Summary

Directed Differentiation of Induced Pluripotent Stem Cells towards T Lymphocytes

Published: May 14, 2012
doi:

Summary

Generation of T lymphocytes from induced pluripotent stem (iPS) cells gives an alternative approach of using embryonic stem cells for T cell-based immunotherapy. The method shows that by utilizing either in vitro or in vivo induction system, iPS cells are able to differentiate into both conventional and antigen-specific T lymphocytes.

Abstract

Adoptive cell transfer (ACT) of antigen-specific CD8+ cytotoxic T lymphocytes (CTLs) is a promising treatment for a variety of malignancies 1. CTLs can recognize malignant cells by interacting tumor antigens with the T cell receptors (TCR), and release cytotoxins as well as cytokines to kill malignant cells. It is known that less-differentiated and central-memory-like (termed highly reactive) CTLs are the optimal population for ACT-based immunotherapy, because these CTLs have a high proliferative potential, are less prone to apoptosis than more differentiated cells and have a higher ability to respond to homeostatic cytokines 2-7. However, due to difficulties in obtaining a high number of such CTLs from patients, there is an urgent need to find a new approach to generate highly reactive Ag-specific CTLs for successful ACT-based therapies.

TCR transduction of the self-renewable stem cells for immune reconstitution has a therapeutic potential for the treatment of diseases 8-10. However, the approach to obtain embryonic stem cells (ESCs) from patients is not feasible. Although the use of hematopoietic stem cells (HSCs) for therapeutic purposes has been widely applied in clinic 11-13, HSCs have reduced differentiation and proliferative capacities, and HSCs are difficult to expand in in vitro cell culture 14-16. Recent iPS cell technology and the development of an in vitro system for gene delivery are capable of generating iPS cells from patients without any surgical approach. In addition, like ESCs, iPS cells possess indefinite proliferative capacity in vitro, and have been shown to differentiate into hematopoietic cells. Thus, iPS cells have greater potential to be used in ACT-based immunotherapy compared to ESCs or HSCs.

Here, we present methods for the generation of T lymphocytes from iPS cells in vitro, and in vivo programming of antigen-specific CTLs from iPS cells for promoting cancer immune surveillance. Stimulation in vitro with a Notch ligand drives T cell differentiation from iPS cells, and TCR gene transduction results in iPS cells differentiating into antigen-specific T cells in vivo, which prevents tumor growth. Thus, we demonstrate antigen-specific T cell differentiation from iPS cells. Our studies provide a potentially more efficient approach for generating antigen-specific CTLs for ACT-based therapies and facilitate the development of therapeutic strategies for diseases.

Protocol

1. Cell Culture Preparation of irradiated SNL76/7 (irSNL76/7) feeder cells for culture. SNL76/7 cells are generally maintained in 10% fetal bovine serum (FBS) Dulbecco’s Modified Eagle Medium (DMEM) media. A culture dish or flask will be coated with 0.1% gelatin solution in 37 °C; incubator for 30 minutes before recovering SNL76/7 cells from liquid nitrogen. When SNL76/7 cells reach confluency, cells will be trypsinized off, centrifuged at 400 g for 5 min and resuspen…

Discussion

For ACT-based therapies, the in vitro generation of large numbers of highly reactive Ag-specific T cells for in vivo re-infusion is an optimal approach. Although our in vitro method gives rise of functional T cells from iPS cells, large numbers of iPS cell-derived cells die in four weeks, especially in the fourth week. We conclude that the survival signals from Notch signaling mediated by the DL1 as well as IL-7 and FLt3L are not sufficient to maintain the survival of iPS cell-derived prog…

Declarações

The authors have nothing to disclose.

Acknowledgements

We thank Dr. Shinya Yamanaka (Kyoto University) for providing iPS-MEF-Ng-20D-17 cell line, Dr. Dario Vignali (St. Jude Children’s Research Hospital) for supporting the OT1-2A•pMig II construct, Dr. Juan Carlos Zuniga-Pflucker (Department of Immunology, University of Toronto) for supporting the OP9-DL1 cell line, and Dr. Kent E Vrana (Department of Pharmacology, Penn State University College of Medicine) for helping the design of this study. This project is funded, under grants with the Grant Number K18CA151798 from the National Cancer Institute, the Barsumian Trust and the Melanoma Research Foundation (J. Song).

Materials

Name of the reagent Company Catalogue number
C57BL/6J mice Jackson Laboratory 000664
B6.129S7-Rag1tm1Mom/J Jackson Laboratory 002216
Anti-CD3 (2C11) antibody BD PharMingen 553058
Anti-CD28 (37.51) antibody BD PharMingen 553295
Anti-CD3 (17A2) antibody BioLegend 100202
Anti-CD4 (GK1.5) antibody BioLegend 100417
Anti-CD8 (53-6.7) antibody BioLegend 100714
Anti-CD25 (3C7) antibody BioLegend 101912
Anti-CD44 (1M7) antibody BioLegend 103012
Anti-CD117 (2B8) antibody BioLegend 105812
Anti-TCR-β (H57597) antibody BioLegend 109220
Anti-IL-2 (JES6-5H4) antibody BioLegend 503810
Anti-IFN-γ (XMG1.2) antibody BioLegend 505822
DMEM Invitrogen ABCD1234
α-MEM Invitrogen A10490-01
FBS HyClone SH3007.01
Brefeldin A Sigma B7651
Polybrene Sigma 107689
GeneJammer Integrated Sciences 204130
RNA kit Qiagen 74104
DNA kit Qiagen 69504
CD8 Isolation Kit Miltenyi Biotec 130-095-236
ACK lysis buffer Lonza 10-548E
mFlt-3L PeproTech 250-31L
mIL-7 PeproTech 217-17
Gelatin Sigma G9391
FITC-anti-OVA antibody Rockland Immunochemicals 200-4233
Permeabilization buffer Biolegend 421002
BSA Sigma A7906
Formaldehyde Sigma F8775
0.4 μm filter MIllipore  
Moflo Cell Sorter Dake Cytomation  
Calibur Flow Cytometer BD  
LSR II Flow Cytometer BD  
Mouse restrainer Braintree Scientific  

Referências

  1. Brenner, M. K., Heslop, H. E. Adoptive T cell therapy of cancer. Curr. Opin. Immunol. 22, 251-257 (2010).
  2. Hataye, J., Moon, J. J., Khoruts, A., Reilly, C., Jenkins, M. K. Naive and memory CD4+ T cell survival controlled by clonal abundance. Science. 312, 114-116 (2006).
  3. Seki, Y. IL-7/STAT5 cytokine signaling pathway is essential but insufficient for maintenance of naive CD4 T cell survival in peripheral lymphoid organs. J. Immunol. 178, 262-270 (2007).
  4. Stemberger, C. A single naive CD8+ T cell precursor can develop into diverse effector and memory subsets. Immunity. 27, 985-997 (2007).
  5. Siewert, C. Experience-driven development: effector/memory-like alphaE+Foxp3+ regulatory T cells originate from both naive T cells and naturally occurring naive-like regulatory T cells. J. Immunol. 180, 146-155 (2008).
  6. Wang, L. X., Plautz, G. E. Tumor-primed, in vitro-activated CD4+ effector T cells establish long-term memory without exogenous cytokine support or ongoing antigen exposure. J. Immunol. 184, 5612-5618 (2010).
  7. Hinrichs, C. S. Human effector CD8+ T cells derived from naive rather than memory subsets possess superior traits for adoptive immunotherapy. Blood. 117, 808-814 (2011).
  8. Alajez, N. M., Schmielau, J., Alter, M. D., Cascio, M., Finn, O. J. Therapeutic potential of a tumor-specific, MHC-unrestricted T-cell receptor expressed on effector cells of the innate and the adaptive immune system through bone marrow transduction and immune reconstitution. Blood. 105, 4583-4589 (2005).
  9. Yang, L., Baltimore, D. Long-term in vivo provision of antigen-specific T cell immunity by programming hematopoietic stem cells. Proc. Natl. Acad. Sci. U.S.A. 102, 4518-4523 (2005).
  10. Zhao, Y. Extrathymic generation of tumor-specific T cells from genetically engineered human hematopoietic stem cells via Notch signaling. Cancer Res. 67, 2425-2429 (2007).
  11. Boztug, K., Med, N. .. E. n. g. l. .. J. .. Stem-cell gene therapy for the Wiskott-Aldrich syndrome. N. Engl. J. Med. 363, 1918-1927 (2010).
  12. Peerani, R., Zandstra, P. W. Enabling stem cell therapies through synthetic stem cell-niche engineering. J. Clin. Invest. 120, 60-70 (2010).
  13. Mendez-Ferrer, S. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 466, 829-834 (2010).
  14. Daley, G. Q. Towards the generation of patient-specific pluripotent stem cells for combined gene and cell therapy of hematologic disorders. Hematology Am. Soc. Hematol. Educ. Program. , 17-22 (2007).
  15. Boitano, A. E. Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science. 329, 1345-1348 (2010).
  16. Himburg, H. A. Pleiotrophin regulates the expansion and regeneration of hematopoietic stem cells. Nat. Med. 16, 475-482 (2010).
  17. Tanigaki, K., Honjo, T. Regulation of lymphocyte development by Notch signaling. Nature immunology. 8, 451-456 (2007).
  18. Zhao, T., Zhang, Z. N., Rong, Z., Xu, Y. Immunogenicity of induced pluripotent stem cells. Nature. 474, 212-215 (2011).
check_url/pt/3986?article_type=t

Play Video

Citar este artigo
Lei, F., Haque, R., Xiong, X., Song, J. Directed Differentiation of Induced Pluripotent Stem Cells towards T Lymphocytes. J. Vis. Exp. (63), e3986, doi:10.3791/3986 (2012).

View Video