Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
Sci Rep ; 14(1): 3961, 2024 02 17.
Article in English | MEDLINE | ID: mdl-38368436

ABSTRACT

Lysosomal storage diseases (LSDs) are a group of monogenic diseases characterized by mutations in genes coding for proteins associated with the lysosomal function. Despite the monogenic nature, LSDs patients exhibit variable and heterogeneous clinical manifestations, prompting investigations into epigenetic factors underlying this phenotypic diversity. In this study, we focused on the potential role of epigenetic mechanisms in the pathogenesis of mucopolysaccharidosis IIIB (MPS IIIB) and mucopolysaccharidosis IVA (MPS IVA). We analyzed DNA methylation (5mC) and histone modifications (H3K14 acetylation and H3K9 trimethylation) in MPS IIIB and MPS IVA patients' fibroblasts and healthy controls. The findings revealed that global DNA hypomethylation is present in cell lines for both diseases. At the same time, histone acetylation was increased in MPS IIIB and MPS IVA cells in a donor-dependent way, further indicating a shift towards relaxed open chromatin in these MPS. Finally, the constitutive heterochromatin marker, histone H3K9 trimethylation, only showed reduced clustering in MPS IIIB cells, suggesting limited alterations in heterochromatin organization. These findings collectively emphasize the significance of epigenetic mechanisms in modulating the phenotypic variations observed in LSDs. While global DNA hypomethylation could contribute to the MPS pathogenesis, the study also highlights individual-specific epigenetic responses that might contribute to phenotypic heterogeneity. Further research into the specific genes and pathways affected by these epigenetic changes could provide insights into potential therapeutic interventions for these MPS and other LSDs.


Subject(s)
Mucopolysaccharidosis III , Mucopolysaccharidosis IV , Humans , Mucopolysaccharidosis III/metabolism , Heterochromatin , Histones/genetics , DNA
2.
J Transl Med ; 21(1): 437, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37407981

ABSTRACT

BACKGROUND: Mucopolysaccharidosis IIIC (MPSIIIC) is one of four Sanfilippo diseases sharing clinical symptoms of severe cognitive decline and shortened lifespan. The missing enzyme, heparan sulfate acetyl-CoA: α-glucosaminide-N-acetyltransferase (HGSNAT), is bound to the lysosomal membrane, therefore cannot cross the blood-brain barrier or diffuse between cells. We previously demonstrated disease correction in MPSIIIC mice using an Adeno-Associated Vector (AAV) delivering HGSNAT via intraparenchymal brain injections using an AAV2 derived AAV-truetype (AAV-TT) serotype with improved distribution over AAV9. METHODS: Here, intraparenchymal AAV was delivered in sheep using catheters or Hamilton syringes, placed using Brainlab cranial navigation for convection enhanced delivery, to reduce proximal vector expression and improve spread. RESULTS: Hamilton syringes gave improved AAV-GFP distribution, despite lower vector doses and titres. AAV-TT-GFP displayed moderately better transduction compared to AAV9-GFP but both serotypes almost exclusively transduced neurons. Functional HGSNAT enzyme was detected in 24-37% of a 140g gyrencephalic sheep brain using AAV9-HGSNAT with three injections in one hemisphere. CONCLUSIONS: Despite variabilities in volume and titre, catheter design may be critical for efficient brain delivery. These data help inform a clinical trial for MPSIIIC.


Subject(s)
Mucopolysaccharidosis III , Animals , Acetyltransferases/genetics , Acetyltransferases/metabolism , Brain , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Heparitin Sulfate/metabolism , Mucopolysaccharidoses/genetics , Mucopolysaccharidoses/therapy , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/therapy , Sheep , Genetic Therapy
3.
J Med Chem ; 66(3): 1790-1808, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36696678

ABSTRACT

Sanfilippo syndrome comprises a group of four genetic diseases due to the lack or decreased activity of enzymes involved in heparan sulfate (HS) catabolism. HS accumulation in lysosomes and other cellular compartments results in tissue and organ dysfunctions, leading to a wide range of clinical symptoms including severe neurodegeneration. To date, no approved treatments for Sanfilippo disease exist. Here, we report the ability of N-substituted l-iminosugars to significantly reduce substrate storage and lysosomal dysfunctions in Sanfilippo fibroblasts and in a neuronal cellular model of Sanfilippo B subtype. Particularly, we found that they increase the levels of defective α-N-acetylglucosaminidase and correct its proper sorting toward the lysosomal compartment. Furthermore, l-iminosugars reduce HS accumulation by downregulating protein levels of exostosin glycosyltransferases. These results highlight an interesting pharmacological potential of these glycomimetics in Sanfilippo syndrome, paving the way for the development of novel therapeutic approaches for the treatment of such incurable disease.


Subject(s)
Mucopolysaccharidosis III , Humans , Mucopolysaccharidosis III/drug therapy , Mucopolysaccharidosis III/metabolism , Heparitin Sulfate/metabolism , Lysosomes/metabolism , Fibroblasts/metabolism , Neurons/metabolism
4.
Hum Mol Genet ; 32(3): 417-430, 2023 01 13.
Article in English | MEDLINE | ID: mdl-35997776

ABSTRACT

Mucopolysaccharidosis type IIIB (MPS IIIB) is an autosomal recessive lysosomal storage disease caused by mutations in the gene that encodes the protein N-acetyl-glucosaminidase (NAGLU). Defective NAGLU activity results in aberrant retention of heparan sulfate within lysosomes leading to progressive central nervous system (CNS) degeneration. Intravenous treatment options are limited by the need to overcome the blood-brain barrier and gain successful entry into the CNS. Additionally, we have demonstrated that AAV8 provides a broader transduction area in the MPS IIIB mouse brain compared with AAV5, 9 or rh10. A triple-capsid mutant (tcm) modification of AAV8 further enhanced GFP reporter expression and distribution. Using the MPS IIIB mouse model, we performed a study using either intracranial six site or intracisterna magna injection of AAVtcm8-codon-optimized (co)-NAGLU using untreated MPS IIIB mice as controls to assess disease correction. Disease correction was evaluated based on enzyme activity, heparan sulfate storage levels, CNS lysosomal signal intensity, coordination, activity level, hearing and survival. Both histologic and enzymatic assessments show that each injection method results in supranormal levels of NAGLU expression in the brain. In this study, we have shown correction of lifespan and auditory deficits, increased CNS NAGLU activity and reduced lysosomal storage levels of heparan sulfate following AAVtcm8-coNAGLU administration and partial correction of NAGLU activity in several peripheral organs in the murine model of MPS IIIB.


Subject(s)
Mucopolysaccharidosis III , Animals , Mice , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/therapy , Mucopolysaccharidosis III/metabolism , Capsid/metabolism , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Heparitin Sulfate/metabolism
5.
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
6.
J Biol Chem ; 298(8): 102159, 2022 08.
Article in English | MEDLINE | ID: mdl-35750212

ABSTRACT

Lysosomal storage diseases result in various developmental and physiological complications, including cachexia. To study the causes for the negative energy balance associated with cachexia, we assessed the impact of sulfamidase deficiency and heparan sulfate storage on energy homeostasis and metabolism in a mouse model of type IIIa mucopolysaccharidosis (MPS IIIa, Sanfilippo A syndrome). At 12-weeks of age, MPS IIIa mice exhibited fasting and postprandial hypertriglyceridemia compared with wildtype mice, with a reduction of white and brown adipose tissues. Partitioning of dietary [3H]triolein showed a marked increase in intestinal uptake and secretion, whereas hepatic production and clearance of triglyceride-rich lipoproteins did not differ from wildtype controls. Uptake of dietary triolein was also elevated in brown adipose tissue (BAT), and notable increases in beige adipose tissue occurred, resulting in hyperthermia, hyperphagia, hyperdipsia, and increased energy expenditure. Furthermore, fasted MPS IIIa mice remained hyperthermic when subjected to low temperature but became cachexic and profoundly hypothermic when treated with a lipolytic inhibitor. We demonstrated that the reliance on increased lipid fueling of BAT was driven by a reduced ability to generate energy from stored lipids within the depot. These alterations arose from impaired autophagosome-lysosome fusion, resulting in increased mitochondria content in beige and BAT. Finally, we show that increased mitochondria content in BAT and postprandial dyslipidemia was partially reversed upon 5-week treatment with recombinant sulfamidase. We hypothesize that increased BAT activity and persistent increases in energy demand in MPS IIIa mice contribute to the negative energy balance observed in patients with MPS IIIa.


Subject(s)
Hypertriglyceridemia , Mucopolysaccharidosis III , Adipose Tissue, Brown/metabolism , Animals , Cachexia , Mice , Mitophagy , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/therapy , Triolein
7.
Cardiovasc Pathol ; 60: 107430, 2022.
Article in English | MEDLINE | ID: mdl-35537562

ABSTRACT

PURPOSE: To report two unusual presentations of mucopolysaccharidosis type III (Sanfilippo syndrome) and provide evidence for the cardiac involvement. PATIENTS AND METHODS: We report two siblings with cardiac involvement that were diagnosed in childhood with Sanfilippo A Syndrome (SAS). All patients' diagnosis was confirmed by the excess of heparan sulfate in the urine and the reduction of heparan sulfamidase protein activity. The heart specimens were studied. RESULTS: We report two sibling patients (15-years-old female and 12-years-old female) occurring in sisters both with onset in childhood with no neurological, ophthalmic, hepatic symptoms or coarsening of features as classically described. Both patients underwent bilateral hip arthroplasty in their early 30`s. The older sister had an orthotopic heart transplant because of end-stage heart failure of her cardiomyopathy at the age of 45. She is alive and well. The youngest sister died due to heart failure before a transplantation took place. In the two siblings a thin right ventricular free wall was seen, which triggered the differential diagnosis with arrhythmogenic right ventricular cardiomyopathy or lamin A/C cardiomyopathy. CONCLUSIONS: Early recognition of solitary or mainly cardiac involvement is essential for patients with mucopolysaccharidosis type III (SAS).


Subject(s)
Cardiomyopathies , Heart Failure , Mucopolysaccharidosis III , Adolescent , Female , Heart Failure/etiology , Heparitin Sulfate/metabolism , Humans , Lamin Type A , Mucopolysaccharidosis III/complications , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/pathology
8.
Mol Genet Metab ; 134(4): 323-329, 2021 12.
Article in English | MEDLINE | ID: mdl-34844863

ABSTRACT

Sanfilippo D syndrome (mucopolysaccharidosis type IIID) is a lysosomal storage disorder caused by the deficiency of N-acetylglucosamine-6-sulfatase (GNS). A mouse model was generated by constitutive knockout of the Gns gene. We studied affected mice and controls at 12, 24, 36, and 48 weeks of age for neuropathological markers of disease in the somatosensory cortex, primary motor cortex, ventral posterior nuclei of the thalamus, striatum, hippocampus, and lateral and medial entorhinal cortex. We found significantly increased immunostaining for glial fibrillary associated protein (GFAP), CD68 (a marker of activated microglia), and lysosomal-associated membrane protein-1 (LAMP-1) in Sanfilippo D mice compared to controls at 12 weeks of age in all brain regions. Intergroup differences were marked for GFAP and CD68 staining, with levels in Sanfilippo D mice consistently above controls at all age groups. Intergroup differences in LAMP-1 staining were more pronounced in 12- and 24-week age groups compared to 36- and 48-week groups, as control animals showed some LAMP-1 staining at later timepoints in some brain regions. We also evaluated the somatosensory cortex, medial entorhinal cortex, reticular nucleus of the thalamus, medial amygdala, and hippocampal hilus for subunit c of mitochondrial ATP synthase (SCMAS). We found a progressive accumulation of SCMAS in most brain regions of Sanfilippo D mice compared to controls by 24 weeks of age. Cataloging the regional neuropathology of Sanfilippo D mice may aid in understanding the disease pathogenesis and designing preclinical studies to test brain-directed treatments.


Subject(s)
Brain/pathology , Mucopolysaccharidosis III/pathology , Animals , Female , Gliosis/etiology , Lysosomal Membrane Proteins/analysis , Male , Mice , Microglia/physiology , Mitochondrial Proton-Translocating ATPases/analysis , Mucopolysaccharidosis III/etiology , Mucopolysaccharidosis III/metabolism
9.
Exp Cell Res ; 407(1): 112785, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34411609

ABSTRACT

Mucopolysaccharidosis type IIIB (MPS IIIB) is a lysosomal disease caused by mutations in the NAGLU gene encoding α-N-acetylglucosaminidase (NAGLU) which degrades heparan sulfate in lysosomes. Deficiency in NAGLU results in lysosomal accumulation of glycosaminoglycans (GAGs) and neurological symptoms. Currently, there is no effective treatment or cure for this disease. In this study, induced pluripotent stem cell lines were established from two MPS IIIB patient fibroblast lines and differentiated into neural stem cells and neurons. MPS IIIB neural stem cells exhibited NAGLU deficiency accompanied with GAG accumulation, as well as lysosomal enlargement and secondary lipid accumulation. Treatments with recombinant NAGLU, δ-tocopherol, and 2-hydroxypropyl-b-cyclodextrin significantly reduced the disease phenotypes in these cells. These results indicate the MPS IIIB neural stem cells and neurons have the disease relevant phenotype and can be used as a cell-based disease model system for evaluation of drug efficacy and compound screening for drug development.


Subject(s)
Acetylglucosaminidase/metabolism , Induced Pluripotent Stem Cells/metabolism , Mucopolysaccharidosis III/metabolism , Neural Stem Cells/metabolism , Acetylglucosaminidase/genetics , Cell Differentiation/physiology , Heparitin Sulfate/metabolism , Humans , Lysosomes/metabolism , Mucopolysaccharidosis III/genetics , Neurons/metabolism , Phenotype
10.
Biochim Biophys Acta Mol Cell Res ; 1868(11): 119113, 2021 10.
Article in English | MEDLINE | ID: mdl-34329663

ABSTRACT

Mucopolysaccharidoses (MPSs) are a group of inherited lysosomal storage disorders associated with the deficiency of lysosomal enzymes involved in glycosaminoglycan (GAG) degradation. The resulting cellular accumulation of GAGs is responsible for widespread tissue and organ dysfunctions. The MPS III, caused by mutations in the genes responsible for the degradation of heparan sulfate (HS), includes four subtypes (A, B, C, and D) that present significant neurological manifestations such as progressive cognitive decline and behavioral disorders. The established treatments for the MPS III do not cure the disease but only ameliorate non-neurological clinical symptoms. We previously demonstrated that the natural variant of the hepatocyte growth factor NK1 reduces the lysosomal pathology and reactivates impaired growth factor signaling in fibroblasts from MPS IIIB patients. Here, we show that the recombinant NK1 is effective in rescuing the morphological and functional dysfunctions of lysosomes in a neuronal cellular model of the MPS IIIB. More importantly, NK1 treatment is able to stimulate neuronal differentiation of neuroblastoma SK-NBE cells stable silenced for the NAGLU gene causative of the MPS IIIB. These results provide the basis for the development of a novel approach to possibly correct the neurological phenotypes of the MPS IIIB as well as of other MPSs characterized by the accumulation of HS and progressive neurodegeneration.


Subject(s)
Heparitin Sulfate/metabolism , Models, Biological , Mucopolysaccharidosis III/metabolism , Neurons/metabolism , Binding Sites , Cell Differentiation , Humans , Lysosomes/metabolism , Mucopolysaccharidosis III/pathology , Neurons/pathology , Tumor Cells, Cultured
11.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073041

ABSTRACT

Mucopolysaccharidosis IIIA (MPS IIIA, Sanfilippo syndrome type A), a paediatric neurological lysosomal storage disease, is caused by impaired function of the enzyme N-sulfoglucosamine sulfohydrolase (SGSH) resulting in impaired catabolism of heparan sulfate glycosaminoglycan (HS GAG) and its accumulation in tissues. MPS IIIA represents a significant proportion of childhood dementias. This condition generally leads to patient death in the teenage years, yet no effective therapy exists for MPS IIIA and a complete understanding of the mechanisms of MPS IIIA pathogenesis is lacking. Here, we employ targeted CRISPR/Cas9 mutagenesis to generate a model of MPS IIIA in the zebrafish, a model organism with strong genetic tractability and amenity for high-throughput screening. The sgshΔex5-6 zebrafish mutant exhibits a complete absence of Sgsh enzymatic activity, leading to progressive accumulation of HS degradation products with age. sgshΔex5-6 zebrafish faithfully recapitulate diverse CNS-specific features of MPS IIIA, including neuronal lysosomal overabundance, complex behavioural phenotypes, and profound, lifelong neuroinflammation. We further demonstrate that neuroinflammation in sgshΔex5-6 zebrafish is largely dependent on interleukin-1ß and can be attenuated via the pharmacological inhibition of Caspase-1, which partially rescues behavioural abnormalities in sgshΔex5-6 mutant larvae in a context-dependent manner. We expect the sgshΔex5-6 zebrafish mutant to be a valuable resource in gaining a better understanding of MPS IIIA pathobiology towards the development of timely and effective therapeutic interventions.


Subject(s)
Disease Models, Animal , Hydrolases/genetics , Mucopolysaccharidosis III , Animals , Humans , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/pathology , Mutation , Phenotype , Zebrafish
12.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34156977

ABSTRACT

The majority of patients affected with lysosomal storage disorders (LSD) exhibit neurological symptoms. For mucopolysaccharidosis type IIIC (MPSIIIC), the major burdens are progressive and severe neuropsychiatric problems and dementia, primarily thought to stem from neurodegeneration. Using the MPSIIIC mouse model, we studied whether clinical manifestations preceding massive neurodegeneration arise from synaptic dysfunction. Reduced levels or abnormal distribution of multiple synaptic proteins were revealed in cultured hippocampal and CA1 pyramidal MPSIIIC neurons. These defects were rescued by virus-mediated gene correction. Dendritic spines were reduced in pyramidal neurons of mouse models of MPSIIIC and other (Tay-Sachs, sialidosis) LSD as early as at P10. MPSIIIC neurons also presented alterations in frequency and amplitude of miniature excitatory and inhibitory postsynaptic currents, sparse synaptic vesicles, reduced postsynaptic densities, disorganized microtubule networks, and partially impaired axonal transport of synaptic proteins. Furthermore, postsynaptic densities were reduced in postmortem cortices of human MPS patients, suggesting that the pathology is a common hallmark for neurological LSD. Together, our results demonstrate that lysosomal storage defects cause early alterations in synaptic structure and abnormalities in neurotransmission originating from impaired synaptic vesicular transport, and they suggest that synaptic defects could be targeted to treat behavioral and cognitive defects in neurological LSD patients.


Subject(s)
Lysosomal Storage Diseases/metabolism , Mucopolysaccharidosis III , Pyramidal Cells , Secretory Vesicles/metabolism , Synaptic Transmission/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cells, Cultured , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Disease Progression , Drug Discovery , Hippocampus/pathology , Mice , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/psychology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Protein Transport , Pyramidal Cells/metabolism , Pyramidal Cells/pathology
13.
Nat Commun ; 12(1): 3495, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34108486

ABSTRACT

Lysosomal storage disorders characterized by altered metabolism of heparan sulfate, including Mucopolysaccharidosis (MPS) III and MPS-II, exhibit lysosomal dysfunctions leading to neurodegeneration and dementia in children. In lysosomal storage disorders, dementia is preceded by severe and therapy-resistant autistic-like symptoms of unknown cause. Using mouse and cellular models of MPS-IIIA, we discovered that autistic-like behaviours are due to increased proliferation of mesencephalic dopamine neurons originating during embryogenesis, which is not due to lysosomal dysfunction, but to altered HS function. Hyperdopaminergia and autistic-like behaviours are corrected by the dopamine D1-like receptor antagonist SCH-23390, providing a potential alternative strategy to the D2-like antagonist haloperidol that has only minimal therapeutic effects in MPS-IIIA. These findings identify embryonic dopaminergic neurodevelopmental defects due to altered function of HS leading to autistic-like behaviours in MPS-II and MPS-IIIA and support evidence showing that altered HS-related gene function is causative of autism.


Subject(s)
Autism Spectrum Disorder/metabolism , Dopamine/metabolism , Heparitin Sulfate/metabolism , Lysosomal Storage Diseases/metabolism , Animals , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/pathology , Benzazepines/therapeutic use , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Dopamine Antagonists/therapeutic use , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Heparitin Sulfate/pharmacology , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/pathology , Mesencephalon/drug effects , Mesencephalon/embryology , Mesencephalon/pathology , Mice , Mucopolysaccharidosis III/drug therapy , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/pathology , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/metabolism
14.
Mol Genet Metab ; 133(2): 193-200, 2021 06.
Article in English | MEDLINE | ID: mdl-33962822

ABSTRACT

OBJECTIVE: To quantify changes in segmented brain volumes over 12 months in children with mucopolysaccharidosis types IIIA and IIIB (MPS IIIA and IIIB). METHODS: In order to establish suitable outcome measures for clinical trials, twenty-five children greater than 2 years of age were enrolled in a prospective natural history study of MPS IIIA and IIIB at Nationwide Children's Hospital. Data from sedated non-contrast brain 3 T MRIs and neuropsychological measures were reviewed from the baseline visit and at 12-month follow-up. No intervention beyond standard clinical care was provided. Age- and sex-matched controls were gathered from the National Institute of Mental Health Data Archive. Automated brain volume segmentation with longitudinal processing was performed using FreeSurfer. RESULTS: Of the 25 subjects enrolled with MPS III, 17 children (4 females, 13 males) completed at least one MRI with interpretable volumetric data. The ages ranged from 2.8 to 13.7 years old (average 7.2 years old) at enrollment, including 8 with MPS IIIA and 9 with MPS IIIB. At baseline, individuals with MPS III demonstrated reduced cerebral white matter and corpus callosum volumes, but greater volumes of the lateral ventricles, cerebellar cortex, and cerebellar white matter compared to controls. Among the 13 individuals with MPS III with two interpretable MRIs, there were annualized losses or plateaus in supratentorial brain tissue volumes (cerebral cortex -42.10 ± 18.52 cm3/year [mean ± SD], cerebral white matter -4.37 ± 11.82 cm3/year, subcortical gray matter -6.54 ± 3.63 cm3/year, corpus callosum -0.18 ± 0.62 cm3/yr) and in cerebellar cortex (-0.49 ± 12.57 cm3/year), with a compensatory increase in lateral ventricular volume (7.17 ± 6.79 cm3/year). Reductions in the cerebral cortex and subcortical gray matter were more striking in individuals younger than 8 years of age. Greater cerebral cortex volume was associated with higher fine and gross motor functioning on the Mullen Scales of Early Learning, while greater subcortical gray matter volume was associated with higher nonverbal functioning on the Leiter International Performance Scale. Larger cerebellar cortex was associated with higher receptive language performance on the Mullen, but greater cerebellar white matter correlated with worse adaptive functioning on the Vineland Adaptive Behavioral Scales and visual problem-solving on the Mullen. CONCLUSIONS: Loss or plateauing of supratentorial brain tissue volumes may serve as longitudinal biomarkers of MPS III age-related disease progression compared to age-related growth in typically developing controls. Abnormally increased cerebellar white matter in MPS III, and its association with worse performance on neuropsychological measures, suggest the possibility of pathophysiological mechanisms distinct from neurodegeneration-associated atrophy that warrant further investigation.


Subject(s)
Brain/diagnostic imaging , Learning/physiology , Mucopolysaccharidosis III/diagnostic imaging , Adolescent , Brain/metabolism , Child , Child, Preschool , Corpus Callosum/diagnostic imaging , Corpus Callosum/metabolism , Corpus Callosum/pathology , Female , Humans , Longitudinal Studies , Magnetic Resonance Imaging , Male , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/pathology , White Matter/diagnostic imaging , White Matter/metabolism , White Matter/pathology
15.
Cells ; 10(4)2021 04 08.
Article in English | MEDLINE | ID: mdl-33918094

ABSTRACT

Mucopolysaccharidosis IIIA (MPS IIIA) is a lysosomal storage disease with significant neurological and skeletal pathologies. Respiratory dysfunction is a secondary pathology contributing to mortality in MPS IIIA patients. Pulmonary surfactant is crucial to optimal lung function and has not been investigated in MPS IIIA. We measured heparan sulphate (HS), lipids and surfactant proteins (SP) in pulmonary tissue and bronchoalveolar lavage fluid (BALF), and surfactant activity in healthy and diseased mice (20 weeks of age). Heparan sulphate, ganglioside GM3 and bis(monoacylglycero)phosphate (BMP) were increased in MPS IIIA lung tissue. There was an increase in HS and a decrease in BMP and cholesteryl esters (CE) in MPS IIIA BALF. Phospholipid composition remained unchanged, but BALF total phospholipids were reduced (49.70%) in MPS IIIA. There was a reduction in SP-A, -C and -D mRNA, SP-D protein in tissue and SP-A, -C and -D protein in BALF of MPS IIIA mice. Captive bubble surfactometry showed an increase in minimum and maximum surface tension and percent surface area compression, as well as a higher compressibility and hysteresis in MPS IIIA surfactant upon dynamic cycling. Collectively these biochemical and biophysical changes in alveolar surfactant are likely to be detrimental to lung function in MPS IIIA.


Subject(s)
Heparitin Sulfate/metabolism , Mucopolysaccharidosis III/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Surfactants/metabolism , Animals , Biophysical Phenomena , Bronchoalveolar Lavage Fluid , Cholesterol/metabolism , Chromatography, Liquid , G(M3) Ganglioside/metabolism , Gene Expression Regulation , Lysophospholipids/metabolism , Mice, Inbred C57BL , Monoglycerides/metabolism , Phospholipids/metabolism , Reference Standards , Tandem Mass Spectrometry
16.
Mol Pharm ; 18(1): 214-227, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33320673

ABSTRACT

There is currently no cure or effective treatment available for mucopolysaccharidosis type IIID (MPS IIID, Sanfilippo syndrome type D), a lysosomal storage disorder (LSD) caused by the deficiency of α-N-acetylglucosamine-6-sulfatase (GNS). The clinical symptoms of MPS IIID, like other subtypes of Sanfilippo syndrome, are largely localized to the central nervous system (CNS), and any treatments aiming to ameliorate or reverse the catastrophic and fatal neurologic decline caused by this disease need to be delivered across the blood-brain barrier. Here, we report a proof-of-concept enzyme replacement therapy (ERT) for MPS IIID using recombinant human α-N-acetylglucosamine-6-sulfatase (rhGNS) via intracerebroventricular (ICV) delivery in a neonatal MPS IIID mouse model. We overexpressed and purified rhGNS from CHO cells with a specific activity of 3.9 × 104 units/mg protein and a maximal enzymatic activity at lysosomal pH (pH 5.6), which was stable for over one month at 4 °C in artificial cerebrospinal fluid (CSF). We demonstrated that rhGNS was taken up by MPS IIID patient fibroblasts via the mannose 6-phosphate (M6P) receptor and reduced intracellular glycosaminoglycans to normal levels. The delivery of 5 µg of rhGNS into the lateral cerebral ventricle of neonatal MPS IIID mice resulted in normalization of the enzymatic activity in brain tissues; rhGNS was found to be enriched in lysosomes in MPS IIID-treated mice relative to the control. Furthermore, a single dose of rhGNS was able to reduce the accumulated heparan sulfate and ß-hexosaminidase. Our results demonstrate that rhGNS delivered into CSF is a potential therapeutic option for MPS IIID that is worthy of further development.


Subject(s)
Mucopolysaccharidosis III/drug therapy , Recombinant Proteins/pharmacology , Sulfatases/pharmacology , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , CHO Cells , Cricetulus , Disease Models, Animal , Enzyme Replacement Therapy/methods , Glycosaminoglycans/metabolism , Heparitin Sulfate/metabolism , Humans , Liver/drug effects , Liver/metabolism , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mucopolysaccharidosis III/metabolism , Neurons/drug effects , Neurons/metabolism , Receptor, IGF Type 2/metabolism
17.
Acta Neuropathol Commun ; 8(1): 194, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33203474

ABSTRACT

Sanfilippo syndrome is an untreatable form of childhood-onset dementia. Whilst several therapeutic strategies are being evaluated in human clinical trials including i.v. delivery of AAV9-based gene therapy, an urgent unmet need is the availability of non-invasive, quantitative measures of neurodegeneration. We hypothesise that as part of the central nervous system, the retina may provide a window through which to 'visualise' degenerative lesions in brain and amelioration of them following treatment. This is reliant on the age of onset and the rate of disease progression being equivalent in retina and brain. For the first time we have assessed in parallel, the nature, age of onset and rate of retinal and brain degeneration in a mouse model of Sanfilippo syndrome. Significant accumulation of heparan sulphate and expansion of the endo/lysosomal system was observed in both retina and brain pre-symptomatically (by 3 weeks of age). Robust and early activation of micro- and macroglia was also observed in both tissues. There was substantial thinning of retina and loss of rod and cone photoreceptors by ~ 12 weeks of age, a time at which cognitive symptoms are noted. Intravenous delivery of a clinically relevant AAV9-human sulphamidase vector to neonatal mice prevented disease lesion appearance in retina and most areas of brain when assessed 6 weeks later. Collectively, the findings highlight the previously unrecognised early and significant involvement of retina in the Sanfilippo disease process, lesions that are preventable by neonatal treatment with AAV9-sulphamidase. Critically, our data demonstrate for the first time that the advancement of retinal disease parallels that occurring in brain in Sanfilippo syndrome, thus retina may provide an easily accessible neural tissue via which brain disease development and its amelioration with treatment can be monitored.


Subject(s)
Brain/pathology , Mucopolysaccharidosis III/pathology , Neurodegenerative Diseases/pathology , Retina/pathology , Retinal Degeneration/pathology , Animals , Asymptomatic Diseases , Disease Models, Animal , Endosomes/pathology , Genetic Therapy , Heparitin Sulfate/metabolism , Humans , Hydrolases/genetics , Lysosomes/pathology , Mice , Microglia/pathology , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/metabolism , Neurodegenerative Diseases/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/pathology
18.
Sci Rep ; 10(1): 20365, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33230178

ABSTRACT

Mucopolysaccharidosis type IIIB (MPS IIIB; Sanfilippo syndrome B) is an autosomal recessive lysosomal storage disorder caused by the deficiency of alpha-N-acetylglucosaminidase activity, leading to increased levels of nondegraded heparan sulfate (HS). A mouse model has been useful to evaluate novel treatments for MPS IIIB, but has limitations. In this study, we evaluated the naturally occurring canine model of MPS IIIB for the onset and progression of biochemical and neuropathological changes during the preclinical stages (onset approximately 24-30 months of age) of canine MPS IIIB disease. Even by 1 month of age, MPS IIIB dogs had elevated HS levels in brain and cerebrospinal fluid. Analysis of histopathology of several disease-relevant regions of the forebrain demonstrated progressive lysosomal storage and microglial activation despite a lack of cerebrocortical atrophy in the oldest animals studied. More pronounced histopathology changes were detected in the cerebellum, where progressive lysosomal storage, astrocytosis and microglial activation were observed. Microglial activation was particularly prominent in cerebellar white matter and within the deep cerebellar nuclei, where neuron loss also occurred. The findings in this study will form the basis of future assessments of therapeutic efficacy in this large animal disease model.


Subject(s)
Acetylglucosaminidase/deficiency , Cerebellum/pathology , Cerebral Cortex/pathology , Dog Diseases/pathology , Mucopolysaccharidosis III/pathology , Prosencephalon/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cerebellum/metabolism , Cerebral Cortex/metabolism , Disease Models, Animal , Disease Progression , Dog Diseases/metabolism , Dogs , Female , Heparitin Sulfate/metabolism , Histocytochemistry , Humans , Lysosomes/metabolism , Lysosomes/pathology , Male , Microglia/metabolism , Microglia/pathology , Mucopolysaccharidosis III/metabolism , Neurons/metabolism , Neurons/pathology , Prosencephalon/metabolism , White Matter/metabolism , White Matter/pathology
19.
Mol Genet Metab ; 131(1-2): 197-205, 2020.
Article in English | MEDLINE | ID: mdl-32739280

ABSTRACT

The cause of neurodegeneration in MPS mouse models is the focus of much debate and what the underlying cause of disease pathology in MPS mice is. The timing of development of pathology and when this can be reversed or impacted is the key to developing suitable therapies in MPS. This study is the first of its kind to correlate the biochemical changes with the functional outcome as assessed using non-invasive behaviour testing across multiple mucopolysaccharidosis (MPS) mouse models. In the MPS brain, the primary lysosomal enzyme dysfunction leads to accumulation of primary glycosaminoglycans (GAGs) with gangliosides (GM2 and GM3) being the major secondary storage products. With a focus on the neuropathology, a time course experiment was conducted in MPS I, MPS IIIA, MPS VII (severe and attenuated models) in order to understand the relative timing and level of GAG and ganglioside accumulation and how this correlates to behaviour deficits. Time course analysis from 1 to 6 months of age was conducted on brain samples to assess primary GAG (uronic acid), ß-hexosaminidase enzyme activity and levels of GM2 and GM3 gangliosides. This was compared to a battery of non-invasive behaviour tests including open field, inverted grid, rotarod and water cross maze were assessed to determine effects on motor function, activity and learning ability. The results show that the GAG and ganglioside accumulation begins prior to the onset of detectable changes in learning ability and behaviour. Interestingly, the highest levels of GAG and ganglioside accumulation was observed in the MPS IIIA mouse despite having 3% residual enzyme activity. Deficits in motor function were clearly observed in the severe Gusmps/mps, which were significantly delayed in the attenuated Gustm(L175F)Sly model despite their minimal increase in detectable enzyme activity. This suggests that genotype and residual enzyme activity are not indicative of severity of disease pathology in MPS disease and there exists a window when there are considerable storage products without detectable functional deficits which may allow an alteration to occur with therapy.


Subject(s)
Brain/metabolism , Glucuronidase/genetics , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis VII/metabolism , Animals , Brain/pathology , Disease Models, Animal , G(M2) Ganglioside/genetics , G(M2) Ganglioside/metabolism , G(M3) Ganglioside/genetics , G(M3) Ganglioside/metabolism , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Heparitin Sulfate/metabolism , Humans , Male , Maze Learning/physiology , Mice , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/pathology , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/pathology
20.
Int J Mol Sci ; 21(12)2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32545699

ABSTRACT

Mucopolysaccharidoses (MPSs) are inherited disorders of the glycosaminoglycan (GAG) metabolism. The defective digestion of GAGs within the intralysosomal compartment of affected patients leads to a broad spectrum of clinical manifestations ranging from cardiovascular disease to neurological impairment. The molecular mechanisms underlying the progression of the disease downstream of the genetic mutation of genes encoding for lysosomal enzymes still remain unclear. Here, we applied a targeted metabolomic approach to a mouse model of PS IIIB, using a platform dedicated to the diagnosis of inherited metabolic disorders, in order to identify amino acid and fatty acid metabolic pathway alterations or the manifestations of other metabolic phenotypes. Our analysis highlighted an increase in the levels of branched-chain amino acids (BCAAs: Val, Ile, and Leu), aromatic amino acids (Tyr and Phe), free carnitine, and acylcarnitines in the liver and heart tissues of MPS IIIB mice as compared to the wild type (WT). Moreover, Ala, Met, Glu, Gly, Arg, Orn, and Cit amino acids were also found upregulated in the liver of MPS IIIB mice. These findings show a specific impairment of the BCAA and fatty acid catabolism in the heart of MPS IIIB mice. In the liver of affected mice, the glucose-alanine cycle and urea cycle resulted in being altered alongside a deregulation of the BCAA metabolism. Thus, our data demonstrate that an accumulation of BCAAs occurs secondary to lysosomal GAG storage, in both the liver and the heart of MPS IIIB mice. Since BCAAs regulate the biogenesis of lysosomes and autophagy mechanisms through mTOR signaling, impacting on lipid metabolism, this condition might contribute to the progression of the MPS IIIB disease.


Subject(s)
Liver/chemistry , Metabolomics/methods , Mucopolysaccharidosis III/metabolism , Myocardium/chemistry , Amino Acids, Aromatic/analysis , Amino Acids, Branched-Chain/analysis , Animals , Carnitine/analogs & derivatives , Carnitine/analysis , Disease Models, Animal , Humans , Lipid Metabolism , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...