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.

  • Brief Communication
  • Published:

Lentiviral gene therapy and vitamin B3 treatment enable granulocytic differentiation of G6PC3-deficient induced pluripotent stem cells

Abstract

Induced pluripotent stem cells (iPSCs) from patients with genetic disorders are a valuable source for in vitro disease models, which enable drug testing and validation of gene and cell therapies. We generated iPSCs from a severe congenital neutropenia (SCN) patient, who presented with a nonsense mutation in the glucose-6-phosphatase catalytic subunit 3 (G6PC3) gene causing profound defects in granulopoiesis, associated with increased susceptibility of neutrophils to apoptosis. Generated SCN iPSC clones exhibited the capacity to differentiate into hematopoietic cells of the myeloid lineage and we identified two cytokine conditions, i.e., using granulocyte-colony stimulating factor or granulocyte-macrophage colony stimulating factor in combination with interleukin-3, to model the SCN phenotype in vitro. Reduced numbers of granulocytes were produced by SCN iPSCs compared with control iPSCs in both settings, which reflected the phenotype in patients. Interestingly, our model showed increased monocyte/macrophage production from the SCN iPSCs. Most importantly, lentiviral genetic correction of SCN iPSCs with a codon-optimized G6PC3 transgene restored granulopoiesis and reduced apoptosis of in vitro differentiated myeloid cells. Moreover, addition of vitamin B3 clearly induced granulocytic differentiation of SCN iPSCs and increased the number of neutrophils to levels comparable with those obtained from healthy control iPSCs. In summary, we established an iPSC-derived in vitro disease model, which will serve as a tool to test the potency of alternative treatment options for SCN patients, such as small molecules and gene therapeutic vectors.

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

Access options

Buy this article

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

Fig. 1: Defective granulocyte production from G6PC3-deficient iPSCs under different differentiation conditions.
Fig. 2: Lentiviral vector mediated gene transfer reconstituted granulocytic differentiation in G6PC3-deficient iPSCs.
Fig. 3: Stimulation of G6PC3-deficient myeloid differentiation cultures with vitamin B3 increases the production of granulocytes.

Similar content being viewed by others

Data availability

Datasets generated and analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Klein C. Genetic defects in severe congenital neutropenia: emerging insights into life and death of human neutrophil granulocytes. Annu Rev Immunol. 2011;29:399–413.

    Article  CAS  Google Scholar 

  2. Skokowa J, Dale DC, Touw IP, Zeidler C, Welte K. Severe congenital neutropenias. Nat Rev Dis Primer. 2017;3:17032.

    Article  Google Scholar 

  3. Freedman MH, Bonilla MA, Fier C, Bolyard AA, Scarlata D, Boxer LA, et al. Myelodysplasia syndrome and acute myeloid leukemia in patients with congenital neutropenia receiving G-CSF therapy. Blood. 2000;96:429–36.

    CAS  PubMed  Google Scholar 

  4. Rosenberg PS, Zeidler C, Bolyard AA, Alter BP, Bonilla MA, Boxer LA, et al. Stable long-term risk of leukaemia in patients with severe congenital neutropenia maintained on G-CSF therapy. Br J Haematol. 2010;150:196–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. De Ravin SS, Wu X, Moir S, Kardava L, Anaya-O’Brien S, Kwatemaa N, et al. Lentiviral hematopoietic stem cell gene therapy for X-linked severe combined immunodeficiency. Sci Transl Med. 2016;8:335ra57.

    Article  Google Scholar 

  6. Hacein-Bey-Abina S, Pai S-Y, Gaspar HB, Armant M, Berry CC, Blanche S, et al. A modified γ-retrovirus vector for X-linked severe combined immunodeficiency. N Engl J Med. 2014;371:1407–17.

    Article  Google Scholar 

  7. Aiuti A, Biasco L, Scaramuzza S, Ferrua F, Cicalese MP, Baricordi C, et al. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskot–Aldrich syndrome. Science. 2013;341:1233151.

    Article  Google Scholar 

  8. Braun CJ, Boztug K, Paruzynski A, Witzel M, Schwarzer A, Rothe M, et al. Gene therapy for Wiskott-Aldrich syndrome—long-term efficacy and genotoxicity. Sci Transl Med. 2014;6:227ra33.

    Article  Google Scholar 

  9. Nanua S, Murakami M, Xia J, Grenda DS, Woloszynek J, Strand M, et al. Activation of the unfolded protein response is associated with impaired granulopoiesis in transgenic mice expressing mutant Elane. Blood. 2011;117:3539–47.

    Article  CAS  Google Scholar 

  10. Cheung YY, Kim SY, Yiu WH, Pan C-J, Jun H-S, Ruef RA, et al. Impaired neutrophil activity and increased susceptibility to bacterial infection in mice lacking glucose-6-phosphatase–β. J Clin Investig. 2007;117:784–93.

    Article  CAS  Google Scholar 

  11. Nayak RC, Trump LR, Aronow BJ, Myers K, Mehta P, Kalfa T, et al. Pathogenesis of ELANE-mutant severe neutropenia revealed by induced pluripotent stem cells. J Clin Investig. 2015;125:3103–16.

    Article  Google Scholar 

  12. Hiramoto T, Ebihara Y, Mizoguchi Y, Nakamura K, Yamaguchi K, Ueno K, et al. Wnt3a stimulates maturation of impaired neutrophils developed from severe congenital neutropenia patient-derived pluripotent stem cells. Proc Natl Acad Sci USA. 2013;110:3023–8.

    Article  CAS  Google Scholar 

  13. Dannenmann B, Zahabi A, Mir P, Oswald B, Bernhard R, Klimiankou M, et al. Human iPSC-based model of severe congenital neutropenia reveals elevated UPR and DNA damage in CD34+ cells preceding leukemic transformation. Exp Hematol. 2019;71:51–60.

    Article  CAS  Google Scholar 

  14. Morishima T, Watanabe K-i, Niwa A, Hirai H, Saida S, Tanaka T, et al. Genetic correction of HAX1 in induced pluripotent stem cells from a patient with severe congenital neutropenia improves defective granulopoiesis. Haematologica. 2014;99:19–27.

    Article  CAS  Google Scholar 

  15. Pittermann E, Lachmann N, Maclean G, Emmrich S, Ackermann M, Ohring G, et al. Gene correction of HAX1 reversed Kostmann disease phenotype in patient-specific induced pluripotent stem cells. Blood Adv. 2017;1:903–14.

    Article  CAS  Google Scholar 

  16. Satoh D, Maeda T, Ito T, Nakajima Y, Ohte M, Ukai A, et al. Establishment and directed differentiation of induced pluripotent stem cells from glycogen storage disease type Ib patient. Genes Cells. 2013;18:1053–69.

    Article  CAS  Google Scholar 

  17. Banka S, Newman WG. A clinical and molecular review of ubiquitous glucose-6-phosphatase deficiency caused by G6PC3 mutations. Orphanet J Rare Dis. 2013;8:1.

    Article  Google Scholar 

  18. Boztug K, Appaswamy G, Ashikov A, Schäffer AA, Salzer U, Diestelhorst J, et al. A syndrome with congenital neutropenia and mutations in G6PC3. N Engl J Med. 2009;360:32–43.

    Article  CAS  Google Scholar 

  19. Hoffmann D, Schott JW, Geis FK, Lange L, Müller F-J, Lenz D, et al. Detailed comparison of retroviral vectors and promoter configurations for stable and high transgene expression in human induced pluripotent stem cells. Gene Ther. 2017;24:298–307.

    Article  CAS  Google Scholar 

  20. Dull T, Zufferey R, Kelly M, Mandel RJ, Nguyen M, Trono D, et al. A third-generation lentivirus vector with a conditional packaging system. J Virol. 1998;72:8463–71.

    Article  CAS  Google Scholar 

  21. Ai HW, Shaner NC, Cheng Z, Tsien RY, Campbell RE. Exploration of new chromophore structures leads to the identification of improved blue fluorescent proteins. Biochemistry. 2007;46:5904–10.

    Article  CAS  Google Scholar 

  22. Lachmann N, Ackermann M, Frenzel E, Liebhaber S, Brennig S, Happle C, et al. Large-scale hematopoietic differentiation of human induced pluripotent stem cells provides granulocytes or macrophages for cell replacement therapies. Stem Cell Rep. 2015;4:282–96.

    Article  CAS  Google Scholar 

  23. Warlich E, Kuehle J, Cantz T, Brugman MH, Maetzig T, Galla M, et al. Lentiviral vector design and imaging approaches to visualize the early stages of cellular reprogramming. Mol Ther. 2011;19:782–9.

    Article  CAS  Google Scholar 

  24. Voelkel C, Galla M, Maetzig T, Warlich E, Kuehle J, Zychlinski D, et al. Protein transduction from retroviral Gag precursors. Proc Natl Acad Sci USA. 2010;107:7805–10.

    Article  CAS  Google Scholar 

  25. Rothe M, Rittelmeyer I, Iken M, Rüdrich U, Schambach A, Glage S, et al. Epidermal growth factor improves lentivirus vector gene transfer into primary mouse hepatocytes. Gene Ther. 2012;19:425–34.

    Article  CAS  Google Scholar 

  26. Naldini L. Gene therapy returns to centre stage. Nature. 2015;526:351–60.

    Article  CAS  Google Scholar 

  27. Müller-Kuller U, Ackermann M, Kolodziej S, Brendel C, Fritsch J, Lachmann N, et al. A minimal ubiquitous chromatin opening element (UCOE) effectively prevents silencing of juxtaposed heterologous promoters by epigenetic remodeling in multipotent and pluripotent stem cells. Nucleic Acids Res. 2015;43:1577–92.

    Article  Google Scholar 

  28. Donadieu J, Fenneteau O, Beaupain B, Mahlaoui N, Chantelot C. Congenital neutropenia: diagnosis, molecular bases and patient management. Orphanet J Rare Dis. 2011;6:26.

    Article  Google Scholar 

  29. Koch C, Samareh B, Morishima T, Mir P, Kanz L, Zeidler C, et al. GM-CSF treatment is not effective in congenital neutropenia patients due to its inability to activate NAMPT signaling. Ann Hematol. 2017;96:345–53.

    Article  CAS  Google Scholar 

  30. Skokowa J, Lan D, Thakur BK, Wang F, Gupta K, Cario G, et al. NAMPT is essential for the G-CSF-induced myeloid differentiation via a NAD+-sirtuin-1-dependent pathway. Nat Med. 2009;15:151–8.

    Article  CAS  Google Scholar 

  31. Notarangelo LD, Savoldi G, Cavagnini S, Bennato V, Vasile S, Pilotta A, et al. Severe congenital neutropenia due to G6PC3 deficiency: early and delayed phenotype in two patients with two novel mutations. Ital J Pediatr. 2014;40:80.

    Article  Google Scholar 

  32. Jun HS, Cheung YY, Lee YM, Mansfield BC, Chou JY. Glucose-6-phosphatase-β, implicated in a congenital neutropenia syndrome, is essential for macrophage energy homeostasis and functionality. Blood. 2012;119:4047–55.

    Article  CAS  Google Scholar 

  33. Mistry A, Scambler T, Parry D, Wood M, Barcenas-Morales G, Carter C, et al. Glucose-6-phosphatase catalytic subunit 3 (G6PC3) deficiency associated with autoinflammatory complications. Front Immunol. 2017;8:1485.

    Article  Google Scholar 

  34. Boztug K, Rosenberg PS, Dorda M, Banka S, Moulton T, Curtin J, et al. Extended spectrum of human glucose-6-phosphatase catalytic subunit 3 deficiency: novel genotypes and phenotypic variability in severe congenital neutropenia. J Pediatr. 2012;160:679.

    Article  CAS  Google Scholar 

  35. Ackermann M, Kempf H, Hetzel M, Hesse C, Hashtchin AR, Brinkert K, et al. Bioreactor-based mass production of human iPSC-derived macrophages enables immunotherapies against bacterial airway infections. Nat Commun. 2018;9:5088.

    Article  Google Scholar 

  36. Cugno C, Deola S, Filippini P, Stroncek DF, Rutella S. Granulocyte transfusions in children and adults with hematological malignancies: benefits and controversies. J Transl Med. 2015;13:362.

    Article  Google Scholar 

  37. Trump LR, Nayak RC, Singh AK, Emberesh S, Wellendorf AM, Lutzko CM, et al. Neutrophils derived from genetically modified human induced pluripotent stem cells circulate and phagocytose bacteria in vivo. Stem Cells Transl Med. 2019;8:557–67.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the Bundesministerium für Bildung und Forschung (PID-NET FK2016M1517F; MyPred), the Deutsche Forschungsgemeinschaft (Cluster of Excellence REBIRTH (EXC 62/2) and SFB738), and the Deutsche Akademische Austauschdienst (DAAD). We thank Gerald Draeger and Thomas Scheper for providing Rock inhibitor Y-27632 and bFGF (Leibniz University Hannover, Hannover, Germany) and Malte Sgodda and Tobias Cantz for H9 ESC RNA (Hannover Medical School, Hannover, Germany), and Robert E. Campbell (University of Alberta, Alberta, Canada) for providing EBFP2.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Axel Schambach.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hoffmann, D., Kuehle, J., Lenz, D. et al. Lentiviral gene therapy and vitamin B3 treatment enable granulocytic differentiation of G6PC3-deficient induced pluripotent stem cells. Gene Ther 27, 297–306 (2020). https://doi.org/10.1038/s41434-020-0127-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41434-020-0127-y

This article is cited by

Search

Quick links