Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 19(22): 4497-514, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20826447

ABSTRACT

Neuronal ceroid lipofuscinoses (NCLs) constitute a group of progressive neurodegenerative disorders resulting from mutations in at least eight different genes. Mutations in the most recently identified NCL gene, MFSD8/CLN7, underlie a variant of late-infantile NCL (vLINCL). The MFSD8/CLN7 gene encodes a polytopic protein with unknown function, which shares homology with ion-coupled membrane transporters. In this study, we confirmed the lysosomal localization of the native CLN7 protein. This localization of CLN7 is not impaired by the presence of pathogenic missense mutations or after genetic ablation of the N-glycans. Expression of chimeric and full-length constructs showed that lysosomal targeting of CLN7 is mainly determined by an N-terminal dileucine motif, which specifically binds to the heterotetrameric adaptor AP-1 in vitro. We also show that CLN7 mRNA is more abundant in neurons than astrocytes and microglia, and that it is expressed throughout rat brain, with increased levels in the granular layer of cerebellum and hippocampal pyramidal cells. Interestingly, this cellular and regional distribution is in good agreement with the autofluorescent lysosomal storage and cell loss patterns found in brains from CLN7-defective patients. Overall, these data highlight lysosomes as the primary site of action for CLN7, and suggest that the pathophysiology underpinning CLN7-associated vLINCL is a cell-autonomous process.


Subject(s)
Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses/genetics , Animals , Animals, Newborn , Brain/metabolism , Cells, Cultured , HEK293 Cells , HeLa Cells , Homozygote , Humans , Lysosomes/genetics , Lysosomes/metabolism , Mutation , Neuronal Ceroid-Lipofuscinoses/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transfection
3.
Hum Mol Genet ; 10(19): 2123-31, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11590129

ABSTRACT

Batten disease (juvenile neuronal ceroid lipofuscinosis, JNCL), the most common neurodegenerative disease of childhood, is caused by mutations in the CLN3 gene encoding a putative transmembrane protein. The function of CLN3 is currently unknown but it has been shown to localize in the endosomal/lysosomal compartments of non-neuronal cells. In addition, several other intracellular localizations have been proposed and the controversy of the reports suggests that CLN3 may have different intracellular localization in different cell types. Batten disease severely affects neuronal cells but leaves other organs clinically unaffected, and thus it is of utmost importance to approach the disease mechanism by studying the expression and localization of CLN3 in the brain and neuronal cells. We have analysed here CLN3 in the mouse brain using in situ hybridization, immunohistochemical staining and western blot analysis of subcellular fractions. As visual deterioration is the hallmark of Batten disease we have set up primary retinal cultures from the mouse and analysed both endogenous mouse CLN3 and Semliki Forest virus-mediated human CLN3 localization using immunofluorescence staining and confocal microscopy. We demonstrate that CLN3 is abundantly expressed in neuronal cells, especially in the cortex, hippocampus and cerebellum of the adult mouse brain. Furthermore, our results indicate that in neurons CLN3 is not solely a lysosomal protein. It is localized in the synaptosomes but, interestingly, is not targeted to the synaptic vesicles. The novel localization of CLN3 directs attention towards molecular alterations at the synapses. This should yield important clues about the mechanisms of neurodegeneration in Batten disease.


Subject(s)
Brain/metabolism , Molecular Chaperones , Neuronal Ceroid-Lipofuscinoses/metabolism , Proteins/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , Animals , Blotting, Western , Fluorescent Antibody Technique, Indirect , GAP-43 Protein/metabolism , Genetic Vectors , Humans , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Nerve Tissue Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/metabolism , Proteins/genetics , Retina/metabolism , Semliki forest virus/genetics , Transfection
4.
Mol Cell Neurosci ; 18(2): 131-40, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11520175

ABSTRACT

Infantile neuronal ceroid lipofuscinosis (INCL) is a severe neurodegenerative storage disorder in children caused by mutations in the palmitoyl protein thioesterase gene (PPT1). We have investigated here four naturally occurring previously described PPT1 mutations and show that all cause severe effects on PPT1 enzyme activity in transiently transfected COS-1 cells. Two of the mutations (delPhe84 and insCys45) cause a classical INCL phenotype and two (Thr75Pro and Leu219Gln) result in a late onset disease phenotype. All these mutated PPT1 molecules have severely altered intracellular localization in transiently transfected BHK-cells, whereas in mouse primary neuron cultures different effects were observed. In neurons the delPhe84 and insCys45 mutant polypeptides were targeted to the ER. Interestingly the Thr75Pro and Leu219Gln mutations had only minor effects on the neuronal trafficking of PPT1 and the mutated polypeptides were observed in neuronal shafts and showed colocalization with the presynaptic marker SV2. Our data indicates that neuronal cells provide an excellent model to study the genotype-phenotype correlation in INCL.


Subject(s)
Models, Biological , Mutation/physiology , Neuronal Ceroid-Lipofuscinoses/enzymology , Neuronal Ceroid-Lipofuscinoses/genetics , Neurons/metabolism , Palmitoyl-CoA Hydrolase/genetics , Palmitoyl-CoA Hydrolase/metabolism , Protein Transport/genetics , Age of Onset , Animals , COS Cells/metabolism , COS Cells/ultrastructure , Cricetinae , Disease Progression , Endoplasmic Reticulum/metabolism , Fetus , Fluorescent Antibody Technique , Golgi Apparatus/metabolism , Humans , Mice , Microscopy, Electron , Neuronal Ceroid-Lipofuscinoses/physiopathology , Phenotype , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
Hum Mol Genet ; 10(9): 983-95, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309371

ABSTRACT

A deficiency of functional aspartylglucosaminidase (AGA) causes a lysosomal storage disease, aspartylglucosaminuria (AGU). The recessively inherited disease is enriched in the Finnish population, where 98% of AGU alleles contain one founder mutation, AGU(Fin). Elsewhere in the world, we and others have described 18 different sporadic AGU mutations. Many of these are predicted to interfere with the complex intracellular maturation and processing of the AGA polypeptide. Proper initial folding of AGA in the endoplasmic reticulum (ER) is dependent on intramolecular disulfide bridge formation and dimerization of two precursor polypeptides. The subsequent activation of AGA occurs autocatalytically in the ER and the protein is transported via the Golgi to the lysosomal compartment using the mannose-6-phosphate receptor pathway. Here we use the three-dimensional structure of AGA to predict structural consequences of AGU mutations, including six novel mutations, and make an effort to characterize every known disease mutation by dissecting the effect of mutations on intracellular stability, maturation, transport and the activity of AGA. Most mutations are substitutions replacing the original amino acid with a bulkier residue. Mutations of the dimer interface prevent dimerization in the ER, whereas active site mutations not only destroy the activity but also affect maturation of the precursor. Depending on their effects on the AGA polypeptide the mutations can be categorized as mild, moderate or severe. These data contribute to the expanding body of knowledge pertaining to molecular pathogenesis of AGU.


Subject(s)
Aspartylglucosylaminase/genetics , Lysosomal Storage Diseases/genetics , Mutation/physiology , Amino Acid Sequence , Aspartylglucosaminuria , Aspartylglucosylaminase/blood , Aspartylglucosylaminase/chemistry , Binding Sites , Cell Line, Transformed , DNA/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , Ligands , Lysosomal Storage Diseases/enzymology , Lysosomes/chemistry , Lysosomes/enzymology , Microscopy, Confocal , Microscopy, Immunoelectron , Molecular Sequence Data , Mutagenesis, Site-Directed , Precipitin Tests , Protein Conformation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Transfection
6.
Hum Mol Genet ; 10(1): 69-75, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11136716

ABSTRACT

A deficiency of palmitoyl protein thioesterase (PPT) leads to the neurodegenerative disease infantile neuronal ceroid lipofuscinosis (INCL), which is characterized by an almost complete loss of cortical neurons. PPT expressed in COS-1 cells is recognized by the mannose-6-phosphate receptor (M6PR) and is routed to lysosome, but a substantial fraction of PPT is secreted. We have here determined the neuronal localization of PPT by confocal microscopy, cryoimmunoelectron microscopy and cell fractionation. In mouse primary neurons and brain tissue, PPT is localized in synaptosomes and synaptic vesicles but not in lysosomes. Furthermore, in polarized epithelial Caco-2 cells, PPT is localized exclusively to the basolateral site, in contrast to the classical lysosomal enzyme, aspartylglucosaminidase (AGA), which is localized in the apical site. The current data imply that PPT has a role outside the lysosomes in the brain and may be associated with synaptic functioning. This finding opens a new route to study the neuropathological events associated with INCL.


Subject(s)
Neuronal Ceroid-Lipofuscinoses/genetics , Neurons/enzymology , Synaptic Vesicles/enzymology , Synaptosomes/enzymology , Thiolester Hydrolases/metabolism , Animals , Blotting, Western , Brain/enzymology , CHO Cells , Caco-2 Cells , Cell Fractionation , Cell Line , Cricetinae , Humans , Lysosomes/enzymology , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Immunoelectron , Phenotype , Thiolester Hydrolases/pharmacokinetics , Transfection
7.
J Comp Neurol ; 426(3): 406-12, 2000 Oct 23.
Article in English | MEDLINE | ID: mdl-10992246

ABSTRACT

Mutations in the CLN-1 and CLN-5 genes underlie the infantile, and Finnish variant of the late-infantile, neuronal ceroid lipofuscinoses, respectively. These disorders are characterized by a massive neuronal death early in childhood. We have studied mRNA and protein expression of CLN-1 and CLN-5 in embryonic human brains. The spatial and temporal distributions of CLN-1 and CLN-5 were similar in the embryonic human brain. Both genes are expressed at the beginning of cortical neurogenesis, and this expression increases as cortical development proceeds. In the developing cortical plate, expression is found in postmitotic migrating neuroblasts and neuroblasts that have completed migration. Expression was intense also in cells of the thalamus as well as in the future Purkinje cell layer of the cerebellum. These findings indicate that expression of CLN-1 and CLN-5 may be significant for development of a wide range of maturating neurons.


Subject(s)
Brain/embryology , Gene Expression , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Embryo, Mammalian/metabolism , Embryo, Mammalian/physiology , Humans , Immunohistochemistry , In Situ Hybridization , Lysosomal Membrane Proteins , Membrane Proteins/metabolism , RNA, Messenger/metabolism , Thiolester Hydrolases
8.
Hum Mol Genet ; 9(11): 1691-7, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861296

ABSTRACT

Progressive epilepsy with mental retardation (EPMR) is a new member of the neuronal ceroid lipofuscinoses (NCLs). The CLN8 gene underlying EPMR was recently identified. It encodes a novel 286 amino acid transmembrane protein that contains an endoplasmic reticulum (ER)-retrieval signal (KKRP) in its C-terminus. A homozygous mutation in the orthologous mouse gene (Cln8) underlies the phenotype of a naturally occurring NCL model, the motor neuron degeneration mouse (mnd). To characterize the product of the CLN8 gene and to determine its intracellular localization, we expressed CLN8 cDNA in BHK, HeLa and CHO cell lines. In western blotting and pulse-chase analyses an approximately 33 kDa protein that does not undergo proteolytic processing steps was detected. Using CLN8 and cell organelle specific antibodies with confocal immunofluorescence microscopy the CLN8 protein was shown to localize in the ER. Partial localization to the ER-Golgi intermediate compartment (ERGIC) was also observed. The ER-ERGIC localization was not altered in the CLN8 protein representing the EPMR mutation. However, mnd mutant protein was only found in the ER. Mutations in the ER retrieval signal KKRP resulted in localization of CLN8 to the Golgi apparatus. Taken together, these data strongly suggest that CLN8 is an ER resident protein that recycles between ER and ERGIC.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Amino Acid Motifs , Animals , Biological Transport , CHO Cells , COS Cells , Cell Line , Cricetinae , Epilepsy/genetics , Golgi Apparatus/metabolism , HeLa Cells , Humans , Intellectual Disability/genetics , Lysosomes/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microscopy, Confocal , Molecular Weight , Motor Neuron Disease/genetics , Mutation , Neuronal Ceroid-Lipofuscinoses/genetics , Protein Processing, Post-Translational
9.
Mol Genet Metab ; 69(2): 123-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10720439

ABSTRACT

Infantile neuronal ceroid lipofuscinosis (INCL) is a severe neurodegenerative disorder in childhood that is caused by mutations in the gene encoding lysosomal palmitoyl protein thioesterase (PPT). INCL is characterized by massive and selective loss of cortical neurons. Here we have analyzed the intracellular processing and localization of adenovirus-mediated PPT in mouse primary neurons and NGF-induced PC-12 cells. The neuronal processing of PPT was found to be similar to that observed in peripheral cells, and a significant amount of the PPT enzyme was secreted in the primary neurons. Immunofluorescence analysis of the neuronal cells infected with wild-type PPT showed a granular staining pattern in the cell soma and neuronal shafts. Interestingly, PPT was also found in the synaptic ends of the neuronal cells and the staining pattern of the enzyme colocalized to a significant extent with the synaptic markers SV2 and synaptophysin. These in vitro data correspond with the distribution of endogeneous PPT in mouse brain and suggest that PPT may not solely be a lysosomal hydrolase. The specific targeting of PPT into the neuritic shafts and nerve terminals indicates that PPT may be associated with the maintenance of synaptic function. Furthermore, since a substantial amount of PPT is secreted by neurons, it is tempting to speculate that the enzyme could also have an extracellular substrate.


Subject(s)
Neurons/enzymology , Thiolester Hydrolases/metabolism , Adenoviridae/genetics , Animals , Brain/enzymology , Cells, Cultured , Genetic Vectors , Immunohistochemistry , Mice , Mice, Inbred Strains , Neurons/cytology , PC12 Cells , Rats , Recombination, Genetic
10.
Hum Mutat ; 15(3): 273-9, 2000.
Article in English | MEDLINE | ID: mdl-10679943

ABSTRACT

The infantile form of neuronal ceroid lipofuscinosis (INCL; CLN1) is the earliest onset form of the neuronal ceroid lipofuscinoses (NCL), a group of progressive encephalopathies of children. INCL is caused by mutations in the palmitoyl protein thioesterase (PPT) gene, and we report here eight novel INCL mutations in PPT. Five of the mutations, c.456C>A, c.162-163insA, c.174-175delG, c.774-775insA, and a splice acceptor mutation IVS1-2A>G in intron 1, caused the generation of a premature STOP codon either directly or after a frameshift. One mutation was a three-bp insertion in exon 2 (c. 132-133insTGT) leading to insertion of one extra cysteine (Ser44-insCys-Cys45), and another mutation, a 3-bp deletion in exon 3 (c.249-251delCTT), led to deletion of Phe84 in PPT. A splice acceptor mutation IVS6-1G>T in intron 6 can be predicted to cause skipping of exon 7 in PPT. All of these novel mutations were associated with the classical phenotype of INCL, with the first symptoms starting around 12 months of age. The severe phenotypes could be explained by the nature of the novel mutations: five are predicted to lead to premature translational termination, thus abolishing the active site of PPT, and three will probably cause a misfolding of the nascent polypeptide. Thirty-five percent (7/20) of the disease alleles in these 11 families contained the most prevalent c.451C>T missense mutation outside Finland [Das et al., 1998]. Consequently, 31 different mutations underlying INCL have been found so far, the majority leading to classical INCL.


Subject(s)
Neuronal Ceroid-Lipofuscinoses/genetics , Thiolester Hydrolases/genetics , Amino Acid Sequence , Amino Acid Substitution , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Exons/genetics , Family Health , Humans , Infant , Molecular Sequence Data , Mutagenesis, Insertional , Mutation , Neuronal Ceroid-Lipofuscinoses/enzymology , Point Mutation , Sequence Deletion , Sequence Homology, Amino Acid
11.
Brain Res Dev Brain Res ; 118(1-2): 1-11, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10611498

ABSTRACT

Deficiency in palmitoyl protein thioesterase (PPT) results in the rapid death of neocortical neurons in human. Very little is known about the developmental and cell-specific expression of this lysosomal enzyme. Here we show that PPT is expressed as a major 2.65 kb and a minor 1.85 kb transcript in the mouse brain. Transcript levels gradually increase between postnatal days 10 and 30. In situ hybridization analysis revealed that PPT transcripts are found widely but not homogeneously in the brain. The most intense signal was detected in the cerebral cortex (layers II, IV-V), hippocampal CA1-CA3 pyramidal cells, dentate gyrus granule cells and the hypothalamus. Immunostaining of PPT was localized in the cell soma, axons and dendrites, especially in the pyramidal and granular cells of the hippocampus, correlating well, both spatially and temporally, with the immunoreactivity of a presynaptic vesicle membrane protein, synaptophysin. In whole embryos, at embryonic day 8, the PPT mRNA expression was most apparent throughout the neuroepithelium, and from day 9 onwards it was seen in all tissues. The expression pattern of PPT suggests its general significance for the brain cells and reflects the response to maturation and growth of the neural networks. Strong PPT immunoreactivity in the axons and dentrites would imply that PPT may not be exclusively a lysosomal enzyme. A notable correlation with synaptophysin would suggest that PPT may have a role in the function of the synaptic machinery.


Subject(s)
Aging/metabolism , Animals, Newborn/metabolism , Brain/embryology , Brain/metabolism , Fetus/metabolism , Thiolester Hydrolases/metabolism , Animals , Animals, Newborn/growth & development , Embryonic and Fetal Development , Fetus/physiology , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred Strains , RNA, Messenger/metabolism , Reference Values , Thiolester Hydrolases/genetics , Tissue Distribution
12.
Hum Mol Genet ; 8(10): 1913-23, 1999.
Article in English | MEDLINE | ID: mdl-10469845

ABSTRACT

Finland, located at the edge of the inhabitable world, is one of the best-studied genetic isolates. The characteristic features of population isolates-founder effect, genetic drift and isolation-have, over the centuries, shaped the gene pool of the Finns. Finnish diseases have been a target of extensive genetic research and the majority of some 35 disease genes enriched in this population have been identified; the molecular and cellular consequences of disease mutations are currently being characterized. Special strategies taking advantage of linkage disequilibrium have been efficiently used in the initial mapping and restriction of Finnish disease loci and this has stimulated development of novel statistical approaches in the disease gene hunt. Identification of mutated genes has provided tools for detailed analyses of molecular pathogenesis in Finnish diseases, many of which reveal a distinct tissue specificity of clinical phenotype. Often these studies have not only clarified the molecular detail of Finnish diseases, but also provided novel information on biological processes and metabolic pathways essential for normal development and function of human cells and tissues.


Subject(s)
Chromosome Mapping , Gene Pool , Genetic Diseases, Inborn/genetics , Finland , Genetic Testing , Humans , Linkage Disequilibrium/genetics
13.
Exp Hematol ; 27(9): 1467-74, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10480438

ABSTRACT

OBJECTIVE: Bone marrow transplantation has been shown to alleviate symptoms outside the CNS in many lysosomal storage diseases depending on the type and stage of the disease, but the effect on neurological symptoms is variable or still unclear. Aspartylglucosaminuria (AGU) is a lysosomal storage disease characterized by mental retardation, recurrent infections in childhood, hepatosplenomegaly and coarse facial features. Vacuolized storage lysosomes are found in all tissues of patients and uncleaved enzyme substrate is excreted in the urine. The recently generated AGU mouse model closely mimicks the human disease and serves as a good model to study the efficiency of bone marrow transplantation in this disease. METHODS: Eight-week-old AGU mice were lethally irradiated and transplanted with bone marrow from normal donors. The AGA enzyme activity was measured in the liver and the brain and the degree of correction of tissue pathology was analyzed by light and electron microscopy. Reverse bone marrow transplantation (AGU bone marrow to wild-type mice) was also performed. RESULTS: Six months after transplantation the AGA enzyme activity was 13% of normal in the liver, but only 3% in the brain. Tissue pathology was reversed in the liver and the spleen, but not in the brain and the kidney. The urinary excretion of enzyme substrate was diminished but still detectable. No storage vacuoles were found in the tissues after reverse transplantation, but subtle excretion of uncleaved substrate was detected in the urine. CONCLUSION: Liver and spleen pathology of AGU was corrected by bone marrow transplantation, but there was no effect on lysosomal accumulation in the CNS and in the kidneys.


Subject(s)
Acetylglucosamine/analogs & derivatives , Amino Acid Metabolism, Inborn Errors/therapy , Aspartylglucosaminuria , Bone Marrow Transplantation , Lysosomal Storage Diseases/therapy , Lysosomes/pathology , Acetylglucosamine/urine , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Aspartylglucosylaminase/analysis , Aspartylglucosylaminase/genetics , Brain/enzymology , Brain/pathology , Humans , Intellectual Disability/etiology , Intellectual Disability/prevention & control , Kidney/enzymology , Kidney/pathology , Liver/enzymology , Liver/pathology , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Lysosomes/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/analysis , Organ Specificity , Polymerase Chain Reaction , Radiation Chimera , Specific Pathogen-Free Organisms , Spleen/enzymology , Spleen/pathology , Vacuoles/pathology
14.
Mol Genet Metab ; 67(4): 294-307, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10444340

ABSTRACT

The deficiency of a lysosomal enzyme, aspartylglucosaminidase, results in a lysosomal storage disorder, aspartylglucosaminuria, manifesting as progressive mental retardation. To understand tissue pathogenesis and disease progression we analyzed the developmental expression of the enzyme, especially in brain, which is the major source of the pathological symptoms. Highest mRNA levels in brain were detected during embryogenesis, the levels decreased neonatally and started to increase again from Day 7 on. In Western analyses, a defective processing of aspartylglucosaminidase was observed in brain as compared to other tissues, resulting in very low levels of the mature, active form of the enzyme. Interestingly immunohistochemical analyses of mouse brain revealed that aspartylglucosaminidase immunoreactivity closely mimicked the myelin basic protein immunostaining pattern. The only evident neuronal staining was observed in the developing Purkinje cells of the cerebellum from Days 3 to 10, reflecting well the mRNA expression. In human infant brain, the immunostaining was also present in myelinated fibers as well as in the Purkinje cells and, additionally, in the soma and extensions of other neurons. In the adult human brain neurons and oligodendrocytes displayed immunoreactivity whereas myelinated fibers were not stained. Our results of aspartylglucosaminidase immunostaining in myelinated fibers of infant brain might imply the involvement of aspartylglucosaminidase in the early myelination process. This is consistent with previous magnetic resonance imaging findings in the brains of aspartylglucosaminuria patients, revealing delayed myelination in childhood.


Subject(s)
Brain/enzymology , Lysosomal Storage Diseases/enzymology , Adult , Animals , Aspartylglucosylaminase/genetics , Aspartylglucosylaminase/metabolism , Blotting, Western , Brain/embryology , Brain/pathology , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Humans , Immunohistochemistry , In Situ Hybridization , Infant, Newborn , Liver/enzymology , Lysosomal Storage Diseases/etiology , Lysosomal Storage Diseases/genetics , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic
15.
FEBS Lett ; 454(3): 233-9, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10431814

ABSTRACT

Gelsolin, an actin-modulating protein, derived from a single gene exists in intracellular and secreted forms. A point mutation at position 187 of both forms of gelsolin causes familial amyloidosis of the Finnish type (FAF). Here, we expressed both isoforms of the wild-type and FAF mutant gelsolin in mouse embryonic gelsolin-null fibroblasts. We demonstrate that the FAF mutation does not interfere with the normal actin-modulating function of intracellular gelsolin, and that aberrant processing of secreted FAF gelsolin to FAF amyloid precursor takes place in the gelsolin-negative background. These results suggest that, in patients with FAF, symptoms are caused by the accumulation in their tissues of amyloid derived from plasma gelsolin and are not due to functional differences in cytoplasmic gelsolin.


Subject(s)
Actins/metabolism , Amyloidosis/metabolism , Fibroblasts/metabolism , Gelsolin/genetics , Gelsolin/metabolism , Amyloidosis/genetics , Animals , Cells, Cultured , Mice , Mice, Knockout , Mutation
16.
Hum Mol Genet ; 8(6): 1091-8, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10332042

ABSTRACT

Batten disease [juvenile-onset neuronal ceroid lipofuscinosis (JNCL)], the most common progressive encephalopathy of childhood, is caused by mutations in a novel lysosomal membrane protein (CLN3) with unknown function. In this study, we have confirmed the lysosomal localization of the CLN3 protein by immunoelectron microscopy by co-localizing it with soluble and membrane-associated lysosomal proteins. We have analysed the intracellular processing and localization of two mutants, 461-677del, which is present in 85% of CLN3 alleles and causes the classical JNCL, and E295K [corrected], which is a rare missense mutation associated with an atypical form of JNCL. Pulse-chase labelling and immunoprecipitation of the two mutant proteins in COS-1-cells indicated that 461-677del is synthesized as an approximately 24 kDa truncated polypeptide, whereas the maturation of E295K [corrected] resembles that of the wild-type CLN3 polypeptide. Transient expression of the two mutants in BHK cells showed that 461-677del is retained in the endoplasmic reticulum, whereas E295K [corrected] was capable of reaching the lysosomal compartment. The CLN3 polypeptides were expressed further in mouse primary neurons where the wild-type CLN3 protein was localized both in the cell soma and in neuronal extensions, whereas the 461-677del mutant was arrested in the cell soma. Interestingly, co-localization of the wild-type CLN3 and E295K [corrected] proteins with a synaptic vesicle marker indicates that the CLN3 protein might participate in synaptic vesicle transport/transmission. The data presented here provide clear evidence for a cellular distinction between classical and atypical forms of Batten disease both in neural and non-neural cells.


Subject(s)
Membrane Glycoproteins , Molecular Chaperones , Neuronal Ceroid-Lipofuscinoses/genetics , Proteins/genetics , Amino Acid Substitution , Animals , Biological Transport/genetics , COS Cells , Cell Line , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Female , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Mutation , Neurons/metabolism , Pregnancy , Proteins/metabolism , Recombinant Fusion Proteins/genetics , Sequence Deletion , Telencephalon/cytology , Transfection
17.
J Neuropathol Exp Neurol ; 57(12): 1154-63, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9862638

ABSTRACT

Aspartylglucosaminuria (AGU) is a recessively inherited lysosomal storage disorder caused by the deficiency of the aspartylglucosaminidase (AGA) enzyme. The hallmark of AGU is slowly progressing mental retardation but the progression of brain pathology has remained uncharacterized in humans. Here we describe the long-term follow-up of mice carrying a targeted AGU-mutation in both alleles. Immunohistochemistry, histology, electron microscopy, quantitative magnetic resonance imaging (MRI) and behavioral studies were carried out to evaluate the CNS affection of the disease during development. The lysosomal storage vacuoles of the AGA -/- mice were most evident in central brain regions where MRI also revealed signs of brain atrophy similar to that seen in the older human patients. By immunohistochemistry and MRI examinations, a subtle delay of myelination was observed in AGA -/- mice. The life span of the AGA -/- mice was not shortened. Similar to the slow clinical course observed in human patients, the AGA -/- mice have behavioral symptoms that emerge at older age. Thus, the AGU knock-out mice represent an accurate model for AGU, both histopathologically and phenotypically.


Subject(s)
Aspartylglucosaminuria , Central Nervous System/pathology , Monitoring, Physiologic/methods , Animals , Aspartylglucosylaminase/urine , Behavior, Animal/physiology , Humans , Immunoblotting , Immunohistochemistry , Intellectual Disability/enzymology , Intellectual Disability/pathology , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Myelin Sheath/physiology , Nerve Tissue Proteins/metabolism , RNA, Messenger/analysis
18.
J Neurosci ; 18(19): 7750-6, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9742145

ABSTRACT

Aspartylglucosaminuria (AGU) is a neurodegenerative lysosomal storage disease that is caused by mutations in the gene encoding for a soluble hydrolase, aspartylglucosaminidase (AGA). In this study, we have used our recently developed mouse model for AGU and analyzed processing, intracellular localization, and endocytosis of recombinant AGA in telencephalic AGU mouse neurons in vitro. The processing steps of AGA were found to be similar to the peripheral cells, but both the accumulation of the inactive precursor molecule and delayed lysosomal processing of the enzyme were detected. AGA was distributed to the cell soma and neuronal processes but was not found in the nerve terminals. Endocytotic capability of cultured telencephalic neurons was comparable to that of fibroblasts, and endocytosis of AGA was blocked by free mannose-6-phosphate (M6P), indicating that uptake of the enzyme was mediated by M6P receptors (M6PRs). Uptake of extracellular AGA was also studied in the tumor-derived cell lines rat pheochromocytoma (PC12) and mouse neuroblastoma cells (N18), which both endocytosed AGA poorly as compared with cultured primary neurons. Expression of cation-independent M6PRs (CI-M6PRs) in different cell lines correlated well with the endocytotic capability of these cells. Although a punctate expression pattern of CI-M6PRs was found in fibroblasts and cultured primary neurons, the expression was beyond the detection limit in PC12 and N18 cells. This indicates that PC12 and N18 are not feasible cell lines to describe neuronal uptake of mannose-6-phosphate-tagged proteins. This in vitro data will form an important basis for the brain-targeted therapy of AGU.


Subject(s)
Aspartylglucosylaminase/genetics , Aspartylglucosylaminase/metabolism , Endocytosis/physiology , Lysosomes/enzymology , Neurons/enzymology , Animals , Fibroblasts/chemistry , Fibroblasts/metabolism , Gene Expression Regulation, Viral , Lysosomes/chemistry , Mice , Mice, Mutant Strains , Mutagenesis/physiology , Neuroblastoma , Neurons/chemistry , Neurons/ultrastructure , PC12 Cells , Rats , Receptor, IGF Type 2/analysis , Receptor, IGF Type 2/biosynthesis , Receptor, IGF Type 2/metabolism , Recombinant Proteins/pharmacology , Semliki forest virus , Telencephalon/cytology
19.
J Biol Chem ; 273(39): 25320-8, 1998 Sep 25.
Article in English | MEDLINE | ID: mdl-9737998

ABSTRACT

Secretory, membrane, and lysosomal proteins undergo covalent modifications and acquire their secondary and tertiary structure in the lumen of the endoplasmic reticulum (ER). In order to pass the ER quality control system and become transported to their final destinations, many of them are also assembled into oligomers. We have recently determined the three-dimensional structure of lysosomal aspartylglucosaminidase (AGA), which belongs to a newly discovered family of homologous amidohydrolases, the N-terminal nucleophile hydrolases. Members of this protein family are activated from an inactive precursor molecule by an autocatalytic proteolytic processing event whose exact mechanism has not been thoroughly determined. Here we have characterized in more detail the initial events in the ER required for the formation of active AGA enzyme using transient expression of polypeptides carrying targeted amino acid substitutions. We show that His124 at an interface between two heterodimers of AGA is crucial for the thermodynamically stable oligomeric structure of AGA. Furthermore, the side chain of Thr206 is essential both for the proteolytic activation and enzymatic activity of AGA. Finally, the proper geometry of the residues His204-Asp205 seems to be crucial for the activation of AGA precursor polypeptides. We propose here a reaction mechanism for the activation of AGA which could be valid for homologous enzymes as well.


Subject(s)
Aspartylglucosylaminase/metabolism , Amino Acid Sequence , Animals , Aspartylglucosylaminase/chemistry , Aspartylglucosylaminase/genetics , Biopolymers , Calcium-Binding Proteins/metabolism , Calnexin , Calreticulin , Catalysis , Conserved Sequence , DNA, Complementary , Endoplasmic Reticulum/enzymology , Enzyme Activation , Humans , Molecular Chaperones/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational , Ribonucleoproteins/metabolism , Sequence Homology, Amino Acid
20.
Genome Res ; 8(7): 724-30, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9685319

ABSTRACT

Palmitoyl protein thioesterase (PPT) is the defective enzyme in infantile neuronal ceroid lipofuscinosis (INCL), which is a recessively inherited, progressive neurodegenerative disorder. We present here the cloning, chromosomal mapping, genomic structure, and the expression of the cDNA of mouse PPT. The mouse PPT gene spans >21 kb of genomic DNA and contains nine exons with a coding sequence of 918 bp. Fluorescence in situ hybridization to metaphase chromosomes localized the mouse PPT gene to the chromosome 4 conserved syntenic region with human chromosome 1p32 where the human PPT is located. PPT is expressed widely in a variety of mouse tissues. The mouse PPT cDNA is conserved highly with the human and rat PPT both at the nucleotide and amino acid sequence level. Transient expression of mouse PPT in COS-1 cells yielded a 38/36-kD differentially glycosylated polypeptide that was also secreted into culture media. Immunofluorescence analysis of transiently transfected HeLa cells indicated lysosomal localization of mouse PPT. Based on the high conservation of the gene and polypeptide structure as well as similar processing and intracellular localization, the function of PPT in mouse and human are likely to be very similar.


Subject(s)
DNA, Complementary/biosynthesis , Gene Expression Regulation , Genes , Thiolester Hydrolases/genetics , Amino Acid Sequence , Amino Acids/analysis , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/analysis , HeLa Cells , Humans , Intracellular Fluid/enzymology , Male , Mice , Molecular Sequence Data , Thiolester Hydrolases/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...