#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Gene editing of PKLR gene in human hematopoietic progenitors through 5’ and 3’ UTR modified TALEN mRNA


Autoři: Oscar Quintana-Bustamante aff001;  Sara Fañanas-Baquero aff001;  Israel Orman aff001;  Raul Torres aff003;  Philippe Duchateau aff005;  Laurent Poirot aff005;  Agnès Gouble aff005;  Juan A. Bueren aff001;  Jose C. Segovia aff001
Působiště autorů: Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas/Centro de Investigación Biomédica en Red de Enfermedades Raras (CIEMAT/CIBERER), Madrid, Spain aff001;  Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain aff002;  Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain aff003;  Instituto Josep Carreras, Barcelona, Spain aff004;  CELLECTIS, Paris, France aff005
Vyšlo v časopise: PLoS ONE 14(10)
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pone.0223775

Souhrn

Pyruvate Kinase Deficiency (PKD) is a rare erythroid metabolic disease caused by mutations in the PKLR gene, which encodes the erythroid specific Pyruvate Kinase enzyme. Erythrocytes from PKD patients show an energetic imbalance and are susceptible to hemolysis. Gene editing of hematopoietic stem cells (HSCs) would provide a therapeutic benefit and improve safety of gene therapy approaches to treat PKD patients. In previous studies, we established a gene editing protocol that corrected the PKD phenotype of PKD-iPSC lines through a TALEN mediated homologous recombination strategy. With the goal of moving toward more clinically relevant stem cells, we aim at editing the PKLR gene in primary human hematopoietic progenitors and hematopoietic stem cells (HPSCs). After nucleofection of the gene editing tools and selection with puromycin, up to 96% colony forming units showed precise integration. However, a low yield of gene edited HPSCs was associated to the procedure. To reduce toxicity while increasing efficacy, we worked on i) optimizing gene editing tools and ii) defining optimal expansion and selection times. Different versions of specific nucleases (TALEN and CRISPR-Cas9) were compared. TALEN mRNAs with 5’ and 3’ added motifs to increase RNA stability were the most efficient nucleases to obtain high gene editing frequency and low toxicity. Shortening ex vivo manipulation did not reduce the efficiency of homologous recombination and preserved the hematopoietic progenitor potential of the nucleofected HPSCs. Lastly, a very low level of gene edited HPSCs were detected after engraftment in immunodeficient (NSG) mice. Overall, we showed that gene editing of the PKLR gene in HPSCs is feasible, although further improvements must to be done before the clinical use of the gene editing to correct PKD.

Klíčová slova:

DNA – Genetic loci – Polymerase chain reaction – DNA sequence analysis – Nucleases – Nested polymerase chain reaction – TALENs – Nuclear matrix


Zdroje

1. Beutler E, Gelbart T. Estimating the prevalence of pyruvate kinase deficiency from the gene frequency in the general white population. Blood. 2000;95(11):3585–8. 10828047.

2. Zanella A, Fermo E, Bianchi P, Valentini G. Red cell pyruvate kinase deficiency: molecular and clinical aspects. Br J Haematol. 2005;130(1):11–25. Epub 2005/06/29. doi: 10.1111/j.1365-2141.2005.05527.x 15982340.

3. van Straaten S, Bierings M, Bianchi P, Akiyoshi K, Kanno H, Serra IB, et al. Worldwide study of hematopoietic allogeneic stem cell transplantation in pyruvate kinase deficiency. Haematologica. 2018;103(2):e82–e6. doi: 10.3324/haematol.2017.177857 29242305; PubMed Central PMCID: PMC5792292.

4. Dunbar CE, High KA, Joung JK, Kohn DB, Ozawa K, Sadelain M. Gene therapy comes of age. Science. 2018;359(6372). Epub 2018/01/13. doi: 10.1126/science.aan4672 29326244.

5. Cavazzana-Calvo M, Payen E, Negre O, Wang G, Hehir K, Fusil F, et al. Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature. 2010;467(7313):318–22. Epub 2010/09/17. nature09328 [pii] doi: 10.1038/nature09328 20844535.

6. Thompson AA, Walters MC, Kwiatkowski J, Rasko JEJ, Ribeil JA, Hongeng S, et al. Gene Therapy in Patients with Transfusion-Dependent beta-Thalassemia. N Engl J Med. 2018;378(16):1479–93. doi: 10.1056/NEJMoa1705342 29669226.

7. Ribeil JA, Hacein-Bey-Abina S, Payen E, Magnani A, Semeraro M, Magrin E, et al. Gene Therapy in a Patient with Sickle Cell Disease. N Engl J Med. 2017;376(9):848–55. Epub 2017/03/02. doi: 10.1056/NEJMoa1609677 28249145.

8. Garcia-Gomez M, Calabria A, Garcia-Bravo M, Benedicenti F, Kosinski P, Lopez-Manzaneda S, et al. Safe and Efficient Gene Therapy for Pyruvate Kinase Deficiency. Mol Ther. 2016;24(7):1187–98. doi: 10.1038/mt.2016.87 27138040; PubMed Central PMCID: PMC5088764.

9. Zou J, Sweeney CL, Chou BK, Choi U, Pan J, Wang H, et al. Oxidase-deficient neutrophils from X-linked chronic granulomatous disease iPS cells: functional correction by zinc finger nuclease-mediated safe harbor targeting. Blood. 2011;117(21):5561–72. Epub 2011/03/18. blood-2010-12-328161 [pii] doi: 10.1182/blood-2010-12-328161 21411759.

10. Chang CJ, Bouhassira EE. Zinc-finger nuclease-mediated correction of alpha-thalassemia in iPS cells. Blood. 2012;120(19):3906–14. Epub 2012/09/25. doi: 10.1182/blood-2012-03-420703 23002118; PubMed Central PMCID: PMC3543982.

11. Niu X, He W, Song B, Ou Z, Fan D, Chen Y, et al. Combining Single Strand Oligodeoxynucleotides and CRISPR/Cas9 to Correct Gene Mutations in beta-Thalassemia-induced Pluripotent Stem Cells. J Biol Chem. 2016;291(32):16576–85. doi: 10.1074/jbc.M116.719237 27288406; PubMed Central PMCID: PMC4974373.

12. Xu P, Tong Y, Liu XZ, Wang TT, Cheng L, Wang BY, et al. Both TALENs and CRISPR/Cas9 directly target the HBB IVS2-654 (C > T) mutation in beta-thalassemia-derived iPSCs. Sci Rep. 2015;5:12065. doi: 10.1038/srep12065 26156589; PubMed Central PMCID: PMC4496796.

13. Sebastiano V, Maeder ML, Angstman JF, Haddad B, Khayter C, Yeo DT, et al. In situ genetic correction of the sickle cell anemia mutation in human induced pluripotent stem cells using engineered zinc finger nucleases. Stem Cells. 2011;29(11):1717–26. doi: 10.1002/stem.718 21898685; PubMed Central PMCID: PMC3285772.

14. Huang X, Wang Y, Yan W, Smith C, Ye Z, Wang J, et al. Production of Gene-Corrected Adult Beta Globin Protein in Human Erythrocytes Differentiated from Patient iPSCs After Genome Editing of the Sickle Point Mutation. Stem Cells. 2015;33(5):1470–9. doi: 10.1002/stem.1969 25702619; PubMed Central PMCID: PMC4628786.

15. Garcon L, Ge J, Manjunath SH, Mills JA, Apicella M, Parikh S, et al. Ribosomal and hematopoietic defects in induced pluripotent stem cells derived from Diamond Blackfan anemia patients. Blood. 2013;122(6):912–21. doi: 10.1182/blood-2013-01-478321 23744582; PubMed Central PMCID: PMC3739037.

16. Rio P, Banos R, Lombardo A, Quintana-Bustamante O, Alvarez L, Garate Z, et al. Targeted gene therapy and cell reprogramming in Fanconi anemia. EMBO Mol Med. 2014;6(6):835–48. doi: 10.15252/emmm.201303374 24859981.

17. Garate Z, Quintana-Bustamante O, Crane AM, Olivier E, Poirot L, Galetto R, et al. Generation of a High Number of Healthy Erythroid Cells from Gene-Edited Pyruvate Kinase Deficiency Patient-Specific Induced Pluripotent Stem Cells. Stem Cell Reports. 2015;5(6):1053–66. doi: 10.1016/j.stemcr.2015.10.002 26549847.

18. Genovese P, Schiroli G, Escobar G, Di Tomaso T, Firrito C, Calabria A, et al. Targeted genome editing in human repopulating haematopoietic stem cells. Nature. 2014;510(7504):235–40. doi: 10.1038/nature13420 24870228; PubMed Central PMCID: PMC4082311.

19. Chang CW, Lai YS, Westin E, Khodadadi-Jamayran A, Pawlik KM, Lamb LS Jr., et al. Modeling Human Severe Combined Immunodeficiency and Correction by CRISPR/Cas9-Enhanced Gene Targeting. Cell Rep. 2015;12(10):1668–77. doi: 10.1016/j.celrep.2015.08.013 26321643.

20. De Ravin SS, Li L, Wu X, Choi U, Allen C, Koontz S, et al. CRISPR-Cas9 gene repair of hematopoietic stem cells from patients with X-linked chronic granulomatous disease. Sci Transl Med. 2017;9(372). doi: 10.1126/scitranslmed.aah3480 28077679.

21. Ye L, Wang J, Tan Y, Beyer AI, Xie F, Muench MO, et al. Genome editing using CRISPR-Cas9 to create the HPFH genotype in HSPCs: An approach for treating sickle cell disease and beta-thalassemia. Proc Natl Acad Sci U S A. 2016;113(38):10661–5. doi: 10.1073/pnas.1612075113 27601644; PubMed Central PMCID: PMC5035856.

22. Vierstra J, Reik A, Chang KH, Stehling-Sun S, Zhou Y, Hinkley SJ, et al. Functional footprinting of regulatory DNA. Nature methods. 2015;12(10):927–30. doi: 10.1038/nmeth.3554 26322838.

23. Traxler EA, Yao Y, Wang YD, Woodard KJ, Kurita R, Nakamura Y, et al. A genome-editing strategy to treat beta-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition. Nat Med. 2016;22(9):987–90. doi: 10.1038/nm.4170 27525524; PubMed Central PMCID: PMC5706766.

24. Dever DP, Bak RO, Reinisch A, Camarena J, Washington G, Nicolas CE, et al. CRISPR/Cas9 beta-globin gene targeting in human haematopoietic stem cells. Nature. 2016;539(7629):384–9. doi: 10.1038/nature20134 27820943.

25. Hoban MD, Cost GJ, Mendel MC, Romero Z, Kaufman ML, Joglekar AV, et al. Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells. Blood. 2015;125(17):2597–604. doi: 10.1182/blood-2014-12-615948 25733580; PubMed Central PMCID: PMC4408287.

26. Hoban MD, Lumaquin D, Kuo CY, Romero Z, Long J, Ho M, et al. CRISPR/Cas9-Mediated Correction of the Sickle Mutation in Human CD34+ cells. Mol Ther. 2016;24(9):1561–9. doi: 10.1038/mt.2016.148 27406980; PubMed Central PMCID: PMC5113113.

27. Liu H, Agarwal S, Kmiec E, Davis BR. Targeted beta-globin gene conversion in human hematopoietic CD34(+)and Lin(-)CD38(-)cells. Gene therapy. 2002;9(2):118–26. doi: 10.1038/sj.gt.3301610 11857070.

28. DeWitt MA, Magis W, Bray NL, Wang T, Berman JR, Urbinati F, et al. Selection-free genome editing of the sickle mutation in human adult hematopoietic stem/progenitor cells. Sci Transl Med. 2016;8(360):360ra134. doi: 10.1126/scitranslmed.aaf9336 27733558; PubMed Central PMCID: PMC5500303.

29. Diez B, Genovese P, Roman-Rodriguez FJ, Alvarez L, Schiroli G, Ugalde L, et al. Therapeutic gene editing in CD34(+) hematopoietic progenitors from Fanconi anemia patients. EMBO molecular medicine. 2017;9(11):1574–88. doi: 10.15252/emmm.201707540 28899930; PubMed Central PMCID: PMC5666315.

30. Hyde JL, Gardner CL, Kimura T, White JP, Liu G, Trobaugh DW, et al. A viral RNA structural element alters host recognition of nonself RNA. Science. 2014;343(6172):783–7. doi: 10.1126/science.1248465 24482115; PubMed Central PMCID: PMC4209899.

31. Russell JE, Liebhaber SA. The stability of human beta-globin mRNA is dependent on structural determinants positioned within its 3' untranslated region. Blood. 1996;87(12):5314–23. 8652847.

32. Garate Z, Quintana-Bustamante O, Crane AM, Olivier E, Poirot L, Galetto R, et al. Generation of a High Number of Healthy Erythroid Cells from Gene-Edited Pyruvate Kinase Deficiency Patient-Specific Induced Pluripotent Stem Cells. Stem Cell Rep. 2015;5(6):1053–66. doi: 10.1016/j.stemcr.2015.10.002 WOS:000366142900012. 26549847

33. Cutler C, Multani P, Robbins D, Kim HT, Le T, Hoggatt J, et al. Prostaglandin-modulated umbilical cord blood hematopoietic stem cell transplantation. Blood. 2013;122(17):3074–81. doi: 10.1182/blood-2013-05-503177 23996087; PubMed Central PMCID: PMC3811179.

34. Goessling W, Allen RS, Guan X, Jin P, Uchida N, Dovey M, et al. Prostaglandin E2 enhances human cord blood stem cell xenotransplants and shows long-term safety in preclinical nonhuman primate transplant models. Cell stem cell. 2011;8(4):445–58. doi: 10.1016/j.stem.2011.02.003 21474107; PubMed Central PMCID: PMC3148081.

35. Charrier S, Ferrand M, Zerbato M, Precigout G, Viornery A, Bucher-Laurent S, et al. Quantification of lentiviral vector copy numbers in individual hematopoietic colony-forming cells shows vector dose-dependent effects on the frequency and level of transduction. Gene therapy. 2011;18(5):479–87. doi: 10.1038/gt.2010.163 21160533; PubMed Central PMCID: PMC3130191.

36. Zou J, Mali P, Huang X, Dowey SN, Cheng L. Site-specific gene correction of a point mutation in human iPS cells derived from an adult patient with sickle cell disease. Blood. 2011;118(17):4599–608. doi: 10.1182/blood-2011-02-335554 21881051; PubMed Central PMCID: PMC3208277.

37. Mandai M, Watanabe A, Kurimoto Y, Hirami Y, Morinaga C, Daimon T, et al. Autologous Induced Stem-Cell-Derived Retinal Cells for Macular Degeneration. N Engl J Med. 2017;376(11):1038–46. doi: 10.1056/NEJMoa1608368 28296613.

38. Doulatov S, Vo LT, Chou SS, Kim PG, Arora N, Li H, et al. Induction of multipotential hematopoietic progenitors from human pluripotent stem cells via respecification of lineage-restricted precursors. Cell Stem Cell. 2013;13(4):459–70. doi: 10.1016/j.stem.2013.09.002 24094326; PubMed Central PMCID: PMC3888026.

39. Suzuki N, Yamazaki S, Yamaguchi T, Okabe M, Masaki H, Takaki S, et al. Generation of engraftable hematopoietic stem cells from induced pluripotent stem cells by way of teratoma formation. Mol Ther. 2013;21(7):1424–31. doi: 10.1038/mt.2013.71 23670574; PubMed Central PMCID: PMC3705943.

40. Rahman N, Brauer PM, Ho L, Usenko T, Tewary M, Zuniga-Pflucker JC, et al. Engineering the haemogenic niche mitigates endogenous inhibitory signals and controls pluripotent stem cell-derived blood emergence. Nat Commun. 2017;8:15380. doi: 10.1038/ncomms15380 28541275; PubMed Central PMCID: PMC5477512.

41. Kuo CY, Long JD, Campo-Fernandez B, de Oliveira S, Cooper AR, Romero Z, et al. Site-Specific Gene Editing of Human Hematopoietic Stem Cells for X-Linked Hyper-IgM Syndrome. Cell Rep. 2018;23(9):2606–16. doi: 10.1016/j.celrep.2018.04.103 29847792; PubMed Central PMCID: PMC6181643.

42. Meza NW, Quintana-Bustamante O, Puyet A, Rio P, Navarro S, Diez A, et al. In Vitro and in vivo expression of human erythrocyte pyruvate kinase in erythroid cells: A gene therapy approach. Human Gene Therapy. 2007;18(6):502–14. doi: 10.1089/hum.2006.052 WOS:000247721600003. 17547515

43. Huerfano S, Ryabchenko B, Forstova J. Nucleofection of expression vectors induces a robust interferon response and inhibition of cell proliferation. DNA Cell Biol. 2013;32(8):467–79. doi: 10.1089/dna.2012.1950 23745681; PubMed Central PMCID: PMC3725941.

44. Wu X, Li Y, Crise B, Burgess SM. Transcription start regions in the human genome are favored targets for MLV integration. Science. 2003;300(5626):1749–51. 12805549.

45. Ma Y, Han X, Quintana Bustamante O, Bessa de Castro R, Zhang K, Zhang P, et al. Highly efficient genome editing of human hematopoietic stem cells via a nano-silicon-blade delivery approach. Integr Biol (Camb). 2017;9(6):548–54. doi: 10.1039/c7ib00060j 28513735; PubMed Central PMCID: PMC5598083.

46. Kim JS. Genome editing comes of age. Nature protocols. 2016;11(9):1573–8. doi: 10.1038/nprot.2016.104 27490630.

47. Sander JD, Joung JK. CRISPR-Cas systems for editing, regulating and targeting genomes. Nature biotechnology. 2014;32(4):347–55. doi: 10.1038/nbt.2842 24584096; PubMed Central PMCID: PMC4022601.

48. Nerys-Junior A, Braga-Dias LP, Pezzuto P, Cotta-de-Almeida V, Tanuri A. Comparison of the editing patterns and editing efficiencies of TALEN and CRISPR-Cas9 when targeting the human CCR5 gene. Genet Mol Biol. 2018:0. doi: 10.1590/1678-4685-GMB-2017-0065 29583154.

49. Boitano AE, Wang J, Romeo R, Bouchez LC, Parker AE, Sutton SE, et al. Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science. 2010;329(5997):1345–8. doi: 10.1126/science.1191536 20688981; PubMed Central PMCID: PMC3033342.

50. Fares I, Chagraoui J, Gareau Y, Gingras S, Ruel R, Mayotte N, et al. Cord blood expansion. Pyrimidoindole derivatives are agonists of human hematopoietic stem cell self-renewal. Science. 2014;345(6203):1509–12. doi: 10.1126/science.1256337 25237102; PubMed Central PMCID: PMC4372335.

51. Hoggatt J, Singh P, Sampath J, Pelus LM. Prostaglandin E2 enhances hematopoietic stem cell homing, survival, and proliferation. Blood. 2009;113(22):5444–55. doi: 10.1182/blood-2009-01-201335 19324903; PubMed Central PMCID: PMC2689046.

52. Pelus LM, Hoggatt J, Singh P. Pulse exposure of haematopoietic grafts to prostaglandin E2 in vitro facilitates engraftment and recovery. Cell Prolif. 2011;44 Suppl 1:22–9. doi: 10.1111/j.1365-2184.2010.00726.x 21481039.


Článok vyšiel v časopise

PLOS One


2019 Číslo 10
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#