Viable mouse models of acid beta-glucosidase deficiency: the defect in Gaucher disease

You-Hai Xu, Brian Quinn, David Witte, Gregory A Grabowski, You-Hai Xu, Brian Quinn, David Witte, Gregory A Grabowski

Abstract

Gaucher disease is an autosomal recessively inherited disease caused by mutations at the acid beta-glucosidase (GCase) locus (GBA). To develop viable models of Gaucher disease, point mutations (pmuts), encoding N370S, V394L, D409H, or D409V were introduced into the mouse GCase (gba) locus. DNA sequencing verified each unique pmut. Mutant GCase mRNAs were near wild-type (WT) levels. GCase activities were reduced to 2 to 25% of WT in liver, lung, spleen, and cultured fibroblasts from pmut/pmut or pmut/null mice. The corresponding brain GCase activities were approximately 25% of WT. N370S homozygosity was lethal in the neonatal period. For the other pmut mice, a few storage cells appeared in the spleen at > or =7 months (D409H or D409V homozygotes) or > or =1 year (V394L homozygotes). V394L/null, D409H/null, or D409V/null mice showed scattered storage cells in spleen at approximately 3 to 4 months. Occasional storage cells (sinusoidal cells) were present in liver. In D409V/null mice, large numbers of Mac-3-positive storage cells (ie, macrophages) accumulated in the lung. Glycosphingolipid analyses showed varying rates of progressive glucosylceramide accumulation in visceral organs of pmut/pmut or pmut/null mice, but not in brain. These GCase-deficient mice provide tools for gaining insight into the pathophysiology of Gaucher disease and developing improved therapies.

Figures

Figure 1.
Figure 1.
Mouse gba targeting strategy. The WT gba is shown at the top. The replacement fragment from the targeting construct (second line) contained exons 5 to 11, parts of intron 4 and the 3′ flanking region, and a floxed neo selection marker in intron 8. The fourth line shows the resultant recombination with gba (gba with neo) and, following cre excision of neo (gba without neo) the recombined allele containing one loxP () site and the individual exon 9 mutations (*).
Figure 2.
Figure 2.
Genotype analyses for gba point mutations. A 391 bp PCR fragment from WT (bottom arrow) or a 485-bp PCR fragment (391+loxP, top arrow) from point-mutated gba mice. The genotypes are indicated above each lane. +/+, WT homozygote; +/−, heterozygote (V394L, D409H, and D409V); and −/−, homozygote (eg, D409V). Ctrl is the PCR control without template DNA. MW = size standards.
Figure 3.
Figure 3.
CBE inhibition of mouse N370S and V394L mGCases: WT (▾), V394L (♦) and N370S (▴) mGCase were harvested from media of Sf21 cells infected with the desired recombinant baculovirus. The enzymes were delipidated and dialyzed before being incubated (30 minutes; 37°C) with conduritol B epoxide (CBE, 1 to 500 μmol/L) and 4MU-Glc (4 mmol/L). The control sample (▪) was from media of uninfected Sf21 cells.
Figure 4.
Figure 4.
Storage cells in spleen and liver of gba point-mutated mice. A: Representative samples from spleen of D409V/null mice. A large storage cell (arrow) is illustrated with prominent amounts of pale staining vacuolated cytoplasm. (H&E, magnification, ×400). B: A representative section from liver. A small cluster of pale staining storage cells are illustrated in the sinusoidal space (H&E, magnification, ×400).
Figure 5.
Figure 5.
Storage cell staining with Mac-3. A: Section of wild-type lung with no accumulation of storage cells in the alveolar spaces. B: D409V/null lung with large storage cells (arrows) in the alveolar space. C: Wild-type lung stained for Mac-3. D: Mac-3 staining of lung from the D409V/null mouse indicating macrophage origin of the storage cells. Magnification: ×165 (A), ×250 (B), ×350 (C and D).
Figure 6.
Figure 6.
Glycosphingolipid (GSL) profiles from various tissue of point-mutated gba mice. Cerebellum, liver, spleen, and lung (∼100 mg wet weight) were from gba mutant mice at various ages. The tissues were processed for thin-layer chromatography. The genotypes and ages are indicated only on the bottom for clarity of presentation. Migration standards for of sphingolipids are on the right: GluCer, glucosylceramide; GalCer, galactosylceramide; SO4-GalCer, sulfogalactosylceramide; LacCer, lactosylceramide; THCer, ceramide trihexoside; Spm, sphingomyelin. The borate-impregnated plates were developed in chloroform/methanol/water (65/25/4) using borate-impregnated plates. Mature WT mice showed little variation in GSL levels in the respective tissues within the time frame of these experiments.

References

    1. Beutler E, Grabowski GA: Gaucher disease. Scriver CR Beaudet AL Sly WS Valle D eds. The Metabolic and Molecular Bases of Inherited Diseases. 2001:pp 3635-3668 McGraw-Hill New York
    1. Blom S, Erikson A: Gaucher disease–Norrbottnian type: neurodevelopmental, neurological, and neurophysiological aspects. Eur J Pediatr 1983, 140:316-322
    1. Kolodny EH, Ullman MD, Mankin HJ, Raghavan SS, Topol J, Sullivan JL: Phenotypic manifestations of Gaucher disease: clinical features in 48 biochemically verified type 1 patients and comment on type 2 patients. Prog Clin Biol Res 1982, 95:33-65
    1. Volk BW, Wallace BJ, Adachi M: Infantile Gaucher’s disease: electron microscopic and histochemical studies of a cerebral biopsy. J Neuropathol Exp Neurol 1967, 26:176-177
    1. Adachi M, Wallace BJ, Schneck L, Volk BW: Fine structure of central nervous system in early infantile Gaucher’s disease. Arch Pathol (Chicago) 1967, 83:513-526
    1. Charrow J, Andersson HC, Kaplan P, Kolodny E, Mistry PK, Pastores G, Rosenbloom B, Scott CR, Wappner RS, Weinreb N, Zimran A: The Gaucher registry: demographics and disease characteristics of 1698 patients with Gaucher disease. Arch Intern Med 2000, 160:2835-2843
    1. Sibille A, Eng CM, Kim SJ, Pastores G, Grabowski GA: Phenotype/genotype correlations in Gaucher disease type I: clinical and therapeutic implications. Am J Hum Genet 1993, 52:1094-1101
    1. Abrahamov A, Elstein D, Gross-Tsur V, Farber B, Glaser Y, Hadas-Halpern I, Ronen S, Tafakjdi M, Horowitz M, Zimran A: Gaucher’s disease variant characterised by progressive calcification of heart valves and unique genotype. Lancet 1995, 346:1000-1003
    1. Theophilus B, Latham T, Grabowski GA, Smith FI: Gaucher disease: molecular heterogeneity and phenotype-genotype correlations. Am J Hum Genet 1989, 45:212-225
    1. Theophilus BD, Latham T, Grabowski GA, Smith FI: Comparison of RNase A, a chemical cleavage and GC-clamped denaturing gradient gel electrophoresis for the detection of mutations in exon 9 of the human acid β-glucosidase gene. Nucleic Acids Res 1989, 17:7707-7722
    1. Grace ME, Newman KM, Scheinker V, Berg-Fussman A, Grabowski GA: Analysis of human acid β-glucosidase by site-directed mutagenesis and heterologous expression. J Biol Chem 1994, 269:2283-2291
    1. Sidransky E, Tayebi N, Stubblefield BK, Eliason W, Klineburgess A, Pizzolato GP, Cox JN, Porta J, Bottani A, DeLozier-Blanchet CD: The clinical, molecular, and pathological characterisation of a family with two cases of lethal perinatal type 2 Gaucher disease. J Med Genet 1996, 33:132-136
    1. Finn LS, Zhang M, Chen SH, Scott CR: Severe type II Gaucher disease with ichthyosis, arthrogryposis, and neuronal apoptosis: molecular and pathological analyses. Am J Med Genet 2000, 91:222-226
    1. Tayebi N, Cushner SR, Kleijer W, Lau EK, Damschroder-Williams PJ, Stubblefield BK, Den Hollander J, Sidransky E: Prenatal lethality of a homozygous null mutation in the human glucocerebrosidase gene. Am J Med Genet 1997, 73:41-47
    1. Tybulewicz VLJ, Tremblay ML, LaMarca ME, Willemsen R, Stubblefield BK, Winfield S, Zablocka B, Sidransky E, Martin BM, Huang SP, Mintzer KA, Westphal H, Mulligan RC, Ginns EI: Animal model of Gaucher’s disease from targeted disruption of the mouse glucocerebrosidase gene. Nature 1992, 357:407-410
    1. Liu Y, Suzuki K, Reed JD, Grinberg A, Westphal H, Hoffmann A, Doring T, Sandhoff K, Proia RL: Mice with type 2 and 3 Gaucher disease point mutations generated by a single insertion mutagenesis procedure. Proc Natl Acad Sci USA 1998, 95:2503-2508
    1. Kyle JW, Birkenmeier EH, Gwynn B, Vogler C, Hoppe PC, Hoffmann JW, Sly WS: Correction of murine mucopolysaccharidosis VII by a human β-glucuronidase transgene. Proc Natl Acad Sci USA 1990, 87:3914-3918
    1. Vogler C, Birkenmeier EH, Sly WS, Levy B, Pegors C, Kyle JW, Beamer WG: A murine model of mucopolysaccharidosis VII: gross and microscopic findings in β-glucuronidase-deficient mice. Am J Pathol 1990, 136:207-217
    1. Sands MS, Barker JE, Vogler C, Levy B, Gwynn B, Galvin N, Sly WS, Birkenmeier E: Treatment of murine mucopolysaccharidosis type VII by syngeneic bone marrow transplantation in neonates. Lab Invest 1993, 68:676-686
    1. Clarke LA, Russell CS, Pownall S, Warrington CL, Borowski A, Dimmick JE, Toone J, Jirik FR: Murine mucopolysaccharidosis type I: targeted disruption of the murine α-L-iduronidase gene. Hum Mol Genet 1997, 6:503-511
    1. de Geest N, Bonten E, Mann L, de Sousa-Hitzler J, Hahn C, d’Azzo A: Systemic and neurologic abnormalities distinguish the lysosomal disorders sialidosis and galactosialidosis in mice. Hum Mol Genet 2002, 11:1455-1464
    1. Marathe S, Miranda SR, Devlin C, Johns A, Kuriakose G, Williams KJ, Schuchman EH, Tabas I: Creation of a mouse model for non-neurological (type B) Niemann-Pick disease by stable, low level expression of lysosomal sphingomyelinase in the absence of secretory sphingomyelinase: relationship between brain intra-lysosomal enzyme activity and central nervous system function. Hum Mol Genet 2000, 9:1967-1976
    1. Li CM, Park JH, Simonaro CM, He X, Gordon RE, Friedman AH, Ehleiter D, Paris F, Manova K, Hepbildikler S, Fuks Z, Sandhoff K, Kolesnick R, Schuchman EH, Hepbiloikler S: Insertional mutagenesis of the mouse acid ceramidase gene leads to early embryonic lethality in homozygotes and progressive lipid storage disease in heterozygotes. Genomics 2002, 79:218-224
    1. Sandhoff K, Kolter T, Harzer K: Sphingolipid activator proteins. Scriver CR Beaudet A Sly W Valle D eds. The Metabolic and Molecular Bases of Inherited Disease. 2001:pp 3371-3388 McGraw-Hill New York
    1. Dahl N, Lagerström M, Erikson A, Pettersson U: Gaucher disease type III (Norrbottnian type) is caused by a single mutation in exon 10 of the glucocerebrosidase gene. Am J Hum Genet 1990, 47:275-278
    1. Lewis BD, Nelson PV, Robertson EF, Morris CP: Mutation analysis of 28 Gaucher disease patients: the australasian experience. Am J Med Genet 1994, 49:218-223
    1. O’Neill RR, Tokoro T, Kozak CA, Brady RO: Comparison of the chromosomal localization of murine and human glucocerebrosidase genes and of the deduced amino acid sequences. Proc Natl Acad Sci USA 1989, 86:5049-5053
    1. Sambrook J, Fritsch EF, Maniatis T: Molecular Cloning, A Laboratory Manual. 1989. Cold Spring Harbor Laboratory New York
    1. Sabath DE, Broome HE, Prystowsky MB: Glyceraldehyde-3-phosphate dehydrogenase mRNA is a major interleukin 2-induced transcript in a cloned T-helper lymphocyte. Gene 1990, 91:185-191
    1. Ausubel FM, Brent R, Kingston RE, Moore DD, Seidman JG, Smith JA, Struhl K: Current Protocols in Molecular Biology. 1992:pp 10.5.1-10.5.5 Greene Publishing Associates and Wiley-Interscience New York
    1. Fabbro D, Desnick RJ, Grabowski GA: Gaucher disease: genetic heterogeneity within and among the subtypes detected by immunoblotting. Am J Hum Genet 1987, 40:15-31
    1. O’Reilly DR, Miller LK, Luckow VA: Baculovirus Expression Vectors: A Laboratory Manual. 1992. W. H. Freeman and Company New York
    1. Grabowski GA, Osiecki-Newman K, Dinur T, Fabbro D, Legler G, Gatt S, Desnick RJ: Human acid β-glucosidase: use of conduritol B epoxide derivatives to investigate the catalytically active normal and Gaucher disease enzymes. J Biol Chem 1986, 261:8263-8269
    1. Fujita N, Suzuki K, Vanier MT, Popko B, Maeda N, Klein A, Henseler M, Sandhoff K, Nakayasu H: Targeted disruption of the mouse sphingolipid activator protein gene: a complex phenotype, including severe leukodystrophy and wide-spread storage of multiple sphingolipids. Hum Mol Genet 1996, 5:711-725
    1. Wells MA, Dittmer JC: The use of Sephadex for the removal of non-lipid contaminants from lipid extracts. Biochemistry 1963, 2:1259-1263
    1. Dreyfus H, Guerold B, Freysz L, Hicks D: Successive isolation and separation of the major lipid fractions including gangliosides from single biological samples. Anal Biochem 1997, 249:67-78
    1. Igisu H, Takahashi H, Suzuki K, Suzuki K: Abnormal accumulation of galactosylceramide in the kidney of twitcher mouse. Biochem Biophys Res Commun 1983, 110:940-944
    1. Daniels LB, Coyle PJ, Glew RH, Radin NS, Labow RS: Brain glucocerebrosidase in Gaucher’s disease. Arch Neurol 1982, 39:550-556
    1. Mizukami H, Mi Y, Wada R, Kono M, Yamashita T, Liu Y, Werth N, Sandhoff R, Sandhoff K, Proia RL: Systemic inflammation in glucocerebrosidase-deficient mice with minimal glucosylceramide storage. J Clin Invest 2002, 109:1215-1221
    1. Sauer B: Inducible gene targeting in mice using the Cre/lox system. Methods 1998, 14:381-392
    1. Conzelmann E, Sandhoff K: Partial enzyme deficiencies: residual activities and the development of neurological disorders. Dev Neurosci 1983, 6:58-71
    1. Svennerholm L, Hakansson G, Mansson JE, Nilsson O: Chemical differentiation of the Gaucher subtypes. Prog Clin Biol Res 1982, 95:231-252
    1. Conradi NG, Sourander P, Nilsson O, Svennerholm L, Erikson A: Neuropathology of the Norrbottnian type of Gaucher disease: morphological and biochemical studies. Acta Neuropathol (Berl) 1984, 65:99-109
    1. Pasmanik-Chor M, Laadan S, Elroy-Stein O, Zimran A, Abrahamov A, Gatt S, Horowitz M: The glucocerebrosidase D409H mutation in Gaucher disease. Biochem Mol Med 1996, 59:125-133
    1. Latham TE, Theophilus BD, Grabowski GA, Smith FI: Heterogeneity of mutations in the acid β-glucosidase gene of Gaucher disease patients. DNA Cell Biol 1991, 10:15-21

Source: PubMed

Подписаться