Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 267
Filter
1.
Cell Mol Gastroenterol Hepatol ; 17(6): 983-1005, 2024.
Article in English | MEDLINE | ID: mdl-38307491

ABSTRACT

Microvillus inclusion disease (MVID) is a rare condition that is present from birth and affects the digestive system. People with MVID experience severe diarrhea that is difficult to control, cannot absorb dietary nutrients, and struggle to grow and thrive. In addition, diverse clinical manifestations, some of which are life-threatening, have been reported in cases of MVID. MVID can be caused by variants in the MYO5B, STX3, STXBP2, or UNC45A gene. These genes produce proteins that have been functionally linked to each other in intestinal epithelial cells. MVID associated with STXBP2 variants presents in a subset of patients diagnosed with familial hemophagocytic lymphohistiocytosis type 5. MVID associated with UNC45A variants presents in most patients diagnosed with osteo-oto-hepato-enteric syndrome. Furthermore, variants in MYO5B or STX3 can also cause other diseases that are characterized by phenotypes that can co-occur in subsets of patients diagnosed with MVID. Recent studies involving clinical data and experiments with cells and animals revealed connections between specific phenotypes occurring outside of the digestive system and the type of gene variants that cause MVID. Here, we have reviewed these patterns and correlations, which are expected to be valuable for healthcare professionals in managing the disease and providing personalized care for patients and their families.


Subject(s)
Malabsorption Syndromes , Microvilli , Mucolipidoses , Phenotype , Humans , Mucolipidoses/genetics , Mucolipidoses/pathology , Microvilli/pathology , Microvilli/genetics , Malabsorption Syndromes/genetics , Malabsorption Syndromes/pathology , Animals , Myosin Type V/genetics , Myosin Type V/metabolism , Mutation , Genetic Predisposition to Disease
2.
Am J Med Genet A ; 194(6): e63545, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38264826

ABSTRACT

Mucolipidosis type-II (ML-II) is an ultra-rare disorder caused by deficiency of N-acetylglucosaminyl-1-phosphotransferase enzyme due to biallelic pathogenic variants in GNPTAB gene. There are a few known about the natural history of ML-II. In this study, we presented the natural course of 24 patients diagnosed with ML-II. Mean age at diagnosis was 9.3 ± 5.7 months. All patients had coarse face, developmental delay, and hypotonia. The mean survival time was 3.01 ± 1.4 years. The oldest patient was 6.5 years old. Twelve patients died due to lung infection and respiratory failure. We observed early and significant radiological findings of ML-II were different from typical dysostosis multiplex such as femoral cloaking, rickets-like changes, and talocalcaneal stippling. These are significant findings observed in the fetal or newborn period which is considered to be highly characteristic of ML-II and disappears in the first year. Cloaking, rickets-like changes, and stippling were not observed in patients older than three months of age and this suggests that these findings disappear within the first year. These radiological features can be used as important clues for diagnosis. We detected eight different pathogenic variants in GNPTAB gene, three of them were novel.


Subject(s)
Mucolipidoses , Humans , Mucolipidoses/genetics , Mucolipidoses/diagnosis , Mucolipidoses/diagnostic imaging , Mucolipidoses/pathology , Male , Female , Infant , Child, Preschool , Child , Transferases (Other Substituted Phosphate Groups)/genetics , Mutation/genetics , Radiography , Early Diagnosis , Infant, Newborn , Phenotype
3.
Am J Ophthalmol ; 258: 183-195, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37972748

ABSTRACT

PURPOSE: To report the genetic etiology of Lisch epithelial corneal dystrophy (LECD). DESIGN: Multicenter cohort study. METHODS: A discovery cohort of 27 individuals with LECD from 17 families, including 7 affected members from the original LECD family, 6 patients from 2 new families and 14 simplex cases, was recruited. A cohort of 6 individuals carrying a pathogenic MCOLN1 (mucolipin 1) variant was reviewed for signs of LECD. Next-generation sequencing or targeted Sanger sequencing were used in all patients to identify pathogenic or likely pathogenic variants and penetrance of variants. RESULTS: Nine rare heterozygous MCOLN1 variants were identified in 23 of 27 affected individuals from 13 families. The truncating nature of 7 variants and functional testing of 1 missense variant indicated that they result in MCOLN1 haploinsufficiency. Importantly, in the homozygous and compound-heterozygous state, 4 of 9 LECD-associated variants cause the rare lysosomal storage disorder mucolipidosis IV (MLIV). Autosomal recessive MLIV is a systemic disease and comprises neurodegeneration as well as corneal opacity of infantile-onset with epithelial autofluorescent lysosomal inclusions. However, the 6 parents of 3 patients with MLIV confirmed to carry pathogenic MCOLN1 variants did not have the LECD phenotype, suggesting MCOLN1 haploinsufficiency may be associated with reduced penetrance and variable expressivity. CONCLUSIONS: MCOLN1 haploinsufficiency is the major cause of LECD. Based on the overlapping clinical features of corneal epithelial cells with autofluorescent inclusions reported in both LECD and MLIV, it is concluded that some carriers of MCOLN1 haploinsufficiency-causing variants present with LECD.


Subject(s)
Corneal Dystrophies, Hereditary , Mucolipidoses , Transient Receptor Potential Channels , Humans , Transient Receptor Potential Channels/genetics , Cohort Studies , Mucolipidoses/diagnosis , Mucolipidoses/genetics , Mucolipidoses/pathology , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/genetics
4.
JCI Insight ; 8(20)2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37698928

ABSTRACT

Sialidosis is an ultra-rare multisystemic lysosomal disease caused by mutations in the neuraminidase 1 (NEU1) gene. The severe type II form of the disease manifests with a prenatal/infantile or juvenile onset, bone abnormalities, severe neuropathology, and visceromegaly. A subset of these patients present with nephrosialidosis, characterized by abrupt onset of fulminant glomerular nephropathy. We studied the pathophysiological mechanism of the disease in 2 NEU1-deficient mouse models, a constitutive Neu1-knockout, Neu1ΔEx3, and a conditional phagocyte-specific knockout, Neu1Cx3cr1ΔEx3. Mice of both strains exhibited terminal urinary retention and severe kidney damage with elevated urinary albumin levels, loss of nephrons, renal fibrosis, presence of storage vacuoles, and dysmorphic mitochondria in the intraglomerular and tubular cells. Glycoprotein sialylation in glomeruli, proximal distal tubules, and distal tubules was drastically increased, including that of an endocytic reabsorption receptor megalin. The pool of megalin bearing O-linked glycans with terminal galactose residues, essential for protein targeting and activity, was reduced to below detection levels. Megalin levels were severely reduced, and the protein was directed to lysosomes instead of the apical membrane. Together, our results demonstrated that desialylation by NEU1 plays a crucial role in processing and cellular trafficking of megalin and that NEU1 deficiency in sialidosis impairs megalin-mediated protein reabsorption.


Subject(s)
Kidney Diseases , Mucolipidoses , Animals , Humans , Mice , Kidney Diseases/metabolism , Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Mucolipidoses/genetics , Mucolipidoses/pathology , Neuraminidase/genetics
5.
J Clin Invest ; 132(10)2022 05 16.
Article in English | MEDLINE | ID: mdl-35575086

ABSTRACT

Variants in the UNC45A cochaperone have been recently associated with a syndrome combining diarrhea, cholestasis, deafness, and bone fragility. Yet the mechanism underlying intestinal failure in UNC45A deficiency remains unclear. Here, biallelic variants in UNC45A were identified by next-generation sequencing in 6 patients with congenital diarrhea. Corroborating in silico prediction, variants either abolished UNC45A expression or altered protein conformation. Myosin VB was identified by mass spectrometry as client of the UNC45A chaperone and was found misfolded in UNC45AKO Caco-2 cells. In keeping with impaired myosin VB function, UNC45AKO Caco-2 cells showed abnormal epithelial morphogenesis that was restored by full-length UNC45A, but not by mutant alleles. Patients and UNC45AKO 3D organoids displayed altered luminal development and microvillus inclusions, while 2D cultures revealed Rab11 and apical transporter mislocalization as well as sparse and disorganized microvilli. All those features resembled the subcellular abnormalities observed in duodenal biopsies from patients with microvillus inclusion disease. Finally, microvillus inclusions and shortened microvilli were evidenced in enterocytes from unc45a-deficient zebrafish. Taken together, our results provide evidence that UNC45A plays an essential role in epithelial morphogenesis through its cochaperone function of myosin VB and that UNC45A loss causes a variant of microvillus inclusion disease.


Subject(s)
Diarrhea, Infantile , Malabsorption Syndromes , Mucolipidoses , Myosin Type V , Animals , Caco-2 Cells , Diarrhea, Infantile/metabolism , Diarrhea, Infantile/pathology , Facies , Fetal Growth Retardation , Hair Diseases , Humans , Infant , Intracellular Signaling Peptides and Proteins/metabolism , Malabsorption Syndromes/metabolism , Microvilli/genetics , Microvilli/pathology , Mucolipidoses/genetics , Mucolipidoses/metabolism , Mucolipidoses/pathology , Myosin Type V/genetics , Myosin Type V/metabolism , Phenotype , Zebrafish/genetics , Zebrafish/metabolism
6.
Cells ; 11(3)2022 02 04.
Article in English | MEDLINE | ID: mdl-35159355

ABSTRACT

BACKGROUND: Mucolipidosis IV (MLIV) is an autosomal recessive pediatric disease that leads to motor and cognitive deficits and loss of vision. It is caused by a loss of function of the lysosomal channel transient receptor potential mucolipin-1 and is associated with an early pro-inflammatory brain phenotype, including increased cytokine expression. The goal of the current study was to determine whether blood cytokines are linked to motor dysfunction in patients with MLIV and reflect brain inflammatory changes observed in an MLIV mouse model. METHODS: To determine the relationship between blood cytokines and motor function, we collected plasma from MLIV patients and parental controls concomitantly with assessment of motor function using the Brief Assessment of Motor Function and Modified Ashworth scales. We then compared these profiles with cytokine profiles in brain and plasma samples collected from the Mcoln1-/- mouse model of MLIV. RESULTS: We found that MLIV patients had prominently increased cytokine levels compared to familial controls and identified profiles of cytokines correlated with motor dysfunction, including IFN-γ, IFN-α2, and IP-10. We found that IP-10 was a key differentiating factor separating MLIV cases from controls based on data from human plasma, mouse plasma, and mouse brain. CONCLUSIONS: Our data indicate that MLIV is characterized by increased blood cytokines, which are strongly related to underlying neurological and functional deficits in MLIV patients. Moreover, our data identify the interferon pro-inflammatory axis in both human and mouse signatures, suggesting that interferon signaling is an important aspect of MLIV pathology.


Subject(s)
Mucolipidoses , Transient Receptor Potential Channels , Animals , Chemokine CXCL10/metabolism , Cytokines/metabolism , Disease Models, Animal , Humans , Interferons/metabolism , Mice , Mucolipidoses/genetics , Mucolipidoses/metabolism , Mucolipidoses/pathology , Transient Receptor Potential Channels/metabolism
7.
Liver Int ; 42(2): 402-411, 2022 02.
Article in English | MEDLINE | ID: mdl-34811877

ABSTRACT

BACKGROUND & AIMS: Biallelic pathogenic variants in MYO5B cause microvillus inclusion disease (MVID), or familial intrahepatic cholestasis (FIC). The reported FIC patients are scarce and so the genotype-phenotype correlation has not been fully characterised. This study aimed to report more MYO5B-associated FIC patients and correlate genotypes to phenotypes in more detail. METHODS: The phenotype and genetic data of 12 newly diagnosed MYO5B-associated (including 11 FIC) patients, as well as 118 previously reported patients with available genotypes, were summarised. Only patients with biallelic MYO5B variants were enrolled. Nonsense, frameshift, canonical splice sites, initiation codon loss, and single exon or multiexon deletion were defined as null MYO5B variants. RESULTS: Phenotypically, 50 were isolated MVID, 47 involved both liver and intestine (combined), and 33 were isolated FIC (9 persistent, 15 recurrent, 3 transient, and 6 un-sub-classified) patients. The severity of intestinal manifestation was positively correlated to an increased number of null variants (ρ = 0.299, P = .001). All FIC patients carried at least one non-null variant, and the severity of cholestasis was correlated to the presence of a null variant (ρ = 0.420, P = .029). The proportion of FIC patients (16/29, 55%) harbouring missense/in-frame variants affecting the non-motor regions of MYO5B was significantly higher than that of MVID (3/25, 12%, P = .001) and combined patients (3/31, 10%, P = .000). 10 of the 29 FIC patients harboured missense/in-frame variants at the IQ motifs comparing to none in the 56 MVID and combined patients (P = .000). CONCLUSIONS: The phenotype of MYO5B deficiency was associated with MYO5B genotypes, the nullity or the domain affected.


Subject(s)
Cholestasis, Intrahepatic/genetics , Mucolipidoses , Myosin Heavy Chains , Myosin Type V , Genetic Association Studies , Humans , Liver/pathology , Mucolipidoses/genetics , Mucolipidoses/pathology , Mutation , Myosin Heavy Chains/genetics , Myosin Type V/genetics
8.
Genet Med ; 23(12): 2369-2377, 2021 12.
Article in English | MEDLINE | ID: mdl-34341521

ABSTRACT

PURPOSE: Pathogenic variants in GNPTAB and GNPTG, encoding different subunits of GlcNAc-1-phosphotransferase, cause mucolipidosis (ML) II, MLIII alpha/beta, and MLIII gamma. This study aimed to investigate the cellular and molecular bases underlying skeletal abnormalities in patients with MLII and MLIII. METHODS: We analyzed bone biopsies from patients with MLIII alpha/beta or MLIII gamma by undecalcified histology and histomorphometry. The skeletal status of Gnptgko and Gnptab-deficient mice was determined and complemented by biochemical analysis of primary Gnptgko bone cells. The clinical relevance of the mouse data was underscored by systematic urinary collagen crosslinks quantification in patients with MLII, MLIII alpha/beta, and MLIII gamma. RESULTS: The analysis of iliac crest biopsies revealed that bone remodeling is impaired in patients with GNPTAB-associated MLIII alpha/beta but not with GNPTG-associated MLIII gamma. Opposed to Gnptab-deficient mice, skeletal remodeling is not affected in Gnptgko mice. Most importantly, patients with variants in GNPTAB but not in GNPTG exhibited increased bone resorption. CONCLUSION: The gene-specific impact on bone remodeling in human individuals and in mice proposes distinct molecular functions of the GlcNAc-1-phosphotransferase subunits in bone cells. We therefore appeal for the necessity to classify MLIII based on genetic in addition to clinical criteria to ensure appropriate therapy.


Subject(s)
Bone Resorption , Mucolipidoses , Transferases (Other Substituted Phosphate Groups) , Animals , Humans , Mice , Mucolipidoses/genetics , Mucolipidoses/pathology , Transferases (Other Substituted Phosphate Groups)/genetics
9.
JCI Insight ; 6(16)2021 08 23.
Article in English | MEDLINE | ID: mdl-34197342

ABSTRACT

Functional loss of myosin Vb (MYO5B) induces a variety of deficits in intestinal epithelial cell function and causes a congenital diarrheal disorder, microvillus inclusion disease (MVID). The impact of MYO5B loss on differentiated cell lineage choice has not been investigated. We quantified the populations of differentiated epithelial cells in tamoxifen-induced, epithelial cell-specific MYO5B-knockout (VilCreERT2 Myo5bfl/fl) mice utilizing digital image analysis. Consistent with our RNA-sequencing data, MYO5B loss induced a reduction in tuft cells in vivo and in organoid cultures. Paneth cells were significantly increased by MYO5B deficiency along with expansion of the progenitor cell zone. We further investigated the effect of lysophosphatidic acid (LPA) signaling on epithelial cell differentiation. Intraperitoneal LPA significantly increased tuft cell populations in both control and MYO5B-knockout mice. Transcripts for Wnt ligands were significantly downregulated by MYO5B loss in intestinal epithelial cells, whereas Notch signaling molecules were unchanged. Additionally, treatment with the Notch inhibitor dibenzazepine (DBZ) restored the populations of secretory cells, suggesting that the Notch pathway is maintained in MYO5B-deficient intestine. MYO5B loss likely impairs progenitor cell differentiation in the small intestine in vivo and in vitro, partially mediated by Wnt/Notch imbalance. Notch inhibition and/or LPA treatment may represent an effective therapeutic approach for treatment of MVID.


Subject(s)
Malabsorption Syndromes/genetics , Microvilli/pathology , Mucolipidoses/genetics , Myosin Type V/deficiency , Receptors, Notch/metabolism , Wnt Signaling Pathway/genetics , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Dibenzazepines/pharmacology , Disease Models, Animal , Enterocytes/drug effects , Enterocytes/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Jejunum/cytology , Jejunum/drug effects , Jejunum/pathology , Lysophospholipids/pharmacology , Lysophospholipids/therapeutic use , Malabsorption Syndromes/drug therapy , Malabsorption Syndromes/pathology , Mice , Mice, Knockout , Microvilli/genetics , Mucolipidoses/drug therapy , Mucolipidoses/pathology , Myosin Type V/genetics , Organoids , Primary Cell Culture , Receptors, Notch/antagonists & inhibitors , Stem Cells/physiology , Wnt Signaling Pathway/drug effects
10.
Am J Med Genet A ; 185(10): 2873-2877, 2021 10.
Article in English | MEDLINE | ID: mdl-34037310

ABSTRACT

Trichohepatoenteric syndrome (THES) is a very rare autosomal recessive genetic disorder, which is characterized by intractable diarrhea during infancy, dysmorphic features, immunodeficiency, and a failure to thrive. There are still significant difficulties for patients and clinicians in terms of the management of THES, even though its molecular basis has been uncovered in the last decade. In this article, we have presented two cases relating to siblings that have been diagnosed with the condition. Concerning one of the patients, we described a novel variation (c.2114 + 5G > A) in the TTC37 gene and a mild clinical course; meanwhile, the other one was clinically diagnosed with THES at 17 years of age, but they had seizures and died suddenly. These cases expand the spectrum of clinical findings in relation to THES.


Subject(s)
Carrier Proteins/genetics , Diarrhea, Infantile/genetics , Failure to Thrive/genetics , Fetal Growth Retardation/genetics , Hair Diseases/genetics , Malabsorption Syndromes/genetics , Microvilli/pathology , Mucolipidoses/genetics , Adolescent , Diarrhea, Infantile/complications , Diarrhea, Infantile/diagnosis , Diarrhea, Infantile/pathology , Facies , Failure to Thrive/complications , Failure to Thrive/diagnosis , Failure to Thrive/pathology , Female , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/pathology , Genetic Predisposition to Disease , Hair Diseases/complications , Hair Diseases/diagnosis , Hair Diseases/pathology , Humans , Infant , Malabsorption Syndromes/complications , Malabsorption Syndromes/diagnosis , Malabsorption Syndromes/pathology , Male , Microvilli/genetics , Mucolipidoses/complications , Mucolipidoses/diagnosis , Mucolipidoses/pathology , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/pathology , Siblings
11.
Neurosci Lett ; 755: 135944, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33965501

ABSTRACT

Mucolipidosis IV (MLIV) is an autosomal-recessive disease caused by loss-of-function mutations in the MCOLN1 gene encoding the non-selective cationic lysosomal channel transient receptor potential mucolipin-1 (TRPML1). Patients with MLIV suffer from severe motor and cognitive deficits that manifest in early infancy and progressive loss of vision leading to blindness in the second decade of life. There are no therapies available for MLIV and the unmet medical need is extremely high. Here we review the spectrum of clinical presentations and the latest research in the MLIV pre-clinical model, with the aim of highlighting the progress in understanding the pathophysiology of the disease. These highlights include elucidation of the neurodevelopmental versus neurodegenerative features over the course of disease, hypomyelination as one of the major brain pathological disease hallmarks, and dysregulation of cytokines, with emerging evidence of IFN-gamma pathway upregulation in response to TRPML1 loss and pro-inflammatory activation of astrocytes and microglia. These scientific advances in the MLIV field provide a basis for future translational research, including biomarker and therapy development, that are desperately needed for this patient population.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Mucolipidoses/diagnostic imaging , Mucolipidoses/metabolism , Transient Receptor Potential Channels/metabolism , Brain/pathology , Humans , Lysosomes/genetics , Lysosomes/metabolism , Lysosomes/pathology , Mucolipidoses/genetics , Mucolipidoses/pathology , Myelin Sheath/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Transient Receptor Potential Channels/genetics
12.
Hum Genet ; 140(8): 1143-1156, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33974130

ABSTRACT

Biallelic STX3 variants were previously reported in five individuals with the severe congenital enteropathy, microvillus inclusion disease (MVID). Here, we provide a significant extension of the phenotypic spectrum caused by STX3 variants. We report ten individuals of diverse geographic origin with biallelic STX3 loss-of-function variants, identified through exome sequencing, single-nucleotide polymorphism array-based homozygosity mapping, and international collaboration. The evaluated individuals all presented with MVID. Eight individuals also displayed early-onset severe retinal dystrophy, i.e., syndromic-intestinal and retinal-disease. These individuals harbored STX3 variants that affected both the retinal and intestinal STX3 transcripts, whereas STX3 variants affected only the intestinal transcript in individuals with solitary MVID. That STX3 is essential for retinal photoreceptor survival was confirmed by the creation of a rod photoreceptor-specific STX3 knockout mouse model which revealed a time-dependent reduction in the number of rod photoreceptors, thinning of the outer nuclear layer, and the eventual loss of both rod and cone photoreceptors. Together, our results provide a link between STX3 loss-of-function variants and a human retinal dystrophy. Depending on the genomic site of a human loss-of-function STX3 variant, it can cause MVID, the novel intestinal-retinal syndrome reported here or, hypothetically, an isolated retinal dystrophy.


Subject(s)
Eye Diseases, Hereditary/genetics , Intestinal Mucosa/metabolism , Malabsorption Syndromes/genetics , Microvilli/pathology , Mucolipidoses/genetics , Polymorphism, Single Nucleotide , Qa-SNARE Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Dystrophies/genetics , Aged , Aged, 80 and over , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Autopsy , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Eye Diseases, Hereditary/metabolism , Eye Diseases, Hereditary/pathology , Female , Gene Expression Regulation , Homozygote , Humans , Intestinal Mucosa/pathology , Malabsorption Syndromes/metabolism , Malabsorption Syndromes/pathology , Mice , Mice, Knockout , Microvilli/genetics , Microvilli/metabolism , Mucolipidoses/metabolism , Mucolipidoses/pathology , Phenotype , Qa-SNARE Proteins/deficiency , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Dystrophies/metabolism , Retinal Dystrophies/pathology , Sensory Rhodopsins/genetics , Sensory Rhodopsins/metabolism , Exome Sequencing
13.
Int J Mol Sci ; 22(9)2021 Apr 22.
Article in English | MEDLINE | ID: mdl-33922276

ABSTRACT

Sialidosis, caused by a genetic deficiency of the lysosomal sialidase gene (NEU1), is a systemic disease involving various tissues and organs, including the nervous system. Understanding the neurological dysfunction and pathology associated with sialidosis remains a challenge, partially due to the lack of a human model system. In this study, we have generated two types of induced pluripotent stem cells (iPSCs) with sialidosis-specific NEU1G227R and NEU1V275A/R347Q mutations (sialidosis-iPSCs), and further differentiated them into neural precursor cells (iNPCs). Characterization of NEU1G227R- and NEU1V275A/R347Q- mutated iNPCs derived from sialidosis-iPSCs (sialidosis-iNPCs) validated that sialidosis-iNPCs faithfully recapitulate key disease-specific phenotypes, including reduced NEU1 activity and impaired lysosomal and autophagic function. In particular, these cells showed defective differentiation into oligodendrocytes and astrocytes, while their neuronal differentiation was not notably affected. Importantly, we found that the phenotypic defects of sialidosis-iNPCs, such as impaired differentiation capacity, could be effectively rescued by the induction of autophagy with rapamycin. Our results demonstrate the first use of a sialidosis-iNPC model with NEU1G227R- and NEU1V275A/R347Q- mutation(s) to study the neurological defects of sialidosis, particularly those related to a defective autophagy-lysosome pathway, and may help accelerate the development of new drugs and therapeutics to combat sialidosis and other LSDs.


Subject(s)
Astrocytes/pathology , Induced Pluripotent Stem Cells/pathology , Mucolipidoses/pathology , Neural Stem Cells/pathology , Neuraminidase/metabolism , Oligodendroglia/pathology , Teratoma/pathology , Astrocytes/metabolism , Autophagy , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/metabolism , Lysosomes , Mucolipidoses/genetics , Mucolipidoses/metabolism , Mutation , Neural Stem Cells/metabolism , Neuraminidase/genetics , Oligodendroglia/metabolism , Phenotype , Teratoma/genetics , Teratoma/metabolism
14.
Hum Mol Genet ; 30(10): 908-922, 2021 05 29.
Article in English | MEDLINE | ID: mdl-33822942

ABSTRACT

Mucolipidosis IV (MLIV) is an orphan disease leading to debilitating psychomotor deficits and vision loss. It is caused by loss-of-function mutations in the MCOLN1 gene that encodes the lysosomal transient receptor potential channel mucolipin1, or TRPML1. With no existing therapy, the unmet need in this disease is very high. Here, we showed that AAV-mediated CNS-targeted gene transfer of the human MCOLN1 gene rescued motor function and alleviated brain pathology in the MLIV mouse model. Using the AAV-PHP.b vector in symptomatic mice, we showed long-term reversal of declined motor function and significant delay of paralysis. Next, using self-complementary AAV9 clinical candidate vector, we showed that its intracerebroventricular administration in post-natal day 1 mice significantly improved motor function, myelination and reduced lysosomal storage load in the MLIV mouse brain. Based on our data and general advancements in the gene therapy field, we propose scAAV9-mediated CSF-targeted MCOLN1 gene transfer as a therapeutic strategy in MLIV.


Subject(s)
Genetic Therapy , Mucolipidoses/therapy , Nervous System Diseases/therapy , Transient Receptor Potential Channels/genetics , Animals , Brain/metabolism , Brain/pathology , Dependovirus/genetics , Disease Models, Animal , Humans , Loss of Function Mutation/genetics , Lysosomes/genetics , Lysosomes/pathology , Mice , Mucolipidoses/cerebrospinal fluid , Mucolipidoses/genetics , Mucolipidoses/pathology , Nervous System Diseases/cerebrospinal fluid , Nervous System Diseases/genetics , Nervous System Diseases/pathology
16.
Sci Rep ; 11(1): 3556, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33574442

ABSTRACT

Severe skeletal alterations are common symptoms in patients with mucolipidosis type II (MLII), a rare lysosomal storage disorder of childhood. We have previously reported that progressive bone loss in a mouse model for MLII is caused by an increased number of bone-resorbing osteoclasts, which is accompanied by elevated expression of the cytokine interleukin-6 (IL-6) in the bone microenvironment. In the present study we addressed the question, if pharmacological blockade of IL-6 can prevent the low bone mass phenotype of MLII mice. Since the cellular IL-6 response can be mediated by either the membrane-bound (classic signaling) or the soluble IL-6 receptor (trans-signaling), we first performed cell culture assays and found that both pathways can increase osteoclastogenesis. We then crossed MLII mice with transgenic mice expressing the recombinant soluble fusion protein sgp130Fc, which represents a natural inhibitor of IL-6 trans-signaling. By undecalcified histology and bone-specific histomorphometry we found that high circulating sgp130Fc levels do not affect skeletal growth or remodeling in wild-type mice. Most importantly, blockade of IL-6 trans-signaling did neither reduce osteoclastogenesis, nor increase bone mass in MLII mice. Therefore, our data clearly demonstrate that the bone phenotype of MLII mice cannot be corrected by blocking the IL-6 trans-signaling.


Subject(s)
Interleukin-6/genetics , Mucolipidoses/genetics , Osteogenesis/genetics , Skeleton/pathology , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Disease Models, Animal , Humans , Mice , Mice, Transgenic/genetics , Mucolipidoses/pathology , Osteoclasts/metabolism , Osteoclasts/pathology
17.
Mol Biol Rep ; 48(2): 1465-1474, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33507475

ABSTRACT

Mucolipidosis III gamma (ML III γ) is a slowly progressive disorder that affects multiple parts of the body such as the skeleton, joints, and connective tissue structures. It is caused by pathogenic variants in the GNPTG gene that provides instructions for producing the γ subunit of GlcNAc-1-phosphotransferase. In this study we aim to characterize clinical findings and biological insights on two novel GNPTG variants causing ML III γ phenotypes with varying severity. We report on two siblings with ML III γ bearing the previously undescribed c.477C > G (p.Y159*) nonsense variant in a homozygous state as well as a patient with ML III γ bearing the novel c.110 + 19_111-17del variant in a homozygous state. These variants were revealed by whole-exome sequencing and Sanger sequencing, respectively. Their parents, who are heterozygotes for the same mutation, are healthy. The clinical and radiographic presentation of ML III γ in our patients who had c.477C > G (p.Y159*) variant is consistent with a relatively severe form of the disease, which is further supported by a working three-dimensional model of the GlcNAc-1-phosphotransferase γ subunit. On the other hand, it is seen that our patient who carries the c.110 + 19_111-17del variant has a milder phenotype. Our findings help broaden the spectrum of GNPTG variants causing ML III γ and offer structural and mechanistic insights into loss of GlcNAc-1-phosphotransferase γ subunit function.


Subject(s)
Genetic Predisposition to Disease , Mucolipidoses/genetics , Transferases (Other Substituted Phosphate Groups)/genetics , Codon, Nonsense/genetics , Female , Homozygote , Humans , Joints/pathology , Male , Mucolipidoses/pathology , Phenotype , Severity of Illness Index , Siblings , Skeleton/pathology , Exome Sequencing
18.
Tokai J Exp Clin Med ; 45(4): 243-248, 2020 Dec 20.
Article in English | MEDLINE | ID: mdl-33300597

ABSTRACT

OBJECTIVE: We generated induced pluripotent stem (iPS) cells from a patient with distal myopathy with rimmed vacuoles (DMRV), in which sialic acids synthesis is reported to be defective. In this study, we examined whether the differentiation to retinal pigment epithelial (RPE) cells and autophagy was affected in the patient derived cells. METHODS: Patient derived iPS cells were established through the transduction of re-programming factors into peripheral mononuclear cells via retrovirus vectors. RPE cells were induced from iPS cells through aggregation culture. Then the autophagy induced by amino acid starvation was estimated by measuring LC3-containing "puncta" structure. RESULTS: A 3D aggregate culture of patient-derived iPS cells resulted in some irregular shapes, and the aggregate contained large vacuoles filled with lipid droplets and cellular components such as damaged mitochondria. RPE cells induced from patient-derived iPS cells showed impaired autophagy flux under amino acid starvation. CONCLUSION: These findings were similar to those of sialidosis patient-derived iPS cells, in which cleavage of terminal sialic acids in oligosaccharide chains is defective. This suggests that the control of both the addition and removal of sialic acids are pivotal for autophagy progression.


Subject(s)
Autophagy , Distal Myopathies/pathology , Epithelial Cells/physiology , Pluripotent Stem Cells/pathology , Retinal Pigments , Sialic Acids , Vacuoles/pathology , Adult , Amino Acids/deficiency , Cell Differentiation , Cells, Cultured , Distal Myopathies/etiology , Distal Myopathies/metabolism , Female , Humans , Mucolipidoses/pathology , Oligosaccharides/metabolism , Pluripotent Stem Cells/cytology , Sialic Acids/deficiency , Sialic Acids/metabolism
19.
Dis Model Mech ; 13(11)2020 11 18.
Article in English | MEDLINE | ID: mdl-33023972

ABSTRACT

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.


Subject(s)
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
20.
Int J Mol Sci ; 21(17)2020 Aug 23.
Article in English | MEDLINE | ID: mdl-32842549

ABSTRACT

The endosomal recycling pathway lies at the heart of the membrane trafficking machinery in the cell. It plays a central role in determining the composition of the plasma membrane and is thus critical for normal cellular homeostasis. However, defective endosomal recycling has been linked to a wide range of diseases, including cancer and some of the most common neurological disorders. It is also frequently subverted by many diverse human pathogens in order to successfully infect cells. Despite its importance, endosomal recycling remains relatively understudied in comparison to the endocytic and secretory transport pathways. A greater understanding of the molecular mechanisms that support transport through the endosomal recycling pathway will provide deeper insights into the pathophysiology of disease and will likely identify new approaches for their detection and treatment. This review will provide an overview of the normal physiological role of the endosomal recycling pathway, describe the consequences when it malfunctions, and discuss potential strategies for modulating its activity.


Subject(s)
Endosomes/metabolism , Neoplasms/metabolism , Small Molecule Libraries/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Drug Delivery Systems/methods , Endocytosis/physiology , Endosomes/drug effects , Humans , Malabsorption Syndromes/metabolism , Malabsorption Syndromes/pathology , Microvilli/metabolism , Microvilli/pathology , Mucolipidoses/metabolism , Mucolipidoses/pathology , Neoplasms/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Transport/physiology , Secretory Pathway , rab GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...