Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 224
Filter
1.
Glycoconj J ; 40(6): 611-619, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38147151

ABSTRACT

Neuraminidase 1 (NEU1) is a lysosomal sialidase that cleaves terminal α-linked sialic acid residues from sialylglycans. NEU1 is biosynthesized in the rough endoplasmic reticulum (RER) lumen as an N-glycosylated protein to associate with its protective protein/cathepsin A (CTSA) and then form a lysosomal multienzyme complex (LMC) also containing ß-galactosidase 1 (GLB1). Unlike other mammalian sialidases, including NEU2 to NEU4, NEU1 transport to lysosomes requires association of NEU1 with CTSA, binding of the CTSA carrying terminal mannose 6-phosphate (M6P)-type N-glycan with M6P receptor (M6PR), and intralysosomal NEU1 activation at acidic pH. In contrast, overexpression of the single NEU1 gene in mammalian cells causes intracellular NEU1 protein crystallization in the RER due to self-aggregation when intracellular CTSA is reduced to a relatively low level. Sialidosis (SiD) and galactosialidosis (GS) are autosomal recessive lysosomal storage diseases caused by the gene mutations of NEU1 and CTSA, respectively. These incurable diseases associate with the NEU1 deficiency, excessive accumulation of sialylglycans in neurovisceral organs, and systemic manifestations. We established a novel GS model mouse carrying homozygotic Ctsa IVS6 + 1 g/a mutation causing partial exon 6 skipping with simultaneous deficiency of Ctsa and Neu1. Symptoms developed in the GS mice like those in juvenile/adult GS patients, such as myoclonic seizures, suppressed behavior, gargoyle-like face, edema, proctoptosis due to Neu1 deficiency, and sialylglycan accumulation associated with neurovisceral inflammation. We developed a modified NEU1 (modNEU1), which does not form protein crystals but is transported to lysosomes by co-expressed CTSA. In vivo gene therapy for GS and SiD utilizing a single adeno-associated virus (AAV) carrying modNEU1 and CTSA genes under dual promoter control will be created.


Subject(s)
Lysosomal Storage Diseases , Mucolipidoses , Neuraminidase , Animals , Humans , Mice , Neuraminidase/chemistry , Mucolipidoses/genetics , Mucolipidoses/metabolism , Lysosomes/metabolism , Mammals/metabolism
2.
J Clin Invest ; 133(20)2023 10 16.
Article in English | MEDLINE | ID: mdl-37643022

ABSTRACT

Microvillus inclusion disease (MVID), caused by loss-of-function mutations in the motor protein myosin Vb (MYO5B), is a severe infantile disease characterized by diarrhea, malabsorption, and acid/base instability, requiring intensive parenteral support for nutritional and fluid management. Human patient-derived enteroids represent a model for investigation of monogenic epithelial disorders but are a rare resource from MVID patients. We developed human enteroids with different loss-of function MYO5B variants and showed that they recapitulated the structural changes found in native MVID enterocytes. Multiplex immunofluorescence imaging of patient duodenal tissues revealed patient-specific changes in localization of brush border transporters. Functional analysis of electrolyte transport revealed profound loss of Na+/H+ exchange (NHE) activity in MVID patient enteroids with near-normal chloride secretion. The chloride channel-blocking antidiarrheal drug crofelemer dose-dependently inhibited agonist-mediated fluid secretion. MVID enteroids exhibited altered differentiation and maturation versus healthy enteroids. γ-Secretase inhibition with DAPT recovered apical brush border structure and functional Na+/H+ exchange activity in MVID enteroids. Transcriptomic analysis revealed potential pathways involved in the rescue of MVID cells including serum/glucocorticoid-regulated kinase 2 (SGK2) and NHE regulatory factor 3 (NHERF3). These results demonstrate the utility of patient-derived enteroids for developing therapeutic approaches to MVID.


Subject(s)
Malabsorption Syndromes , Mucolipidoses , Myosin Type V , Humans , Microvilli/genetics , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Enterocytes/metabolism , Malabsorption Syndromes/genetics , Malabsorption Syndromes/therapy , Malabsorption Syndromes/metabolism , Mucolipidoses/genetics , Mucolipidoses/therapy , Mucolipidoses/metabolism
3.
Autophagy ; 19(7): 2143-2145, 2023 07.
Article in English | MEDLINE | ID: mdl-36633450

ABSTRACT

Degradation of macromolecules delivered to lysosomes by processes such as autophagy or endocytosis is crucial for cellular function. Lysosomes require more than 60 soluble hydrolases in order to catabolize such macromolecules. These soluble hydrolases are tagged with mannose6-phosphate (M6P) moieties in sequential reactions by the Golgi-resident GlcNAc-1-phosphotransferase complex and NAGPA/UCE/uncovering enzyme (N-acetylglucosamine-1-phosphodiester alpha-N-acetylglucosaminidase), which allows their delivery to endosomal/lysosomal compartments through trafficking mediated by cation-dependent and -independent mannose 6-phosphate receptors (MPRs). We and others recently identified TMEM251 as a novel regulator of the M6P pathway via independent genome-wide genetic screening strategies. We renamed TMEM251 to LYSET (lysosomal enzyme trafficking factor) to establish nomenclature reflective to this gene's function. LYSET is a Golgi-localized transmembrane protein important for the retention of the GlcNAc-1-phosphotransferase complex in the Golgi-apparatus. The current understanding of LYSET's importance regarding human biology is 3-fold: 1) highly pathogenic viruses that depend on lysosomal hydrolase activity require LYSET for infection. 2) The presence of LYSET is critical for cancer cell proliferation in nutrient-deprived environments in which extracellular proteins must be catabolized. 3) Inherited pathogenic alleles of LYSET can cause a severe inherited disease which resembles GlcNAc-1-phosphotransferase deficiency (i.e., mucolipidosis type II).Abbreviations: GlcNAc-1-PT: GlcNAc-1-phosphotransferase; KO: knockout; LSD: lysosomal storage disorder; LYSET: lysosomal enzyme trafficking factor; M6P: mannose 6-phosphate; MPRs: mannose-6-phosphate receptors, cation-dependent or -independent; MBTPS1/site-1 protease: membrane bound transcription factor peptidase, site 1; MLII: mucolipidosis type II; WT: wild-type.


Subject(s)
Mucolipidoses , Humans , Mucolipidoses/genetics , Mucolipidoses/metabolism , Mannose/metabolism , Autophagy , Lysosomes/metabolism , Hydrolases/metabolism , Receptor, IGF Type 2/metabolism , Cations/metabolism , Phosphotransferases/metabolism
4.
Science ; 378(6615): eabn5648, 2022 10 07.
Article in English | MEDLINE | ID: mdl-36074821

ABSTRACT

Lysosomes are key degradative compartments of the cell. Transport to lysosomes relies on GlcNAc-1-phosphotransferase-mediated tagging of soluble enzymes with mannose 6-phosphate (M6P). GlcNAc-1-phosphotransferase deficiency leads to the severe lysosomal storage disorder mucolipidosis II (MLII). Several viruses require lysosomal cathepsins to cleave structural proteins and thus depend on functional GlcNAc-1-phosphotransferase. We used genome-scale CRISPR screens to identify lysosomal enzyme trafficking factor (LYSET, also named TMEM251) as essential for infection by cathepsin-dependent viruses including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). LYSET deficiency resulted in global loss of M6P tagging and mislocalization of GlcNAc-1-phosphotransferase from the Golgi complex to lysosomes. Lyset knockout mice exhibited MLII-like phenotypes, and human pathogenic LYSET alleles failed to restore lysosomal sorting defects. Thus, LYSET is required for correct functioning of the M6P trafficking machinery and mutations in LYSET can explain the phenotype of the associated disorder.


Subject(s)
COVID-19 , Lysosomes , Mucolipidoses , Proteins , Animals , COVID-19/genetics , Cathepsins/metabolism , Humans , Lysosomes/metabolism , Mannose/metabolism , Mice , Mice, Knockout , Mucolipidoses/genetics , Mucolipidoses/metabolism , Proteins/genetics , Transferases (Other Substituted Phosphate Groups)
5.
Nat Commun ; 13(1): 5351, 2022 09 12.
Article in English | MEDLINE | ID: mdl-36096887

ABSTRACT

The mannose-6-phosphate (M6P) biosynthetic pathway for lysosome biogenesis has been studied for decades and is considered a well-understood topic. However, whether this pathway is regulated remains an open question. In a genome-wide CRISPR/Cas9 knockout screen, we discover TMEM251 as the first regulator of the M6P modification. Deleting TMEM251 causes mistargeting of most lysosomal enzymes due to their loss of M6P modification and accumulation of numerous undigested materials. We further demonstrate that TMEM251 localizes to the Golgi and is required for the cleavage and activity of GNPT, the enzyme that catalyzes M6P modification. In zebrafish, TMEM251 deletion leads to severe developmental defects including heart edema and skeletal dysplasia, which phenocopies Mucolipidosis Type II. Our discovery provides a mechanism for the newly discovered human disease caused by TMEM251 mutations. We name TMEM251 as GNPTAB cleavage and activity factor (GCAF) and its related disease as Mucolipidosis Type V.


Subject(s)
Membrane Proteins , Mucolipidoses , Zebrafish , Animals , Humans , Lysosomes/metabolism , Mannosephosphates/metabolism , Membrane Proteins/metabolism , Mucolipidoses/genetics , Mucolipidoses/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism , Zebrafish/metabolism
6.
EMBO Mol Med ; 14(9): e15377, 2022 09 07.
Article in English | MEDLINE | ID: mdl-35929194

ABSTRACT

Lysosomes are cell organelles that degrade macromolecules to recycle their components. If lysosomal degradative function is impaired, e.g., due to mutations in lysosomal enzymes or membrane proteins, lysosomal storage diseases (LSDs) can develop. LSDs manifest often with neurodegenerative symptoms, typically starting in early childhood, and going along with a strongly reduced life expectancy and quality of life. We show here that small molecule activation of the Ca2+ -permeable endolysosomal two-pore channel 2 (TPC2) results in an amelioration of cellular phenotypes associated with LSDs such as cholesterol or lipofuscin accumulation, or the formation of abnormal vacuoles seen by electron microscopy. Rescue effects by TPC2 activation, which promotes lysosomal exocytosis and autophagy, were assessed in mucolipidosis type IV (MLIV), Niemann-Pick type C1, and Batten disease patient fibroblasts, and in neurons derived from newly generated isogenic human iPSC models for MLIV and Batten disease. For in vivo proof of concept, we tested TPC2 activation in the MLIV mouse model. In sum, our data suggest that TPC2 is a promising target for the treatment of different types of LSDs, both in vitro and in-vivo.


Subject(s)
Lysosomal Storage Diseases , Mucolipidoses , Neuronal Ceroid-Lipofuscinoses , Animals , Child, Preschool , Humans , Lysosomes/metabolism , Mice , Mucolipidoses/genetics , Mucolipidoses/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Quality of Life
7.
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
8.
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
9.
Glycoconj J ; 38(6): 649-667, 2021 12.
Article in English | MEDLINE | ID: mdl-34686927

ABSTRACT

Sialidases catalyze the removal of sialic acid residues from glycoproteins, oligosaccharides, and sialylated glycolipids. Sialidase Neu4 is in the lysosome and has broad substrate specificity. Previously generated Neu4-/- mice were viable, fertile and lacked gross morphological abnormalities, but displayed a marked vacuolization and lysosomal storage in lung and spleen cells. In addition, we showed that there is an increased level of GD1a ganglioside and a markedly decreased level of GM1 ganglioside in the brain of Neu4-/- mice. In this study, we further explored whether sialidase Neu4 deficiency causes neuroinflammation. We demostrated that elevated level of GD1a and GT1b is associated with an increased level of LAMP1-positive lysosomal vesicles and Tunel-positive neurons correlated with alterations in the expression of cytokines and chemokines in adult Neu4-/- mice. Astrogliosis and microgliosis were also significantly enhanced in the hippocampus, and cerebellum. These changes in brain immunity were accompanied by motor impairment in these mice. Our results indicate that sialidase Neu4 is a novel mediator of an inflammatory response in the mouse brain due to the altered catabolism of gangliosides.


Subject(s)
Mucolipidoses , Neuraminidase/metabolism , Animals , Gangliosides/metabolism , Lysosomes/metabolism , Mice , Mucolipidoses/metabolism , Neuraminidase/chemistry , Neuroinflammatory Diseases , Substrate Specificity
10.
J Invest Dermatol ; 141(12): 2957-2965.e5, 2021 12.
Article in English | MEDLINE | ID: mdl-34116066

ABSTRACT

UDP-GlcNAc-1-phosphotransferase, a product of two separate genes (GNPTAB, GNPTG), is essential for the sorting and transportation of lysosomal enzymes to lysosomes. GNPTAB gene defects cause extracellular missorting of lysosomal enzymes resulting in lysosomal storage diseases, namely mucolipidosis type II and mucolipidosis type III alpha/beta, which is associated with hair discoloration. Yet, the physiological functions of GNPTAB in the control of hair follicle (HF) pigmentation remain unknown. To elucidate these, we have silenced GNPTAB in organ-cultured human HFs as a human ex vivo model for mucolipidosis type II. GNPTAB silencing profoundly inhibited intrafollicular melanin production, the correct sorting of melanosomes, tyrosinase activity, and HMB45 expression in the HF pigmentary unit and altered HF melanocyte morphology in situ. In isolated primary human HF melanocytes, GNPTAB knockdown significantly reduced melanogenesis, tyrosinase activity, and correct tyrosinase protein sorting as well as POMC expression and caused the expected lysosomal enzyme missorting in vitro. Moreover, transgenic mice overexpressing an inserted missense mutation corresponding to that seen in human mucolipidosis type II and mucolipidosis type III alpha/beta showed significantly reduced HF pigmentation, thus corroborating the in vivo relevance of our ex vivo and in vitro findings in the human system. This identifies GNPTAB as a clinically important enzymatic control of human HF pigmentation, likely by directly controlling tyrosinase sorting and POMC transcription in HF melanocytes.


Subject(s)
Gene Expression Regulation , Hair Follicle/metabolism , Hair/metabolism , Pigmentation , Transferases (Other Substituted Phosphate Groups)/metabolism , Animals , Female , Gene Silencing , Humans , Lysosomes/metabolism , Melanins/metabolism , Melanocytes/metabolism , Mice , Mice, Transgenic , Middle Aged , Monophenol Monooxygenase/metabolism , Mucolipidoses/metabolism , Mutation, Missense , Phenotype , Pigmentation Disorders/metabolism , Pro-Opiomelanocortin/metabolism , RNA, Small Interfering/metabolism , Scalp/metabolism , Skin/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics
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.
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
15.
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
16.
Proc Natl Acad Sci U S A ; 117(32): 19266-19275, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32703809

ABSTRACT

Mitochondria and lysosomes are critical for cellular homeostasis, and dysfunction of both organelles has been implicated in numerous diseases. Recently, interorganelle contacts between mitochondria and lysosomes were identified and found to regulate mitochondrial dynamics. However, whether mitochondria-lysosome contacts serve additional functions by facilitating the direct transfer of metabolites or ions between the two organelles has not been elucidated. Here, using high spatial and temporal resolution live-cell microscopy, we identified a role for mitochondria-lysosome contacts in regulating mitochondrial calcium dynamics through the lysosomal calcium efflux channel, transient receptor potential mucolipin 1 (TRPML1). Lysosomal calcium release by TRPML1 promotes calcium transfer to mitochondria, which was mediated by tethering of mitochondria-lysosome contact sites. Moreover, mitochondrial calcium uptake at mitochondria-lysosome contact sites was modulated by the outer and inner mitochondrial membrane channels, voltage-dependent anion channel 1 and the mitochondrial calcium uniporter, respectively. Since loss of TRPML1 function results in the lysosomal storage disorder mucolipidosis type IV (MLIV), we examined MLIV patient fibroblasts and found both altered mitochondria-lysosome contact dynamics and defective contact-dependent mitochondrial calcium uptake. Thus, our work highlights mitochondria-lysosome contacts as key contributors to interorganelle calcium dynamics and their potential role in the pathophysiology of disorders characterized by dysfunctional mitochondria or lysosomes.


Subject(s)
Calcium/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Mucolipidoses/metabolism , Transient Receptor Potential Channels/metabolism , Biological Transport , Humans , Lysosomes/genetics , Mitochondria/genetics , Mitochondrial Dynamics , Mucolipidoses/genetics , Transient Receptor Potential Channels/genetics
17.
J Am Soc Nephrol ; 31(8): 1796-1814, 2020 08.
Article in English | MEDLINE | ID: mdl-32641396

ABSTRACT

BACKGROUND: The mechanisms balancing proteostasis in glomerular cells are unknown. Mucolipidosis (ML) II and III are rare lysosomal storage disorders associated with mutations of the Golgi-resident GlcNAc-1-phosphotransferase, which generates mannose 6-phosphate residues on lysosomal enzymes. Without this modification, lysosomal enzymes are missorted to the extracellular space, which results in lysosomal dysfunction of many cell types. Patients with MLII present with severe skeletal abnormalities, multisystemic symptoms, and early death; the clinical course in MLIII is less progressive. Despite dysfunction of a major degradative pathway, renal and glomerular involvement is rarely reported, suggesting organ-specific compensatory mechanisms. METHODS: MLII mice were generated and compared with an established MLIII model to investigate the balance of protein synthesis and degradation, which reflects glomerular integrity. Proteinuria was assessed in patients. High-resolution confocal microscopy and functional assays identified proteins to deduce compensatory modes of balancing proteostasis. RESULTS: Patients with MLII but not MLIII exhibited microalbuminuria. MLII mice showed lysosomal enzyme missorting and several skeletal alterations, indicating that they are a useful model. In glomeruli, both MLII and MLIII mice exhibited reduced levels of lysosomal enzymes and enlarged lysosomes with abnormal storage material. Nevertheless, neither model had detectable morphologic or functional glomerular alterations. The models rebalance proteostasis in two ways: MLII mice downregulate protein translation and increase the integrated stress response, whereas MLIII mice upregulate the proteasome system in their glomeruli. Both MLII and MLIII downregulate the protein complex mTORC1 (mammalian target of rapamycin complex 1) signaling, which decreases protein synthesis. CONCLUSIONS: Severe lysosomal dysfunction leads to microalbuminuria in some patients with mucolipidosis. Mouse models indicate distinct compensatory pathways that balance proteostasis in MLII and MLIII.


Subject(s)
Kidney Glomerulus/metabolism , Mucolipidoses/metabolism , Proteinuria/prevention & control , Proteostasis/physiology , Albuminuria/etiology , Animals , Blood Urea Nitrogen , Cells, Cultured , Disease Models, Animal , Humans , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mucolipidoses/complications , Proteasome Endopeptidase Complex/physiology
18.
Biosci Rep ; 40(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32542330

ABSTRACT

Homologues of the putative dehydrogenase YjhC are found in operons involved in the metabolism of N-acetylneuraminate (Neu5Ac) or related compounds. We observed that purified recombinant YjhC forms Neu5Ac from two dehydrated forms of this compound, 2,7-anhydro-N-acetylneuraminate (2,7-AN) and 2-deoxy-2,3-didehydro-N-acetylneuraminate (2,3-EN) that are produced during the degradation of sialoconjugates by some sialidases. The conversion of 2,7-AN into Neu5Ac is reversible and reaches its equilibrium when the ratio of 2,7-AN to Neu5Ac is ≈1/6. The conversion of 2,3-EN is irreversible, leading to a mixture of Neu5Ac and 2,7-AN. NMR analysis of the reaction catalysed by YjhC on 2,3-EN indicated that Neu5Ac was produced as the α-anomer. All conversions require NAD+ as a cofactor, which is regenerated in the reaction. They appear to involve the formation of keto (presumably 4-keto) intermediates of 2,7-AN, 2,3-EN and Neu5Ac, which were detected by liquid chromatography-mass spectrometry (LC-MS). The proposed reaction mechanism is reminiscent of the one catalysed by family 4 ß-glycosidases, which also use NAD+ as a cofactor. Both 2,7-AN and 2,3-EN support the growth of Escherichia coli provided the repressor NanR, which negatively controls the expression of the yjhBC operons, has been inactivated. Inactivation of either YjhC or YjhB in NanR-deficient cells prevents the growth on 2,7-AN and 2,3-EN. This confirms the role of YjhC in 2,7-AN and 2,3-EN metabolism and indicates that transport of 2,7-AN and 2,3-EN is carried out by YjhB, which is homologous to the Neu5Ac transporter NanT.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Mucolipidoses/metabolism , N-Acetylneuraminic Acid/metabolism , Oxidoreductases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Kinetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , NAD/metabolism , Oxidoreductases/genetics , Substrate Specificity
19.
Dis Model Mech ; 13(7)2020 07 30.
Article in English | MEDLINE | ID: mdl-32586947

ABSTRACT

Mucolipidosis type IV (MLIV) is a lysosomal disease caused by mutations in the MCOLN1 gene that encodes the endolysosomal transient receptor potential channel mucolipin-1, or TRPML1. MLIV results in developmental delay, motor and cognitive impairments, and vision loss. Brain abnormalities include thinning and malformation of the corpus callosum, white-matter abnormalities, accumulation of undegraded intracellular 'storage' material and cerebellar atrophy in older patients. Identification of the early events in the MLIV course is key to understanding the disease and deploying therapies. The Mcoln1-/- mouse model reproduces all major aspects of the human disease. We have previously reported hypomyelination in the MLIV mouse brain. Here, we investigated the onset of hypomyelination and compared oligodendrocyte maturation between the cortex/forebrain and cerebellum. We found significant delays in expression of mature oligodendrocyte markers Mag, Mbp and Mobp in the Mcoln1-/- cortex, manifesting as early as 10 days after birth and persisting later in life. Such delays were less pronounced in the cerebellum. Despite our previous finding of diminished accumulation of the ferritin-bound iron in the Mcoln1-/- brain, we report no significant changes in expression of the cytosolic iron reporters, suggesting that iron-handling deficits in MLIV occur in the lysosomes and do not involve broad iron deficiency. These data demonstrate very early deficits of oligodendrocyte maturation and critical regional differences in myelination between the forebrain and cerebellum in the mouse model of MLIV. Furthermore, they establish quantitative readouts of the MLIV impact on early brain development, useful to gauge efficacy in pre-clinical trials.


Subject(s)
Brain/metabolism , Cell Differentiation , Mucolipidoses/metabolism , Oligodendroglia/metabolism , Transient Receptor Potential Channels/metabolism , Age Factors , Animals , Brain/pathology , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Mice, Inbred C57BL , Mice, Knockout , Mucolipidoses/genetics , Mucolipidoses/pathology , Myelin Basic Protein/metabolism , Myelin Proteins/metabolism , Myelin-Associated Glycoprotein/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/pathology , Oligodendroglia/pathology , Prosencephalon/metabolism , Prosencephalon/pathology , Transient Receptor Potential Channels/genetics
20.
Biomolecules ; 10(5)2020 04 26.
Article in English | MEDLINE | ID: mdl-32357547

ABSTRACT

Mucolipidosis II (ML-II) is a lysosomal disease caused by defects in the carbohydrate-dependent sorting of soluble hydrolases to lysosomes. Altered growth factor signaling has been identified as a contributor to the phenotypes associated with ML-II and other lysosomal disorders but an understanding of how these signaling pathways are affected is still emerging. Here, we investigated transforming growth factor beta 1 (TGFß1) signaling in the context of ML-II patient fibroblasts, observing decreased TGFß1 signaling that was accompanied by impaired TGFß1-dependent wound closure. We found increased intracellular latent TGFß1 complexes, caused by reduced secretion and stable localization in detergent-resistant lysosomes. Sortilin, a sorting receptor for hydrolases and TGFß-related cytokines, was upregulated in ML-II fibroblasts as well as GNPTAB-null HeLa cells, suggesting a mechanism for inappropriate lysosomal targeting of TGFß. Co-expression of sortilin and TGFß in HeLa cells resulted in reduced TGFß1 secretion. Elevated sortilin levels correlated with normal levels of cathepsin D in ML-II cells, consistent with a compensatory role for this receptor in lysosomal hydrolase targeting. Collectively, these data support a model whereby sortilin upregulation in cells with lysosomal storage maintains hydrolase sorting but suppresses TGFß1 secretion through increased lysosomal delivery. These findings highlight an unexpected link between impaired lysosomal sorting and altered growth factor bioavailability.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Mucolipidoses/metabolism , Transforming Growth Factor beta/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Cathepsin D/metabolism , Cell Line , Cells, Cultured , Fibroblasts/metabolism , HeLa Cells , Humans , Lysosomes/metabolism , Protein Transport , Signal Transduction , Transferases (Other Substituted Phosphate Groups)/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...