaha, N.; Tibbo, E.; Niu, M.Y. Cytoplasmic mutant p53 increases Bcl-2 expression in estrogen receptor-positive breast cancer cells. Apoptosis 2007, 12, 657–669. [Google Scholar] [CrossRef]
  410. Marques, M.A.; de Andrade, G.C.; Silva, J.L.; de Oliveira, G.A.P. Protein of a thousand faces: The tumor-suppressive and oncogenic responses of p53. Front. Mol. Biosci. 2022, 9, 944955. [Google Scholar] [CrossRef]
  411. Yang, C.; Huang, X.; Li, Y.; Chen, J.; Lv, Y.; Dai, S. Prognosis and personalized treatment prediction in TP53-mutant hepatocellular carcinoma: An in silico strategy towards precision oncology. Brief Bioinform. 2021, 22, 164. [Google Scholar] [CrossRef] [PubMed]
  412. Huang, Y.; Ge, W.; Zhou, J.; Gao, B.; Qian, X.; Wang, W. The role of tumor associated macrophages in hepatocellular carcinoma. J. Cancer 2021, 12, 1284–1294. [Google Scholar] [CrossRef] [PubMed]
  413. Ngabire, D.; Kim, G.D. Autophagy and inflammatory response in the tumor microenvironment. Int. J. Mol. Sci. 2017, 18, 2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  414. Sun, K.; Xu, L.; Jing, Y.; Han, Z.; Chen, X.; Cai, C.; Zhao, P.; Zhao, X.; Yang, L.; Wei, L. Autophagy-deficient Kupffer cells promote tumorigenesis by enhancing mtROS-NF-κB-IL1α/β-dependent inflammation and fibrosis during the preneoplastic stage of hepatocarcinogenesis. Cancer Lett. 2017, 388, 198–207. [Google Scholar] [CrossRef] [PubMed]
  415. Chang, C.P.; Su, Y.C.; Lee, P.H.; Lei, H.Y. Targeting NFKB by autophagy to polarize hepatoma-associated macrophage differentiation. Autophagy 2013, 9, 619–621. [Google Scholar] [CrossRef] [Green Version]
  416. Lin, H.; Yan, J.; Wang, Z.; Hua, F.; Yu, J.; Sun, W.; Li, K.; Liu, H.; Yang, H.; Lv, Q.; et al. Loss of immunity-supported senescence enhances susceptibility to hepatocellular carcinogenesis and progression in Toll-like receptor 2-deficient mice. Hepatology 2013, 57, 171–182. [Google Scholar] [CrossRef]
  417. Tan, H.Y.; Wang, N.; Man, K.; Tsao, S.W.; Che, C.M.; Feng, Y. Autophagy-induced RelB/p52 activation mediates tumour-associated macrophage repolarisation and suppression of hepatocellular carcinoma by natural compound baicalin. Cell Death Dis. 2015, 6, 1942. [Google Scholar] [CrossRef] [Green Version]
  418. Zhao, M.; Finlay, D.; Liddington, R.; Vuori, K. SRC plays a specific role in the cross-talk between apoptosis and autophagy via phosphorylation of a novel regulatory site on AMPK. Autophagy Rep. 2022, 1, 38–41. [Google Scholar] [CrossRef]
  419. Xu, H.; Ye, D.; Ren, M.; Zhang, H.; Bi, F. Ferroptosis in the tumor microenvironment: Perspectives for immunotherapy. Trends Mol Med. 2021, 27, 856–867. [Google Scholar] [CrossRef]
  420. Gu, X.; Liu, Y.; Dai, X.; Yang, Y.G.; Zhang, X. Deciphering the potential roles of ferroptosis in regulating tumor immunity and tumor immunotherapy. Front Immunol. 2023, 14, 1137107. [Google Scholar] [CrossRef]
  421. Cai, H.; Ren, Y.; Chen, S.; Wang, Y.; Chu, L. Ferroptosis and tumor immunotherapy: A promising combination therapy for tumors. Front Oncol. 2023, 13, 1119369. [Google Scholar] [CrossRef]
  422. Guo, J.Y.; Chen, H.Y.; Mathew, R.; Fan, J.; Strohecker, A.M.; Karsli-Uzunbas, G.; Kamphorst, J.J.; Chen, G.; Lemons, J.M.; Karantza, V.; et al. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev. 2011, 25, 460–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  423. Qu, X.; Sheng, J.; Shen, L.; Su, J.; Xu, Y.; Xie, Q.; Wu, Y.; Zhang, X.; Sun, L. Autophagy inhibitor chloroquine increases sensitivity to cisplatin in QBC939 cholangiocarcinoma cells by mitochondrial ROS. PLoS ONE 2017, 12, 0173712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  424. Hori, Y.S.; Hosoda, R.; Akiyama, Y.; Sebori, R.; Wanibuchi, M.; Mikami, T.; Sugino, T.; Suzuki, K.; Maruyama, M.; Tsukamoto, M.; et al. Chloroquine potentiates temozolomide cytotoxicity by inhibiting mitochondrial autophagy in glioma cells. J. Neurooncol. 2015, 122, 11–20. [Google Scholar] [CrossRef]
  425. Goldsmith, J.; Levine, B.; Debnath, J. Autophagy and cancer metabolism. Methods Enzymol. 2014, 542, 25–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  426. Shetty, S.; Kumar, R.; Bharati, S. Mito-TEMPO, a mitochondria-targeted antioxidant, prevents N-nitrosodiethylamine-induced hepatocarcinogenesis in mice. Free Radic. Biol. Med. 2019, 136, 76–86. [Google Scholar] [CrossRef]
  427. Shetty, S.; Anushree, U.; Kumar, R.; Bharati, S. Mitochondria-targeted antioxidant, mito-TEMPO mitigates initiation phase of N-Nitrosodiethylamine-induced hepatocarcinogenesis. Mitochondrion 2021, 58, 123–130. [Google Scholar] [CrossRef]
  428. Li, J.; Jiang, R.; Cong, X.; Zhao, Y. UCP2 gene polymorphisms in obesity and diabetes, and the role of UCP2 in cancer. FEBS Lett. 2019, 593, 2525–2534. [Google Scholar] [CrossRef] [Green Version]
  429. Fernández-Tussy, P.; Rodríguez-Agudo, R.; Fernández-Ramos, D.; Barbier-Torres, L.; Zubiete-Franco, I.; Davalillo, S.L.; Herraez, E.; Goikoetxea-Usandizaga, N.; Lachiondo-Ortega, S.; Simón, J.; et al. Anti-miR-518d-5p overcomes liver tumor cell death resistance through mitochondrial activity. Cell Death Dis. 2021, 12, 555. [Google Scholar] [CrossRef]
  430. Hou, Z.L.; Han, F.Y.; Lou, L.L.; Zhao, W.Y.; Huang, X.X.; Yao, G.D.; Song, S.J. The nature compound dehydrocrenatidine exerts potent antihepatocellular carcinoma by destroying mitochondrial complexes in vitro and in vivo. Phytother. Res. 2022, 36, 1353–1371. [Google Scholar] [CrossRef]
  431. Yao, J.; Wang, J.; Xu, Y.; Guo, Q.; Sun, Y.; Liu, J.; Li, S.; Guo, Y.; Wei, L. CDK9 inhibition blocks the initiation of PINK1-PRKN-mediated mitophagy by regulating the SIRT1-FOXO3-BNIP3 axis and enhances the therapeutic effects involving mitochondrial dysfunction in hepatocellular carcinoma. Autophagy 2022, 18, 1879–1897. [Google Scholar] [CrossRef]
  432. Abate, M.; Festa, A.; Falco, M.; Lombardi, A.; Luce, A.; Grimaldi, A.; Zappavigna, S.; Sperlongano, P.; Irace, C.; Caraglia, M.; et al. Mitochondria as playmakers of apoptosis, autophagy and senescence. Semin. Cell. Dev. Biol. 2020, 98, 139–153. [Google Scholar] [CrossRef]
  433. Zhang, C.; Zhao, Y.; Yu, M.; Qin, J.; Ye, B.; Wang, Q. Mitochondrial dysfunction and chronic liver disease. Curr. Issues Mol. Biol. 2022, 44, 3156–3165. [Google Scholar] [CrossRef] [PubMed]
  434. Han, Z.; Liu, D.; Chen, L.; He, Y.; Tian, X.; Qi, L.; Chen, L.; Luo, Y.; Chen, Z.; Hu, X.; et al. PNO1 regulates autophagy and apoptosis of hepatocellular carcinoma via the MAPK signaling pathway. Cell Death Dis. 2021, 12, 552. [Google Scholar] [CrossRef]
  435. Li, J.; Cao, F.; Yin, H.L.; Huang, Z.J.; Lin, Z.T.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, present and future. Cell Death Dis. 2020, 11, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  436. Latunde-Dada, G.O. Ferroptosis: Role of lipid peroxidation, iron and ferritinophagy. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 1893–1900. [Google Scholar] [CrossRef] [Green Version]
  437. Chen, X.; Comish, P.B.; Tang, D.; Kang, R. Characteristics and biomarkers of ferroptosis. Front. Cell. Dev. Biol. 2021, 9, 637162. [Google Scholar] [CrossRef]
  438. Zheng, J.; Conrad, M. The metabolic underpinnings of ferroptosis. Cell Metab. 2020, 32, 920–937. [Google Scholar] [CrossRef] [PubMed]
  439. Koppula, P.; Zhuang, L.; Gan, B. Cystine transporter SLC7A11/xCT in cancer: Ferroptosis, nutrient dependency, and cancer therapy. Protein Cell 2021, 12, 599–620. [Google Scholar] [CrossRef]
  440. Hou, W.; Xie, Y.; Song, X.; Sun, X.; Lotze, M.T.; Zeh, H.J., III; Kang, R.; Tang, D. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 2016, 12, 1425–1428. [Google Scholar] [CrossRef]
  441. Sun, X.; Ou, Z.; Chen, R.; Niu, X.; Chen, D.; Kang, R.; Tang, D. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology 2016, 63, 173–184. [Google Scholar] [CrossRef] [Green Version]
  442. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular mechanisms and health implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef] [PubMed]
  443. Dodson, M.; de la Vega, M.R.; Cholanians, A.B.; Schmidlin, C.J.; Chapman, E.; Zhang, D.D. Modulating NRF2 in disease: Timing is everything. Annu. Rev. Pharmacol. Toxicol. 2019, 59, 555–575. [Google Scholar] [CrossRef]
  444. Song, X.; Zhu, S.; Chen, P.; Hou, W.; Wen, Q.; Liu, J.; Xie, Y.; Liu, J.; Klionsky, D.J.; Kroemer, G.; et al. AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc-activity. Curr. Biol. 2018, 28, 2388–2399. [Google Scholar] [CrossRef] [Green Version]
  445. Zhou, B.; Liu, J.; Kang, R.; Klionsky, D.J.; Kroemer, G.; Tang, D. Ferroptosis is a type of autophagy-dependent cell death. Semin. Cancer Biol. 2020, 66, 89–100. [Google Scholar] [CrossRef]
  446. Hong, S.H.; Lee, D.H.; Lee, Y.S.; Jo, M.J.; Jeong, Y.A.; Kwon, W.T.; Choudry, H.A.; Bartlett, D.L.; Lee, Y.J. Molecular crosstalk between ferroptosis and apoptosis: Emerging role of ER stress-induced p53-independent PUMA expression. Oncotarget 2017, 8, 115164–115178. [Google Scholar] [CrossRef] [Green Version]
  447. Huang, C.; Yang, M.; Deng, J.; Li, P.; Su, W.; Jiang, R. Upregulation and activation of p53 by erastin-induced reactive oxygen species contribute to cytotoxic and cytostatic effects in A549 lung cancer cells. Oncol. Rep. 2018, 40, 2363–2370. [Google Scholar] [CrossRef] [PubMed]
  448. Kew, M.C. Hepatic iron overload and hepatocellular carcinoma. Liver Cancer 2014, 3, 31–40. [Google Scholar] [CrossRef] [PubMed]
  449. Torti, S.V.; Manz, D.H.; Paul, B.T.; Blanchette-Farra, N.; Torti, F.M. Iron and cancer. Annu. Rev. Nutr. 2018, 38, 97–125. [Google Scholar] [CrossRef]
  450. Liao, H.; Shi, J.; Wen, K.; Lin, J.; Liu, Q.; Shi, B.; Yan, Y.; Xiao, Z. Molecular targets of ferroptosis in hepatocellular carcinoma. J. Hepatocell. Carcinoma 2021, 8, 985–996. [Google Scholar] [CrossRef]
  451. Jennis, M.; Kung, C.P.; Basu, S.; Budina-Kolomets, A.; Leu, J.I.; Khaku, S.; Scott, J.P.; Cai, K.Q.; Campbell, M.R.; Porter, D.K.; et al. An African-specific polymorphism in the TP53 gene impairs p53 tumor suppressor function in a mouse model. Genes Dev. 2016, 30, 918–930. [Google Scholar] [CrossRef] [Green Version]
  452. Nie, J.; Lin, B.; Zhou, M.; Wu, L.; Zheng, T. Role of ferroptosis in hepatocellular carcinoma. J. Cancer Res. Clin. Oncol. 2018, 144, 2329–2337. [Google Scholar] [CrossRef]
  453. Sun, X.; Niu, X.; Chen, R.; He, W.; Chen, D.; Kang, R.; Tang, D. Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology 2016, 64, 488–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  454. Shang, Y.; Luo, M.; Yao, F.; Wang, S.; Yuan, Z.; Yang, Y. Ceruloplasmin suppresses ferroptosis by regulating iron homeostasis in hepatocellular carcinoma cells. Cell. Signal. 2020, 72, 109633. [Google Scholar] [CrossRef] [PubMed]
  455. Doll, S.; Proneth, B.; Tyurina, Y.Y.; Panzilius, E.; Kobayashi, S.; Ingold, I.; Irmler, M.; Beckers, J.; Aichler, M.; Walch, A.; et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 2017, 13, 91–98. [Google Scholar] [CrossRef] [PubMed]
  456. Feng, J.; Lu, P.Z.; Zhu, G.Z.; Hooi, S.C.; Wu, Y.; Huang, X.W.; Dai, H.Q.; Chen, P.H.; Li, Z.J.; Su, W.J.; et al. ACSL4 is a predictive biomarker of sorafenib sensitivity in hepatocellular carcinoma. Acta Pharmacol. Sin. 2021, 42, 160–170. [Google Scholar] [CrossRef]
  457. Sun, X.J.; Xu, G.L. Overexpression of Acyl-CoA Ligase 4 (ACSL4) in patients with hepatocellular carcinoma and its prognosis. Med. Sci. Monit. 2017, 23, 4343–4350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  458. Quan, J.; Bode, A.M.; Luo, X. ACSL family: The regulatory mechanisms and therapeutic implications in cancer. Eur. J. Pharmacol. 2021, 909, 174397. [Google Scholar] [CrossRef] [PubMed]
  459. Chen, X.; Li, J.; Kang, R.; Klionsky, D.J.; Tang, D. Ferroptosis: Machinery and regulation. Autophagy 2021, 17, 2054–2081. [Google Scholar] [CrossRef]
  460. Chen, J.; Ding, C.; Chen, Y.; Hu, W.; Yu, C.; Peng, C.; Feng, X.; Cheng, Q.; Wu, W.; Lu, Y.; et al. ACSL4 reprograms fatty acid metabolism in hepatocellular carcinoma via c-Myc/SREBP1 pathway. Cancer Lett. 2021, 502, 154–165. [Google Scholar] [CrossRef]
  461. Li, H.; Song, J.; He, Y.; Liu, Y.; Liu, Z.; Sun, W.; Hu, W.; Lei, Q.Y.; Hu, X.; Chen, Z.; et al. CRISPR/Cas9 screens reveal that hexokinase 2 enhances cancer stemness and tumorigenicity by activating the ACSL4-fatty acid β-oxidation pathway. Adv. Sci. 2022, 9, e2105126. [Google Scholar] [CrossRef]
  462. Han, Y.M.; Jeong, M.; Park, J.M.; Kim, M.Y.; Go, E.J.; Cha, J.Y.; Kim, K.J.; Hahm, K.B. The ω-3 polyunsaturated fatty acids prevented colitis-associated carcinogenesis through blocking dissociation of β-catenin complex, inhibiting COX-2 through repressing NF-κB, and inducing 15-prostaglandin dehydrogenase. Oncotarget 2016, 7, 63583–63595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  463. Ou, W.; Mulik, R.S.; Anwar, A.; McDonald, J.G.; He, X.; Corbin, I.R. Low-density lipoprotein docosahexaenoic acid nanoparticles induce ferroptotic cell death in hepatocellular carcinoma. Free Radic. Biol. Med. 2017, 112, 597–607. [Google Scholar] [CrossRef] [PubMed]
  464. Weylandt, K.H.; Krause, L.F.; Gomolka, B.; Chiu, C.Y.; Bilal, S.; Nadolny, A.; Waechter, S.F.; Fischer, A.; Rothe, M.; Kang, J.X. Suppressed liver tumorigenesis in fat-1 mice with elevated omega-3 fatty acids is associated with increased omega-3 derived lipid mediators and reduced TNF-α. Carcinogenesis 2011, 32, 897–903. [Google Scholar] [CrossRef] [Green Version]
  465. Lim, L.J.; Wong, S.Y.S.; Huang, F.; Lim, S.; Chong, S.S.; Ooi, L.L.; Kon, O.L.; Lee, C.G. Roles and regulation of long noncoding RNAs in hepatocellular carcinoma. Cancer Res. 2019, 79, 5131–5139. [Google Scholar] [CrossRef]
  466. Qi, W.; Li, Z.; Xia, L.; Dai, J.; Zhang, Q.; Wu, C.; Xu, S. LncRNA GABPB1-AS1 and GABPB1 regulate oxidative stress during erastin-induced ferroptosis in HepG2 hepatocellular carcinoma cells. Sci. Rep. 2019, 9, 16185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  467. Xu, Z.; Peng, B.; Liang, Q.; Chen, X.; Cai, Y.; Zeng, S.; Gao, K.; Wang, X.; Yi, Q.; Gong, Z.; et al. Construction of a ferroptosis-related nine-lncRNA signature for predicting prognosis and immune response in hepatocellular carcinoma. Front. Immunol. 2021, 12, 719175. [Google Scholar] [CrossRef]
  468. Xiong, Y.; Ouyang, Y.; Fang, K.; Sun, G.; Tu, S.; Xin, W.; Wei, Y.; Xiao, W. Prediction of prognosis and molecular mechanism of ferroptosis in hepatocellular carcinoma based on bioinformatics methods. Comput. Math. Methods Med. 2022, 2022, 4558782. [Google Scholar] [CrossRef]
  469. Lyu, N.; Zeng, Y.; Kong, Y.; Chen, Q.; Deng, H.; Ou, S.; Bai, Y.; Tang, H.; Wang, X.; Zhao, M. Ferroptosis is involved in the progression of hepatocellular carcinoma through the circ0097009/miR-1261/SLC7A11 axis. Ann. Transl. Med. 2021, 9, 675. [Google Scholar] [CrossRef]
  470. Zhang, B.; Zhao, J.; Liu, B.; Shang, Y.; Chen, F.; Zhang, S.; He, J.; Fan, Y.; Tan, K. Development and validation of a novel ferroptosis-related gene signature for prognosis and immunotherapy in hepatocellular carcinoma. Front. Mol. Biosci. 2022, 9, 940575. [Google Scholar] [CrossRef]
  471. Liang, J.Y.; Wang, D.S.; Lin, H.C.; Chen, X.X.; Yang, H.; Zheng, Y.; Li, Y.H. A novel ferroptosis-related gene signature for overall survival prediction in patients with hepatocellular carcinoma. Int. J. Biol. Sci. 2020, 16, 2430–2441. [Google Scholar] [CrossRef] [PubMed]
  472. Pan, F.; Lin, X.; Hao, L.; Wang, T.; Song, H.; Wang, R. The critical role of ferroptosis in hepatocellular carcinoma. Front. Cell Dev. Biol. 2022, 10, 882571. [Google Scholar] [CrossRef]
  473. Huang, Z.; Xia, H.; Cui, Y.; Yam, J.W.P.; Xu, Y. Ferroptosis: From basic research to clinical therapeutics in hepatocellular carcinoma. J. Clin. Transl. Hepatol. 2023, 11, 207–218. [Google Scholar] [CrossRef] [PubMed]
  474. Brunetti, O.; Gnoni, A.; Licchetta, A.; Longo, V.; Calabrese, A.; Argentiero, A.; Delcuratolo, S.; Solimando, A.G.; Casadei-Gardini, A.; Silvestris, N. Predictive and prognostic factors in HCC patients treated with sorafenib. Medicina 2019, 55, 707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  475. Facciorusso, A.; Abd El Aziz, M.A.; Singh, S.; Pusceddu, S.; Milione, M.; Giacomelli, L.; Sacco, R. Statin use decreases the incidence of hepatocellular carcinoma: An updated meta-analysis. Cancers 2020, 12, 874. [Google Scholar] [CrossRef] [Green Version]
  476. Solimando, A.G.; Susca, N.; Argentiero, A.; Brunetti, O.; Leone, P.; De Re, V.; Fasano, R.; Krebs, M.; Petracci, E.; Azzali, I.; et al. Second-line treatments for advanced hepatocellular carcinoma: A systematic review and Bayesian network meta-analysis. Clin. Exp. Med. 2022, 22, 65–74. [Google Scholar] [CrossRef] [PubMed]
  477. Tang, B.; Zhu, J.; Li, J.; Fan, K.; Gao, Y.; Cheng, S.; Kong, C.; Zheng, L.; Wu, F.; Weng, Q.; et al. The ferroptosis and iron-metabolism signature robustly predicts clinical diagnosis, prognosis and immune microenvironment for hepatocellular carcinoma. Cell Commun. Signal. 2020, 18, 174. [Google Scholar] [CrossRef]
  478. Brun, S.; Bestion, E.; Raymond, E.; Bassissi, F.; Jilkova, Z.M.; Mezouar, S.; Rachid, M.; Novello, M.; Tracz, J.; Hamaï, A.; et al. GNS561, a clinical-stage PPT1 inhibitor, is efficient against hepatocellular carcinoma via modulation of lysosomal functions. Autophagy 2022, 18, 678–694. [Google Scholar] [CrossRef]
  479. Wang, W.; Green, M.; Choi, J.E.; Gijón, M.; Kennedy, P.D.; Johnson, J.K.; Liao, P.; Lang, X.; Kryczek, I.; Sell, A.; et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019, 569, 270–274. [Google Scholar] [CrossRef]
  480. Zheng, C.; Peng, Y.; Wang, H.; Wang, Y.; Liu, L.; Zhao, Q. Identification and validation of ferroptosis-related subtypes and a predictive signature in hepatocellular carcinoma. Pharmgenomics Pers. Med. 2023, 16, 39–58. [Google Scholar] [CrossRef]
  481. Fan, G.; Wei, X.; Xu, X. Is the era of sorafenib over? A review of the literature. Ther. Adv. Med. Oncol. 2020, 12, 1758835920927602. [Google Scholar] [CrossRef]
  482. Louandre, C.; Ezzoukhry, Z.; Godin, C.; Barbare, J.C.; Mazière, J.C.; Chauffert, B.; Galmiche, A. Iron-dependent cell death of hepatocellular carcinoma cells exposed to sorafenib. Int. J. Cancer 2013, 133, 1732–1742. [Google Scholar] [CrossRef]
  483. Gao, R.; Kalathur, R.K.R.; Coto-Llerena, M.; Ercan, C.; Buechel, D.; Shuang, S.; Piscuoglio, S.; Dill, M.T.; Camargo, F.D.; Christofori, G.; et al. YAP/TAZ and ATF4 drive resistance to Sorafenib in hepatocellular carcinoma by preventing ferroptosis. EMBO Mol. Med. 2021, 13, e14351. [Google Scholar] [CrossRef] [PubMed]
  484. Li, D.; Yao, Y.; Rao, Y.; Huang, X.; Wei, L.; You, Z.; Zheng, G.; Hou, X.; Su, Y.; Varghese, Z.; et al. Cholesterol sensor SCAP contributes to sorafenib resistance by regulating autophagy in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2022, 41, 116. [Google Scholar] [CrossRef]
  485. Jing, Z.; Ye, X.; Ma, X.; Hu, X.; Yang, W.; Shi, J.; Chen, G.; Gong, L. SNGH16 regulates cell autophagy to promote Sorafenib resistance through suppressing miR-23b-3p via sponging EGR1 in hepatocellular carcinoma. Cancer Med. 2020, 9, 4324–4338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  486. Lu, Y.; Chan, Y.T.; Tan, H.Y.; Zhang, C.; Guo, W.; Xu, Y.; Sharma, R.; Chen, Z.S.; Zheng, Y.C.; Wang, N.; et al. Epigenetic regulation of ferroptosis via ETS1/miR-23a-3p/ACSL4 axis mediates sorafenib resistance in human hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2022, 41, 3. [Google Scholar] [CrossRef] [PubMed]
  487. Xu, W.P.; Liu, J.P.; Feng, J.F.; Zhu, C.P.; Yang, Y.; Zhou, W.P.; Ding, J.; Huang, C.K.; Cui, Y.L.; Ding, C.H.; et al. miR-541 potentiates the response of human hepatocellular carcinoma to sorafenib treatment by inhibiting autophagy. Gut 2020, 69, 1309–1321. [Google Scholar] [CrossRef]
  488. Fondevila, F.; Méndez-Blanco, C.; Fernández-Palanca, P.; Payo-Serafín, T.; van Pelt, J.; Verslype, C.; González-Gallego, J.; Mauriz, J.L. Autophagy-related chemoprotection against sorafenib in human hepatocarcinoma: Role of FOXO3 upregulation and modulation by regorafenib. Int. J. Mol. Sci. 2021, 22, 11770. [Google Scholar] [CrossRef] [PubMed]
  489. Liu, L.; Lv, Z.; Wang, M.; Zhang, D.; Liu, D.; Zhu, F. HBV enhances sorafenib resistance in hepatocellular carcinoma by reducing ferroptosis via SRSF2-mediated abnormal PCLAF splicing. Int. J. Mol. Sci. 2023, 24, 3263. [Google Scholar] [CrossRef]
  490. Sun, J.; Zhou, C.; Zhao, Y.; Zhang, X.; Chen, W.; Zhou, Q.; Hu, B.; Gao, D.; Raatz, L.; Wang, Z.; et al. Quiescin sulfhydryl oxidase 1 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by driving EGFR endosomal trafficking and inhibiting NRF2 activation. Redox Biol. 2021, 41, 101942. [Google Scholar] [CrossRef]
  491. Shao, W.Q.; Zhu, W.W.; Luo, M.J.; Fan, M.H.; Li, Q.; Wang, S.H.; Lin, Z.F.; Zhao, J.; Zheng, Y.; Dong, Q.Z.; et al. Cholesterol suppresses GOLM1-dependent selective autophagy of RTKs in hepatocellular carcinoma. Cell Rep. 2022, 39, 110712. [Google Scholar] [CrossRef]
  492. Li, Y.; Xu, B.; Ren, X.; Wang, L.; Xu, Y.; Zhao, Y.; Yang, C.; Yuan, C.; Li, H.; Tong, X.; et al. Inhibition of CISD2 promotes ferroptosis through ferritinophagy-mediated ferritin turnover and regulation of p62-Keap1-NRF2 pathway. Cell. Mol. Biol. Lett. 2022, 27, 81. [Google Scholar] [CrossRef]
  493. Li, H.; Zhao, J.; Zhong, X.L.; Xu, P.Y.; Du, L.J.; Fang, P.; Tan, L.J.; Li, M.J.; Zhang, C.F.; Cao, T.S. CPLX2 regulates ferroptosis and apoptosis through NRF2 pathway in human hepatocellular carcinoma cells. Appl. Biochem. Biotechnol. 2023, 195, 597–609. [Google Scholar] [CrossRef] [PubMed]
  494. Bai, T.; Wang, S.; Zhao, Y.; Zhu, R.; Wang, W.; Sun, Y. Haloperidol, a sigma receptor 1 antagonist, promotes ferroptosis in hepatocellular carcinoma cells. Biochem. Biophys. Res. Commun. 2017, 491, 919–925. [Google Scholar] [CrossRef] [PubMed]
  495. Zhao, F.; Feng, G.; Zhu, J.; Su, Z.; Guo, R.; Liu, J.; Zhang, H.; Zhai, Y. 3-Methyladenine-enhanced susceptibility to sorafenib in hepatocellular carcinoma cells by inhibiting autophagy. Anti-Cancer Drugs 2021, 32, 386–393. [Google Scholar] [CrossRef]
  496. Wu, H.; Wang, T.; Liu, Y.; Li, X.; Xu, S.; Wu, C.; Zou, H.; Cao, M.; Jin, G.; Lang, J.; et al. Mitophagy promotes sorafenib resistance through hypoxia-inducible ATAD3A dependent Axis. J. Exp. Clin. Cancer Res. 2020, 39, 274. [Google Scholar] [CrossRef]
  497. Zheng, Y.; Huang, C.; Lu, L.; Yu, K.; Zhao, J.; Chen, M.; Liu, L.; Sun, Q.; Lin, Z.; Zheng, J.; et al. STOML2 potentiates metastasis of hepatocellular carcinoma by promoting PINK1-mediated mitophagy and regulates sensitivity to lenvatinib. J. Hematol. Oncol. 2021, 14, 16. [Google Scholar] [CrossRef] [PubMed]
  498. Chen, Y.; Chen, H.N.; Wang, K.; Zhang, L.; Huang, Z.; Liu, J.; Zhang, Z.; Luo, M.; Lei, Y.; Peng, Y.; et al. Ketoconazole exacerbates mitophagy to induce apoptosis by downregulating cyclooxygenase-2 in hepatocellular carcinoma. J. Hepatol. 2019, 70, 66–77. [Google Scholar] [CrossRef]
  499. Wang, Z.; Zhu, Q.; Li, X.; Ren, X.; Li, J.; Zhang, Y.; Zeng, S.; Xu, L.; Dong, X.; Zhai, B. TOP2A inhibition reverses drug resistance of hepatocellular carcinoma to regorafenib. Am. J. Cancer Res. 2022, 12, 4343–4360. [Google Scholar]
  500. Chang, W.T.; Bow, Y.D.; Fu, P.J.; Li, C.Y.; Wu, C.Y.; Chang, Y.H.; Teng, Y.N.; Li, R.N.; Lu, M.C.; Liu, Y.C.; et al. A marine terpenoid, heteronemin, induces both the apoptosis and ferroptosis of hepatocellular carcinoma cells and involves the ROS and MAPK pathways. Oxid. Med. Cell. Longev. 2021, 2021, 7689045. [Google Scholar] [CrossRef] [PubMed]
  501. Sun, C.; Zhang, J.; Hou, J.; Hui, M.; Qi, H.; Lei, T.; Zhang, X.; Zhao, L.; Du, H. Induction of autophagy via the PI3K/Akt/mTOR signaling pathway by Pueraria flavonoids improves non-alcoholic fatty liver disease in obese mice. Biomed. Pharmacother. 2023, 157, 114005. [Google Scholar] [CrossRef] [PubMed]
  502. Zhang, J.; Shang, L.; Jiang, W.; Wu, W. Shikonin induces apoptosis and autophagy via downregulation of pyrroline-5-carboxylate reductase1 in hepatocellular carcinoma cells. Bioengineered 2022, 13, 7904–7918. [Google Scholar] [CrossRef] [PubMed]
  503. Ji, A.; Hu, L.; Ma, D.; Qiang, G.; Yan, D.; Zhang, G.; Jiang, C. Myricetin induces apoptosis and protective autophagy through endoplasmic reticulum stress in hepatocellular carcinoma. Evid. Based Complement. Altern. Med. 2022, 2022, 3115312. [Google Scholar] [CrossRef]
  504. Jiang, Z.; Gao, L.; Liu, C.; Wang, J.; Han, Y.; Pan, J. Sarmentosin induces autophagy-dependent apoptosis via activation of Nrf2 in hepatocellular carcinoma. J. Clin. Transl. Hepatol. 2023. [Google Scholar] [CrossRef]
  505. Oura, K.; Morishita, A.; Hamaya, S.; Fujita, K.; Masaki, T. The roles of epigenetic regulation and the tumor microenvironment in the mechanism of resistance to systemic therapy in hepatocellular carcinoma. Int. J. Mol. Sci. 2023, 24, 2805. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Regulatory pathways of autophagy. Black arrows indicate induction and red arrows indicate inhibition. See text for details.
Figure 1. Regulatory pathways of autophagy. Black arrows indicate induction and red arrows indicate inhibition. See text for details.
Biomedicines 11 01166 g001
Figure 2. A simplified diagram of the interplay of autophagy and apoptosis. Most controllers of the interplay are presented. p53, the main gatekeeper, increases autophagy directly or indirectly through the DRAM activation of AMPK, but cytoplasmic p53 may inhibit autophagy. p53 increases apoptosis via the inhibition of Bcl-2 or the overexpression of BAX. Bcl-2, associated with Beclin1, inactivates the complex, leading to increased autophagy (only Vps34 is shown here). The phosphorylation of either Beclin1 by DAPK or Bcl-2 by JNK liberates the pro-autophagy complex. DAPK also increases apoptosis by an unknown mechanism. The cleavage of Beclin1 or Atg5 (an autophagy inducer promoted by ER stress) by apoptosis-induced caspases inhibits autophagy. Several intermediate components have been omitted for clarity. Intermittent line: Cleavage.
Figure 2. A simplified diagram of the interplay of autophagy and apoptosis. Most controllers of the interplay are presented. p53, the main gatekeeper, increases autophagy directly or indirectly through the DRAM activation of AMPK, but cytoplasmic p53 may inhibit autophagy. p53 increases apoptosis via the inhibition of Bcl-2 or the overexpression of BAX. Bcl-2, associated with Beclin1, inactivates the complex, leading to increased autophagy (only Vps34 is shown here). The phosphorylation of either Beclin1 by DAPK or Bcl-2 by JNK liberates the pro-autophagy complex. DAPK also increases apoptosis by an unknown mechanism. The cleavage of Beclin1 or Atg5 (an autophagy inducer promoted by ER stress) by apoptosis-induced caspases inhibits autophagy. Several intermediate components have been omitted for clarity. Intermittent line: Cleavage.
Biomedicines 11 01166 g002
Figure 3. The p27 switch between autophagy and apoptosis is dependent on the phosphorylation and cellular localization of p27kip1.
Figure 3. The p27 switch between autophagy and apoptosis is dependent on the phosphorylation and cellular localization of p27kip1.
Biomedicines 11 01166 g003
Figure 4. Mechanisms of ferroptosis. The Xc–antiporter system consisting of the SLC7A11 and SLC3A2 subunits allows for the extrusion and internalization of glutamate and cysteine. Glutathione peroxidase 4 (GPX 4) is produced by the glutamate–cystine exchange system. Xc- is the main inhibitor of ROS. See text for details.
Figure 4. Mechanisms of ferroptosis. The Xc–antiporter system consisting of the SLC7A11 and SLC3A2 subunits allows for the extrusion and internalization of glutamate and cysteine. Glutathione peroxidase 4 (GPX 4) is produced by the glutamate–cystine exchange system. Xc- is the main inhibitor of ROS. See text for details.
Biomedicines 11 01166 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kouroumalis, E.; Tsomidis, I.; Voumvouraki, A. Pathogenesis of Hepatocellular Carcinoma: The Interplay of Apoptosis and Autophagy. Biomedicines 2023, 11, 1166. https://doi.org/10.3390/biomedicines11041166

AMA Style

Kouroumalis E, Tsomidis I, Voumvouraki A. Pathogenesis of Hepatocellular Carcinoma: The Interplay of Apoptosis and Autophagy. Biomedicines. 2023; 11(4):1166. https://doi.org/10.3390/biomedicines11041166

Chicago/Turabian Style

Kouroumalis, Elias, Ioannis Tsomidis, and Argyro Voumvouraki. 2023. "Pathogenesis of Hepatocellular Carcinoma: The Interplay of Apoptosis and Autophagy" Biomedicines 11, no. 4: 1166. https://doi.org/10.3390/biomedicines11041166

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop