Myeloid/Microglial driven autologous hematopoietic stem cell gene therapy corrects a neuronopathic lysosomal disease

Ana Sergijenko, Alexander Langford-Smith, Ai Y Liao, Claire E Pickford, John McDermott, Gabriel Nowinski, Kia J Langford-Smith, Catherine L R Merry, Simon A Jones, J Edmond Wraith, Robert F Wynn, Fiona L Wilkinson, Brian W Bigger, Ana Sergijenko, Alexander Langford-Smith, Ai Y Liao, Claire E Pickford, John McDermott, Gabriel Nowinski, Kia J Langford-Smith, Catherine L R Merry, Simon A Jones, J Edmond Wraith, Robert F Wynn, Fiona L Wilkinson, Brian W Bigger

Abstract

Mucopolysaccharidosis type IIIA (MPSIIIA) is a lysosomal storage disorder caused by mutations in N-sulfoglucosamine sulfohydrolase (SGSH), resulting in heparan sulfate (HS) accumulation and progressive neurodegeneration. There are no treatments. We previously demonstrated improved neuropathology in MPSIIIA mice using lentiviral vectors (LVs) overexpressing SGSH in wild-type (WT) hematopoietic stem cell (HSC) transplants (HSCTs), achieved via donor monocyte/microglial engraftment in the brain. However, neurological disease was not corrected using LVs in autologous MPSIIIA HSCTs. To improve brain expression via monocyte/microglial specificity, LVs expressing enhanced green fluorescent protein (eGFP) under ubiquitous phosphoglycerate kinase (PGK) or myeloid-specific promoters were compared in transplanted HSCs. LV-CD11b-GFP gave significantly higher monocyte/B-cell eGFP expression than LV-PGK-GFP or LV-CD18-GFP after 6 months. Subsequently, autologous MPSIIIA HSCs were transduced with either LV-PGK-coSGSH or LV-CD11b-coSGSH vectors expressing codon-optimized SGSH and transplanted into MPSIIIA mice. Eight months after HSCT, LV-PGK-coSGSH vectors produced bone marrow SGSH (576% normal activity) similar to LV-CD11b-coSGSH (473%), but LV-CD11b-coSGSH had significantly higher brain expression (11 versus 7%), demonstrating improved brain specificity. LV-CD11b-coSGSH normalized MPSIIIA behavior, brain HS, GM2 ganglioside, and neuroinflammation to WT levels, whereas LV-PGK-coSGSH partly corrected neuropathology but not behavior. We demonstrate compelling evidence of neurological disease correction using autologous myeloid driven lentiviral-HSC gene therapy in MPSIIIA mice.

Figures

Figure 1
Figure 1
Comparison of eGFP expression from myeloid promoters. (a) pCCL LVs containing human CD18, CD11b, and PGK promoters driving eGFP expression. (b) WT bone marrow was lineage depleted and the enriched stem cell population transduced with LV-hCD18-eGFP, LV-hCD11b-eGFP, or LV-hPGK-eGFP at an MOI of 40–70 and transplanted into myeloablated WT mice. n = 8–10 (c) Donor and eGFP+ chimerism was determined 24 weeks post-transplantation. The vector copy number (VCN) was determined in WBCs. (de) Mean fluorescent intensity of eGFP expression was determined for all leukocytes and for CD11b+, CD19+, and CD3+ cells 24 weeks post-transplantation. (fh) Mice with most equivalent copy numbers (VCN of 2.3–4.1) in WBCs were further compared. Error bars represent the SEM. Significant differences between the two groups marked with a line are demonstrated with *P < 0.05; **P < 0.01; and ***P < 0.001. CMV, cytomegalovirus; eGFP, enhanced green fluorescent protein; hPGK, human phosphoglycerate kinase; LV, lentiviral vector; MOI, multiplicity of infection; ns, nonsignificant; WBC, white blood cell; WT, wild-type.
Figure 2
Figure 2
LV-CD11b–modified autologous hematopoietic stem cells correct MPSIIIA mouse behavior. (a) pCCL LVs were made containing either human CD11b or hPGK promoters driving normal or coSGSH. (b) Expression of hSGSH and coSGSH was compared by transient transfection of the human CHME3 microglial cell line. (c) MPSIIIA bone marrow was lineage depleted and transduced with LV-hCD11b-coSGSH or LV-hPGK-coSGSH at an MOI of 80, then transplanted into busulfan myeloablated MPSIIIA mice. (d,e) VCN was determined in WBCs (behavior n = 8–12; biochemistry n = 5). (f) 4 months post-transplantation, the donor chimerism in WBCs was determined by flow cytometry (n = 3–7). (gj) Open-field behavior was evaluated at 6 months (24 weeks) of age (n = 8–15 female mice per group). The measures of hyperactivity were: (g) path length, (i) frequency, and (j) duration spent moving over 100 mm/second, while (h) frequency of centre entries measures thigmotaxis and may be a measure of sense of danger. Error bars represent the SEM. Significant differences to MPSIIIA, between the two groups marked with a line, are demonstrated with *P < 0.05; **P < 0.01; and ***P < 0.001. CMV, cytomegalovirus; coSGSH, codon-optimized human N-sulfoglucosamine sulfohydrolase; hPGK, human phosphoglycerate kinase; hSGSH, human N-sulfoglucosamine sulfohydrolase; LV, lentiviral vector; MOI, multiplicity of infection; MPSIIIA, mucopolysaccharidosis type IIIA; VCN, vector copy number; WBC, white blood cell; WT, wild-type.
Figure 3
Figure 3
LV-CD11b improves brain-specific SGSH activity over LV-PGK and corrects primary HS storage of MPSIIIA mice. At 8 months of age, SGSH enzyme activity was measured in (a) bone marrow, (b) spleen, (c) liver, and (d) brain of perfused mice (n = 5). Secondary upregulation of β-Hex was measured in (e) liver and (f) brain. (g) Total sulfated GAG in the brain was measured with the dye binding Blyscan assay. (h) Total HS storage was measured by 2-aminoacridone (AMAC)–labeled disaccharide analysis. The different HS disaccharides were quantified by (i) AMAC, and (j) the relative proportion of HS that was NAc, NS, 6S, and 2S was also determined. Error bars represent the SEM. Significant differences to MPSIIIA are marked with *P < 0.05 and ***P < 0.001. Treatments that are not significantly different to WT are marked with a line and ns (nonsignificant) (P > 0.05). 6S, 6-O-sulfated; 2S, 2-O-sulfated; β-Hex, β-N-acetyl hexosaminidase; GAG, glycosaminoglycan; HS, heparan sulfate; LV, lentiviral vector; MPSIIIA, mucopolysaccharidosis type IIIA; NAc, N-acetylated; NS, N-sulfated; PGK, phosphoglycerate kinase; SGSH, N-sulfoglucosamine sulfohydrolase; WT, wild-type.
Figure 4
Figure 4
LV-CD11b corrects lysosomal compartment size in MPSIIIA brains. Representative confocal images of (a) brain cortex (layer IV/V) and (b) amygdala (medial nucleus) from approximately −1.18 mm relative to bregma were stained with LAMP2 for lysosomal compartment (green) and NeuN for neuronal nuclei (red), and representative images are shown. Bar = 20 µm. LV, lentiviral vector; MPSIIIA, mucopolysaccharidosis type IIIA; PGK, phosphoglycerate kinase; WT, wild-type.
Figure 5
Figure 5
LV-CD11b normalizes neuroinflammation in the cerebral cortex and amygdala of MPSIIIA mice, while LV-PGK mediates improvements in neuroinflammation. Four brain sections per mouse (n = 5 per group), taken from approximately bregma 0.26, −0.46, −1.18, and −1.94 mm, were stained and quantified with isolectin B4 for microglia number in the (a) cerebral cortex and (b) amygdala or with GFAP for astrocytes in the (c) cerebral cortex and (d) amygdala. Representative images from the cerebral cortex (layer IV/V) and the amygdala, both from approximately −1.18 mm relative to bregma, are shown. Sections were counterstained with Mayer's hematoxylin to highlight the nuclei (blue). Error bars represent the SEM. Significant differences to MPSIIIA, between the two groups marked with a line, are demonstrated with *P < 0.05, **P < 0.01; and ***P < 0.001. Bar = 50 µm. LV, lentiviral vector; MPSIIIA, mucopolysaccharidosis type IIIA; PGK, phosphoglycerate kinase; WT, wild-type.
Figure 6
Figure 6
LV-CD11b and LV-PGK both correct secondary storage and presynaptic vesicles in the cerebral cortex, while LV-PGK is less effective in the amygdala of MPSIIIA mice. Four brain sections per mouse (n = 5 per group), taken from approximately bregma 0.14, −0.58, −1.3, and −2.06 mm, were stained and quantified for GM2 ganglioside in the (a) cerebral cortex and (b) amygdala, and VAMP2 for presynaptic vesicles in the (c) cerebral cortex and (d) amygdala. Representative images from the cerebral cortex (layer IV/V) and the amygdala, both from approximately −1.3 mm relative to bregma, are shown. Error bars represent the SEM. Significant differences to MPSIIIA, between the two groups marked with a line, are demonstrated with ***P < 0.001. Bar = 50 µm. LV, lentiviral vector; MPSIIIA, mucopolysaccharidosis type IIIA; PGK, phosphoglycerate kinase; WT, wild-type.

References

    1. Valstar MJ, Ruijter GJ, van Diggelen OP, Poorthuis BJ, Wijburg FA. Sanfilippo syndrome: a mini-review. J Inherit Metab Dis. 2008;31:240–252.
    1. Holley RJ, Deligny A, Wei W, Watson HA, Niñonuevo MR, Dagälv A, et al. Mucopolysaccharidosis type I, unique structure of accumulated heparan sulfate and increased N-sulfotransferase activity in mice lacking α-L-iduronidase. J Biol Chem. 2011;286:37515–37524.
    1. Langford-Smith A, Wilkinson FL, Langford-Smith KJ, Holley RJ, Sergijenko A, Howe SJ, et al. Hematopoietic stem cell and gene therapy corrects primary neuropathology and behavior in mucopolysaccharidosis IIIA mice. Mol Ther. 2012;20:1610–1621.
    1. Wilkinson FL, Holley RJ, Langford-Smith KJ, Badrinath S, Liao A, Langford-Smith A, et al. Neuropathology in mouse models of mucopolysaccharidosis type I, IIIA and IIIB. PLoS ONE. 2012;7:e35787.
    1. Héron B, Mikaeloff Y, Froissart R, Caridade G, Maire I, Caillaud C, et al. Incidence and natural history of mucopolysaccharidosis type III in France and comparison with United Kingdom and Greece. Am J Med Genet A. 2011;155A:58–68.
    1. Meyer A, Kossow K, Gal A, Mühlhausen C, Ullrich K, Braulke T, et al. Scoring evaluation of the natural course of mucopolysaccharidosis type IIIA (Sanfilippo syndrome type A). Pediatrics. 2007;120:e1255–e1261.
    1. Bhaumik M, Muller VJ, Rozaklis T, Johnson L, Dobrenis K, Bhattacharyya R, et al. A mouse model for mucopolysaccharidosis type III A (Sanfilippo syndrome). Glycobiology. 1999;9:1389–1396.
    1. McGlynn R, Dobrenis K, Walkley SU. Differential subcellular localization of cholesterol, gangliosides, and glycosaminoglycans in murine models of mucopolysaccharide storage disorders. J Comp Neurol. 2004;480:415–426.
    1. Arfi A, Richard M, Gandolphe C, Bonnefont-Rousselot D, Thérond P, Scherman D. Neuroinflammatory and oxidative stress phenomena in MPS IIIA mouse model: the positive effect of long-term aspirin treatment. Mol Genet Metab. 2011;103:18–25.
    1. Fraldi A, Hemsley K, Crawley A, Lombardi A, Lau A, Sutherland L, et al. Functional correction of CNS lesions in an MPS-IIIA mouse model by intracerebral AAV-mediated delivery of sulfamidase and SUMF1 genes. Hum Mol Genet. 2007;16:2693–2702.
    1. Ruzo A, Marcó S, García M, Villacampa P, Ribera A, Ayuso E, et al. Correction of pathological accumulation of glycosaminoglycans in central nervous system and peripheral tissues of MPSIIIA mice through systemic AAV9 gene transfer. Hum Gene Ther. 2012;23:1237–1246.
    1. Savas PS, Hemsley KM, Hopwood JJ. Intracerebral injection of sulfamidase delays neuropathology in murine MPS-IIIA. Mol Genet Metab. 2004;82:273–285.
    1. Boelens JJ, Rocha V, Aldenhoven M, Wynn R, O'Meara A, Michel G, EUROCORD, Inborn error Working Party of EBMT and Duke University et al. Risk factor analysis of outcomes after unrelated cord blood transplantation in patients with Hurler syndrome. Biol Blood Marrow Transplant. 2009;15:618–625.
    1. Boelens JJ, Wynn RF, O'Meara A, Veys P, Bertrand Y, Souillet G, et al. Outcomes of hematopoietic stem cell transplantation for Hurler's syndrome in Europe: a risk factor analysis for graft failure. Bone Marrow Transplant. 2007;40:225–233.
    1. Saif MA, Bigger BW, Brookes KE, Mercer J, Tylee KL, Church HJ, et al. Hematopoietic stem cell transplantation improves the high incidence of neutralizing allo-antibodies observed in Hurler's syndrome after pharmacological enzyme replacement therapy. Haematologica. 2012;97:1320–1328.
    1. Wynn RF, Wraith JE, Mercer J, O'Meara A, Tylee K, Thornley M, et al. Improved metabolic correction in patients with lysosomal storage disease treated with hematopoietic stem cell transplant compared with enzyme replacement therapy. J Pediatr. 2009;154:609–611.
    1. Krivit W, Sung JH, Shapiro EG, Lockman LA. Microglia: the effector cell for reconstitution of the central nervous system following bone marrow transplantation for lysosomal and peroxisomal storage diseases. Cell Transplant. 1995;4:385–392.
    1. Prasad VK, Mendizabal A, Parikh SH, Szabolcs P, Driscoll TA, Page K, et al. Unrelated donor umbilical cord blood transplantation for inherited metabolic disorders in 159 pediatric patients from a single center: influence of cellular composition of the graft on transplantation outcomes. Blood. 2008;112:2979–2989.
    1. Shapiro EG, Lockman LA, Balthazor M, Krivit W. Neuropsychological outcomes of several storage diseases with and without bone marrow transplantation. J Inherit Metab Dis. 1995;18:413–429.
    1. Sivakumur P, Wraith JE. Bone marrow transplantation in mucopolysaccharidosis type IIIA: a comparison of an early treated patient with his untreated sibling. J Inherit Metab Dis. 1999;22:849–850.
    1. Visigalli I, Delai S, Politi LS, Di Domenico C, Cerri F, Mrak E, et al. Gene therapy augments the efficacy of hematopoietic cell transplantation and fully corrects mucopolysaccharidosis type I phenotype in the mouse model. Blood. 2010;116:5130–5139.
    1. Biffi A, Capotondo A, Fasano S, del Carro U, Marchesini S, Azuma H, et al. Gene therapy of metachromatic leukodystrophy reverses neurological damage and deficits in mice. J Clin Invest. 2006;116:3070–3082.
    1. Cartier N, Hacein-Bey-Abina S, Bartholomae CC, Veres G, Schmidt M, Kutschera I, et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science. 2009;326:818–823.
    1. 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;72:8463–8471.
    1. Agura ED, Howard M, Collins SJ. Identification and sequence analysis of the promoter for the leukocyte integrin β-subunit (CD18): a retinoic acid-inducible gene. Blood. 1992;79:602–609.
    1. Hickstein DD, Baker DM, Gollahon KA, Back AL. Identification of the promoter of the myelomonocytic leukocyte integrin CD11b. Proc Natl Acad Sci USA. 1992;89:2105–2109.
    1. Langford-Smith A, Langford-Smith KJ, Jones SA, Wynn RF, Wraith JE, Wilkinson FL, et al. Female mucopolysaccharidosis IIIA mice exhibit hyperactivity and a reduced sense of danger in the open field test. PLoS ONE. 2011;6:e25717.
    1. Sardiello M, Palmieri M, di Ronza A, Medina DL, Valenza M, Gennarino VA, et al. A gene network regulating lysosomal biogenesis and function. Science. 2009;325:473–477.
    1. Cohen MJ, Park YD, Kim H, Pillai JJ. Long-term neuropsychological follow-up of a child with Klüver-Bucy syndrome. Epilepsy Behav. 2010;19:643–646.
    1. Malik P, Krall WJ, Yu XJ, Zhou C, Kohn DB. Retroviral-mediated gene expression in human myelomonocytic cells: a comparison of hematopoietic cell promoters to viral promoters. Blood. 1995;86:2993–3005.
    1. Rozaklis T, Beard H, Hassiotis S, Garcia AR, Tonini M, Luck A, et al. Impact of high-dose, chemically modified sulfamidase on pathology in a murine model of MPS IIIA. Exp Neurol. 2011;230:123–130.
    1. Gaspar HB, Cooray S, Gilmour KC, Parsley KL, Zhang F, Adams S, et al. Hematopoietic stem cell gene therapy for adenosine deaminase-deficient severe combined immunodeficiency leads to long-term immunological recovery and metabolic correction. Sci Transl Med. 2011;3:97–80.
    1. Ausseil J, Desmaris N, Bigou S, Attali R, Corbineau S, Vitry S, et al. Early neurodegeneration progresses independently of microglial activation by heparan sulfate in the brain of mucopolysaccharidosis IIIB mice. PLoS ONE. 2008;3:e2296.
    1. Piccinini AM, Midwood KS. Mediators Inflamm. epub ahead of print; 2010. DAMPening inflammation by modulating TLR signalling.
    1. Archer LD, Langford-Smith KJ, Bigger BW, Fildes JE. J Inherit Metab Dis. epub ahead of print; 2013. Mucopolysaccharide diseases: a complex interplay between neuroinflammation, microglial activation and adaptive immunity.
    1. Langford-Smith KJ, Mercer J, Petty J, Tylee K, Church H, Roberts J, et al. Heparin cofactor II-thrombin complex and dermatan sulphate:chondroitin sulphate ratio are biomarkers of short- and long-term treatment effects in mucopolysaccharide diseases. J Inherit Metab Dis. 2011;34:499–508.
    1. Langford-Smith K, Arasaradnam M, Wraith JE, Wynn R, Bigger BW. Evaluation of heparin cofactor II-thrombin complex as a biomarker on blood spots from mucopolysaccharidosis I, IIIA and IIIB mice. Mol Genet Metab. 2010;99:269–274.
    1. de Ruijter J, de Ru MH, Wagemans T, Ijlst L, Lund AM, Orchard PJ, et al. Heparan sulfate and dermatan sulfate derived disaccharides are sensitive markers for newborn screening for mucopolysaccharidoses types I, II and III. Mol Genet Metab. 2012;107:705–710.
    1. de Ru MH, van der Tol L, van Vlies N, Bigger BW, Hollak CE, Ijlst L, et al. Plasma and urinary levels of dermatan sulfate and heparan sulfate derived disaccharides after long-term enzyme replacement therapy (ERT) in MPS I: correlation with the timing of ERT and with total urinary excretion of glycosaminoglycans. J Inherit Metab Dis. 2013;36:247–255.
    1. Wilkinson FL, Sergijenko A, Langford-Smith KJ, Malinowska M, Wynn RF, Bigger BW. Busulfan conditioning enhances engraftment of hematopoietic donor-derived cells in the brain compared with irradiation. Mol Ther. 2013;21:868–876.
    1. Capotondo A, Milazzo R, Politi LS, Quattrini A, Palini A, Plati T, et al. Brain conditioning is instrumental for successful microglia reconstitution following hematopoietic stem cell transplantation. Proc Natl Acad Sci USA. 2012;109:15018–15023.
    1. Gentner B, Visigalli I, Hiramatsu H, Lechman E, Ungari S, Giustacchini A, et al. Identification of hematopoietic stem cell-specific miRNAs enables gene therapy of globoid cell leukodystrophy. Sci Transl Med. 2010;2:58–84.
    1. Zanta-Boussif MA, Charrier S, Brice-Ouzet A, Martin S, Opolon P, Thrasher AJ, et al. Validation of a mutated PRE sequence allowing high and sustained transgene expression while abrogating WHV-X protein synthesis: application to the gene therapy of WAS. Gene Ther. 2009;16(5):605–619.
    1. Bigger BW, Siapati EK, Mistry A, Waddington SN, Nivsarkar MS, Jacobs L, et al. Permanent partial phenotypic correction and tolerance in a mouse model of hemophilia B by stem cell gene delivery of human factor IX. Gene Ther. 2006;13:117–126.
    1. Siapati EK, Bigger BW, Miskin J, Chipchase D, Parsley KL, Mitrophanous K, et al. Comparison of HIV- and EIAV-based vectors on their efficiency in transducing murine and human hematopoietic repopulating cells. Mol Ther. 2005;12:537.
    1. Langford-Smith A, Malinowska M, Langford-Smith KJ, Wegrzyn G, Jones S, Wynn R, et al. Hyperactive behaviour in the mouse model of mucopolysaccharidosis IIIB in the open field and home cage environments. Genes Brain Behav. 2011;10:673–682.
    1. Malinowska M, Wilkinson FL, Langford-Smith KJ, Langford-Smith A, Brown JR, Crawford BE, et al. Genistein improves neuropathology and corrects behaviour in a mouse model of neurodegenerative metabolic disease. PLoS ONE. 2010;5:e14192.
    1. Canal MM, Wilkinson FL, Cooper JD, Wraith JE, Wynn R, Bigger BW. Circadian rhythm and suprachiasmatic nucleus alterations in the mouse model of mucopolysaccharidosis IIIB. Behav Brain Res. 2010;209:212–220.

Source: PubMed

3
Suscribir