Functional expression of complement factor I following AAV-mediated gene delivery in the retina of mice and human cells

Anna K Dreismann, Michelle E McClements, Alun R Barnard, Elise Orhan, Jane P Hughes, Peter J Lachmann, Robert E MacLaren, Anna K Dreismann, Michelle E McClements, Alun R Barnard, Elise Orhan, Jane P Hughes, Peter J Lachmann, Robert E MacLaren

Abstract

Dry age-related macular degeneration (AMD) is characterised by loss of central vision and currently has no approved medical treatment. Dysregulation of the complement system is thought to play an important role in disease pathology and supplementation of Complement Factor I (CFI), a key regulator of the complement system, has the potential to provide a treatment option for AMD. In this study, we demonstrate the generation of AAV constructs carrying the human CFI sequence and expression of CFI in cell lines and in the retina of C57BL/6 J mice. Four codon optimised constructs were compared to the most common human CFI sequence. All constructs expressed CFI protein; however, most codon optimised sequences resulted in significantly reduced CFI secretion compared to the non-optimised CFI sequence. In vivo expression analysis showed that CFI was predominantly expressed in the RPE and photoreceptors. Secreted protein in vitreous humour was demonstrated to be functionally active. The findings presented here have led to the formulation of an AAV-vectored gene therapy product currently being tested in a first-in-human clinical trial in subjects with geographic atrophy secondary to dry AMD (NCT03846193).

Conflict of interest statement

AKD and JPH are employed by Gyroscope Therapeutics Ltd, EO was employed by Gyroscope Therapeutics Ltd while engaged in the research project, PJL and REM are consultants to Gyroscope Therapeutics Ltd, MEM and ARB have no conflict of interest. AKD, JPH, PJL and REM have stock options in Gyroscope Therapeutics Limited.

Figures

Fig. 1. Schematic diagram of the role…
Fig. 1. Schematic diagram of the role of CFI in alternative pathway regulation and AAV design.
A CFI is a key regulator of the alternative pathway and degrades C3b into iC3b and C3dg in the presence of cofactor. Figure adapted with permission from [12]. B Schematic diagram of AAV2 backbone coding sequence. AAV = adeno-associated virus, bGHpA = bovine growth hormone poly adenylation sequence, bp = base pair, C3 = Complement C3, CAG = chicken b-actin promoter with a CMV enhancer and a β-globin intron, CFB = complement factor B, CFD = complement factor D, CFH = complement factor H, CFI = complement factor I, CR1 = Complement Receptor 1, WPRE = Woodchuck hepatitis virus post-transcriptional regulator element. Biorender.com was used to generate Fig. 1A, and B.
Fig. 2. Immunoblot analysis of supernatants and…
Fig. 2. Immunoblot analysis of supernatants and cell lysates of pAAV.CFI, pAAV.CFI.co and pCMV.GFP co-transfected ARPE-19 cells.
A Upper immunoblot: Supernatants of transiently transfected cells. CFI and codon-optimised CFI were detected with mouse anti-human CFI. Plasmid backbone containing AAV sequences without transgene served as a negative control (representative result). Supernatant was loaded non-reduced to prevent separation of heavy and light chain. Lower immunoblot: Lysate of transiently transfected cells. GFP was detected with anti-turbo-GFP and implies that there were not large differences in transfection efficiency between the groups. B Quantification of CFI expression immunoblots by densitometry analysis using ImageJ software. Equal volumes were loaded in each experiment and summary data from 7 independent experiments is shown. Results are presented as relative density of CFI signal relative to signal intensity of AAV.CFI.wt. Data are shown as mean + standard deviation. AAV = adeno-associated virus, CFI = complement factor I, co = codon optimised. Asterisks represent significant differences between codon optimised constructs and the most common CFI sequence (reference) analysed by one-way ANOVA and Dunn’s multiple comparison test: * = P < 0.05, ** = P < 0.01, ns = not significant.
Fig. 3. In vivo expression of CFI…
Fig. 3. In vivo expression of CFI RNA and protein.
AD RT-qPCR to analyse gene expression of AAV.CFI and AAV.CFI.co-1 (n = 3 per condition) of whole eye cup (A), RPE (B), retina (C), and a comparison of all tissues analysed (D). RPE samples generated more CFI mRNA than retina (p < 0.0001) and multiple comparisons revealed significant differences between injection materials, which are described in the text. Data are shown as mean + standard deviation. E Immunoblot of whole eye cups, retina, and RPE of mice injected with AAV.CFI.wt or AAV.CFI.co-1, samples of three mice per condition were pooled for analysis. β-actin staining was performed as a loading control. AAV = adeno-associated virus, CFI = complement factor I, co = codon optimised, ec = eye cup, ret = retina, RPE = retinal pigment epithelium. Asterisks represent significant differences between constructs analysed by one-way ANOVA with Tukey’s multiple comparison: * = P < 0.05, ** = P < 0.01, *** = P < 0.001, **** = P < 0.0001.
Fig. 4. Immunohistological analysis of retinal sections…
Fig. 4. Immunohistological analysis of retinal sections of AAV.CFI and AAV.CFI.co-1 injected mouse eyes.
10 µm retinal sections of sham (AC), AAV.CFI (DF) or AAV.CFI.co-1 (GI) injected mice stained with anti-phalloidin, DAPI, and anti-CFI to demonstrate that CFI protein localises to the RPE cell layer and photoreceptors. AAV = adeno-associated virus, CFI = complement factor I, co = codon optimised, GCL = ganglion cell layer, IS = inner segment of photoreceptors, NFL = nerve fibre layer; ONL = outer nuclear layer, OPL = outer plexiform layer, OS = outer segment of photoreceptors, RPE = retinal pigment epithelium, Scl = Sclera. Magnification: ×20.
Fig. 5. Immunohistological analysis retinal sections of…
Fig. 5. Immunohistological analysis retinal sections of sham and AAV.CFI injected mouse eyes – higher magnification of RPE.
Immunohistological analysis retinal sections of sham (A-C) and AAV.CFI (D-F) injected mouse eyes. Retinal sections were double labelled with phalloidin to stain actin filaments (A, D) and CFI (B, E). Nuclei were stained with DAPI and are shown in merge (C, F). Vesicular staining is depicted with arrows, RPE microvilli are highlighted with asterisk. AAV = adeno-associated virus, BrM: Bruch’s membrane, CFI = complement factor I, Chr: choriocapillaris, IPM: inter photoreceptor matrix, RPE: retinal pigment epithelium, Scl: sclera. Magnification: 189x.
Fig. 6. Immunohistological analysis of whole mounted…
Fig. 6. Immunohistological analysis of whole mounted RPE of AAV.CFI and AAV.CFI.co-1 injected mouse eyes.
Whole mounted RPE of sham (A-C), AAV.CFI (D-F) and AAV.CFI.co-1 (G-I) injected mice was stained with anti-phalloidin and anti-CFI. CFI is highly expressed in RPE cells of AAV.CFI.wt and AAV.CFI.gs treated mice. Abbreviations: AAV = adeno-associated virus, CFI = complement factor I, co = codon optimised. Magnification: 40x.
Fig. 7. Ex vivo demonstration of CFI…
Fig. 7. Ex vivo demonstration of CFI functional activity by a cofactor assay following subretinal administration.
C3b, the substrate, is incubated with CFH as a cofactor; in the absence of CFI no degradation occurs (lane ‘C3b + CFH’, assay negative control). As soon as C3b is incubated with CFH and CFI, degradation of C3b into iC3b occurs, visible as 68 and 43 kDa bands (lane ‘C3b + CFH + CFI, assay positive control). C3 and CFH are incubated with ‘wash’ sample derived from either sham injected or AAV.CFI injected mice as a source of CFI and degradation of the C3 alpha band into the iC3b bands is observed. Abbreviations: AAV = adeno-associated virus, C3 = complement C3, CFI = complement factor I, CFH = complement factor H.

References

    1. Heesterbeek TJ, Lorés-Motta L, Hoyng CB, Lechanteur YT, den Hollander AI. Risk factors for progression of age-related macular degeneration. Ophthal Physiol Optics. 2020;40:140–70. doi: 10.1111/opo.12675.
    1. Lechner J, Chen M, Hogg RE, Toth L, Silvestri G, Chakravarthy U, et al. Higher plasma levels of complement C3a, C4a and C5a increase the risk of subretinal fibrosis in neovascular age-related macular degeneration. Immunity Ageing. 2016;13:1–9. doi: 10.1186/s12979-016-0060-5.
    1. Reynolds R, Hartnett ME, Atkinson JP, Giclas PC, Rosner B, Seddon JM. Plasma complement components and activation fragments: associations with age-related macular degeneration genotypes and phenotypes. Investig Ophthalmol Vis Sci. 2009;50:5818–27. doi: 10.1167/iovs.09-3928.
    1. Scholl HP, Issa PC, Walier M, Janzer S, Pollok-Kopp B, Börncke F, et al. Systemic complement activation in age-related macular degeneration. PLoS ONE. 2008;3:e2593. doi: 10.1371/journal.pone.0002593.
    1. Fritsche LG, Igl W, Bailey JN, Grassmann F, Sengupta S, Bragg-Gresham JL, et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat Genet. 2016;48:134–43. doi: 10.1038/ng.3448.
    1. Geerlings MJ, de Jong EK, den Hollander AI. The complement system in age-related macular degeneration: a review of rare genetic variants and implications for personalized treatment. Mol Immunol. 2017;84:65–76. doi: 10.1016/j.molimm.2016.11.016.
    1. Hollyfield JG, Bonilha VL, Rayborn ME, Yang X, Shadrach KG, Lu L, et al. Oxidative damage–induced inflammation initiates age-related macular degeneration. Nat Med. 2008;14:194–8. doi: 10.1038/nm1709.
    1. Joseph K, Kulik L, Coughlin B, Kunchithapautham K, Bandyopadhyay M, Thiel S, et al. Oxidative stress sensitizes retinal pigmented epithelial (RPE) cells to complement-mediated injury in a natural antibody-, lectin pathway-, and phospholipid epitope-dependent manner. J Biol Chem. 2013;288:12753–65. doi: 10.1074/jbc.M112.421891.
    1. Kavanagh D, Yu Y, Schramm EC, Triebwasser M, Wagner EK, Raychaudhuri S, et al. Rare genetic variants in the CFI gene are associated with advanced age-related macular degeneration and commonly result in reduced serum factor I levels. Human Mol Genet. 2015;24:3861–70.
    1. Hallam TM, Marchbank KJ, Harris CL, Osmond C, Shuttleworth VG, Griffiths H, et al. Rare genetic variants in complement factor I lead to low FI plasma levels resulting in increased risk of age-related macular degeneration. Investig Ophthalmol Vis Sci. 2020;61:18. doi: 10.1167/iovs.61.6.18.
    1. Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: a key system for immune surveillance and homeostasis. Nat Immunol. 2010;11:785–97. doi: 10.1038/ni.1923.
    1. Fearon DT, Austen KF. Activation of the alternative complement pathway due to resistance of zymosan-bound. Proc Natl Acad Sci USA. 1977;74:1683–7. doi: 10.1073/pnas.74.4.1683.
    1. Lachmann PJ The amplification loop of the complement pathways. In Advances in immunology 2009 Jan, 104, 115-49. Academic Press.
    1. Ekdahl KN, Mohlin C, Adler A, Åman A, Manivel VA, Sandholm K, et al. Is generation of C3 (H2O) necessary for activation of the alternative pathway in real life? Mol Immunol. 2019;114:353–61. doi: 10.1016/j.molimm.2019.07.032.
    1. Spitzer D, Mitchell LM, Atkinson JP, Hourcade DE. Properdin can initiate complement activation by binding specific target surfaces and providing a platform for de novo convertase assembly. J Immunol. 2007;179:2600–8. doi: 10.4049/jimmunol.179.4.2600.
    1. Harboe M, Johnson C, Nymo S, Ekholt K, Schjalm C, Lindstad JK, et al. Properdin binding to complement activating surfaces depends on initial C3b deposition. Proc Natl Acad Sci USA. 2017;114:E534–9. doi: 10.1073/pnas.1612385114.
    1. Harrison RA The properdin pathway: an “alternative activation pathway” or a “critical amplification loop” for C3 and C5 activation?. In Seminars in immunopathology 2018 Jan, 40, No. 1, pp. 15-35). Springer Berlin Heidelberg.
    1. Lachmann PJ, Halbwachs LI. The influence of C3b inactivator (KAF) concentration on the ability of serum to support complement activation. Clin Exp Immunol. 1975;21:109.
    1. Harboe M, Garred P, Karlstrøm E, Lindstad JK, Stahl GL, Mollnes TE. The down-stream effects of mannan-induced lectin complement pathway activation depend quantitatively on alternative pathway amplification. Mol Immunol. 2009;47:373–80. doi: 10.1016/j.molimm.2009.09.005.
    1. Lachmann PJ, Lay E, Seilly DJ, Buchberger A, Schwaeble W, Khadake J. Further studies of the down‐regulation by Factor I of the C3b feedback cycle using endotoxin as a soluble activator and red cells as a source of CR1 on sera of different complotype. Clin Exp Immunol. 2016;183:150–6. doi: 10.1111/cei.12714.
    1. Loeb JE, Cordier WS, Harris ME, Weitzman MD, Hope TJ. Enhanced expression of transgenes from adeno-associated virus vectors with the woodchuck hepatitis virus posttranscriptional regulatory element: implications for gene therapy. Human gene therapy. 1999;10:2295–305. doi: 10.1089/10430349950016942.
    1. Patrício MI, Barnard AR, Orlans HO, McClements ME, MacLaren RE. Inclusion of the woodchuck hepatitis virus posttranscriptional regulatory element enhances AAV2-driven transduction of mouse and human retina. Molecular Therapy-Nucleic Acids. 2017;6:198–208. doi: 10.1016/j.omtn.2016.12.006.
    1. Paterna JC, Moccetti T, Mura A, Feldon J, Büeler H. Influence of promoter and WHV post-transcriptional regulatory element on AAV-mediated transgene expression in the rat brain. Gene therapy. 2000;7:1304–11. doi: 10.1038/sj.gt.3301221.
    1. LeWitt PA, Rezai AR, Leehey MA, Ojemann SG, Flaherty AW, Eskandar EN, et al. AAV2-GAD gene therapy for advanced Parkinson’s disease: a double-blind, sham-surgery controlled, randomised trial. Lancet Neurol. 2011;10:309–19. doi: 10.1016/S1474-4422(11)70039-4.
    1. Xue K, Jolly JK, Barnard AR, Rudenko A, Salvetti AP, Patrício MI, et al. Beneficial effects on vision in patients undergoing retinal gene therapy for choroideremia. Nat Med. 2018;24:1507–12. doi: 10.1038/s41591-018-0185-5.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2- ΔΔCT method. Methods. 2001;25:402–8. doi: 10.1006/meth.2001.1262.
    1. Wong MJ, Goldberger G, Isenman DE, Minta JO. Processing of human factor I in COS-1 cells co-transfected with factor I and paired basic amino acid cleaving enzyme (PACE) cDNA. Mol Immunol. 1995;32:379–87. doi: 10.1016/0161-5890(94)00151-P.
    1. Roversi P, Johnson S, Caesar JJ, McLean F, Leath KJ, Tsiftsoglou SA, et al. Structural basis for complement factor I control and its disease-associated sequence polymorphisms. Proc Natl Acad Sci. 2011;108:12839–44. doi: 10.1073/pnas.1102167108.
    1. Dittmar KA, Goodenbour JM, Pan T. Tissue-specific differences in human transfer RNA expression. PLoS Genet. 2006;2:e221. doi: 10.1371/journal.pgen.0020221.
    1. Mauro VP, Chappell SA. A critical analysis of codon optimization in human therapeutics. Trends Mol Med. 2014;20:604–13. doi: 10.1016/j.molmed.2014.09.003.
    1. Boyer DS, Rosenfeld PJ. New Pathways for Dry AMD Treatment - Several potential therapies are in clinical trials. Retinal Physician. 2019;16:24–25.
    1. Liao DS, Metlapally R, Ribeiro R, Saroj N. Inhibiting Complement C3 in Dry AMD - C3 inhibition may be the dark horse to pursue for treating dry AMD. Retinal Physician. 2019;16:34–36.
    1. Sohn JH, Kaplan HJ, Suk HJ, Bora PS, Bora NS. Chronic low level complement activation within the eye is controlled by intraocular complement regulatory proteins. Investig Ophthalmol Vis Sci. 2000;41:3492–502.

Source: PubMed

3
Abonnieren