Telomeres in Liver Cancer: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Oncology
Contributor: , , ,

Liver cancer is one of the most common cancer types worldwide and the fourth leading cause of cancer-related death. Liver carcinoma is distinguished by a high heterogeneity in pathogenesis, histopathology and biological behavior. Dysregulated signaling pathways and various gene mutations are frequent in hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA), which represent the two most common types of liver tumors. Both tumor types are characterized by telomere shortening and reactivation of telomerase during carcinogenesis.

The observation that TERT promoter mutations occur early during liver carcinogenesis highlights the importance of telomerase activity for tumor cell survival. Two possible scenarios are conceivable how telomerase contributes to tumorigenesis in liver cancer: On the one side, telomerase reactivation before entering the crisis checkpoint may stabilize critically short telomeres, providing growth advantage for cells with oncogenic mutations. On the other side, early reactivation of telomerase may be related to its non-canonical functions. 

Similarities and differences between HCC and iCCA in telomere biology are depicted in this review article.

  • liver cancer
  • hepatocellular carcinoma
  • intrahepatic cholangiocarcinoma
  • telomere shortening
  • TERT promoter mutation
  • telomerase

Liver cancer is one of the most common cancer types worldwide and the fourth leading cause of cancer-related death. Liver carcinoma is distinguished by a high heterogeneity in pathogenesis, histopathology and biological behavior. Dysregulated signaling pathways and various gene mutations are frequent in hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA), which represent the two most common types of liver tumors. Both tumor types are characterized by telomere shortening and reactivation of telomerase during carcinogenesis. Continuous cell proliferation, e.g., by oncogenic mutations, can cause extensive telomere shortening in the absence of sufficient telomerase activity, leading to dysfunctional telomeres and genome instability by breakage–fusion–bridge cycles, which induce senescence or apoptosis as a tumor suppressor mechanism. Telomerase reactivation is required to stabilize telomere functionality and for tumor cell survival, representing a genetic risk factor for the development of liver cirrhosis and liver carcinoma. Therefore, telomeres and telomerase could be useful targets in hepatocarcinogenesis. Here, we review similarities and differences between HCC and iCCA in telomere biology.

1. Introduction

 

Liver cancer is predicted to be the sixth most common tumor disease worldwide and the fourth leading cause of cancer-related death [‎1]. Liver cancer presents with a high heterogeneity in pathogenesis, histopathology and biological behavior. The heterogeneous disease in terms of etiologies reflects the poor prognosis of patients with liver cancer. The two most common types of liver cancer are hepatocellular carcinoma (HCC) (75–85% of cases) and intrahepatic cholangiocarcinoma (iCCA) (10–15% of cases) [1]. Most liver carcinomas are diagnosed at advanced stages despite the surveillance program of patients with liver cirrhosis to diagnose early liver tumors. To date, therapy options are limited to the multikinase inhibitors sorafenib and lenvatinib as first-line treatment options and regorafenib and cabozantinib as second-line treatment options for liver cancer patients [2][3][4]. A recent clinical trial revealed significantly longer overall and progression-free survival in patients with unresectable hepatocellular carcinoma, who received atezolizumab, a programmed death ligand 1 (PD-L1) inhibitor, combined with bevacizumab, a monoclonal antibody targeting the vascular endothelial growth factor (VEGF), in comparison to sorafenib only [5]. These findings point to new treatment options in patients with unresectable hepatocellular carcinoma and support the development of new therapy options.

The major risk factors in hepatocarcinogenesis are chronic viral infections with hepatitis B virus (HBV) and hepatitis C virus (HCV), heavy alcohol consumption, obesity, type 2 diabetes, smoking and long-term exposure to aflatoxin B [6]. Most liver tumors arise on the basis of chronic liver diseases and often result in liver fibrosis and cirrhosis formation, which itself represents a risk factor for tumor development. In western countries, HCC is mainly related to HCV, high alcohol consumption and non-alcoholic steatohepatitis (NASH) and connected with cirrhosis formation [7][8], whereas in Asia, most HCC patients are related to HBV infection; in addition, HCC can also develop in normal liver without fibrosis/cirrhosis or liver with limited fibrosis formation [9]. Interestingly, in Japan, chronic HCV infection is more common than HBV, and HCV infection accounts for the majority of HCC [10]. The burden of hepatocellular carcinoma is continuously growing due to increased rates of obesity, type 2 diabetes and nonalcoholic fatty liver disease (NAFLD), especially in low-risk HCC areas and thereby replaces viral- and alcohol-related chronic liver diseases [11]. Due to the diversity of risk factors, a high heterogeneity in liver tumors is reported. The molecular heterogeneity in terms of various gene mutations in liver cancer requires the identification of molecular targets for designing individualized therapies. Recent studies have described various sub-classification of HCC and iCCA tumor types [12][13][14]. Individual studies underline the importance of specific treatment options based on the tumor subtypes as a key to achieving a better overall survival of liver cancer patients [15][16][17]. The identification of dysregulated molecular pathways in premalignant lesions is required for an early disease detection in hepatocarcinogenesis [18]. A detailed description of molecular targets in the diversity of liver cancer subtypes would be beneficial for targeted therapies.

Telomere shortening and reactivation of telomerase, two common hallmarks of carcinogenesis, are described in a broad range of human cancers, including liver cancer [19][20][21]. Telomere shortening and reactivation of telomerase, through TERT promoter mutations, for example, represent genetic risk factors for the development of liver cirrhosis and liver cancer [22]. Therefore, disruptions in telomere biology could be a useful target for the treatment of liver cancer. In the following, we will present the current knowledge of telomere biology in HCC and iCCA.

2. Telomere Shortening in Liver Cirrhosis and Hepatocellular Carcinoma

 

Chronic liver disease is associated with chronic liver inflammation, which can lead to cell death and compensatory cell regeneration. The liver is characterized by a high regenerative reserve [23], which decreases in the context of chronic liver disease, consequently leading to telomere shortening and limiting the regenerative reserve of the liver. The frequent appearance of senescent hepatocytes in liver cirrhosis is the result of the loss of telomeric repeats and the extensive proliferation [24][25][26][27]. These senescent hepatocytes exhibit markers like p16INK4a and p21WAF1/Cip1 and are positive for senescence-associated β-galactosidase staining. A disruption of the p53-signaling pathway overcomes the senescence checkpoint and leads to further cell division of hepatocytes with already-shortened telomeres until the telomeres become critically short. At this point, the cells enter the crisis checkpoint, which is characterized by massive cell death [27][28].

In the liver, telomerase activity is downregulated during early embryonic development, and telomerase activity is absent in the adult liver. The healthy liver is a slowly proliferating organ, and most hepatocytes are in a quiescent stage (only one out of 20,000 cells (0.005%) is in the cell cycle [29]). Thus, telomerase activity seems not to be essential for hepatocyte function in a healthy liver. However, telomere shortening occurs in the absence of sufficient telomerase activity in hepatocytes under conditions of chronic liver diseases or upon injury [25]. Importantly, low levels of telomerase activity were observed under regenerative conditions, indicating the potential physiological activation of telomerase in adult hepatocytes [30][31]. So far, two distinct mechanisms were made responsible for telomere shortening upon increased proliferative signals, either due to oncogene activation (e.g., Ras mutations or c-Myc amplification) or expression of viral oncogenic proteins: (i) in most of the cases, absence of sufficient telomerase reactivation [32][33] and (ii) in some cases, germline mutations within the coding region of telomerase, which impair the enzymatic activity of telomerase in proliferating hepatocytes (see Section 5) [34][35].

Reactivation of telomerase activity has been shown in more than 80% of HCCs, which suggests that telomerase activation is a rate-limiting process for liver cancer formation [36][37]. The reactivation of telomerase correlates with the upregulation of both essential components TERT and TERC, respectively. It has been reported that the re-expression of TERT and activation of telomerase occurs at early premalignant stages in regenerative nodules and cirrhotic livers [38][39]. Importantly, telomerase activity was detected both in HCC and in iCCA to a similar extent (80–85%) [40]. Thus, it is important to emphasize at this point that the majority of both HCCs and iCCAs are similarly telomerase-positive, highlighting the necessity of telomerase activity for telomere functionality and tumor progression.

Irrespective of the mechanism, insufficient telomerase activity leads to accelerated telomere shortening in proliferating liver cells and as a result to genomic instability by breakage–fusion–bridge cycles (see review by Meena et al. [41]). In cells with intact DNA-damage response (DDR) checkpoints, telomere shortening leads to senescence/apoptosis and functions as a tumor suppressor mechanism. On the other hand, in cells lacking functional DDR, telomere shortening promotes genome instability and tumor formation.

Consequently, telomere shortening is an important risk factor for tumor initiation in liver carcinogenesis. The risk of tumor formation drastically increases at the cirrhosis stage, which is characterized by increased hepatocyte senescence, and upon further cell division at the crisis checkpoint by apoptosis of hepatocytes. Several studies have shown that telomere shortening is more pronounced in liver carcinoma compared to the surrounding liver tissue (see review by Satyanarayana et al. [42]). Furthermore, the progressive shortening of telomeres and the inactivation of cell cycle checkpoints in premalignant lesions led to the identification of a preneoplastic-sequence in human hepatocarcinogenesis, suggesting that small cell changes (SCC) are more advanced precursor lesions compared to large cell changes (LCC) [43]. In addition, telomere shortening was more pronounced in HCCs with a high degree of aneuploidy compared to diploid HCCs [44][45]. In fact, several studies provide evidence for a role of telomere shortening in the induction of chromosomal instability and increased risk for tumor formation [46][47][48][49]. The importance of telomere shortening and dysfunctional telomeres in HCC initiation was shown in transgenic mouse models (see Section 3).

3. Mouse Models of Telomere Dysfunction in Hepatocarcinogenesis

 

To understand the severe situation of dysfunctional telomeres and telomere shortening during chronic liver disease, transgenic mouse models were used to analyze the functions of telomeres. To this end, it is important to note that, firstly, there is a substantial difference in the regulation of telomerase between mouse and human liver. Telomerase activity is detectable in resting mouse liver but not in resting human liver [50][51][52][53]. The limiting component, restricting telomerase activity in human tissues, is the catalytic subunit of the telomerase TERT [54]. Concordantly, there is a marked difference in TERT mRNA levels in human and mouse livers [52][53][54]. In fact, in vivo experimental evidence supports the idea that the species-specific differential regulation is based on different promoter organization [53][55]. Secondly, the average telomere length in laboratory mice is about five times longer than that of human telomeres, to some extent due to constitutive telomerase activity in mouse cells [56][57].

The telomerase knockout mouse (Terc−/−) lacking the RNA component of the telomerase enzyme was used to analyze telomere shortening in liver regeneration, chronic liver disease, and hepatocarcinogenesis [58][59][60]. Mice are characterized by the existence of longer telomeres compared to humans [56]. Mice of different backgrounds differ a lot in telomere length [61]. For this reason, the Terc−/− mouse has to be crossed until the third to the sixth generation, depending on the used background strain to generate mice with critically short telomere lengths [58][60][62]. In an experimental model of liver regeneration involving the removal of two-thirds of the liver by partial hepatectomy of G3 Terc−/− and Terc+/+ mice, telomere shortening was observed to be a heterogeneous event at the cellular level, which led to the inhibition of a subpopulation of cells with critically short telomeres to enter the cell cycle and prevent those cells from participating in liver regeneration [58]. By comparing the mean telomere fluorescence intensities measured by FISH analysis, Satyanarayana and colleagues [58] observed no significant differences in TERC+/+ mice between BrdU-positive cells (952.53 ± 144.19) and BrdU-negative cells (957.44 ± 130.57) but saw a dramatic reduction of the mean telomere fluorescence intensity of BrdU-negative cells (364.94 ± 116.45) in comparison to BrdU-positive cells (509.65 ± 101.30) in G3 TERC−/− mice. In a mouse model of experimentally induced acute liver damage in which Terc−/− mice were subjected to genetic, surgical and chemical impairment of the liver, dysfunctional telomeres were associated with defective liver regeneration and accelerated formation of liver cirrhosis, which could be partly rescued by adenoviral delivery of the telomerase RNA [24]. In an approach comprising three different cancer-prone model systems—1) treatment with CCl4 (carbon tetrachloride), (2) treatment with DEN (diethylnitrosamine) and (3) a genetic model (urokinase plasminogen activator transgenic mice) in Terc−/− mice—it could be shown that telomere dysfunction has a differential impact on tumor initiation and tumor progression. In all three model systems, dysfunctional telomeres were associated with higher amounts of tumor initiation and a decline in tumor progression [63]. In mouse models of chronic liver damage achieved by crossing HBsAg-expressing mice (the mice express the hepatitis B surface antigen under the liver-specific albumin promoter [64]) with Terc−/− mice, contrary effects of telomere shortening were shown between the beneficial effect on suppression of tumor growth and the negative effect on organismal survival [59]. In another mouse model of chronic liver disease, HBsAg mice were crossed with Terc−/− and Trp53 cKO mice. We generated mice with critically short telomeres by an intercross of Terc−/− and Terc+/− to generate siblings with loss of telomerase function in one group and telomerase expression in the other group. This study yielded the evidence for telomerase to be a critical component in the progression of Trp53-deficient hepatocellular carcinoma with short telomeres in the setting of chronic liver damage [60]. In addition, it was also shown that telomerase limits the accumulation of telomere dysfunction and the generation of aneuploidy by the activation of TRP53-independent checkpoints which suppress carcinogenesis [60]. Increased rates of chromosomal aberrations could be also shown in a DEN-induced liver cancer mouse model with dysfunctional telomeres. Telomerase knockout mice (Terc−/−) with chronic telomere dysfunction as well as a model of transient telomere dysfunction by inducing a dominant-negative variant of the TRF2 (telomeric repeat-binding factor 2) protein exhibited higher levels of chromosomal aberrations. In summary, the model of transient telomere dysfunction promotes chromosomal instability and liver carcinogenesis in telomerase-competent mice [65]. RAP1 (Ras-proximate-1 or Ras-related protein 1), like TRF2, is a component of the shelterin complex, which caps the telomere end for the protection of chromosome ends [66]. A recent publication suggested an important role of RAP1 in the protection of liver damage and liver carcinogenesis. DEN-induced Rap1−/− female mice were more prone to liver damage and hepatocellular carcinoma [67]. These models reflect the complexity and opposing roles of dysfunctional telomeres in hepatocarcinogenesis.

4. Telomere Shortening in Cholangiocarcinoma

 

Intrahepatic cholangiocarcinoma (iCCA) is the second most common malignant liver tumor which arises from the biliary tract and is characterized by a very poor prognosis with rising incidence and mortality in recent years [68][69]. The main risk factors described for HCC are also reported for iCCA. Additional risk factors are primary sclerosing cholangitis (PSC), hepatobiliary flukes, biliary duct cysts and hepatolithiasis. A study by Verma and colleagues analyzed telomere shortening during aging in normal liver with no history of liver disease. Interestingly, they observed that the cholangiocytes exhibited the longest telomeres compared to all other analyzed intrahepatic lineages [70]. Similar to CD4+ and CD8+ lymphocytes, no significant telomere shortening was observed in cholangiocytes and hepatocytes of individuals without liver disease during aging. The authors only observed an age-related telomere shortening in Kupffer cells and stellate cells [70]. On the other hand, a consistent telomere shortening was reported during the development of biliary tract carcinoma, starting early in carcinogenesis in the inflamed biliary tract, metaplasia, dysplasia and carcinoma [71]. In contrast, the normal and the inflamed epithelium of the biliary tract showed a uniform telomere length [71]. Within cholangiocarcinoma, a frequent intratumoral heterogeneity of telomere length is reported [71]. As indicated above, telomere shortening in hepatocytes triggers cellular senescence in the context of intact DDR checkpoints. Similarly, an investigation of telomere shortening and senescence in the pathogenesis of primary biliary cirrhosis (PBC) showed telomere shortening. Moreover, DNA damage accumulation was detectable in biliary epithelial cells in the damaged small bile ducts and bile ductules in PBC in comparison to normal-looking bile ducts and bile ductules in PBC, chronic viral hepatitis and normal livers [72]. Of note, the accumulation of DNA damage foci correlated with increased expression of p16INK4a and p21WAF1/Cip1, which characterize biliary cellular senescence [72].

5. Loss of Function Mutations in Telomerase Components

 

Germline loss-of-function mutations in the telomerase components were found in a variety of human diseases, including dyskeratosis congenita, aplastic anemia, familial idiopathic fibrosis and acute myeloid leukemia [73][74][75][76][77][78][79][80]. These mutations provoked an impaired tissue regeneration due to telomere dysfunction and stem/progenitor cell exhaustion. Similar mutations were also reported in a subset of liver cancer samples. The authors analyzed TERT and TERC mutations in buccal mucosa tissue and peripheral blood of patients with liver cirrhosis and compared them with healthy non-cirrhotic controls. An increased number of telomerase mutations were found in the group with liver cirrhosis. The study by Calado et al. reported nine patients with a mutation in the TERT gene and one patient with a mutation in the TERC gene among 134 patients with liver cirrhosis. Similarly, Hartmann et al.  reported mutations in the TERT and TERC genes in 16 out of 521 patients. The Calado study reported a significantly higher allele frequency for the gene variants in the TERT and TERC genes in patients with cirrhosis (allele frequency 0.037) compared to controls (0.008; p = 0.0011). A similar result was shown by the Hartmann study, which stated an increased incidence of telomerase mutations detected in cirrhosis patients (allele frequency 0.017) compared to non-cirrhotic controls (0.003, p = 0.0007). The mutations in telomerase components led to decreased telomerase activity in comparison to wildtype telomerase enzyme activity. Consequently, patients with these mutations showed shorter telomeres in peripheral white blood cells . Rare TERT mutations were also reported in patients with nonalcoholic fatty liver disease (NAFLD). Here, an enrichment of TERT mutations could be found in NAFLD-associated HCC [81]. Functional evaluation of these mutations exposed reduced protein synthesis from some of the mutations compared to the TERT wild-type protein. It is speculated that these TERT mutations could also impair the DNA-binding function of TERT. In summary, these results indicate that TERT mutations result in impaired telomerase activity, accelerated telomere shortening and impaired regeneration in chronic liver disease. These findings are supported by the above-mentioned studies indicating low/absent telomerase activity in resting liver and telomerase activation in the regenerating liver . Taken together, these studies show that telomerase activity acts as a protective mechanism in chronic diseases to prevent telomere shortening during accelerated cell proliferation, whereas TERT mutations result in telomere shortening and may promote hepatocarcinogenesis by dysfunctional telomeres.

This entry is adapted from the peer-reviewed paper 10.3390/cancers12082048

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J. Clin. 2018, 68, 394–424, doi:10.3322/caac.21492.
  2. Llovet, J.M.; Ricci, S.; Mazzaferro, V.M.; Hilgard, P.; Gane, E.; Blanc, J.-F.; De Oliveira, A.C.; Santoro, A.; Raoul, J.L.; Forner, A.; et al. Sorafenib in Advanced Hepatocellular Carcinoma. N. Engl. J. Med. 2008, 359, 378–390, doi:10.1056/nejmoa0708857.
  3. Bruix, J.; Qin, S.; Merle, P.; Granito, A.; Huang, Y.-H.; Bodoky, G.; Pracht, M.; Yokosuka, O.; Rosmorduc, O.; Breder, V.V.; et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 389, 56–66, doi:10.1016/s0140-673632453-9.
  4. Kudo, M.; Finn, R.S.; Qin, S.; Han, K.-H.; Ikeda, K.; Piscaglia, F.; Baron, A.; Park, J.W.; Han, G.; Jassem, J.; et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: A randomised phase 3 non-inferiority trial. Lancet 2018, 391, 1163–1173, doi:10.1016/S0140-673630207-1.
  5. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905, doi:10.1056/nejmoa1915745.
  6. London, W.T.; Petrick, J.L.; McGlynn, K.A. Liver Cancer. In: Cancer Epidemiology and Prevention Thun, M.J., Linet, M.S., Cerhan, J.R., Haiman, C.A., Schottenfeld, D., Eds 4th ed Oxford University Press, Cambridge, MA, USA 2018 pp. 635–660.
  7. Llovet, J.M.; Zucman-Rossi, J.; Pikarsky, E.; Sangro, B.; Schwartz, M.; Sherman, M.; Gores, G. Hepatocellular carcinoma. Nat. Rev. Dis. Prim. 2016, 2, 16019, doi:10.1038/nrdp.2016.19.
  8. El-Serag, H.B. Hepatocellular Carcinoma. J. Clin. Gastroenterol. 2002, 35, S72–S78, doi:10.1097/00004836-200211002-00002.
  9. Nault, J.-C. Pathogenesis of hepatocellular carcinoma according to aetiology. Best Pr. Res. Clin. Gastroenterol. 2014, 28, 937–947, doi:10.1016/j.bpg.2014.08.006.
  10. Umemura, T.; Ichijo, T.; Yoshizawa, K.; Tanaka, E.; Kiyosawa, K. Epidemiology of hepatocellular carcinoma in Japan. J. Gastroenterol. 2009, 44, 102–107, doi:10.1007/s00535-008-2251-0.
  11. Marengo, A.; Rosso, C.; Bugianesi, E. Liver Cancer: Connections with Obesity, Fatty Liver, and Cirrhosis. Annu. Rev. Med. 2016, 67, 103–117, doi:10.1146/annurev-med-090514-013832.
  12. Zucman‐Rossi, J.; Villanueva, A.; Nault, J.-C.; Llovet, J.M. Genetic Landscape and Biomarkers of Hepatocellular Carcinoma. Gastroenterology 2015, 149, 1226–1239.e4, doi:10.1053/j.gastro.2015.05.061.
  13. Moeini, A.; Sia, D.; Bardeesy, N.; Mazzaferro, V.; Llovet, J.M. Molecular Pathogenesis and Targeted Therapies for Intrahepatic Cholangiocarcinoma. Clin. Cancer Res. 2015, 22, 291–300, doi:10.1158/1078-0432.ccr-14-3296.
  14. Haga, H.; Patel, T. Molecular diagnosis of intrahepatic cholangiocarcinoma. J. Hepato-Biliary-Pancreatic Sci. 2014, 22, 114–23, doi:10.1002/jhbp.156.
  15. Dauch, D.; Rudalska, R.; Cossa, G.; Nault, J.C.; Kang, T.W.; Wuestefeld, T.; Hohmeyer, A.; Imbeaud, S.; Yevsa, T.; Hoenicke, L.; et al. A MYC–aurora kinase A protein complex represents an actionable drug target in p53-altered liver cancer. Nat. Med. 2016, 22, 744–753, doi:10.1038/nm.4107.
  16. Fujita, M.; Yamaguchi, R.; Hasegawa, T.; Shimada, S.; Arihiro, K.; Hayashi, S.; Maejima, K.; Nakano, K.; Fujimoto, A.; Ono, A.; et al. Classification of primary liver cancer with immunosuppression mechanisms and correlation with genomic alterations. EBioMedicine 2020, 53, 102659, doi:10.1016/j.ebiom.2020.102659.
  17. Ahn, K.S.; O’Brien, D.; Kang, Y.N.; Mounajjed, T.; Kim, Y.H.; Kim, T.S.; Kocher, J.A.; Allotey, L.K.; Borad, M.J.; Roberts, L.R.; et al. Prognostic subclass of intrahepatic cholangiocarcinoma by integrative molecular-clinical analysis and potential targeted approach. Hepatol Int. 2019, 13, 490–500.
  18. Yang, Y.; Lin, X.; Lu, X.; Luo, G.; Zeng, T.; Tang, J.; Jiang, F.; Li, L.; Cui, X.; Huang, W.; et al. Interferon-microRNA signalling drives liver precancerous lesion formation and hepatocarcinogenesis. Gut 2016, 65, 1186–201, doi:10.1136/gutjnl-2015-310318.
  19. Kim, N.W.; A.; Piatyszek, M.; Prowse, K.R.; Harley, C.B.; West, M.D.; Ho, P.L.; Coviello, G.M.; E.; Wright, W.; Weinrich, S.L.; Shay, J.W. Specific association of human telomerase activity with immortal cells and cancer. Science 1994, 266, 2011–2015, doi:10.1126/science.7605428.
  20. Shay, J.; Bacchetti, S. A survey of telomerase activity in human cancer. Eur. J. Cancer 1997, 33, 787–791, doi:10.1016/s0959-804900062-2.
  21. Miura, N.; Horikawa, I.; Nishimoto, A.; Ohmura, H.; Ito, H.; Hirohashi, S.; Shay, J.W.; Oshimura, M. Progressive telomere shortening and telomerase reactivation during hepatocellular carcinogenesis. Cancer Genet. Cytogenet. 1997, 93, 56–62, doi:10.1016/s0165-460800329-9.
  22. Kumar, M.; Lechel, A.; Günes, C. Telomerase: The Devil Inside. Genes 2016, 7, 43, doi:10.3390/genes7080043.
  23. Michalopoulos, G.K.; DeFrances, M.C. Liver regeneration. Science 1997, 276, 60–66.
  24. Rudolph, K.L.; Chang, S.; Millard, M.; Schreiber-Agus, N.; Depinho, R.A. Inhibition of Experimental Liver Cirrhosis in Mice by Telomerase Gene Delivery. Science 2000, 287, 1253–1258, doi:10.1126/science.287.5456.1253.
  25. Wiemann, S.U.; Satyanarayana, A.; Tsahuridu, M.; Tillmann, H.L.; Zender, L.; Klempnauer, J.; Flemming, P.; Franco, S.; Blasco, M.A.; Manns, M.P.; et al. Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis. FASEB J. 2002, 16, 935–942.
  26. Kitada, T.; Seki, S.; Kawakita, N.; Kuroki, T.; Monna, T. Telomere Shortening in Chronic Liver Diseases. Biochem. Biophys. Res. Commun. 1995, 211, 33–39, doi:10.1006/bbrc.1995.1774.
  27. Urabe, Y.; Nouso, K.; Higashi, T.; Nakatsukasa, H.; Hino, N.; Ashida, K.; Kinugasa, N.; Yoshida, K.; Uematsu, S.; Tsuji, T. Telomere length in human liver diseases. Liver Int. 2008, 16, 293–297, doi:10.1111/j.1600-0676.1996.tb00748.x.
  28. Paradis, V.; Youssef, N.; Dargere, D.; Bâ, N.; Bonvoust, F.; Deschatrette, J.; Bedossa, P. Replicative senescence in normal liver, chronic hepatitis C, and hepatocellular carcinomas. Hum. Pathol. 2001, 32, 327–332, doi:10.1053/hupa.2001.22747.
  29. Mangnall, D.; Bird, N.C.; Majeed, A.W. The molecular physiology of liver regeneration following partial hepatectomy. Liver Int. 2003, 23, 124–138, doi:10.1034/j.1600-0676.2003.00812.x.
  30. Sirma, H.; Kumar, M.; Meena, J.K.; Witt, B.; Weise, J.M.; Lechel, A.; Ande, S.; Sakk, V.; Guguen–Guillouzo, C.; Zender, L.; et al. The Promoter of Human Telomerase Reverse Transcriptase Is Activated During Liver Regeneration and Hepatocyte Proliferation. Gastroenterology 2011, 141, 326–337.e3, doi:10.1053/j.gastro.2011.03.047.
  31. Kotoula, V.; Hytiroglou, P.; Pyrpasopoulou, A.; Saxena, R.; Thung, S.N.; Papadimitriou, C.S. Expression of human telomerase reverse transcriptase in regenerative and precancerous lesions of cirrhotic livers. Liver Int. 2002, 22, 57–69, doi:10.1046/j.0106-9543.2001.01594.x.
  32. Allsopp, R.C.; Chang, E.; Kashefi-Aazam, M.; Rogaev, E.I.; Piatyszek, M.A.; Shay, J.W.; Harley, C.B. Telomere Shortening Is Associated with Cell Division in Vitro and in Vivo. Exp. Cell Res. 1995, 220, 194–200, doi:10.1006/excr.1995.1306.
  33. Holt, S.; Gollahon, L.; Willingham, T.; Barbosa, M.; Shay, J. p53 levels in human mammary epithelial cells expressing wild-type and mutant human papillomavirus type 16 (HPV-16) E6 proteins. Int. J. Oncol. 1996, 8, 263–270, doi:10.3892/ijo.8.2.263.
  34. Calado, R.T.; Brudno, J.; Mehta, P.; Kovacs, J.J.; Wu, C.; Zago, M.A.; Chanock, S.J.; Boyer, T.D.; Young, N.S. Constitutional telomerase mutations are genetic risk factors for cirrhosis. Hepatology 2011, 53, 1600–1607, doi:10.1002/hep.24173.
  35. Hartmann, D.; Srivastava, U.; Thaler, M.; Kleinhans, K.N.; N’Kontchou, G.; Scheffold, A.; Bauer, K.; Kratzer, R.F.; Kloos, N.; Katz, S.-F.; et al. Telomerase gene mutations are associated with cirrhosis formation. Hepatology 2011, 53, 1608–1617, doi:10.1002/hep.24217.
  36. Tahara, H.; Nakanishi, T.; Kitamoto, M.; Nakashio, R.; Shay, J.W.; Tahara, E.; Kajiyama, G.; Ide, T. Telomerase activity in human liver tissues: Comparison between chronic liver disease and hepatocellular carcinomas. Cancer Res. 1995, 55, 2734–2736.
  37. Ide, T.; Tahara, H.; Nakashio, R.; Kitamoto, M.; Nakanishi, T.; Kajiyama, G. Telomerase in hepatocellular carcinogenesis. Hum. Cell 1996, 9, 283–286.
  38. Hytiroglou, P.; Kotoula, V.; Thung, S.N.; Tsokos, M.; Fiel, M.I.; Papadimitriou, C.S. Telomerase activity in precancerous hepatic nodules. Cancer 1998, 82, 1831–1838.
  39. Youssef, N.; Paradis, V.; Ferlicot, S.; Bedossa, P. In situdetection of telomerase enzymatic activity in human hepatocellular carcinogenesis. J. Pathol. 2001, 194, 459–465, doi:10.1002/path.901.
  40. Ozaki, S.; Harada, K.; Sanzen, T.; Watanabe, K.; Tsui, W.M.S.; Nakanuma, Y. In situ nucleic acid detection of human telomerase in intrahepatic cholangiocarcinoma and its preneoplastic lesion. Hepatology. 1999, 30, 914–919, doi:10.1002/hep.510300419.
  41. Meena, J.; Rudolph, K.L.; Günes, C. Telomere Dysfunction, Chromosomal Instability and Cancer. Recent Results Cancer Res. 2015, 200, 61–79.
  42. Satyanarayana, A.; Manns, M.P.; Rudolph, K.L. Telomeres and telomerase: A dual role in hepatocarcinogenesis. Hepatology 2004, 40, 276–283, doi:10.1002/hep.20308.
  43. Plentz, R.R.; Park, Y.N.; Lechel, A.; Kim, H.; Nellessen, F.; Langkopf, B.H.E.; Wilkens, L.; Destro, A.; Fiamengo, B.; Manns, M.P.; et al. Telomere shortening and inactivation of cell cycle checkpoints characterize human hepatocarcinogenesis. Hepatology 2007, 45, 968–976, doi:10.1002/hep.21552.
  44. Plentz, R.R.; Caselitz, M.; Bleck, J.S.; Gebel, M.; Flemming, P.; Kubicka, S.; Manns, M.P.; Rudolph, K.L. Hepatocellular telomere shortening correlates with chromosomal instability and the development of human hepatoma. Hepatology 2004, 40, 80–86, doi:10.1002/hep.20271.
  45. Plentz, R.R.; Schlegelberger, B.; Flemming, P.; Gebel, M.; Kreipe, H.; Manns, M.P.; Rudolph, K.L.; Wilkens, L. Telomere shortening correlates with increasing aneuploidy of chromosome 8 in human hepatocellular carcinoma. Hepatology 2005, 42, 522–526, doi:10.1002/hep.20847.
  46. Davoli, T.; Denchi, E.L.; De Lange, T. Persistent Telomere Damage Induces Bypass of Mitosis and Tetraploidy. Cell 2010, 141, 81–93, doi:10.1016/j.cell.2010.01.031.
  47. Davoli, T.; De Lange, T. Telomere-Driven Tetraploidization Occurs in Human Cells Undergoing Crisis and Promotes Transformation of Mouse Cells. Cancer Cell 2012, 21, 765–776, doi:10.1016/j.ccr.2012.03.044.
  48. Pampalona, J.; Soler, D.; Genesca, A.; Tusell, L. Whole chromosome loss is promoted by telomere dysfunction in primary cells. Genes, Chromosom. Cancer 2010, 49, 368–378, doi:10.1002/gcc.20749.
  49. Pampalona, J.; Frías, C.; Genesca, A.; Tusell, L. Progressive Telomere Dysfunction Causes Cytokinesis Failure and Leads to the Accumulation of Polyploid Cells. PLoS Genet. 2012, 8, e1002679, doi:10.1371/journal.pgen.1002679.
  50. Prowse, K.R.; Greider, C.W. Developmental and tissue-specific regulation of mouse telomerase and telomere length. Proc. Natl. Acad. Sci. USA 1995, 92, 4818–4822, doi:10.1073/pnas.92.11.4818.
  51. Wright, W.E.; Piatyszek, M.A.; Rainey, W.E.; Byrd, W.; Shay, J.W. Telomerase activity in human germline and embryonic tissues and cells. Dev Genet. 1996, 18, 173–179.
  52. Ritz, J.M.; Kühle, O.; Riethdorf, S.; Sipos, B.; Deppert, W.; Englert, C.; Gunes, C. A Novel Transgenic Mouse Model Reveals Humanlike Regulation of an 8-kbp Human TERT Gene Promoter Fragment in Normal and Tumor Tissues. Cancer Res. 2005, 65, 1187–1196, doi:10.1158/0008-5472.can-04-3046.
  53. Horikawa, I.; Chiang, Y.J.; Patterson, T.; Feigenbaum, L.; Leem, S.-H.; Michishita, E.; Larionov, V.; Hodes, R.J.; Barrett, J.C. Differential cis-regulation of human versus mouse TERT gene expression in vivo: Identification of a human-specific repressive element. Proc. Natl. Acad. Sci. USA 2005, 102, 18437–18442, doi:10.1073/pnas.0508964102.
  54. Kolquist, K.A.; Ellisen, L.W.; Counter, C.M.; Meyerson, M.M.; Tan, L.K.; Weinberg, R.A.; Haber, D.A.; Gerald, W.L. Expression of TERT in early premalignant lesions and a subset of cells in normal tissues. Nat. Genet. 1998, 19, 182–186, doi:10.1038/554.
  55. A.; Greenberg, R.; O’Hagan, R.C.; Deng, H.; Xiao, Q.; Hann, S.R.; Adams, R.R.; Lichtsteiner, S.; Chin, L.; Morin, G.B.; A. DePinho, R. Telomerase reverse transcriptase gene is a direct target of c-Myc but is not functionally equivalent in cellular transformation. Oncogene 1999, 18, 1219–1226, doi:10.1038/sj.onc.1202669.
  56. Kipling, D.; Cooke, H.J. Hypervariable ultra-long telomeres in mice. Nature 1990, 347, 400–402, doi:10.1038/347400a0.
  57. Kipling, D. Telomere structure and telomerase expression during mouse development and tumorigenesis. Eur. J. Cancer 1997, 33, 792–800, doi:10.1016/s0959-804900060-9.
  58. Satyanarayana, A.; Wiemann, S.; Buer, J.; Lauber, J.; Dittmar, K.; Wüstefeld, T.; A Blasco7M.; Manns, M.; Rudolph, K.L. Telomere shortening impairs organ regeneration by inhibiting cell cycle re-entry of a subpopulation of cells. EMBO J. 2003, 22, 4003–4013, doi:10.1093/emboj/cdg367.
  59. Wiemann, S.U.; Satyanarayana, A.; Buer, J.; Kamino, K.; Manns, M.P.; Rudolph, K.L. Contrasting effects of telomere shortening on organ homeostasis, tumor suppression, and survival during chronic liver damage. Oncogene 2004, 24, 1501–1509, doi:10.1038/sj.onc.1208308.
  60. Lechel, A.; Holstege, H.; Begus, Y.; Schienke, A.; Kamino, K.; Lehmann, U.; Kubicka, S.; Schirmacher, P.; Jonkers, J.; Rudolph, K.L. Telomerase Deletion Limits Progression of p53-Mutant Hepatocellular Carcinoma With Short Telomeres in Chronic Liver Disease. Gastroenterology 2007, 132, 1465–1475, doi:10.1053/j.gastro.2007.01.045.
  61. Hemann, M.T. Wild-derived inbred mouse strains have short telomeres. Nucleic Acids Res. 2000, 28, 4474–4478, doi:10.1093/nar/28.22.4474.
  62. Rudolph, K.L.; Chang, S.; Lee, H.-W.; A Blasco7M.; Gottlieb, G.J.; Greider, C.W.; Depinho, R.A.; Longevity, Stress Response, and Cancer in Aging Telomerase-Deficient Mice. Cell 1999, 96, 701–712, doi:10.1016/s0092-867480580-2.
  63. A.; Farazi, P.; Glickman, J.; Jiang, S.; Yu, A.; Rudolph, K.L.; A. DePinho, R. Differential impact of telomere dysfunction on initiation and progression of hepatocellular carcinoma. Cancer Res. 2003, 63, 5021–5027.
  64. Chisari, F.V.; Klopchin, K.; Moriyama, T.; Pasquinelli, C.; Dunsford, H.A.; Sell, S.; Pinkert, C.; Brinster, R.L.; Palmiter, R.D. Molecular pathogenesis of hepatocellular carcinoma in hepatitis B virus transgenic mice. Cell 1989, 59, 1145–1156, doi:10.1016/0092-867490770-8.
  65. Begus-Nahrmann, Y.; Hartmann, D.; Kraus, J.; Eshraghi, P.; Scheffold, A.; Grieb, M.; Rasche, V.; Schirmacher, P.; Lee, H.-W.; Kestler, H.A.; et al. Transient telomere dysfunction induces chromosomal instability and promotes carcinogenesis. J. Clin. Investig. 2012, 122, 2283–8, doi:10.1172/JCI61745.
  66. De Lange, T. Shelterin: The protein complex that shapes and safeguards human telomeres. Genes Dev. 2005, 19, 2100–2110, doi:10.1101/gad.1346005.
  67. Ferrara-Romeo, I.; Martínez, P.; A Blasco7M. Mice lacking RAP1 show early onset and higher rates of DEN-induced hepatocellular carcinomas in female mice. PLoS ONE 2018, 13, e0204909, doi:10.1371/journal.pone.0204909.
  68. Bridgewater, J.; Galle, P.R.; Khan, S.A.; Llovet, J.M.; Park, J.-W.; Patel, T.; Pawlik, T.M.; Gores, G.J. Guidelines for the diagnosis and management of intrahepatic cholangiocarcinoma. J. Hepatol. 2014, 60, 1268–1289, doi:10.1016/j.jhep.2014.01.021.
  69. Njei, B. Changing pattern of epidemiology in intrahepatic cholangiocarcinoma. Hepatol. 2014, 60, 1107–1108, doi:10.1002/hep.26958.
  70. Verma, S.; Tachtatzis, P.; Penrhyn-Lowe, S.; Scarpini, C.G.; Jurk, D.; Zglinicki, T.; Coleman, N.; Alexander, G.J.M. Sustained telomere length in hepatocytes and cholangiocytes with increasing age in normal liver. Hepatology 2012, 56, 1510–1520, doi:10.1002/hep.25787.
  71. E.; Hansel, D.; Meeker, A.K.; Hicks, J.; De Marzo, A.M.; Lillemoe, K.D.; Schulick, R.; Hruban, R.H.; Maitra, A.; Argani, P. Telomere length variation in biliary tract metaplasia, dysplasia, and carcinoma. Mod. Pathol. 2006, 19, 772–779, doi:10.1038/modpathol.3800591.
  72. Sasaki, M.; Ikeda, H.; Yamaguchi, J.; Nakada, S.; Nakanuma, Y. Telomere shortening in the damaged small bile ducts in primary biliary cirrhosis reflects ongoing cellular senescence. Hepatològy 2008, 48, 186–195, doi:10.1002/hep.22348.
  73. Heiss, N.S.; Knight, S.W.; Vulliamy, T.; Klauck, S.M.; Wiemann, S.; Mason, P.J.; Poustka, A.; Dokal, I. X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions. Nat. Genet. 1998, 19, 32–38, doi:10.1038/ng0598-32.
  74. Vulliamy, T.; Marrone, A.; Goldman, F.; Dearlove, A.; Bessler, M.; Mason, P.J.; Dokal, I. The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature 2001, 413, 432–435, doi:10.1038/35096585.
  75. Fogarty, P.F.; Yamaguchi, H.; Wiestner, A.; Baerlocher, G.M.; Sloand, E.; Zeng, W.; Read, E.J.; Lansdorp, P.M.; Young, N.S. Late presentation of dyskeratosis congenita as apparently acquired aplastic anaemia due to mutations in telomerase RNA. Lancet 2003, 362, 1628–1630, doi:10.1016/s0140-673614797-6.
  76. Yamaguchi, H.; Calado, R.T.; Ly, H.; Kajigaya, S.; Baerlocher, G.M.; Chanock, S.J.; Lansdorp, P.M.; Young, N.S. Mutations inTERT,the Gene for Telomerase Reverse Transcriptase, in Aplastic Anemia. New Engl. J. Med. 2005, 352, 1413–1424, doi:10.1056/nejmoa042980.
  77. Armanios, M.Y.; Chen, J.J.-L.; Cogan, J.; Alder, J.K.; Ingersoll, R.G.; Markin, C.; Lawson, W.E.; Xie, M.; Vulto, I.; Phillips, J.A.; et al. Telomerase Mutations in Families with Idiopathic Pulmonary Fibrosis. New Engl. J. Med. 2007, 356, 1317–1326, doi:10.1056/nejmoa066157.
  78. Tsakiri, K.D.; Cronkhite, J.T.; Kuan, P.J.; Xing, C.; Raghu, G.; Weissler, J.C.; Rosenblatt, R.L.; Shay, J.W.; Garcia, C.K. Adult-onset pulmonary fibrosis caused by mutations in telomerase. Proc. Natl. Acad. Sci. USA; 2007 104, 7552–7557.
  79. Calado, R.T.; Regal, J.A.; Hills, M.; Yewdell, W.T.; Dalmazzo, L.F.; Zago, M.A.; Lansdorp, P.M.; Hogge, N.; Chanock, S.J.; Estey, E.H.; et al. Constitutional hypomorphic telomerase mutations in patients with acute myeloid leukemia. Proceedings of the National Academy of Sciences; Proc. Natl. Acad. Sci. USA, 2009; 106, 1187–1192.
  80. Kirwan, M.; Vulliamy, T.; Marrone, A.; Walne, A.J.; Beswick, R.; Hillmen, P.; Kelly, R.; Stewart, A.; Bowen, D.; Schönland, S.; et al. Defining the pathogenic role of telomerase mutations in myelodysplastic syndrome and acute myeloid leukemia. Hum. Mutat. 2009, 30, 1567–1573, doi:10.1002/humu.21115.
  81. Donati, B.; Pietrelli, A.; Pingitore, P.; Dongiovanni, P.; Caddeo, A.; Walker, L.; Baselli, G.; Pelusi, S.; Rosso, C.; Vanni, E.; et al. Telomerase reverse transcriptase germline mutations and hepatocellular carcinoma in patients with nonalcoholic fatty liver disease. Cancer Med. 2017, 6, 1930–1940, doi:10.1002/cam4.1078.
More
This entry is offline, you can click here to edit this entry!