Reprogramming of T Cells from Human Peripheral Blood

Slides:



Advertisements
Similar presentations
Volume 64, Issue 5, Pages (November 2013)
Advertisements

Volume 143, Issue 4, Pages (November 2010)
Volume 9, Issue 1, Pages (July 2017)
Volume 13, Issue 2, Pages (August 2013)
Generation of Induced Pluripotent Stem Cell Lines from Adult Rat Cells
Erythropoietic differentiation of a human embryonic stem cell line harbouring the sickle cell anaemia mutation  Marina V Pryzhkova, Ann Peters, Elias.
Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors  Kazutoshi Takahashi, Shinya Yamanaka  Cell 
Volume 64, Issue 5, Pages (November 2013)
Hypoxia Enhances the Generation of Induced Pluripotent Stem Cells
Volume 9, Issue 5, Pages (November 2017)
Volume 4, Issue 6, Pages (June 2009)
Volume 7, Issue 5, Pages (November 2010)
Generation of iPSCs from cultured human malignant cells
Metastable Pluripotent States in NOD-Mouse-Derived ESCs
Disease-Specific Induced Pluripotent Stem Cells
Volume 3, Issue 3, Pages (September 2008)
Volume 3, Issue 4, Pages (October 2008)
Volume 11, Issue 2, Pages (August 2012)
Volume 3, Issue 3, Pages (September 2014)
Patient-Specific Naturally Gene-Reverted Induced Pluripotent Stem Cells in Recessive Dystrophic Epidermolysis Bullosa  Jakub Tolar, John A. McGrath, Lily.
Volume 8, Issue 6, Pages (June 2011)
Volume 29, Issue 1, Pages (April 2014)
Volume 11, Issue 1, Pages 1-3 (July 2018)
Transduction of Human Embryonic Stem Cells by Foamy Virus Vectors
Cells Isolated from the Epidermis by Hoechst Dye Exclusion, Small Size, and Negative Selection for Hematopoietic Markers Can Generate B Lymphocyte Precursors 
Volume 4, Issue 5, Pages (May 2009)
Volume 13, Issue 6, Pages (December 2013)
Yang Li, Ph.D., Ji-chun Tan, M.D., Ph.D., Ling-song Li, M.D., Ph.D. 
Claire A. Higgins, Munenari Itoh, Keita Inoue, Gavin D
Volume 12, Issue 6, Pages (June 2013)
Volume 13, Issue 1, Pages (July 2013)
Volume 13, Issue 2, Pages (August 2013)
Volume 6, Issue 1, Pages (January 2016)
Volume 7, Issue 1, Pages (July 2010)
Wnt Signaling Promotes Reprogramming of Somatic Cells to Pluripotency
Generation of Induced Pluripotent Stem Cells from Human Cord Blood
Volume 8, Issue 6, Pages (June 2011)
Volume 7, Issue 1, Pages 1-10 (July 2016)
Disease-Specific Induced Pluripotent Stem Cells
Volume 6, Issue 2, Pages (February 2016)
Volume 46, Issue 1, Pages (April 2012)
Volume 9, Issue 2, Pages (August 2011)
Volume 7, Issue 4, Pages (October 2016)
Volume 12, Issue 1, Pages (January 2013)
Volume 16, Issue 9, Pages (August 2016)
Volume 2, Issue 3, Pages (March 2014)
Volume 131, Issue 5, Pages (November 2007)
Volume 11, Issue 1, Pages (July 2012)
Volume 3, Issue 5, Pages (November 2008)
Histone Modifications Associated with Somatic Hypermutation
Generation of Melanocytes from Induced Pluripotent Stem Cells
Volume 3, Issue 6, Pages (December 2008)
The synthetic Oct6 molecule contributes to epigenetic reprogramming of mouse embryonic fibroblasts The synthetic Oct6 molecule contributes to epigenetic.
Volume 13, Issue 3, Pages (September 2013)
Volume 7, Issue 1, Pages (July 2010)
Kiran Batta, Magdalena Florkowska, Valerie Kouskoff, Georges Lacaud 
Volume 3, Issue 3, Pages (September 2008)
Jakub Tolar, Lily Xia, Megan J. Riddle, Chris J. Lees, Cindy R
Volume 7, Issue 5, Pages (November 2010)
Volume 4, Issue 1, Pages (January 2009)
Volume 8, Issue 5, Pages (May 2017)
Short Telomeres in ESCs Lead to Unstable Differentiation
Dynamic transcriptional and epigenomic reprogramming from pediatric nasal epithelial cells to induced pluripotent stem cells  Hong Ji, PhD, Xue Zhang,
Volume 7, Issue 2, Pages (August 2016)
Volume 2, Issue 2, Pages (February 2014)
Volume 7, Issue 6, Pages (December 2010)
Derivation of Mouse Haploid Trophoblast Stem Cells
Volume 7, Issue 3, Pages (September 2010)
Safeguarding Nonhuman Primate iPS Cells With Suicide Genes
Periodic Activation of Wnt/β-Catenin Signaling Enhances Somatic Cell Reprogramming Mediated by Cell Fusion  Frederic Lluis, Elisa Pedone, Stefano Pepe,
Presentation transcript:

Reprogramming of T Cells from Human Peripheral Blood Yuin-Han Loh, Odelya Hartung, Hu Li, Chunguang Guo, Julie M. Sahalie, Philip D. Manos, Achia Urbach, Garrett C. Heffner, Marica Grskovic, Francois Vigneault, M. William Lensch, In-Hyun Park, Suneet Agarwal, George M. Church, James J. Collins, Stefan Irion, George Q. Daley  Cell Stem Cell  Volume 7, Issue 1, Pages 15-19 (July 2010) DOI: 10.1016/j.stem.2010.06.004 Copyright © 2010 Elsevier Inc. Terms and Conditions

Figure 1 Reprogramming of Peripheral Blood Cells to Pluripotent iPSCs (A) Scheme for reprogramming human peripheral blood (PB) mononuclear cells (PBMCs) and CD34+ cells (PB CD34+). Morphology of the typical peripheral blood cells and images of hESC-like iPSC colonies are shown. (B) Images of PB34 iPSC colonies. Bright-field images were acquired with a standard microscope (Nikon, Japan) with a 10× objective. Immunohistochemistry of PB-derived iPSC colonies expressing markers for OCT4, NANOG, Tra-1-60, and alkaline phosphatase (AP). Hoechst staining indicates the total cell content per field. Fibroblasts surrounding human iPSC colonies serve as internal negative controls for immunohistochemistry staining. Images were acquired with a standard microscope (Nikon, Japan) with a 10× objective. (C) Images of PBMC (Donor GH) iPSC colonies. Bright-field images were acquired with a standard microscope (Nikon, Japan) with a 10× objective. Immunohistochemistry of PB-derived iPSC colonies expressing markers for OCT4, NANOG, Tra-1-60, and alkaline phosphatase (AP). Hoechst staining indicates the total cell content per field. (D) Images of PBMC (donors 34, 50, 76) iPSC colonies. Bright-field images were acquired with a standard microscope (Nikon, Japan) with a 10× objective. Immunohistochemistry of PB-derived iPSC colonies expressing marker for NANOG. DAPI staining indicates the total cell content per field. (E) Quantitative reverse transcription-PCR analyses for the expression of ESC-marker genes NANOG, hTERT, GDF3, and REX1 in PB CD34+ and PBMC-derived iPSCs and human H1 ESCs (with their respective standard errors). Individual PCR reactions were normalized against β-ACTIN and plotted (Log10 scale) relative to the expression level in the H1 ESCs, which was set to 1. (F) Scatter plots comparing PB34 iPSCs and PBMC iPSCs global gene expression profiles to parental (left) and H1 human ESCs (right). The black lines indicate the linear equivalent and 2-fold changes in gene expression levels between the paired cell types. Positions of pluripotency genes OCT4, SOX2, NANOG, and LIN28 in scatter plots are indicated. (G) Bisulfite genomic sequencing of the NANOG promoters reveals demethylation in the iPSC lines. Each horizontal row of circles represents an individual sequencing reaction for a given amplicon. Open and filled circles represent unmethylated and methylated CpGs dinucleotides, respectively. Percentage of methylation is indicated for each cell line. For further characterization of the peripheral blood-derived iPSC clones, see also Figure S1 and Table S1. Cell Stem Cell 2010 7, 15-19DOI: (10.1016/j.stem.2010.06.004) Copyright © 2010 Elsevier Inc. Terms and Conditions

Figure 2 Pluripotency and V(D)J Rearrangement of Peripheral Blood-Derived iPSCs (A) Embryoid bodies derived from PB34 and PBMC iPSCs yield hematopoetic colonies in semisolid methylcellulose media: burst forming unit-erythroid (BFU-E), colony forming unit-granulocyte (CFU-G), colony forming unit-macrophage (CFU-M), colony forming unit-granulocyte, macrophage (CFU-GM), and colony forming unit-granulocyte, erythroid, macrophage (CFU-GEMM). Total number of each type of colony was counted. (B) Representative images of various types of hematopoietic colonies. Images were acquired with a standard microscope (Nikon, Japan) with a 20× objective. (C and D) Hematoxylin and eosin staining of teratomas derived from immunodeficient mice injected with PB34 iPSCs (C) and PBMC iPSCs (D) show tissues representing all three embryonic germ layers. (E) Genomic DNA from peripheral blood-derived iPSC lines grown was digested with NcoI and analyzed for V(D)J rearrangements at the TCR-δ (T cell receptor Delta) locus by Southern blotting with a 3′Jδ3 probe. (F) TCR-δ V(D)J recombination of blood-derived iPSC clones. Lanes 2–8 and lanes 13–17 are PBMC iPSC lines. B cell lines on lanes 8 and 9 showed no rearrangement. TCR-δ rearrangement was observed for some PBMC-derived iPSC lines (lanes 2–6 and 8). Lanes 1, 11, 12, 18, and 19 are H1 hESCs, PB34 iPSCs, fibroblast cells, fibroblast-derived iPSCs via retrovirus and lentivirus, respectively. The red arrow indicates expected size of the germline band. Orange arrow indicates rearranged bands. For further information on the pluripotency, V(D)J rearrangement, and fingerprint analysis performed on the peripheral blood-derived iPSC clones, see also Figure S2 and Table S2. Cell Stem Cell 2010 7, 15-19DOI: (10.1016/j.stem.2010.06.004) Copyright © 2010 Elsevier Inc. Terms and Conditions