DOI QR코드

DOI QR Code

Ahnak depletion accelerates liver regeneration by modulating the TGF-β/Smad signaling pathway

  • Yang, Insook (Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University) ;
  • Son, Yeri (Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University) ;
  • Shin, Jae Hoon (Department of Surgery, University of Michigan) ;
  • Kim, Il Yong (Korea Mouse Phenotyping Center, Seoul National University) ;
  • Seong, Je Kyung (Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University)
  • 투고 : 2022.04.20
  • 심사 : 2022.06.30
  • 발행 : 2022.08.31

초록

Ahnak, a large protein first identified as an inhibitor of TGF-β signaling in human neuroblastoma, was recently shown to promote TGF-β in some cancers. The TGF-β signaling pathway regulates cell growth, various biological functions, and cancer growth and metastasis. In this study, we used Ahnak knockout (KO) mice that underwent a 70% partial hepatectomy (PH) to investigate the function of Ahnak in TGF-β signaling during liver regeneration. At the indicated time points after PH, we analyzed the mRNA and protein expression of the TGF -β/Smad signaling pathway and cell cycle-related factors, evaluated the cell cycle through proliferating cell nuclear antigen (PCNA) immunostaining, analyzed the mitotic index by hematoxylin and eosin staining. We also measured the ratio of liver tissue weight to body weight. Activation of TGF-β signaling was confirmed by analyzing the levels of phospho-Smad 2 and 3 in the liver at the indicated time points after PH and was lower in Ahnak KO mice than in WT mice. The expression levels of cyclin B1, D1, and E1; proteins in the Rb/E2F transcriptional pathway, which regulates the cell cycle; and the numbers of PCNA-positive cells were increased in Ahnak KO mice and showed tendencies opposite that of TGF-β expression. During postoperative regeneration, the liver weight to body weight ratio tended to increase faster in Ahnak KO mice. However, 7 days after PH, both groups of mice showed similar rates of regeneration, following which their active regeneration stopped. Analysis of hepatocytes undergoing mitosis showed that there were more mitotic cells in Ahnak KO mice, consistent with the weight ratio. Our findings suggest that Ahnak enhances TGF-β signaling during postoperative liver regeneration, resulting in cell cycle disruption; this highlights a novel role of Ahnak in liver regeneration. These results provide new insight into liver regeneration and potential treatment targets for liver diseases that require surgical treatment.

키워드

과제정보

This research was supported by Korea Mouse Phenotyping Project (2013M3A9D5072550) of the National Research Foundation (NRF) funded by the Ministry of Science and ICT (2012M3A 9D1054622) and partially supported by the Brain Korea 21 Plus Program and the Research Institute for Veterinary Science of Seoul National University.

참고문헌

  1. Shtivelman E, Cohen FE and Bishop JM (1992) A human gene (AHNAK) encoding an unusually large protein with a 1.2-microns polyionic rod structure. Proc Natl Acad Sci U S A 89, 5472-5476 https://doi.org/10.1073/pnas.89.12.5472
  2. Han H and Kursula P (2014) Periaxin and AHNAK nucleoprotein 2 form intertwined homodimers through domain swapping. J Biol Chem 289, 14121-14131 https://doi.org/10.1074/jbc.M114.554816
  3. Dempsey BR, Rezvanpour A, Lee TW, Barber KR, Junop MS and Shaw GS (2012) Structure of an asymmetric ternary protein complex provides insight for membrane interaction. Structure 20, 1737-1745 https://doi.org/10.1016/j.str.2012.08.004
  4. Shankar J, Messenberg A, Chan J, Underhill TM, Foster LJ and Nabi IR (2010) Pseudopodial actin dynamics control epithelial-mesenchymal transition in metastatic cancer cells. Cancer Res 70, 3780-3790
  5. Park JW, Kim IY, Choi JW et al (2018) AHNAK loss in mice promotes type II pneumocyte hyperplasia and lung tumor development. Mol Cancer Res 16, 1287-1298 https://doi.org/10.1158/1541-7786.MCR-17-0726
  6. Peng R, Zhang PF, Yang X et al (2019) Overexpression of RNF38 facilitates TGF-beta signaling by Ubiquitinating and degrading AHNAK in hepatocellular carcinoma. J Exp Clin Cancer Res 38, 113
  7. Lee IH, Sohn M, Lim HJ et al (2014) Ahnak functions as a tumor suppressor via modulation of TGFbeta/Smad signaling pathway. Oncogene 33, 4675-4684 https://doi.org/10.1038/onc.2014.69
  8. Michalopoulos GK (2010) Liver regeneration after partial hepatectomy: critical analysis of mechanistic dilemmas. Am J Pathol 176, 2-13 https://doi.org/10.2353/ajpath.2010.090675
  9. Fausto N (2000) Liver regeneration. J Hepatol 32, 19-31 https://doi.org/10.1016/S0168-8278(00)80412-2
  10. Taub R (2004) Liver regeneration: from myth to mechanism. Nat Rev Mol Cell Biol 5, 836-847 https://doi.org/10.1038/nrm1489
  11. Bottinger EP, Factor VM, Tsang ML et al (1996) The recombinant proregion of transforming growth factor beta1 (latency-associated peptide) inhibits active transforming growth factor beta1 in transgenic mice. Proc Natl Acad Sci U S A 93, 5877-5882 https://doi.org/10.1073/pnas.93.12.5877
  12. Jakowlew SB, Mead JE, Danielpour D, Wu J, Roberts AB and Fausto N (1991) Transforming growth factor-beta (TGF-beta) isoforms in rat liver regeneration: messenger RNA expression and activation of latent TGF-beta. Cell Regul 2, 535-548 https://doi.org/10.1091/mbc.2.7.535
  13. Fausto N, Campbell JS and Riehle KJ (2006) Liver regeneration. Hepatology 43, S45-53 https://doi.org/10.1002/hep.20969
  14. Higgins GM (1931) Experimental pathology of the liver. I. Restoration of the liver of the white rat following partial surgical removal. Arch Pathol 12, 186-202
  15. Mitchell C and Willenbring H (2008) A reproducible and well-tolerated method for 2/3 partial hepatectomy in mice. Nat Protoc 3, 1167-1170 https://doi.org/10.1038/nprot.2008.80
  16. Michalopoulos GK (2017) Hepatostat: liver regeneration and normal liver tissue maintenance. Hepatology 65, 1384-1392 https://doi.org/10.1002/hep.28988
  17. Michalopoulos GK (2007) Liver regeneration. J Cell Physiol 213, 286-300 https://doi.org/10.1002/jcp.21172
  18. Kokura K, Kim H, Shinagawa T, Khan MM, Nomura T and Ishii S (2003) The Ski-binding protein C184M negatively regulates tumor growth factor-beta signaling by sequestering the Smad proteins in the cytoplasm. J Biol Chem 278, 20133-20139 https://doi.org/10.1074/jbc.M210855200
  19. Kitamura T, Watanabe S and Sato N (1998) Liver regeneration, liver cancers and cyclins. J Gastroenterol Hepatol 13, S96-S99 https://doi.org/10.1111/jgh.1998.13.s1.96
  20. LK M (2008) Distinct proliferative and transcriptional effects of the D-type cyclins in vivo. Cell Cycle 7, 2215-2224 https://doi.org/10.4161/cc.7.14.6274
  21. Liu HX, Fang Y, Hu Y, Gonzalez FJ, Fang J and Wan YJ (2013) PPARbeta regulates liver regeneration by modulating Akt and E2f signaling. PLoS One 8, e65644
  22. Assy N, Gong Y, Zhang M, Pettigrew NM, Pashniak D and Minuk GY (1998) Use of proliferating cell nuclear antigen as a marker of liver regeneration after partial hepatectomy in rats. J Lab Clin Med 131, 251-256 https://doi.org/10.1016/S0022-2143(98)90097-X
  23. Michalopoulos GK and Bhushan B (2021) Liver regeneration: biological and pathological mechanisms and implications. Nat Rev Gastroenterol Hepatol 18, 40-55 https://doi.org/10.1038/s41575-020-0342-4
  24. Webber EM, Bruix J, Pierce RH and Fausto N (1998) Tumor necrosis factor primes hepatocytes for DNA replication in the rat. Hepatology 28, 1226-1234 https://doi.org/10.1002/hep.510280509
  25. Albrecht JH, Poon RY, Ahonen CL, Rieland BM, Deng C and Crary GS (1998) Involvement of p21 and p27 in the regulation of CDK activity and cell cycle progression in the regenerating liver. Oncogene 16, 2141-2150 https://doi.org/10.1038/sj.onc.1201728
  26. Goggin MM, Nelsen CJ, Kimball SR, Jefferson LS, Morley SJ and Albrecht JH (2004) Rapamycin-sensitive induction of eukaryotic initiation factor 4F in regenerating mouse liver. Hepatology 40, 537-544 https://doi.org/10.1002/hep.20338
  27. Kogure K, Zhang YQ, Maeshima A, Suzuki K, Kuwano H and Kojima I (2000) The role of activin and transforming growth factor-beta in the regulation of organ mass in the rat liver. Hepatology 31, 916-921 https://doi.org/10.1053/he.2000.6100
  28. Wrighton KH, Lin X and Feng XH (2009) Phospho-control of TGF-beta superfamily signaling. Cell Res 19, 8-20 https://doi.org/10.1038/cr.2008.327
  29. Moriuchi A, Hirono S, Ido A et al (2001) Additive and inhibitory effects of simultaneous treatment with growth factors on DNA synthesis through MAPK pathway and G1 cyclins in rat hepatocytes. Biochem Biophys Res Commun 280, 368-373 https://doi.org/10.1006/bbrc.2000.4063
  30. Nevins JR (2001) The Rb/E2F pathway and cancer. Hum Mol Genet 10, 699-703 https://doi.org/10.1093/hmg/10.7.699
  31. Marongiu F, Marongiu M, Contini A et al (2017) Hyperplasia vs hypertrophy in tissue regeneration after extensive liver resection. World J Gastroenterol 23, 1764-1770 https://doi.org/10.3748/wjg.v23.i10.1764
  32. Oe S, Lemmer ER, Conner EA et al (2004) Intact signaling by transforming growth factor beta is not required for termination of liver regeneration in mice. Hepatology 40, 1098-1105 https://doi.org/10.1002/hep.20426
  33. Romero-Gallo J, Sozmen EG, Chytil A et al (2005) Inactivation of TGF-beta signaling in hepatocytes results in an increased proliferative response after partial hepatectomy. Oncogene 24, 3028-3041 https://doi.org/10.1038/sj.onc.1208475
  34. Lee IH, Lim HJ, Yoon S et al (2008) Ahnak protein activates protein kinase C (PKC) through dissociation of the PKC-protein phosphatase 2A complex. J Biol Chem 283, 6312-6320 https://doi.org/10.1074/jbc.M706878200
  35. Baak JP (1990) Mitosis counting in tumors. Hum Pathol 21, 683-685 https://doi.org/10.1016/0046-8177(90)90026-2