Summary

ヒト多能性のための代替培養はセル生産、メンテナンス、および遺伝子解析幹

Published: July 24, 2014
doi:

Summary

Here, we present human pluripotent stem cell (hPSC) culture protocols, based on non-colony type monolayer (NCM) growth of dissociated single cells. This new method, utilizing Rho-associated kinase inhibitors or the laminin isoform 521 (LN-521), is suitable for producing large amounts of homogeneous hPSCs, genetic manipulation, and drug discovery.

Abstract

Human pluripotent stem cells (hPSCs) hold great promise for regenerative medicine and biopharmaceutical applications. Currently, optimal culture and efficient expansion of large amounts of clinical-grade hPSCs are critical issues in hPSC-based therapies. Conventionally, hPSCs are propagated as colonies on both feeder and feeder-free culture systems. However, these methods have several major limitations, including low cell yields and generation of heterogeneously differentiated cells. To improve current hPSC culture methods, we have recently developed a new method, which is based on non-colony type monolayer (NCM) culture of dissociated single cells. Here, we present detailed NCM protocols based on the Rho-associated kinase (ROCK) inhibitor Y-27632. We also provide new information regarding NCM culture with different small molecules such as Y-39983 (ROCK I inhibitor), phenylbenzodioxane (ROCK II inhibitor), and thiazovivin (a novel ROCK inhibitor). We further extend our basic protocol to cultivate hPSCs on defined extracellular proteins such as the laminin isoform 521 (LN-521) without the use of ROCK inhibitors. Moreover, based on NCM, we have demonstrated efficient transfection or transduction of plasmid DNAs, lentiviral particles, and oligonucleotide-based microRNAs into hPSCs in order to genetically modify these cells for molecular analyses and drug discovery. The NCM-based methods overcome the major shortcomings of colony-type culture, and thus may be suitable for producing large amounts of homogeneous hPSCs for future clinical therapies, stem cell research, and drug discovery.

Introduction

多系列成体組織に向かって差別化するhPSCsの容量は、心血管、肝臓、膵臓、および神経システム1-4を伴う重篤な疾患に苦しむ患者の治療に新たな道を開いた。 hPSCsから派生した様々な細胞型はまた、疾患のモデリング、遺伝子工学、薬物スクリーニング、および毒性試験1,4のための堅牢な携帯プラットフォームを提供するであろう。将来の臨床的および薬理学的用途を保証する重要な問題は、 インビトロでの細胞培養を介して臨床グレードhPSCs多数の生成である。しかし、現在の培養系ではコロニー5,6としてhPSCsの様々なフィーダーフィーダーフリーの培養液を含む、不十分であったり、本質的に可変のどちらかである。

hPSCs株哺乳動物の初期胚の内部細胞塊(ICM)の多くの構造的特徴のコロニー型の増殖。 ICMは3生殖層に分化する傾向がある多細胞の環境であるため、異種シグナル勾配の存在。このように、初期胚発生における不均一性の獲得は、分化に必要なプロセスであると考えますが、HPSC文化の不要な機能です。 HPSC培養における不均一性は、多くの場合、過度のアポトーシスシグナルおよび準最適な成長条件による自発的分化によって誘導される。従って、コロニータイプの培養で、不均一な細胞は、しばしば、コロニー7,8の周囲に観察される。また、ヒト胚性幹細胞(hESCの)中の細胞が、例えばBMP-4 9とシグナル伝達分子に展示差動応答をコロニーことが示されている。また、コロニー培養法は、制御不能の成長率およびアポトーシスシグナル伝達経路6,9に低い細胞収量だけでなく、凍結保存と非常に低い細胞回収率を生成する。近年、種々の懸濁培養は、培養のためにhPSCs particulが開発されているフィーダーマトリックスを含まない条件下6,10-13中hPSCs、大量の拡張のためアルリー。明らかに、別の培養システムは、独自の長所と短所を有する。一般に、hPSCsの不均一な性質を遺伝子工学6 hPSCsにDNAとRNAの物質を送達するための準最適であるのコロニー型および凝集培養法における主な欠点のうちの1つを表す。

明らかに、現在の培養方法のいくつかの欠点を回避する新しいシステムを開発する必要性が不可欠である。単一細胞の生存を改善する(このようなROCK阻害剤Y-27632およびJAK阻害剤1のような)低分子阻害剤の発見は解離し、HPSC文化14,15のための道を開く。これらの小分子を使用することで、我々は最近、非コロニー型(NCM)を解離·hPSCs 9の成長に基づいた培養法を開発した。この新規培養法は、単一細胞継代と高密度の両方を兼ね備え、メッキ法、私たちは主要な染色体異常9なしで一貫性のある成長サイクルの下で均質hPSCsを大量に生成することができます。あるいは、NCM培養は広い用途のための培養方法を最適化するために、異なる小分子と(例えば、ラミニンなど)が定義行列を用いて実施され得る。ここでは、NCMのカルチャに基づいて、いくつかの詳細なプロトコルを提示し、遺伝子工学のための詳細な手順を描く。 NCMプロトコルの汎用性を実証するために、我々はまた、多様なROCK阻害剤で、単一のラミニンアイソフォーム521( すなわち 、LN-521)でNCM文化をテストしました。

Protocol

hPSCsの単一細胞に基づく非植民地型単層(NCM)の文化。 1。準備 10%FBS、2mMのL-グルタミン、および0.1mM非必須アミノ酸(NEAA)を補充したDMEM培地:マウス胚線維芽細胞(MEF)の培養のための培地500mlを加える。 DMEM培地中の0.1%ゼラチンでコーティングされた6ウェル細胞培養プレート上のルーチンプロトコル16と培養したMEF以下CF1株に由来するマウ?…

Representative Results

NCM文化の一般的なスキーマ 図1に、ROCK阻害剤Y-27632の存在下での高密度単一細胞めっき後hPSCsの動的な変化を示す典型的なNCM培養スキーマを表します。これらの形態学的変化は、細胞のクラスター形成をプレーティングし、細胞縮合、続いて指数関数的な細胞増殖( 図1A)の後に細胞間接続を含む。代表的な実験は、1日目に、10μMのY…

Discussion

従来の(フィーダまたは細胞外マトリックス上の細胞の)コロニー型の文化やフィーダー6のない集合体としてhPSCsの懸濁培養:in vitroでの培養hPSCsには、2つの主要な方法があります。コロニー型とサスペンションの両方培養法の制限は累積異質と継承エピジェネティックな変化があります。 NCM培養物は、単一細胞継代及び高密度細胞播種の両方に基づいて、HPSC成長6,18?…

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was supported by the Intramural Research Program of the National Institutes of Health (NIH) at the National Institute of Neurological Disorders and Stroke. We would like to thank Dr. Ronald D. McKay for his discussion and comments on this project.

Materials

Countess automated cell counter   Invitrogen Inc.  C10227 Automatic cell counting
Faxitron Cabinet X-ray System Faxitron X-ray Corporation, Wheeling, IL  Model RX-650 X-ray irradiation of MEFs
MULTIWELL six-well plates  Becton Dickinson Labware 353046 Polystyrene plates 
DMEM Invitrogen Inc. 11965–092 For MEF medium
mitomycin C Roche  107 409 Mitotic inhibitor
Trypsin Invitrogen Inc. 25300-054 For MEF dissociation
DMEM/F12  Invitrogen Inc. 11330–032 For hPSC medium
Opti-MEM I Reduced Serum Medium  Invitrogen Inc. 31985-062 For hPSC transfection
Heat-inactivated FBS Invitrogen Inc. 16000–044 Component of MEF medium
Knockout Serum Replacer  Invitrogen Inc. 10828–028 KSR, Component of hPSC medium
Dulbecco’s Phosphate-Buffered Saline Invitrogen Inc. 14190-144 D-PBS, free of Ca2+/Mg2+
Non-essential amino acids  Invitrogen  11140–050 NEAA, component of hPSC medium
L-Glutamine  Invitrogen  25030–081 Component of hPSC medium
mTeSR1 & Supplements StemCell Technologies 5850 Animal protein-free
medium
TeSR2 & Supplements StemCell Technologies 5860 Xeno-free medium
β-mercaptoethanol  Sigma  7522 Component of hPSC medium

MEF (CF-1) ATCC
American Type Culture Collection (ATCC)  SCRC-1040 For feeder culture of hPSCs
hESC-qualified Matrigel BD Bioscience 354277 For feeder-free culture of hPSCs
Laminin-521 BioLamina LN521-02 Human recombinant protein
FGF-2 (recombinant FGF, basic) R&D Systems, MN 223-FB Growth factor in hPSC medium
CryoStor CA10  StemCell Technologies 7930
Accutase Innovative Cell Technologies AT-104 1X mixed enzymatic solution
JAK inhibitor I EMD4 Biosciences 420099 An inhibitor of Janus kinase
Y-27632 EMD4 Biosciences 688000 ROCK inhibitor
Y-27632 Stemgent 04-0012 ROCK inhibitor
Y-39983 Stemgent 04-0029 ROCK I inhibitor
Phenylbenzodioxane  Stemgent 04-0030 ROCK II inhibitor
Thiazovivin Stemgent 04-0017 A novel ROCK inhibitor
BD Falcon Cell Strainer  BD Bioscience 352340 40-µm cell strainer
Nalgene 5100-0001 Cryo 1°C Thermo Scientific  C6516F-1 “Mr. Frosty” Freezing Container
Lipofectamine 2000  Invitrogen Inc. 11668-027 Transfection reagents
DharmaFECT Duo  Thermo Scientific T-2010-02 Transfection reagent
Non-targeting miRIDIAN miRNA Transfection Control Thermo Scientific IP-004500-01-05 Labeled with Dy547, to monitor the delivery of microRNAs 
SMART-shRNA Thermo Scientific  To be determined Lentiviral vector
pmaxGFP amaxa Inc (Lonza) Included in every transfection kit Expression plasmid for transfection control
4-Oct Santa Cruz Biotechnology sc-5279 Mouse IgG2b, pluripotent marker
SSEA-1 Santa Cruz Biotechnology sc-21702 Mouse IgM, differentiation marker
SSEA-4 Santa Cruz Biotechnology sc-21704 Mouse IgG3, pluripotent marker
Tra-1-60 Santa Cruz Biotechnology sc-21705  Mouse IgM, pluripotent marker
Tra-1-81 Santa Cruz Biotechnology sc-21706 Mouse IgM, pluripotent marker
CK8 (C51) Santa Cruz Biotechnology sc-8020 Mouse IgG1, against cytokeratin 8
α-fetoprotein Santa Cruz Biotechnology sc-8399 AFP, mouse IgG2a
HNF-3β (P-19) Santa Cruz Biotechnology sc-9187 FOXA2, goat polyclonal antibody
Troponin T (Av-1) Thermo Scientific MS-295-P0 Mouse IgG1
Desmin  Thermo Scientific RB-9014-P1 Rabbit IgG
Anti-NANOG ReproCELL Inc, Japan RCAB0004P-F Polyclonal antibody 
Rat anti-GFAP Zymed 13-0300 Glial fibrillary acidic protein
Albumin (clone HSA1/25.1.3) Cedarlane Laboratories Ltd. ( CL2513A Mouse IgG1,
Smooth muscle actin (clone 1A4) DakoCytomation Inc IR611/IS611 Mouse IgG2a
Nestin Chemicon International MAB5326 Rabbit polyclonal antibody
TUBB3 Convance Inc MMS-435P Tuj1, mouse IgG2a
HNF4α (C11F12) Cell Signaling Technologies 3113 Rabbit monoclonal antibody
Paraformaldehyde (solution) Electron Microscopy Sciences 15710 PFA, fixative, diluted in D-PBS

References

  1. Cherry, A. B., Daley, G. Q. Reprogrammed cells for disease modeling and regenerative medicine. Annu Rev Med. 64, 277-290 (2013).
  2. Takahashi, K., Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 126, 663-676 (2006).
  3. Thomson, J. A., et al. Embryonic stem cell lines derived from human blastocysts. Science. 282, 1145-1147 (1998).
  4. Burridge, P. W., et al. Production of de novo cardiomyocytes: human pluripotent stem cell differentiation and direct reprogramming. Cell Stem Cell. 10, 16-28 (2012).
  5. Mallon, B. S., et al. StemCellDB: The Human Pluripotent Stem Cell Database at the National Institutes of Health. Stem Cell Res. 10, 57-66 (2012).
  6. Chen, K. G., et al. Human pluripotent stem cell culture: considerations for maintenance, expansion, and therapeutics. Cell Stem Cell. 14, 13-26 (2014).
  7. Bendall, S. C., et al. IGF and FGF cooperatively establish the regulatory stem cell niche of pluripotent human cells in vitro. Nature. 448, 1015-1021 (2007).
  8. Moogk, D., et al. Human ESC colony formation is dependent on interplay between self-renewing hESCs and unique precursors responsible for niche generation. Cytometry A. 77, 321-327 (2010).
  9. Chen, K. G., et al. Non-colony type monolayer culture of human embryonic stem cells. Stem Cell Res. 9, 237-248 (2012).
  10. Amit, M., et al. Suspension culture of undifferentiated human embryonic and induced pluripotent stem cells. Stem Cell Rev. 6, 248-259 (2010).
  11. Dang, S. M., et al. scalable embryonic stem cell differentiation culture. Stem Cells. 22, 275-282 (2004).
  12. Serra, M., et al. Process engineering of human pluripotent stem cells for clinical application. Trends Biotechnol. 30, 350-359 (2012).
  13. Steiner, D., et al. propagation and controlled differentiation of human embryonic stem cells in suspension. Nat Biotechnol. 28, 361-364 (2010).
  14. Watanabe, K., et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol. 25, 681-686 (2007).
  15. Androutsellis-Theotokis, A., et al. Notch signalling regulates stem cell numbers in vitro and in vivo. Nature. 442, 823-826 (2006).
  16. Jozefczuk, J., et al. Preparation of mouse embryonic fibroblast cells suitable for culturing human embryonic and induced pluripotent stem cells. J Vis Exp. , (2012).
  17. Kozhich, O. A., et al. Standardized Generation and Differentiation of Neural Precursor Cells from Human Pluripotent Stem Cells. Stem Cell Rev. , (2012).
  18. Kunova, M., et al. Adaptation to robust monolayer expansion produces human pluripotent stem cells with improved viability. Stem Cells Transl Med. 2, 246-254 (2013).
  19. Tsutsui, H., et al. An optimized small molecule inhibitor cocktail supports long-term maintenance of human embryonic stem cells. Nat Commun. 2, 167 (2011).
  20. Amps, K., et al. Screening ethnically diverse human embryonic stem cells identifies a chromosome 20 minimal amplicon conferring growth advantage. Nat Biotechnol. 29, 1132-1144 (2011).
  21. Baker, D. E., et al. Adaptation to culture of human embryonic stem cells and oncogenesis in vivo. Nat Biotechnol. 25, 207-215 (2007).
  22. Lee, A. S., et al. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat Med. 19, 998-1004 (2013).
  23. Domogatskaya, A., et al. Laminin-511 but not -332, -111, or -411 enables mouse embryonic stem cell self-renewal in vitro. Stem Cells. 26, 2800-2809 (2008).
  24. Domogatskaya, A., et al. Functional diversity of laminins. Annu Rev Cell Dev Biol. 28, 523-553 (2012).
  25. Rodin, S., et al. Long-term self-renewal of human pluripotent stem cells on human recombinant laminin-511. Nat Biotechnol. 28, 611-615 (2010).
  26. Liew, C. G., et al. Transient and stable transgene expression in human embryonic stem cells. Stem Cells. 25, 1521-1528 (2007).
  27. Braam, S. R., et al. Genetic manipulation of human embryonic stem cells in serum and feeder-free media. Methods Mol Biol. 584, 413-423 (2010).
  28. Braam, S. R., et al. Improved genetic manipulation of human embryonic stem cells. Nat Methods. 5, 389-392 (2008).

Play Video

Cite This Article
Chen, K. G., Hamilton, R. S., Robey, P. G., Mallon, B. S. Alternative Cultures for Human Pluripotent Stem Cell Production, Maintenance, and Genetic Analysis. J. Vis. Exp. (89), e51519, doi:10.3791/51519 (2014).

View Video