Review Special Issues

Stem Cell Engineering and Differentiation for Disease Modeling and Cell-based Therapies

  • The identification and characterization of stem cells, especially human embryonic stem cells, has revolutionized the field of developmental biology by providing an in vitro system to study human development. In addition, reprogramming adult cells from patients into an embryonic stem cell-like state using induced pluripotent stem cell (iPSC) technology can potentially generate an unlimited source of human tissue carrying genetic mutations that caused or facilitated disease development, providing unprecedented possibilities to model human disease in the culture dish. To do this, however, efficient differentiation methods to direct iPSCs through multiple progenitor stages to yield homogeneous populations of somatic cells must be established. Furthermore, disease modeling using iPSCs requires proper controls for this “disease-in-a-dish” approach. Therefore, methods to efficiently engineer the genome of iPSCs to correct the mutations become vital in stem cell research. Here we reviewed the iPSC generation techniques and several genome-editing tools, such as TALENs and CRISPR-Cas9, for performing iPSC gene knock-in and knockout. We also present several efficient stem cell directed differentiation methods for converting iPSCs to neural, hematopoietic, cardiac, and pancreatic lineages. Together, this knowledge will provide insight into design principles for disease modeling using iPSCs and stem cell-based therapies.

    Citation: Lauren N. Randolph, Yuqian Jiang, Xiaojun Lian. Stem Cell Engineering and Differentiation for Disease Modeling and Cell-based Therapies[J]. AIMS Cell and Tissue Engineering, 2017, 1(2): 140-157. doi: 10.3934/celltissue.2017.2.140

    Related Papers:

  • The identification and characterization of stem cells, especially human embryonic stem cells, has revolutionized the field of developmental biology by providing an in vitro system to study human development. In addition, reprogramming adult cells from patients into an embryonic stem cell-like state using induced pluripotent stem cell (iPSC) technology can potentially generate an unlimited source of human tissue carrying genetic mutations that caused or facilitated disease development, providing unprecedented possibilities to model human disease in the culture dish. To do this, however, efficient differentiation methods to direct iPSCs through multiple progenitor stages to yield homogeneous populations of somatic cells must be established. Furthermore, disease modeling using iPSCs requires proper controls for this “disease-in-a-dish” approach. Therefore, methods to efficiently engineer the genome of iPSCs to correct the mutations become vital in stem cell research. Here we reviewed the iPSC generation techniques and several genome-editing tools, such as TALENs and CRISPR-Cas9, for performing iPSC gene knock-in and knockout. We also present several efficient stem cell directed differentiation methods for converting iPSCs to neural, hematopoietic, cardiac, and pancreatic lineages. Together, this knowledge will provide insight into design principles for disease modeling using iPSCs and stem cell-based therapies.


    加载中
    [1] Takahashi K, Tanabe K, Ohnuki M, et al. (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131: 861-872. doi: 10.1016/j.cell.2007.11.019
    [2] Yu J, Vodyanik MA, Smugaotto K, et al. (2007) Induced Pluripotent Stem Cell Lines Derived from Human Somatic Cells. Stem Cell Rev 8: 693-702.
    [3] Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. (1998) Embryonic stem cell lines derived from human blastocysts. Science 282: 1145-1147.
    [4] Takahashi K, Yamanaka S (2006) Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell 126: 663-676. doi: 10.1016/j.cell.2006.07.024
    [5] Okita K, Ichisaka T, Yamanaka S (2007) Generation of germline-competent induced pluripotent stem cells. Nature 448: 313-317. doi: 10.1038/nature05934
    [6] Wernig M, Meissner A, Foreman R, et al. (2007) In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448: 318-324. doi: 10.1038/nature05944
    [7] Maherali N, Sridharan R, Xie W, et al. (2017) Directly Reprogrammed Fibroblasts Show Global Epigenetic Remodeling and Widespread Tissue Contribution. Cell Stem Cell 1: 55-70.
    [8] Sumi T, Tsuneyoshi N, Nakatsuji N, et al. (2007) Apoptosis and differentiation of human embryonic stem cells induced by sustained activation of c-Myc. Oncogene 26: 5564-5576. doi: 10.1038/sj.onc.1210353
    [9] Baharvand H, Hassani S (2013) Pluripotent Stem Cells. 997: 13-23.
    [10] Faravelli I, Bucchia M, Rinchetti, et al. (2014) Motor neuron derivation from human embryonic and induced pluripotent stem cells: experimental approaches and clinical perspectives. Stem Cell Res Ther 5: 87. doi: 10.1186/scrt476
    [11] Du ZW, Chen H, Liu H, et al. (2015) Generation and expansion of highly pure motor neuron progenitors from human pluripotent stem cells. Nat Commun 6: 6626. doi: 10.1038/ncomms7626
    [12] Wichterle H, Lieberam I, Porter JA, et al. (2002) Directed differentiation of embryonic stem cells into motor neurons. Cell 110: 385-397. doi: 10.1016/S0092-8674(02)00835-8
    [13] Takazawa T, Croft GF, Amoroso MW, et al. (2012) Maturation of spinal motor neurons derived from human embryonic stem cells. PLoS One 7: 1-9.
    [14] Broccoli V, Rubio A, Taverna S, et al. (2015) Overcoming the hurdles for a reproducible generation of human functionally mature reprogrammed neurons. Exp Biol Med 240: 787-794. doi: 10.1177/1535370215577585
    [15] Miller JD, Ganat YM, Kishinevsky S, et al. (2013) Human iPSC-Based Modeling of Late-Onset Disease via Progerin-Induced Aging. Cell Stem Cell 13: 691-705. doi: 10.1016/j.stem.2013.11.006
    [16] Clevers H (2016) Modeling Development and Disease with Organoids. Cell 165: 1586-1597. doi: 10.1016/j.cell.2016.05.082
    [17] Doetschman TC, Eistetter H, Katz M, et al. (1985) The in vitro development of blastocyst-derived embryonic stem cell lines: formation of visceral yolk sac, blood islands and myocardium. J Embryol Exp Morphol 87: 27-45.
    [18] Bu L, Jiang X, Martinpuig S, et al. (2009) Human ISL1 heart progenitors generate diverse multipotent cardiovascular cell lineages. Nature 460: 113-117. doi: 10.1038/nature08191
    [19] Elliott DA, Braam SR, Koutsis K, et al. (2011) NKX2-5(eGFP/w) hESCs for isolation of human cardiac progenitors and cardiomyocytes. Nat Methods 8: 1037-1040. doi: 10.1038/nmeth.1740
    [20] Bao ZZ, Bruneau BG, Seidman JG, et al. (1999) Regulation of chamber-specific gene expression in the developing heart by Irx4. Science 283: 1161-1164. doi: 10.1126/science.283.5405.1161
    [21] O'Brien TX, Lee KJ, Chien KR (1993) Positional specification of ventricular myosin light chain 2 expression in the primitive murine heart tube. Proc Natl Acad Sci USA 90: 5157-5161. doi: 10.1073/pnas.90.11.5157
    [22] Protze SI, Liu J, Nussinovitch U, et al. (2017) Sinoatrial node cardiomyocytes derived from human pluripotent cells function as a biological pacemaker. Nat Biotechnol 35: 56-68.
    [23] Xu C, Police S, Rao N, et al. (2002) Characterization and enrichment of cardiomyocytes derived from human embryonic stem cells. Circ Res 91: 501-508. doi: 10.1161/01.RES.0000035254.80718.91
    [24] Laflamme MA, Gold J, Xu C, et al. (2005) Formation of human myocardium in the rat heart from human embryonic stem cells. Am J Pathol 167: 663-671. doi: 10.1016/S0002-9440(10)62041-X
    [25] Laflamme MA, Chen KY, Naumova AV, et al. (2007) Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat Biotechnol 25: 1015-1024. doi: 10.1038/nbt1327
    [26] Paige SL, Osugi T, Afanasiev OK, et al. (2010) Endogenous Wnt/Beta-Catenin signaling is required for cardiac differentiation in human embryonic stem cells. PLoS One 5: e11134. doi: 10.1371/journal.pone.0011134
    [27] Lian X, Palecek SP (2012) Cozzarelli Prize Winner: Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci 109: E1848-E1857. doi: 10.1073/pnas.1200250109
    [28] Lian X, Zhang J, Azarin SM, et al. (2013) Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions. Nat Protoc 8: 162-175.
    [29] Lian X, Bao X, Zilberter M, et al. (2015) Chemically defined, albumin-free human cardiomyocyte generation. Nat Methods 12: 595-596. doi: 10.1038/nmeth.3448
    [30] Pei F, Jiang J, Bai S, et al. (2017) Chemical-de fi ned and albumin-free generation of human atrial and ventricular myocytes from human pluripotent stem cells. Stem Cell Res 19: 94-103. doi: 10.1016/j.scr.2017.01.006
    [31] Kehat I, Khimovich L, Caspi O, et al. (2004) Electromechanical integration of cardiomyocytes derived from human embryonic stem cells. Nat Biotechnol 22: 1282-1289. doi: 10.1038/nbt1014
    [32] Zhang Q, Jiang J, Han P, et al. (2011) Direct differentiation of atrial and ventricular myocytes from human embryonic stem cells by alternating retinoid signals. Cell Res 21: 579-587. doi: 10.1038/cr.2010.163
    [33] Cao N, Huang Y, Zheng J, et al. (2016) Conversion of human fibroblasts into functional cardiomyocytes by small molecules. Science 352: 1216-1220. doi: 10.1126/science.aaf1502
    [34] Maizels L, Huber I, Arbel G, et al. (2017) Patient-Specific Drug Screening Using a Human Induced Pluripotent Stem Cell Model of Catecholaminergic Polymorphic Ventricular Tachycardia Type 2. Circ Arrhythmia Electrophysiol 10: 1-15.
    [35] Ackermann M, Liebhaber S, Klusmann JH, et al. (2015) Lost in translation: pluripotent stem cell-derived hematopoiesis. EMBO Mol Med 7: 1388-1402. doi: 10.15252/emmm.201505301
    [36] Chadwick K, Wang L, Li L, et al. (2003) Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells. Blood 102: 906-915. doi: 10.1182/blood-2003-03-0832
    [37] Woods NB, Parker AS, Moraghebi R, et al. (2011) Brief report: efficient generation of hematopoietic precursors and progenitors from human pluripotent stem cell lines. Stem Cells 29: 1158-1164. doi: 10.1002/stem.657
    [38] Ng ES, Azzola L, Bruveris FF, et al. (2016) Differentiation of human embryonic stem cells to HOXA+ hemogenic vasculature that resembles the aorta-gonad-mesonephros. Nat Biotechnol 34: 1168-1179. doi: 10.1038/nbt.3702
    [39] Galat Y, Dambaeva S, Elcheva I, et al. (2017) Cytokine-free directed differentiation of human pluripotent stem cells efficiently produces hemogenic endothelium with lymphoid potential. Stem Cell Res Ther 8: 67. doi: 10.1186/s13287-017-0519-0
    [40] Lian X, Bao X, Ai-Ahmad A, et al. (2014) Efficient Differentiation of Human Pluripotent Stem Cells to Endothelial Progenitors via Small-Molecule Activation of WNT Signalling. Stem Cell Rep 3: 804-816. doi: 10.1016/j.stemcr.2014.09.005
    [41] Sugimura R, Jha DK, Han A, et al. (2017) Haematopoietic stem and progenitor cells from human pluripotent stem cells. Nature 545: 432-438. doi: 10.1038/nature22370
    [42] Lumelsky N, Blondel O, Laeng P, et al. (2001) Differentiation of Embryonic Stem Cells to Insulin-Secreting Structures Similar to Pancreatic Islets. Science 292: 1389-1394. doi: 10.1126/science.1058866
    [43] Shi Y, Hou L, Tang F, et al. (2005) Inducing Embryonic Stem Cells to Differentiate into Pancreatic β Cells by a Novel Three-Step Approach with Activin A and All- Trans Retinoic Acid. Stem Cells 23: 656-662. doi: 10.1634/stemcells.2004-0241
    [44] D'Amour KA, Agulnick AD, Eliazer S, et al. (2005) Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol 23: 1534-1541. doi: 10.1038/nbt1163
    [45] D'Amour KA, Bang AG, Eliazer S, et al. (2006) Production of pancreatic hormone–expressing endocrine cells from human embryonic stem cells. Nat Biotechnol 24: 1392-1401. doi: 10.1038/nbt1259
    [46] Jiang J, Au M, Lu K, et al. (2007) Generation of Insulin-Producing Islet-Like Clusters from Human Embryonic Stem Cells. Stem Cells 25: 1940-1953. doi: 10.1634/stemcells.2006-0761
    [47] Nostro MC, Sarangi F, Ogawa S, et al. (2011) Stage-specific signaling through TGFβ family members and WNT regulates patterning and pancreatic specification of human pluripotent stem cells. Development 138: 861-871. doi: 10.1242/dev.055236
    [48] Pagliuca FW, Millman JR, Gurtler M, et al. (2014) Generation of functional human pancreatic β cells in vitro. Cell 159: 428-439. doi: 10.1016/j.cell.2014.09.040
    [49] Millman JR, Xie C, Van DA, et al. (2016) Generation of stem cell-derived β-cells from patients with type 1 diabetes. Nat Commun 7: 11463. doi: 10.1038/ncomms11463
    [50] Kim YG, Cha J, Chandrasegaran S (1996) Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc Natl Acad Sci USA 93: 1156-1160. doi: 10.1073/pnas.93.3.1156
    [51] Porteus MH, Carroll D (2005) Gene targeting using zinc finger nucleases. Nat Biotechnol 23: 967-973. doi: 10.1038/nbt1125
    [52] Sanjana NE, Cong L, Zhou Y, et al. (2012) A transcription activator-like effector toolbox for genome engineering. Nat Protoc 7: 171-192. doi: 10.1038/nprot.2011.431
    [53] Ishino Y, Shinagawa H, Makino K, et al. (1987) Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. J Bacteriol 169: 5429-5433. doi: 10.1128/jb.169.12.5429-5433.1987
    [54] Jansen R, Embden JDA, Gaastra W, at al. (2002) Identification of genes that are associated with DNA repeats in prokaryotes. Mol Microbiol 43: 1565-1575. doi: 10.1046/j.1365-2958.2002.02839.x
    [55] Mojica FJM, Díez-Villaseñor C, García-Martínez J, et al. (2005) Intervening Sequences of Regularly Spaced Prokaryotic Repeats Derive from Foreign Genetic Elements. J Mol Evol 60: 174-182. doi: 10.1007/s00239-004-0046-3
    [56] Hsu PD, Lander ES, Zhang, F (2014) Development and Applications of CRISPR-Cas9 for Genome Engineering. Cell 157: 1262-1278. doi: 10.1016/j.cell.2014.05.010
    [57] Cong L, Ran FA, Cox D, et al. (2013) Multiplex Genome Engineering Using CRISPR/Cas Systems. Science 339: 819-823. doi: 10.1126/science.1231143
    [58] Zhang JP, Li XL, Li GH, et al. (2017) Efficient precise knockin with a double cut HDR donor after CRISPR / Cas9-mediated double-stranded DNA cleavage. Genome Biol 18: 35. doi: 10.1186/s13059-017-1164-8
    [59] Zhang XH, Tee LY, Wang XG, et al. (2015) Off-target Effects in CRISPR/Cas9-mediated Genome Engineering. Mol Ther Nucleic Acids 4: e264. doi: 10.1038/mtna.2015.37
    [60] Tsai SQ, Wyvekens N, Khayter C, et al. (2014) Dimeric CRISPR RNA-guided FokI nucleases for highly specific genome editing. Nat Biotechnol 32: 569-576. doi: 10.1038/nbt.2908
    [61] Davis RP, Ng ES, Costa M, et al. (2008) Targeting a GFP reporter gene to the MIXL1 locus of human embryonic stem cells identifies human primitive streak – like cells and enables isolation of primitive hematopoietic precursors. Blood 111: 1876-1884. doi: 10.1182/blood-2007-06-093609
    [62] Lian X, Xu J, Bao X, et al. (2016) Interrogating Canonical Wnt Signaling Pathway in Human Pluripotent Stem Cell Fate Decisions Using CRISPR-Cas9. Cell Mol Bioeng 9: 325-334. doi: 10.1007/s12195-016-0453-8
    [63] Wang H, Luo X, Yao L, et al. (2015) Improvement of Cell Survival During Human Pluripotent Stem Cell Definitive Endoderm Differentiation. Stem Cells Dev 24: 2536-2546. doi: 10.1089/scd.2015.0018
    [64] Clarke RL, Yzaguirre AD, Yashiro-Ohtani Y, et al. (2013) The expression of Sox17 identifies and regulates haemogenic endothelium. Nat Cell Biol 15: 502-510. doi: 10.1038/ncb2724
    [65] Wang P, Rodriguez RT, Wang J, et al. (2011) Targeting SOX17 in human embryonic stem cells creates unique strategies for isolating and analyzing developing endoderm. Cell Stem Cell 8: 335-346. doi: 10.1016/j.stem.2011.01.017
    [66] Hinson JT, Chopra A, Nafissi N, et al. (2015) Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 349: 982-987. doi: 10.1126/science.aaa5458
    [67] Nelson CE, Hakim CH, Ousterout DG, et al. (2016) In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystropy. Science 351: 403-408. doi: 10.1126/science.aad5143
    [68] Chen Y, Yu J, Niu Y, et al. (2017) Modeling Rett Syndrome Using TALEN-Edited MECP2 Mutant Cynomolgus Monkeys. Cell 169: 945-955. doi: 10.1016/j.cell.2017.04.035
  • Reader Comments
  • © 2017 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(4102) PDF downloads(1063) Cited by(5)

Article outline

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog