Genetically modified animal models recapitulating molecular events altered in human...

7
Abstract New advancements have been made in recent years in the understanding of the molecular mechanisms that govern human liver tumorigenesis. Experimental ani- mal models have been widely used, especially mouse models. In this review we highlight some of the genetically engineered mouse models that have proved to be excellent tools to study the intracellular signalling pathways altered in hepatocarcinogenesis and establish potential correlations with data from humans, with special focus on hepatocel- lular carcinoma (HCC), the most common type of primary liver cancer. Information obtained from these animal mod- els will help to design future therapeutic approaches to HCC, particularly those that explore drugs that specifically target the altered molecular pathways. Keywords Liver cancer · HCC · Growth factors · TGF-beta · p53 · Mdr-2 Introduction Liver cancer comprises diverse, histologically distinct primary hepatic neoplasms. Among these hepatocellular carcinoma (HCC) is the most common, representing 83% of all cases and one of the world’s deadliest cancers [1]. In recent years impressive progress has been made in under- standing its molecular pathogenesis and several experimen- tal animal models have contributed to the definition of the signal transduction pathways involved [2]. In spite of the interspecies differences and the obvious fact that there is no mouse model that can possibly mirror all human HCC variants, there is no doubt that animal models have allowed us to learn much about the molecular mechanisms underly- ing the pathogenesis of HCC. In fact, common alterations in specific growth factors and signal transduction pathways playing a critical role in the development and progression of liver cancer in mice and humans have been identified, thus providing promising targets for HCC therapies. Here, we highlight some of the genetically engineered mouse models that have proved to be excellent tools to study the intracellular molecular mechanisms altered in liver tumori- genesis and summarise the most important and clinically relevant findings achieved. Experimental models recapitulating overactivation of growth factor pathways Both in vitro and in vivo studies provided early evidence of an association of transforming growth factor-alpha (TGF- α) overexpression and TGF- α-driven signalling activation with hepatocarcinogenesis. Thus, deregulation of the TGF-α signalling pathway is correlated with the *Supported by an unrestricted educational grant from Merck Serono A. Sánchez Dep. Bioquímica y Biología Molecular II Facultad de Farmacia Universidad Complutense ES-28040 Madrid, Spain I. Fabregat (Y) Laboratori d’Oncologia Molecular Institut d’Investigació Biomèdica de Bellvitge (IDIBELL) and Universitat de Barcelona (IDIBELL-UB) Gran Via de l’Hospitalet, 199 ES-08907 L’Hospitalet, Barcelona, Spain e-mail: [email protected] Clin Transl Oncol (2009) 11:208-214 DOI 10.1007/s12094-009-0342-x EDUCATIONAL SERIES Red Series* Genetically modified animal models recapitulating molecular events altered in human hepatocarcinogenesis Aránzazu Sánchez · Isabel Fabregat Received: 8 January 2009 / Accepted: 10 February 2009 CURRENT TECHNOLOGY IN CANCER RESEARCH AND TREATMENT

Transcript of Genetically modified animal models recapitulating molecular events altered in human...

Abstract New advancements have been made in recent years in the understanding of the molecular mechanisms that govern human liver tumorigenesis. Experimental ani-mal models have been widely used, especially mouse models. In this review we highlight some of the genetically engineered mouse models that have proved to be excellent tools to study the intracellular signalling pathways altered in hepatocarcinogenesis and establish potential correlations with data from humans, with special focus on hepatocel-lular carcinoma (HCC), the most common type of primary liver cancer. Information obtained from these animal mod-els will help to design future therapeutic approaches to HCC, particularly those that explore drugs that specifically target the altered molecular pathways.

Keywords Liver cancer · HCC · Growth factors · TGF-beta · p53 · Mdr-2

Introduction

Liver cancer comprises diverse, histologically distinct primary hepatic neoplasms. Among these hepatocellular carcinoma (HCC) is the most common, representing 83% of all cases and one of the world’s deadliest cancers [1]. In recent years impressive progress has been made in under-standing its molecular pathogenesis and several experimen-tal animal models have contributed to the definition of the signal transduction pathways involved [2]. In spite of the interspecies differences and the obvious fact that there is no mouse model that can possibly mirror all human HCC variants, there is no doubt that animal models have allowed us to learn much about the molecular mechanisms underly-ing the pathogenesis of HCC. In fact, common alterations in specific growth factors and signal transduction pathways playing a critical role in the development and progression of liver cancer in mice and humans have been identified, thus providing promising targets for HCC therapies. Here, we highlight some of the genetically engineered mouse models that have proved to be excellent tools to study the intracellular molecular mechanisms altered in liver tumori-genesis and summarise the most important and clinically relevant findings achieved.

Experimental models recapitulating overactivation of growth factor pathways

Both in vitro and in vivo studies provided early evidence of an association of transforming growth factor-alpha (TGF-α) overexpression and TGF-α-driven signalling activation with hepatocarcinogenesis. Thus, deregulation of the TGF-α signalling pathway is correlated with the

*Supported by an unrestricted educational grantfrom Merck Serono

A. SánchezDep. Bioquímica y Biología Molecular IIFacultad de FarmaciaUniversidad ComplutenseES-28040 Madrid, Spain

I. Fabregat (Y) Laboratori d’Oncologia MolecularInstitut d’Investigació Biomèdica de Bellvitge (IDIBELL)and Universitat de Barcelona (IDIBELL-UB)Gran Via de l’Hospitalet, 199ES-08907 L’Hospitalet, Barcelona, Spaine-mail: [email protected]

Clin Transl Oncol (2009) 11:208-214DOI 10.1007/s12094-009-0342-x

E D U C AT I O N A L S E R I E S R e d S e r i e s *

Genetically modified animal models recapitulating molecular events altered in human hepatocarcinogenesis

Aránzazu Sánchez · Isabel Fabregat

Received: 8 January 2009 / Accepted: 10 February 2009

CURRENT TECHNOLOGY IN CANCER RESEARCH AND TREATMENT

Clin Transl Oncol (2009) 11:208-214 209

transformation of liver epithelial cell lines by carcinogens and oncogenes [3, 4]. Mice overexpressing TGF-α showed increased hepatocyte proliferation and hyperplasia, which ultimately resulted in development of hepatocellular tu-mours [5–8]. Molecular characterisation of these tumours evidenced an enhanced expression of the endogenous c-myc and insulin-like growth factor II genes, suggesting that these, and possibly other factors, may collaborate in TGF-α-induced hepatocarcinogenesis [7].

Liver tumours occur in transgenic mice selectively expressing c-myc in the liver, but the tumour incidence in these c-myc transgenic mice is relatively low with a long latency period [9]. However, coexpression of TGF-α and c-myc transgenes leads to a tremendous acceleration of tumour growth [10], proving the cooperation or synergy between growth factors and oncogenes in tumorigenesis. Double transgenic c-myc/TGF-α mice have been particu-larly useful for delineating the molecular events occurring during HCC formation and progression. A quite thorough analysis of the preneoplastic and neoplastic lesions has been performed and a few important molecules and signal-ling pathways have been found to be altered and contribute to the neoplasia development and malignant conversion. The highly malignant phenotype of the HCC arising in these mice is the result of the increased proliferation via a disruption of the cyclin D/pRb/E2F pathway and the TGF-α-dependent reduced apoptosis [11, 12].

The discovery of abnormally elevated E2F activity in the HCC lesions in the double transgenic c-myc/TGF-α mice prompted Conner et al. [13] to generate liver specific-E2F transgenic mice. These animals displayed a 100% in-cidence of adenomas, but only some of these (about 30%) showed malignant transformation to HCC at 12 months. The authors argue that this is a reflection of the dual role of E2F-1, as oncogene, during the early stages of hepatocar-cinogenesis, mediated by a persistent increase in cell pro-liferation, and tumour suppressor, during the tumour pro-gression phase, mediated by a steady-state increase in the apoptotic rate, and potentially responsible for the delayed or diminished malignant conversion [13]. Similarly to the TGF-α transgenic mice, a strong induction of endogenous c-myc was observed in the E2F-transgenic livers, sug-gesting cooperation between these two oncogenes in liver oncogenesis. Supporting this hypothesis, double transgenic c-myc/E2F mice showed accelerated HCC development with 100% incidence of tumours by 6–8 months.

Interestingly, molecular characterisation of the tumours developed in the various animal models has revealed im-

portant differences. Thus, the c-myc/TGF-α tumours, char-acterised as fast growing and very aggressive, displayed extensive genomic instability and a low rate of beta-catenin activation while the c-myc or c-myc/E2F-1 tumours were characterised by a high frequency of beta-catenin activa-tion in a relatively stable genome [14–16]. These data were consistent with observations in human HCC showing that ß-catenin activation occurs in a HCC subset with a rela-tively stable genome [17, 18] and a more favourable prog-nosis [19]. Direct comparison of the global gene expression patterns of HCCs from these different mouse models and human HCCs showed that gene expression signatures re-flecting similar phenotypes are indeed conserved in mice and humans during tumour development. Thus, HCCs from c-myc, E2F1 and c-myc/E2F1 mice grouped together in the better survival subset of human HCC, while c-myc/TGF-α mouse HCCs fit into the poorer survival set of human HCC [20] (Table 1). These studies were exceedingly important for several reasons. On the one hand, they served to identify two classes of mouse models that appear to closely reca-pitulate the molecular patterns of the two subclasses of hu-man HCC: the c-myc/TGF-α HCCs typically having a poor prognosis, including earlier and higher incidence of HCC development, higher mortality and genomic instability; and the c-myc, E2F1 and c-myc/E2F1, which have a relatively higher frequency of ß-catenin mutations and nuclear accu-mulation, which are indicative of lower genomic instability and better prognosis. On the other hand, they demonstrated the usefulness of the genetically engineered mice for the study of human cancer and provided the basis for future studies aimed at better understanding the molecular patho-genesis of human neoplastic development. In addition to this, they hold promise for the discovery of relevant thera-peutic targets and the testing of potential therapeutic agents.

Hepatocyte growth factor (HGF) is a pleiotropic growth factor that regulates multiple cellular functions, including proliferation, differentiation, migration, morphogenesis, survival and angiogenesis, through binding to its tyrosine kinase receptor, Met [21–23]. The protooncogene c-Met is implicated in many types of malignant tumours [24, 25]. Activating mutations, amplification and increases in c-met and HGF mRNA and protein expression have been described in rodent and human HCC [26–29] but the rele-vance of these alterations for tumour formation or progres-sion remains somehow elusive. Transforming activity of HGF has been demonstrated in vitro utilising immortalised mouse hepatocytes [30]. However, both direct and inverse correlations between the expression of c-Met and HGF and

Table 1 TGF-α/c-Myc/E2F transgenic mouse models

TGF-α/c-Myc mice c-Myc/E2F mice

Highly malignant phenotype as a result of disruption Accelerated HCC development. 100% incidence of tumoursin the cyclinD/pRb/E2F pathway. Extensive genomic instability, by 6–8 months. High frequency of beta catenin activationlow rate of beta-catenin activation. in a relatively stable genome.Fit into the poorer survival set of human HCC Fit into the better survival subset of human HCC

210 Clin Transl Oncol (2009) 11:208-214

the growth, malignancy and metastatic potential of HCC cell lines have been reported [31, 32]. Moreover, HGF en-hances motility, but inhibits growth and induces apoptosis in spontaneously and aflatoxin-transformed rat liver epithe-lial (RLE) cells [33].

There are also conflictive reports in HGF and c-Met transgenic and knock-out mice. Overexpression of mouse HGF in transgenic mice under the metallothionein (MT-HGF) gene promoter resulted in spontaneous formation of benign and malignant liver tumours in most transgenic mice after 17 months of age, which was associated with a chronic activation of c-met [34]. MT-HGF overexpression also promoted DEN-induced hepatocarcinogenesis, which was often accompanied by abnormal blood vessel forma-tion and up-regulation of the expression of vascular en-dothelial growth factor (VEGF), suggesting that HGF can directly promote tumour progression through enhancement of angiogenesis and/or indirectly through VEGF induc-tion [35]. Evidence that Met can contribute to genesis and maintenance of HCC was provided by targeting expression of human Met in mouse hepatocytes. Tumours initiated by Met transgene regressed when the transgene was inactivat-ed, suggesting that continuous expression of Met is needed for the maintenance of the lesions [36].

In contrast, c-myc-driven hepatocarcinogenesis is strik-ingly inhibited by coexpression of HGF, which delayed the appearance of preneoplastic lesions and prevented malig-nant conversion. Furthermore, c-myc/HGF double trans-genic mice were completely resistant to tumour promotion by phenobarbital. These results suggested that HGF might function as a tumour suppressor during the early stages of liver carcinogenesis [37]. A similar inhibitory effect of HGF on HCC development was found in double transgenic mice HGF/TGF-α [38]. The puzzle is further amplified by recent data from Takami et al. showing that hepatocyte-specific c-Met conditional knockout mice display greatly increased susceptibility to N-nitrosodiethylamine (DEN)-induced hepatocarcinogenesis [39]. The tumour-promoting effect of c-Met signalling deficiency was associated with dysregula-tion of genes associated with stress responses and increased oxidative stress, and was reversed by administration of the antioxidant N-acetyl-L-cysteine, suggesting that intact HGF/c-Met signalling is critical for maintaining normal re-dox homeostasis and has tumour suppressor properties dur-ing the early stages of DEN-induced hepatocarcinogenesis.

The most illustrative demonstration of the importance of the HGF/c-Met signalling pathway in HCC has been provided by Kaposi-Novak et al. These authors defined a Met-dependent gene expression signature by comparing wt- and Met-deficient primary hepatocytes through mi-croarray analysis, and used it for comparative functional genomic analysis applied to human HCC samples [40]. This approach has revealed a clinically significant sub-group of human HCCs with a prominent Met-dependent gene expression signature, associated with aggressive phenotype and poor prognosis as evidenced by increased vascular invasion rate and microvessel density as well as

decreased mean survival time. These results demonstrated the predictive value of the Met expression signature on the disease outcome in HCC patients. Although further under-standing of the significance of HGF/c-Met signalling in carcinogenesis is needed, these data have important clinical implications and help validate Met as a potential target in human HCC therapy.

Experimental models recapitulating loss in tumour suppressor pathways

TGF-ß1 is a multifunctional cytokine that regulates many biological processes, including proliferation, differentiation and death, being considered a tumour suppressor. However, it also mediates disruption of cell adhesion, increase in mi-gration and invasion, immune suppression and angiogen-esis, being considered a tumour promoter [41]. The final effects might depend on cell context and microenviron-ment. In hepatocytes, TGF-ß1 is well known as an impor-tant regulatory suppressor factor that inhibits proliferation and induces cell death [42–44]. However, an overexpres-sion of TGF-ß1 has been found in human HCC [27, 45, 46] and hepatoblastomas [47], and during the early stages of hepatocarcinogenesis in mice co-expressing c-myc and TGF-α transgenes [11]. Furthermore, HCC cell lines show autocrine expression of TGF-ß [48].

Experiments in mouse models have revealed that over-expression of TGF-ß1 in the liver predisposes to both spon-taneous and chemically induced hepatocarcinogenesis [49]. Spontaneous development of hepatocellular tumours with a 59% incidence occurred at 16–18 months of age, which was enhanced and accelerated in the c-myc/TGF-ß1 mice. DEN treatment dramatically accelerated hepatocarcinogen-esis in the double transgenic mice compared to both single parental lines, leading to a higher tumour frequency and size, and a high frequency of malignant conversion. Inter-estingly, progression from benign to malignant phenotype correlated with an early and maintained loss of TGF-ßRII expression (78% of HCCs). Inactivation of TGF-ßRII seemed necessary but not sufficient for tumour progression because no tumours were detected in the single transgenic TGF-ß1 mice. These results suggested that TGF-ß1-in-duced transformation requires cooperation with additional mutations or genetic alterations, i.e., c-myc up-regulation, to overcome the inhibitory actions of TGF-ß1 and facilitate tumour development [49].

The importance of disruption of TGF-ß1 suppressor signalling, particularly at the receptor level, for hepatocel-lular carcinogenesis has also been demonstrated by using TßRII heterozygous knock-out mice. These mice did not develop tumours spontaneously but exhibited increased susceptibility to DEN-induced liver tumorigenesis [50], highlighting a role for TβRII as a suppressor of hepato-carcinogenesis. In agreement with this, reintroduction of the TβRII receptor in hepatoma cells from TβRII-negative

Clin Transl Oncol (2009) 11:208-214 211

cancers restores sensitivity to TGF-β1 and reduces their tumorigenicity [51]. Additionally, Santoni-Rugiu et al. demonstrated that enhancement of c-myc-induced hepato-carcinogenesis by co-expression of TGF-α is also associ-ated with early and frequent occurrence of TßRII-negative preneoplastic and neoplastic lesions and with reduced levels of p27 in HCC cells, indicating that disruption of TGF-ß1 growth-suppressive pathways may play a cru-cial role in the promotion and progression of liver cancer [52]. These observations supported a model in which the autocrine production of active TGF-ß1 by the hepatocytes creates a selective environment that facilitates transformed cells with down-regulated TßRII expression to escape from the growth-inhibitory effects of this cytokine and progress towards a more malignant phenotype.

It is now very well established that TGF-ß1 can induce both tumour suppressive and oncogenic properties [41]. Consistent with this, experiments with cultured hepatocytes and hepatoma cells have demonstrated that apart from inducing cell death, TGF-ß1 can activate antiapoptotic sig-nals [53–56]. Furthermore, TGF-ß1 induces an epithelial–mesenchymal transition [56–58], an important late event during development of epithelial tumours [59]. An associa-tion between the TGF-ß-induced growth arrest/apoptosis and EMT is suggested, since cells that have acquired a mesenchymal phenotype become refractory to TGF-ß sup-pressor effects [60–62]. These results support a crucial role for TGF-ß in tumour development and progression. The mechanisms that allow cells to escape from the inhibitory effects of TGF-ß are not completely understood. Interest-ingly, preneoplastic and neoplastic lesions arising from TGF-ß1, TGF-α/c-myc transgenic mice and human HCC cell lines showed constitutive activation of NF-kB signal-ling, which might contribute to accelerate epithelial neo-plastic progression, by protecting from TGF-ß1-induced apoptosis [63, 64]. In agreement with this, ROS-dependent

NFkB activation induced by TGF-ß1 in foetal hepatocytes mediates the up-regulation of EGFR ligands and the anti-apoptotic signals in TGF-ß-treated cells [65].

On the basis of the dual role of TGF-ß1 in tumorigene-sis, e.g., tumour-suppressive and tumour-promoting proper-ties, Coulouarn et al. have recently applied gene expression microarray technology to identify human HCC subgroups based on a TGF-ß1 signalling pathway-specific signa-ture, by direct comparison between TGF-ß1-treated wt vs. TßRII knock-out primary hepatocytes [66]. Interestingly, two subsets of TGF-ß-responsive genes, reflecting both the suppressive and oncogenic properties of TGF-ß, were iden-tified. Comparative functional genomic analysis identified subgroups of human HCC either positive or negative for the TGF-ß gene expression signature. Furthermore, within the TGF-ß-positive HCC subgroup, two distinct subsets of tumours that preferentially expressed early or late TGF-ß-responsive genes were discriminated. Importantly, these two distinct HCC subgroups differed greatly in survival and recurrence. Patients with the late TGF-ß signature had a considerably shortened mean survival time and increased tumour recurrence compared to the patients with the early TGF-ß signature (Fig. 1). These results constituted more proof for the usefulness of the experimental models and comparative functional genomic approaches for the mo-lecular classification of human HCC, and the clinical relevance and predictive and prognostic value of these ap-proaches for the development of personalised therapies.

Among the most common alterations observed in hu-man HCC are mutations in the p53 tumour suppressor gene (TP53) [67]. The presence of specific mutational hotspots in TP53 in different types of human cancer implicates en-vironmental carcinogens and endogenous processes. In this sense, somatic mutations at the third base in codon 249 of TP53 in HCC have been related to exposure to aflatoxin B1 (AFB1), in cooperation with HBV infection [67]. Chronic

Early stagesTGF-beta

Suppressor arm

Protumorigenic pathways

Late stagesTGF-beta

Suppressor arm

Protumorigenic pathways

Early TGF-β signature in HCC patients. Better prognosis

Late TGF-β signature in HCCpatients. Shortened mean survival

time and increased tumor recurrence

Fig. 1 Dual role of TGF-ß in liver tumorigenesis. In early stages preneoplastic cells respond to TGF-ß inhibiting growth and inducing apoptosis, being considered a tumour suppressor factor. Later stages correlate with molecular alterations that allow cells to escape from its suppressor effects, favouring responses that mediate cell migration and invasion

212 Clin Transl Oncol (2009) 11:208-214

infection with HBV and HCV viruses and exposure to oxi-dative stress, including haemochromatosis or inflammation, induce damage in the DNA and mutations in cancer-related genes, including TP53. Thus, it would seem plausible that p53 mutation might operate in either HCC initiation or pro-gression, depending on the context. Indeed, Trp53 knockout mice develop more metastatic tumours than the wild-type mice when HCC is induced by using somatic delivery of oncogene-bearing avian retroviral vectors to the liver [68]. Concomitant loss of the Ink4a/Arf tumour suppressor locus accelerated tumour formation and metastasis in the same animal model, suggesting potential roles for the p16 and p19 tumour suppressors in this process [69]. However, in spite of these clear results, adenoviral delivery of p53 re-combinant DNA into mouse models bearing HCCs did not apparently suppress tumour growth [70]. Farazi et al., in a recent work [71], have helped to clarify this point. They have demonstrated that the effect of p53 loss in chronic liver disease-associated HCC is dependent on cellular context, in particular, intact or dysfunctional telomeres. In mice, reduction in telomere length is not clearly observed, which is related to the existence of long telomeres and high expression of telomerase (mTert). However, in Tert–/– mice p53 mutation enabled advanced HCC disease [71]. These authors propose that in the face of chronic liver damage, at-tenuated p53 function and telomere-induced chromosomal instability might play critical and cooperative roles in the progression of HCC. In the context of intact telomeres, p53 mutation might not have such a relevant effect on liver tum-origenesis.

Experimental models recapitulating inflammation- associated hepatocellular carcinoma

Knock-out animals of the multidrug-resistant gene Mdr2 represent a model of inflammation-associated HCC. The liver-specific phospho-glycoprotein responsible for the transport of phosphatidylcholine through the bile canali-cular membrane is absent in these animals, which results in bile regurgitation into the portal tracts, with the sub-sequent portal inflammation and fibrosis, reminiscent of human intrahepatic cholestasis [72, 73]. Due to the liver inflammation the Mdr2–/– mice develop hepatocyte dys-plasia and show liver tumorigenic foci at 16 months of age (almost 100%). Gene expression profiling has shown that Mdr2–/– animals differ from other published murine HCC models and are of great interest because they share several important deregulated pathways and many coordi-nately differentially expressed genes with human data sets [74]. The Mdr2–/– mouse may serve as a model for the ß-catenin-negative subgroup of human HCCs characterised by low nuclear cyclin D1 levels and down-regulation of multiple tumour suppressor genes. Most Mdr2–/– liver samples were more similar to the better-survival human HCC samples and better-survival-like mouse models.

A major link between inflammation and cancer is pro-vided by NF-kappaB transcription factors. Different stud-ies in genetically modified animals have indicated that IkappaB kinase beta (IKKbeta), required for NF-kappaB activation, links chronic inflammation with liver carcino-genesis. Indeed, mice lacking IKKbeta only in hepatocytes (Ikkbeta(Deltahep) mice) exhibited a marked increase in chemical hepatocarcinogenesis caused by DEN. This corre-lated with enhanced reactive oxygen species (ROS) produc-tion, increased c-Jun N-terminal kinase (JNK) activation and hepatocyte death, which give rise to augmented compensato-ry proliferation of surviving hepatocytes [75, 76]. However, decreased hepatocarcinogenesis was found in mice lacking IKKbeta in both hepatocytes and haematopoietic-derived Kupffer cells, due to the lack of inflammatory processes. These results indicated that chemical liver carcinogenesis depends on inflammation and suggested the usefulness of anti-inflammatory intervention targeting Kupffer cells in chemoprevention of HCC. Chemicals or viruses that inter-fere with NF-κB activation in hepatocytes, but not in sur-rounding cells, might promote HCC development (Fig. 2).

A relevant aspect of HCC epidemiology is the gender difference of incidence. Men are about three to five times more likely to develop HCC than women. Similar, or even more pronounced, gender differences are observed in rodent chemical-induced HCC models. Interleukin-6 knock-out (IL-6–/–) mice have helped to understand this gender disparity. IL-6 is a multifunctional cytokine largely responsible for the hepatic response to systemic inflam-mation (acute phase response). IL-6 concentrations are increased in patients with HCC [77]. IL-6 is necessary for compensatory regeneration in chemical models of hepatocarcinogenesis [78] and deletion of the suppressor of cytokine signalling-3 (SOCS3), a negative regulator of IL-6-related cytokines, enhances hepatitis- or chemical-induced liver carcinogenesis [79, 80]. In a recent work, Naugler et al. have elegantly described that ablation of IL-6 in IL-6–/– mice abolished the gender differences in a model of DEN-induced hepatocarcinogenesis. Oestrogen inhibited secretion of IL-6 from Kupffer cells exposed to necrotic hepatocytes and reduced circulating concentra-tions of IL-6 in DEN-treated male mice [81]. These results further evidence the connection between inflammation and HCC and open the way for therapeutic anti-inflammatory approaches to prevent or control this malignancy.

In conclusion, genetic modification of growth fac-tor pathways has revealed the relevance of the epidermal growth factor receptor (EGFR) ligands in hepatocarcino-genesis, which in cooperation with c-myc might mimic the poorer survival set of human HCC. Results obtained after genetic modification of the HGF/c-Met or TGF-ß pathways have revealed that they play dual roles both as suppres-sor, in early stages, or promoters, in later stages, of HCC. Genetic modification of the p53 pathway has revealed its relevance in hepatocarcinogenesis, particularly under telomere-induced chromosomal instability. Finally, differ-ent genetically modified mice have pointed out the straight

Clin Transl Oncol (2009) 11:208-214 213

link between inflammation and liver carcinogenesis, NF-kappaB and IL-6 pathways playing pivotal roles.

Acknowledgements The authors acknowledge the help of Javier Marquez in the writing of this manuscript.

NF-kappaB Kupffer cellsViral infections

Chemicals

Inflammatory cytokinesGrowth and survival factors

Angiogenic factors

PreneoplasticLiver cell

Growth, survival, invasive properties

Fig. 2 NF-kappaB activation in inflammatory cells can promote malignant conversion and liver tumour progression. Viral infections or toxic reactions activate inflammatory cells to produce cytokines and growth factors that aug-ment compensatory proliferation of surround-ing cells. Chemicals or viruses that interfere with NF-kappaB activation in hepatocytes, but not in preneoplastic cells, would promote HCC development

References

1. Farazi PA, DePinho RA (2006) Hepatocellular carcinoma pathogenesis: from genes to environ-ment. Nat Rev Cancer 6:674–687

2. Newell P, Villanueva A, Friedman SL et al (2008) Experimental models of hepatocellular carcinoma. J Hepatol 48:858–879

3. Lee LW, Raymond VW, Tsao MS et al (1991) Clonal cosegregation of tumorigenicity with over-expression of c-myc and transforming growth fac-tor alpha genes in chemically transformed rat liver epithelial cells. Cancer Res 51:5238–5244

4. Presnell SC, Thompson MT, Strom SC (1995) Investigation of the cooperative effects of trans-forming growth factor alpha and c-myc overex-pression in rat liver epithelial cells. Mol Carcinog 13:233–244

5. Sandgren EP, Luetteke NC, Palmiter RD et al (1990) Overexpression of TGF alpha in transgenic mice: in-duction of epithelial hyperplasia, pancreatic metapla-sia, and carcinoma of the breast. Cell 61:1121–1135

6. Lee GH, Merlino G, Fausto N (1992) Develop-ment of liver tumors in transforming growth factor alpha transgenic mice. Cancer Res 52:5162–5170

7. Takagi H, Sharp R, Hammermeister C et al (1992) Molecular and genetic analysis of liver oncogene-sis in transforming growth factor alpha transgenic mice. Cancer Res 52:5171–5177

8. Webber EM, Wu JC, Wang L et al (1994) Over-expression of transforming growth factor-alpha causes liver enlargement and increased hepatocyte proliferation in transgenic mice. Am J Pathol 145:398–408

9. Sandgren EP, Quaife CJ, Pinkert CA et al (1989) Oncogene-induced liver neoplasia in transgenic mice. Oncogene 4:715–724

10. Murakami H, Sanderson ND, Nagy P et al (1993) Transgenic mouse model for synergistic effects of nuclear oncogenes and growth factors in tum-origenesis: interaction of c-myc and transforming growth factor alpha in hepatic oncogenesis. Can-cer Res 53:1719–1723

11. Santoni-Rugiu E, Nagy P, Jensen MR et al (1996) Evolution of neoplastic development in the liver of transgenic mice co-expressing c-myc and transform-ing growth factor-alpha. Am J Pathol 149:407–428

12. Santoni-Rugiu E, Jensen MR, Thorgeirsson SS (1998) Disruption of the pRb/E2F pathway and in-hibition of apoptosis are major oncogenic events in liver constitutively expressing c-myc and transform-ing growth factor alpha. Cancer Res 58:123–134

13. Conner EA, Lemmer ER, Omori M et al (2000) Du-al functions of E2F-1 in a transgenic mouse model of liver carcinogenesis. Oncogene 19:5054–5062

14. Sargent LM, Sanderson ND, Thorgeirsson SS (1996) Ploidy and karyotypic alterations asso-ciated with early events in the development of hepatocarcinogenesis in transgenic mice harbor-ing c-myc and transforming growth factor alpha transgenes. Cancer Res 56:2137–2142

15. Calvisi DF, Factor VM, Loi R, Thorgeirsson SS (2001) Activation of beta-catenin during hepato-carcinogenesis in transgenic mouse models: re-lationship to phenotype and tumor grade. Cancer Res 61:2085–2091

16. Calvisi DF, Ladu S, Conner EA et al (2004) Disregulation of E-cadherin in transgenic mouse models of liver cancer. Lab Invest 84:1137–1147

17. Legoix P, Bluteau O, Bayer J et al (1999) Beta-catenin mutations in hepatocellular carcinoma correlate with a low rate of loss of heterozygosity. Oncogene 8:4044–4046

18. Hsu HC, Jeng YM, Mao TL et al (2000) Beta-catenin mutations are associated with a subset of low-stage hepatocellular carcinoma negative for hepatitis B virus and with favorable prognosis. Am J Pathol 157:763–770

19. Laurent-Puig P, Legoix P, Bluteau O et al (2001) Genetic alterations associated with hepatocellular carcinomas define distinct pathways of hepatocar-cinogenesis. Gastroenterology 120:1763–1773

20. Lee JS, Chu IS, Mikaelyan A et al (2004) Ap-plication of comparative functional genomics to identify best-fit mouse models to study human cancer. Nat Genet 36:1306–1311

21. Zarnegar R, Michalopoulos GK (1995) The many faces of hepatocyte growth factor: from hepatopoi-esis to hematopoiesis. J Cell Biol 129:1177–1180

22. Matsumoto K, Nakamura T (1996) Emerging multipotent aspects of hepatocyte growth factor. J Biochem 119:591–600

23. Furge KA, Zhang YW, Vande Woude GF (2000) Met receptor tyrosine kinase: enhanced signaling through adapter proteins. Oncogene 19:5582–5589

24. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF (2003) Met, metastasis, motility and more. Nat Rev Mol Cell Biol 4:915–925

25. Gentile A, Comoglio PM (2004) Invasive growth: a genetic program. Int J Dev Biol 48:451–456

26. Imai T, Masui T, Nakanishi H et al (1996) Ex-pression of hepatocyte growth factor and c-met mRNAs during rat chemically induced hepatocar-cinogenesis. Carcinogenesis 17:19–24

214 Clin Transl Oncol (2009) 11:208-214

27. Kiss A, Wang NJ, Xie JP, Thorgeirsson SS (1997) Analysis of transforming growth factor (TGF)-alpha/epidermal growth factor receptor, hepato-cyte growth Factor/c-met,TGF-beta receptor type II, and p53 expression in human hepatocellular carcinomas. Clin Cancer Res 3:1059–1066

28. Ueki T, Fujimoto J, Suzuki T et al (1997) Expres-sion of hepatocyte growth factor and its receptor, the c-met proto-oncogene, in hepatocellular carci-noma. Hepatology 25:619–623

29. Park WS, Dong SM, Kim SY et al (1999) Somatic mutations in the kinase domain of the Met/hepato-cyte growth factor receptor gene in childhood he-patocellular carcinomas. Cancer Res 59:307–310

30. Kanda H, Tajima H, Lee GH et al (1993) Hepato-cyte growth factor transforms immortalized mouse liver epithelial cells. Oncogene 8:3047–3053

31. Shiota G, Rhoads DB, Wang TC et al (1992) Hepa-tocyte growth factor inhibits growth of hepatocellu-lar carcinoma cells. Proc Natl Acad Sci 89:373–377

32. Xie Q, Liu K-D, Hu M-Y, Zhou K (2001) SF/HGF-c-Met autocrine and paracrine promote me-tastasis of hepatocellular carcinoma. World J Gas-troenterol 7:816–820

33. Conner EA, Wirth PJ, Kiss A et al (1997) Growth inhibition and induction of apoptosis by HGF in transformed rat liver epithelial cells. Biochem Biophys Res Commun 236:396–401

34. Sakata H, Takayama H, Sharp R et al (1996) He-patocyte growth factor/scatter factor overexpres-sion induces growth, abnormal development, and tumor formation in transgenic mouse livers. Cell Growth Differ 7:1513–523

35. Horiguchi N, Takayama H, Toyoda M et al (2002) Hepatocyte growth factor promotes hepatocarcino-genesis through c-Met autocrine activation and enhanced angiogenesis in transgenic mice treated with diethylnitrosamine. Oncogene 21:1791–1799

36. Wang R, Ferrell LD, Faouzi S et al (2001) Ac-tivation of the Met receptor by cell attachment induces and sustains hepatocellular carcinomas in transgenic mice. J Cell Biol 153:1023–1034

37. Santoni-Rugiu E, Preisegger KH, Kiss A et al (1996) Inhibition of neoplastic development in the liver by hepatocyte growth factor in a transgenic mouse model. Proc Natl Acad Sci USA 93:9577–9582

38. Shiota G, Kawasaki H, Nakamura T, Schmidt EV (1995) Characterization of double transgenic mice expressing hepatocyte growth factor and trans-forming growth factor alpha. Res Commun Mol Pathol Pharmacol 90:17–24

39. Takami T, Kaposi-Novak P, Uchida K et al (2007) Loss of hepatocyte growth factor/c-Met signaling pathway accelerates early stages of N-nitrosodi-ethylamine induced hepatocarcinogenesis. Cancer Res 67:9844–9851

40. Kaposi-Novak P, Lee J-S, Gómez-Quiroz L et al (2006) Met-regulated expression signature defines a subset of human hepatocellular carcinomas with poor prognosis and aggressive phenotype. J Clin Invest 116:1582–1595

41. Massague J (2008) TGFbeta in cancer. Cell 134: 215–230

42. Carr BI, Hayashi I, Branum EL, Moses HL (1986) Inhibition of DNA synthesis in rat hepatocytes by platelet-derived type beta transforming growth factor. Cancer Res 46:2330–2334

43. Oberhammer FA, Pavelka M, Sharma S et al (1992) Induction of apoptosis in cultured hepa-tocytes and in regressing liver by transforming growth factor beta 1. Proc Natl Acad Sci USA 89:5408–5412

44. Sánchez A, Alvarez AM, Benito M, Fabregat I (1996) Apoptosis induced by transforming growth factor-beta in fetal hepatocyte primary cultures: involvement of reactive oxygen intermediates. J Biol Chem 271:7416–7422

45. Ito N, Kawata S, Tamura S et al (1991) Elevated levels of transforming growth factor beta messen-ger RNA and its polypeptide in human hepatocel-lular carcinoma. Cancer Res 51:4080–4083

46. Bedossa P, Peltier E, Terris B et al (1995) Trans-forming growth factor ß1 (TGF-ß1) and TGF-ß1

receptors in normal, cirrhotic, and neoplastic hu-man livers. Hepatology 21:760–766

47. Luo JH, Ren B, Keryanov S et al (2006) Tran-scriptomic and genomic analysis of human he-patocellular carcinomas and hepatoblastomas. Hepatology 44:1012–1024

48. Matsuzaki K, Date M, Furukawa F et al (2000) Regulatory mechanisms for transforming growth factor beta as an autocrine inhibitor in human hepatocellular carcinoma: implications for roles of smads in its growth. Hepatology 32:218–227

49. Factor VM, Kao C-Y, Santoni-Rugiu E et al (1997) Constitutive expression of mature Trans-forming growth factor ß1 in the liver accelerates hepatocarcinogenesis in transgenic mice. Cancer Res 57:2089–2095

50. Im Y-H, Kim HT, Kim IY et al (2001) Heterozy-gous mice for the transforming growth factor-ß type II receptor gene have increased susceptibil-ity to hepatocellular carcinogenesis. Cancer Res 61:6665–6668

51. Inagaki M, Moustakas A, Lin HY et al (1993) Growth inhibition by transforming growth fac-tor-ß (TGF-ß) type I is restored in TGF-ß resistant hepatoma cells after expression of TGF-ß receptor type II cDNA. Proc Natl Acad Sci USA 90:5359–5363

52. Santoni-Rugiu E, Jensen MR, Factor VM, Thor-geirsson SS (1999) Acceleration of c-myc-induced hepatocarcinogenesis by Co-expression of trans-forming growth factor (TGF)-alpha in transgenic mice is associated with TGF-beta1 signaling dis-ruption. Am J Pathol 154:1693–1700

53. Valdés F, Murillo MM, Valverde AM et al (2004) Transforming growth factor-beta activates both pro-apoptotic and survival signals in fetal rat he-patocytes. Exp Cell Res 292:209–218

54. Murillo MM, Del Castillo G, Sanchez A et al (2005) Involvement of EGF receptor and c-Src in the survival signals induced by TGF-beta1 in hepatocytes. Oncogene 24:4580–4587

55. Shin I, Bakin AV, Rodeck U et al (2001) Trans-forming growth factor beta enhances epithelial cell survival via Akt-dependent regulation of FKHRL1. Mol Biol Cell 12:3328–3339

56. Caja L, Ortiz C, Bertran E et al (2007) Differ-ential intracellular signaling induced by TGF-beta in rat adult hepatocytes and hepatoma cells: implications in liver carcinogenesis. Cell Signal 19:683–694

57. Gotzmann J, Huber H, Thallinger C et al (2002) Hepatocytes convert to a fibroblastoid pheno-type through the cooperation of TGF-beta1 and Ha-Ras: steps towards invasiveness. J Cell Sci 115:1189–1202

58. Valdés F, Alvarez AM, Locascio A et al (2002) The epithelial mesenchymal transition confers resistance to the apoptotic effects of transforming growth factor Beta in fetal rat hepatocytes. Mol Cancer Res 1:68–78

59. Thiery JP (2003) Epithelial-mesenchymal transi-tions in development and pathologies. Curr Opin Cell Biol 15:740–746

60. Nicolás FJ, Lehmann K, Warne PH et al (2003) Epithelial to mesenchymal transition in Madin-Darby canine kidney cells is accompanied by down-regulation of Smad3 expression, leading to resistance to transforming growth factor-beta-induced growth arrest. J Biol Chem 278:3251–3256

61. Del Castillo G, Murillo MM, Alvarez-Barrientos A et al (2006) Autocrine production of TGF-beta confers resistance to apoptosis alter an epithe-lial-mesenchymal transition process in hepato-cytes: Role of EGF receptor ligands. Exp Cell Res 312:2860–2871

62. Gal A, Sjöblom T, Fedorova L et al (2008) Sus-tained TGF beta exposure suppresses Smad and non-Smad signaling in mammary epithelial cells, leading to EMT and inhibition of growth arrest and apoptosis. Oncogene 27:1218–1230

63. Arsura M, Panta GR, Bilyeu JD et al (2003) Tran-sient activation of NF-kappaB through a TAK1/

IKK kinase pathway by TGF-beta1 inhibits AP-1/SMAD signaling and apoptosis: implications in liver tumor formation. Oncogene 22:412–425

64. Cavin LG, Romieu-Mourez R, Panta GR et al (2003) Inhibition of CK2 activity by TGF-b1 promotes IkB-a protein stabilization and apoptosis of immor-talized hepatocytes. Hepatology 38:1540–1551

65. Murillo MM, Carmona-Cuenca I, del Castillo G et al (2007) Activation of NADPH oxidase by transforming growth factor-beta in hepato-cytes mediates up-regulation of epidermal growth factor receptor ligands through a nuclear fac-tor-kappaB-dependent mechanism. Biochem J 405:251–259

66. Coulouarn C, Factor VM, Thorgeirsson SS (2008) Transforming growth factor-β gene expression sig-nature in mouse hepatocytes predicts clinical out-come in human cancer. Hepatology 47:2059–2067

67. Hussain SP, Schwank J, Staib F et al (2007) TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene 26:2166–2176

68. Lewis BC, Klimstra DS, Socci ND et al (2005) The absence of p53 promotes metastasis in a novel somatic mouse model for hepatocellular carcinoma. Mol Cell Biol 25:1228–1237

69. Chen YW, Klimstra DS, Mongeau ME et al (2007) Loss of p53 and Ink4a/Arf cooperate in a cell autonomous fashion to induce metastasis of hepatocellular carcinoma cells. Cancer Res 67:7589–7596

70. Bao JJ, Zhang WW, Kuo MT (1996) Adenoviral delivery of recombinant DNA into transgenic mice bearing hepatocellular carcinomas. Hum Gene Ther 7:355–365

71. Farazi PA, Glickman J, Horner J, Depinho RA (2006) Cooperative interactions of p53 mutation, telomere dysfunction, and chronic liver damage in hepatocellular carcinoma progression. Cancer Res 66:4766–4773

72. Mauad TH, van Nieuwkerk CM, Dingemans KP et al (1994) Mice with homozygous disruption of the mdr2 P-glycoprotein gene. A novel animal model for studies of nonsuppurative inflamma-tory cholangitis and hepatocarcinogenesis. Am J Pathol 145:1237–1245

73. Fickert P, Fuchsbichler A, Wagner M et al (2004) Regurgitation of bile acids from leaky bile ducts causes sclerosing cholangitis in Mdr2 (Abcb4) knockout mice. Gastroenterology 127:261–274

74. Katzenellenbogen M, Mizrahi L, Pappo O et al (2007) Molecular mechanisms of liver carcino-genesis in the mdr2-knockout mice. Mol Cancer Res 5:1159–1170

75. Maeda S, Kamata H, Luo JL et al (2005) IKKbeta couples hepatocyte death to cytokine-driven com-pensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 121:977–990

76. Sakurai T, Maeda S, Chang L, Karin M (2006) Loss of hepatic NF-kappa B activity enhances chemical hepatocarcinogenesis through sustained c-Jun N-terminal kinase 1 activation. Proc Natl Acad Sci USA 103:10544–10551

77. Soresi M, Giannitrapani L, D’Antona F et al (2006) Interleukin-6 and its soluble receptor in patients with liver cirrhosis and hepatocellular carcinoma. World J Gastroenterol 12:2563–2568

78. Cressman DE, Greenbaum LE, DeAngelis RA et al (1996) Liver failure and defective hepatocyte regeneration in interleukin-6-deficient mice. Sci-ence 274:1379–1383

79. Ogata H, Kobayashi T, Chinen T et al (2006) Deletion of the SOCS3 gene in liver parenchymal cells promotes hepatitis-induced hepatocarcino-genesis. Gastroenterology 131:179–193

80. Riehle KJ, Campbell JS, McMahan RS et al (2008) Regulation of liver regeneration and hepa-tocarcinogenesis by suppressor of cytokine signal-ing 3. J Exp Med 205:91–103

81. Naugler WE, Sakurai T, Kim S et al (2007) Gen-der disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science 317:121–124