Next Article in Journal
Gestational Viral Infections: Role of Host Immune System
Next Article in Special Issue
The Role of Fusobacterium nucleatum in Oral and Colorectal Carcinogenesis
Previous Article in Journal
The Gut Microbiota of Farmed and Wild Brook Trout (Salvelinus fontinalis): Evaluation of Feed-Related Differences Using 16S rRNA Gene Metabarcoding
Previous Article in Special Issue
The Immune Response Generated against HPV Infection in Men and Its Implications in the Diagnosis of Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Infectious Agents Induce Wnt/β-Catenin Pathway Deregulation in Primary Liver Cancers

by
Teresa Catalano
1,*,
Federico Selvaggi
2,
Diana Liberata Esposito
3,4,
Roberto Cotellese
5,6 and
Gitana Maria Aceto
5,*
1
Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
2
Unit of General Surgery, ASL2 Lanciano-Vasto-Chieti, Ospedale Clinicizzato SS Annunziata, 66100 Chieti, Italy
3
Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy
4
Department of Innovative Technologies in Medicine & Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
5
Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
6
Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy
*
Authors to whom correspondence should be addressed.
Microorganisms 2023, 11(7), 1632; https://doi.org/10.3390/microorganisms11071632
Submission received: 25 May 2023 / Revised: 18 June 2023 / Accepted: 20 June 2023 / Published: 22 June 2023
(This article belongs to the Special Issue Oncogenic Role of Viruses and Bacteria)

Abstract

:
Interaction between infectious agents and liver tissue, as well as repeated and extreme biological events beyond adaptive capacities, may result in pathological conditions predisposing people to development of primary liver cancers (PLCs). In adults, PLCs mainly comprise hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). Various infectious agents in the hepatic microenvironment can destabilize normal liver cell functions by modulating the Wnt/β-catenin pathway components. Among them, hepatotropic viruses B, C, and D are involved in Wnt/β-catenin signaling dysregulation. Other microbial agents, including oncogenic viruses such as Epstein–Barr virus (EBV) and human papilloma virus (HPV), bacteria, e.g., Mycoplasma hyorhinis and Salmonella Typhi, the protozoan parasite Toxoplasma gondii, the fungus Aspergillus flavus, and liver flukes such as Clonorchissinensis or Opisthorchis viverrini, may induce malignant transformation in hepatocytes or in target cells of the biliary tract through aberrant Wnt signaling activation. This review focuses on new insights into infectious agents implicated in the deregulation of Wnt signaling and PLC development. Since the Wnt/β-catenin pathway is a driver of cancer following viral and bacterial infections, molecules inhibiting the complex axis of Wnt signaling could represent novel therapeutic approaches in PLC treatment.

1. Introduction

According to global cancer 2020 estimates, primary liver cancer (PLC) is the sixth most common cancer worldwide and represents the third cause of tumor death [1]. In adults, PLCs include hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA) in proportions of 90% and 5–10%, respectively [2]. HCC is the third most common cause of cancer-related deaths worldwide, and CCA is the second most prevalent type of liver cancer [3,4]. Despite efforts in basic and clinical research, the five-year survival rate of patients affected with PLCs is less than 5%, since diagnosis is often delayed and is associated with advanced stage [5]. Therefore, it is of fundamental importance to reduce incidence and impact of these diseases through knowledge of risk factors and primary prevention strategies.
Different risk factors are involved in HCC and CCA pathogenesis and are related to diet, environmental exposure, lifestyle, and genetic predisposition [6,7,8]. Indeed, the interaction of liver tissue with infectious agents through the course of life or the detrimental impact of frequent and extreme biological events exceeding adaptive capacity, may determine numerous disorders predisposing humans to the onset of PLC [6,9,10]. Pathological liver conditions that predispose humans to PLCs include metabolic dysfunction-associated fatty liver disease (MAFLD), chronic hepatitis, alcoholic cirrhosis and non-alcoholic fatty liver disease (NAFLD) [9].
Patients affected with NAFLD show a higher risk of developing bacterial, fungal, and viral infections, but the exact mechanism by which this occurs is still unknown [11]. Hepatitis B virus (HBV) infection could stimulate NAFLD progression to liver steatosis, through the induction of cholesterol synthesis gene expression [12]. Even though the prevalence of HCC from NAFLD is less common than that from chronic viral hepatitis B and C, hepatocellular carcinoma usually arises in the setting of chronic liver damage and cirrhosis [10]. Indeed, both hepatitis C virus (HCV) and HBV are associated with HCC development, whereas HCC related to other underlying hepatic diseases is linked to the onset of cirrhosis [6].
Multiple risk factors have been linked to the development of CCA. These include hepatic parasites, chronic biliary tract, and liver diseases, including primary sclerosing cholangitis, choledochal cysts, Caroli’s disease and HCV cirrhosis, as well as lifestyle factors associated with chronic hepatic inflammation and cholestasis [13,14,15,16]. CCA occurs along the biliary tree and is classified into intrahepatic, perihilar, or distal [17]. The prevalence of CCA varies in different geographical areas, because of environmental and genetic conditions. This disease is observed after the fourth decade of life and more often in men [16].
In recent decades, a clear causal link has been found between liver damage, inflammation and regeneration and the occurrence of PLCs. Chronic inflammation determined by long-term infections is observed in more than 80% of the HCCs and represents an important factor able to modify the hepatic microenvironment [2]. The liver, which receives portal and systemic circulation, contains about one third of the reticuloendothelial system and acts as a defense against infection [18]. This allows the liver to maintain the equilibrium between host immune system activation and tolerance to circumvent inappropriate immune responses against non-pathogenic exogenous molecules, such as food-antigens [19]. The involvement of the gut–liver axis in the pathophysiological mechanism responsible for the development of HCC has recently been revealed [20,21,22]. The gut barrier integrity protects the host from the microbiota residing in the intestinal ecosystem. If this barrier is leaky or disrupted and/or in the presence of gut dysbiosis, the liver is exposed to factors from the intestine, such as bacteria and bacterial endotoxins (e.g., LPS) that can lead to hepatic diseases [23,24]. Increased bacterial translocation from the gut could be a key factor associated with HCC development in animal models, although data coming from human studies are still incomplete [22]. Moreover, depletion of the gut microbiota in mice could reduce T cell antiviral immunity in the liver, and protract HBV infection [25]. Various microorganisms can exert different clinical effects on the liver, such as increasing levels of aminotransaminases and causing acute liver failure and hepatic fibrosis, rapidly progressing to cirrhosis [19,26]. Significant modifications occur in tissue metabolism throughout inflammatory and immune responses. They consist of local reduction of nutrients, increased oxygen consumption, and enhanced generation of reactive oxygen and nitrogen intermediates. In addition, evolutionary adaptations allow pathogens to utilize host metabolic pathways and survival mechanisms [27].
The Wingless/It (Wnt)/β-catenin signaling pathway is a pathway preserved throughout animal evolution. It is involved in the regulation of cells’ differentiative fates both during embryonic development and in the homeostasis of tissue regeneration in the adult organism [28,29]. Given its involvement in both stem capacity and cell differentiation, alterations that overregulate the functions of the Wnt signaling are often observed in tumors, particularly in those tissues that physiologically depend on it for their ability to renew or repair themselves [30]. The signal is activated by a family of Wnt ligands consisting of 19 secreted glycoproteins that can act in both autocrine and paracrine manners, binding to frizzled (Fzd) receptor complexes on the cell membrane to transduce downstream intracellular signals [31]. At the cytoplasmic level, the signal network is defined according to whether it is dependent on its key mediator, β-catenin; thus, the pathway is divided into canonical (β-catenin-dependent) or non-canonical (β-catenin-independent) [32]. β-catenin, as a scaffold protein, can link the cytoplasmic tail of E-cadherin to the actin of the cytoskeleton [33]. In the presence of receptor stimulation by Wnt ligands, cytoplasmic and nuclear levels of β-catenin increase due to the reduction of its proteasomal degradation. The process is controlled by a multi-protein destruction complex consisting of Axin, Adenomatous polyposis coli (APC), Casein kinase1a (CK1a) and Glycogen synthase kinase 3-β (GSK3-β) [34] (Figure 1). The continuous elimination of β-catenin prevents it from reaching the nucleus and the Wnt target genes are then repressed by the DNA-bound T cell factor/lymphoid enhancer factor (TCF/LEF) proteins [35] (Figure 1).
Wnt signaling plays key physiological functions in the liver, from development to metabolic zonation and regeneration after damage [36,37]. Specifically, β-catenin and its negative regulator APC are localized in the perivenous and periportal areas, where they control liver metabolic activity [38]. Molecular dysregulation of the Wnt pathway is strongly associated with the hepatic microenvironment and may drive the progression from precancerous dysplasia to PLC metastasis [26,39,40,41]. This is a common condition also reported in liver metastasis from colorectal cancer [42].
Upregulation of Wnt ligands and their receptors/coreceptors, as well as downregulation of Wnt/β-catenin signaling inhibitors, have been reported in liver cancer and its adjacent precancerous lesions [43]. Hepatic tumors recurrently show mutations in genes encoding key elements of the Wnt/β-catenin pathway [26,39,40]. Mutations of the AXIN and APC genes, and the CTNNB1 gene encoding β-catenin, have been detected in PLCs [41,44,45,46].
Infectious agents implicated in PLC development can modulate Wnt pathway molecular components to destabilize normal cell functions [47]. In fact, oncogenic viruses and bacteria toxins often activate an aberrant Wnt signaling that promotes malignant transformation in target cells [24,47].
A better understanding of the events that lead to dysregulation of the Wnt signal following infection by microbial agents in PLCs could lead to a targeted therapeutic approach in liver cancer treatment. Therefore, it is important to identify the infectious agents that alter the Wnt pathway in liver cells, with the aim to understand the mechanisms used by bacteria or viruses or parasites to modify the Wnt signal and evaluate their involvement in the pathogenesis of HCC and CCA. This review considers what is currently known about deregulation of Wnt signaling due to the presence of infectious agents in the hepatic microenvironment as a driver of liver carcinogenesis. Comprehension of the cellular and molecular events occurring in the microenvironment in the presence of infectious agents could improve targeted therapeutic approaches in the treatment of PLCs.

2. Infectious Agents Involved in HCC

2.1. Hepatitis B Virus (HBV)

In approximately 80% of HCCs, the development of disease is associated with HBV and/or HCV chronic infections [48]. Without any doubt, the vaccination and therapeutic treatment of hepatitis B, in addition to programs to prevent HBV and HCV transmission, are currently changing the epidemiology of HCC, although its non-viral etiology is increasing [49,50,51]. HBV seroprevalence shows the highest incidence rate in Africa and Western Pacific Asia, while chronic HBV disease is decreasing in many European countries; males over 40 years of age are most affected worldwide [52,53]. From a molecular point of view, Wnt is one of the main pathways modulated by HBV [54] (Table 1). Indeed, regulatory hepatitis B Viral X protein (HBx) and hepatitis B surface antigen (HBsAg) can activate Wnt/β-catenin signaling components [55]. In particular, HBx protein regulates the expression of the secreted frizzled-related proteins, SFRP1 and SFRP5, which antagonize Wnt This occurs through hypermethylation of their gene promoter by the DNA methyltransferases DNMT1 and DNMT3 [56]. In addition, HBx impairs the cell destruction complex through multiple mechanisms [41], such as the competitive binding to the β-catenin domain of APC, or the Src kinase activation that suppresses GSK-3β, or the induction of the androgen receptor pathway by cell-cycle-related kinase [41,57]. Furthermore, HBx increases the expression of ETS variant 4 (ETV4) through Disheveled 2 (DVL2) transcriptional activation and consequent inhibition of β-catenin degradation; this Wnt deregulation is associated with HCC progression and poor prognosis [58]. ETV4 also occurs in Wnt/β-catenin pathway activation through the upregulation of Annexin A2 (ANXA2) in HBV-liver cancers [59]. On the other hand, HBsAg upregulates the expression of LEF-1 transcription factor in HCC cell models and tissues [60,61]. A recent study shows that the intermediate protein p22, a precursor to HBVe antigen (HBeAg), can activate TCF/β-catenin transcription in in vitro and in vivo models [62]. Regarding the role played by gene mutations, changes in Hbx modulate the expression of Wnt-5a in hepatoma cells, whereas inactivating mutations in APC, AXIN1 and AXIN2 genes or methylation in APC aberrantly modify the Wnt signaling response [63,64]. In HBV-dependent HCC, AXIN1 mutations are less frequently associated with CTNNB1 ones than those found in HCV-related HCC [39,65]. Polymorphisms in AXIN1, AXIN2, CTNNB1, and WNT2 genes seem to confer a different HBV-dependent HCC susceptibility and progression [66]. In addition, WNT2 and WNT1 gene expression can be considered as diagnostic or prognostic biomarkers, respectively, while Wnt-3a protein expression could be a novel HCC target in early diagnosis [67,68]. Activation of the Wnt/β-catenin pathway could also be induced by integration of the HBV genome into long interspersed nuclear element 1 (LINE1) with production of an oncogenic chimeric transcript, although this hypothesis is not fully confirmed [64,69]. HBV can also indirectly influence the Wnt signaling in hepatocarcinogenesis through the modulation of the expression of different miRNAs, such as miR-26a, miR-15a, miR-16-1, miR-148a, miR-132, miR-122, miR-34a, miR-21, miR-29a, miR-222 and miR-119a/b-3p [70]. In HBV-related HCCs and matched adjacent non-cancerous liver tissues, a differential regulation of the Wnt-β-catenin pathway, but no variability of protein levels or phosphorylation status, was identified by integrated proteogenomic profiling [71]. Recently, mRNAs of some Wnt target genes in HBV-associated/CTNNB1-mutant and TP53-mutant HCCs were found to be differentially expressed and associated with the loss of epithelial phenotype. This condition most likely resulted from altered phosphorylation of proteins implicated in the actin filament organization or decreased expression of the epithelial markers E-cadherin (CDH1) and Keratin-19 (KRT19). CTNNB1-mutant HCCs are often low grade and linked with the immune-desert phenotype, while TP53-mutant HCCs are related to HBV infection [72]. In addition, the presence of HBV can affect normal liver cell processes when it crosses a developmental pathway, such as Wnt/β-catenin [26,47].

2.2. Hepatitis C Virus (HCV)

Chronic HCV infection affects an estimated 115 million people worldwide; this disease is more common in East/Central Asia and North Africa than in Europe [53].
HCV core protein, non-structural protein 4B (NS4B) and 5A (NS5A) are able to activate Wnt/β-catenin signaling in HCC pathogenesis (Table 1). The core protein upregulates Wnt ligands, Fzd and LRP5/6 receptors, increases the TCF-dependent transcription stimulated by Wnt3a and downregulates Wnt antagonists Dickkopf (DKK), SFRP1 and E-cadherin [73,74,75,76]. HCV core protein can reduce E-cadherin expression levels due to CDH1 gene promoter hypermethylation, with consequent dissociation of the β-catenin/E-cadherin complex, and stimulates tumor cell growth by GSK-3β inactivation and Wnt3a release [76,77]. A study performed on core proteins of HCV subtypes from Cambodia, Romania and Cameroon detected a critical role of the core region encompassing amino acids Ser64 and Thr71 in the upregulation of Wnt target genes c-MYC and Cyclin D1 (CCND1), and different nuclear levels of β-catenin in cells infected, according to various HCV subtypes [78].
NS4B activates the Wnt3a-induced Wnt/β-catenin signaling in HCC Huh7 cells and normal human liver LO2 cell lines [77,79]. NS5A stabilizes β-catenin and stimulates its transcriptional activity with a mechanism mediated by phosphoinositide 3-kinase (PI3K)-AKT pathway activation and subsequent inactivation of GSK-3β [80,81,82].
The progression from chronic HCV hepatitis to HCC is favored by binding of Wnt1 and Wnt5a to Fzd, which transactivates the EGFR pathway [83]. In a model of HCV infection in vitro, activation of Wnt/β-catenin signaling remains even after antiviral treatment and is related to GSK-3β inhibition [84]. In HCV-related HCC, FGF signaling induces the release of phosphorylated β-catenin from the complexes with E cadherin [83].
Downregulation of APC or Axin2 induced by HCV increases de-phospho-β-catenin, which translocates to the nucleus to activate the target genes and induce malignant changes in hepatocytes [85]. Chronic HBV and HCV hepatitis shows increased activity of cyclooxygenase 2 (COX2) that in turn activates the Wnt pathway through the release of prostaglandin E2 [86]. HCV also downregulates APC and Axin2, while it upregulates miR-155, which promotes Wnt signaling activation [87]. Conversely, miR-125b correlates with APC through suppression of cell growth, arrest of cell cycle at G1 phase, and inhibition of HCC cell proliferation, migration and invasion [88].

2.3. Hepatitis D Virus (HDV)

HDV is a defective RNA virus that needs co-infection with HBV. At a global level, approximately 5% (15–20 million) of chronic HBV carriers are co-infected with HDV. HDV infection may occur with HBV co-infection or superinfection in persistent HBV hepatitis [89]. HDV produces the small and large hepatitis delta antigens (S-HDAg and L-HDAg, respectively). Nuclei of Huh-7 cells transfected with the L-HDAg expression plasmid show β-catenin immunoreactivity. L-HDAg activates the transforming growth factor-β (TGF-β), which may induce epithelial–mesenchymal transition (EMT) and fibrosis [90].

2.4. Epstein–Barr Virus (EBV)

EBV has a high global prevalence and is present in more than 90% of adults [91]. In HCV-related HCC, EBV coinfection may promote carcinogenesis through increased HCV replication and inflammation [92,93,94]. Research has not confirmed the direct involvement of EBV in liver carcinogenesis [95,96,97]. A more recent study detected the frequent activation of EBV in infiltrating lymphocytes of clinically low aggressive HCCs [94]. Based on results coming from other cancers, EBV might induce epigenetic reprogramming of LEF1 and Wnt5a. In other terms, EBV might indirectly contribute to hepatocarcinogenesis [98,99]. In light of this preliminary evidence, it might be interesting to investigate the role of EBV in the dysregulation of Wnt signaling in HCC. Viral interference, which can inhibit both GSK-3 and promote β-catenin functions, has been observed with DNA virus-encoded proteins, such as the X protein of HBV and the EBV LMP2A protein [100].

2.5. Human Papilloma Virus (HPV)

Prevalence and distribution of over 207 HPV genotypes vary according to different populations and geographical areas [101]. Some reports have investigated the role of HPV-16 in hepatocarcinogenesis by detecting the common involvement of E6 oncoprotein and HBx in AP-1 activation to increase HBV transcriptional activity in human liver cells [102]. E6 and E7 oncoproteins are involved in the regulation of the Wnt/β-catenin pathway, which results in hyperactivated HPV-associated cancers [103,104]. There is currently no further experimental evidence on the role of HPV in liver cancer.

2.6. Non-Viral Infectious Agents Involved in HCC

Regarding the role of non-viral agents in liver carcinogenesis, it has been reported that Toxoplasma gondii, a protozoan parasite, is involved in liver disease and fibrosis, with a global distribution. Indeed, in China, the highest seroprevalence of T. gondii was found among patients with liver disease, who ranged in age between 41–50 years [105]. T. gondii secretes GRA18, a protein exported to the cytoplasm of infected cells, in which it is a potential inhibitor of the β-catenin destruction complex [106]. In the presence of GRA18, β-catenin moves towards the nucleus where it induces the expression of anti-inflammatory chemokines Ccl17, Ccl22, and Ccl24 [106]. Echinococcus granulosus seems to significantly extend the survival and play a protective effect in HCC patients [107]. The presence of Mycoplasma hyorhinis, which predominantly infects swine, has been also detected in human cancer tissues [108]. M. hyorhinis infection promotes HCC progression and cell migration. The interaction between mycoplasma p37 membrane protein and the epithelial cell adhesion molecule (EpCAM) involves the Akt/mTOR pathway [109], which in turn can control Wnt/β-catenin signaling. Since in gastric cancer M. hyorhinis infection has been reported to induce nuclear β-catenin accumulation and increase its target gene expression [110], further research is needed to assess the direct involvement of M. hyorhinis in hepatocarcinogenesis, by Wnt signaling dysregulation.
In some countries, such as India, Mexico, Pakistan, Sudan, and Saudi Arabia, Aspergillus flavus has a high prevalence and is frequently isolated in the respiratory tract [111]. Aflatoxin-B1 (AFB1) mycotoxin, produced by Aspergillus fungi (flavus), is a genotoxic compound responsible for HCC. Persistent AFB1 exposure activates the Wnt pathway [112]. Other studies have revealed the role of AFB1 in miR-33 and miR-34a activation to downregulate the Wnt/β-catenin pathway in HCC cells, suggesting an involvement for these miRNAs in therapeutic applications [113,114]. Moreover, macrophages infected with A. flavus, A. fumigatus, and Candida albicans strongly activate Wnt signaling [115]. These contradictory data indicate that probably other unknown factors are involved in the modulation of Wnt signaling by mycotoxins produced by A. flavus. Finally, AFB1 may increase the HCC risk in the presence of HBV or HCV infections [116,117].

3. Infectious Agents Involved in CCA

The onset of CCA is related to a chronic inflammatory condition due to long exposure of the cholangiocytes to predisposing risk factors, such as transmissible agents (Table 1). In bile ducts, this persistent phlogistic state is responsible for the induction of several pathways, including Wnt signaling, as well as the activation of cell proliferation and CCA-predisposing genetic mutations and epigenetic changes [118].
In this tumor, mutations in the CTNNB1 (1.5%), AXIN1 (4%) and APC (2%) genes are the most commonly identified [41]. The host response to infection causes differential expression of Wnt signaling molecules, in particular overexpression of canonical ligands such as Wnt7b and non-canonical ligands such as Wnt5a, in infiltrating macrophages activated in both inflammation and cancer [99,119]. Moreover, macrophages determine the secretion of pro-inflammatory cytokines activating Wnt5a [120]. Tumor-associated macrophages (TAMs) are implicated in CCA cell proliferation, metastasis, and angiogenesis as well as in the activation of Wnt/β-catenin signaling [121]. In fact, this pathway contributes to CCA induction, progression, EMT and multidrug resistance [26]. Other signaling pathways, such as PI3K/AKT/PTEN/GSK-3β, in association with microRNA upregulation, are involved [122].

3.1. Hepatitis B Virus (HBV)

HBV genes are present in the genome of CCA. HBx protein may be implicated in cholangiocarcinogenesis, since HBx gene insertion induces cis-activation of Telomerase reverse transcriptase (TERT) mRNA transcription in CCA cell lines [123]. An aberrant activation of Wnt/β-catenin signaling was observed in carcinogen-treated HBx transgenic mice, which developed mixed HCC and CCA in the same liver [124].

3.2. Hepatitis C Virus (HCV)

Differing from HBV-associated CCA, HCV infection is associated with a poor prognosis of this type of cancer [83,125]. Molecular mechanisms inducing CCA after HCV infection are still unclear [126]. To the best of our knowledge, there is not enough scientific evidence regarding the involvement of the Wnt pathway in the pathogenesis of HCV-induced CCA, and there is a lack of studies on a broader case series. Analysis of few samples of CCA and hepatocholangiocarcinoma showed no mutations in TERT promoter and CTNNB1 exon 3 [127].

3.3. Human Polyomavirus (HPyV)

In CCA lesions and adjacent peritumoral cells of bile duct epithelium, the occurrence of human polyomavirus 7 (HPyV7) is more frequent than human polyomavirus 6 (HPyV6) and oncogenic Merkel cell polyomavirus (MCPyV) [128].

3.4. Epstein–Barr Virus (EBV)

EBV can synergistically cooperate with HBV in the onset of a rare subtype of intrahepatic CCA (EBVaICC), a tumoral form distinguished from others by exclusive clinicopathological and genetic alterations [129] and probably with a different pathogenesis that does not seem to involve Wnt signals.

3.5. Non-Viral Infectious Agents Involved in CCA

Mechanisms leading to CCA development are dependent on genetic alterations induced by different trematodes. Liver flukes such as Clonorchissinensis, Opisthorchis viverrini and Opisthorchis felineus are trematodes of the family Opisthorchiidae. In particular, the genera Clonorchis and Opisthorchis are responsible for CCA [130,131].
C. sinensis determines clonorchiasis, which is correlated with hyperplasia of biliary epithelium and metaplasia of mucin-secreting cells in bile ducts, probably through upregulation of Wnt7b, Fzd6 and cell cycle controllers, which leads to CCA development [132]. An excretory/secretory (ES) product of C. sinensis, the growth factor-like protein termed granulin, seems to be involved in cell migration and invasion in CCA as well as in HCC, with activation of EMT by upregulation of β-catenin and decline of E-cadherin [133]. To support this, in a mouse model of C. sinensis infection, macrophages showed variations in polarization and phenotype during different phases of disease [121]. On the other hand, ES products of O. viverrini contain growth factors and other molecules, including liver fluke granulin. In Opisthorchiasis, chronic inflammation is induced by mechanical damage to biliary epithelia caused by O. viverrini, which continuously exposes cholangiocytes to granulin and other molecules [134]. The chronic inflammation may act through receptor tyrosin kinase (RTK) signaling, which is related to MAPK, PI3K/AKT and Wnt/β-catenin pathways [135].
In particular, the development of opisthorchiasis-dependent CCA is associated with upregulation of the Wnt/β-catenin signaling pathway. In cholangiocarcinogenesis of O. viverrini-infected hamsters, increased expression of Wnt3, Wnt3a, Wnt5a, Wnt7b as well as β-catenin nuclear translocation were found [136]. Similarly, these molecules were also upregulated in human CCA tissues. On the other hand, some Wnt ligands (i.e., Wnt3a) released into the tumor microenvironment could be produced by both CCA and inflammatory cells [137].
Cryptosporidium parvum has been isolated in human immunodeficiency virus (HIV) cholangiopathy. C. parvum induces a Toll-like receptor 4-dependent response that is inhibited by HIV-1 trans-activator of transcription (Tat) protein [138]. Possible retrograde invasion of C. parvum from the small intestine to biliary ducts favors a pro-inflammatory cholangiopathy that involves molecules such as micro-RNA let-7i, NF-kB p50-CCAAT/enhancer-binding protein beta (C/EBPbeta) and NRAS [139]. Moreover, HIV-1 Tat increases apoptosis in cholangiocytes induced by C. parvum through activation of the Fas/Fas ligand pathway [140]. In individuals with CD40L deficiency, C. parvum infection predisposes humans to sclerosing cholangitis, which may evolve to CCA [141]. Wnt/β-catenin signaling interferes with maturation and activation of dendritic cells in the tumor microenvironment. Inflammatory dendritic cells express elevated levels of costimulatory molecules, including CD40 [142].
Helicobacter pylori was found to be associated with opisthorchiasis and hepatobiliary disease [143]. It induces the Wnt/β-catenin pathway and macrophage recruitment [144].
Salmonella typhi. Chronic carriers for S. typhi show a 200-fold risk of developing CCA [131]. In intestinal stem cells of a murine model, Salmonella enterica serovar Typhimurium increases mRNA expression of Wnt2, Wnt3, Wnt6, Wnt9a, Wnt11 as well as Fzd2, Fzd4, Fzd6, Fzd7, Fzd8, and Fzd9 [145,146,147]. S. enterica serovar Typhimurium could dysregulate the Wnt/β-catenin signaling by the translocation of antigens through the gut–vascular barrier, with a mechanism based on its pathogenicity island (Spi) 2–encoded type III secretion system that permits its dissemination in the liver and bloodstream [148]. Induction of Wnt2 and Wnt11 expression has been partially ascribed to Salmonella AvrA, which is a bacterial effector involved in the control of β-catenin ubiquitination and stabilization [99]. These data suggest a deep role of Salmonella in the activation of the Wnt pathway in CCA.
Table 1. Summary of infectious agents interfering with Wnt/β-catenin signalling in hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA).
Table 1. Summary of infectious agents interfering with Wnt/β-catenin signalling in hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA).
Diseases
and Pathogens
Mode of Entry
into the Liver
Wnt Pathway DeregulationDeterminantsReferences
HCC
Hepatitis B VirusbloodstreamSFRP1 and SFRP5 downregulation. Competitive binding to β-catenin domain of APC; GSK3-β suppression. LEF-1 upregulation. Activation of TCF/β-catenin transcription. Inactivating mutations in APC, AXIN1 and AXIN2. APC methylation. Polymorphisms in AXIN1, AXIN2, CTNNB1, and WNT2. HBx
HBsAg
p22
[41,54,57,60,61,62,63,64]
Hepatitis C VirusbloodstreamWnt, Fzd and LRP5/6 overexpression; increased TCF-dependent transcription; DKK and SFRP1 downregulation. CDH1 hypermethylation. Wnt3a induction. Stabilization of β-catenin by GSK-3β inactivation. β-catenin-EGFR pathway transactivation by Wnt1 and Wnt5a. APC or Axin2 downregulation. Wnt signaling activation by miR-155 upregulation. HCV core protein
NS4B
NS5A
[73,74,75,76,77,79,81,82,83,85,87,90]
Hepatitis D virusbloodstreamβ-catenin increased expression L-HDAg[90]
Human papilloma viruspossibly bloodstreamWnt/β-catenin pathway
hyperactivation
E6
E7
[103,104]
Toxoplasma gondiimesenteric and
portal vessels
β-catenin destruction complex inhibitionGRA18 [106]
Aspergillus flavusmesenteric and
portal vessels
Wnt pathway activation.
Wnt/β-catenin signaling downregulation by AFB1 activated by miR-33a and miR-34a
AFB1[112,113,114]
CCA
Hepatitis B Virus Aberrant activation of Wnt/β-catenin signalingHBx[124]
Hepatitis C Virus Unknown
Human polyomavirus 6
Human polyomavirus 7
Merkel cell polyomavirus
Unknown
Epstein-Barr virus Unknown
Clonorchis sinensisretrograde invasion from the small intestine to biliary ductsWnt7b and Fzd6 overexpression. β-catenin upregulationGranulin[132,133]
Opisthorchis viverriniretrograde invasion from the small intestine to biliary ductsWnt3, Wnt3a, Wnt5a, Wnt7b, and β-catenin overexpressionGranulin[136]
Cryptosporidium parvumretrograde invasion from the small intestine to biliary ductsWnt/β-catenin signaling affects dendritic cells maturation and activation in tumor microenvironmentUnknown[142]
Salmonella Typhihematogenous
seeding in the liver during bacteremic phase, and reticuloendothelial cells infection
Wnt2, Wnt3, Wnt6, Wnt9a, Wnt11 and Fzd2, Fzd4, Fzd6, Fzd7, Fzd8, Fzd9 mRNA upregulation. Wnt2 and Wnt11 expression inductionAvrA[99,145,146,147]
Hepatitis B Viral X protein (HBx); hepatitis B surface antigen (HBsAg); Hepatitis C Virus (HCV) core protein; non-structural protein 4B (NS4B); non-structural protein 5A (NS5A); large hepatitis delta antigen (L-HDAg); E6 oncoprotein; E7 oncoprotein; dense granule (GRA) protein (GRA18); aflatoxin-B1 (AFB1); Salmonella bacterial type III secretion effector protein AvrA.

4. Therapeutic Perspectives

Kinases involved in the Wnt signaling pathway could represent attractive molecular targets for their inhibitors and antiviral agents in the treatment of infectious agents responsible for liver cancer. A novel therapeutic target in HCV-infected patients could be GSK-3, which constitutively phosphorylates cytosolic β-catenin. GSK-3 is expressed as GSK-3α and GSK-3β isozymes, which are structurally similar in catalytic domains [149]. Under physiological conditions, GSK-3 activity is inhibited through phosphorylation of its serine residues by several kinases, such as protein kinase A (PKA), protein kinase C, Akt. These changes lead to Wnt cascade attenuation and maintenance of cellular homeostasis. Conversely, overexpression of GSK-3 or imbalance of its phosphorylation status induce overactivity of the enzyme [149,150]. HCV infection increases cyclic adenosine monophosphate (cAMP) levels necessary for PKA activation. Therefore, the use of endoplasmic reticulum (ER) stress/PKA/GSK-3β-dependent Wnt/β-catenin signaling as targets of PKA inhibitor could constitute a new therapeutic approach for patients infected with HCV, in which the Wnt/β-catenin pathway is activated despite treatment with direct-acting antiviral agents (DAAs) and HCV clearance [150]. Another therapeutic strategy could be based on the reduction of GSK3β in tumor-associated macrophages (TAMs) to prevent HCC development through the inhibition of M2 phenotype and increase the sensitivity of anti-programmed death 1 (PD1) immunotherapy by the decrease of Programmed Death-Ligand 1 (PD-L1) ubiquitination [151].
Other interesting therapeutic perspective is represented by targeting of the β-catenin destruction complex (APC, Axin, CK1, and GSK3β) with the aim to induce the inactivation of Wnt signaling in HCC. The protein destruction complex induces degradation through CK1- and GSK3-mediated phosphorylation of β-catenin, followed by enrollment of the beta-transducin repeats-containing protein (β-TrCP) ligase [43]. Targeting the intracellular protein complex is difficult since APC mutations are often present. Stabilization of the β-catenin protein destruction complex allows inactivation of the Wnt pathway. Tankyrase (TNKS) is an activator of the Wnt/β-catenin cascade since it degrades the negative regulators of β-catenin, AXIN1 and AXIN2, through proteasomal degradation mediated by ubiquitin [43,152]. TNKS inhibitors negatively regulate Wnt signaling by stabilizing AXIN and antagonizing the Wnt/β-catenin pathway. Among them, XAV939 and WXL-8 reduce Wnt/β-catenin cascade and inhibit HCC cell proliferation. NVP-TNKS656 inhibits HCC cell line proliferation, as well as invasion, metastasis, and EMT features [153]. Indeed, inhibitors of TNKS could represent a novel attractive therapeutic target in liver cancers induced by infective microorganisms. Additional molecules act on the protein destruction complex at different levels. Pyrvinium blocks Wnt signaling through the increase of CK1 kinase activity, whereas the non-steroidal anti-inflammatory drug Sulindac binds to Dvl [154]. Currently, these therapies are still in the experimental stage, and it is hoped that they can be used in the treatment of primary liver cancers with infectious etiology. Deubiquitinase inhibitors may represent an alternative therapeutic option to reduce β-catenin levels by promoting its degradation [155]. In their evolution, pathogens have developed molecular signals that enable them to survive host defenses. Recent studies have unveiled the role of the ubiquitin system in innate immunity pathways in controlling responses to infectious agents. The ubiquitination process is involved in post-translational regulation of intracellular proteins through the control of their degradation via the proteasome system [156,157]. Deubiquitinating enzymes (DUB) are a large group of proteases implicated in deubiquitylation, which can rescue labelled substrate proteins by removing their conjugated ubiquitin chains [158]. Recent findings have revealed the role of viral-derived DUBs in counteracting host immune responses [159] and DUBs have also been proposed as possible therapeutic targets for the treatment of infectious diseases [160,161]. Viral-derived DUBs are not structurally homologous to those of the host, but they can act on the same target proteins [162,163]. In the case of viruses, the production of DUBs prevents them from being eliminated by host cells.
The identification of microbial DUBs, capable of interfering with the molecular pathways of the host cell, could be important for the development of inhibitory drugs. Therefore, it would be interesting to understand whether infectious agents, related to the occurrence of primary liver tumors, produce DUBs that inhibit the proteasomal degradation of β-catenin.

5. Conclusions

Pathogens alter Wnt-associated processes to increase both infection and survival in the human host. In conserved eukaryotes, Wnt signaling is involved in the interplay between the human host and extracellular and intracellular bacterial pathogens [164]. These aspects have important implications in the knowledge of microbic carcinogenesis, especially in the human liver. Since the Wnt/β-catenin pathway is considered a driver of viral and bacterial tumors, the development of targeted molecules that are able to inhibit this intracellular signaling might improve the therapeutic approaches of patients with PLCs. In recent years, studies have focused on the mechanisms used by GSK-3 and β-catenin to control the antiviral innate immune response toward RNA and DNA virus infections. Targeting the GSK-3/β-catenin axis could avoid harmful effects consequent to chronic infection, such as viral oncogenesis. Nevertheless, detection of small molecules inhibiting the interaction of β-catenin with TCF and TNKS has not given the expected results [100]. DUB inhibitors that reduce the levels of β-catenin by promoting its degradation [156] represent an alternative therapeutic option.
A better insight into the molecular mechanisms leading to dysregulation of Wnt signaling during infections could open up new perspectives in clinical practice with integrated therapies in PLC patients.

Author Contributions

Conceptualization, T.C. and G.M.A.; resources, D.L.E. and G.M.A.; data curation, T.C., F.S. and D.L.E.; writing—original draft preparation, T.C.; writing—review and editing, G.M.A., F.S. and D.L.E.; supervision, R.C. and G.M.A.; funding acquisition, R.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the “G. d’Annunzio” University of Chieti-Pescara (Fondi di. Ateneo per la Ricerca–F.A.R.) grant 2020 to R.C.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rumgay, H.; Arnold, M.; Ferlay, J.; Lesi, O.; Cabasag, C.J.; Vignat, J.; Laversanne, M.; McGlynn, K.A.; Soerjomataram, I. Global burden of primary liver cancer in 2020 and predictions to 2040. J. Hepatol. 2022, 77, 1598–1606. [Google Scholar] [CrossRef]
  2. Kumar, M.; Zhao, X.; Wang, X.W. Molecular carcinogenesis of hepatocellular carcinoma and intrahepatic cholangiocarcinoma: One step closer to personalized medicine? Cell Biosci. 2011, 1, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Zhuo, Y.; Chen, Q.; Chhatwal, J. Changing Epidemiology of Hepatocellular Carcinoma and Role of Surveillance. In Hepatocellular Carcinoma: Translational Precision Medicine Approaches; Hoshida, Y., Ed.; Humana Press: Cham, Switzerland, 2019; Chapter 3. [Google Scholar] [CrossRef]
  4. Shin, D.W.; Moon, S.H.; Kim, J.H. Diagnosis of Cholangiocarcinoma. Diagnostics 2023, 13, 233. [Google Scholar] [CrossRef]
  5. Anwanwan, D.; Singh, S.K.; Singh, S.; Saikam, V.; Singh, R. Challenges in liver cancer and possible treatment approaches. Biochim. Biophys. Acta Rev. Cancer 2020, 1873, 188314. [Google Scholar] [CrossRef] [PubMed]
  6. Shiani, A.; Narayanan, S.; Pena, L.; Friedman, M. The Role of Diagnosis and Treatment of Underlying Liver Disease for the Prognosis of Primary Liver Cancer. Cancer Control. 2017, 24, 1073274817729240. [Google Scholar] [CrossRef] [Green Version]
  7. Yang, W.S.; Zeng, X.F.; Liu, Z.N.; Zhao, Q.H.; Tan, Y.; Gao, J.; Li, H.L.; Xiang, Y.B. Diet and liver cancer risk: A narrative review of epidemiological evidence. Br. J. Nutr. 2020, 124, 330–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Wang, G.; Heij, L.R.; Liu, D.; Dahl, E.; Lang, S.A.; Ulmer, T.F.; Luedde, T.; Neumann, U.P.; Bednarsch, J. The Role of Single-Nucleotide Polymorphisms in Cholangiocarcinoma: A Systematic Review. Cancers 2022, 14, 5969. [Google Scholar] [CrossRef] [PubMed]
  9. Polyzos, S.A.; Kang, E.S.; Tsochatzis, E.A.; Kechagias, S.; Ekstedt, M.; Xanthakos, S.; Lonardo, A.; Mantovani, A.; Tilg, H.; Côté, I.; et al. Commentary: Nonalcoholic or metabolic dysfunction-associated fatty liver disease? The epidemic of the 21st century in search of the most appropriate name. Metabolism 2020, 113, 154413. [Google Scholar] [CrossRef] [PubMed]
  10. Negro, F. Natural history of NASH and HCC. Liver Int. 2020, 40, 72–76. [Google Scholar] [CrossRef] [Green Version]
  11. Adenote, A.; Dumic, I.; Madrid, C.; Barusya, C.; Nordstrom, C.W.; Rueda Prada, L. NAFLD and Infection, a Nuanced Relationship. Can. J. Gastroenterol. Hepatol. 2021, 2021, 5556354. [Google Scholar] [CrossRef]
  12. Tourkochristou, E.; Assimakopoulos, S.F.; Thomopoulos, K.; Marangos, M.; Triantos, C. NAFLD and HBV interplay-related mechanisms underlying liver disease progression. Front. Immunol. 2022, 13, 965548. [Google Scholar] [CrossRef] [PubMed]
  13. He, S.; Tang, S. WNT/β-catenin signaling in the development of liver cancers. Biomed. Pharmacother. 2020, 132, 110851. [Google Scholar] [CrossRef] [PubMed]
  14. Massironi, S.; Pilla, L.; Elvevi, A.; Longarini, R.; Rossi, R.E.; Bidoli, P.; Invernizzi, P. New and Emerging Systemic Therapeutic Options for Advanced Cholangiocarcinoma. Cells 2020, 9, 688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Sarcognato, S.; Sacchi, D.; Fassan, M.; Fabris, L.; Cadamuro, M.; Zanus, G.; Cataldo, I.; Capelli, P.; Baciorri, F.; Cacciatore, M.; et al. Cholangiocarcinoma. Patologica 2021, 113, 158–169. [Google Scholar] [CrossRef]
  16. Kirstein, M.M.; Vogel, A. Epidemiology and Risk Factors of Cholangiocarcinoma. Visc. Med. 2016, 32, 395–400. [Google Scholar] [CrossRef]
  17. Brindley, P.J.; Bachini, M.; Ilyas, S.I.; Khan, S.A.; Loukas, A.; Sirica, A.E.; The, B.T.; Wongkham, S.; Gores, G.J. Cholangiocarcinoma. Nat. Rev. Dis. Prim. 2021, 7, 65. [Google Scholar] [CrossRef]
  18. Talwani, R.; Gilliam, B.L.; Howell, C. Infectious diseases and the liver. Clin. Liver Dis. 2011, 15, 111–130. [Google Scholar] [CrossRef] [Green Version]
  19. Jenne, C.N.; Kubes, P. Immune surveillance by the liver. Nat. Immunol. 2013, 14, 996–1006. [Google Scholar] [CrossRef]
  20. Das, B.K. Altered gut microbiota in hepatocellular carcinoma: Insights into the pathogenic mechanism and preclinical to clinical findings. APMIS 2022, 130, 719–740. [Google Scholar] [CrossRef]
  21. Ohtani, N.; Hara, E. Gut-liver axis-mediated mechanism of liver cancer: A special focus on the role of gut microbiota. Cancer Sci. 2021, 112, 4433–4443. [Google Scholar] [CrossRef]
  22. Gupta, H.; Youn, G.S.; Shin, M.J.; Suk, K.T. Role of Gut Microbiota in Hepatocarcinogenesis. Microorganisms 2019, 7, 121. [Google Scholar] [CrossRef] [Green Version]
  23. Nishimura, N.; Kaji, K.; Kitagawa, K.; Sawada, Y.; Furukawa, M.; Ozutsumi, T.; Fujinaga, Y.; Tsuji, Y.; Takaya, H.; Kawaratani, H.; et al. Intestinal Permeability Is a Mechanical Rheostat in the Pathogenesis of Liver Cirrhosis. Int. J. Mol. Sci. 2021, 22, 6921. [Google Scholar] [CrossRef]
  24. Giannelli, V.; Di Gregorio, V.; Iebba, V.; Giusto, M.; Schippa, S.; Merli, M.; Thalheimer, U. Mi-crobiota and the gut-liver axis: Bacterial translocation, inflammation and infection in cirrhosis. World J. Gastroenterol. 2014, 20, 16795–16810. [Google Scholar] [CrossRef] [PubMed]
  25. Guo, W.; Zhou, X.; Li, X.; Zhu, Q.; Peng, J.; Zhu, B.; Zheng, X.; Lu, Y.; Yang, D.; Wang, B.; et al. Depletion of Gut Microbiota Impairs Gut Barrier Function and Antiviral Immune Defense in the Liver. Front. Immunol. 2021, 12, 636803. [Google Scholar] [CrossRef] [PubMed]
  26. Selvaggi, F.; Catalano, T.; Cotellese, R.; Aceto, G.M. Targeting Wnt/β-Catenin Pathways in Primary Liver Tumours: From Microenvironment Signaling to Therapeutic Agents. Cancers 2022, 14, 1912. [Google Scholar] [CrossRef]
  27. Bhavsar, A.P.; Guttman, J.; Finlay, B.B. Manipulation of host-cell pathways by bacterial pathogens. Nature 2007, 449, 827–834. [Google Scholar] [CrossRef] [PubMed]
  28. Nusse, R.; Clevers, H. Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017, 169, 985–999. [Google Scholar] [CrossRef] [Green Version]
  29. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef]
  30. Clevers, H.; Nusse, R. Wnt/beta-catenin signaling and disease. Cell 2012, 149, 1192–1205. [Google Scholar] [CrossRef] [Green Version]
  31. Hua, Y.; Yang, Y.; Li, Q.; He, X.; Zhu, W.; Wang, J.; Gan, X. Oligomerization of Frizzled and LRP5/6 protein initiates intracellular signaling for the canonical WNT/β-catenin pathway. J. Biol. Chem. 2018, 293, 19710–19724. [Google Scholar] [CrossRef] [Green Version]
  32. Florian, M.C.; Nattamai, K.J.; Dörr, K.; Marka, G.; Uberle, B.; Vas, V.; Eckl, C.; Andrä, I.; Schiemann, M.; Oostendorp, R.A.; et al. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature 2013, 503, 392–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Huber, A.H.; Weis, W.I. The structure of the beta-catenin/E-cadherin complex and the molecular basis of diverse ligand recognition by beta-catenin. Cell 2001, 105, 391–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Gordon, M.D.; Nusse, R. Wnt signaling: Multiple pathways, multiple receptors, and multiple transcription factors. J. Biol. Chem. 2006, 281, 22429–22433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Clevers, H.; Loh, K.M.; Nusse, R. Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science 2014, 34, 1248012. [Google Scholar] [CrossRef]
  36. Vilchez, V.; Turcios, L.; Marti, F.; Gedaly, R. Targeting Wnt/β-catenin pathway in hepatocellular carcinoma treatment. World J. Gastroenterol. 2016, 22, 823–832. [Google Scholar] [CrossRef]
  37. Perugorria, M.J.; Olaizola, P.; Labiano, I.; Esparza-Baquer, A.; Marzioni, M.; Marin, J.J.G.; Bujanda, L.; Banales, J.M. Wnt-β-catenin signalling in liver development, health and disease. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 121–136. [Google Scholar] [CrossRef] [PubMed]
  38. Benhamouche, S.; Decaens, T.; Godard, C.; Chambrey, R.; Rickman, D.S.; Moinard, C.; Vasseur-Cognet, M.; Kuo, C.J.; Kahn, A.; Perret, C.; et al. Apc tumor suppressor gene is the “zonation-keeper” of mouse liver. Dev. Cell 2006, 10, 759–770. [Google Scholar] [CrossRef] [Green Version]
  39. Khalaf, A.M.; Fuentes, D.; Morshid, A.I.; Burke, M.R.; Kaseb, A.O.; Hassan, M.; Hazle, J.D.; Elsayes, K.M. Role of Wnt/β-catenin signaling in hepatocellular carcinoma, pathogenesis, and clinical significance. J. Hepatocell. Carcinoma 2018, 5, 61–73. [Google Scholar] [CrossRef] [Green Version]
  40. Elpek, G.O. Molecular pathways in viral hepatitis-associated liver carcinogenesis: An update. World J. Clin. Cases 2021, 9, 4890–4917. [Google Scholar] [CrossRef]
  41. Wang, W.; Smits, R.; Hao, H.; He, C. Wnt/β-Catenin Signaling in Liver Cancers. Cancers 2019, 11, 926. [Google Scholar] [CrossRef] [Green Version]
  42. Selvaggi, F.; Catalano, T.; Lattanzio, R.; Cotellese, R.; Aceto, G.M. Wingless/It/β-catenin signaling in liver metastasis from colorectal cancer: A focus on biological mechanisms and therapeutic opportunities. World J. Gastroenterol. 2023, 29, 2764–2783. [Google Scholar] [CrossRef] [PubMed]
  43. Pez, F.; Lopez, A.; Kim, M.; Wands, J.R.; Caron de Fromentel, C.; Merle, P. Wnt signaling and hepatocarcinogenesis: Molecular targets for the development of innovative anticancer drugs. J. Hepatol. 2013, 59, 1107–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Taniguchi, K.; Roberts, L.R.; Aderca, I.N.; Dong, X.; Qian, C.; Murphy, L.M.; Nagorney, D.M.; Burgart, L.J.; Roche, P.C.; Smith, D.I.; et al. Mutational spectrum of beta-catenin, AXIN1, and AXIN2 in hepatocellular carcinomas and hepatoblastomas. Oncogene 2002, 21, 4863–4871. [Google Scholar] [CrossRef] [Green Version]
  45. Russell, J.O.; Monga, S.P. Wnt/β-Catenin Signaling in Liver Development, Homeostasis, and Pathobiology. Annu. Rev. Pathol. 2018, 13, 351–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Mavila, N.; Thundimadathil, J. The Emerging Roles of Cancer Stem Cells and Wnt/Beta-Catenin Signaling in Hepatoblastoma. Cancers 2019, 11, 1406. [Google Scholar] [CrossRef] [Green Version]
  47. Trivedi, P.; Patel, S.K.; Bellavia, D.; Messina, E.; Palermo, R.; Ceccarelli, S.; Marchese, C.; Anastasiadou, E.; Minter, L.M.; Felli, M.P. When Viruses Cross Developmental Pathways. Front. Cell Dev. Biol. 2021, 9, 691644. [Google Scholar] [CrossRef] [PubMed]
  48. Vedham, V.; Verma, M.; Mahabir, S. Early-life exposures to infectious agents and later cancer development. Cancer Med. 2015, 4, 1908–1922. [Google Scholar] [CrossRef]
  49. 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] [PubMed]
  50. Singal, A.G.; Lampertico, P.; Nahon, P. Epidemiology and surveillance for hepatocellular carcinoma: New trends. J. Hepatol. 2020, 72, 250–261. [Google Scholar] [CrossRef] [Green Version]
  51. Markakis, G.E.; Koulouris, A.; Tampaki, M.; Cholongitas, E.; Deutsch, M.; Papatheodoridis, G.V.; Koskinas, J. The changing epidemiology of hepatocellular carcinoma in Greece. Ann. Gastroenterol. 2022, 35, 88–94. [Google Scholar] [CrossRef]
  52. Chang, M.S.; Nguyen, M.H. Epidemiology of hepatitis B and the role of vaccination. Best Pract. Res. Clin. Gastroenterol. 2017, 31, 239–247. [Google Scholar] [CrossRef] [PubMed]
  53. Stella, L.; Santopaolo, F.; Gasbarrini, A.; Pompili, M.; Ponziani, F.R. Viral hepatitis and hepatocellular carcinoma: From molecular pathways to the role of clinical surveillance and antiviral treatment. World J. Gastroenterol. 2022, 28, 2251–2281. [Google Scholar] [CrossRef] [PubMed]
  54. Yu, J.; Zhang, H.; Zhang, Y.; Zhang, X. Integrated Analysis of the Altered lncRNA, microRNA, and mRNA Expression in HBV-Positive Hepatocellular Carcinoma. Life 2022, 12, 701. [Google Scholar] [CrossRef] [PubMed]
  55. Daud, M.; Rana, M.A.; Husnain, T.; Ijaz, B. Modulation of Wnt signaling pathway by hepatitis B virus. Arch. Virol. 2017, 162, 2937–2947. [Google Scholar] [CrossRef]
  56. Xie, Q.; Chen, L.; Shan, X.; Shan, X.; Tang, J.; Zhou, F.; Chen, Q.; Quan, H.; Nie, D.; Zhang, W.; et al. Epigenetic silencing of SFRP1 and SFRP5 by hepatitis B Virus X protein enhances hepatoma cell tumorigenicity through Wnt signaling pathway. Int. J. Cancer 2014, 135, 635–646. [Google Scholar] [CrossRef]
  57. Hsieh, A.; Kim, H.S.; Lim, S.O.; Yu, D.Y.; Jung, G. Hepatitis B Viral X protein interacts with tumor suppressor adenomatous polyposis coli to activate Wnt/β-catenin signaling. Cancer Lett. 2011, 300, 162–172. [Google Scholar] [CrossRef]
  58. Zheng, C.; Liu, M.; Ge, Y.; Qian, Y.; Fan, H. HBx increases chromatin accessibility and ETV4 expression to regulate dishevelled-2 and promote HCC progression. Cell Death Dis. 2022, 13, 116. [Google Scholar] [CrossRef]
  59. Sun, T.; Zhang, J. ETV4 mediates the Wnt/β-catenin pathway through transcriptional activation of ANXA2 to promote hepatitis B virus-associated liver hepatocellular carcinoma progression. J. Biochem. 2021, 170, 663–673. [Google Scholar] [CrossRef]
  60. Tian, X.; Zhao, C.; Ren, J.; Ma, Z.M.; Xie, Y.H.; Wen, Y.M. Gene-expression profiles of a hepatitis B small surface antigen-secreting cell line reveal upregulation of lymphoid enhancer-binding factor 1. J. Gen. Virol. 2007, 88, 2966–2976. [Google Scholar] [CrossRef]
  61. Tian, X.; Li, J.; Ma, Z.M.; Zhao, C.; Wan, D.F.; Wen, Y.M. Role of hepatitis B surface antigen in the development of hepatocellular carcinoma: Regulation of lymphoid enhancer-binding factor 1. J. Exp. Clin. Cancer Res. 2009, 28, 58. [Google Scholar] [CrossRef] [Green Version]
  62. Tran, B.M.; Flanagan, D.J.; Ebert, G.; Warner, N.; Tran, H.; Fifis, T.; Kastrappis, G.; Christophi, C.; Pellegrini, M.; Torresi, J.; et al. The Hepatitis B Virus Pre-Core Protein p22 Activates Wnt Signaling. Cancers 2020, 12, 1435. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, X.; Wang, L.; Zhang, S.; Lin, J.; Zhang, S.; Feitelson, M.A.; Gao, H.; Zhu, M. Mutations in the C-terminus of the X protein of hepatitis B virus regulate Wnt-5a expression in hepatoma Huh7 cells: cDNA microarray and proteomic analyses. Carcinogenesis 2008, 29, 1207–1214. [Google Scholar] [CrossRef] [PubMed]
  64. Ding, S.L.; Yang, Z.W.; Wang, J.; Zhang, X.L.; Chen, X.M.; Lu, F.M. Integrative analysis of aberrant Wnt signaling in hepatitis B virus-related hepatocellular carcinoma. World J. Gastroenterol. 2015, 21, 6317–6328. [Google Scholar] [CrossRef]
  65. Nault, J.C.; Zucman-Rossi, J. Genetics of hepatocellular carcinoma: The next generation. J. Hepatol. 2014, 60, 224–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Kim, S.S.; Cho, H.J.; Lee, H.Y.; Park, J.H.; Noh, C.K.; Shin, S.J.; Lee, K.M.; Yoo, B.M.; Lee, K.J.; Cho, S.W.; et al. Genetic polymorphisms in the Wnt/β-catenin pathway genes as predictors of tumor development and survival in patients with hepatitis B virus-associated hepatocellular carcinoma. Clin. Biochem. 2016, 49, 792–801. [Google Scholar] [CrossRef]
  67. Han, Q.; Wang, X.; Liao, X.; Han, C.; Yu, T.; Yang, C.; Li, G.; Han, B.; Huang, K.; Zhu, G.; et al. Diagnostic and prognostic value of WNT family gene expression in hepatitis B virus related hepatocellular carcinoma. Oncol. Rep. 2019, 42, 895–910. [Google Scholar] [CrossRef] [PubMed]
  68. Zheng, W.; Yao, M.; Fang, M.; Pan, L.; Wang, L.; Yang, J.; Dong, Z.; Yao, D. Oncogenic Wnt3a: A Candidate Specific Marker and Novel Molecular Target for Hepatocellular Carcinoma. J. Cancer 2019, 10, 5862–5873. [Google Scholar] [CrossRef]
  69. Niknafs, Y.S.; Chinnaiyan, A.M. RNA identity crisis: Hepatitis B walks the LINE. Cancer Cell 2014, 25, 259–260. [Google Scholar] [CrossRef] [Green Version]
  70. 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]
  71. Gao, Q.; Zhu, H.; Dong, L.; Shi, W.; Chen, R.; Song, Z.; Huang, C.; Li, J.; Dong, X.; Zhou, Y.; et al. Integrated Proteogenomic Characterization of HBV-Related Hepatocellular Carcinoma. Cell 2019, 179, 1240. [Google Scholar] [CrossRef]
  72. Ng, C.K.Y.; Dazert, E.; Boldanova, T.; Coto-Llerena, M.; Nuciforo, S.; Ercan, C.; Suslov, A.; Meier, M.A.; Bock, T.; Schmidt, A.; et al. Integrative proteogenomic characterization of hepatocellular carcinoma across etiologies and stages. Nat. Commun. 2022, 13, 2436. [Google Scholar] [CrossRef]
  73. 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, e27496. [Google Scholar] [CrossRef] [PubMed]
  74. Quan, H.; Zhou, F.; Nie, D.; Chen, Q.; Cai, X.; Shan, X.; Zhou, Z.; Chen, K.; Huang, A.; Li, S.; et al. Hepatitis C virus core protein epigenetically silences SFRP1 and enhances HCC aggressiveness by inducing epithelial-mesenchymal transition. Oncogene 2014, 33, 2826–2835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Umer, M.; Qureshi, S.A.; Hashmi, Z.Y.; Raza, A.; Ahmad, J.; Rahman, M.; Iqbal, M. Promoter hypermethylation of Wnt pathway inhibitors in hepatitis C virus-induced multistep hepatocarcinogenesis. Virol. J. 2014, 11, 117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Ripoli, M.; Barbano, R.; Balsamo, T.; Piccoli, C.; Brunetti, V.; Coco, M.; Mazzoccoli, G.; Vinciguerra, M.; Pazienza, V. Hypermethylated levels of E-cadherin promoter in Huh-7 cells expressing the HCV core protein. Virus Res. 2011, 160, 74–81. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, J.; Wang, Z.; Tang, J.; Tang, R.; Shan, X.; Zhang, W.; Chen, Q.; Zhou, F.; Chen, K.; Huang, A.; et al. Hepatitis C virus core protein activates Wnt/β-catenin signaling through multiple regulation of upstream molecules in the SMMC-7721 cell line. Arch. Virol. 2011, 156, 1013–1023. [Google Scholar] [CrossRef]
  78. Aicher, S.; Kakkanas, A.; Cohen, L.; Blumen, B.; Oprisan, G.; Njouom, R.; Meurs, E.F.; Mavromara, P.; Martin, A. Differential regulation of the Wnt/β-catenin pathway by hepatitis C virus recombinants expressing core from various genotypes. Sci. Rep. 2018, 8, 11185. [Google Scholar] [CrossRef] [Green Version]
  79. Jiang, X.H.; Xie, Y.T.; Cai, Y.P.; Ren, J.; Ma, T. Effects of hepatitis C virus core protein and nonstructural protein 4B on the Wnt/β-catenin pathway. BMC Microbiol. 2017, 17, 124. [Google Scholar] [CrossRef] [Green Version]
  80. 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]
  81. Milward, A.; Mankouri, J.; Harris, M. Hepatitis C virus NS5A protein interacts with beta-catenin and stimulates its transcriptional activity in a phosphoinositide-3 kinase-dependent fashion. J. Gen. Virol. 2010, 91, 373–381. [Google Scholar] [CrossRef]
  82. 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]
  83. Wang, Z.; Sheng, Y.Y.; Dong, Q.Z.; Qin, L.X. Hepatitis B virus and hepatitis C virus play different prognostic roles in intrahepatic cholangiocarcinoma: A meta-analysis. World J. Gastroenterol. 2016, 22, 3038–3051. [Google Scholar] [CrossRef] [PubMed]
  84. Lin, D.; Reddy, V.; Osman, H.; Lopez, A.; Koksal, A.R.; Rhadhi, S.M.; Dash, S.; Aydin, Y. Additional Inhibition of Wnt/β-Catenin Signaling by Metformin in DAA Treatments as a Novel Therapeutic Strategy for HCV-Infected Patients. Cells 2021, 10, 790. [Google Scholar] [CrossRef] [PubMed]
  85. Zhao, P.; Malik, S.; Xing, S. Epigenetic Mechanisms Involved in HCV-Induced Hepatocellular Carcinoma (HCC). Front. Oncol. 2021, 11, 677926. [Google Scholar] [CrossRef] [PubMed]
  86. Mahmoudvand, S.; Shokri, S.; Taherkhani, R.; Farshadpour, F. Hepatitis C virus core protein modulates several signaling pathways involved in hepatocellular carcinoma. World J. Gastroenterol. 2019, 25, 42–58. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, Y.; Wei, W.; Cheng, N.; Wang, K.; Li, B.; Jiang, X.; Sun, S. Hepatitis C virus-induced up-regulation of microRNA-155 promotes hepatocarcinogenesis by activating Wnt signaling. Hepatology 2012, 56, 1631–1640. [Google Scholar] [CrossRef]
  88. Ashmawy, A.M.; Elgeshy, K.M.; Abdel-Salam, E.T.; Ghareeb, M.; Kobaisi, M.H.; Amin, H.A.A.; Sharawy, S.K.; Abdel Wahab, A.H.A. Crosstalk between liver-related microRNAs and Wnt/β-catenin pathway in hepatocellular carcinoma patients. Arab. J. Gastroenterol. 2017, 18, 144–150. [Google Scholar] [CrossRef]
  89. Vlachogiannakos, J.; Papatheodoridis, G.V. New epidemiology of hepatitis delta. Liver Int. 2020, 40, 48–53. [Google Scholar] [CrossRef] [Green Version]
  90. Shih, H.H.; Sheen, I.J.; Su, C.W.; Peng, W.L.; Lin, L.H.; Wu, J.C. Hepatitis D virus isolates with low replication and epithelial-mesenchymal transition-inducing activity are associated with disease remission. J. Virol. 2012, 86, 9044–9054. [Google Scholar] [CrossRef] [Green Version]
  91. Wong, Y.; Meehan, M.T.; Burrows, S.R.; Doolan, D.L.; Miles, J.J. Estimating the global burden of Epstein-Barr virus-related cancers. J. Cancer Res. Clin. Oncol. 2022, 148, 31–46. [Google Scholar] [CrossRef]
  92. Sugawara, Y.; Makuuchi, M.; Takada, K. Detection of Epstein-Barr virus DNA in hepatocellular carcinoma tissues from hepatitis C-positive patients. Scand. J. Gastroenterol. 2000, 35, 981–984. [Google Scholar] [CrossRef] [PubMed]
  93. Li, W.; Wu, B.A.; Zeng, Y.M.; Chen, G.C.; Li, X.X.; Chen, J.T.; Guo, Y.W.; Li, M.H.; Zeng, Y. Epstein-Barr virus in hepatocellular carcinogenesis. World J. Gastroenterol. 2004, 10, 3409–3413. [Google Scholar] [CrossRef] [PubMed]
  94. Kang, H.J.; Oh, J.H.; Chun, S.M.; Kim, D.; Ryu, Y.M.; Hwang, H.S.; Kim, S.Y.; An, J.; Cho, E.J.; Lee, H.; et al. Immunogenomic landscape of hepatocellular carcinoma with immune cell stroma and EBV-positive tumor-infiltrating lymphocytes. J. Hepatol. 2019, 71, 91–103. [Google Scholar] [CrossRef] [PubMed]
  95. Chu, P.G.; Chen, Y.Y.; Chen, W.; Weiss, L.M. No direct role for Epstein-Barr virus in American hepatocellular carcinoma. Am. J. Pathol. 2001, 159, 1287–1292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Akhter, S.; Liu, H.; Prabhu, R.; DeLucca, C.; Bastian, F.; Garry, R.F.; Schwartz, M.; Thung, S.N.; Dash, S. Epstein-Barr virus and human hepatocellular carcinoma. Cancer Lett. 2003, 192, 49–57. [Google Scholar] [CrossRef]
  97. Junying, J.; Herrmann, K.; Davies, G.; Lissauer, D.; Bell, A.; Timms, J.; Reynolds, G.M.; Hubscher, S.G.; Young, L.S.; Niedobitek, G.; et al. Absence of Epstein-Barr virus DNA in the tumor cells of European hepatocellular carcinoma. Virology 2003, 306, 236–243. [Google Scholar] [CrossRef] [Green Version]
  98. Birdwell, C.E.; Prasai, K.; Dykes, S.; Jia, Y.; Munroe, T.G.C.; Bienkowska-Haba, M.; Scott, R.S. Epstein-Barr virus stably confers an invasive phenotype to epithelial cells through reprogramming of the WNT pathway. Oncotarget 2018, 9, 10417–10435. [Google Scholar] [CrossRef] [Green Version]
  99. Ljungberg, J.K.; Kling, J.C.; Tran, T.T.; Blumenthal, A. Functions of the WNT Signaling Network in Shaping Host Responses to Infection. Front. Immunol. 2019, 10, 2521. [Google Scholar] [CrossRef] [Green Version]
  100. Marineau, A.; Khan, K.A.; Servant, M.J. Roles of GSK-3 and β-Catenin in Antiviral Innate Immune Sensing of Nucleic Acids. Cells 2020, 9, 897. [Google Scholar] [CrossRef] [Green Version]
  101. Rideg, O.; Dergez, T.; Farkas, K.; Kovács, K.; Kálmán, E.; Tornóczky, T.; Oszter, A. High Prevalence of Non-Vaccinated Oncogenic Human Papillomavirus Genotypes in High-Grade Squamous Intraepithelial Lesions of the Cervix: Thought-Provoking Results of a Detailed HPV Genotype Analysis. Vaccines 2022, 10, 748. [Google Scholar] [CrossRef]
  102. Ma, T.; Su, Z.; Chen, L.; Liu, S.; Zhu, N.; Wen, L.; Yuan, Y.; Lv, L.; Chen, X.; Huang, J.; et al. Human papillomavirus type 18 E6 and E7 genes integrate into human hepatoma derived cell line Hep G2. PLoS ONE 2012, 7, e37964. [Google Scholar] [CrossRef] [PubMed]
  103. Bello, J.O.; Nieva, L.O.; Paredes, A.C.; Gonzalez, A.M.; Zavaleta, L.R.; Lizano, M. Regulation of the Wnt/β-Catenin Signaling Pathway by Human Papillomavirus E6 and E7 Oncoproteins. Viruses 2015, 7, 4734–4755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Marônek, M.; Link, R.; Monteleone, G.; Gardlík, R.; Stolfi, C. Viruses in Cancers of the Digestive System: Active Contributors or Idle Byskluders? Int. J. Mol. Sci. 2020, 21, 8133. [Google Scholar] [CrossRef] [PubMed]
  105. Tian, A.L.; Li, G.X.; Elsheikha, H.M.; Gardner, D.S.; Zhang, X.Y.; Dong, W.; Yang, X.P.; Luo, Y.Y.; Li, H.L.; Cong, W.; et al. Seroepidemiology of Toxoplasma gondii infection in patients with liver disease in eastern China. Epidemiol. Infect. 2017, 145, 2296–2302. [Google Scholar] [CrossRef] [Green Version]
  106. He, H.; Brenier-Pinchart, M.P.; Braun, L.; Kraut, A.; Touquet, B.; Couté, Y.; Tardieux, I.; Hakimi, M.A.; Bougdour, A. Characterization of a Toxoplasma effector uncovers an alternative GSK3/β-catenin-regulatory pathway of inflammation. Elife 2018, 7, e39887. [Google Scholar] [CrossRef]
  107. Bo, R.; Yasen, A.; Shao, Y.; Zhang, W.; Lin, R.; Jiang, T.; Wen, H.; Xiao, H.; Aji, T. Co-existence of hepatocellular carcinoma and cystic echinococcosis. Infect. Agents Cancer 2020, 15, 5. [Google Scholar] [CrossRef] [Green Version]
  108. Xu, Y.; Li, H.; Chen, W.; Yao, X.; Xing, Y.; Wang, X.; Zhong, J.; Meng, G. Mycoplasma hyorhinis activates the NLRP3 inflammasome and promotes migration and invasion of gastric cancer cells. PLoS ONE 2013, 8, e77955. [Google Scholar] [CrossRef] [Green Version]
  109. Kim, M.K.; Shin, S.J.; Lee, H.M.; Choi, H.S.; Jeong, J.; Kim, H.; Paik, S.S.; Kim, M.; Choi, D.; Ryu, C.J. Mycoplasma infection promotes tumor progression via interaction of the mycoplasmal protein p37 and epithelial cell adhesion molecule in hepatocellular carcinoma. Cancer Lett. 2019, 454, 44–52. [Google Scholar] [CrossRef]
  110. Liu, X.; Rong, Z.; Shou, C. Mycoplasma hyorhinis infection promotes gastric cancer cell motility via β-catenin signaling. Cancer Med. 2019, 8, 5301–5312. [Google Scholar] [CrossRef] [Green Version]
  111. Rudramurthy, S.M.; Paul, R.A.; Chakrabarti, A.; Mouton, J.W.; Meis, J.F. Invasive Aspergillosis by Aspergillus flavus: Epidemiology, Diagnosis, Antifungal Resistance, and Management. J. Fungi 2019, 5, 55. [Google Scholar] [CrossRef] [Green Version]
  112. Hussain, S.P.; Schwank, J.; Staib, F.; Wang, X.W.; Harris, C.C. TP53 mutations and hepatocellular carcinoma: Insights into the etiology and pathogenesis of liver cancer. Oncogene 2007, 26, 2166–2176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Fang, Y.; Feng, Y.; Wu, T.; Srinivas, S.; Yang, W.; Fan, J.; Yang, C.; Wang, S. Aflatoxin B1 negatively regulates Wnt/β-catenin signaling pathway through activating miR-33a. PLoS ONE 2013, 8, e73004. [Google Scholar] [CrossRef] [PubMed]
  114. Zhu, L.; Gao, J.; Huang, K.; Luo, Y.; Zhang, B.; Xu, W. miR-34a screened by miRNA profiling negatively regulates Wnt/β-catenin signaling pathway in Aflatoxin B1 induced hepatotoxicity. Sci. Rep. 2015, 5, 16732. [Google Scholar] [CrossRef] [Green Version]
  115. Trinath, J.; Holla, S.; Mahadik, K.; Prakhar, P.; Singh, V.; Balaji, K.N. The WNT signaling pathway contributes to dectin-1-dependent inhibition of Toll-like receptor-induced inflammatory signature. Mol. Cell. Biol. 2014, 34, 4301–4314. [Google Scholar] [CrossRef] [Green Version]
  116. Marchese, S.; Polo, A.; Ariano, A.; Velotto, S.; Costantini, S.; Severino, L. Aflatoxin B1 and M1: Biological Properties and Their Involvement in Cancer Development. Toxins 2018, 10, 214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Chu, Y.J.; Yang, H.I.; Wu, H.C.; Lee, M.H.; Liu, J.; Wang, L.Y.; Lu, S.N.; Jen, C.L.; You, S.L.; Santella, R.M.; et al. Aflatoxin B1 exposure increases the risk of hepatocellular carcinoma associated with hepatitis C virus infection or alcohol consumption. Eur. J. Cancer 2018, 94, 37–46. [Google Scholar] [CrossRef]
  118. Labib, P.L.; Goodchild, G.; Pereira, S.P. Molecular Pathogenesis of Cholangiocarcinoma. BMC Cancer 2019, 19, 185. [Google Scholar] [CrossRef] [Green Version]
  119. Boulter, L.; Guest, R.V.; Kendall, T.J.; Wilson, D.H.; Wojtacha, D.; Robson, A.J.; Ridgway, R.A.; Samuel, K.; Van Rooijen, N.; Barry, S.T.; et al. WNT signaling drives cholangiocarcinoma growth and can be pharmacologically inhibited. J. Clin. Investig. 2015, 125, 1269–1285. [Google Scholar] [CrossRef] [Green Version]
  120. Shao, Y.; Zheng, Q.; Wang, W.; Xin, N.; Song, X.; Zhao, C. Biological functions of macrophage-derived Wnt5a, and its roles in human diseases. Oncotarget 2016, 7, 67674–67684. [Google Scholar] [CrossRef] [Green Version]
  121. Zhou, M.; Wang, C.; Lu, S.; Xu, Y.; Li, Z.; Jiang, H.; Ma, Y. Tumor-associated macrophages in cholangiocarcinoma: Complex interplay and potential therapeutic target. EBioMedicine 2021, 67, 103375. [Google Scholar] [CrossRef]
  122. Zhang, G.F.; Qiu, L.; Yang, S.L.; Wu, J.C.; Liu, T.J. Wnt/β-catenin signaling as an emerging potential key pharmacological target in cholangiocarcinoma. Biosci. Rep. 2020, 40, BSR20193353. [Google Scholar] [CrossRef] [Green Version]
  123. Tao, L.Y.; He, X.D.; Xiu, D.R. Hepatitis B virus is associated with the clinical features and survival rate of patients with intrahepatic cholangiocarcinoma. Clin. Res. Hepatol. Gastroenterol. 2016, 40, 682–687. [Google Scholar] [CrossRef]
  124. Wang, C.; Yang, W.; Yan, H.X.; Luo, T.; Zhang, J.; Tang, L.; Wu, F.Q.; Zhang, H.L.; Yu, L.X.; Zheng, L.Y.; et al. Hepatitis B Virus X (HBx) induces tumorigenicity of hepatic progenitor cells in 3,5-diethoxycarbonyl-1,4-dihydrocollidine-treated HBx transgenic mice. Hepatology 2012, 55, 108–120. [Google Scholar] [CrossRef]
  125. Zhou, H.B.; Hu, J.Y.; Hu, H.P. Hepatitis B virus infection and intrahepatic cholangiocarcinoma. World J. Gastroenterol. 2014, 20, 5721–5729. [Google Scholar] [CrossRef]
  126. Navas, M.C.; Glaser, S.; Dhruv, H.; Celinski, S.; Alpini, G.; Meng, F. Hepatitis C Virus Infection and Cholangiocarcinoma: An Insight into Epidemiologic Evidences and Hypothetical Mechanisms of Oncogenesis. Am. J. Pathol. 2019, 189, 1122–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Pezzuto, F.; Izzo, F.; Buonaguro, L.; Annunziata, C.; Tatangelo, F.; Botti, G.; Buonaguro, F.M.; Tornesello, M.L. Tumor specific mutations in TERT promoter and CTNNB1 gene in hepatitis B and hepatitis C related hepatocellular carcinoma. Oncotarget 2016, 7, 54253–54262. [Google Scholar] [CrossRef] [Green Version]
  128. Klufah, F.; Mobaraki, G.; Chteinberg, E.; Alharbi, R.A.; Winnepenninckx, V.; Speel, E.J.M.; Rennspiess, D.; Olde Damink, S.W.; Neumann, U.P.; Kurz, A.K.; et al. High Prevalence of Human Polyomavirus 7 in Cholangiocarcinomas and Adjacent Peritumoral Hepatocytes: Preliminary Findings. Microorganisms 2020, 8, 1125. [Google Scholar] [CrossRef] [PubMed]
  129. Huang, Y.H.; Zhang, C.Z.; Huang, Q.S.; Yeong, J.; Wang, F.; Yang, X.; He, Y.F.; Zhang, X.L.; Zhang, H.; Chen, S.L.; et al. Clinicopathologic features, tumor immune microenvironment and genomic landscape of Epstein-Barr virus-associated intrahepatic cholangiocarcinoma. J. Hepatol. 2021, 74, 838–849. [Google Scholar] [CrossRef] [PubMed]
  130. Saijuntha, W.; Sithithaworn, P.; Petney, T.N.; Andrews, R.H. Foodborne zoonotic parasites of the family Opisthorchiidae. Res. Vet. Sci. 2021, 135, 404–411. [Google Scholar] [CrossRef] [PubMed]
  131. Samaras, V.; Rafailidis, P.I.; Mourtzoukou, E.G.; Peppas, G.; Falagas, M.E. Chronic bacterial and parasitic infections and cancer: A review. J. Infect. Dev. Ctries. 2010, 4, 267–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Kim, D.M.; Ko, B.S.; Ju, J.W.; Cho, S.H.; Yang., S.J.; Yeom, Y.I.; Kim., T.S.; Won, Y.; Kim, I.C. Gene expression profiling in mouse liver infected with Clonorchis sinensis metacercariae. Parasitol. Res. 2009, 106, 269–278. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, C.; Lei, H.; Tian, Y.; Shang, M.; Wu, Y.; Li, Y.; Zhao, L.; Shi, M.; Tang, X.; Chen, T.; et al. Clonorchis sinensis granulin: Identification, immunolocalization, and function in promoting the metastasis of cholangiocarcinoma and hepatocellular carcinoma. Parasites Vectors 2017, 10, 262. [Google Scholar] [CrossRef] [Green Version]
  134. Smout, M.J.; Sripa, B.; Laha, T.; Mulvenna, J.; Gasser, R.B.; Young, N.D.; Bethony, J.M.; Brindley, P.J.; Loukas, A. Infection with the carcinogenic human liver fluke, Opisthorchis viverrini. Mol. Biosyst. 2011, 7, 1367–1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Arunsan, P.; Chaidee, A.; Cochran, C.J.; Mann, V.H.; Tanno, T.; Kumkhaek, C.; Smout, M.J.; Karinshak, S.E.; Rodpai, R.; Sotillo, J.; et al. Liver fluke granulin promotes extracellular vesicle-mediated crosstalk and cellular microenvironment conducive to cholangiocarcinoma. Neoplasia 2020, 22, 203–216. [Google Scholar] [CrossRef] [PubMed]
  136. Yothaisong, S.; Thanee, M.; Namwat, N.; Yongvanit, P.; Boonmars, T.; Puapairoj, A.; Loilome, W. Opisthorchis viverrini infection activates the PI3K/AKT/PTEN and Wnt/β-catenin signaling pathways in a Cholangiocarcinogenesis model. Asian Pac. J. Cancer Prev. 2014, 15, 10463–10468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Loilome, W.; Bungkanjana, P.; Techasen, A.; Namwat, N.; Yongvanit, P.; Puapairoj, A.; Khuntikeo, N.; Riggins, G.J. Activated macrophages promote Wnt/β-catenin signaling in cholangiocarcinoma cells. Tumour Biol. 2014, 35, 5357–5367. [Google Scholar] [CrossRef] [Green Version]
  138. O’Hara, S.P.; Small, A.J.; Gajdos, G.B.; Badley, A.D.; Chen, X.M.; Larusso, N.F. HIV-1 Tat protein suppresses cholangiocyte toll-like receptor 4 expression and defense against Cryptosporidium parvum. J. Infect. Dis. 2009, 199, 1195–1204. [Google Scholar] [CrossRef] [Green Version]
  139. O’Hara, S.P.; Karlsen, T.H.; LaRusso, N.F. Cholangiocytes and the environment in primary sclerosing cholangitis: Where is the link? Gut. 2017, 66, 1873–1877. [Google Scholar] [CrossRef] [Green Version]
  140. O’Hara, S.P.; Small, A.J.; Nelson, J.B.; Badley, A.D.; Chen, X.M.; Gores, G.J.; Larusso, N.F. The human immunodeficiency virus type 1 tat protein enhances Cryptosporidium parvum-induced apoptosis in cholangiocytes via a Fas ligand-dependent mechanism. Infect. Immun. 2007, 75, 684–696. [Google Scholar] [CrossRef] [Green Version]
  141. Rahman, M.; Chapel, H.; Chapman, R.W.; Collier, J.D. Cholangiocarcinoma complicating secondary sclerosing cholangitis from cryptosporidiosis in an adult patient with CD40 ligand deficiency: Case report and review of the literature. Int. Arch. Allergy Immunol. 2012, 159, 204–220. [Google Scholar] [CrossRef]
  142. Suryawanshi, A.; Hussein, M.S.; Prasad, P.D.; Manicassamy, S. Wnt Signaling Cascade in Dendritic Cells and Regulation of Anti-tumor Immunity. Front. Immunol. 2020, 11, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Deenonpoe, R.; Mairiang, E.; Mairiang, P.; Pairojkul, C.; Chamgramol, Y.; Rinaldi, G.; Loukas, A.; Brindley, P.J.; Sripa, B. Elevated prevalence of Helicobacter species and virulence factors in opisthorchiasis and associated hepatobiliary disease. Sci. Rep. 2017, 7, 42744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Song, X.; Xin, N.; Wang, W.; Zhao, C. Wnt/β-catenin, an oncogenic pathway targeted by H. pylori in gastric carcinogenesis. Oncotarget 2015, 6, 35579–35588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Liu, X.; Lu, R.; Wu, S.; Sun, J. Salmonella regulation of intestinal stem cells through the Wnt/beta-catenin pathway. FEBS Lett. 2010, 584, 911–916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Liu, X.; Lu, R.; Wu, S.; Zhang, Y.G.; Xia, Y.; Sartor, R.B.; Sun, J. Wnt2 inhibits enteric bacterial-induced inflammation in intestinal epithelial cells. Inflamm. Bowel Dis. 2012, 18, 418–429. [Google Scholar] [CrossRef] [Green Version]
  147. Liu, X.; Wu, S.; Xia, Y.; Li, X.E.; Xia, Y.; Zhou, Z.D.; Sun, J. Wingless homolog Wnt11 suppresses bacterial invasion and inflammation in intestinal epithelial cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 301, G992–G1003. [Google Scholar] [CrossRef] [Green Version]
  148. Spadoni, I.; Zagato, E.; Bertocchi, A.; Paolinelli, R.; Hot, E.; Di Sabatino, A.; Caprioli, F.; Bottiglieri, L.; Oldani, A.; Viale, G.; et al. gut-vascular barrier controls the systemic dissemination of bacteria. Science 2015, 350, 830–834. [Google Scholar] [CrossRef]
  149. Arciniegas Ruiz, S.M.; Eldar-Finkelman, H. Glycogen Synthase Kinase-3 Inhibitors: Preclinical and Clinical Focus on CNS-A Decade Onward. Front. Mol. Neurosci. 2022, 14, 792364. [Google Scholar] [CrossRef]
  150. Lin, D.; Chen, Y.; Koksal, A.R.; Dash, S.; Aydin, Y. Targeting ER stress/PKA/GSK-3β/β-catenin pathway as a potential novel strategy for hepatitis C virus-infected patients. Cell Commun. Signal. 2023, 21, 102. [Google Scholar] [CrossRef]
  151. Sun, G.; Liu, H.; Zhao, J.; Zhang, J.; Huang, T.; Sun, G.; Zhao, S.; Zhang, Z.; Cao, H.; Rong, D.; et al. Macrophage GSK3β-deficiency inhibits the progression of hepatocellular carcinoma and enhances the sensitivity of anti-PD1 immunotherapy. J. Immunother. Cancer 2022, 10, e005655. [Google Scholar] [CrossRef]
  152. Park, H.; Park, H.; Baek, J.; Moon, H.; Ro, S.W. Target Therapy for Hepatocellular Carcinoma: Beyond Receptor Tyrosine Kinase Inhibitors and Immune Checkpoint Inhibitors. Biology 2022, 11, 585. [Google Scholar] [CrossRef] [PubMed]
  153. Huang, J.; Qu, Q.; Guo, Y.; Xiang, Y.; Feng, D. Tankyrases/β-catenin Signaling Pathway as an Anti-proliferation and Anti-metastatic Target in Hepatocarcinoma Cell Lines. J. Cancer 2020, 11, 432–440. [Google Scholar] [CrossRef] [PubMed]
  154. Park, W.J.; Kim, M.J. A New Wave of Targeting ‘Undruggable’ Wnt Signaling for Cancer Therapy: Challenges and Opportunities. Cells 2023, 12, 1110. [Google Scholar] [CrossRef] [PubMed]
  155. Park, H.B.; Kim, J.W.; Baek, K.H. Regulation of Wnt Signaling through Ubiquitination and Deubiquitination in Cancers. Int. J. Mol. Sci. 2020, 21, 3904. [Google Scholar] [CrossRef] [PubMed]
  156. Shi, J.; Liu, Y.; Xu, X.; Zhang, W.; Yu, T.; Jia, J.; Liu, C. Deubiquitinase USP47/UBP64E Regulates β-Catenin Ubiquitination and Degradation and Plays a Positive Role in Wnt Signaling. Mol. Cell. Biol. 2015, 35, 3301–3311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Kwon, Y.T.; Ciechanover, A. The Ubiquitin Code in the Ubiquitin-Proteasome System and Autophagy. Trends Biochem. Sci. 2017, 42, 873–886. [Google Scholar] [CrossRef]
  158. Reyes-Turcu, F.E.; Ventii, K.H.; Wilkinson, K.D. Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu. Rev. Biochem. 2009, 78, 363–397. [Google Scholar] [CrossRef] [Green Version]
  159. Proulx, J.; Borgmann, K.; Park, I.W. Role of Virally-Encoded Deubiquitinating Enzymes in Regulation of the Virus Life Cycle. Int. J. Mol. Sci. 2021, 22, 4438. [Google Scholar] [CrossRef]
  160. Zong, Z.; Zhang, Z.; Wu, L.; Zhang, L.; Zhou, F. The Functional Deubiquitinating Enzymes in Control of Innate Antiviral Immunity. Adv. Sci. 2020, 8, 2002484. [Google Scholar] [CrossRef]
  161. Kumar, P.; Kumar, P.; Mandal, D.; Velayutham, R. The emerging role of Deubiquitinases (DUBs) in parasites: A foresight review. Front. Cell. Infect. Microbiol. 2022, 12, 985178. [Google Scholar] [CrossRef]
  162. Calistri, A.; Munegato, D.; Carli, I.; Parolin, C.; Palù, G. The ubiquitin-conjugating system: Multiple roles in viral replication and infection. Cells 2014, 3, 386–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Kusakabe, S.; Suzuki, T.; Sugiyama, Y.; Haga, S.; Horike, K.; Tokunaga, M.; Hirano, J.; Zhang, H.; Chen, D.V.; Ishiga, H.; et al. USP15 Participates in Hepatitis C Virus Propagation through Regulation of Viral RNA Translation and Lipid Droplet Formation. J. Virol. 2019, 93, e01708-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Rogan, M.R.; Patterson, L.L.; Wang, J.Y.; McBride, J.W. Bacterial Manipulation of Wnt Signaling: A Host-Pathogen Tug-of-Wnt. Front. Immunol. 2019, 10, 2390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Overview of the Wnt/β-catenin signal pathway. Wnt ligands o-acylated by Porcupine acylase (PORCN) can be secreted and exert both paracrine and autocrine actions on cells. In the absence of adequate ligand production, β-catenin is initiated for proteasomal degradation by the destruction complex (Axin, APC, GSK-3β), sequentially phosphorylated by CK1 and ubiquitinated by β-TrCP. In the nucleus, in the absence of β-catenin, transcription of target genes is repressed by the binding of TLE-1 to the transcription factor TCF/LEF. In the presence of Wnt ligand overproduction, Frizzled and LRP coreceptors associate by recruiting Axin and Dvl to the membrane; the inactive signal then becomes active, the stabilized β-Catenin can enter the nucleus, through binding with the transcription factor TCF/LEF, and can induce the expression of Wnt target genes (e.g., cyclin D1 or c-Myc). The main infectious agents that have been shown to interfere with the Wnt/β-catenin pathway are: Hepatitis B virus (HBV); Hepatitis C virus (HCV); Hepatitis D virus (HDV); Human papilloma virus (HPV); T. (Toxoplasma) gondii; A. (Aspergillus) flavus; C. (Clonorchis) sinensis; O. (Opisthorchis) viverrini; S. (Salmonella) typhi.
Figure 1. Overview of the Wnt/β-catenin signal pathway. Wnt ligands o-acylated by Porcupine acylase (PORCN) can be secreted and exert both paracrine and autocrine actions on cells. In the absence of adequate ligand production, β-catenin is initiated for proteasomal degradation by the destruction complex (Axin, APC, GSK-3β), sequentially phosphorylated by CK1 and ubiquitinated by β-TrCP. In the nucleus, in the absence of β-catenin, transcription of target genes is repressed by the binding of TLE-1 to the transcription factor TCF/LEF. In the presence of Wnt ligand overproduction, Frizzled and LRP coreceptors associate by recruiting Axin and Dvl to the membrane; the inactive signal then becomes active, the stabilized β-Catenin can enter the nucleus, through binding with the transcription factor TCF/LEF, and can induce the expression of Wnt target genes (e.g., cyclin D1 or c-Myc). The main infectious agents that have been shown to interfere with the Wnt/β-catenin pathway are: Hepatitis B virus (HBV); Hepatitis C virus (HCV); Hepatitis D virus (HDV); Human papilloma virus (HPV); T. (Toxoplasma) gondii; A. (Aspergillus) flavus; C. (Clonorchis) sinensis; O. (Opisthorchis) viverrini; S. (Salmonella) typhi.
Microorganisms 11 01632 g001
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

Catalano, T.; Selvaggi, F.; Esposito, D.L.; Cotellese, R.; Aceto, G.M. Infectious Agents Induce Wnt/β-Catenin Pathway Deregulation in Primary Liver Cancers. Microorganisms 2023, 11, 1632. https://doi.org/10.3390/microorganisms11071632

AMA Style

Catalano T, Selvaggi F, Esposito DL, Cotellese R, Aceto GM. Infectious Agents Induce Wnt/β-Catenin Pathway Deregulation in Primary Liver Cancers. Microorganisms. 2023; 11(7):1632. https://doi.org/10.3390/microorganisms11071632

Chicago/Turabian Style

Catalano, Teresa, Federico Selvaggi, Diana Liberata Esposito, Roberto Cotellese, and Gitana Maria Aceto. 2023. "Infectious Agents Induce Wnt/β-Catenin Pathway Deregulation in Primary Liver Cancers" Microorganisms 11, no. 7: 1632. https://doi.org/10.3390/microorganisms11071632

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