Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(12): 102625, 2022 12.
Article in English | MEDLINE | ID: mdl-36306823

ABSTRACT

Mucopolysaccharidosis type IIIA (MPS IIIA) is a lysosomal storage disorder caused by N-sulfoglucosamine sulfohydrolase (SGSH) deficiency. SGSH removes the sulfate from N-sulfoglucosamine residues on the nonreducing end of heparan sulfate (HS-NRE) within lysosomes. Enzyme deficiency results in accumulation of partially degraded HS within lysosomes throughout the body, leading to a progressive severe neurological disease. Enzyme replacement therapy has been proposed, but further evaluation of the treatment strategy is needed. Here, we used Chinese hamster ovary cells to produce a highly soluble and fully active recombinant human sulfamidase (rhSGSH). We discovered that rhSGSH utilizes both the CI-MPR and LRP1 receptors for uptake into patient fibroblasts. A single intracerebroventricular (ICV) injection of rhSGSH in MPS IIIA mice resulted in a tissue half-life of 9 days and widespread distribution throughout the brain. Following a single ICV dose, both total HS and the MPS IIIA disease-specific HS-NRE were dramatically reduced, reaching a nadir 2 weeks post dose. The durability of effect for reduction of both substrate and protein markers of lysosomal dysfunction and a neuroimmune response lasted through the 56 days tested. Furthermore, seven weekly 148 µg doses ICV reduced those markers to near normal and produced a 99.5% reduction in HS-NRE levels. A pilot study utilizing every other week dosing in two animals supports further evaluation of less frequent dosing. Finally, our dose-response study also suggests lower doses may be efficacious. Our findings show that rhSGSH can normalize lysosomal HS storage and markers of a neuroimmune response when delivered ICV.


Subject(s)
Brain Diseases , Mucopolysaccharidosis III , Cricetinae , Animals , Humans , Mice , Mucopolysaccharidosis III/drug therapy , Mucopolysaccharidosis III/metabolism , CHO Cells , Pilot Projects , Cricetulus , Hydrolases/metabolism , Brain/metabolism , Heparitin Sulfate/metabolism , Brain Diseases/metabolism , Lysosomes/metabolism , Disease Models, Animal
3.
Blood ; 136(22): 2524-2534, 2020 11 26.
Article in English | MEDLINE | ID: mdl-32915950

ABSTRACT

Adeno-associated virus (AAV)-based gene therapies can restore endogenous factor VIII (FVIII) expression in hemophilia A (HA). AAV vectors typically use a B-domain-deleted FVIII transgene, such as human FVIII-SQ in valoctocogene roxaparvovec (AAV5-FVIII-SQ). Surprisingly, the activity of transgene-produced FVIII-SQ was between 1.3 and 2.0 times higher in one-stage clot (OS) assays than in chromogenic-substrate (CS) assays, whereas recombinant FVIII-SQ products had lower OS than CS activity. Transgene-produced and recombinant FVIII-SQ showed comparable specific activity (international units per milligram) in the CS assay, demonstrating that the diverging activities arise in the OS assay. Higher OS activity for transgene-produced FVIII-SQ was observed across various assay kits and clinical laboratories, suggesting that intrinsic molecular features are potential root causes. Further experiments in 2 participants showed that transgene-produced FVIII-SQ accelerated early factor Xa and thrombin formation, which may explain the higher OS activity based on a kinetic bias between OS and CS assay readout times. Despite the faster onset of coagulation, global thrombin levels were unaffected. A correlation with joint bleeds suggested that both OS and CS assay remained clinically meaningful to distinguish hemophilic from nonhemophilic FVIII activity levels. During clinical development, the CS activity was chosen as a surrogate end point to conservatively assess hemostatic efficacy and enable comparison with recombinant FVIII-SQ products. Relevant trials are registered on clinicaltrials.gov as #NCT02576795 and #NCT03370913 and, respectively, on EudraCT (European Union Drug Regulating Authorities Clinical Trials Database; https://eudract.ema.europa.eu) as #2014-003880-38 and #2017-003215-19.


Subject(s)
Factor VIII , Genetic Therapy , Hemophilia A , Parvovirinae , Transgenes , Dependovirus , Factor VIII/genetics , Factor VIII/metabolism , Hemophilia A/blood , Hemophilia A/genetics , Hemophilia A/therapy , Humans , Male
4.
J Biol Chem ; 295(39): 13556-13569, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32727849

ABSTRACT

Mutations in the galactosidase ß 1 (GLB1) gene cause lysosomal ß-galactosidase (ß-Gal) deficiency and clinical onset of the neurodegenerative lysosomal storage disease, GM1 gangliosidosis. ß-Gal and neuraminidase 1 (NEU1) form a multienzyme complex in lysosomes along with the molecular chaperone, protective protein cathepsin A (PPCA). NEU1 is deficient in the neurodegenerative lysosomal storage disease sialidosis, and its targeting to and stability in lysosomes strictly depend on PPCA. In contrast, ß-Gal only partially depends on PPCA, prompting us to investigate the role that ß-Gal plays in the multienzyme complex. Here, we demonstrate that ß-Gal negatively regulates NEU1 levels in lysosomes by competitively displacing this labile sialidase from PPCA. Chronic cellular uptake of purified recombinant human ß-Gal (rhß-Gal) or chronic lentiviral-mediated GLB1 overexpression in GM1 gangliosidosis patient fibroblasts coincides with profound secondary NEU1 deficiency. A regimen of intermittent enzyme replacement therapy dosing with rhß-Gal, followed by enzyme withdrawal, is sufficient to augment ß-Gal activity levels in GM1 gangliosidosis patient fibroblasts without promoting NEU1 deficiency. In the absence of ß-Gal, NEU1 levels are elevated in the GM1 gangliosidosis mouse brain, which are restored to normal levels following weekly intracerebroventricular dosing with rhß-Gal. Collectively, our results highlight the need to carefully titrate the dose and dosing frequency of ß-Gal augmentation therapy for GM1 gangliosidosis. They further suggest that intermittent intracerebroventricular enzyme replacement therapy dosing with rhß-Gal is a tunable approach that can safely augment ß-Gal levels while maintaining NEU1 at physiological levels in the GM1 gangliosidosis brain.


Subject(s)
Enzyme Replacement Therapy , Fibroblasts/enzymology , Lysosomes/enzymology , Mucolipidoses , beta-Galactosidase/therapeutic use , Animals , CHO Cells , Cricetulus , Humans , Lysosomes/genetics , Mice , Mice, Mutant Strains , Mucolipidoses/drug therapy , Mucolipidoses/enzymology , Mucolipidoses/genetics , Neuraminidase/genetics , Neuraminidase/metabolism
5.
Biotechnol Prog ; 36(3): e2974, 2020 05.
Article in English | MEDLINE | ID: mdl-31990124

ABSTRACT

Difficult-to-express (DTE) recombinant proteins such as multi-specific proteins, DTE monoclonal antibodies, and lysosomal enzymes have seen difficulties in manufacturability using Chinese hamster ovary (CHO) cells or other mammalian cells as production platforms. CHO cells are preferably used for recombinant protein production for their ability to secrete human-like recombinant proteins with posttranslational modification, resistance to viral infection, and familiarity with drug regulators. However, despite huge progress made in engineering CHO cells for high volumetric productivity, DTE proteins like recombinant lysosomal sulfatase represent one of the poorly understood proteins. Furthermore, there is growing interest in the use of microRNA (miRNA) to engineer CHO cells expressing DTE proteins to improve cell performance of relevant bioprocess phenotypes. To our knowledge, no research has been done to improve CHO cell production of DTE recombinant lysosomal sulfatase using miRNA. We identified miR-23a and miR-377 as miRNAs predicted to target SUMF1, an activator of sulfatases, using in silico prediction tools. Transient inhibition of CHO endogenous miR-23a/miR-377 significantly enhanced recombinant sulfatase enzyme-specific activity by ~15-21% compared to scramble without affecting cell growth. Though inhibition of miR-23a/miR-377 had no significant effect on the mRNA and protein levels of SUMF1, overexpression of miR-23a/377 caused ~30% and ~27-29% significant reduction in endogenous SUMF1 protein and mRNA expression levels, respectively. In summary, our data demonstrate the importance of using miRNA to optimize the CHO cell line secreting DTE recombinant lysosomal sulfatase.


Subject(s)
MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Sulfatases/biosynthesis , Animals , CHO Cells , Cell Proliferation , Cricetulus , Gene Expression Regulation, Enzymologic/genetics , Humans , Lysosomes/enzymology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sulfatases/genetics
6.
Drug Deliv Transl Res ; 10(2): 425-439, 2020 04.
Article in English | MEDLINE | ID: mdl-31942701

ABSTRACT

BMN 250 is being developed as enzyme replacement therapy for Sanfilippo type B, a primarily neurological rare disease, in which patients have deficient lysosomal alpha-N-acetylglucosaminidase (NAGLU) enzyme activity. BMN 250 is taken up in target cells by the cation-independent mannose 6-phosphate receptor (CI-MPR, insulin-like growth factor 2 receptor), which then facilitates transit to the lysosome. BMN 250 is dosed directly into the central nervous system via the intracerebroventricular (ICV) route, and the objective of this work was to compare systemic intravenous (IV) and ICV delivery of BMN 250 to confirm the value of ICV dosing. We first assess the ability of enzyme to cross a potentially compromised blood-brain barrier in the Naglu-/- mouse model and then assess the potential for CI-MPR to be employed for receptor-mediated transport across the blood-brain barrier. In wild-type and Naglu-/- mice, CI-MPR expression in brain vasculature is high during the neonatal period but virtually absent by adolescence. In contrast, CI-MPR remains expressed through adolescence in non-affected non-human primate and human brain vasculature. Combined results from IV administration of BMN 250 in Naglu-/- mice and IV and ICV administration in healthy juvenile non-human primates suggest a limitation to therapeutic benefit from IV administration because enzyme distribution is restricted to brain vascular endothelial cells: enzyme does not reach target neuronal cells following IV administration, and pharmacological response following IV administration is likely restricted to clearance of substrate in endothelial cells. In contrast, ICV administration enables central nervous system enzyme replacement with biodistribution to target cells.


Subject(s)
Acetylglucosaminidase/administration & dosage , Acetylglucosaminidase/genetics , Blood-Brain Barrier/chemistry , Insulin-Like Growth Factor II/administration & dosage , Mucopolysaccharidosis III/drug therapy , Receptor, IGF Type 2/metabolism , Recombinant Fusion Proteins/administration & dosage , Acetylglucosaminidase/therapeutic use , Administration, Intravenous , Animals , Disease Models, Animal , Enzyme Replacement Therapy , Female , Infusions, Intraventricular , Insulin-Like Growth Factor II/therapeutic use , Male , Mice , Mice, Transgenic , Mucopolysaccharidosis III/genetics , Primates , Recombinant Fusion Proteins/therapeutic use , Translational Research, Biomedical
7.
J Biol Chem ; 295(39): 13532-13555, 2020 09 25.
Article in English | MEDLINE | ID: mdl-31481471

ABSTRACT

Autosomal recessive mutations in the galactosidase ß1 (GLB1) gene cause lysosomal ß-gal deficiency, resulting in accumulation of galactose-containing substrates and onset of the progressive and fatal neurodegenerative lysosomal storage disease, GM1 gangliosidosis. Here, an enzyme replacement therapy (ERT) approach in fibroblasts from GM1 gangliosidosis patients with recombinant human ß-gal (rhß-gal) produced in Chinese hamster ovary cells enabled direct and precise rhß-gal delivery to acidified lysosomes. A single, low dose (3 nm) of rhß-gal was sufficient for normalizing ß-gal activity and mediating substrate clearance for several weeks. We found that rhß-gal uptake by the fibroblasts is dose-dependent and saturable and can be competitively inhibited by mannose 6-phosphate, suggesting cation-independent, mannose 6-phosphate receptor-mediated endocytosis from the cell surface. A single intracerebroventricularly (ICV) administered dose of rhß-gal (100 µg) resulted in broad bilateral biodistribution of rhß-gal to critical regions of pathology in a mouse model of GM1 gangliosidosis. Weekly ICV dosing of rhß-gal for 8 weeks substantially reduced brain levels of ganglioside and oligosaccharide substrates and reversed well-established secondary neuropathology. Of note, unlike with the ERT approach, chronic lentivirus-mediated GLB1 overexpression in the GM1 gangliosidosis patient fibroblasts caused accumulation of a prelysosomal pool of ß-gal, resulting in activation of the unfolded protein response and endoplasmic reticulum stress. This outcome was unsurprising in light of our in vitro biophysical findings for rhß-gal, which include pH-dependent and concentration-dependent stability and dynamic self-association. Collectively, our results highlight that ICV-ERT is an effective therapeutic intervention for managing GM1 gangliosidosis potentially more safely than with gene therapy approaches.


Subject(s)
Enzyme Replacement Therapy , Gangliosidosis, GM1/therapy , beta-Galactosidase/metabolism , Animals , Gangliosidosis, GM1/metabolism , Gangliosidosis, GM1/pathology , Mice
8.
Mol Ther Methods Clin Dev ; 14: 56-63, 2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31309128

ABSTRACT

Sanfilippo syndrome type B, or mucopolysaccharidosis IIIB (MPS IIIB), is a rare autosomal recessive lysosomal storage disease caused by a deficiency of α-N-acetylglucosaminidase (NAGLU). Deficiency in NAGLU disrupts the lysosomal turnover of heparan sulfate (HS), which results in the abnormal accumulation of partially degraded HS in cells and tissues. BMN 250 (NAGLU-insulin-like growth factor 2 [IGF2]) is a recombinant fusion protein developed as an investigational enzyme replacement therapy for MPS IIIB. The IGF2 peptide on BMN 250 promotes enhanced targeting of the enzyme to lysosomes through its interaction with the mannose 6-phosphate receptor. The focus of these studies was to further characterize the ability of NAGLU-IGF2 to clear accumulated HS compared to unmodified NAGLU in primary cellular models of MPS IIIB. Here, we establish distinct primary cell models of MPS IIIB with HS accumulation. These cellular models revealed distinct NAGLU uptake characteristics that depend on the duration of exposure. We found that with sustained exposure, NAGLU uptake and HS clearance occurred independent of known lysosomal targeting signals. In contrast, under conditions of limited exposure duration, NAGLU-IGF2 was taken up more rapidly than the unmodified NAGLU into MPS IIIB primary fibroblasts, astrocytes, and cortical neurons, where it efficiently degraded accumulated HS. These studies illustrate the importance of using physiologically relevant conditions in the evaluation of enzyme replacement therapies in cellular models.

9.
PLoS One ; 14(1): e0207836, 2019.
Article in English | MEDLINE | ID: mdl-30657762

ABSTRACT

Sanfilippo syndrome type B (Sanfilippo B; Mucopolysaccharidosis type IIIB) occurs due to genetic deficiency of lysosomal alpha-N-acetylglucosaminidase (NAGLU) and subsequent lysosomal accumulation of heparan sulfate (HS), which coincides with devastating neurodegenerative disease. Because NAGLU expressed in Chinese hamster ovary cells is not mannose-6-phosphorylated, we developed an insulin-like growth factor 2 (IGF2)-tagged NAGLU molecule (BMN 250; tralesinidase alfa) that binds avidly to the IGF2 / cation-independent mannose 6-phosphate receptor (CI-MPR) for glycosylation independent lysosomal targeting. BMN 250 is currently being developed as an investigational enzyme replacement therapy for Sanfilippo B. Here we distinguish two cellular uptake mechanisms by which BMN 250 is targeted to lysosomes. In normal rodent-derived neurons and astrocytes, the majority of BMN250 uptake over 24 hours reaches saturation, which can be competitively inhibited with IGF2, suggestive of CI-MPR-mediated uptake. Kuptake, defined as the concentration of enzyme at half-maximal uptake, is 5 nM and 3 nM in neurons and astrocytes, with a maximal uptake capacity (Vmax) corresponding to 764 nmol/hr/mg and 5380 nmol/hr/mg, respectively. Similar to neurons and astrocytes, BMN 250 uptake in Sanfilippo B patient fibroblasts is predominantly CI-MPR-mediated, resulting in augmentation of NAGLU activity with doses of enzyme that fall well below the Kuptake (5 nM), which are sufficient to prevent HS accumulation. In contrast, uptake of the untagged recombinant human NAGLU (rhNAGLU) enzyme in neurons, astrocytes and fibroblasts is negligible at the same doses tested. In microglia, receptor-independent uptake, defined as enzyme uptake resistant to competition with excess IGF2, results in appreciable lysosomal delivery of BMN 250 and rhNAGLU (Vmax = 12,336 nmol/hr/mg and 5469 nmol/hr/mg, respectively). These results suggest that while receptor-independent mechanisms exist for lysosomal targeting of rhNAGLU in microglia, BMN 250, by its IGF2 tag moiety, confers increased CI-MPR-mediated lysosomal targeting to neurons and astrocytes, two additional critical cell types of Sanfilippo B disease pathogenesis.


Subject(s)
Acetylglucosaminidase/metabolism , Endocytosis , Insulin-Like Growth Factor II/therapeutic use , Mucopolysaccharidosis III/drug therapy , Mucopolysaccharidosis III/pathology , Recombinant Fusion Proteins/therapeutic use , Acetylglucosaminidase/pharmacokinetics , Acetylglucosaminidase/therapeutic use , Animals , Astrocytes/metabolism , Axons/metabolism , Cations , Fibroblasts/metabolism , Heparitin Sulfate/metabolism , Hippocampus/pathology , Humans , Insulin-Like Growth Factor II/pharmacokinetics , Lysosomes/enzymology , Microglia/metabolism , Rats , Receptor, IGF Type 2/metabolism , Recombinant Fusion Proteins/pharmacokinetics
10.
Mol Ther Methods Clin Dev ; 6: 43-53, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28664165

ABSTRACT

Sanfilippo syndrome type B (mucopolysaccharidosis IIIB), caused by inherited deficiency of α-N-acetylglucosaminidase (NAGLU), required for lysosomal degradation of heparan sulfate (HS), is a pediatric neurodegenerative disorder with no approved treatment. Intracerebroventricular (ICV) delivery of a modified recombinant NAGLU, consisting of human NAGLU fused with insulin-like growth factor 2 (IGF2) for enhanced lysosomal targeting, was previously shown to result in marked enzyme uptake and clearance of HS storage in the Naglu-/- mouse brain. To further evaluate regional, cell type-specific, and dose-dependent biodistribution of NAGLU-IGF2 (BMN 250) and its effects on biochemical and histological pathology, Naglu-/- mice were treated with 1-100 µg ICV doses (four times over 2 weeks). 1 day after the last dose, BMN 250 (100 µg doses) resulted in above-normal NAGLU activity levels, broad biodistribution, and uptake in all cell types, with NAGLU predominantly localized to neurons in the Naglu-/- mouse brain. This led to complete clearance of disease-specific HS and reduction of secondary lysosomal defects and neuropathology across various brain regions lasting for at least 28 days after the last dose. The substantial brain uptake of NAGLU attainable by this highest ICV dosage was required for nearly complete attenuation of disease-driven storage accumulations and neuropathology throughout the Naglu-/- mouse brain.

11.
J Biol Chem ; 292(10): 4255-4265, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28115520

ABSTRACT

Neutrophil myeloperoxidase (MPO) catalyzes the H2O2-dependent oxidation of chloride anion to generate hypochlorous acid, a potent antimicrobial agent. Besides its well defined role in innate immunity, aberrant degranulation of neutrophils in several inflammatory diseases leads to redistribution of MPO to the extracellular space, where it can mediate tissue damage by promoting the oxidation of several additional substrates. Here, we demonstrate that mannose 6-phosphate receptor-mediated cellular uptake and delivery of MPO to lysosomes of retinal pigmented epithelial (RPE) cells acts to clear this harmful enzyme from the extracellular space, with lysosomal-delivered MPO exhibiting a half-life of 10 h. Lysosomal-targeted MPO exerts both cell-protective and cytotoxic functions. From a therapeutic standpoint, MPO catalyzes the in vitro degradation of N-retinylidene-N-retinylethanolamine, a toxic form of retinal lipofuscin that accumulates in RPE lysosomes and drives the pathogenesis of Stargardt macular degeneration. Furthermore, chronic cellular uptake and accumulation of MPO in lysosomes coincides with N-retinylidene-N-retinylethanolamine elimination in a cell-based model of macular degeneration. However, lysosomal-delivered MPO also disrupts lysosomal acidification in RPE cells, which coincides with nuclear translocation of the lysosomal stress-sensing transcription factor EB and, eventually, cell death. Based on these findings we predict that under periods of acute exposure, cellular uptake and lysosomal degradation of MPO mediates elimination of this harmful enzyme, whereas chronic exposure results in progressive accumulation of MPO in lysosomes. Lysosomal-accumulated MPO can be both cell-protective, by promoting the degradation of toxic retinal lipofuscin deposits, and cytotoxic, by triggering lysosomal stress and cell death.


Subject(s)
Lipofuscin/metabolism , Lysosomes/metabolism , Lysosomes/pathology , Peroxidase/metabolism , Receptor, IGF Type 2/metabolism , Retinal Pigment Epithelium/metabolism , Stress, Physiological , Cells, Cultured , Humans , Retinal Pigment Epithelium/pathology
12.
Proc Natl Acad Sci U S A ; 111(41): 14870-5, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25267636

ABSTRACT

Mucopolysaccharidosis type IIIB (MPS IIIB, Sanfilippo syndrome type B) is a lysosomal storage disease characterized by profound intellectual disability, dementia, and a lifespan of about two decades. The cause is mutation in the gene encoding α-N-acetylglucosaminidase (NAGLU), deficiency of NAGLU, and accumulation of heparan sulfate. Impediments to enzyme replacement therapy are the absence of mannose 6-phosphate on recombinant human NAGLU and the blood-brain barrier. To overcome the first impediment, a fusion protein of recombinant NAGLU and a fragment of insulin-like growth factor II (IGFII) was prepared for endocytosis by the mannose 6-phosphate/IGFII receptor. To bypass the blood-brain barrier, the fusion protein ("enzyme") in artificial cerebrospinal fluid ("vehicle") was administered intracerebroventricularly to the brain of adult MPS IIIB mice, four times over 2 wk. The brains were analyzed 1-28 d later and compared with brains of MPS IIIB mice that received vehicle alone or control (heterozygous) mice that received vehicle. There was marked uptake of the administered enzyme in many parts of the brain, where it persisted with a half-life of approximately 10 d. Heparan sulfate, and especially disease-specific heparan sulfate, was reduced to control level. A number of secondary accumulations in neurons [ß-hexosaminidase, LAMP1(lysosome-associated membrane protein 1), SCMAS (subunit c of mitochondrial ATP synthase), glypican 5, ß-amyloid, P-tau] were reduced almost to control level. CD68, a microglial protein, was reduced halfway. A large amount of enzyme also appeared in liver cells, where it reduced heparan sulfate and ß-hexosaminidase accumulation to control levels. These results suggest the feasibility of enzyme replacement therapy for MPS IIIB.


Subject(s)
Acetylglucosaminidase/therapeutic use , Brain/metabolism , Drug Delivery Systems , Insulin-Like Growth Factor II/therapeutic use , Mucopolysaccharidosis III/drug therapy , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use , Animals , Biomarkers/metabolism , Brain/pathology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Endocytosis , Fibroblasts/metabolism , Fibroblasts/pathology , Heparitin Sulfate/metabolism , Humans , Injections, Intraventricular , Liver/metabolism , Lysosomal Membrane Proteins/metabolism , Mice , Mucopolysaccharidosis III/pathology , Neurons/metabolism , Neurons/pathology , Protein Binding , beta-N-Acetylhexosaminidases/metabolism
13.
PLoS One ; 5(8): e12194, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20808938

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA; Morquio A syndrome) is a lysosomal storage disorder caused by deficiency of N-acetylgalactosamine-6-sulfatase (GALNS), an enzyme that degrades keratan sulfate (KS). Currently no therapy for MPS IVA is available. We produced recombinant human (rh)GALNS as a potential enzyme replacement therapy for MPS IVA. Chinese hamster ovary cells stably overexpressing GALNS and sulfatase modifying factor-1 were used to produce active ( approximately 2 U/mg) and pure (>or=97%) rhGALNS. The recombinant enzyme was phosphorylated and was dose-dependently taken up by mannose-6-phosphate receptor (K(uptake) = 2.5 nM), thereby restoring enzyme activity in MPS IVA fibroblasts. In the absence of an animal model with a skeletal phenotype, we established chondrocytes isolated from two MPS IVA patients as a disease model in vitro. MPS IVA chondrocyte GALNS activity was not detectable and the cells exhibited KS storage up to 11-fold higher than unaffected chondrocytes. MPS IVA chondrocytes internalized rhGALNS into lysosomes, resulting in normalization of enzyme activity and decrease in KS storage. rhGALNS treatment also modulated gene expression, increasing expression of chondrogenic genes Collagen II, Collagen X, Aggrecan and Sox9 and decreasing abnormal expression of Collagen I. Intravenous administration of rhGALNS resulted in biodistribution throughout all layers of the heart valve and the entire thickness of the growth plate in wild-type mice. We show that enzyme replacement therapy with recombinant human GALNS results in clearance of keratan sulfate accumulation, and that such treatment ameliorates aberrant gene expression in human chondrocytes in vitro. Penetration of the therapeutic enzyme throughout poorly vascularized, but clinically relevant tissues, including growth plate cartilage and heart valve, as well as macrophages and hepatocytes in wild-type mouse, further supports development of rhGALNS as enzyme replacement therapy for MPS IVA.


Subject(s)
Cartilage/metabolism , Enzyme Replacement Therapy/methods , Mucopolysaccharidosis IV/drug therapy , Mucopolysaccharidosis IV/metabolism , Animals , Biological Transport , Cattle , Chondrocytes/metabolism , Chondrocytes/pathology , Chondroitinsulfatases/isolation & purification , Chondroitinsulfatases/metabolism , Chondroitinsulfatases/pharmacokinetics , Chondroitinsulfatases/therapeutic use , Female , Gene Expression Regulation/drug effects , Growth Plate/metabolism , Heart Valves/metabolism , Humans , Keratan Sulfate/metabolism , Liver/pathology , Lysosomes/metabolism , Macrophages/metabolism , Mice , Mucopolysaccharidosis IV/pathology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use
14.
Biochem J ; 382(Pt 2): 581-7, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15175008

ABSTRACT

Arylsulphatases B (ASB) and A (ASA) are subject to a unique post-translational modification that is required for their function. The modification reaction, conversion of an active-site cysteine into a formylglycine, becomes saturated when these enzymes are overexpressed. We have removed the possibility of in vivo modification by expressing mutants of ASB and ASA in which the active-site cysteine is substituted with a serine. These mutants are expressed much more efficiently when compared with the native enzymes under identical conditions. The purified ASB mutant can then be converted into catalytically active ASB in vitro using vanadate and light.


Subject(s)
Arylsulfatases/biosynthesis , Arylsulfatases/metabolism , Light , Mutation, Missense , Vanadates/metabolism , Animals , Arylsulfatases/genetics , CHO Cells/chemistry , CHO Cells/metabolism , Cell Line , Cricetinae , DNA, Complementary/genetics , Enzyme Activation , Humans , Liver/enzymology , Mutation, Missense/genetics , Oxidation-Reduction , Transfection/methods , Vanadates/chemistry
15.
J Biol Chem ; 279(33): 35037-46, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15170390

ABSTRACT

Enzyme replacement therapy for lysosomal storage disorders depends on efficient uptake of recombinant enzyme into the tissues of patients. This uptake is mediated by oligosaccharide receptors including the cation-independent mannose 6-phosphate receptor and the mannose receptor. We have sought to exploit alternative receptor systems that are independent of glycosylation but allow for efficient delivery to the lysosome. Fusions of the human lysosomal enzymes alpha-l-iduronidase or acid alpha-glucosidase with the receptor-associated protein were efficiently endocytosed by lysosomal storage disorder patient fibroblasts, rat C6 glioma cells, mouse C2C12 myoblasts, and recombinant Chinese hamster ovary cells expressing individual members of the low-density lipoprotein receptor family. Uptake of the fusions exceeded that of phosphorylated enzyme in all cases, often by an order of magnitude or greater. Uptake was specifically mediated by members of the low-density lipoprotein receptor protein family and was followed by delivery of the fusions to the lysosome. The advantages of the lipoprotein receptor system over oligosaccharide receptor systems include more efficient cellular delivery and the potential for transcytosis of ligands across tight endothelia, including the blood-brain barrier.


Subject(s)
Iduronidase/metabolism , Lysosomes/metabolism , Receptors, Lipoprotein/metabolism , alpha-Glucosidases/metabolism , Animals , Blotting, Western , CHO Cells , Carbohydrates/chemistry , Cell Line, Tumor , Cricetinae , Dose-Response Relationship, Drug , Electrophoresis , Endocytosis , Fibroblasts/metabolism , Glioma/metabolism , Glycosaminoglycans/chemistry , Humans , Kinetics , Ligands , Lipoproteins, LDL/metabolism , Mice , Oligosaccharides/chemistry , Phosphorylation , Plasmids/metabolism , Protein Binding , Rats , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...