New frontiers to cure Alport syndrome: COL4A3 and COL4A5 gene editing in podocyte-lineage cells

Sergio Daga, Francesco Donati, Katia Capitani, Susanna Croci, Rossella Tita, Annarita Giliberti, Floriana Valentino, Elisa Benetti, Chiara Fallerini, Francesca Niccheri, Margherita Baldassarri, Maria Antonietta Mencarelli, Elisa Frullanti, Simone Furini, Silvestro Giovanni Conticello, Alessandra Renieri, Anna Maria Pinto, Sergio Daga, Francesco Donati, Katia Capitani, Susanna Croci, Rossella Tita, Annarita Giliberti, Floriana Valentino, Elisa Benetti, Chiara Fallerini, Francesca Niccheri, Margherita Baldassarri, Maria Antonietta Mencarelli, Elisa Frullanti, Simone Furini, Silvestro Giovanni Conticello, Alessandra Renieri, Anna Maria Pinto

Abstract

Alport syndrome (AS) is an inherited genetic disorder characterized by range of alterations from glomerular basement membrane abnormalities up to end-stage renal disease. Pathogenic variants in the collagen α3, α4, and α5 encoding genes are causative both of the autosomal dominant and of the X-linked forms of AS. Podocytes are the only renal cells that are able to produce the COL(IV)a3-a4a5 heterotrimer. We have previously demonstrated how it is possible to isolate podocyte-lineage cells from urine of patients, providing an easily accessible cellular model closer to the podocytes' physiological conditions. Taking advantage of disease-relevant cell lines, we employed a two-plasmid approach in order to achieve a beneficial and stable variant-specific correction using CRISPR/Cas9 genome editing. One plasmid carries a Donor DNA and a reporter system mCherry/GFP to track the activity of Cas9 in cells. The other plasmid carries a self-cleaving SpCas9 and the variant-specific sgRNA. We have analyzed two stable podocyte-lineage cell lines, harboring a variant in the X-linked COL4A5 (p.(Gly624Asp)) and in the autosomal COL4A3 gene (p.(Gly856Glu)). We have achieved reversion of variants greater than 40% with undesired insertions/deletions lower than 15%. Overall, we have demonstrated a new gene therapy approach directly on patients' cells, key players of Alport pathogenesis, and we have reverted COL4 causative variants towards the wild type state. These results, in combination with preclinical models, could open new frontiers in the management and the treatment of the disorder.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1
Fig. 1
sgRNA identification and design and dual plasmid cloning strategy to achieved CRISPR/Cas9 correction. In the upper lane of panel a, the sgRNA is represented in red and blue: PAM in blue and in red the mutated nucleotide for the COL4A5 CRISPR/Cas9 design. Below, the same design is reported for the COL4A3 variant. Plasmid functionality is based on the cloning of two plasmids for CRISPR/Cas9 components delivery. In the first plasmid, the sgRNA is under the control of Human U6 Promoter (b), while the mCherry Red/Green Reporter System is under the control of a CMV Promoter. The Cas9 plasmid is cloned with spCas9 endonuclease gene under control of a CMV promoter flanked by two Self-Cleaving Cas9 for auto-inactivation (c). Representative FACS analysis on HEK293T cells after 48 h of transfection with reporter harboring the wild type sequence and mutated reporter, for COL4A3 and COL4A5 variants, respectively. The population of cells mCherry+/GFP+ is gated in the UR quadrant. The double fluorescence is present only in the cells co-transfected with Donor/Reporter plasmid with specific variant and spCas9 (41.9% and 27.6% for COL4A3 and COL4A5, respectively) demonstrating the specificy of sgRNA (d)
Fig. 2
Fig. 2
Transfection efficacy in urine-derived podocyte-lineage cells. a Type I (Upper left panel) and Type II (Upper right panel) urinary cell colonies (arrowheads) at day 4. At day seven the isolated cells start to aggregate in egg shape conformation (Bottom left panel). On the eighth day, cells starts to have clonal expansion and they can splitted (Bottom right panel) Scale bars, 400 μm. b Both plasmids were transfected on cells in clonal growth stage (Bottom panel) and mCherry and GFP fluorescence was observed in vivo through fluorescence microscope. cRepresentative FACS analysis of GFP expressing. Cells were transfected either with Donor/Reporter plasmid alone and sgRNA/Cas9 coupled with the Donor/Reporter plasmids. The cells were analyzed by FACS 48 h after transient transfection. The percentage of GFP + cells is indicated in the upper right quadrant (UR). At least three independent experiments were performed. The average population in the UR quadrant was 0.2% for Reporter alone; 21.7% for Cas9 + Donor/Reporter
Fig. 3
Fig. 3
CasAnalyzer results on editing efficiency on COL4A5 podocyte-lineage cells. CasAnalzyer Tool operating on the CRISPR/Cas9 treated sample and control has reported a HDR of 58.8% in patients’ mutated podocyte-lineage cells. IGV Visualization Software for the variant shows how there is a substantial loss of heterozygosity with the restoration of the wild-type base, confirming the functionality of the system
Fig. 4
Fig. 4
CasAnalyzer results on editing efficiency on COL4A3 podocyte-lineage cells. The CasAnalzyer Tool performed on the CRISPR/Cas9 treated sample and control has reported a HDR of 44.2% in patients’ mutated podocyte-lineage cells. IGV Visualization Software for this variant also exhibited the loss of heterozygosity with the restoration of the wt base in COL4A3 gene

References

    1. Hertz JM, Thomassen M, Storey H, Flinter F. Clinical utility gene card for: Alport syndrome. Eur J Hum Genet. 2012;20.
    1. Abrahamson DR, Hudson BG, Stroganova L, Borza DB, St John PL. Cellular origins of Type IV Collagen Networks in developing Glomeruli. J Am Soc Nephrol. 2009;20:1471–9. doi: 10.1681/ASN.2008101086.
    1. Daga S, Baldassarri M, Lo Rizzo C, Fallerini C, Imperatore V, Longo I, et al. Urine derived podocytes lineage cells: a promising tool for precision medicine in Alport syndrome. Hum Mutat. 2018;39:302–14. doi: 10.1002/humu.23364.
    1. Renieri A, Bruttini M, Galli L, Zanelli P, Neri T, Rossetti S, et al. X-linked Alport syndrome: a SSCP-based mutation survey over all 51 exons of the COL4A5 gene. Am J Hum Genet. 1996;58:1192–204.
    1. Jais JP, Knebelmann B, Giatras I, De Marchi M, Rizzoni G, Renieri A, et al. X-linked Alport syndrome: natural history in 195 families and genotype- phenotype correlations in males. J Am Soc Nephrol. 2000;11:649–57. doi: 10.1681/ASN.V114649.
    1. Jais JP, Knebelmann B, Giatras I, De Marchi M, Rizzoni G, Renieri A, et al. X-linked Alport syndrome: natural history and genotype-phenotype correlations in girls and women belonging to 195 families: a “European Community Alport Syndrome Concerted Action” study. J Am Soc Nephrol. 2003;14:2603–10. doi: 10.1097/01.ASN.0000090034.71205.74.
    1. Longo I, Porcedda P, Mari F, Giachino D, Meloni I, Deplano C, et al. COL4A3/COL4A4 mutations: from familial hematuria to autosomal-dominant or recessive Alport syndrome. Kidney Int. 2002;61:1947–56. doi: 10.1046/j.1523-1755.2002.00379.x.
    1. Longo I, Scala E, Mari F, Caselli R, Pescucci C, Mencarelli MA, et al. Autosomal recessive Alport syndrome: an in-depth clinical and molecular analysis of five families. Nephrol Dial Transpl. 2006;21:665–71. doi: 10.1093/ndt/gfi312.
    1. Artuso R, Fallerini C, Dosa L, Scionti F, Clementi M, Garosi G, et al. Advances in Alport syndrome diagnosis using next-generation sequencing. Eur J Hum Genet. 2012;20:50–57. doi: 10.1038/ejhg.2011.164.
    1. Pescucci C, Mari F, Longo I, Voqiatzi P, Caselli R, Scala E, et al. Autosomal-dominant Alport syndrome: natural history of a disease due to COL4A3 or COL4A4 gene. Kidney Int. 2004;65:1598–603. doi: 10.1111/j.1523-1755.2004.00560.x.
    1. Karginov FV & Hannon GJ. The CRISPR system: small RNA-guided defense in bacteria and archaea. Mol Cell. 2010;37:7–19.
    1. Casini A, Olivieri M, Petris G, Montagna C, Reginato G, Maule G, et al. A highly specific SpCas9 variant is identified by in vivo screening in yeast. Nat Biotechnol. 2018;36:265–71. doi: 10.1038/nbt.4066.
    1. Lee J, Jung MH, Jeong E, Lee JK. Using Sniper-Cas9 to minimize off-target effects of CRISPR-Cas9 without the loss of on-target activity via directed evolution. J Vis Exp. 2019;26.
    1. Luther DC, Lee YW, Nagaraj H, Scaletti F, Rotello VM. Delivery approaches for CRISPR/Cas9 therapeutics in-vivo: advances and challenges. Exp Opin Drug Deliv. 2018;15:905–13. doi: 10.1080/17425247.2018.1517746.
    1. WareJoncas Z, Campbell JM, Martìnez-Gàlvez G, Gendron WAC, Barry MA, Harris PC, et al. Precision gene editing technology and applications in neprhology. Nat Rev Nephrol. 2018;14:663–77. doi: 10.1038/s41581-018-0047-x.
    1. Li HL, Fujimoto N, Sasakawa N, Shirai S, Ohkame T, Sakuma T, et al. Precise correction of the dystrophin gene in duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Cell Rep. 2015;4:143–54. doi: 10.1016/j.stemcr.2014.10.013.
    1. Ousterout DG, Kabadi AM, Thakore PI, Wh Majoros, Reddy TE, Gersbach CA. Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy. Nat Commun. 2015;18:6244. doi: 10.1038/ncomms7244.
    1. Xie F, Ye L, Chang JC, Beyer AL, Wang J, Muench MO, et al. Seamless gene correction of b-thalassemia mutations in patient-specific iPSCs using CRISPR/Cas9 and piggyback. Genome Res. 2014;24:1526–33. doi: 10.1101/gr.173427.114.
    1. Auricchio A, O’Connor E, Hildinger M, Wilson JM. A single-step affinity column for purification of serotype-5 based adeno-associated viral vectors. Mol Ther. 2001;4:372–4. doi: 10.1006/mthe.2001.0462.
    1. Park J, Lim K, Kim SJ, Bae S. Cas-analyzer: an online tool for assessing genome editing results using NGS data. Bioinformatics. 2017;33:286–8. doi: 10.1093/bioinformatics/btw561.
    1. Haapaniemi E, Botla S, Persson J, Schmierer B, Taipale J. CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nat Med. 2018;24:927–30. doi: 10.1038/s41591-018-0049-z.
    1. Rabai A, Reisser L, Reina-San-Martin B, Mamchaoui K, Cowling BS, Nicot AS, et al. Allele-specific CRISPR/Cas9 correction of a heterozygous DNM2 mutation rescues centronuclear myopathy cell phenotypes. Mol Ther Nucleic Acids. 2019;16:246–56.
    1. Bak RO, Dever DP, Porteus MH. CRISPR/Cas9 genome editing in human hematopoietic stem cells. Nat Protoc. 2018;13:358–76. doi: 10.1038/nprot.2017.143.
    1. Ma H, Marti-Gutierrez N, Park SW, Wu J, Lee Y, Suzuki K, et al. Correction of a pathogenic gene mutation in human embryos. Nature. 2017;548:413–9. doi: 10.1038/nature23305.
    1. Thomas M, Kalita A, Labrecque S, Pim D, Banks L, Matlashewki G. Two polymorphic variants of wild-type p53 differ biochemically and biologically. Mol Cell Biol. 1999;19:1092–100. doi: 10.1128/MCB.19.2.1092.
    1. Pim D, Banks L. p53 polymorphic variants at codon 72 exert different effects on cell cycle progression. Int J Cancer. 2004;108:196–9. doi: 10.1002/ijc.11548.
    1. Suzuki K, Tsunekawa Y, Hernandez-Benitez R, Wu K, Zhu J, Kim EJ, et al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature. 2016;540:144–9. doi: 10.1038/nature20565.
    1. Ertl HCJ, High KA. Impact of AAV capsid-specific T-cell response on design and outcome of clinical gene transfer trials with recombinant adeno-associated viral vectors: an evolving controversy. Hum Gene Ther. 2017;6:1574–83.
    1. Qi YF, Li QH, Shenoy V, Zingler M, Jun JY, Verma A, et al. Comparison of the transduction efficiency of tyrosine-mutant adeno-associated virus serotype vectors in kidney. Clin Exp Pharm Physiol. 2013;40:53–5. doi: 10.1111/1440-1681.12037.
    1. Colella P, Trapani I, Cesi G, Sommella A, Manfredi A, Puppo A, et al. Efficient gene delivery to the cone-enriched pig retina by dual AAV vectors. Gene Ther. 2014;21:450–6. doi: 10.1038/gt.2014.8.
    1. Trapani I, Colella P, Sommella A, Iodice C, Cesi G, de Simone S, et al. Effective delivery of large genes to the retina by dual AAV vectors. EMBO Mol Med. 2014;6:194–211. doi: 10.1002/emmm.201302948.

Source: PubMed

3
Se inscrever