Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38928321

ABSTRACT

Glucocerebrosidase (GCase) is a lysosomal enzyme that catalyzes the breakdown of glucosylceramide in the presence of its activator saposin C (SapC). SapC arises from the proteolytical cleavage of prosaposin (encoded by PSAP gene), which gives rise to four saposins. GCase is targeted to the lysosomes by LIMP-2, encoded by SCARB2 gene. GCase deficiency causes Gaucher Disease (GD), which is mainly due to biallelic pathogenetic variants in the GCase-encoding gene, GBA1. However, impairment of GCase activity can be rarely caused by SapC or LIMP-2 deficiencies. We report a new case of LIMP-2 deficiency and a new case of SapC deficiency (missing all four saposins, PSAP deficiency), and measured common biomarkers of GD and GCase activity. Glucosylsphingosine and chitotriosidase activity in plasma were increased in GCase deficiencies caused by PSAP and GBA1 mutations, whereas SCARB2-linked deficiency showed only Glucosylsphingosine elevation. GCase activity was reduced in fibroblasts and leukocytes: the decrease was sharper in GBA1- and SCARB2-mutant fibroblasts than PSAP-mutant ones; LIMP-2-deficient leukocytes displayed higher residual GCase activity than GBA1-mutant ones. Finally, we demonstrated that GCase mainly undergoes proteasomal degradation in LIMP-2-deficient fibroblasts and lysosomal degradation in PSAP-deficient fibroblasts. Thus, we analyzed the differential biochemical profile of GCase deficiencies due to the ultra-rare PSAP and SCARB2 biallelic pathogenic variants in comparison with the profile observed in GBA1-linked GCase deficiency.


Subject(s)
Gaucher Disease , Glucosylceramidase , Lysosomal Membrane Proteins , Receptors, Scavenger , Saposins , Glucosylceramidase/genetics , Glucosylceramidase/deficiency , Glucosylceramidase/metabolism , Humans , Gaucher Disease/genetics , Gaucher Disease/metabolism , Saposins/deficiency , Saposins/genetics , Saposins/metabolism , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/genetics , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Fibroblasts/metabolism , Mutation , Lysosomes/metabolism , Lysosomes/enzymology , Hexosaminidases/metabolism , Hexosaminidases/genetics , Hexosaminidases/deficiency , Male , Female
2.
Mol Genet Metab ; 129(2): 161-164, 2020 02.
Article in English | MEDLINE | ID: mdl-31439510

ABSTRACT

Saposin A is a post-translation product of the prosaposin (PSAP) gene that serves as an activator protein of the galactocerebrosidase (GALC) enzyme, and is necessary for the degradation of certain glycosphingolipids. Deficiency of saposin A leads to a clinical picture identical to that of early-infantile Krabbe disease caused by GALC enzyme deficiency. Galactosylsphingosine, also known as psychosine, is a substrate of the GALC enzyme that is known to be elevated in classic Krabbe disease. We present the case of an 18-month-old male with clinical and radiological findings concerning for Krabbe disease who had preserved GALC enzyme activity and negative GALC gene sequencing, but was found to have a homozygous variant, c.257 T > A (p.I86N), in the saposin A peptide of PSAP. Psychosine determination on dried blood spot at 18 months of age was elevated to 12 nmol/L (normal <3 nmol/L). We present this case to add to the literature on the rare diagnosis of atypical Krabbe disease due to saposin A deficiency, to report a novel presumed pathogenic variant within PSAP, and to suggest that individuals with saposin A deficiency may have elevated levels of psychosine, similar to children with classic Krabbe disease due to GALC deficiency.


Subject(s)
Galactosylceramidase/genetics , Homozygote , Leukodystrophy, Globoid Cell/diagnostic imaging , Psychosine/blood , Saposins/deficiency , Dried Blood Spot Testing , Genetic Variation , Humans , Infant , Leukodystrophy, Globoid Cell/blood , Leukodystrophy, Globoid Cell/genetics , Magnetic Resonance Imaging , Male , Saposins/blood , Saposins/genetics
3.
Neuropediatrics ; 50(5): 318-321, 2019 10.
Article in English | MEDLINE | ID: mdl-31319425

ABSTRACT

Metachromatic leukodystrophy (MLD) is a rare sphingolipid storage disorder caused by arylsulfatase A (ARSA) deficiency, resulting in central and peripheral demyelination. However, an uncommon form of MLD caused by saposin B deficiency is also described (around 10 mutations reported till date). MLD is a systemic disorder affecting the central and peripheral nervous system, gall bladder, and kidneys. Acute flaccid paralysis as the initial clinical presentation is previously known in ARSA-deficient MLD. Hereby, we report a child with acute flaccid paralysis with brain magnetic resonance imaging showing nonspecific periventricular leukodystrophy. He had progressive cognitive decline with gall bladder polyposis. ARSA levels were within normal limits. Leukodystrophy gene panel revealed a homozygous pathogenic deletion (Lys227del variant) in prosaposin (PSAP) gene. Hence, a final diagnosis of saposin B-deficient MLD was established. The index case highlights the importance of clinical and electrophysiological clues in the diagnosis of such atypical presentations of MLD.


Subject(s)
Leukodystrophy, Metachromatic/diagnosis , Paralysis/diagnosis , Saposins/deficiency , Abdomen/diagnostic imaging , Brain/diagnostic imaging , Child, Preschool , Diagnosis, Differential , Humans , Leukodystrophy, Metachromatic/complications , Leukodystrophy, Metachromatic/genetics , Male , Mutation , Paralysis/complications , Paralysis/genetics , Saposins/genetics
5.
Sci Rep ; 9(1): 5571, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30944381

ABSTRACT

Gaucher disease is caused by mutations in GBA1 encoding acid ß-glucosidase (GCase). Saposin C enhances GCase activity and protects GCase from intracellular proteolysis. Structure simulations indicated that the mutant GCases, N370S (0 S), V394L (4L) and D409V(9V)/H(9H), had altered function. To investigate the in vivo function of Gba1 mutants, mouse models were generated by backcrossing the above homozygous mutant GCase mice into Saposin C deficient (C*) mice. Without saposin C, the mutant GCase activities in the resultant mouse tissues were reduced by ~50% compared with those in the presence of Saposin C. In contrast to 9H and 4L mice that have normal histology and life span, the 9H;C* and 4L;C* mice had shorter life spans. 9H;C* mice developed significant visceral glucosylceramide (GC) and glucosylsphingosine (GS) accumulation (GC¼GS) and storage macrophages, but lesser GC in the brain, compared to 4L;C* mice that presents with a severe neuronopathic phenotype and accumulated GC and GS primarily in the brain. Unlike 9V mice that developed normally for over a year, 9V;C* pups had a lethal skin defect as did 0S;C* mice resembled that of 0S mice. These variant Gaucher disease mouse models presented a mutation specific phenotype and underscored the in vivo role of Saposin C in the modulation of Gaucher disease.


Subject(s)
Gaucher Disease/genetics , Glucosylceramidase/genetics , Mutation/genetics , Saposins/deficiency , beta-Glucosidase/genetics , Animals , Brain/pathology , Disease Models, Animal , Glucosylceramides/genetics , Mice , Mice, Inbred C57BL , Phenotype
6.
Blood Cells Mol Dis ; 68: 60-65, 2018 02.
Article in English | MEDLINE | ID: mdl-28457694

ABSTRACT

Gaucher disease is mainly caused by the deficiency of lysosomal acid ß-glucosidase. Gaucher disease caused by the deficiency of saposin C is rare. Here we report a patient mainly presenting with hepatosplenomegaly, thrombocytopenia and anemia. EEG examination revealed increased theta waves. Gaucher cells identified in his bone marrow and the highly elevated plasma chitotriosidase activity and glucosylsphingosine supported a diagnosis of Gaucher disease. However, the leukocyte ß-glucosidase activity was in a normal range. Sanger sequencing revealed a novel maternal exonic mutation c.1133C>G (p.Pro378Arg) in exon 10 of the PSAP gene, which codes the Sap C domain of PSAP protein. To search for other underlying mutations in this patient, whole genome sequencing was applied and revealed a deletion involving exon 2 to 7 of PSAP gene. The deletion appears as a de novo event on paternal chromosome. We concluded that biallelic mutations of PSAP gene were the cause of this patient's Gaucher disease. Our finding expands the mutation spectrum of Gaucher disease with saposin C deficiency.


Subject(s)
Gaucher Disease/etiology , Gaucher Disease/genetics , Point Mutation , Saposins/deficiency , Base Sequence , Child , Exons , Gene Deletion , Humans , Male , Mutation, Missense , Saposins/genetics , Sequence Deletion
8.
Neurobiol Dis ; 98: 77-87, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27913291

ABSTRACT

Saposin deficiency is a childhood neurodegenerative lysosomal storage disorder (LSD) that can cause premature death within three months of life. Saposins are activator proteins that promote the function of lysosomal hydrolases that mediate the degradation of sphingolipids. There are four saposin proteins in humans, which are encoded by the prosaposin gene. Mutations causing an absence or impaired function of individual saposins or the whole prosaposin gene lead to distinct LSDs due to the storage of different classes of sphingolipids. The pathological events leading to neuronal dysfunction induced by lysosomal storage of sphingolipids are as yet poorly defined. We have generated and characterised a Drosophila model of saposin deficiency that shows striking similarities to the human diseases. Drosophila saposin-related (dSap-r) mutants show a reduced longevity, progressive neurodegeneration, lysosomal storage, dramatic swelling of neuronal soma, perturbations in sphingolipid catabolism, and sensory physiological deterioration. Our data suggests a genetic interaction with a calcium exchanger (Calx) pointing to a possible calcium homeostasis deficit in dSap-r mutants. Together these findings support the use of dSap-r mutants in advancing our understanding of the cellular pathology implicated in saposin deficiency and related LSDs.


Subject(s)
Disease Models, Animal , Drosophila Proteins/deficiency , Lysosomal Storage Diseases, Nervous System/metabolism , Neurodegenerative Diseases/metabolism , Saposins/deficiency , Aging/metabolism , Aging/pathology , Animals , Animals, Genetically Modified , Antiporters/genetics , Antiporters/metabolism , Brain/metabolism , Brain/pathology , Calcium/metabolism , Ceramides/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Homeostasis/physiology , Lysosomal Storage Diseases, Nervous System/pathology , Neurodegenerative Diseases/pathology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Phenotype , Saposins/genetics , Sphingosine/metabolism , Survival Analysis
9.
Clin Genet ; 90(3): 220-9, 2016 09.
Article in English | MEDLINE | ID: mdl-26831127

ABSTRACT

Prosaposin (PSAP) deficiency is an ultra-rare, fatal infantile lysosomal storage disorder (LSD) caused by variants in the PSAP gene, with seven subjects reported so far. Here, we provide the clinical, biochemical and molecular characterization of two additional PSAP deficiency cases. Lysoplex, a targeted resequencing approach was utilized to identify the variant in the first patient, while quantification of plasma lysosphingolipids (lysoSLs), assessed by liquid chromatography mass spectrometry (LC-MS/MS) and brain magnetic resonance imaging (MRI), followed by Sanger sequencing allowed to attain diagnosis in the second case. Functional studies were carried out on patients' fibroblast lines to explore the functional impact of variants. The two patients were homozygous for two different truncating PSAP mutations (c.895G>T, p.Glu299*; c.834_835delGA, p.Glu278Aspfs*27). Both variants led to a complete lack of processed transcript. LC-MS/MS and brain MRI analyses consistently provided a distinctive profile in the two children. Quantification of specific plasma lysoSLs revealed elevated levels of globotriaosylsphingosine (LysoGb3) and glucosylsphingosine (GlSph), and accumulation of autophagosomes, due to a decreased autophagic flux, was observed. This report documents the successful use of plasma lysoSLs profiling in the PSAP deficiency diagnosis, as a reliable and informative tool to obtain a preliminary information in infantile cases with complex traits displaying severe neurological signs and visceral involvement.


Subject(s)
Brain/metabolism , Leukodystrophy, Metachromatic/genetics , Saposins/deficiency , Sphingolipids/blood , Brain/diagnostic imaging , Brain/pathology , Chromatography, Liquid , Consanguinity , Female , Humans , Infant , Leukodystrophy, Metachromatic/blood , Leukodystrophy, Metachromatic/diagnostic imaging , Leukodystrophy, Metachromatic/pathology , Magnetic Resonance Imaging , Male , Mutation , Saposins/blood , Saposins/genetics
10.
J Cell Biol ; 210(6): 991-1002, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26370502

ABSTRACT

Mutations in the progranulin (PGRN) gene have been linked to two distinct neurodegenerative diseases, frontotemporal lobar degeneration (FTLD) and neuronal ceroid lipofuscinosis (NCL). Accumulating evidence suggests a critical role of PGRN in lysosomes. However, how PGRN is trafficked to lysosomes is still not clear. Here we report a novel pathway for lysosomal delivery of PGRN. We found that prosaposin (PSAP) interacts with PGRN and facilitates its lysosomal targeting in both biosynthetic and endocytic pathways via the cation-independent mannose 6-phosphate receptor and low density lipoprotein receptor-related protein 1. PSAP deficiency in mice leads to severe PGRN trafficking defects and a drastic increase in serum PGRN levels. We further showed that this PSAP pathway is independent of, but complementary to, the previously identified PGRN lysosomal trafficking mediated by sortilin. Collectively, our results provide new understanding on PGRN trafficking and shed light on the molecular mechanisms behind FTLD and NCL caused by PGRN mutations.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Neurons/metabolism , Saposins/metabolism , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Animals , Endocytosis , Genotype , Granulins , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Low Density Lipoprotein Receptor-Related Protein-1 , Lysosomes/pathology , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration , Neurons/pathology , Phenotype , Progranulins , Protein Binding , Protein Transport , Receptor, IGF Type 2/metabolism , Receptors, LDL/metabolism , Saposins/deficiency , Saposins/genetics , Transfection , Tumor Suppressor Proteins/metabolism
11.
Hum Mol Genet ; 24(15): 4198-211, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25926625

ABSTRACT

Saposin (Sap) C deficiency is a rare variant form of Gaucher disease caused by impaired Sap C expression or accelerated degradation, and associated with accumulation of glucosylceramide and other lipids in the endo/lysosomal compartment. No effective therapies are currently available for the treatment of Sap C deficiency. We previously reported that a reduced amount and enzymatic activity of cathepsin (Cath) B and Cath D, and defective autophagy occur in Sap C-deficient fibroblasts. Here, we explored the use of two compounds, BCM-95, a curcumin derivative, and (2-hydroxypropyl)-ß-cyclodextrin (HP-ß-CD), to improve lysosomal function of Sap C-deficient fibroblasts. Immunofluorescence and biochemical studies documented that each compound promotes an increase of the expression levels and activities of Cath B and Cath D, and efficient clearance of cholesterol (Chol) and ceramide (Cer) in lysosomes. We provide evidence that BCM-95 and HP-ß-CD enhance lysosomal function promoting autophagic clearance capacity and lysosome reformation. Our findings suggest a novel pharmacological approach to Sap C deficiency directed to treat major secondary pathological aspects in this disorder.


Subject(s)
Curcumin/adverse effects , Gaucher Disease/drug therapy , Saposins/genetics , beta-Cyclodextrins/administration & dosage , Autophagy/drug effects , Cathepsin B/biosynthesis , Cathepsin B/genetics , Cathepsin D/biosynthesis , Cathepsin D/genetics , Curcumin/analogs & derivatives , Fibroblasts/metabolism , Fibroblasts/pathology , Gaucher Disease/genetics , Gaucher Disease/pathology , Glucosylceramides/metabolism , Humans , Lysosomes/genetics , Lysosomes/pathology , Saposins/deficiency
12.
J Neurosci ; 35(7): 3263-75, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25698761

ABSTRACT

Saposin B (Sap B) is an essential activator protein for arylsulfatase A in the hydrolysis of sulfatide, a lipid component of myelin. To study Sap B's role in hearing and balance, a Sap B-deficient (B(-/-)) mouse was evaluated. At both light and electron microscopy (EM) levels, inclusion body accumulation was seen in satellite cells surrounding spiral ganglion (SG) neurons from postnatal month 1 onward, progressing into large vacuoles preceding satellite cell degeneration, and followed by SG degeneration. EM also revealed reduced or absent myelin sheaths in SG neurons from postnatal month 8 onwards. Hearing loss was initially seen at postnatal month 6 and progressed thereafter for frequency-specific stimuli, whereas click responses became abnormal from postnatal month 13 onward. The progressive hearing loss correlated with the accumulation of inclusion bodies in the satellite cells and their subsequent degeneration. Outer hair cell numbers and efferent function measures (distortion product otoacoustic emissions and contralateral suppression) were normal in the B(-/-) mice throughout this period. Alcian blue staining of SGs demonstrated that these inclusion bodies corresponded to sulfatide accumulation. In contrast, changes in the vestibular system were much milder, but caused severe physiologic deficits. These results demonstrate that loss of Sap B function leads to progressive sulfatide accumulation in satellite cells surrounding the SG neurons, leading to satellite cell degeneration and subsequent SG degeneration with a resultant loss of hearing. Relative sparing of the efferent auditory and vestibular neurons suggests that alternate glycosphingolipid metabolic pathways predominate in these other systems.


Subject(s)
Hearing Disorders/etiology , Leukodystrophy, Metachromatic/complications , Leukodystrophy, Metachromatic/genetics , Nerve Degeneration/etiology , Saposins/deficiency , Satellite Cells, Perineuronal/pathology , Spiral Ganglion/pathology , Acoustic Stimulation , Animals , Cell Death/genetics , Cochlea/metabolism , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Functional Laterality , Hearing Tests , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Otoacoustic Emissions, Spontaneous/genetics , Saposins/genetics , Spiral Ganglion/ultrastructure , Swimming/psychology
13.
Hum Mol Genet ; 23(21): 5814-26, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24925315

ABSTRACT

Saposin (Sap) C is an essential cofactor for the lysosomal degradation of glucosylceramide (GC) by glucosylceramidase (GCase) and its functional impairment underlies a rare variant form of Gaucher disease (GD). Sap C promotes rearrangement of lipid organization in lysosomal membranes favoring substrate accessibility to GCase. It is characterized by six invariantly conserved cysteine residues involved in three intramolecular disulfide bonds, which make the protein remarkably stable to acid environment and degradation. Five different mutations (i.e. p.C315S, p.342_348FDKMCSKdel, p.L349P, p.C382G and p.C382F) have been identified to underlie Sap C deficiency. The molecular mechanism by which these mutations affect Sap C function, however, has not been delineated in detail. Here, we characterized biochemically and functionally four of these gene lesions. We show that all Sap C mutants are efficiently produced, and exhibit lipid-binding properties, modulatory behavior on GCase activity and subcellular localization comparable with those of the wild-type protein. We then delineated the structural rearrangement of these mutants, documenting that most proteins assume diverse aberrant disulfide bridge arrangements, which result in a substantial diminished half-life, and rapid degradation via autophagy. These findings further document the paramount importance of disulfide bridges in the stability of Sap C and provide evidence that accelerated degradation of the Sap C mutants is the underlying pathogenetic mechanism of Sap C deficiency.


Subject(s)
Gaucher Disease/genetics , Gaucher Disease/metabolism , Lysosomes/metabolism , Mutation , Saposins/genetics , Saposins/metabolism , Amino Acid Sequence , Animals , Cell Line , Gene Expression , Humans , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Stability , Protein Transport , Proteolysis , Saposins/chemistry , Saposins/deficiency , Sequence Alignment
14.
J Biol Chem ; 289(29): 20026-38, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24872419

ABSTRACT

A proteomics-based search for molecules interacting with caspase-14 identified prosaposin and epidermal mesotrypsin as candidates. Prosaposin is a precursor of four sphingolipid activator proteins (saposins A-D) that are essential for lysosomal hydrolysis of sphingolipids. Thus, we hypothesized that caspase-14 and mesotrypsin participate in processing of prosaposin. Because we identified a saposin A sequence as an interactor with these proteases, we prepared a specific antibody to saposin A and focused on saposin A-related physiological reactions. We found that mesotrypsin generated saposins A-D from prosaposin, and mature caspase-14 contributed to this process by activating mesotrypsinogen to mesotrypsin. Knockdown of these proteases markedly down-regulated saposin A synthesis in skin equivalent models. Saposin A was localized in granular cells, whereas prosaposin was present in the upper layer of human epidermis. The proximity ligation assay confirmed interaction between prosaposin, caspase-14, and mesotrypsin in the granular layer. Oil Red staining showed that the lipid envelope was significantly reduced in the cornified layer of skin from saposin A-deficient mice. Ultrastructural studies revealed severely disorganized cornified layer structure in both prosaposin- and saposin A-deficient mice. Overall, our results indicate that epidermal mesotrypsin and caspase-14 work cooperatively in prosaposin processing. We propose that they thereby contribute to permeability barrier formation in vivo.


Subject(s)
Caspases/metabolism , Saposins/metabolism , Skin/metabolism , Trypsin/metabolism , Animals , Caspases/genetics , Cells, Cultured , Gene Knockdown Techniques , Humans , Keratinocytes/metabolism , Mice , Mice, Knockout , Models, Biological , Permeability , Protein Processing, Post-Translational , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saposins/deficiency , Saposins/genetics , Skin/ultrastructure , Trypsin/genetics
15.
PLoS One ; 8(11): e80032, 2013.
Article in English | MEDLINE | ID: mdl-24244600

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy caused by a mutation in the X-linked dystrophin gene induces metabolic and structural disorders in the brain. A lack of dystrophin in brain structures is involved in impaired cognitive function. Prosaposin (PS), a neurotrophic factor, is abundant in the choroid plexus and various brain regions. We investigated whether PS serves as a link between dystrophin loss and gross and/or ultrastructural brain abnormalities. METHODOLOGY/PRINCIPAL FINDINGS: The distribution of PS in the brains of juvenile and adult mdx mice was investigated by immunochemistry, Western blotting, and in situ hybridization. Immunochemistry revealed lower levels of PS in the cytoplasm of neurons of the cerebral cortex, hippocampus, cerebellum, and choroid plexus in mdx mice. Western blotting confirmed that PS levels were lower in these brain regions in both juveniles and adults. Even with low PS production in the choroids plexus, there was no significant PS decrease in cerebrospinal fluid (CSF). In situ hybridization revealed that the primary form of PS mRNA in both normal and mdx mice was Pro+9, a secretory-type PS, and the hybridization signals for Pro+9 in the above-mentioned brain regions were weaker in mdx mice than in normal mice. We also investigated mitogen-activated protein kinase signalling. Stronger activation of ERK1/2 was observed in mdx mice, ERK1/2 activity was positively correlated with PS activity, and exogenous PS18 stimulated both p-ERK1/2 and PS in SH-SY5Y cells. CONCLUSIONS/SIGNIFICANCE: Low levels of PS and its receptors suggest the participation of PS in some pathological changes in the brains of mdx mice.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Saposins/genetics , Signal Transduction , Age Factors , Animals , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Choroid Plexus/metabolism , Choroid Plexus/pathology , Disease Models, Animal , Dystrophin/deficiency , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred mdx , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Muscular Dystrophy, Duchenne/cerebrospinal fluid , Muscular Dystrophy, Duchenne/pathology , Neurons/metabolism , Neurons/pathology , Saposins/deficiency
16.
Autophagy ; 9(7): 1115-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23697974

ABSTRACT

Combined saposin A and saposin B deficiency (AB(-/-)) was created in mice by knock-in of point mutations into the saposin A and B domains of the Psap (encoding prosaposin) locus. PSAP is the precursor of saposin A, saposin B and two other members, saposin C and saposin D. Those four saposins have multiple functions including their roles as glycosphingolipid activator proteins in a lysosomal glycosphingolipid degradation pathway. Saposin A participates in the removal of galactose from galactosylceramide and galactosylsphingosine by enhancing ß-galactosylceramidase activity. Saposin B has lipid binding properties and is involved in glycosphingolipid metabolism by presenting the substrates to specific enzymes for degradation, i.e., sulfatide to ARSA/arylsulfatase A, lactosylceramide to GALC/GM-1-ß-galactosylceramidase, and globotriaosylceramide to GLA/α-galactosidase. Galactosylceramide and sulfatide are myelin glycosphingolipids involved in carbohydrate interaction between synapses. The AB(-/-) mice develop accumulation of multiple glycosphingolipids in various organs. Sulfatide and galactosylsphingosine, a deacylated form of galactosylceramide, are the major substrates accumulated in the CNS of AB(-/-) mice. The latter is a toxic metabolite to oligodendrocytes and results in demyelination and cell death.


Subject(s)
Autophagy , Saposins/deficiency , Saposins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain Stem/metabolism , Brain Stem/pathology , Brain Stem/ultrastructure , Heat-Shock Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Phagosomes/metabolism , Sequestosome-1 Protein
17.
Hum Mol Genet ; 22(12): 2435-50, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23446636

ABSTRACT

Individual saposin A (A-/-) and saposin B (B-/-)-deficient mice show unique phenotypes caused by insufficient degradation of myelin-related glycosphingolipids (GSLs): galactosylceramide and galactosylsphingosine and sulfatide, respectively. To gain insight into the interrelated functions of saposins A and B, combined saposin AB-deficient mice (AB-/-) were created by knock-in point mutations into the saposins A and B domains on the prosaposin locus. Saposin A and B proteins were undetectable in AB-/- mice, whereas prosaposin, saposin C and saposin D were expressed near wild-type (WT) levels. AB-/- mice developed neuromotor deterioration at >61 days and exhibited abnormal locomotor activity and enhanced tremor. AB-/- mice (~96 days) lived longer than A-/- mice (~85 days), but shorter than B-/- mice (~644 days). Storage materials were observed in Schwann cells and neuronal processes by electron microscopy. Accumulation of p62 and increased levels of LC3-II were detected in the brainstem suggesting altered autophagy. GSL analyses by (liquid chromatography) LC/MS identified substantial increases in lactosylceramide in AB-/- mouse livers. Sulfatide accumulated, but galactosylceramide remained at WT levels, in the AB-/- mouse brains and kidneys. Brain galactosylsphingosine in AB-/- mice was ~68% of that in A-/- mice. These findings indicate that combined saposins A and B deficiencies attenuated GalCer-ß-galactosylceramidase and GM1-ß-galactosidase functions in the degradation of lactosylceramide preferentially in the liver. Blocking sulfatide degradation from the saposin B deficiency diminished galactosylceramide accumulation in the brain and kidney and galctosylsphingosine in the brain. These analyses of AB-/- mice continue to delineate the tissue differential interactions of saposins in GSL metabolism.


Subject(s)
Glycosphingolipids/metabolism , Nervous System Diseases/metabolism , Saposins/deficiency , Animals , Brain/metabolism , Female , Galactosylceramidase/metabolism , Humans , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Nervous System Diseases/enzymology , Nervous System Diseases/genetics , Nervous System Diseases/psychology , Organ Specificity , Phenotype , Saposins/genetics , beta-Galactosidase/metabolism
18.
Autophagy ; 9(2): 241-3, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23108186

ABSTRACT

Saposin C deficiency, a rare variant form of Gaucher disease, is due to mutations in the prosaposin gene (PSAP) affecting saposin C expression and/or function. We previously reported that saposin C mutations affecting one cysteine residue result in autophagy dysfunction. We further demonstrated that the accumulation of autophagosomes, observed in saposin C-deficient fibroblasts, is due to an impairment of autolysosome degradation, partially caused by the reduced amount and enzymatic activity of CTSB (cathepsin B) and CTSD (cathepsin D). The restoration of both proteases in pathological fibroblasts results in almost completely recovery of autophagic flux and lysosome homeostasis.


Subject(s)
Autophagy , Cathepsin B/metabolism , Cathepsin D/metabolism , Saposins/deficiency , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Models, Biological , Saposins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
19.
Hum Mol Genet ; 21(23): 5159-73, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22949512

ABSTRACT

Saposin (Sap) C deficiency, a rare variant form of Gaucher disease, is due to mutations in the Sap C coding region of the prosaposin (PSAP) gene. Sap C is required as an activator of the lysosomal enzyme glucosylceramidase (GCase), which catalyzes glucosylceramide (GC) degradation. Deficit of either GCase or Sap C leads to the accumulation of undegraded GC and other lipids in lysosomes of monocyte/macrophage lineage. Recently, we reported that Sap C mutations affecting a cysteine residue result in increased autophagy. Here, we characterized the basis for the autophagic dysfunction. We analyzed Sap C-deficient and GCase-deficient fibroblasts and observed that autophagic disturbance was only associated with lack of Sap C. By a combined fluorescence microscopy and biochemical studies, we demonstrated that the accumulation of autophagosomes in Sap C-deficient fibroblasts is not due to enhanced autophagosome formation but to delayed degradation of autolysosomes caused, in part, to decreased amount and reduced enzymatic activity of cathepsins B and D. On the contrary, in GCase-deficient fibroblasts, the protein level and enzymatic activity of cathepsin D were comparable with control fibroblasts, whereas those of cathepsin B were almost doubled. Moreover, the enhanced expression of both these lysosomal proteases in Sap C-deficient fibroblasts resulted in close to functional autophagic degradation. Our data provide a novel example of altered autophagy as secondary event resulting from insufficient lysosomal function.


Subject(s)
Autophagy/genetics , Cathepsin B/genetics , Cathepsin B/metabolism , Cathepsin D/genetics , Cathepsin D/metabolism , Fibroblasts/metabolism , Saposins/deficiency , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Beclin-1 , Cell Line , Enzyme Activation , Fibroblasts/drug effects , Gaucher Disease/genetics , Gaucher Disease/metabolism , Gene Expression , Humans , Lysosomes/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Saposins/genetics , Sirolimus/pharmacology , Ubiquitin-Activating Enzymes/metabolism
20.
Neurosci Res ; 72(4): 296-305, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22326583

ABSTRACT

Prosaposin, a precursor of four glycoprotein activators (saposin A, B, C and D) for lysosomal hydrolases, has previously been shown to be important for normal adult cochlear innervation and the maintenance of normal hearing. In these studies, we now investigate prosaposin in normal vestibular epithelium and the functional impairment of balance caused by prosaposin ablation. In normal mice, prosaposin is localized to all 3 vestibular end-organs (ampullae, saccule, and utricle) and Scarpa's ganglion by RT-PCR, Western blot analysis and immunofluorescence. Ablation of prosaposin function caused severe vestibular dysfunction on a battery of behavioral tasks. Histologically, the KO mice demonstrated an exuberant cellular proliferation below the vestibular hair cells with disruption of the supporting cells. Electron microscopy further demonstrated inclusion bodies and cellular proliferation disturbing the normal neuroepithelial structure of the vestibular end-organs. Lastly, immunofluorescence (neurofilament 200 and synaptophysin) staining suggests that this cellular proliferation corresponds to afferent and efferent neuronal overgrowth. These data suggest that prosaposin plays a role not only in the maintenance of normal hearing but also an important role in the neuronal maturation processes of the vestibular sensory epithelium and the maintenance of normal vestibular system function.


Subject(s)
Saposins/deficiency , Vestibule, Labyrinth/innervation , Vestibule, Labyrinth/metabolism , Animals , Blotting, Western , Evoked Potentials, Auditory, Brain Stem/physiology , Fluorescent Antibody Technique , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Reverse Transcriptase Polymerase Chain Reaction , Vestibule, Labyrinth/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...