Mannose receptor-mediated delivery of moss-made α-galactosidase A efficiently corrects enzyme deficiency in Fabry mice

Jin-Song Shen, Andreas Busch, Taniqua S Day, Xing-Li Meng, Chun I Yu, Paulina Dabrowska-Schlepp, Benjamin Fode, Holger Niederkrüger, Sabrina Forni, Shuyuan Chen, Raphael Schiffmann, Thomas Frischmuth, Andreas Schaaf, Jin-Song Shen, Andreas Busch, Taniqua S Day, Xing-Li Meng, Chun I Yu, Paulina Dabrowska-Schlepp, Benjamin Fode, Holger Niederkrüger, Sabrina Forni, Shuyuan Chen, Raphael Schiffmann, Thomas Frischmuth, Andreas Schaaf

Abstract

Enzyme replacement therapy (ERT) is an effective treatment for several lysosomal storage disorders (LSDs). Intravenously infused enzymes are taken up by tissues through either the mannose 6-phosphate receptor (M6PR) or the mannose receptor (MR). It is generally believed that M6PR-mediated endocytosis is a key mechanism for ERT in treating LSDs that affect the non-macrophage cells of visceral organs. However, the therapeutic efficacy of MR-mediated delivery of mannose-terminated enzymes in these diseases has not been fully evaluated. We tested the effectiveness of a non-phosphorylated α-galactosidase A produced from moss (referred to as moss-aGal) in vitro and in a mouse model of Fabry disease. Endocytosis of moss-aGal was MR-dependent. Compared to agalsidase alfa, a phosphorylated form of α-galactosidase A, moss-aGal was more preferentially targeted to the kidney. Cellular localization of moss-aGal and agalsidase alfa in the heart and kidney was essentially identical. A single injection of moss-aGal led to clearance of accumulated substrate in the heart and kidney to an extent comparable to that achieved by agalsidase alfa. This study suggested that mannose-terminated enzymes may be sufficiently effective for some LSDs in which non-macrophage cells are affected, and that M6P residues may not always be a prerequisite for ERT as previously considered.

Figures

Fig. 1
Fig. 1
In vitro characterization and uptake studies a Enzyme preparations separated in SDS-PAGE and stained with Coomassie blue. Lanes 1 and 2 are moss-aGal and agalsidase alfa respectively. Lanes 3 and 4 are moss-aGal and agalsidase alfa digested with PNGase F. Arrow, α-gal A enzymes after digestion; arrowhead, PNGase F (36 KDa). Protein standard and molecular weights are shown on left. b Moss-aGal and agalsidase alfa (1 ng each) detected by western blot using a polyclonal antibody specific to human α-gal A. Representative data from three independent experiments was shown. c Specific α-gal A activities of enzyme preparations determined using artificial substrate 4-MU-α-D-galactopyranoside. d Plots of reaction velocities of moss-aGal and agalsidase alfa assessed with artificial substrate 4-nitrophenyl α-D-galactopyranoside (pNP-Gal). e Stability of the enzymes diluted in buffered human plasma and heated at 37 °C (data are means of triplicates). f Intracellular α-gal A activities of Fabry patient fibroblasts after overnight incubation with different enzymes in the presence or absence of 5 mM M6P or 2 mg/ml yeast mannan. g Gb3 immunofluorescence staining shows massive lysosomal accumulation of Gb3 in untreated Fabry patient fibroblasts (upper) and significantly decreased Gb3 in the cells that were treated with moss-aGal (lower). h and i MR expression in Fabry patient fibroblasts and microvascular endothelial cells IMFE1. IMFE1 cells were MR-positive determined by both western blot (h) and immunofluorescence staining (i), while the fibroblasts were MR-negative. j Intracellular α-gal A activities of IMFE1 cells after overnight incubation with different enzymes in the presence or absence of M6P or mannan. k Uptake rates of different enzymes in IMFE1 cells. Cells were harvested at indicated time points and intracellular activities were measured. ***P < 0.001, moss-aGal vs. high-mann moss-aGal or agalsidase alfa. l Western blot analysis of internalized α-gal A in IMFE1 cells after 3 h incubation with different enzyme preparations. m Binding of different enzymes (10 μg/ml) to IMFE1 cells. After 3 h incubation at 4 °C, cell surface-bound enzymes were determined by enzyme assay. The dotted line indicates activity level of mock-treated IMFE1 cells in this assay (i.e., background level). *P < 0.05, ***P < 0.001. All the data in graphs are presented as mean ± SEM (n = 3-4). High-mann: high-mann moss-aGal; Agal-alfa: agalsidase alfa; Agal-beta: agalsidase beta
Fig. 2
Fig. 2
Tissue and cellular distribution of infused enzymes a-c Enzyme preparations were injected into Fabry mice, and α-gal A activities in the kidney, heart, spleen, and liver were measured 2 h post-injection. a Specific activities in organs. Data are presented as mean ± SEM (n = 5). *P < 0.05, **P < 0.01. b Activities in whole organs were calculated and data are presented as % of total activity recovered from four organs. c α-gal A protein in kidney homogenates detected by western blot. Arrow, specific α-gal A band in moss-aGal-injected mice. No detectable specific band was seen in agalsidase alfa-injected mice. Arrowhead, approximate position where agalsidase alfa band may migrate (based on findings shown in Fig. 1b). d Cellular distribution of infused enzymes in the heart and kidney was determined by immunohistochemistry (n = 2). Heart: asterisks indicate the blood vessels with immunostaining positive cells (most likely endothelial cells), and arrows indicate positive perivascular cells (presumably macrophages). Kidney: arrows indicate immunostaining positive tubular epithelial cells. Scale bar: 25 μm. Original magnification: 400×. Agal-alfa: agalsidase alfa
Fig. 3
Fig. 3
Efficacy of moss-aGal in clearing accumulated Gb3 in tissues. Gb3 contents in kidney (a), heart (b), and liver (c) were analyzed 7 days after a single infusion of either moss-aGal or agalsidase alfa at various doses. Data are presented as mean ± SEM (n = 4-5). *P < 0.05, ***P < 0.001. Statistical significance shown on top of each agalsidase alfa-injected group indicates difference between agalsidase alfa and the same dose of moss-aGal. Agal-alfa: agalsidase alfa

References

    1. Ashe KM, Budman E, Bangari DS, et al (2015) Efficacy of enzyme and substrate reduction therapy with a novel antagonist of glucosylceramide synthase for Fabry disease. Mol Med
    1. Ballou CE. Isolation, characterization, and properties of Saccharomyces cerevisiae mnn mutants with nonconditional protein glycosylation defects. Methods Enzymol. 1990;185:440–470. doi: 10.1016/0076-6879(90)85038-P.
    1. Barton NW, Brady RO, Dambrosia JM, et al. Replacement therapy for inherited enzyme deficiency--macrophage-targeted glucocerebrosidase for Gaucher’s disease. N Engl J Med. 1991;324:1464–1470. doi: 10.1056/NEJM199105233242104.
    1. Bonten EJ, Wang D, Toy JN, et al. Targeting macrophages with baculovirus-produced lysosomal enzymes: implications for enzyme replacement therapy of the glycoprotein storage disorder galactosialidosis. FASEB J. 2004;18:971–973.
    1. Brady R, Gal AE, Bradley RM, Martensson E, Warshaw AL, Laster L. Enzymatic defect in Fabry disease: ceramide trihexosidase deficiency. N Engl J Med. 1967;276:1163–1167. doi: 10.1056/NEJM196705252762101.
    1. Chen Y, Jin M, Egborge T, Coppola G, Andre J, Calhoun DH. Expression and characterization of glycosylated and catalytically active recombinant human alpha-galactosidase A produced in Pichia pastoris. Protein Expr Purif. 2000;20:472–484. doi: 10.1006/prep.2000.1325.
    1. Chen Y, Jin M, Goodrich L, Smith G, Coppola G, Calhoun DH. Purification and characterization of human alpha-galactosidase A expressed in insect cells using a baculovirus vector. Protein Expr Purif. 2000;20:228–236. doi: 10.1006/prep.2000.1284.
    1. Chiba Y, Sakuraba H, Kotani M, et al. Production in yeast of alpha-galactosidase A, a lysosomal enzyme applicable to enzyme replacement therapy for Fabry disease. Glycobiology. 2002;12:821–828. doi: 10.1093/glycob/cwf096.
    1. Christensen EI, Zhou Q, Sorensen SS, et al. Distribution of alpha-galactosidase A in normal human kidney and renal accumulation and distribution of recombinant alpha-galactosidase A in Fabry mice. J Am Soc Nephrol. 2007;18:698–706. doi: 10.1681/ASN.2006080822.
    1. Costa AR, Rodrigues ME, Henriques M, Oliveira R, Azeredo J. Glycosylation: impact, control and improvement during therapeutic protein production. Crit Rev Biotechnol. 2014;34:281–299. doi: 10.3109/07388551.2013.793649.
    1. Deegan PB. Fabry disease, enzyme replacement therapy and the significance of antibody responses. J Inherit Metab Dis. 2012;35:227–243. doi: 10.1007/s10545-011-9400-y.
    1. Desnick RJ, Ioannou YA, Eng CM. α-Galactosidase A deficiency: Fabry disease. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The metabolic and molecular bases of inherited disease. New York: McGraw-Hill; 2001. pp. 3733–3774.
    1. Du H, Cameron TL, Garger SJ, et al. Wolman disease/cholesteryl ester storage disease: efficacy of plant-produced human lysosomal acid lipase in mice. J Lipid Res. 2008;49:1646–1657. doi: 10.1194/jlr.M700482-JLR200.
    1. Durant B, Forni S, Sweetman L, et al. Sex differences of urinary and kidney globotriaosylceramide and lyso-globotriaosylceramide in Fabry mice. J Lipid Res. 2011;52:1742–1746. doi: 10.1194/jlr.M017178.
    1. Eng CM, Banikazemi M, Gordon RE, et al. A phase 1/2 clinical trial of enzyme replacement in fabry disease: pharmacokinetic, substrate clearance, and safety studies. Am J Hum Genet. 2001;68:711–722. doi: 10.1086/318809.
    1. Eng CM, Guffon N, Wilcox WR, et al. Safety and efficacy of recombinant human alpha-galactosidase A—replacement therapy in Fabry’s disease. N Engl J Med. 2001;345:9–16. doi: 10.1056/NEJM200107053450102.
    1. Gomord V, Faye L. Posttranslational modification of therapeutic proteins in plants. Curr Opin Plant Biol. 2004;7:171–181. doi: 10.1016/j.pbi.2004.01.015.
    1. Groger M, Holnthoner W, Maurer D, et al. Dermal microvascular endothelial cells express the 180-kDa macrophage mannose receptor in situ and in vitro. J Immunol. 2000;165:5428–5434. doi: 10.4049/jimmunol.165.10.5428.
    1. He X, Haselhorst T, von Itzstein M, et al. Production of alpha-L-iduronidase in maize for the potential treatment of a human lysosomal storage disease. Nat Commun. 2012;3:1062. doi: 10.1038/ncomms2070.
    1. Ioannou YA, Zeidner KM, Gordon RE, Desnick RJ. Fabry disease: preclinical studies demonstrate the effectiveness of alpha-galactosidase A replacement in enzyme-deficient mice. Am J Hum Genet. 2001;68:14–25. doi: 10.1086/316953.
    1. Kizhner T, Azulay Y, Hainrichson M, et al. Characterization of a chemically modified plant cell culture expressed human alpha-Galactosidase-A enzyme for treatment of Fabry disease. Mol Genet Metab. 2015;114:259–267. doi: 10.1016/j.ymgme.2014.08.002.
    1. Koprivova A, Stemmer C, Altmann F, et al. Targeted knockouts of Physcomitrella lacking plant-specific immunogenic N-glycans. Plant Biotechnol J. 2004;2:517–523. doi: 10.1111/j.1467-7652.2004.00100.x.
    1. Kornfeld S. Structure and function of the mannose 6-phosphate/insulinlike growth factor II receptors. Annu Rev Biochem. 1992;61:307–330. doi: 10.1146/annurev.bi.61.070192.001515.
    1. Lee K, Jin X, Zhang K, et al. A biochemical and pharmacological comparison of enzyme replacement therapies for the glycolipid storage disorder Fabry disease. Glycobiology. 2003;13:305–313. doi: 10.1093/glycob/cwg034.
    1. Linehan SA, Martinez-Pomares L, Stahl PD, Gordon S. Mannose receptor and its putative ligands in normal murine lymphoid and nonlymphoid organs: In situ expression of mannose receptor by selected macrophages, endothelial cells, perivascular microglia, and mesangial cells, but not dendritic cells. J Exp Med. 1999;189:1961–1972. doi: 10.1084/jem.189.12.1961.
    1. Marchesan D, Cox TM, Deegan PB. Lysosomal delivery of therapeutic enzymes in cell models of Fabry disease. J Inherit Metab Dis. 2012;35:1107–1117. doi: 10.1007/s10545-012-9472-3.
    1. Murray GJ, Anver MR, Kennedy MA, Quirk JM, Schiffmann R. Cellular and tissue distribution of intravenously administered agalsidase alfa. Mol Genet Metab. 2007;90:307–312. doi: 10.1016/j.ymgme.2006.11.008.
    1. Ohshima T, Murray GJ, Swaim WD, et al. alpha-Galactosidase A deficient mice: a model of Fabry disease. Proc Natl Acad Sci U S A. 1997;94:2540–2544. doi: 10.1073/pnas.94.6.2540.
    1. Pastores GM. Agalsidase alfa (Replagal) in the treatment of Anderson-Fabry disease. Biogeosciences. 2007;1:291–300.
    1. Prabakaran T, Nielsen R, Larsen JV, et al. Receptor-mediated endocytosis of alpha-galactosidase A in human podocytes in Fabry disease. PLoS One. 2011;6 doi: 10.1371/journal.pone.0025065.
    1. Prabakaran T, Nielsen R, Satchell SC, et al. Mannose 6-phosphate receptor and sortilin mediated endocytosis of alpha-galactosidase A in kidney endothelial cells. PLoS One. 2012;7 doi: 10.1371/journal.pone.0039975.
    1. Sands MS, Vogler CA, Ohlemiller KK, et al. Biodistribution, kinetics, and efficacy of highly phosphorylated and non-phosphorylated beta-glucuronidase in the murine model of mucopolysaccharidosis VII. J Biol Chem. 2001;276:43160–43165. doi: 10.1074/jbc.M107778200.
    1. Schiffmann R, Murray GJ, Treco D, et al. Infusion of alpha-galactosidase A reduces tissue globotriaosylceramide storage in patients with Fabry disease. Proc Natl Acad Sci U S A. 2000;97:365–370. doi: 10.1073/pnas.97.1.365.
    1. Schiffmann R, Kopp JB, Austin HA, 3rd, et al. Enzyme replacement therapy in Fabry disease: a randomized controlled trial. JAMA. 2001;285:2743–2749. doi: 10.1001/jama.285.21.2743.
    1. Shaaltiel Y, Bartfeld D, Hashmueli S, et al. Production of glucocerebrosidase with terminal mannose glycans for enzyme replacement therapy of Gaucher’s disease using a plant cell system. Plant Biotechnol J. 2007;5:579–590. doi: 10.1111/j.1467-7652.2007.00263.x.
    1. Shen JS, Meng XL, Schiffmann R, Brady RO, Kaneski CR. Establishment and characterization of Fabry disease endothelial cells with an extended lifespan. Mol Genet Metab. 2007;92:137–144. doi: 10.1016/j.ymgme.2007.06.003.
    1. Shen JS, Meng XL, Wight-Carter M, et al. Blocking hyperactive androgen receptor signaling ameliorates cardiac and renal hypertrophy in Fabry mice. Hum Mol Genet. 2015;24:3181–3191. doi: 10.1093/hmg/ddv070.
    1. Sly WS, Vogler C, Grubb JH, et al. Enzyme therapy in mannose receptor-null mucopolysaccharidosis VII mice defines roles for the mannose 6-phosphate and mannose receptors. Proc Natl Acad Sci U S A. 2006;103:15172–15177. doi: 10.1073/pnas.0607053103.
    1. Stahl PD, Ezekowitz RA. The mannose receptor is a pattern recognition receptor involved in host defense. Curr Opin Immunol. 1998;10:50–55. doi: 10.1016/S0952-7915(98)80031-9.
    1. Tsukimura T, Kawashima I, Togawa T, et al. Efficient uptake of recombinant alpha-galactosidase A produced with a gene-manipulated yeast by Fabry mice kidneys. Mol Med. 2012;18:76–82. doi: 10.2119/molmed.2011.00248.
    1. Van Patten SM, Hughes H, Huff MR, et al. Effect of mannose chain length on targeting of glucocerebrosidase for enzyme replacement therapy of Gaucher disease. Glycobiology. 2007;17:467–478. doi: 10.1093/glycob/cwm008.
    1. Zhu Y, Jiang JL, Gumlaw NK, et al. Glycoengineered acid alpha-glucosidase with improved efficacy at correcting the metabolic aberrations and motor function deficits in a mouse model of Pompe disease. Mol Ther. 2009;17:954–963. doi: 10.1038/mt.2009.37.
    1. Ziegler RJ, Cherry M, Barbon CM, et al. Correction of the biochemical and functional deficits in fabry mice following AAV8-mediated hepatic expression of alpha-galactosidase A. Mol Ther. 2007;15:492–500. doi: 10.1038/sj.mt.6300066.

Source: PubMed

3
구독하다