J. Bruix and M. Colombo, Hepatocellular carcinoma: current state of the art in diagnosis and treatment, Best practice & research, Clinical gastroenterology, vol.28, p.751, 2014.

A. C. Mcpherron, A. M. Lawler, and S. J. Lee, Regulation of anterior/posterior patterning of the axial skeleton by growth/differentiation factor 11, Nat Genet, vol.22, pp.260-264, 1999.

R. Essalmani, A. Zaid, J. Marcinkiewicz, A. Chamberland, A. Pasquato et al.,

A. Seidah and . Prat, In vivo functions of the proprotein convertase PC5/6 during mouse development: Gdf11 is a likely substrate, Proc Natl Acad Sci U S A, vol.105, pp.5750-5755, 2008.

C. E. Brun and M. A. Rudnicki, GDF11 and the Mythical Fountain of Youth, Cell Metab, vol.22, pp.54-56, 2015.

M. Sinha, Y. C. Jang, J. Oh, D. Khong, E. Y. Wu et al.,

F. S. Regalado, J. R. Loffredo, M. F. Pancoast, J. Hirshman, and J. L. Lebowitz,

M. Shadrach, M. J. Cerletti, T. Kim, L. J. Serwold, B. Goodyear et al.,

A. J. Lee and . Wagers, Restoring systemic GDF11 levels reverses age-related dysfunction in mouse skeletal muscle, Science, vol.344, pp.649-652, 2014.

F. S. Loffredo, M. L. Steinhauser, S. M. Jay, J. Gannon, J. R. Pancoast et al.,

M. Yalamanchi, C. Sinha, D. Dall'osso, J. L. Khong, C. M. Shadrach et al.,

A. Singer, N. Stewart, R. E. Psychogios, A. J. Gerszten, M. J. Hartigan et al.,

A. J. Serwold, R. T. Wagers, and . Lee, Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy, Cell, pp.828-839, 2013.

M. A. Egerman, S. M. Cadena, J. A. Gilbert, A. Meyer, H. N. Nelson et al.,

C. Swalley, C. Mallozzi, L. L. Jacobi, I. Jennings, G. Clay et al.,

E. Brachat, T. Lach-trifilieff, A. U. Shavlakadze, A. S. Trendelenburg, and D. J. Brack,

. Glass, GDF11 Increases with Age and Inhibits Skeletal Muscle Regeneration, Cell Metab, vol.22, pp.164-174, 2015.

D. J. Glass, Elevated GDF11 Is a Risk Factor for Age-Related Frailty and Disease in Humans, Cell Metab, vol.24, pp.7-8, 2016.

M. J. Schafer, E. J. Atkinson, P. M. Vanderboom, B. Kotajarvi, and T. A. White,

M. M. Moore, C. J. Bruce, K. L. Greason, R. M. Suri, S. Khosla et al.,

3. Bergen and N. K. Lebrasseur, Quantification of GDF11 and Myostatin in Human Aging and Cardiovascular Disease, Cell Metab, vol.23, pp.1207-1215, 2016.

G. Finkenzeller, G. B. Stark, and S. Strassburg, Growth differentiation factor 11 supports migration and sprouting of endothelial progenitor cells, J Surg Res, vol.198, pp.50-56, 2015.

F. Jeanplong, S. J. Falconer, J. M. Oldham, N. J. Maqbool, M. Thomas et al.,

C. D. Hennebry and . Mcmahon, Identification and expression of a novel transcript of the growth and differentiation factor-11 gene, Mol Cell Biochem, vol.390, pp.9-18, 2014.

K. K. Gokoffski, H. H. Wu, C. L. Beites, J. Kim, E. J. Kim et al.,

A. D. Johnson, A. L. Lander, and . Calof, Activin and GDF11 collaborate in feedback control of neuroepithelial stem cell proliferation and fate, Development, vol.138, pp.4131-4142, 2011.

T. Yokoe, T. Ohmachi, H. Inoue, K. Mimori, F. Tanaka et al.,

. Mori, Clinical significance of growth differentiation factor 11 in colorectal cancer, Int J Oncol, vol.31, pp.1097-1101, 2007.

C. Wallner, M. Drysch, M. Becerikli, H. Jaurich, J. M. Wagner et al.,

K. Nagler, M. Harati, S. Dadras, M. Philippou, B. Lehnhardt et al., Interaction with the GDF8/11 pathway reveals treatment options for adenocarcinoma of the breast, Breast, vol.37, pp.134-141, 2018.

S. S. Bajikar, C. C. Wang, M. A. Borten, E. J. Pereira, K. A. Atkins et al., Tumor-Suppressor Inactivation of GDF11 Occurs by Precursor Sequestration in Triple-Negative Breast Cancer, Dev Cell, pp.418-435, 2017.

G. Williams, M. P. Zentar, S. Gajendra, M. Sonego, P. Doherty et al., Transcriptional basis for the inhibition of neural stem cell proliferation and migration by the TGFbeta-family member GDF11, PLoS One, vol.8, p.78478, 2013.

C. Enriquez-cortina, M. Almonte-becerril, D. Clavijo-cornejo, M. Palestinodominguez, O. Bello-monroy et al.,

, Hepatocyte growth factor protects against isoniazid/rifampicin-induced oxidative liver damage, Toxicological sciences : an official journal of the Society of Toxicology, vol.135, pp.26-36, 2013.

J. U. Marquardt, D. Seo, L. E. Gomez-quiroz, K. Uchida, M. C. Gillen et al.,

P. Kitade, E. A. Kaposi-novak, V. M. Conner, S. S. Factor, and . Thorgeirsson, Loss of c-Met accelerates development of liver fibrosis in response to CCl(4) exposure through deregulation of multiple molecular pathways, Biochim Biophys Acta, pp.942-951, 2012.

J. P. Quigley and P. B. Armstrong, Tumor cell intravasation alu-cidated: the chick embryo opens the window, Cell, vol.94, pp.281-284, 1998.

D. Ribatti, The chick embryo chorioallantoic membrane (CAM) assay, Reprod Toxicol, vol.70, pp.97-101, 2017.

R. Lazzarini-lechuga, O. Alcantar-ramirez, R. Jaime-cruz, and L. Gómez-quiroz, Efecto teratogénico de nanopartículas de oro de 20 nm durante la septación cardiaca, Munod Nano, vol.7, pp.69-77, 2014.

C. Czauderna, M. Palestino-dominguez, D. Castven, D. Becker, and L. Zanon,

J. Rodriguez, F. L. Hajduk, M. Mahn, D. Herr, S. Strand et al., Ginkgo biloba induces different gene expression signatures and oncogenic pathways in malignant and non-malignant cells of the liver, PLoS One, vol.13, p.209067, 2018.

A. Jemal, E. M. Ward, C. J. Johnson, K. A. Cronin, J. Ma et al.,

A. J. Mariotto, R. Lake, R. L. Wilson, R. N. Sherman, S. J. Anderson et al.,

L. Kohler, E. J. Penberthy, H. K. Feuer, and . Weir, Annual Report to the Nation on the Status of Cancer, Featuring Survival, p.109, 1975.

L. Ghisolfi, A. C. Keates, X. Hu, D. K. Lee, and C. J. Li, Ionizing radiation induces stemness in cancer cells, PLoS One, vol.7, p.43628, 2012.

H. Hirschfield, C. B. Bian, T. Higashi, S. Nakagawa, T. Z. Zeleke et al., In vitro modeling of hepatocellular carcinoma molecular subtypes for anti-cancer drug assessment, Exp Mol Med, vol.50, p.419, 2018.

H. S. Seol, S. E. Lee, J. S. Song, J. K. Rhee, S. R. Singh et al.,

. Jang, Complement proteins C7 and CFH control the stemness of liver cancer cells via LSF-1, Cancer Lett, vol.372, pp.24-35, 2016.

Y. Zhang, P. B. Alexander, and X. F. Wang, TGF-beta Family Signaling in the Control of Cell Proliferation and Survival, Cold Spring Harb Perspect Biol, p.9, 2017.

P. M. Siegel and J. Massague, Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer, Nat Rev Cancer, vol.3, pp.807-821, 2003.

J. Zhang, O. Yamada, S. Kida, Y. Matsushita, and T. Hattori, Down-regulation of osteopontin mediates a novel mechanism underlying the cytostatic activity of TGF-beta, Cell Oncol (Dordr), vol.39, pp.119-128, 2016.

C. W. Lee, S. E. Lin, H. I. Tsai, P. J. Su, C. H. Hsieh et al., Cadherin 17 is related to recurrence and poor prognosis of cytokeratin 19-positive hepatocellular carcinoma, Oncol Lett, vol.15, pp.559-567, 2018.

A. W. Chan, J. H. Tong, S. L. Chan, P. B. Lai, and K. F. To, Expression of stemness markers (CD133 and EpCAM) in prognostication of hepatocellular carcinoma, Histopathology, vol.64, pp.935-950, 2014.

Y. Li, R. Wang, S. Xiong, X. Wang, Z. Zhao et al.,

. Cheng, Cancer-associated fibroblasts promote the stemness of CD24(+) liver cells via paracrine signaling, J Mol Med (Berl), vol.97, pp.243-255, 2019.

W. Chengye, T. Yu, S. Ping, S. Deguang, W. Keyun et al.,

G. Rui, Y. Zhenming, W. Mingliang, and . Liming, Metformin reverses bFGF-induced epithelial-mesenchymal transition in HCC cells, Oncotarget, vol.8, pp.104247-104257, 2017.