Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 250
Filter
1.
Neuropathol Appl Neurobiol ; 45(7): 715-731, 2019 12.
Article in English | MEDLINE | ID: mdl-30907009

ABSTRACT

BACKGROUND: Sanfilippo syndrome (mucopolysaccharidosis type IIIA; MPS IIIA) is an inherited paediatric-onset neurodegenerative disorder caused by the lysosomal deficiency of sulphamidase with subsequent accumulation of heparan sulphate. The pathological mechanisms responsible for clinical disease are unknown; however, intraneuronal accumulation of aggregation-prone proteins such as α-synuclein, phosphorylated tau and amyloid precursor protein suggests inefficient intracellular trafficking and lysosomal degradation. AIM: To investigate the contribution the accumulating α-synuclein plays in early symptom emergence that is, impaired cognition, reduced anxiety and motor deficits, first detectable between 3-5 months of age. METHODS: We have crossed congenic MPS IIIA mice with α-synuclein-deficient (Sncatm1Rosl /J) mice and evaluated phenotype and brain disease lesions. RESULTS: In a battery of behavioural tests performed on mice aged 12-22 weeks, we were unable to differentiate α-synuclein-deficient MPS IIIA mice from those with one or both copies of the α-synuclein gene; all three affected genotypes were significantly impaired in test performance when compared to wild-type littermates. Histological studies revealed that the rate, location and nature of deposition of other proteinaceous lesions, the disruption to endolysosomal protein expression and the inflammatory response seen in the brain of α-synuclein-deficient MPS IIIA mice reflected that seen in MPS IIIA mice homo- or heterozygous for α-synuclein. CONCLUSION: Deletion and/or deficiency of α-synuclein does not influence clinical and neuropathological disease progression in murine MPS IIIA, demonstrating that in and of itself, this protein does not initiate the cognitive and motor symptoms that occur in the first 5 months of life in MPS IIIA mice.


Subject(s)
Brain/pathology , Mucopolysaccharidosis III/genetics , alpha-Synuclein/genetics , Animals , Disease Models, Animal , Disease Progression , Mice , Mice, Knockout , Mucopolysaccharidosis III/pathology
2.
J Inherit Metab Dis ; 36(2): 385-94, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22669363

ABSTRACT

Characteristic cardiac valve abnormalities and left ventricular hypertrophy are present in untreated patients with mucopolysaccharidosis type VI (MPS VI). Cardiac ultrasound was performed to investigate these findings in subjects during long-term enzyme replacement therapy (ERT) with recombinant human arylsulfatase B (rhASB, rhN-acetylgalactosamine 4-sulfatase, galsulfase, Naglazyme®). Studies were conducted in 54 subjects before ERT was begun and at specific intervals for up to 96 weeks of weekly infusions of rhASB at 1 mg/kg during phase 1/2, phase 2, and phase 3 trials of rhASB. At baseline, mitral and aortic valve obstruction was present and was significantly greater in those ≥12 years of age. Mild mitral and trace aortic regurgitation were present, the former being significantly greater in those <12 years. Left ventricular hypertrophy, with averaged z-scores ranging from 1.6-1.9 SD greater than normal, was present for ages both <12 and ≥12 years. After 96 weeks of ERT, ventricular septal hypertrophy regressed in those <12 years. For those ≥12 years, septal hypertrophy was unchanged, and aortic regurgitation increased statistically but not physiologically. Obstructive gradients across mitral and aortic valves remained unchanged. The results suggest that long-term ERT is effective in reducing intraventricular septal hypertrophy and preventing progression of cardiac valve abnormalities when administered to those <12 years of age.


Subject(s)
Enzyme Replacement Therapy/methods , Heart Valves/drug effects , Hypertrophy, Left Ventricular/chemically induced , Mucopolysaccharidosis VI/drug therapy , N-Acetylgalactosamine-4-Sulfatase/adverse effects , N-Acetylgalactosamine-4-Sulfatase/therapeutic use , Adolescent , Adult , Child , Clinical Trials as Topic , Enzyme Replacement Therapy/adverse effects , Female , Humans , Male , Randomized Controlled Trials as Topic , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Treatment Outcome , Young Adult
3.
Neuroscience ; 227: 110-8, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23022219

ABSTRACT

Mucopolysaccharidosis IIIA (MPS IIIA) is a lysosomal storage disorder caused by a deficiency in the activity of the lysosomal hydrolase, sulphamidase, an enzyme involved in the degradation of heparan sulphate. MPS IIIA patients exhibit progressive mental retardation and behavioural disturbance. While neuropathology is the major clinical problem in MPS IIIA patients, there is little understanding of how lysosomal storage generates this phenotype. As reduced neuronal communication can underlie cognitive deficiencies, we investigated whether the secretion of neurotransmitters is altered in MPS IIIA mice; utilising adrenal chromaffin cells, a classical model for studying secretion via exocytosis. MPS IIIA chromaffin cells displayed heparan sulphate storage and electron microscopy revealed large electron-lucent storage compartments. There were also increased numbers of large/elongated chromaffin granules, with a morphology that was similar to immature secretory granules. Carbon fibre amperometry illustrated a significant decrease in the number of exocytotic events for MPS IIIA, when compared to control chromaffin cells. However, there were no changes in the kinetics of release, the amount of catecholamine released per exocytotic event, or the amount of Ca(2+) entry upon stimulation. The increased number of large/elongated granules and reduced number of exocytotic events suggests that either the biogenesis and/or the cell surface docking and fusion potential of these vesicles is impaired in MPS IIIA. If this also occurs in central nervous system neurons, the reduction in neurotransmitter release could help to explain the development of neuropathology in MPS IIIA.


Subject(s)
Chromaffin Cells/physiology , Exocytosis/genetics , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Adrenal Glands/metabolism , Adrenal Glands/pathology , Adrenal Glands/ultrastructure , Analysis of Variance , Animals , Calcium/metabolism , Carbon , Carbon Fiber , Catecholamines/metabolism , Cells, Cultured , Chromaffin Cells/ultrastructure , Disease Models, Animal , Heparitin Sulfate/metabolism , Lysosomes/metabolism , Lysosomes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Statistics, Nonparametric
4.
Neurochem Res ; 37(6): 1372-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22484966

ABSTRACT

Allogenic stem cell transplantation can reduce lysosomal storage of heparan sulfate-derived oligosaccharides by up to 27 % in Sanfilippo MPS3a brain, but does not reduce the abnormal storage of sialolactosylceramide (G(M3)) or improve neurological symptoms, suggesting that ganglioside storage is in a non-lysosomal compartment. To investigate this further we isolated the Triton X100-insoluble at 4 °C, lipid raft (LR) fraction from a sucrose-density gradient from cerebral hemispheres of a 7 month old mouse model of Sanfilippo MPS3a and age-matched control mouse brain. HPLC/MS/MS analysis revealed the expected enrichment of normal complex gangliosides, ceramides, galatosylceramides and sphingomyelin enrichment in this LR fraction. The abnormal HS-derived oligosaccharide storage material was in the Triton X100-soluble at 4 °C fractions (8-12),whereas both GM3 and sialo[GalNAc]lactosylceramide (GM2) were found exclusively in the LR fraction (fractions 3 and 4) and were >90 % C18:0 fatty acid, suggesting a neuronal origin. Further analysis also revealed a >threefold increase in the late-endosome marker bis (monoacylglycerol) phosphate (>70 % as C22:6/22:6-BMP) in non-LR fractions 8-12 whereas different forms of the proposed BMP precursor, phosphatidylglycerol (PG) were in both LR and non-LR fractions and were less elevated in MPS3a brain. Thus heparan sulfate-derived oligosaccharide storage is associated with abnormal lipid accumulation in both lysosomal (BMP) and non-lysosomal (GM3 and GM2) compartments.


Subject(s)
Brain/metabolism , G(M3) Ganglioside/metabolism , Gangliosides/metabolism , Membrane Microdomains/metabolism , Mucopolysaccharidosis III/metabolism , Animals , G(M2) Ganglioside/metabolism , Lysophospholipids/metabolism , Lysosomes/metabolism , Mice , Monoglycerides/metabolism , Tandem Mass Spectrometry
5.
N Z Vet J ; 60(3): 183-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22329490

ABSTRACT

AIM: To investigate and characterise an inborn error of metabolism in a dog with skeletal and ocular abnormalities. METHODS: A 2.5-year-old small male Miniature Poodle-like dog was presented with gross joint laxity and bilateral corneal opacities. Clinical examination was augmented by routine haematology, serum chemistry, radiographs, pathology, enzymology and molecular genetic studies. Euthanasia was requested when the dog was 3 years of age because of progressively decreasing quality of life. RESULTS: Radiology revealed generalised epiphyseal dysplasia, malformed vertebral bodies, luxation/subluxation of appendicular and lumbosacral joints with hypoplasia of the odontoid process and hyoid apparatus. These clinical and radiographic findings, together with a positive urinary Berry spot test for mucopolysaccharides, and metachromatic granules in leucocytes, were indicative of a mucopolysaccharidosis (MPS), a lysosomal storage disease. Histological lesions included vacuolation of stromal cells of the cornea, fibroblasts, chondrocytes, macrophages and renal cells. The brain was essentially normal except for moderate secondary Wallerian-type degeneration in motor and sensory tracts of the hind brain. Dermatan sulphate-uria was present and enzymology revealed negligible activity of N-acetylgalactosamine-4-sulphatase, also known as arylsulphatase B, in cultured fibroblasts and liver tissue. A novel homozygous 22 base pair (bp) deletion in exon 1 of this enzyme's gene was identified (c.103_124del), which caused aframe-shift and subsequent premature stop codon. The "Wisdom pure breed-mixed breed" test reported the dog as a cross between a Miniature and Toy Poodle. CONCLUSIONS: The clinicopathological features are similar to those of MPS type VI as previously described in dogs, cats and other species, and this clinical diagnosis was confirmed by enzymology and molecular genetic studies. This is an autosomal recessively inherited lysosomal storage disease. CLINICAL RELEVANCE: The prevalence of MPS VI in Miniature or Toy Poodles in New Zealand and elsewhere is currently unknown. Due to the congenital nature of the disorder, malformed pups may be subject to euthanasia without investigation and the potential genetic problem in the breed may not be fully recognised. The establishment of a molecular genetic test now permits screening for this mutation as a basis to an informed breeding policy.


Subject(s)
Dog Diseases/genetics , Mucopolysaccharidosis IV/veterinary , N-Acetylgalactosamine-4-Sulfatase/genetics , Amino Acid Sequence , Animals , Base Sequence , Dog Diseases/pathology , Dogs , Gene Deletion , Gene Expression Regulation, Enzymologic , Male , Mucopolysaccharidosis IV/genetics , Mucopolysaccharidosis IV/pathology
6.
Clin Genet ; 77(5): 492-8, 2010 May.
Article in English | MEDLINE | ID: mdl-19968667

ABSTRACT

Mucopolysaccharidosis type VI (MPS VI) is a progressive, multisystem disorder caused by a deficiency of the lysosomal enzyme N-acetylgalactosamine-4-sulphatase (ASB). Enzyme replacement therapy (ERT) has been shown to clinically benefit affected individuals greater than 6 years of age. This case control study of affected siblings assessed the safety, efficacy and benefits of ERT in children less than 5 years of age. Siblings, aged 8 weeks and 3.6 years, were treated weekly with 1 mg/kg recombinant human N-acetylgalactosamine-4-sulphatase (rhASB) with an end-point of 3.6 years. Clinical and biochemical parameters were monitored to assess the benefits of ERT. The treatment was well tolerated by both siblings. In the younger sibling, ERT was associated with the absence of the development of scoliosis and preserved joint movement, cardiac valves and facial morphology. The older sibling had a marked improvement in joint mobility and cardiac valve pathology and scoliosis slowed or stabilized. Corneal clouding and progressive skeletal changes were observed despite treatment. This study demonstrated a clear benefit of early initiation of ERT to slow or prevent the development of significant pathological changes of MPS VI. These results indicate that the earlier ERT is started, the greater the response.


Subject(s)
Enzyme Replacement Therapy , Mucopolysaccharidosis VI/therapy , Siblings , Case-Control Studies , Child, Preschool , Female , Glycosaminoglycans/urine , Humans , Infant , Infant, Newborn , Male , Mucopolysaccharidosis VI/complications , Mucopolysaccharidosis VI/physiopathology , Mucopolysaccharidosis VI/urine , N-Acetylgalactosamine-4-Sulfatase/therapeutic use , Pregnancy , Range of Motion, Articular/physiology , Scoliosis/complications
7.
Mol Genet Metab ; 99(2): 142-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19815439

ABSTRACT

Metachromatic leukodystrophy is a neurodegenerative disease that is characterized by a deficiency of arylsulfatase A, resulting in the accumulation of sulfatide and other lipids in the lysosomal network of affected cells. Accumulation of sulfatide in the nervous system leads to severe impairment of neurological function with a fatal outcome. Prognosis is often poor unless treatment is carried out before the onset of clinical symptoms. Pre-symptomatic detection of affected individuals may be possible with the introduction of newborn screening programs. The ability to accurately predict clinical phenotype and rate of disease progression in asymptomatic individuals will be essential to assist selection of the most appropriate treatment strategy. Biochemical profiling, incorporating the determination of residual enzyme protein/activity using immune-based assays, and metabolite profiling using electrospray ionization-tandem mass spectrometry, was performed on urine and cultured skin fibroblasts from a cohort of patients representing the clinical spectrum of metachromatic leukodystrophy and on unaffected controls. Residual enzyme protein/activity in fibroblasts was able to differentiate unaffected controls, arylsulfatase A pseudo-deficient individuals, pseudo-deficient compound heterozygotes and affected patients. Metachromatic leukodystrophy phenotypes were distinguished by quantification of sulfatide and other secondarily altered lipids in urine and skin fibroblasts; this enabled further differentiation of the late-infantile form of the disorder from the juvenile and adult forms. Prediction of the rate of disease progression for metachromatic leukodystrophy requires a combination of information on genotype, residual arylsulfatase A protein and activity and the measurement of sulfatide and other lipids in urine and cultured skin fibroblasts.


Subject(s)
Leukodystrophy, Metachromatic/diagnosis , Leukodystrophy, Metachromatic/pathology , Severity of Illness Index , Adolescent , Adult , Case-Control Studies , Cell Line , Cerebroside-Sulfatase/deficiency , Child , Child, Preschool , Female , Fibroblasts/enzymology , Fibroblasts/pathology , Heterozygote , Humans , Infant , Leukodystrophy, Metachromatic/enzymology , Leukodystrophy, Metachromatic/urine , Lysophospholipids/metabolism , Male , Middle Aged , Monoglycerides/metabolism , Skin/metabolism , Skin/pathology , Sulfoglycosphingolipids/urine
8.
Int J Clin Pharmacol Ther ; 47 Suppl 1: S118-23, 2009.
Article in English | MEDLINE | ID: mdl-20040322

ABSTRACT

Patients with lysosomal storage diseases (LSDs) have a greatly diminished lifespan and reduced quality of life, particularly those with neurological manifestations. There are few therapeutic options available to treat the neurological signs and symptoms of LSDs. It is, therefore, imperative that efficacious and tolerable treatments are developed. Hematopoietic stem cell transplantation is carried out in some LSDs in which there is neurological involvement. However, this approach is associated with significant morbidity and mortality, and not all patients who receive this treatment exhibit improvements in cognitive signs and symptoms. A growing body of research in animal models of LSDs appears to support the efficacy of repeated delivery of recombinant lysosomal proteins via injection into the cerebrospinal fluid (CSF). Studies in dogs with mucopolysaccharidosis (MPS) Type 1 have shown that this approach enables widespread distribution of the recombinant protein within the brain, leading to a reduction in LSD pathology. Subsequent studies in MPS IIIA mice revealed that this strategy was also effective in ameliorating neuropathology and improving clinical signs in these animals. More recent studies in mice with Krabbe disease or a late infantile form of neuronal ceroid lipofuscinosis have demonstrated that delivery of recombinant proteins into the CSF may be efficacious in reducing disease pathology and neurological signs and symptoms. Whilst there are still important issues that need to be addressed, such as humoral immune responses to therapeutic protein administration and dose/ frequency selection, this approach represents a medium-term option for treating these devastating conditions. This review summarizes some of the findings and challenges ahead.


Subject(s)
Enzyme Replacement Therapy/methods , Lysosomal Storage Diseases/drug therapy , Neurodegenerative Diseases/therapy , Recombinant Proteins/administration & dosage , Animals , Disease Models, Animal , Humans , Injections, Intraventricular , Injections, Spinal , Lysosomal Storage Diseases/complications , Neurodegenerative Diseases/complications , Proteins/metabolism , Proteins/therapeutic use
9.
Genes Brain Behav ; 7(7): 740-53, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18518922

ABSTRACT

Mucopolysaccharidosis type IIIA (MPS IIIA) is an inherited neurodegenerative lysosomal storage disorder characterised by progressive loss of learned skills, sleep disturbance and behavioural problems. Reduced activity of sulphamidase (SGSH; EC 3.10.1.1) results in intracellular accumulation of heparan sulphate (HS), with the brain the primary site of pathology. We have used a naturally-occurring MPS IIIA mouse model to determine the effectiveness of SGSH replacement via the cerebrospinal fluid (CSF) to decrease neuropathology. This is a potential therapeutic option for patients with this disorder. Mice received intra-CSF injections of recombinant human SGSH (30, 50 or 70 mug) fortnightly from six-18 weeks of age, and the cumulative effect on neuropathology was examined and quantified. Anti-SGSH antibodies detected in plasma at euthanasia did not appear to impact upon the health of the mice or the experimental outcome, with significant, but region- and dose-dependent reductions in an HS-derived oligosaccharide observed in the brain and spinal cord using tandem mass spectrometry. SGSH infusion reduced the number of storage inclusions observed in the brain when visualised using electron microscopy and this correlated with a significant decrease in the immunohistochemical staining of a lysosomal membrane marker (LIMP-II). Reduced numbers of activated isolectin-B4-positive microglia and GFAP-positive astrocytes were seen in many, but not all, brain regions. Significant reductions in the number of ubiquitin-positive intracellular inclusions were also observed. These outcomes demonstrate the effectiveness of this method of enzyme delivery in reducing the spectrum of neuropathological changes in murine MPS IIIA brain.


Subject(s)
Brain/drug effects , Hydrolases/administration & dosage , Hydrolases/pharmacology , Mucopolysaccharidosis III/drug therapy , Animals , Brain/pathology , Disease Models, Animal , Heparitin Sulfate/pharmacology , Humans , Male , Mice, Transgenic , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Nervous System Diseases
10.
Cell Mol Neurobiol ; 28(7): 949-59, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18297392

ABSTRACT

In order to evaluate the mechanisms leading to neuropathology in Mucopolysaccharidosis type IIIA (MPS-IIIA, Sanfilippo syndrome), we have harvested and cultured primary neural cells isolated from the cerebellum of newborn and adult MPS-IIIA and unaffected mice. Cell viability and plating efficiency were comparable for brain tissue obtained from either newborn or adult MPS-IIIA and unaffected mice. Cultures (newborn and adult) comprised a mixed brain cell population including astrocytes, oligodendrocytes, and neurons. Newborn MPS-IIIA cells contained inclusions and vacuoles consistent with the pathology present in affected brain tissue. Newborn and adult MPS-IIIA brain cells had approximately 5-7% of the sulfamidase activity present in primary neural cells cultured from unaffected newborn and adult mice. In addition, high levels of glucosamine-N-sulfate[alpha-1,4]hexuronic acid, a heparan sulfate-derived disaccharide, were detected in both newborn and adult MPS-IIIA brain cells. These results suggest that the primary MPS-IIIA brain cells exhibit characteristics of MPS-IIIA phenotype at the histopathological and biochemical level in culture.


Subject(s)
Cerebellum/pathology , Mucopolysaccharidosis III/pathology , Neuroglia/pathology , Neurons/pathology , Age Factors , Aging/metabolism , Aging/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Culture Techniques , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , Cerebellum/metabolism , Cerebellum/physiopathology , Disaccharides/metabolism , Disease Models, Animal , Hexuronic Acids/metabolism , Hydrolases/metabolism , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/physiopathology , Neuroglia/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phenotype , Vacuoles/metabolism , Vacuoles/pathology
11.
Behav Brain Res ; 186(2): 176-84, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-17889945

ABSTRACT

alpha-Mannosidosis is a lysosomal storage disorder resulting from a functional deficiency of the lysosomal enzyme alpha-mannosidase. This deficiency results in the accumulation of various oligosaccharides in the lysosomes of affected individuals, causing somatic pathology and progressive neurological degeneration that results in cognitive deficits, ataxia, and other neurological symptoms. We have a naturally occurring guinea pig model of this disease which exhibits a deficiency of lysosomal alpha-mannosidase and has a similar clinical presentation to human alpha-mannosidosis. Various tests were developed in the present study to characterise and quantitate the loss of neurological function in alpha-mannosidosis guinea pigs and to follow closely the progression of the disease. General neurological examinations showed progressive differences in alpha-mannosidosis animals from approximately 1 month of age. Significant differences were observed in hind limb gait width from 2 months of age and significant cognitive (memory and learning) deficits were observed from 3 months of age. Evoked response tests showed an increase in somatosensory P1 peak latency in alpha-mannosidosis guinea pigs from approximately 2 months of age, as well as progressive hearing loss using auditory brainstem evoked responses. The alpha-mannosidosis guinea pig therefore appears to exhibit many of the characteristics of the human disease, and will be useful in evaluating therapies for treatment of central nervous system pathology.


Subject(s)
Behavior, Animal/physiology , alpha-Mannosidosis/physiopathology , alpha-Mannosidosis/psychology , Acoustic Stimulation/methods , Age Factors , Animals , Disease Models, Animal , Disease Progression , Electric Stimulation/methods , Electroencephalography , Evoked Potentials, Auditory, Brain Stem/physiology , Female , Gait/physiology , Guinea Pigs , Male , Maze Learning/physiology , Neurologic Examination , Reaction Time , Sex Factors , alpha-Mannosidase/deficiency , alpha-Mannosidosis/genetics
12.
Neuropathol Appl Neurobiol ; 33(5): 572-85, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17854439

ABSTRACT

Alpha-mannosidosis is an inherited metabolic disorder characterized by a reduction in alpha-D-mannosidase and intralysosomal accumulation of undegraded mannose-containing oligosaccharides. The alpha-mannosidosis guinea pig exhibits pathological similarities to its human counterpart, which make it a valuable animal model. To trace the progression of alpha-mannosidosis during foetal development, brain and visceral organs from affected and unaffected guinea pigs at 30, 36, 38, 51 and 65 days of gestation (dg) were examined by light and electron microscopy (term: approximately 68 dg). In the affected brain, distended lysosomes (vacuoles) were scarce up to 38 dg and were seen in few differentiating neuronal cells but mostly in macrophages, pericytes and endothelial cells. At 51 and 65 dg, several vacuoles were observed in some neurones, in many Purkinje cells, pericytes, endothelial and microglial cells, and in few cerebellar internal granule cells. Myelination had started by 51 dg. Non-myelinated axonal spheroids were detected in the brainstem at 65 dg. In the kidney cortex and liver, an increase in vacuolation was noticed between 36 and 65 dg. Some vacuolated cells were also noticed in the lungs and spleen at 51 and 65 dg. Altogether, these histological observations suggest that alpha-mannosidosis is unlikely to affect ontogenesis before the second half of gestation in guinea pigs; however, the morphopathological features recorded during the last quarter of gestation (which may roughly correspond to the period covering near term to 1-2 years of age in human) were clearly noticeable and may have had some impact.


Subject(s)
Brain/ultrastructure , Fetus/pathology , Viscera/pathology , alpha-Mannosidosis/pathology , Animals , Disease Progression , Female , Guinea Pigs , Microscopy, Electron, Transmission , Pregnancy
13.
Vet Pathol ; 44(5): 569-78, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17846229

ABSTRACT

Dogs with mucopolysaccharidosis (MPS) IIIA were bred within an experimental colony. As part of characterizing them as a model for testing therapeutic strategies for the analogous disease of children, a pathologic study was undertaken. By histology, there were variably stained storage cytosomes within neurons, including many that stained for gangliosides. On ultrastructure examination, these cytosomes contained either moderately dense granular material, tentatively interpreted as precipitated glycosaminoglycan; a variety of multilaminar bodies, interpreted as being associated with secondary accumulation of gangliosides; or a mixture of both types. In the liver, storage vesicles also contained excess glycogen as a secondary storage product. In various tissues, there were large foamy macrophages. In the brain, many of these were in juxtaposition with neurons, and, on ultrastructure examination, they contained storage cytosomes similar to those in neurons. However, the neuron in association with such a macrophage frequently showed little such material.


Subject(s)
Dog Diseases/pathology , Mucopolysaccharidosis III/veterinary , Animals , Cerebellum/pathology , Cerebral Cortex/pathology , Dogs , Kidney/pathology , Liver/pathology , Microscopy, Electron, Transmission , Mucopolysaccharidosis III/pathology
14.
J Inherit Metab Dis ; 30(3): 358-64, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17458708

ABSTRACT

Mucopolysaccharidosis IIIB, an autosomal recessive lysosomal storage disorder of heparan sulfate caused by mutations in the alpha-N-acetylglucosaminidase (NAGLU) gene, was recently discovered in cattle. Clinical signs include progressive ataxia, stumbling gait, swaying and difficulty in balance and walking. These clinical signs are usually first observed at approximately 2 years of age and then develop progressively over the lifespan of the animals. Affected bulls were found to be homozygous for the missense mutation E452K (c.1354G > A). The availability of mutational analysis permits screening for the NAGLU mutation to eradicate this mutation from the cattle breeding population.


Subject(s)
Acetylglucosaminidase/genetics , Cattle Diseases/genetics , Mucopolysaccharidosis III/veterinary , Mutation, Missense , Animals , Brain/pathology , Cattle , Cattle Diseases/enzymology , Cattle Diseases/pathology , DNA/genetics , DNA/isolation & purification , Genome , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Neurons/pathology , Neurons/ultrastructure , Reference Values , Skin/chemistry , Thalamic Nuclei/pathology
15.
Neurosci Res ; 53(2): 161-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16023750

ABSTRACT

alpha-Mannosidosis is a lysosomal storage disease resulting from a deficiency of the enzyme alpha-D-mannosidase. A major feature of alpha-mannosidosis is progressive neurological decline, for which there is no safe and effective treatment available. We have a guinea pig model of alpha-mannosidosis that models the human condition. This study investigates the feasibility of implanting differentiated mouse embryonic stem cells in the neonatal guinea pig brain in order to provide a source of alpha-mannosidase to the affected central nervous system. Cells implanted at a low dose (1.5 x 10(3)cells per hemisphere) at 1 week of age were found to survive in very low numbers in some immunosuppressed animals out to 8 weeks. Four weeks post-implantation, cells implanted in high numbers (10(5) cells per hemisphere) formed teratomas in the majority of the animals implanted. Although implanted cells were found to migrate extensively within the brain and differentiate into mature cells of neural (and other) lineages, the safety issue related to uncontrolled cell proliferation precluded the use of this cell type for longer-term implantation studies. We conclude that the pluripotent cell type used in this study is unsuitable for achieving safe engraftment in the guinea pig brain.


Subject(s)
Brain/cytology , Graft Survival/physiology , Multipotent Stem Cells/cytology , Stem Cell Transplantation/adverse effects , alpha-Mannosidosis/therapy , Animals , Cell Differentiation , Cell Movement , Disease Models, Animal , Guinea Pigs , Immunohistochemistry , Mice , Microscopy, Confocal , Stem Cell Transplantation/methods , Teratoma/etiology
16.
Mol Genet Metab ; 85(3): 203-12, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15979032

ABSTRACT

Alpha-mannosidosis is a lysosomal storage disorder characterised by the lysosomal accumulation of mannose-containing oligosaccharides and a range of pathological consequences, caused by a deficiency of the lysosomal enzyme alpha-mannosidase. One of the major features of alpha-mannosidosis is progressive neurological decline, for which there is no safe and effective treatment. Implantation of stem cells into the central nervous system has been proposed as a potential therapy for these disorders. We report the construction and characterisation of mouse embryonic stem cell lines for the sustained over-expression of recombinant human lysosomal alpha-mannosidase (rhalphaM). Two vectors (involving recombinant human alpha-mannosidase expression driven by either the chicken beta-actin promoter/CMV enhancer or by the elongation factor 1-alpha promoter) were constructed and used to transfect mouse D3 embryonic stem cells. Selected clonal cell lines were isolated and tested to evaluate their expression of recombinant human alpha-mannosidase. Stem cell clones transfected with the chicken beta-actin promoter/CMV enhancer maintained rhalphaM expression levels throughout differentiation. This expression was not markedly elevated above background. In contrast, the vector incorporating the elongation factor 1-alpha promoter facilitated substantial over-expression of alpha-mannosidase when analysed out to 21 days of differentiation in stably transfected cell lines. The highest expressing cell line was found to qualitatively retain a similar differentiation potential to untransfected cells, and to secrete alpha-mannosidase that could mediate a reduction in the level of oligosaccharides stored by human alpha-mannosidosis skin fibroblasts. These results suggest potential for the use of this cell line for investigation of a stem cell therapy approach to treat alpha-mannosidosis.


Subject(s)
Stem Cells/enzymology , alpha-Mannosidase/metabolism , Animals , Cell Line , Gene Expression , Humans , Mannosephosphates/metabolism , Mice , Stem Cell Transplantation , Stem Cells/cytology , Transfection , Up-Regulation , alpha-Mannosidase/genetics , alpha-Mannosidosis/therapy
17.
Acta Paediatr Suppl ; 94(447): 61-8; discussion 57, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15895715

ABSTRACT

AIM: Mucopolysaccharidosis VI (Maroteaux-Lamy syndrome) is a lysosomal storage disease caused by a deficiency of the enzyme-N-acetylgalactosamine 4-sulphatase (ASB). Enzyme replacement therapy with recombinant human ASB (rhASB) has been studied in a randomized, double-blind, two-dose (0.2 and 1.0 mg/kg/week) phase I/II study (n = 7) followed by an open-label single dose (1.0 mg/kg/week) extension study. We report the pharmacokinetic profile of rhASB and the impact of antibody development. METHODS: Pharmacokinetic analysis was performed at weeks 1, 2, 12, 24, 83, 84 and 96. Infusions were administered over 4 hours using a ramp-up protocol. Plasma ASB and rhASB antibody concentrations and urine glycosaminoglycan (GAG) concentrations were determined. RESULTS: The area under the plasma concentration-time curve (AUC(0-t)) for the high-dose group increased from week 1 to week 2, but remained unchanged at weeks 12 and 24. A large difference in mean AUC(0-t) was observed between the low- and high-dose groups. Pharmacokinetic results at weeks 83, 84 and 96 were similar to those at week 24. Six patients developed antibodies to rhASB. One patient developed high antibody levels in combination with a high ASB concentration, while a second patient also developed high antibody levels with undetectable ASB concentrations. Antibodies from the second patient blocked detection of ASB. By week 72, antibody levels had decreased in all patients. The high-dose rhASB produced a more rapid and greater percentage reduction in urinary GAG concentrations than the lower dose (70% versus 55% at 24 weeks). Antibody levels did not appear to influence urinary GAG concentrations. CONCLUSION: Pharmacokinetic parameters appear to be independent of the duration of treatment and are not linear between the 0.2 and 1.0 mg/kg/week doses. Antibodies to rhASB develop in most patients, but their concentration decreases over time. Antibody formation may influence pharmacokinetic parameters during the early phases of treatment, although it appears to have limited impact on biochemical efficacy.


Subject(s)
Glycosaminoglycans/urine , Mucopolysaccharidosis VI/drug therapy , N-Acetylgalactosamine-4-Sulfatase/pharmacokinetics , N-Acetylgalactosamine-4-Sulfatase/therapeutic use , Adolescent , Child , Double-Blind Method , Female , Humans , Male , Models, Biological , Mucopolysaccharidosis VI/metabolism , N-Acetylgalactosamine-4-Sulfatase/metabolism , Recombinant Proteins/therapeutic use
18.
Mol Genet Metab ; 83(3): 239-45, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542395

ABSTRACT

Mucopolysaccharidosis type IIIA (MPS IIIA) is a lysosomal storage disorder caused by a deficiency in the lysosomal enzyme sulfamidase, which is required for the degradation of heparan sulfate. The disease is characterized by neurological dysfunction but relatively mild somatic manifestations. A naturally occurring mouse model to MPS IIIA exhibits a similar disease progression to that observed in patients. Disease in the mice results from a base substitution at codon 31 in the sulfamidase gene, altering an aspartic acid to an asparagine (D31N). This aspartic 31 is involved in binding of the divalent metal ion needed for catalytic function, and as such reduces the specific activity of the enzyme to about 3% of that of wild-type. The mutant protein has decreased stability and shows increased degradation over a 24 h chase period when compared to wild-type mouse sulfamidase. Mouse sulfamidase that was purified using a two-step ion exchange procedure was shown to have similar kinetic properties to that of purified human sulfamidase. Recombinant murine sulfamidase was able to correct the storage phenotype of MPS IIIA fibroblasts after endocytosis via the mannose-6-phosphate receptor.


Subject(s)
Hydrolases/genetics , Mice/genetics , Mucopolysaccharidosis III/therapy , Mutation, Missense/genetics , Recombinant Proteins/therapeutic use , Animals , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Fibroblasts/metabolism , Hydrolases/deficiency , Hydrolases/metabolism , Immunoprecipitation , Kinetics , Mucopolysaccharidosis III/genetics , Receptor, IGF Type 2/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Staining and Labeling
19.
Hum Mutat ; 24(3): 199-207, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15300847

ABSTRACT

Mucopolysaccharidosis type I (MPS I) is an autosomal recessive lysosomal storage disorder caused by a deficiency of alpha-L-iduronidase (IDUA). Mutations in the gene are responsible for the enzyme deficiency, which leads to the intralysosomal storage of the partially degraded glycosaminoglycans dermatan sulfate and heparan sulfate. Molecular characterization of MPS I patients has resulted in the identification of over 70 distinct mutations in the IDUA gene. The high degree of molecular heterogeneity reflects the wide clinical variability observed in MPS I patients. Six novel mutations, c.1087C>T (p.R363C), c.1804T>A (p.F602I), c.793G>C, c.712T>A (p.L238Q), c.1727+2T>A, and c.1269C>G (p.S423R), in a total of 14 different mutations, and 13 different polymorphic changes, including the novel c.246C>G (p.H82Q), were identified in a cohort of 10 MPS I patients enrolled in a clinical trial of enzyme-replacement therapy. Five novel amino acid substitutions and c.236C>T (p.A79V) were engineered into the wild-type IDUA cDNA and expressed. A p.G265R read-through mutation, arising from the c.793G>C splice mutation, was also expressed. Each mutation reduced IDUA protein and activity levels to varying degrees with the processing of many of the mutant forms also affected by IDUA. The varied properties of the expressed mutant forms of IDUA reflect the broad range of biochemical and clinical phenotypes of the 10 patients in this study. IDUA kinetic data derived from each patient's cultured fibroblasts, in combination with genotype data, was used to predict disease severity. Finally, residual IDUA protein concentration in cultured fibroblasts showed a weak correlation to the degree of immune response to enzyme-replacement therapy in each patient.


Subject(s)
Iduronidase/genetics , Mucopolysaccharidosis I/genetics , Mutation , Amino Acid Substitution , Animals , CHO Cells , Cell Line/enzymology , Codon/genetics , Cohort Studies , Cricetinae , Cricetulus , DNA Mutational Analysis , DNA, Complementary/genetics , Exons/genetics , Fibroblasts/enzymology , Humans , Iduronidase/chemistry , Iduronidase/deficiency , Iduronidase/metabolism , Iduronidase/therapeutic use , Kinetics , Mucopolysaccharidosis I/drug therapy , Mutagenesis, Site-Directed , Mutation, Missense , Phenotype , Point Mutation , Polymorphism, Genetic , Recombinant Fusion Proteins/metabolism
20.
Cell Mol Biol (Noisy-le-grand) ; 49(5): 769-77, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14528914

ABSTRACT

Lysosomal storage disorders represent a group of over 45 distinct genetic diseases, each one resulting from a deficiency of a particular lysosomal protein or, in a few cases, from non-lysosomal proteins that are involved in lysosomal biogenesis. A common biochemical feature of this group of disorders is the accumulation within lysosomes of undegraded or partially degraded substrates that are normally degraded within, and transported out of the lysosome. The particular substrates stored and the site(s) of storage vary with disease type and enzyme/protein deficiency. The nature of the substrate can be used to group the disorders into broad categories including the mucopolysaccharidoses, lipidoses, glycogenoses and oligosaccharidoses. These categories show many clinical similarities within groups as well as significant similarities between groups. For most lysosomal storage disorders the relationship between the stored substrates (type, amount and location) and the disease pathology is not well understood. The use of mass spectrometry and in particular tandem mass spectrometry provides a powerful tool for the investigation of stored substrates in this group of disorders. In this review we will describe the use of mass spectrometry for the analysis of stored substrates. We will discuss progress in the field, limitations of current methods, and summarise issues relating to the diagnosis and treatment of some of the more prevalent lysosomal storage disorders.


Subject(s)
Lysosomal Storage Diseases/metabolism , Mass Spectrometry , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/metabolism , Glycoproteins/analysis , Humans , Lysosomal Storage Diseases/diagnosis , Mucopolysaccharidoses/diagnosis , Mucopolysaccharidoses/metabolism , Oligosaccharides/analysis , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Sphingolipidoses/diagnosis , Sphingolipidoses/metabolism , Sphingolipids/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...