1932

Abstract

Understanding the physiopathology of disease remains an essential step in developing novel therapeutics. Although animal models have certainly contributed to advancing this enterprise, their limitation in modeling all the aspects of complex human disorders is one of the major challenges faced by the biomedical research field. Human induced pluripotent stem cells (hiPSCs) derived from patients represent a great opportunity to overcome this deficiency because these cells cover the genetic diversity needed to fully model human diseases. Here, we provide an overview of the history of hiPSC technology and discuss common challenges and approaches that we and others have faced when using hiPSCs to model disease. Our emphasis is on liver disease, and consequently, we review the progress made using this technology to produce functional liver cells in vitro and how these systems are being used to recapitulate a diversity of developmental, metabolic, genetic, and infectious liver disorders.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pathol-020117-043634
2019-01-24
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/pathmechdis/14/1/annurev-pathol-020117-043634.html?itemId=/content/journals/10.1146/annurev-pathol-020117-043634&mimeType=html&fmt=ahah

Literature Cited

  1. 1. WHO (World Health Organ.). 2018. Genes and human disease. WHO http://www.who.int/genomics/public/geneticdiseases/en/index2.html
  2. 2.  Bomba L, Walter K, Soranzo N 2017. The impact of rare and low-frequency genetic variants in common disease. Genome Biol 18:77
    [Google Scholar]
  3. 3.  Durand C, Rappold GA 2013. Height matters—from monogenic disorders to normal variation. Nat. Rev. Endocrinol. 9:171–77
    [Google Scholar]
  4. 4.  Eilbeck K, Quinlan A, Yandell M 2017. Settling the score: variant prioritization and Mendelian disease. Nat. Rev. Genet. 18:599–612
    [Google Scholar]
  5. 5.  Merkle FT, Eggan K 2013. Modeling human disease with pluripotent stem cells: from genome association to function. Cell Stem Cell 12:656–68
    [Google Scholar]
  6. 6.  Chin MH, Mason MJ, Xie W, Volinia S, Singer M et al. 2009. Induced pluripotent stem cells and embryonic stem cells are distinguished by gene expression signatures. Cell Stem Cell 5:111–23
    [Google Scholar]
  7. 7.  DeBoever C, Li H, Jakubosky D, Benaglio P, Reyna J et al. 2017. Large-scale profiling reveals the influence of genetic variation on gene expression in human induced pluripotent stem cells. Cell Stem Cell 20:533–46.e7
    [Google Scholar]
  8. 8.  Pashos EE, Park Y, Wang X, Raghavan A, Yang W et al. 2017. Large, diverse population cohorts of hiPSCs and derived hepatocyte-like cells reveal functional genetic variation at blood lipid-associated loci. Cell Stem Cell 20:558–70.e10
    [Google Scholar]
  9. 9.  Warren CR, O'Sullivan JF, Friesen M, Becker CE, Zhang X et al. 2017. Induced pluripotent stem cell differentiation enables functional validation of GWAS variants in metabolic disease. Cell Stem Cell 20:547–57.e7
    [Google Scholar]
  10. 10.  Pournasr B, Duncan SA 2017. Modeling inborn errors of hepatic metabolism using induced pluripotent stem cells. Arterioscler. Thromb. Vasc. Biol. 37:1994–99
    [Google Scholar]
  11. 11.  Cayo MA, Cai J, DeLaForest A, Noto FK, Nagaoka M et al. 2012. JD induced pluripotent stem cell–derived hepatocytes faithfully recapitulate the pathophysiology of familial hypercholesterolemia. Hepatology 56:2163–71
    [Google Scholar]
  12. 12.  Guan Y, Xu D, Garfin PM, Ehmer U, Hurwitz M et al. 2017. Human hepatic organoids for the analysis of human genetic diseases. JCI Insight 2:e94954
    [Google Scholar]
  13. 13.  Tafaleng EN, Chakraborty S, Han B, Hale P, Wu W et al. 2015. Induced pluripotent stem cells model personalized variations in liver disease resulting from α1-antitrypsin deficiency. Hepatology 62:147–57
    [Google Scholar]
  14. 14.  Brigida AL, Siniscalco D 2016. Induced pluripotent stem cells as a cellular model for studying Down Syndrome. J. Stem Cells Regen. Med. 12:54–60
    [Google Scholar]
  15. 15.  Doyle MJ, Lohr JL, Chapman CS, Koyano-Nakagawa N, Garry MG, Garry DJ 2015. Human induced pluripotent stem cell–derived cardiomyocytes as a model for heart development and congenital heart disease. Stem Cell Rev 11:710–27
    [Google Scholar]
  16. 16.  Wu AR, Wang J, Streets AM, Huang Y 2017. Single-cell transcriptional analysis. Annu. Rev. Anal. Chem. 10:439–62
    [Google Scholar]
  17. 17.  Goodwin S, McPherson JD, McCombie WR 2016. Coming of age: ten years of next-generation sequencing technologies. Nat. Rev. Genet. 17:333–51
    [Google Scholar]
  18. 18.  Liu Z, Lavis LD, Betzig E 2015. Imaging live-cell dynamics and structure at the single-molecule level. Mol. Cell 58:644–59
    [Google Scholar]
  19. 19.  Specht EA, Braselmann E, Palmer AE 2017. A critical and comparative review of fluorescent tools for live-cell imaging. Annu. Rev. Physiol. 79:93–117
    [Google Scholar]
  20. 20.  Hendriks WT, Warren CR, Cowan CA 2016. Genome editing in human pluripotent stem cells: approaches, pitfalls, and solutions. Cell Stem Cell 18:53–65
    [Google Scholar]
  21. 21.  Hockemeyer D, Jaenisch R 2016. Induced pluripotent stem cells meet genome editing. Cell Stem Cell 18:573–86
    [Google Scholar]
  22. 22.  Kilpinen H, Goncalves A, Leha A, Afzal V, Alasoo K et al. 2017. Common genetic variation drives molecular heterogeneity in human iPSCs. Nature 546:370–75
    [Google Scholar]
  23. 23.  Kyttala A, Moraghebi R, Valensisi C, Kettunen J, Andrus C et al. 2016. Genetic variability overrides the impact of parental cell type and determines iPSC differentiation potential. Stem Cell Rep 6:200–12
    [Google Scholar]
  24. 24.  Engle SJ, Vincent F 2014. Small molecule screening in human induced pluripotent stem cell–derived terminal cell types. J. Biol. Chem. 289:4562–70
    [Google Scholar]
  25. 25.  Iwata Y, Klaren WD, Lebakken CS, Grimm FA, Rusyn I 2017. High-content assay multiplexing for vascular toxicity screening in induced pluripotent stem cell–derived endothelial cells and human umbilical vein endothelial cells. Assay Drug Dev. Technol. 15:267–79
    [Google Scholar]
  26. 26.  Sherman SP, Bang AG 2018. High-throughput screen for compounds that modulate neurite growth of human induced pluripotent stem cell-derived neurons. Dis. Model. Mech. 11:dmm031906
    [Google Scholar]
  27. 27.  Zhou T, Tan L, Cederquist GY, Fan Y, Hartley BJ et al. 2017. High-content screening in hPSC-neural progenitors identifies drug candidates that inhibit Zika virus infection in fetal-like organoids and adult brain. Cell Stem Cell 21:274–83.e5
    [Google Scholar]
  28. 28.  Gurdon JB 1962. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J. Embryol. Exp. Morphol. 10:622–40
    [Google Scholar]
  29. 29.  Gurdon JB 1962. Adult frogs derived from the nuclei of single somatic cells. Dev. Biol. 4:256–73
    [Google Scholar]
  30. 30.  Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ et al. 1998. Embryonic stem cell lines derived from human blastocysts. Science 282:1145–47
    [Google Scholar]
  31. 31.  de Miguel-Beriain I 2015. The ethics of stem cells revisited. Adv. Drug Deliv. Rev. 82–83:176–80
    [Google Scholar]
  32. 32.  Kimbrel EA, Lanza R 2015. Current status of pluripotent stem cells: moving the first therapies to the clinic. Nat. Rev. Drug Discov. 14:681–92
    [Google Scholar]
  33. 33.  Lo B, Parham L 2009. Ethical issues in stem cell research. Endocr. Rev. 30:204–13
    [Google Scholar]
  34. 34.  Tada M, Takahama Y, Abe K, Nakatsuji N, Tada T 2001. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Curr. Biol. 11:1553–58
    [Google Scholar]
  35. 35.  Cowan CA, Atienza J, Melton DA, Eggan K 2005. Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells. Science 309:1369–73
    [Google Scholar]
  36. 36.  Takahashi K, Yamanaka S 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663–76
    [Google Scholar]
  37. 37.  Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS et al. 2005. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 122:947–56
    [Google Scholar]
  38. 38.  Sato N, Sanjuan IM, Heke M, Uchida M, Naef F, Brivanlou AH 2003. Molecular signature of human embryonic stem cells and its comparison with the mouse. Dev. Biol. 260:404–13
    [Google Scholar]
  39. 39.  Niwa H, Ogawa K, Shimosato D, Adachi K 2009. A parallel circuit of LIF signalling pathways maintains pluripotency of mouse ES cells. Nature 460:118–22
    [Google Scholar]
  40. 40.  Adhikary S, Eilers M 2005. Transcriptional regulation and transformation by Myc proteins. Nat. Rev. Mol. Cell Biol. 6:635–45
    [Google Scholar]
  41. 41.  Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T et al. 2007. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131:861–72
    [Google Scholar]
  42. 42.  Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL et al. 2007. Induced pluripotent stem cell lines derived from human somatic cells. Science 318:1917–20
    [Google Scholar]
  43. 43.  Okita K, Ichisaka T, Yamanaka S 2007. Generation of germline-competent induced pluripotent stem cells. Nature 448:313–17
    [Google Scholar]
  44. 44.  Takahashi K, Yamanaka S 2016. A decade of transcription factor–mediated reprogramming to pluripotency. Nat. Rev. Mol. Cell Biol. 17:183–93
    [Google Scholar]
  45. 45.  Fusaki N, Ban H, Nishiyama A, Saeki K, Hasegawa M 2009. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 85:348–62
    [Google Scholar]
  46. 46.  Okita K, Matsumura Y, Sato Y, Okada A, Morizane A et al. 2011. A more efficient method to generate integration-free human iPS cells. Nat. Methods 8:409–12
    [Google Scholar]
  47. 47.  Warren L, Manos PD, Ahfeldt T, Loh YH, Li H et al. 2010. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7:618–30
    [Google Scholar]
  48. 48.  Choi J, Lee S, Mallard W, Clement K, Tagliazucchi GM et al. 2015. A comparison of genetically matched cell lines reveals the equivalence of human iPSCs and ESCs. Nat. Biotechnol. 33:1173–81
    [Google Scholar]
  49. 49.  Nakamura T, Okamoto I, Sasaki K, Yabuta Y, Iwatani C et al. 2016. A developmental coordinate of pluripotency among mice, monkeys and humans. Nature 537:57–62
    [Google Scholar]
  50. 50.  Gafni O, Weinberger L, Mansour AA, Manor YS, Chomsky E et al. 2013. Derivation of novel human ground state naive pluripotent stem cells. Nature 504:282–86
    [Google Scholar]
  51. 51.  Vallier L, Touboul T, Chng Z, Brimpari M, Hannan N et al. 2009. Early cell fate decisions of human embryonic stem cells and mouse epiblast stem cells are controlled by the same signalling pathways. PLOS ONE 4:e6082
    [Google Scholar]
  52. 52.  Passier R, Orlova V, Mummery C 2016. Complex tissue and disease modeling using hiPSCs. Cell Stem Cell 18:309–21
    [Google Scholar]
  53. 53.  Takahashi K, Yamanaka S 2015. A developmental framework for induced pluripotency. Development 142:3274–85
    [Google Scholar]
  54. 54.  Tchieu J, Zimmer B, Fattahi F, Amin S, Zeltner N et al. 2017. A modular platform for differentiation of human PSCs into all major ectodermal lineages. Cell Stem Cell 21:399–410.e7
    [Google Scholar]
  55. 55.  Yoshihara M, Hayashizaki Y, Murakawa Y 2017. Genomic instability of iPSCs: challenges towards their clinical applications. Stem Cell Rev 13:7–16
    [Google Scholar]
  56. 56.  Kim D, Kim CH, Moon JI, Chung YG, Chang MY et al. 2009. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell 4:472–76
    [Google Scholar]
  57. 57.  Lister R, Pelizzola M, Kida YS, Hawkins RD, Nery JR et al. 2011. Hotspots of aberrant epigenomic reprogramming in human induced pluripotent stem cells. Nature 471:68–73
    [Google Scholar]
  58. 58.  Ohi Y, Qin H, Hong C, Blouin L, Polo JM et al. 2011. Incomplete DNA methylation underlies a transcriptional memory of somatic cells in human iPS cells. Nat. Cell Biol. 13:541–49
    [Google Scholar]
  59. 59.  Polo JM, Liu S, Figueroa ME, Kulalert W, Eminli S et al. 2010. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat. Biotechnol. 28:848–55
    [Google Scholar]
  60. 60.  Kim K, Doi A, Wen B, Ng K, Zhao R et al. 2010. Epigenetic memory in induced pluripotent stem cells. Nature 467:285–90
    [Google Scholar]
  61. 61.  Mallon BS, Hamilton RS, Kozhich OA, Johnson KR, Fann YC et al. 2014. Comparison of the molecular profiles of human embryonic and induced pluripotent stem cells of isogenic origin. Stem Cell Res 12:376–86
    [Google Scholar]
  62. 62.  Rouhani F, Kumasaka N, de Brito MC, Bradley A, Vallier L, Gaffney D 2014. Genetic background drives transcriptional variation in human induced pluripotent stem cells. PLOS Genet 10:e1004432
    [Google Scholar]
  63. 63.  Carcamo-Orive I, Hoffman GE, Cundiff P, Beckmann ND, D'Souza SL et al. 2017. Analysis of transcriptional variability in a large human iPSC library reveals genetic and non-genetic determinants of heterogeneity. Cell Stem Cell 20:518–32.e9
    [Google Scholar]
  64. 64.  Warren CR, O'Sullivan JF, Friesen M, Becker CE, Zhang X et al. 2017. Induced pluripotent stem cell differentiation enables functional validation of GWAS variants in metabolic disease. Cell Stem Cell 20:547–57.e7
    [Google Scholar]
  65. 65.  Benam KH, Dauth S, Hassell B, Herland A, Jain A et al. 2015. Engineered in vitro disease models. Annu. Rev. Pathol. 10:195–262
    [Google Scholar]
  66. 66.  Kimbrel EA, Lanza R 2016. Pluripotent stem cells: the last 10 years. Regen. Med. 11:831–47
    [Google Scholar]
  67. 67.  Musah S, Wrighton PJ, Zaltsman Y, Zhong X, Zorn S et al. 2014. Substratum-induced differentiation of human pluripotent stem cells reveals the coactivator YAP is a potent regulator of neuronal specification. PNAS 111:13805–10
    [Google Scholar]
  68. 68.  Ribeiro AJ, Ang YS, Fu JD, Rivas RN, Mohamed TM et al. 2015. Contractility of single cardiomyocytes differentiated from pluripotent stem cells depends on physiological shape and substrate stiffness. PNAS 112:12705–10
    [Google Scholar]
  69. 69.  Smith Q, Chan XY, Carmo AM, Trempel M, Saunders M, Gerecht S 2017. Compliant substratum guides endothelial commitment from human pluripotent stem cells. Sci. Adv. 3:e1602883
    [Google Scholar]
  70. 70.  Takebe T, Sekine K, Enomura M, Koike H, Kimura M et al. 2013. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 499:481–84
    [Google Scholar]
  71. 71.  Camp JG, Sekine K, Gerber T, Loeffler-Wirth H, Binder H et al. 2017. Multilineage communication regulates human liver bud development from pluripotency. Nature 546:533–38
    [Google Scholar]
  72. 72.  Park IH, Arora N, Huo H, Maherali N, Ahfeldt T et al. 2008. Disease-specific induced pluripotent stem cells. Cell 134:877–86
    [Google Scholar]
  73. 73.  Onder TT, Daley GQ 2012. New lessons learned from disease modeling with induced pluripotent stem cells. Curr. Opin. Genet. Dev. 22:500–8
    [Google Scholar]
  74. 74.  Trounson A, Shepard KA, DeWitt ND 2012. Human disease modeling with induced pluripotent stem cells. Curr. Opin. Genet. Dev. 22:509–16
    [Google Scholar]
  75. 75.  Vallier L, Segeritz CP 2016. Inherited metabolic disorders of the liver. Human iPS Cells in Disease Modelling K Fukuda 83–99 Tokyo: Springer
    [Google Scholar]
  76. 76.  Szkolnicka D, Hay DC 2016. Advances in generating hepatocytes from pluripotent stem cells for translational medicine. Stem Cells 34:1421–26
    [Google Scholar]
  77. 77.  Sampaziotis F, Segeritz CP, Vallier L 2015. Potential of human induced pluripotent stem cells in studies of liver disease. Hepatology 62:303–11
    [Google Scholar]
  78. 78.  Rashid ST, Corbineau S, Hannan N, Marciniak SJ, Miranda E et al. 2010. Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells. J. Clin. Investig. 120:3127–36
    [Google Scholar]
  79. 79.  Takebe T, Zhang RR, Koike H, Kimura M, Yoshizawa E et al. 2014. Generation of a vascularized and functional human liver from an iPSC-derived organ bud transplant. Nat. Protoc. 9:396–409
    [Google Scholar]
  80. 80.  Roy-Chowdhury N, Wang X, Guha C, Roy-Chowdhury J 2017. Hepatocyte-like cells derived from induced pluripotent stem cells. Hepatol. Int. 11:54–69
    [Google Scholar]
  81. 81.  Hannan NR, Segeritz CP, Touboul T, Vallier L 2013. Production of hepatocyte-like cells from human pluripotent stem cells. Nat. Protoc. 8:430–37
    [Google Scholar]
  82. 82.  Palakkan AA, Nanda J, Ross JA 2017. Pluripotent stem cells to hepatocytes, the journey so far. Biomed. Rep. 6:367–73
    [Google Scholar]
  83. 83.  Liu H, Ye Z, Kim Y, Sharkis S, Jang YY 2010. Generation of endoderm-derived human induced pluripotent stem cells from primary hepatocytes. Hepatology 51:1810–19
    [Google Scholar]
  84. 84.  Si-Tayeb K, Noto FK, Nagaoka M, Li J, Battle MA et al. 2010. Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells. Hepatology 51:297–305
    [Google Scholar]
  85. 85.  Sullivan GJ, Hay DC, Park IH, Fletcher J, Hannoun Z et al. 2010. Generation of functional human hepatic endoderm from human induced pluripotent stem cells. Hepatology 51:329–35
    [Google Scholar]
  86. 86.  Song Z, Cai J, Liu Y, Zhao D, Yong J et al. 2009. Efficient generation of hepatocyte-like cells from human induced pluripotent stem cells. Cell Res 19:1233–42
    [Google Scholar]
  87. 87.  Yu Y, Liu H, Ikeda Y, Amiot BP, Rinaldo P et al. 2012. Hepatocyte-like cells differentiated from human induced pluripotent stem cells: relevance to cellular therapies. Stem Cell Res 9:196–207
    [Google Scholar]
  88. 88.  Sakurai F, Mitani S, Yamamoto T, Takayama K, Tachibana M et al. 2017. Human induced-pluripotent stem cell–derived hepatocyte-like cells as an in vitro model of human hepatitis B virus infection. Sci. Rep. 7:45698
    [Google Scholar]
  89. 89.  Wu X, Robotham JM, Lee E, Dalton S, Kneteman NM et al. 2012. Productive hepatitis C virus infection of stem cell–derived hepatocytes reveals a critical transition to viral permissiveness during differentiation. PLOS Pathog 8:e1002617
    [Google Scholar]
  90. 90.  Wang Y, Alhaque S, Cameron K, Meseguer-Ripolles J, Lucendo-Villarin B et al. 2017. Defined and scalable generation of hepatocyte-like cells from human pluripotent stem cells. J. Vis. Exp. 121:e55355
    [Google Scholar]
  91. 91.  Cameron K, Tan R, Schmidt-Heck W, Campos G, Lyall MJ et al. 2015. Recombinant laminins drive the differentiation and self-organization of hESC-derived hepatocytes. Stem Cell Rep 5:1250–62
    [Google Scholar]
  92. 92.  Gieseck RL 3rd, Hannan NR, Bort R, Hanley NA, Drake RA et al. 2014. Maturation of induced pluripotent stem cell derived hepatocytes by 3D-culture. PLOS ONE 9:e86372
    [Google Scholar]
  93. 93.  Sampaziotis F, de Brito MC, Madrigal P, Bertero A, Saeb-Parsy K et al. 2015. Cholangiocytes derived from human induced pluripotent stem cells for disease modeling and drug validation. Nat. Biotechnol. 33:845–52
    [Google Scholar]
  94. 94.  Furuyama K, Kawaguchi Y, Akiyama H, Horiguchi M, Kodama S et al. 2011. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat. Genet. 43:34–41
    [Google Scholar]
  95. 95.  Strazzabosco M, Fabris L 2013. The balance between Notch/Wnt signaling regulates progenitor cells’ commitment during liver repair: Mystery solved?. J. Hepatol. 58:181–83
    [Google Scholar]
  96. 96.  Sampaziotis F, de Brito MC, Geti I, Bertero A, Hannan NR, Vallier L 2017. Directed differentiation of human induced pluripotent stem cells into functional cholangiocyte-like cells. Nat. Protoc. 12:814–27
    [Google Scholar]
  97. 97.  Si-Tayeb K, Lemaigre FP, Duncan SA 2010. Organogenesis and development of the liver. Dev. Cell 18:175–89
    [Google Scholar]
  98. 98.  O'Hara SP, Tabibian JH, Splinter PL, LaRusso NF 2013. The dynamic biliary epithelia: molecules, pathways, and disease. J. Hepatol. 58:575–82
    [Google Scholar]
  99. 99.  Schwartz RE, Trehan K, Andrus L, Sheahan TP, Ploss A et al. 2012. Modeling hepatitis C virus infection using human induced pluripotent stem cells. PNAS 109:2544–48
    [Google Scholar]
  100. 100.  Yusa K, Rashid ST, Strick-Marchand H, Varela I, Liu PQ et al. 2011. Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells. Nature 478:391–94
    [Google Scholar]
  101. 101.  Unternaehrer JJ, Daley GQ 2011. Induced pluripotent stem cells for modelling human diseases. Philos. Trans. R. Soc. B 366:2274–85
    [Google Scholar]
  102. 102.  Chou JY 2001. The molecular basis of type 1 glycogen storage diseases. Curr. Mol. Med. 1:25–44
    [Google Scholar]
  103. 103.  Andersson ER, Chivukula IV, Hankeova S, Sjoqvist M, Tsoi YL et al. 2017. Mouse model of Alagille syndrome and mechanisms of Jagged1 missense mutations. Gastroenterology 154:1080–95
    [Google Scholar]
  104. 104.  Gilbert MA, Spinner NB 2017. Alagille syndrome: genetics and functional models. Curr. Pathobiol. Rep. 5:233–41
    [Google Scholar]
  105. 105.  Turnpenny PD, Ellard S 2012. Alagille syndrome: pathogenesis, diagnosis and management. Eur. J. Hum. Genet. 20:251–57
    [Google Scholar]
  106. 106.  Grochowski CM, Loomes KM, Spinner NB 2016. Jagged1 (JAG1): structure, expression, and disease associations. Gene 576:381–84
    [Google Scholar]
  107. 107.  Guegan K, Stals K, Day M, Turnpenny P, Ellard S 2012. JAG1 mutations are found in approximately one third of patients presenting with only one or two clinical features of Alagille syndrome. Clin. Genet. 82:33–40
    [Google Scholar]
  108. 108.  Fon Tacer K, Rozman D 2011. Nonalcoholic fatty liver disease: focus on lipoprotein and lipid deregulation. J. Lipids 2011:783976
    [Google Scholar]
  109. 109.  Tiniakos DG, Vos MB, Brunt EM 2010. Nonalcoholic fatty liver disease: pathology and pathogenesis. Annu. Rev. Pathol. 5:145–71
    [Google Scholar]
  110. 110.  Hannoush ZC, Puerta H, Bauer MS, Goldberg RB 2017. New JAG1 mutation causing Alagille syndrome presenting with severe hypercholesterolemia: case report with emphasis on genetics and lipid abnormalities. J. Clin. Endocrinol. Metab. 102:350–53
    [Google Scholar]
  111. 111.  Wruck W, Graffmann N, Kawala MA, Adjaye J 2017. Current status and future directions on research related to nonalcoholic fatty liver disease. Stem Cells 35:89–96
    [Google Scholar]
  112. 112.  Graffmann N, Ring S, Kawala MA, Wruck W, Ncube A et al. 2016. Modeling nonalcoholic fatty liver disease with human pluripotent stem cell-derived immature hepatocyte-like cells reveals activation of PLIN2 and confirms regulatory functions of peroxisome proliferator-activated receptor alpha. Stem Cells Dev 25:1119–33
    [Google Scholar]
  113. 113.  Rossant J, Tam PPL 2017. New insights into early human development: lessons for stem cell derivation and differentiation. Cell Stem Cell 20:18–28
    [Google Scholar]
  114. 114.  Yamada S, Nakashima T, Hirose A, Yoneyama A, Takeda T, Takakuwa T 2012. Developmental anatomy of the human embryo—3D-imaging and analytical techniques. The Human Embryo S Yamada, T Takakuwa London: Intech Open
    [Google Scholar]
/content/journals/10.1146/annurev-pathol-020117-043634
Loading
/content/journals/10.1146/annurev-pathol-020117-043634
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error