#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Loss of hepatocyte cell division leads to liver inflammation and fibrosis


Autoři: Matthew R. Dewhurst aff001;  Jin Rong Ow aff001;  Gözde Zafer aff001;  Noémi K. M. van Hul aff001;  Heike Wollmann aff001;  Xavier Bisteau aff001;  David Brough aff002;  Hyungwon Choi aff001;  Philipp Kaldis aff001
Působiště autorů: Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore aff001;  Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M aff002;  Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M aff002;  Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M aff002;  Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester... aff002;  Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M aff002;  Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M aff002;  Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M aff002;  Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M aff002;  Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M aff002;  Department of Biochemistry, National University of Singapore (NUS), Singapore aff003;  Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore aff004;  Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Sweden aff005
Vyšlo v časopise: Loss of hepatocyte cell division leads to liver inflammation and fibrosis. PLoS Genet 16(11): e1009084. doi:10.1371/journal.pgen.1009084
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pgen.1009084

Souhrn

The liver possesses a remarkable regenerative capacity based partly on the ability of hepatocytes to re-enter the cell cycle and divide to replace damaged cells. This capability is substantially reduced upon chronic damage, but it is not clear if this is a cause or consequence of liver disease. Here, we investigate whether blocking hepatocyte division using two different mouse models affects physiology as well as clinical liver manifestations like fibrosis and inflammation. We find that in P14 Cdk1Liv-/- mice, where the division of hepatocytes is abolished, polyploidy, DNA damage, and increased p53 signaling are prevalent. Cdk1Liv-/- mice display classical markers of liver damage two weeks after birth, including elevated ALT, ALP, and bilirubin levels, despite the lack of exogenous liver injury. Inflammation was further studied using cytokine arrays, unveiling elevated levels of CCL2, TIMP1, CXCL10, and IL1-Rn in Cdk1Liv-/- liver, which resulted in increased numbers of monocytes. Ablation of CDK2-dependent DNA re-replication and polyploidy in Cdk1Liv-/- mice reversed most of these phenotypes. Overall, our data indicate that blocking hepatocyte division induces biological processes driving the onset of the disease phenotype. It suggests that the decrease in hepatocyte division observed in liver disease may not only be a consequence of fibrosis and inflammation, but also a pathological cue.

Klíčová slova:

Cell cycle and cell division – DNA damage – Fatty liver – Fibrosis – Hepatocytes – Liver fibrosis – Mouse models – Polyploidy


Zdroje

1. Miyaoka Y, Miyajima A. To divide or not to divide: revisiting liver regeneration. Cell Div. 2013;8(1):8. doi: 10.1186/1747-1028-8-8 23786799

2. Macdonald RA. "Lifespan" of liver cells. Autoradio-graphic study using tritiated thymidine in normal, cirrhotic, and partially hepatectomized rats. Arch Intern Med. 1961;107:335–43. doi: 10.1001/archinte.1961.03620030023003 13764742

3. Yang SQ, Lin HZ, Mandal AK, Huang J, Diehl AM. Disrupted signaling and inhibited regeneration in obese mice with fatty livers: implications for nonalcoholic fatty liver disease pathophysiology. Hepatology. 2001;34(4 Pt 1):694–706. doi: 10.1053/jhep.2001.28054 11584365

4. Zhao G, Nakano K, Chijiiwa K, Ueda J, Tanaka M. Inhibited activities in CCAAT/enhancer-binding protein, activating protein-1 and cyclins after hepatectomy in rats with thioacetamide-induced liver cirrhosis. Biochem Biophys Res Commun. 2002;292(2):474–81. doi: 10.1006/bbrc.2002.6630 11906187

5. Vetelainen R, van Vliet AK, van Gulik TM. Severe steatosis increases hepatocellular injury and impairs liver regeneration in a rat model of partial hepatectomy. Ann Surg. 2007;245(1):44–50. doi: 10.1097/01.sla.0000225253.84501.0e 17197964

6. Han MS, Park SY, Shinzawa K, Kim S, Chung KW, Lee JH, et al. Lysophosphatidylcholine as a death effector in the lipoapoptosis of hepatocytes. J Lipid Res. 2008;49(1):84–97. doi: 10.1194/jlr.M700184-JLR200 17951222

7. Karidis NP, Delladetsima I, Theocharis S. Hepatocyte Turnover in Chronic HCV-Induced Liver Injury and Cirrhosis. Gastroenterol Res Pract. 2015;2015:654105. doi: 10.1155/2015/654105 25892989

8. Oh IS, Park SH. Immune-mediated Liver Injury in Hepatitis B Virus Infection. Immune Netw. 2015;15(4):191–8. doi: 10.4110/in.2015.15.4.191 26330805

9. Kitada T, Seki S, Kawakita N, Kuroki T, Monna T. Telomere shortening in chronic liver diseases. Biochem Biophys Res Commun. 1995;211(1):33–9. doi: 10.1006/bbrc.1995.1774 7779103

10. Wiemann SU, Satyanarayana A, Tsahuridu M, Tillmann HL, Zender L, Klempnauer J, et al. Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis. FASEB J. 2002;16(9):935–42. doi: 10.1096/fj.01-0977com 12087054

11. Aravinthan A, Scarpini C, Tachtatzis P, Verma S, Penrhyn-Lowe S, Harvey R, et al. Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease. J Hepatol. 2013;58(3):549–56. doi: 10.1016/j.jhep.2012.10.031 23142622

12. Gentric G, Maillet V, Paradis V, Couton D, L'Hermitte A, Panasyuk G, et al. Oxidative stress promotes pathologic polyploidization in nonalcoholic fatty liver disease. J Clin Invest. 2015;125(3):981–92. doi: 10.1172/JCI73957 25621497

13. Morgan D. The Cell Cycle: Principles of Control. London: New Science Press; 2007. 297 p.

14. Lohka MJ, Hayes MK, Maller JL. Purification of maturation-promoting factor, an intracellular regulator of early mitotic events. Proc Natl Acad Sci U S A. 1988;85(9):3009–13. doi: 10.1073/pnas.85.9.3009 3283736

15. Diril MK, Ratnacaram CK, Padmakumar VC, Du T, Wasser M, Coppola V, et al. Cyclin-dependent kinase 1 (Cdk1) is essential for cell division and suppression of DNA re-replication but not for liver regeneration. Proc Natl Acad Sci U S A. 2012;109(10):3826–31. doi: 10.1073/pnas.1115201109 22355113

16. Itzhaki JE, Gilbert CS, Porter AC. Construction by gene targeting in human cells of a "conditional' CDC2 mutant that rereplicates its DNA. Nat Genet. 1997;15(3):258–65. doi: 10.1038/ng0397-258 9054937

17. Santamaria D, Barriere C, Cerqueira A, Hunt S, Tardy C, Newton K, et al. Cdk1 is sufficient to drive the mammalian cell cycle. Nature. 2007;448(7155):811–5. doi: 10.1038/nature06046 17700700

18. Krude T, Jackman M, Pines J, Laskey RA. Cyclin/Cdk-dependent initiation of DNA replication in a human cell-free system. Cell. 1997;88(1):109–19. doi: 10.1016/s0092-8674(00)81863-2 9019396

19. Ortega S, Prieto I, Odajima J, Martin A, Dubus P, Sotillo R, et al. Cyclin-dependent kinase 2 is essential for meiosis but not for mitotic cell division in mice. Nat Genet. 2003;35(1):25–31. doi: 10.1038/ng1232 12923533

20. Berthet C, Aleem E, Coppola V, Tessarollo L, Kaldis P. Cdk2 knockout mice are viable. Curr Biol. 2003;13(20):1775–85. doi: 10.1016/j.cub.2003.09.024 14561402

21. Margall-Ducos G, Celton-Morizur S, Couton D, Bregerie O, Desdouets C. Liver tetraploidization is controlled by a new process of incomplete cytokinesis. J Cell Sci. 2007;120(Pt 20):3633–9. doi: 10.1242/jcs.016907 17895361

22. Celton-Morizur S, Merlen G, Couton D, Margall-Ducos G, Desdouets C. The insulin/Akt pathway controls a specific cell division program that leads to generation of binucleated tetraploid liver cells in rodents. J Clin Invest. 2009;119(7):1880–7. doi: 10.1172/jci38677 19603546

23. Zhang S, Zhou K, Luo X, Li L, Tu HC, Sehgal A, et al. The Polyploid State Plays a Tumor-Suppressive Role in the Liver. Dev Cell. 2018;44(4):447–59 e5. doi: 10.1016/j.devcel.2018.01.010 29429824

24. Kumar V, Agrawal R, Pandey A, Kopf S, Hoeffgen M, Kaymak S, et al. Compromised DNA repair is responsible for diabetes-associated fibrosis. EMBO J. 2020;39(11):e103477. doi: 10.15252/embj.2019103477 32338774

25. Toyoda H, Bregerie O, Vallet A, Nalpas B, Pivert G, Brechot C, et al. Changes to hepatocyte ploidy and binuclearity profiles during human chronic viral hepatitis. Gut. 2005;54(2):297–302. doi: 10.1136/gut.2004.043893 15647198

26. Nevzorova YA, Tschaharganeh D, Gassler N, Geng Y, Weiskirchen R, Sicinski P, et al. Aberrant cell cycle progression and endoreplication in regenerating livers of mice that lack a single E-type cyclin. Gastroenterology. 2009;137(2):691–703, e1-6. doi: 10.1053/j.gastro.2009.05.003 19445941

27. Chen HZ, Ouseph MM, Li J, Pecot T, Chokshi V, Kent L, et al. Canonical and atypical E2Fs regulate the mammalian endocycle. Nat Cell Biol. 2012;14(11):1192–202. doi: 10.1038/ncb2595 23064266

28. Li D, Cen J, Chen X, Conway EM, Ji Y, Hui L. Hepatic loss of survivin impairs postnatal liver development and promotes expansion of hepatic progenitor cells in mice. Hepatology. 2013;58(6):2109–21. doi: 10.1002/hep.26601 23813590

29. Miettinen TP, Caldez MJ, Kaldis P, Bjorklund M. Cell size control—a mechanism for maintaining fitness and function. Bioessays. 2017;39(9). doi: 10.1002/bies.201700058 28752618

30. Berasain C, Avila MA. Regulation of hepatocyte identity and quiescence. Cell Mol Life Sci. 2015;72(20):3831–51. doi: 10.1007/s00018-015-1970-7 26089250

31. Dai J, Sultan S, Taylor SS, Higgins JM. The kinase haspin is required for mitotic histone H3 Thr3 phosphorylation and normal metaphase chromosome alignment. Genes Dev. 2005;19(4):472–88. doi: 10.1101/gad.1267105 15681610

32. Caldez MJ, Van Hul N, Koh HWL, Teo XQ, Fan JJ, Tan PY, et al. Metabolic Remodeling during Liver Regeneration. Dev Cell. 2018;47(4):425–38 e5. doi: 10.1016/j.devcel.2018.09.020 30344111

33. Moldovan GL, Pfander B, Jentsch S. PCNA, the maestro of the replication fork. Cell. 2007;129(4):665–79. doi: 10.1016/j.cell.2007.05.003 17512402

34. Miettinen TP, Pessa HK, Caldez MJ, Fuhrer T, Diril MK, Sauer U, et al. Identification of transcriptional and metabolic programs related to mammalian cell size. Curr Biol. 2014;24(6):598–608. doi: 10.1016/j.cub.2014.01.071 24613310

35. Di Micco R, Fumagalli M, Cicalese A, Piccinin S, Gasparini P, Luise C, et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature. 2006;444(7119):638–42. doi: 10.1038/nature05327 17136094

36. Zhao H, Piwnica-Worms H. ATR-mediated checkpoint pathways regulate phosphorylation and activation of human Chk1. Mol Cell Biol. 2001;21(13):4129–39. doi: 10.1128/MCB.21.13.4129-4139.2001 11390642

37. Shieh SY, Ikeda M, Taya Y, Prives C. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell. 1997;91(3):325–34. doi: 10.1016/s0092-8674(00)80416-x 9363941

38. Aubrey BJ, Kelly GL, Janic A, Herold MJ, Strasser A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018;25(1):104–13. doi: 10.1038/cdd.2017.169 29149101

39. Yu J, Zhang L, Hwang PM, Kinzler KW, Vogelstein B. PUMA induces the rapid apoptosis of colorectal cancer cells. Mol Cell. 2001;7(3):673–82. doi: 10.1016/s1097-2765(01)00213-1 11463391

40. Nakano K, Vousden KH. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell. 2001;7(3):683–94. doi: 10.1016/s1097-2765(01)00214-3 11463392

41. Li M, He Y, Dubois W, Wu X, Shi J, Huang J. Distinct regulatory mechanisms and functions for p53-activated and p53-repressed DNA damage response genes in embryonic stem cells. Mol Cell. 2012;46(1):30–42. doi: 10.1016/j.molcel.2012.01.020 22387025

42. Schuler M, Bossy-Wetzel E, Goldstein JC, Fitzgerald P, Green DR. p53 induces apoptosis by caspase activation through mitochondrial cytochrome c release. J Biol Chem. 2000;275(10):7337–42. doi: 10.1074/jbc.275.10.7337 10702305

43. Si-Tayeb K, Lemaigre FP, Duncan SA. Organogenesis and development of the liver. Dev Cell. 2010;18(2):175–89. doi: 10.1016/j.devcel.2010.01.011 20159590

44. Ju C, Tacke F. Hepatic macrophages in homeostasis and liver diseases: from pathogenesis to novel therapeutic strategies. Cell Mol Immunol. 2016;13(3):316–27. doi: 10.1038/cmi.2015.104 26908374

45. Austyn JM, Gordon S. F4/80, a monoclonal antibody directed specifically against the mouse macrophage. Eur J Immunol. 1981;11(10):805–15. doi: 10.1002/eji.1830111013 7308288

46. Scott CL, Zheng F, De Baetselier P, Martens L, Saeys Y, De Prijck S, et al. Bone marrow-derived monocytes give rise to self-renewing and fully differentiated Kupffer cells. Nat Commun. 2016;7:10321. doi: 10.1038/ncomms10321 26813785

47. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115(2):209–18. doi: 10.1172/JCI24282 15690074

48. Zhan Z, Chen Y, Duan Y, Li L, Mew K, Hu P, et al. Identification of key genes, pathways and potential therapeutic agents for liver fibrosis using an integrated bioinformatics analysis. PeerJ. 2019;7:e6645. doi: 10.7717/peerj.6645 30923657

49. Rodier F, Coppe JP, Patil CK, Hoeijmakers WA, Munoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009;11(8):973–9. doi: 10.1038/ncb1909 19597488

50. Serbina NV, Pamer EG. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat Immunol. 2006;7(3):311–7. doi: 10.1038/ni1309 16462739

51. Ajuebor MN, Flower RJ, Hannon R, Christie M, Bowers K, Verity A, et al. Endogenous monocyte chemoattractant protein-1 recruits monocytes in the zymosan peritonitis model. J Leukoc Biol. 1998;63(1):108–16. doi: 10.1002/jlb.63.1.108 9469480

52. Ito A, Hong C, Rong X, Zhu X, Tarling EJ, Hedde PN, et al. LXRs link metabolism to inflammation through Abca1-dependent regulation of membrane composition and TLR signaling. Elife. 2015;4:e08009. doi: 10.7554/eLife.08009 26173179

53. Bohuslav J, Chen LF, Kwon H, Mu Y, Greene WC. p53 induces NF-kappaB activation by an IkappaB kinase-independent mechanism involving phosphorylation of p65 by ribosomal S6 kinase 1. J Biol Chem. 2004;279(25):26115–25. doi: 10.1074/jbc.M313509200 15073170

54. Gopinathan L, Tan SL, Padmakumar VC, Coppola V, Tessarollo L, Kaldis P. Loss of Cdk2 and cyclin A2 impairs cell proliferation and tumorigenesis. Cancer Res. 2014;74(14):3870–9. doi: 10.1158/0008-5472.CAN-13-3440 24802190

55. Kanakkanthara A, Jeganathan KB, Limzerwala JF, Baker DJ, Hamada M, Nam HJ, et al. Cyclin A2 is an RNA binding protein that controls Mre11 mRNA translation. Science. 2016;353(6307):1549–52. doi: 10.1126/science.aaf7463 27708105

56. Bou-Nader M, Caruso S, Donne R, Celton-Morizur S, Calderaro J, Gentric G, et al. Polyploidy spectrum: a new marker in HCC classification. Gut. 2020;69(2):355–64. doi: 10.1136/gutjnl-2018-318021 30979717

57. Zheng L, Dai H, Zhou M, Li X, Liu C, Guo Z, et al. Polyploid cells rewire DNA damage response networks to overcome replication stress-induced barriers for tumour progression. Nat Commun. 2012;3:815. doi: 10.1038/ncomms1825 22569363

58. Pandit SK, Westendorp B, de Bruin A. Physiological significance of polyploidization in mammalian cells. Trends Cell Biol. 2013;23(11):556–66. doi: 10.1016/j.tcb.2013.06.002 23849927

59. Vassilev LT, Tovar C, Chen S, Knezevic D, Zhao X, Sun H, et al. Selective small-molecule inhibitor reveals critical mitotic functions of human CDK1. Proc Natl Acad Sci U S A. 2006;103(28):10660–5. doi: 10.1073/pnas.0600447103 16818887

60. Casper AM, Nghiem P, Arlt MF, Glover TW. ATR regulates fragile site stability. Cell. 2002;111(6):779–89. doi: 10.1016/s0092-8674(02)01113-3 12526805

61. Asrih M, Jornayvaz FR. Inflammation as a potential link between nonalcoholic fatty liver disease and insulin resistance. J Endocrinol. 2013;218(3):R25–36. doi: 10.1530/JOE-13-0201 23833274

62. Christian F, Smith EL, Carmody RJ. The Regulation of NF-kappaB Subunits by Phosphorylation. Cells. 2016;5(1):12.

63. Haukeland JW, Damas JK, Konopski Z, Loberg EM, Haaland T, Goverud I, et al. Systemic inflammation in nonalcoholic fatty liver disease is characterized by elevated levels of CCL2. J Hepatol. 2006;44(6):1167–74. doi: 10.1016/j.jhep.2006.02.011 16618517

64. Neuman MG, Benhamou JP, Marcellin P, Valla D, Malkiewicz IM, Katz GG, et al. Cytokine—chemokine and apoptotic signatures in patients with hepatitis C. Transl Res. 2007;149(3):126–36. doi: 10.1016/j.trsl.2006.11.002 17320798

65. Baeck C, Wei X, Bartneck M, Fech V, Heymann F, Gassler N, et al. Pharmacological inhibition of the chemokine C-C motif chemokine ligand 2 (monocyte chemoattractant protein 1) accelerates liver fibrosis regression by suppressing Ly-6C(+) macrophage infiltration in mice. Hepatology. 2014;59(3):1060–72. doi: 10.1002/hep.26783 24481979

66. Anstee QM, Neuschwander-Tetri BA, Wong VW, Abdelmalek MF, Younossi ZM, Yuan J, et al. Cenicriviroc for the treatment of liver fibrosis in adults with nonalcoholic steatohepatitis: AURORA Phase 3 study design. Contemp Clin Trials. 2019;89:105922. doi: 10.1016/j.cct.2019.105922 31881392

67. Yoshiji H, Kuriyama S, Miyamoto Y, Thorgeirsson UP, Gomez DE, Kawata M, et al. Tissue inhibitor of metalloproteinases-1 promotes liver fibrosis development in a transgenic mouse model. Hepatology. 2000;32(6):1248–54. doi: 10.1053/jhep.2000.20521 11093731

68. Ghosh AK, Vaughan DE. PAI-1 in tissue fibrosis. J Cell Physiol. 2012;227(2):493–507. doi: 10.1002/jcp.22783 21465481

69. Yeruva S, Ramadori G, Raddatz D. NF-kappaB-dependent synergistic regulation of CXCL10 gene expression by IL-1beta and IFN-gamma in human intestinal epithelial cell lines. Int J Colorectal Dis. 2008;23(3):305–17. doi: 10.1007/s00384-007-0396-6 18046562

70. Wilczynska KM, Gopalan SM, Bugno M, Kasza A, Konik BS, Bryan L, et al. A novel mechanism of tissue inhibitor of metalloproteinases-1 activation by interleukin-1 in primary human astrocytes. J Biol Chem. 2006;281(46):34955–64. doi: 10.1074/jbc.M604616200 17012236

71. Raskatov JA, Meier JL, Puckett JW, Yang F, Ramakrishnan P, Dervan PB. Modulation of NF-kappaB-dependent gene transcription using programmable DNA minor groove binders. Proc Natl Acad Sci U S A. 2012;109(4):1023–8. doi: 10.1073/pnas.1118506109 22203967

72. Ortica F, Moustrou C, Berthet J, Favaro G, Samat A, Guglielmetti R, et al. Comprehensive photokinetic and NMR study of a biphotochromic supermolecule involving two naphthopyrans linked to a central thiophene unit through acetylenic bonds. Photochem Photobiol. 2003;78(6):558–66. doi: 10.1562/0031-8655(2003)078<0558:cpanso>2.0.co;2 14743863

73. Shang Y, Myers M, Brown M. Formation of the androgen receptor transcription complex. Mol Cell. 2002;9(3):601–10. doi: 10.1016/s1097-2765(02)00471-9 11931767

74. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8. doi: 10.1006/meth.2001.1262 11846609

75. Ji X, Dadon DB, Abraham BJ, Lee TI, Jaenisch R, Bradner JE, et al. Chromatin proteomic profiling reveals novel proteins associated with histone-marked genomic regions. Proc Natl Acad Sci U S A. 2015;112(12):3841–6. doi: 10.1073/pnas.1502971112 25755260

76. Fu Y, Hou B, Weng C, Liu W, Dai J, Zhao C, et al. Functional ectopic neuritogenesis by retinal rod bipolar cells is regulated by miR-125b-5p during retinal remodeling in RCS rats. Sci Rep. 2017;7(1):1011. doi: 10.1038/s41598-017-01261-x 28432360

77. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9(7):671–5. doi: 10.1038/nmeth.2089 22930834

78. Guguen-Guillouzo C. Isolation and culture of animal and human hepatocytes. In: Freshney RI, Freshney MG, editors. Culture of Epithelial Cells. Second Edition ed: Wiley-Liss, Inc.; 2002.

79. Frezza C, Cipolat S, Scorrano L. Organelle isolation: functional mitochondria from mouse liver, muscle and cultured fibroblasts. Nat Protoc. 2007;2(2):287–95. doi: 10.1038/nprot.2006.478 17406588

80. Satyanarayana A, Berthet C, Lopez-Molina J, Coppola V, Tessarollo L, Kaldis P. Genetic substitution of Cdk1 by Cdk2 leads to embryonic lethality and loss of meiotic function of Cdk2. Development. 2008;135(20):3389–400. doi: 10.1242/dev.024919 18787066

81. Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9(7):676–82. doi: 10.1038/nmeth.2019 22743772

82. Palmer N, Talib SZA, Ratnacaram CK, Low D, Bisteau X, Lee JHS, et al. CDK2 regulates the NRF1/Ehmt1 axis during meiotic prophase I. J Cell Biol. 2019. doi: 10.1083/jcb.201903125 31350280

83. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21. doi: 10.1093/bioinformatics/bts635 23104886

84. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323. doi: 10.1186/1471-2105-12-323 21816040

85. Storey JD. A direct approach to false discovery rates. J R Statist Soc B. 2002;64(3):479–98.


Článek vyšel v časopise

PLOS Genetics


2020 Číslo 11
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#