Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 79
1.
Neurobiol Dis ; 152: 105279, 2021 05.
Article En | MEDLINE | ID: mdl-33516873

Sialidosis is a neuropathic lysosomal storage disease caused by a deficiency in the NEU1 gene-encoding lysosomal neuraminidase and characterized by abnormal accumulation of undigested sialyl-oligoconjugates in systemic organs including brain. Although patients exhibit neurological symptoms, the underlying neuropathological mechanism remains unclear. Here, we generated induced pluripotent stem cells (iPSCs) from skin fibroblasts with sialidosis and induced the differentiation into neural progenitor cells (NPCs) and neurons. Sialidosis NPCs and neurons mimicked the disease-like phenotypes including reduced neuraminidase activity, accumulation of sialyl-oligoconjugates and lysosomal expansions. Functional analysis also revealed that sialidosis neurons displayed two distinct abnormalities, defective exocytotic glutamate release and augmented α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor (AMPAR)-mediated Ca2+ influx. These abnormalities were restored by overexpression of the wild-type NEU1 gene, demonstrating causative role of neuraminidase deficiency in functional impairments of disease neurons. Comprehensive proteomics analysis revealed the significant reduction of SNARE proteins and glycolytic enzymes in synaptosomal fraction, with downregulation of ATP production. Bypassing the glycolysis by treatment of pyruvate, which is final metabolite of glycolysis pathway, improved both the synaptsomal ATP production and the exocytotic function. We also found that upregulation of AMPAR and L-type voltage dependent Ca2+ channel (VDCC) subunits in disease neurons, with the restoration of AMPAR-mediated Ca2+ over-load by treatment of antagonists for the AMPAR and L-type VDCC. Our present study provides new insights into both the neuronal pathophysiology and potential therapeutic strategy for sialidosis.


Calcium Signaling/physiology , Mucolipidoses/physiopathology , Neurons/pathology , Neurons/physiology , Exocytosis/physiology , Glycolysis/physiology , Humans , Induced Pluripotent Stem Cells , Synapses/pathology , Synapses/physiology
2.
Dis Model Mech ; 13(11)2020 11 18.
Article En | MEDLINE | ID: mdl-33023972

Mucolipidosis type III (MLIII) gamma is a rare inherited lysosomal storage disorder caused by mutations in GNPTG encoding the γ-subunit of GlcNAc-1-phosphotransferase, the key enzyme ensuring proper intracellular location of multiple lysosomal enzymes. Patients with MLIII gamma typically present with osteoarthritis and joint stiffness, suggesting cartilage involvement. Using Gnptg knockout (Gnptgko ) mice as a model of the human disease, we showed that missorting of a number of lysosomal enzymes is associated with intracellular accumulation of chondroitin sulfate in Gnptgko chondrocytes and their impaired differentiation, as well as with altered microstructure of the cartilage extracellular matrix (ECM). We also demonstrated distinct functional and structural properties of the Achilles tendons isolated from Gnptgko and Gnptab knock-in (Gnptabki ) mice, the latter displaying a more severe phenotype resembling mucolipidosis type II (MLII) in humans. Together with comparative analyses of joint mobility in MLII and MLIII patients, these findings provide a basis for better understanding of the molecular reasons leading to joint pathology in these patients. Our data suggest that lack of GlcNAc-1-phosphotransferase activity due to defects in the γ-subunit causes structural changes within the ECM of connective and mechanosensitive tissues, such as cartilage and tendon, and eventually results in functional joint abnormalities typically observed in MLIII gamma patients. This idea was supported by a deficit of the limb motor function in Gnptgko mice challenged on a rotarod under fatigue-associated conditions, suggesting that the impaired motor performance of Gnptgko mice was caused by fatigue and/or pain at the joint.This article has an associated First Person interview with the first author of the paper.


Cartilage/pathology , Homeostasis , Joints/pathology , Mucolipidoses/metabolism , Mucolipidoses/pathology , Achilles Tendon/pathology , Achilles Tendon/ultrastructure , Aging/pathology , Animals , Cartilage/ultrastructure , Cell Differentiation , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrocytes/ultrastructure , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Fibrillar Collagens/metabolism , Lysosomes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Mucolipidoses/physiopathology , Transferases (Other Substituted Phosphate Groups)/metabolism
3.
JCI Insight ; 5(20)2020 10 15.
Article En | MEDLINE | ID: mdl-33055423

Although congenital heart defects (CHDs) represent the most common birth defect, a comprehensive understanding of disease etiology remains unknown. This is further complicated since CHDs can occur in isolation or as a feature of another disorder. Analyzing disorders with associated CHDs provides a powerful platform to identify primary pathogenic mechanisms driving disease. Aberrant localization and expression of cathepsin proteases can perpetuate later-stage heart diseases, but their contribution toward CHDs is unclear. To investigate the contribution of cathepsins during cardiovascular development and congenital disease, we analyzed the pathogenesis of cardiac defects in zebrafish models of the lysosomal storage disorder mucolipidosis II (MLII). MLII is caused by mutations in the GlcNAc-1-phosphotransferase enzyme (Gnptab) that disrupt carbohydrate-dependent sorting of lysosomal enzymes. Without Gnptab, lysosomal hydrolases, including cathepsin proteases, are inappropriately secreted. Analyses of heart development in gnptab-deficient zebrafish show cathepsin K secretion increases its activity, disrupts TGF-ß-related signaling, and alters myocardial and valvular formation. Importantly, cathepsin K inhibition restored normal heart and valve development in MLII embryos. Collectively, these data identify mislocalized cathepsin K as an initiator of cardiac disease in this lysosomal disorder and establish cathepsin inhibition as a viable therapeutic strategy.


Cathepsin K/genetics , Heart Defects, Congenital/genetics , Heart/growth & development , Mucolipidoses/genetics , Transferases (Other Substituted Phosphate Groups)/genetics , Animals , Disease Models, Animal , Enzyme Activation/genetics , Genetic Predisposition to Disease , Heart/physiopathology , Heart Defects, Congenital/physiopathology , Heart Valves/growth & development , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/physiopathology , Mucolipidoses/physiopathology , Mutation , Transforming Growth Factor beta/genetics , Zebrafish/genetics
4.
Int J Mol Sci ; 21(18)2020 Sep 17.
Article En | MEDLINE | ID: mdl-32957425

Mucolipidosis II and III (ML II/III) are caused by a deficiency of uridine-diphosphate N-acetylglucosamine: lysosomal-enzyme-N-acetylglucosamine-1-phosphotransferase (GlcNAc-1-phosphotransferase, EC2.7.8.17), which tags lysosomal enzymes with a mannose 6-phosphate (M6P) marker for transport to the lysosome. The process is performed by a sequential two-step process: first, GlcNAc-1-phosphotransferase catalyzes the transfer of GlcNAc-1-phosphate to the selected mannose residues on lysosomal enzymes in the cis-Golgi network. The second step removes GlcNAc from lysosomal enzymes by N-acetylglucosamine-1-phosphodiester α-N-acetylglucosaminidase (uncovering enzyme) and exposes the mannose 6-phosphate (M6P) residues in the trans-Golgi network, in which the enzymes are targeted to the lysosomes by M6Preceptors. A deficiency of GlcNAc-1-phosphotransferase causes the hypersecretion of lysosomal enzymes out of cells, resulting in a shortage of multiple lysosomal enzymes within lysosomes. Due to a lack of GlcNAc-1-phosphotransferase, the accumulation of cholesterol, phospholipids, glycosaminoglycans (GAGs), and other undegraded substrates occurs in the lysosomes. Clinically, ML II and ML III exhibit quite similar manifestations to mucopolysaccharidoses (MPSs), including specific skeletal deformities known as dysostosis multiplex and gingival hyperplasia. The life expectancy is less than 10 years in the severe type, and there is no definitive treatment for this disease. In this review, we have described the updated diagnosis and therapy on ML II/III.


Enzyme Replacement Therapy/methods , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Lysosomes/metabolism , Mucolipidoses/diagnosis , Animals , Biological Transport, Active , Disease Models, Animal , Glycosaminoglycans/metabolism , Humans , Mannosephosphates/metabolism , Mucolipidoses/enzymology , Mucolipidoses/physiopathology , Mucolipidoses/therapy
5.
Int J Mol Sci ; 21(12)2020 Jun 26.
Article En | MEDLINE | ID: mdl-32604955

Mucolipidosis type IV (MLIV) is an ultra-rare lysosomal storage disorder caused by biallelic mutations in MCOLN1 gene encoding the transient receptor potential channel mucolipin-1. So far, 35 pathogenic or likely pathogenic MLIV-related variants have been described. Clinical manifestations include severe intellectual disability, speech deficit, progressive visual impairment leading to blindness, and myopathy. The severity of the condition may vary, including less severe psychomotor delay and/or ocular findings. As no striking recognizable facial dysmorphism, skeletal anomalies, organomegaly, or lysosomal enzyme abnormalities in serum are common features of MLIV, the clinical diagnosis may be significantly improved because of characteristic ophthalmological anomalies. This review aims to outline the pathophysiology and genetic defects of this condition with a focus on the genotype-phenotype correlation amongst cases published in the literature. The authors will present their own clinical observations and long-term outcomes in adult MLIV cases.


Genetic Association Studies , Genetic Profile , Mucolipidoses/genetics , Mucolipidoses/physiopathology , Mutation , TRPM Cation Channels/genetics , Adult , Female , Humans , Male , Young Adult
6.
Ann Clin Transl Neurol ; 7(6): 911-923, 2020 06.
Article En | MEDLINE | ID: mdl-32472645

OBJECTIVE: Type I sialidosis (ST-1) is a rare autosomal recessive inherited disorder. To date, there has been no study on ST-1 patients in mainland China. METHODS: We reported in detail the cases of five Chinese ST-1 patients from two centers, and summarized all worldwide cases. Then, we compared the differences between Chinese and foreign patients. RESULTS: A total of 77 genetically confirmed ST-1 patients were identified: 12 from mainland China, 23 from Taiwan, 10 from other Asian regions, and 32 from European and American regions. The mean age of onset was 16.0 ± 6.7 years; the most common symptoms were myoclonus seizures (96.0%), followed by ataxia (94.3%), and blurred vision (67.2%). Compared to other groups, the onset age of patients from mainland China was much younger (10.8 ± 2.7 years). The incidence of visual impairment was lower in patients from other Asian regions than in patients from mainland China and Taiwan (28.6% vs. 81.8%-100%). Cherry-red spots were less frequent in the Taiwanese patients than in patients from other regions (27.3% vs. 55.2%-90.0%). Furthermore, 48 different mutation types were identified. Chinese mainland and Taiwanese patients were more likely to carry the c.544A > G mutation (75% and 100%, respectively) than the patients from other regions (only 0%-10.0%). Approximately 50% of Chinese mainland patients carried the c.239C > T mutation, a much higher proportion than that found in the other populations. In addition, although the brain MRI of most patients was normal, 18 F-FDG-PET analysis could reveal cerebellar and occipital lobe hypometabolism. INTERPRETATION: ST-1 patients in different regions are likely to have different mutation types; environmental factors may influence clinical manifestations. Larger studies enrolling more patients are required.


Mucolipidoses/genetics , Mucolipidoses/physiopathology , Adolescent , Adult , Age of Onset , Cerebellum/diagnostic imaging , Cerebellum/metabolism , China , Female , Humans , Incidence , Male , Mucolipidoses/complications , Mucolipidoses/diagnostic imaging , Occipital Lobe/diagnostic imaging , Occipital Lobe/metabolism , Positron-Emission Tomography , Vision Disorders/epidemiology , Vision Disorders/etiology , Young Adult
7.
Eur J Med Genet ; 63(7): 103927, 2020 Jul.
Article En | MEDLINE | ID: mdl-32298796

BACKGROUND: Mucolipidosis type IV (ML-IV) is a rare autosomal-recessive lysosomal storage disease, caused by mutations in MCOLN1. ML-IV manifests with developmental delay, esotropia and corneal clouding. While the clinical phenotype is well-described, the diagnosis of ML-IV is often challenging and elusive. OBJECTIVE: Our experience with ML-IV patients brought to the clinical observation that they share common and identifiable facial features, not yet described in the literature to date. Here, we utilized a computerized facial analysis tool to establish this association. METHODS: Using the DeepGestalt algorithm, 50 two-dimensional facial images of ten ML-IV patients were analyzed, and compared to unaffected controls (n = 98) and to individuals affected with other genetic disorders (n = 99). Results were expressed in terms of the area-under-the-curve (AUC) of the receiver-operating-characteristic curve (ROC). RESULTS: When compared to unaffected cases and to cases diagnosed with syndromes other than ML-IV, the ML-IV cohort showed an AUC of 0.822 (p value < 0.01) and an AUC of 0.885 (p value < 0.001), respectively. CONCLUSIONS: We describe recognizable facial features typical in patients with ML-IV. Reaffirmed by the DeepGestalt technology, the described common facial phenotype adds to the tools currently available for clinicians and may thus assist in reaching an earlier diagnosis of this rare and underdiagnosed disorder.


Face/diagnostic imaging , Mucolipidoses/diagnostic imaging , Mucolipidoses/genetics , Phenotype , Adolescent , Adult , Automated Facial Recognition/methods , Child , Child, Preschool , Cohort Studies , Face/physiopathology , Family Characteristics , Female , Humans , Infant , Male , Mucolipidoses/physiopathology , Mutation , Patients , Transient Receptor Potential Channels/genetics , Young Adult
8.
Am J Med Genet A ; 182(6): 1500-1505, 2020 06.
Article En | MEDLINE | ID: mdl-32220057

Mucolipidosis type IV (MLIV) is an autosomal recessively inherited lysosomal storage disorder characterized by progressive psychomotor delay and retinal degeneration that is associated with biallelic variants in the MCOLN1 gene. The gene, which is expressed in late endosomes and lysosomes of various tissue cells, encodes the transient receptor potential channel mucolipin 1 consisting of six transmembrane domains. Here, we described 14-year follow-up observation of a 4-year-old Japanese male MLIV patient with a novel homozygous in-frame deletion variant p.(F313del), which was identified by whole-exome sequencing analysis. Neurological examination revealed progressive psychomotor delay, and atrophy of the corpus callosum and cerebellum was observed on brain magnetic resonance images. Ophthalmologically, corneal clouding has remained unchanged during the follow-up period, whereas optic nerve pallor and retinal degenerative changes exhibited progressive disease courses. Light-adapted electroretinography was non-recordable. Transmission electron microscopy of granulocytes revealed characteristic concentric multiple lamellar structures and an electron-dense inclusion in lysosomes. The in-frame deletion variant was located within the second transmembrane domain, which is of putative functional importance for channel properties.


Lysosomal Storage Diseases/genetics , Lysosomes/genetics , Mucolipidoses/genetics , Transient Receptor Potential Channels/genetics , Adolescent , Child , Child, Preschool , Corpus Callosum/diagnostic imaging , Corpus Callosum/physiopathology , Homozygote , Humans , Lysosomal Storage Diseases/diagnostic imaging , Lysosomal Storage Diseases/physiopathology , Lysosomes/pathology , Magnetic Resonance Imaging , Male , Mucolipidoses/diagnostic imaging , Mucolipidoses/physiopathology , Mutation/genetics , Psychomotor Disorders/complications , Psychomotor Disorders/genetics , Psychomotor Disorders/physiopathology , Retinal Degeneration/complications , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology
9.
Am J Med Genet A ; 179(7): 1196-1204, 2019 07.
Article En | MEDLINE | ID: mdl-31038846

Mucolipidosis (ML) is a rare lysosomal storage disorder with a wide spectrum of disease severity according to the type. Sleep-disordered breathing is recognized as a characteristic feature of ML but objective data are scarce. The aim of the study was to describe sleep data and medical management in children with ML α/ß. All patients with ML α/ß followed at a national reference center of ML were included. Five patients had ML II, one patient had ML III and one patient had ML II-III. One patient was started on noninvasive ventilation (NIV) to allow extubation after prolonged invasive mechanical ventilation. The six other patients underwent sleep study at a median age of 1.8 years (range 4 months-17.4 years). Obstructive sleep apnea (OSA) was observed in all patients with a median apnea-hypopnea index (AHI) of 36 events/hr (range 5-52) requiring continuous positive airway pressure (CPAP) or NIV. CPAP/NIV resulted in an improvement of nocturnal gas exchange and was continued in all patients with an excellent compliance. Two patients died. Systematic sleep studies are recommended at time of diagnosis in ML. CPAP or NIV are effective treatments of OSA, well tolerated, and may contribute to improve the quality of life of patients and caregivers.


Continuous Positive Airway Pressure/methods , Mucolipidoses/physiopathology , Mutation , Noninvasive Ventilation/methods , Sleep Apnea, Obstructive/physiopathology , Transferases (Other Substituted Phosphate Groups)/genetics , Adolescent , Child , Child, Preschool , Disease Management , Female , Gene Expression , Humans , Infant , Male , Mucolipidoses/complications , Mucolipidoses/genetics , Mucolipidoses/therapy , Patient Compliance , Polysomnography , Quality of Life , Severity of Illness Index , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/genetics , Sleep Apnea, Obstructive/therapy , Transferases (Other Substituted Phosphate Groups)/deficiency , Treatment Outcome
11.
Brain Imaging Behav ; 11(1): 214-223, 2017 02.
Article En | MEDLINE | ID: mdl-26843009

In order to identify the cortical changes in patients with Sialidosis type 1, diffusion tensor imaging and resting state fMRI were acquired from 11 patients and 11 sex/age matched normal controls after clinical evaluations. The neuroimages from each participant were normalized and parcellated according to the Automatic Anatomical Labeling. Both the mean diffusivity and the corresponding functional connectivity were calculated from each cortical region. The white matter tract integrity was examined. The difference between patients and controls was examined using Student's t-test and between patients with either homozygous or heterozygous mutations by Mann-Whitney U test, both at a threshold of 0.05. Increased mean diffusivity throughout the brain can be noticed in the patients, together with a compromised white matter tracts integrity. The most severely affected cortical regions are in the occipital lobe. Decreased functional connectivity was from the temporal and occipital lobes to the hippocampus and parahippocampus. In contrast, connectivity from thalamus was enhanced. Diffused cortical atrophy with posterior focal lesions was noticed. We concluded that MRI observed functional changes in the posterior cortical pathways in the patients with Sialidosis. The observation might be related to the cortical blindness due to an altered neural network and a compromised visual pathway in the patients.


Brain/diagnostic imaging , Brain/physiopathology , Mucolipidoses/diagnostic imaging , Mucolipidoses/physiopathology , Adult , Atrophy , Brain Mapping , Diffusion Tensor Imaging , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Mucolipidoses/complications , Mucolipidoses/genetics , Rest , Vision Disorders/diagnostic imaging , Vision Disorders/etiology , Vision Disorders/physiopathology , Visual Pathways/diagnostic imaging , Visual Pathways/physiopathology , Young Adult
12.
J Clin Sleep Med ; 12(12): 1695-1696, 2016 12 15.
Article En | MEDLINE | ID: mdl-27707435

ABSTRACT: Mucolipidosis II (Inclusion cell or I-cell disease) is an autosomal recessive lysosomal storage disorder clinically comparable to the mucopolysaccharidoses (MPS), characterized by progressive respiratory and neurologic deterioration. Sleep problems, especially obstructive sleep apnea (OSA) and disrupted sleep architecture, are observed in other lysosomal storage diseases but have not been described in mucolipidosis II. We report the progression of polysomnographic abnormalities in a child with mucolipidosis II, demonstrated by worsening sleep-related hypoventilation, OSA, and sleep state fragmentation despite advancing PAP therapy. Background slowing and reduction in spindle activity on limited EEG may reflect progressive CNS disease affecting thalamic neurons.


Disease Progression , Mucolipidoses/complications , Sleep Apnea, Obstructive/complications , Adolescent , Humans , Male , Mucolipidoses/physiopathology , Polysomnography/statistics & numerical data , Sleep Apnea, Obstructive/physiopathology
13.
Epileptic Disord ; 18(S2): 89-93, 2016 Sep 01.
Article En | MEDLINE | ID: mdl-27621198

Sialidoses are autosomal recessive disorders caused by NEU1 gene mutations and are classified on the basis of their phenotype and onset age. Sialidosis type II, with infantile onset, has a more severe phenotype characterized by coarse facial features, hepatomegaly, dysostosis multiplex, and developmental delay while patients with the late and milder type, known as "cherry red spot-myoclonus syndrome" develop myoclonic epilepsy, visual impairment and ataxia in the second or third decade of life. The diagnosis is usually suggested by increased urinary bound sialic acid excretion. We recently described genetically diagnosed patients with a specially mild phenotype, no retinal abnormalities and normal urinary sialic acid. This observation suggests that genetic analysis or the demonstration of the neuraminidase enzyme deficiency in cultured fibroblasts are needed to detect and diagnose mildest phenotypes.


Mucolipidoses , Humans , Mucolipidoses/diagnosis , Mucolipidoses/physiopathology , Mucolipidoses/therapy
14.
Mech Dev ; 142: 62-74, 2016 11.
Article En | MEDLINE | ID: mdl-27497746

Microvillus inclusion disease (MVID) is a life-threatening enteropathy characterised by malabsorption and incapacitating fluid loss due to chronic diarrhoea. Histological analysis has revealed that enterocytes in MVID patients exhibit reduction of microvilli, presence of microvillus inclusion bodies and intestinal villus atrophy, whereas genetic linkage analysis has identified mutations in myosin Vb gene as the main cause of MVID. In order to understand the cellular basis of MVID and the associated formation of inclusion bodies, an animal model that develops ex utero and is tractable genetically as well as by microscopy would be highly useful. Here we report that the intestine of the zebrafish goosepimples (gsp)/myosin Vb (myoVb) mutant shows severe reduction in intestinal folds - structures similar to mammalian villi. The loss of folds is further correlated with changes in the shape of enterocytes. In striking similarity with MVID patients, zebrafish gsp/myoVb mutant larvae exhibit microvillus atrophy, microvillus inclusions and accumulation of secretory material in enterocytes. We propose that the zebrafish gsp/myoVb mutant is a valuable model to study the pathophysiology of MVID. Furthermore, owing to the advantages of zebrafish in screening libraries of small molecules, the gsp mutant will be an ideal tool to identify compounds having therapeutic value against MVID.


Intestine, Small/physiopathology , Malabsorption Syndromes/genetics , Microvilli/pathology , Mucolipidoses/genetics , Mutant Proteins/genetics , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Animals , Disease Models, Animal , Humans , Malabsorption Syndromes/physiopathology , Microvilli/genetics , Mucolipidoses/physiopathology , Mutation , Zebrafish/genetics , Zebrafish/physiology
15.
Am J Med Genet A ; 170A(5): 1278-82, 2016 May.
Article En | MEDLINE | ID: mdl-26789537

Mucolipidosis (ML) II alpha/beta is an autosomal recessive disease caused by reduced enzyme activity of N-acetylglucosamine-1-phosphotransferase. Clinical symptoms of ML II are severe psychomotor delay and dysostosis multiplex; death usually occurs by 5-8 years of age from cardiopulmonary complications. Allogeneic hematopoietic stem cell transplantation (HSCT) has been attempted for ML; however, few reports have documented the detailed outcomes of HSCT for ML. A 26-month-old girl received a human leukocyte antigen 3/6-allele-matched transplant from cord blood. The preparative regimen consisted of fludarabine, cyclophosphamide, 6-Gy total body irradiation, and rabbit antithymocyte globulin. Although comparing before and after cord blood transplantation results, we observed that lysosomal enzyme activities in the plasma decreased by approximately 20-40%. Low serum levels of immunoglobulin A, G2, and G4 were also observed before HSCT; however, these values normalized after transplantation. Despite undergoing HSCT, she was treated twice for bacterial pneumonia with acute respiratory distress syndrome at ages 37 and 38 months. Although HSCT effects on the clinical manifestations were limited, laboratory data including plasma lysosomal enzyme activities and serum levels of immunoglobulin showed improvement.


Abnormalities, Multiple/genetics , Cord Blood Stem Cell Transplantation , Mucolipidoses/genetics , Psychomotor Disorders/genetics , Transferases (Other Substituted Phosphate Groups)/genetics , Abnormalities, Multiple/blood , Abnormalities, Multiple/physiopathology , Abnormalities, Multiple/therapy , Animals , Child, Preschool , Cyclophosphamide/administration & dosage , Female , Humans , Immunoglobulins/blood , Mucolipidoses/blood , Mucolipidoses/physiopathology , Mucolipidoses/therapy , Psychomotor Disorders/blood , Psychomotor Disorders/physiopathology , Psychomotor Disorders/therapy , Rabbits , Transplantation, Homologous , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
16.
Am J Med Genet A ; 170A(5): 1187-95, 2016 May.
Article En | MEDLINE | ID: mdl-26749367

Mucolipidosis IIIalpha/beta (MLIIIalpha/beta) is a rare lysosomal storage disorder characterized by childhood onset of flexion contractures of fingers, joint stiffness in the shoulders, hips, and knees, and mild short stature. Recessive mutations in the GNPTAB gene have been associated with MLIIIalpha/beta. We present five children aged 9-16 years from a large kindred family whose serum activities of several lysosomal enzymes were significantly elevated. Whole exome sequencing followed by confirmation by Sanger sequencing identified a novel homozygous missense mutation (c.22 A > G; p.R8G) in the GNPTAB gene in all affected subjects. The five patients initially presented with flexion contractures of fingers followed by stiffnes of large joints. Only two affected boys also had a nephrotic-range proteinuria. Renal biopsy showed focal segmental glomerulosclerosis and foamy appearance of glomerular visceral epithelial cells which were compatible with storage disease. No other known causes of proteinuria could be detected by both laboratory and biopsy findings. There was no known family history of hereditary kidney disease, and healthy siblings and parents had normal renal function and urinalysis. These findings suggest that the renal involvement probably due to MLIIIalpha/beta, although it can still be present by coincidence in the two affected patients.


Kidney/physiopathology , Mucolipidoses/genetics , Transferases (Other Substituted Phosphate Groups)/genetics , Adolescent , Base Sequence/genetics , Child , Exome , Female , Glomerulosclerosis, Focal Segmental , Homozygote , Humans , Kidney/diagnostic imaging , Male , Mucolipidoses/diagnostic imaging , Mucolipidoses/physiopathology , Mutation, Missense , Pedigree
17.
Sci Rep ; 5: 12312, 2015 Jul 23.
Article En | MEDLINE | ID: mdl-26201991

Inherited MYO5B mutations have recently been associated with microvillus inclusion disease (MVID), an autosomal recessive syndrome characterized by intractable, life-threatening, watery diarrhea appearing shortly after birth. Characterization of the molecular mechanisms underlying this disease and development of novel therapeutic approaches is hampered by the lack of animal models. In this study we describe the phenotype of a novel mouse model with targeted inactivation of Myo5b. Myo5b knockout mice show perinatal mortality, diarrhea and the characteristic mislocalization of apical and basolateral plasma membrane markers in enterocytes. Moreover, in transmission electron preparations, we observed microvillus atrophy and the presence of microvillus inclusion bodies. Importantly, Myo5b knockout embryos at day 20 of gestation already display all these structural defects, indicating that they are tissue autonomous rather than secondary to environmental cues, such as the long-term absence of nutrients in the intestine. Myo5b knockout mice closely resemble the phenotype of MVID patients and constitute a useful model to further investigate the underlying molecular mechanism of this disease and to preclinically assess the efficacy of novel therapeutic approaches.


Diarrhea/pathology , Diarrhea/physiopathology , Disease Models, Animal , Malabsorption Syndromes/pathology , Malabsorption Syndromes/physiopathology , Microvilli/pathology , Mucolipidoses/pathology , Mucolipidoses/physiopathology , Myosin Type V/genetics , Animals , Diarrhea/etiology , Female , Malabsorption Syndromes/complications , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucolipidoses/complications , Myosin Type V/metabolism
18.
PLoS One ; 9(10): e109768, 2014.
Article En | MEDLINE | ID: mdl-25314316

UDP-GlcNAc:lysosomal enzyme N-acetylglucosamine-1-phosphotransferase is an α2ß2γ2 hexameric enzyme that catalyzes the synthesis of the mannose 6-phosphate targeting signal on lysosomal hydrolases. Mutations in the α/ß subunit precursor gene cause the severe lysosomal storage disorder mucolipidosis II (ML II) or the more moderate mucolipidosis III alpha/beta (ML III α/ß), while mutations in the γ subunit gene cause the mildest disorder, mucolipidosis III gamma (ML III γ). Here we report neurologic consequences of mouse models of ML II and ML III γ. The ML II mice have a total loss of acid hydrolase phosphorylation, which results in depletion of acid hydrolases in mesenchymal-derived cells. The ML III γ mice retain partial phosphorylation. However, in both cases, total brain extracts have normal or near normal activity of many acid hydrolases reflecting mannose 6-phosphate-independent lysosomal targeting pathways. While behavioral deficits occur in both models, the onset of these changes occurs sooner and the severity is greater in the ML II mice. The ML II mice undergo progressive neurodegeneration with neuronal loss, astrocytosis, microgliosis and Purkinje cell depletion which was evident at 4 months whereas ML III γ mice have only mild to moderate astrocytosis and microgliosis at 12 months. Both models accumulate the ganglioside GM2, but only ML II mice accumulate fucosylated glycans. We conclude that in spite of active mannose 6-phosphate-independent targeting pathways in the brain, there are cell types that require at least partial phosphorylation function to avoid lysosomal dysfunction and the associated neurodegeneration and behavioral impairments.


Mucolipidoses/physiopathology , Animals , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Female , Gangliosides/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Motor Activity , Mucolipidoses/genetics , Mucolipidoses/metabolism , Mucolipidoses/pathology , Oligosaccharides/metabolism , Psychomotor Disorders/genetics , Psychomotor Disorders/metabolism , Psychomotor Disorders/pathology , Psychomotor Disorders/physiopathology , Rotarod Performance Test , Sensorimotor Cortex/metabolism , Sensorimotor Cortex/pathology , Sensorimotor Cortex/physiopathology , Spinal Cord/metabolism , Spinal Cord/pathology , Transferases (Other Substituted Phosphate Groups)/genetics
19.
Acta Neuropathol Commun ; 2: 133, 2014 Sep 09.
Article En | MEDLINE | ID: mdl-25200117

Mucolipidosis IV (MLIV) is caused by mutations in the gene MCOLN1. Patients with MLIV have severe neurologic deficits and very little is known about the brain pathology in this lysosomal disease. Using an accurate mouse model of mucolipidosis IV, we observed early behavioral deficits which were accompanied by activation of microglia and astrocytes. The glial activation that persisted during the course of disease was not accompanied by neuronal loss even at the late stage. In vivo [Ca(2+)]-imaging revealed no changes in resting [Ca(2+)] levels in Mcoln1(-/-) cortical neurons, implying their physiological health. Despite the absence of neuron loss, we observed alterations in synaptic plasticity, as indicated by elevated paired-pulse facilitation and enhanced long-term potentiation. Myelination deficits and severely dysmorphic corpus callosum were present early and resembled white matter pathology in mucolipidosis IV patients. These results indicate the early involvement of glia, and challenge the traditional view of mucolipidosis IV as an overtly neurodegenerative condition.


Brain/pathology , Brain/physiopathology , Mucolipidoses/pathology , Mucolipidoses/physiopathology , Animals , Astrocytes/pathology , Disease Models, Animal , Exploratory Behavior/physiology , Gliosis , Male , Mice , Mice, Knockout , Microglia/pathology , Motor Activity/physiology , Myelin Sheath/pathology , Neuronal Plasticity , Neurons/physiology , Transient Receptor Potential Channels/genetics
20.
Nat Commun ; 5: 4681, 2014 Aug 14.
Article En | MEDLINE | ID: mdl-25119295

Mucolipidosis type IV (MLIV) is an autosomal recessive lysosomal storage disorder often characterized by severe neurodevelopmental abnormalities and neuro-retinal degeneration. Mutations in the TRPML1 gene are causative for MLIV. We used lead optimization strategies to identify--and MLIV patient fibroblasts to test--small-molecule activators for their potential to restore TRPML1 mutant channel function. Using the whole-lysosome planar patch-clamp technique, we found that activation of MLIV mutant isoforms by the endogenous ligand PI(3,5)P2 is strongly reduced, while activity can be increased using synthetic ligands. We also found that the F465L mutation renders TRPML1 pH insensitive, while F408Δ impacts synthetic ligand binding. Trafficking defects and accumulation of zinc in lysosomes of MLIV mutant fibroblasts can be rescued by the small molecule treatment. Collectively, our data demonstrate that small molecules can be used to restore channel function and rescue disease associated abnormalities in patient cells expressing specific MLIV point mutations.


Mucolipidoses/genetics , Mucolipidoses/prevention & control , Mutation/genetics , Phosphatidylinositol Phosphates/pharmacology , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/genetics , Cells, Cultured , Electrophysiological Phenomena , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Hydrogen-Ion Concentration , Ligands , Lysosomes/metabolism , Mucolipidoses/physiopathology , Patch-Clamp Techniques , Protein Isoforms , Transient Receptor Potential Channels/physiology , Zinc/metabolism
...