Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Replacing reprogramming factors with antibodies selected from combinatorial antibody libraries

Abstract

The reprogramming of differentiated cells into induced pluripotent stem cells (iPSCs) is usually achieved by exogenous induction of transcription by factors acting in the nucleus. In contrast, during development, signaling pathways initiated at the membrane induce differentiation. The central idea of this study is to identify antibodies that can catalyze cellular de-differentiation and nuclear reprogramming by acting at the cell surface. We screen a lentiviral library encoding 100 million secreted and membrane-bound single-chain antibodies and identify antibodies that can replace either Sox2 and Myc (c-Myc) or Oct4 during reprogramming of mouse embryonic fibroblasts into iPSCs. We show that one Sox2-replacing antibody antagonizes the membrane-associated protein Basp1, thereby de-repressing nuclear factors WT1, Esrrb and Lin28a (Lin28) independent of Sox2. By manipulating this pathway, we identify three methods to generate iPSCs. Our results establish unbiased selection from autocrine combinatorial antibody libraries as a robust method to discover new biologics and uncover membrane-to-nucleus signaling pathways that regulate pluripotency and cell fate.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Autocrine selection from combinatorial libraries identifies antibodies that replace Sox2 and c-Myc in generating iPSCs.
Figure 2: A two-step selection and antibody enrichment method identifies antibodies that can replace Oct4 during reprogramming of MEFs into iPSCs.
Figure 3: Antibodies enable reprogramming to pluripotency without Sox2 and Oct4.
Figure 4: SoxAb2 antagonizes Basp1.
Figure 5: Basp1 repression promotes reprogramming via Wt1-mediated upregulation of Lin28 and Esrrb.

Similar content being viewed by others

References

  1. Boland, M.J. et al. Adult mice generated from induced pluripotent stem cells. Nature 461, 91–94 (2009).

    CAS  PubMed  Google Scholar 

  2. Kang, L., Wang, J., Zhang, Y., Kou, Z. & Gao, S. iPS cells can support full-term development of tetraploid blastocyst-complemented embryos. Cell Stem Cell 5, 135–138 (2009).

    CAS  PubMed  Google Scholar 

  3. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    CAS  PubMed  Google Scholar 

  4. Zhao, X.Y. et al. iPS cells produce viable mice through tetraploid complementation. Nature 461, 86–90 (2009).

    CAS  PubMed  Google Scholar 

  5. Ieda, M. et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142, 375–386 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jopling, C., Boue, S. & Izpisua Belmonte, J.C. Dedifferentiation, transdifferentiation and reprogramming: three routes to regeneration. Nat. Rev. Mol. Cell Biol. 12, 79–89 (2011).

    CAS  PubMed  Google Scholar 

  7. Tsunemoto, R.K., Eade, K.T., Blanchard, J.W. & Baldwin, K.K. Forward engineering neuronal diversity using direct reprogramming. EMBO J. 34, 1445–1455 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Vierbuchen, T. et al. Direct conversion of fibroblasts to functional neurons by defined factors. Nature 463, 1035–1041 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Arkin, M.R., Tang, Y. & Wells, J.A. Small-molecule inhibitors of protein-protein interactions: progressing toward the reality. Chem. Biol. 21, 1102–1114 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Laraia, L., McKenzie, G., Spring, D.R., Venkitaraman, A.R. & Huggins, D.J. Overcoming chemical, biological, and computational challenges in the development of inhibitors targeting protein-protein interactions. Chem. Biol. 22, 689–703 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Carey, B.W. et al. Reprogramming factor stoichiometry influences the epigenetic state and biological properties of induced pluripotent stem cells. Cell Stem Cell 9, 588–598 (2011).

    CAS  PubMed  Google Scholar 

  12. Hanna, J. et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature 462, 595–601 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Stadtfeld, M., Maherali, N., Borkent, M. & Hochedlinger, K. A reprogrammable mouse strain from gene-targeted embryonic stem cells. Nat. Methods 7, 53–55 (2010).

    CAS  PubMed  Google Scholar 

  14. Ichida, J.K. et al. A small-molecule inhibitor of TGF-β signaling replaces sox2 in reprogramming by inducing NANOG. Cell Stem Cell 5, 491–503 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Lyssiotis, C.A. et al. Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of Klf4. Proc. Natl. Acad. Sci. USA 106, 8912–8917 (2009).

    PubMed  PubMed Central  Google Scholar 

  16. Staerk, J. et al. Pan-Src family kinase inhibitors replace Sox2 during the direct reprogramming of somatic cells. Angew. Chem. Int. Ed. 50, 5734–5736 (2011).

    CAS  Google Scholar 

  17. Qin, H. et al. Systematic identification of barriers to human iPSC generation. Cell 158, 449–461 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Borkent, M. et al. A serial shRNA screen for roadblocks to reprogramming identifies the protein modifier SUMO2. Stem Cell Rep. 6, 704–716 (2016).

    CAS  Google Scholar 

  19. Melidoni, A.N., Dyson, M.R., Wormald, S. & McCafferty, J. Selecting antagonistic antibodies that control differentiation through inducible expression in embryonic stem cells. Proc. Natl. Acad. Sci. USA 110, 17802–17807 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Xie, J., Zhang, H., Yea, K. & Lerner, R.A. Autocrine signaling based selection of combinatorial antibodies that transdifferentiate human stem cells. Proc. Natl. Acad. Sci. USA 110, 8099–8104 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhang, H., Wilson, I.A. & Lerner, R.A. Selection of antibodies that regulate phenotype from intracellular combinatorial antibody libraries. Proc. Natl. Acad. Sci. USA 109, 15728–15733 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhang, H. et al. Selecting agonists from single cells infected with combinatorial antibody libraries. Chem. Biol. 20, 734–741 (2013).

    CAS  PubMed  Google Scholar 

  23. Zhang, H., Xie, J. & Lerner, R.A. A proximity based general method for identification of ligand and receptor interactions in living cells. Biochem. Biophys. Res. Commun. 454, 251–255 (2014).

    CAS  PubMed  Google Scholar 

  24. Liu, T. et al. Construction and screening of a lentiviral secretome library. Cell Chem. Biol. 24, 767–771.e3 (2017).

    CAS  PubMed  Google Scholar 

  25. Stadtfeld, M., Maherali, N., Breault, D.T. & Hochedlinger, K. Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. Cell Stem Cell 2, 230–240 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Hou, P. et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science 341, 651–654 (2013).

    CAS  PubMed  Google Scholar 

  27. Radzisheuskaya, A. & Silva, J.C. Do all roads lead to Oct4? the emerging concepts of induced pluripotency. Trends Cell Biol. 24, 275–284 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Hazen, J.L. et al. The complete genome sequences, unique mutational spectra, and developmental potency of adult neurons revealed by cloning. Neuron 89, 1223–1236 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Maherali, N. & Hochedlinger, K. Tgfbeta signal inhibition cooperates in the induction of iPSCs and replaces Sox2 and cMyc. Curr. Biol. 19, 1718–1723 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Shu, J. et al. Induction of pluripotency in mouse somatic cells with lineage specifiers. Cell 153, 963–975 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Korshunova, I. et al. Characterization of BASP1-mediated neurite outgrowth. J. Neurosci. Res. 86, 2201–2213 (2008).

    CAS  PubMed  Google Scholar 

  32. Maekawa, S., Maekawa, M., Hattori, S. & Nakamura, S. Purification and molecular cloning of a novel acidic calmodulin binding protein from rat brain. J. Biol. Chem. 268, 13703–13709 (1993).

    CAS  PubMed  Google Scholar 

  33. Mosevitsky, M.I. et al. The BASP1 family of myristoylated proteins abundant in axonal termini. Primary structure analysis and physico-chemical properties. Biochimie 79, 373–384 (1997).

    CAS  PubMed  Google Scholar 

  34. Hu, X. et al. Tet and TDG mediate DNA demethylation essential for mesenchymal-to-epithelial transition in somatic cell reprogramming. Cell Stem Cell 14, 512–522 (2014).

    CAS  PubMed  Google Scholar 

  35. Li, R. et al. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell 7, 51–63 (2010).

    CAS  PubMed  Google Scholar 

  36. Carpenter, B. et al. BASP1 is a transcriptional cosuppressor for the Wilms' tumor suppressor protein WT1. Mol. Cell. Biol. 24, 537–549 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Toska, E. et al. Repression of transcription by WT1-BASP1 requires the myristoylation of BASP1 and the PIP2-dependent recruitment of histone deacetylase. Cell Rep. 2, 462–469 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Huang, S. Reprogramming cell fates: reconciling rarity with robustness. BioEssays 31, 546–560 (2009).

    CAS  PubMed  Google Scholar 

  39. Buganim, Y. et al. Single-cell expression analyses during cellular reprogramming reveal an early stochastic and a late hierarchic phase. Cell 150, 1209–1222 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Buganim, Y. et al. The developmental potential of iPSCs is greatly influenced by reprogramming factor selection. Cell Stem Cell 15, 295–309 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Feng, B. et al. Reprogramming of fibroblasts into induced pluripotent stem cells with orphan nuclear receptor Esrrb. Nat. Cell Biol. 11, 197–203 (2009).

    CAS  PubMed  Google Scholar 

  42. Motamedi, F.J. et al. WT1 controls antagonistic FGF and BMP-pSMAD pathways in early renal progenitors. Nat. Commun. 5, 4444 (2014).

    CAS  PubMed  Google Scholar 

  43. Melidoni, A.N., Dyson, M.R. & McCafferty, J. Selection of antibodies interfering with cell surface receptor signaling using embryonic stem cell differentiation. Methods Mol. Biol. 1341, 111–132 (2015).

    Google Scholar 

  44. Schlaeger, T.M. et al. A comparison of non-integrating reprogramming methods. Nat. Biotechnol. 33, 58–63 (2015).

    CAS  PubMed  Google Scholar 

  45. Imai, K. & Takaoka, A. Comparing antibody and small-molecule therapies for cancer. Nat. Rev. Cancer 6, 714–727 (2006).

    CAS  PubMed  Google Scholar 

  46. Blanchard, J.W. et al. Selective conversion of fibroblasts into peripheral sensory neurons. Nat. Neurosci. 18, 25–35 (2015).

    CAS  PubMed  Google Scholar 

  47. Bolger, A.M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  49. Anders, S., Pyl, P.T. & Huber, W. HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    CAS  PubMed  Google Scholar 

  50. Love, M.I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank J. Gottesfeld, A. Patapoutian, V. Lo Sardo and J. Hazen for assistance and/or helpful discussions; W. Ferguson for technical assistance and K. Spencer for assistance with microscopy. This research was supported by the National Institute on Deafness and other Communication Disorders (DC012592 to K.K.B.), the National Institute of Mental Health (MH102698 to K.K.B.), the California Institute for Regenerative Medicine (RB3-02186 to K.K.B.), the Baxter Family, Norris and Del Webb Foundations (K.K.B.), Las Patronas and the Dorris Neuroscience Center (K.K.B.), a predoctoral fellowship from the California Institute of Regenerative Medicine (J.W.B.) the Andrea Elizabeth Vogt Memorial Award (J.W.B.).

Author information

Authors and Affiliations

Authors

Contributions

Experiments were initially conceived by R.A.L. and K.K.B. Subsequent experimental design was contributed by all authors, and experiments were designed, performed and analyzed by J.W.B. and J.X. Experiments were also performed by S.L., S.G. and N.E.-M. The manuscript was written and revised by J.W.B., J.X. and K.K.B.

Corresponding authors

Correspondence to Jia Xie or Kristin K Baldwin.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Schematic and results of screening the combinatorial antibody library for Sox2 replacement activity.

A single batch of MEFs were plated and transduced with doxycycline inducible lentiviruses and an rTTA virus. Viral expression was induced for 14 days and images were collected. All MEFs were transduced with viruses encoding Oct4, Klf4 and a previously validated shRNA against p53 (shp53). Additional conditions included the positive control (left panel, with viruses encoding Sox2 and c-Myc), the test for the antibody library (middle panel, since we observed only a few colonies per plate, representative cut outs of colonies are shown) and a negative control with a lentivirus expressing tdTomato (red). Scale bars = 100 μm. Individual colonies arising in the screen were photographed at the same magnification as the other panels but cropped and enlarged variously for visual clarity.

Supplementary Figure 2 Validation of SoxAb- and OctAb-generated iPSCs.

Fig. S2 a and b) Antibody gene sequences were recovered by PCR performed on genomic DNA from iPSC lines. Primer binding sites are depicted in Fig. S1b as “recovery primers”. The amplicons were purified by gel extraction and sub-cloned into lentiviral or other expression vectors and submitted for sequencing. c) Primary colonies induced by Sox2 antibodies were capable of expanding into cell lines maintained under standard mouse ES conditions for at least ten passages. d) Primary colonies induced by Oct4 antibodies can be expanded into cell lines and maintained under standard mouse ES cell conditions for at least ten passages. e) iPSC lines generated with SoxAb2 do not harbor transgenic Sox2 or cMyc. f) iPSC lines generated with OctAb1 and 2 do not harbor transgenic Oct4, but do harbor transgenic human IgG. g) iPSC lines generated with SoxAb2 and OctAb1 express pluripotency markers SSEA1, Sox2, Oct3/4, and Nanog. Scale bars = 100 μm. h) iPS cell lines generated with OctAb2 and SoxAb2 differentiate in vitro to the three embryonic germ layers. Scale bars = 100 μm. i) iPSC lines generated with SoxAb1 differentiate in vivo and contribute to chimeric offspring. Eight-week old chimeric mice formed by injection of KO + SoxAb1 cells (CD1 genetic background) into a C57BL6 blastocyst. j) Chimeric pups display contribution of antibody iPSCs across three embryonic germ layers. k) Quantitative RT-PCR confirmation of the expression of pluripotency genes in the SoxAb1 and SoxAb2 iPSC lines, fully pluripotent iPSCs (iMZ-9 and iiMZ-21) and mESCs (V6.5) compared to control fibroblasts expressing OKM for twelve days under identical culture conditions used to derive the iPSCs. Bars represent the mean of two biological replicates, meaning independent wells of the cell line indicated. Error bars = SEM. l) RNA-seq scatter plots show that global gene expression of antibody-derived iPSC lines is highly similar to TEC competent iPSC (iMZ21) and mESC (MCNT) lines but different from somatic MEFs. Gene expression is plotted as vsd-normalized counts. m) Summary of differential expression analysis (using DESeq2) showing the number of genes that are significantly upregulated (red) and downregulated (blue), that exhibit a greater than 2-fold change in expression in MEFs, and antibody-iPSC lines compared to TEC-competent iPSCs and ESCs.

Supplementary Figure 3 Mechanisms governing reprogramming with SoxAb2 involve Basp1 inhibition.

a) SoxAb2 does not inhibit Tgf-β induced phosphorylation of Smad2/3 whereas small molecule inhibitors RepSox and Tgf-β neutralizing antibodies previously reported to promote Sox2-independent reprogramming do inhibit Tgf-β induced phosphorylation of Smad2/3. For each treatment (SoxAb2, RepSox, SB431542, and Tgf-β neutralizing antibodies) ability to inhibit Smad2/3 phosphorylation was quantified from three independent western blots. The graph depicts the mean from the three blots. Error bars represent SEM. Significance was determined by ANOVA followed by Newman-Keuls post-hoc analysis, *p<0.05, ** p<0.01. b) SoxAb2 does not directly inhibit Src-kinase activity. SoxAb2 ability to inhibit Src-kinase activity was compared to the small molecule PP1 a known Src-kinase inhibitor able to promote Sox2-independent reprogramming. c) SoxAb2 does not antagonize the induction of mesoderm genes. Mesoderm genes Eomes, T, and Ctnnb1 were quantified in three biological replicates of MEFs expressing Klf4 and Oct4 by RT-PCR following 96 hours expression of SoxAb2. Significance was assessed by ANOVA; no significant differences were observed. d) Proteins were immunoprecipitated with SoxAb2 separated by SDS-PAGE and stained with Coomassie brilliant blue. A ~25 KDa based was excised from control (middle) and SoxAb2-expressing (right) lanes and analyzed by trypsin digestion and mass spectrometry which identified candidate proteins enriched in each sample. e) SoxAb2 immunoprecipitates endogenous Basp1. MEFs were transduced with Basp1 (lanes 1,2) or not transduced (lane3). Lysates were immunoprecipitated with SoxAb2 (lanes 2 and 3) and Western blots were performed with either Anti-Basp1 (top) or the SoxAb2 (bottom). Basp1 protein was present and detected by both antibodies in the whole lysate from MEFs overexpressing Basp1 and enriched in pull-downs with the SoxAb2, demonstrating that the SoxAb2 recognized Basp1 protein. f) Quantitative RT-PCR confirming Basp1 shRNA significantly reduces Basp1 expression in MEFs; p=.0000084, Students T-Test. g) The changes in gene expression in MEFs directed by SoxAb2 or knockdown of Basp1 are strongly correlated. Heat map and hierarchical clustering of control MEFs compared to MEFs expressing SoxAb2 or Basp1 shRNA. Clustering represents single linkage Euclidian distance. h) Heat map and hierarchical clustering in MEFs expressing Klf4, Oct4, and cMyc (KOM) compared to KOM MEFs expressing SoxAb2 or Basp1 shRNA. Clustering represents single linkage Euclidian distance. i) Primary colonies induced by Basp1 knockdown + KOM can be expanded into cell lines and cultured under standard mES conditions for at least ten passages. j) Basp1 knockdown iPSC lines do not harbor transgenic viral Sox2 as expected but do harbor viral Oct4 and Klf4 genes. k) RNAseq scatter plots showing that global gene expression of Basp1 knockdown iPSC lines is highly divergent from somatic MEFs relative to iPSCs and mESCs. Gene expression is plotted as vsd-normalized counts.

Supplementary Figure 4 Downstream pathways related to Basp1 inhibition can replace Sox2 in generating iPSCs.

Panel a) Wt1 was co-immunoprecipitated with Basp1 in somatic MEFs. b) Primary colonies induced by Wt1 overexpression + KOM can be expanded into cell lines and cultured under standard mES conditions for at least ten passages. c) Wt1 overexpression + KOM iPSC lines appropriately express pluripotency marker SSEA1, Sox2, Oct4, and Nanog. d and e) RNAseq scatter plots showing global gene expression of KOM + Wt1 iPSC lines are highly similar to TEC competent iPSC (iMZ21) and mESC (MCNT) lines relative to their dissimilarity with somatic MEFs. Gene expression is plotted as vsd-normalized counts. f) Wt1 overexpression + KOM iPSC lines differentiate to the three embryonic germ layers in vitro. g) Scatter plots depicting mean changes in gene expression induced by Wt1 overexpression and Basp1 knockdown on day four and twelve of reprogramming. Expression is normalized to KOM only samples. Error bars = SEM. h) iPSC lines generated with Lin28/Esrrb + Klf4 and Oct4 express pluripotency markers SSEA1, Sox2, Oct4, and Nanog. i) Quantitative RT-PCR showing shRNA-mediated knockdown of Wt1 in iPSC line iMZ9 iPSC and v6.5 mESC reduces expression of Esrrb, Lin28, and the downstream target cMyc. Bars represent means from three biological replicates meaning independent wells of cells treated with the shRNA and analyzed separately. Error bars = SEM. Significance was tested by Students t-test for each comparison, * p<0.05, **p<0.01.

Supplementary Figure 5 Evidence that WT1 can bind to candidate Lin28 promoter regions.

WT1 ChIP-qRT-PCR on LIN28A and B genomic regions. Genomic PCR was performed on regions identified to contain WT1 consensus DNA-binding elements (green line) from in silico analysis. Bars represent the mean of three biologic replicates. Statistical significance was assessed by ANOVA, * p<0.05, **p<0.01. b) Expression of LIN28 (A and B), WT1 (C and D), and BASP1 (E and F) in CML patients. In the left panels, each bar represents a single patient. Right panels are averages of all patients for each group, with significance determined by ANOVA, ***p<0.001.

Supplementary Figure 6 Validation of shRNAs used in the study.

The shRNAs used in the study reduce expression of the expected proteins a) WT-1 protein level in MEF was reduced by three different shRNA sequences (Sigma Mission®), while the housekeeping gene actin is unaffected. b) BASP-1 protein level in MEF cells was reduced by two different shRNA sequences (Sigma Mission®) separately and in combination.

Supplementary Figure 7 Full views of gels used for western blotting.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Blanchard, J., Xie, J., El-Mecharrafie, N. et al. Replacing reprogramming factors with antibodies selected from combinatorial antibody libraries. Nat Biotechnol 35, 960–968 (2017). https://doi.org/10.1038/nbt.3963

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3963

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research