Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Diabetes ; 70(8): 1874-1884, 2021 08.
Article in English | MEDLINE | ID: mdl-34074726

ABSTRACT

We previously reported genotype-phenotype correlations in 12 missense variants causing severe insulin resistance, located in the second and third fibronectin type III (FnIII) domains of the insulin receptor (INSR), containing the α-ß cleavage and part of insulin-binding sites. This study aimed to identify genotype-phenotype correlations in FnIII domain variants of IGF1R, a structurally related homolog of INSR, which may be associated with growth retardation, using the recently reported crystal structures of IGF1R. A structural bioinformatics analysis of five previously reported disease-associated heterozygous missense variants and a likely benign variant in the FnIII domains of IGF1R predicted that the disease-associated variants would severely impair the hydrophobic core formation and stability of the FnIII domains or affect the α-ß cleavage site, while the likely benign variant would not affect the folding of the domains. A functional analysis of these variants in CHO cells showed impaired receptor processing and autophosphorylation in cells expressing the disease-associated variants but not in those expressing the wild-type form or the likely benign variant. These results demonstrated genotype-phenotype correlations in the FnIII domain variants of IGF1R, which are presumably consistent with those of INSR and would help in the early diagnosis of patients with disease-associated IGF1R variants.


Subject(s)
Antigens, CD/genetics , Body Height/genetics , Growth Disorders/genetics , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Animals , Antigens, CD/metabolism , CHO Cells , Cricetinae , Cricetulus , Genetic Association Studies , Humans , Insulin Resistance/genetics , Mutation, Missense , Phenotype , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Retrospective Studies
2.
Ophthalmic Genet ; 41(6): 599-605, 2020 12.
Article in English | MEDLINE | ID: mdl-32940107

ABSTRACT

SIGNIFICANCE: The responsible genetic variants for occult macular dystrophy (OMD) were found at the predicted intrinsically disordered region (IDR) of the RP1L1 gene. PURPOSE: We examined the phenotypes and genotypes of family members from OMD. In addition, the genetic characteristics of the RP1L1 gene in OMD were investigated. METHODS: Whole-exome sequencing was applied on two affected family members, and Sanger sequencing was performed on three members. The structural property of RP1L1 and pathogenic variants was analyzed using predictor of natural disordered regions (PONDR). RESULTS: Two affected members showed moderate visual impairment and relative central scotoma. The spectral domain optical coherence tomography (SD-OCT) images showed an absence of the interdigitation zone (IZ) and ellipsoid zone (EZ) in one case, and an obscure EZ line in the other case. A RP1L1 variant (c.3593 C > T, p.Ser1198Phe) was identified in two affected members but not in the unaffected member. The PONDR analysis showed that the region from p.1189 to p.1248 could be predicted to be an IDR in the RP1L1 molecule. And the p. Ser1198Phe variant showed significant reduction of PONDR score. CONCLUSIONS: Although, the major pathogenic variant of OMD is p.Arg45Trp, multiple reports indicate that the region between p.1194 and p.1201 is another hot spot of OMD. The PONDR analysis predicted that the RP1L1 molecule is one of the intrinsically disordered proteins. It is speculated that the region around p.1200 is essential for the normal function of the RP1L1 molecule, and the missense variants of that area cause the development of OMD.


Subject(s)
Eye Proteins/genetics , Intrinsically Disordered Proteins/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Mutation , Phenotype , Adult , Female , Humans , Male , Middle Aged , Pedigree
3.
Diabetes Res Clin Pract ; 169: 108461, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32971154

ABSTRACT

AIMS: Monogenic diabetes is clinically heterogeneous and differs from common forms of diabetes (type 1 and 2). We aimed to investigate the clinical usefulness of a comprehensive genetic testing system, comprised of targeted next-generation sequencing (NGS) with phenotype-driven bioinformatics analysis in patients with monogenic diabetes, which uses patient genotypic and phenotypic data to prioritize potentially causal variants. METHODS: We performed targeted NGS of 383 genes associated with monogenic diabetes or common forms of diabetes in 13 Japanese patients with suspected (n = 10) or previously diagnosed (n = 3) monogenic diabetes or severe insulin resistance. We performed in silico structural analysis and phenotype-driven bioinformatics analysis of candidate variants from NGS data. RESULTS: Among the patients suspected having monogenic diabetes or insulin resistance, we diagnosed 3 patients as subtypes of monogenic diabetes due to disease-associated variants of INSR, LMNA, and HNF1B. Additionally, in 3 other patients, we detected rare variants with potential phenotypic effects. Notably, we identified a novel missense variant in TBC1D4 and an MC4R variant, which together may cause a mixed phenotype of severe insulin resistance. CONCLUSIONS: This comprehensive approach could assist in the early diagnosis of patients with monogenic diabetes and facilitate the provision of tailored therapy.


Subject(s)
Diabetes Mellitus/diagnosis , Diabetes Mellitus/genetics , Genetic Testing/methods , Insulin Resistance/genetics , Adolescent , Adult , Aged , Computational Biology , Female , GTPase-Activating Proteins/genetics , Genotype , High-Throughput Nucleotide Sequencing , Humans , Infant , Japan , Male , Mass Screening/methods , Middle Aged , Mutation, Missense , Phenotype , Young Adult
4.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(10): 158787, 2020 10.
Article in English | MEDLINE | ID: mdl-32777483

ABSTRACT

Side-chain oxysterols produced from cholesterol either enzymatically or non-enzymatically show various bioactivities. Lecithin-cholesterol acyltransferase (LCAT) esterifies the C3-hydroxyl group of these sterols as well as cholesterol. Lysosomal phospholipase A2 (LPLA2) is related to LCAT but does not catalyze esterification of cholesterol. First, esterification of side-chain oxysterols by LPLA2 was investigated using recombinant mouse LPLA2 and dioleoyl-PC/sulfatide/oxysterol liposomes under acidic conditions. TLC and LC-MS/MS showed that the C3 and C27-hydroxyl groups of 27-hydroxycholesterol could be individually esterified by LPLA2 to form a monoester with the C27-hydroxyl preference. Cholesterol did not inhibit this reaction. Also, LPLA2 esterified other side-chain oxysterols. Their esterifications by mouse serum containing LCAT supported the idea that their esterifications by LPLA2 occur at the C3-hydroxyl group. N-acetylsphingosine (NAS) acting as an acyl acceptor in LPLA2 transacylation inhibited the side-chain oxysterol esterification by LPLA2. This suggests a competition between hydroxycholesterol and NAS on the acyl-LPLA2 intermediate formed during the reaction. Raising cationic amphiphilic drug concentration or ionic strength in the reaction mixture evoked a reduction of the side-chain oxysterol esterification by LPLA2. This indicates that the esterification could progress via an interfacial interaction of LPLA2 with the lipid membrane surface through an electrostatic interaction. The docking model of acyl-LPLA2 intermediate and side-chain oxysterol provided new insight to elucidate the transacylation mechanism of sterols by LPLA2. Finally, exogenous 25-hydroxycholesterol esterification within alveolar macrophages prepared from wild-type mice was significantly higher than that from LPLA2 deficient mice. This suggests that there is an esterification pathway of side-chain oxysterols via LPLA2.


Subject(s)
Cholesterol/metabolism , Oxysterols/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phospholipases A2/genetics , Animals , Catalysis , Esterification/genetics , Humans , Hydroxycholesterols/metabolism , Lysosomes/enzymology , Macrophages/metabolism , Mice , Phospholipases A2/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism
5.
J Biosci Bioeng ; 125(2): 168-174, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28903882

ABSTRACT

Endo-ß-N-acetylglucosaminidase from the methylotrophic yeast Ogataea minuta (Endo-Om) is a glycoside hydrolase family 85 enzyme that has dual catalytic activity in the hydrolysis and transglycosylation of complex N-glycans, in common with the enzymes from the eukaryotic species. In this study, we have conducted mutagenesis of Endo-Om at Trp295, to determine the effect on hydrolytic activity. Structural modeling predicted that Trp295 forms an important interaction with the α-1,3-linked mannose residue of the trimannosyl N-glycan core, rather than being directly involved in catalytic activity. Our results showed that an aromatic amino acid is required at position 295 for the hydrolytic activity of this enzyme. Notably, the tryptophan residue is highly conserved in eukaryotic endo-ß-N-acetylglucosaminidases that show activity toward complex oligosaccharides. Accordingly, our results strongly suggested that Trp295 is involved in the recognition of oligosaccharide substrates by Endo-Om.


Subject(s)
Hydrolysis , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/chemistry , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Saccharomycetales/enzymology , Tryptophan/metabolism , Conserved Sequence , Mannose/metabolism , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/genetics , Oligosaccharides/metabolism , Saccharomycetales/genetics , Tryptophan/genetics
6.
Glycobiology ; 23(4): 495-504, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23363739

ABSTRACT

Human sialidase 2 (NEU2) is a cytoplasmic sialidase that degrades sialylglycoconjugates, including glycoproteins and gangliosides, via hydrolysis of terminal sialic acids to produce asialo-type molecules. Here, we first report the inhibitory effects of a series of synthetic sialyldendrimers comprising three types [Dumbbell(1)6-S-Neu5Ac(6), Fan(0)3-S-Neu5Ac(3) and Ball(0)4-S-NeuAc(4)] toward recombinant human NEU2 in vitro. Among them, Dumbbell(1)6-S-Neu5Ac(6) exhibited the most potent inhibitory activity (concentration causing 50% inhibition (IC(50)), 0.4 ∼ 0.5 mM). In addition, NeuSLac and NeuSCel carrying thiosialyltrisaccharide moieties exhibited more potent inhibitory effects than NeuSGal and NeuSGlc carrying thiosialyldisaccharides. Docking models composed of NEU2 and the thiosialyloligosaccharide suggested that the active pocket of NEU2 prefers the second galactose-ß (Galß) to the glucose-ß (Glcß) residue in the trisaccharide structure, there being a hydrogen bond between the 4-hydroxy group of the second Galß and the side chain of the D46 residue of NEU2. The third Glcß residues of NeuSLac and NeuSCel were also predicted to be stabilized by hydrogen bonds with the side chains of the R21, R304, D358 and Y359 residues of NEU2. NEU2 mutants (D358A and Y359A) exhibited reduced affinity for NeuSLac carrying thiosialyltrisaccharide moieties, suggesting the significant roles of D358 and Y359 residues in recognition of thiosialyltrisaccharide moieties of NeuSLac bound in the active pocket of NEU2. Thus, the present sialyldendrimers could be utilized not only as a new class of NEU2 inhibitors but also as molecular probes for evaluating the biological functions of NEU2, including the catalytic activity and mechanism as to natural substrates carrying sialyloligosaccharides.


Subject(s)
Dendrimers/chemistry , Enzyme Inhibitors/chemistry , N-Acetylneuraminic Acid/chemistry , Neuraminidase/antagonists & inhibitors , Amino Acid Sequence , Binding Sites , Carbohydrate Sequence , Enzyme Inhibitors/pharmacology , Galactose/chemistry , Glucose/chemistry , Hydrogen Bonding , Molecular Docking Simulation , Molecular Sequence Data , Mutation, Missense , Neuraminidase/chemistry , Neuraminidase/genetics , Recombinant Proteins/antagonists & inhibitors , Substrate Specificity , Trisaccharides/chemistry
7.
Mol Ther ; 18(8): 1519-26, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20571546

ABSTRACT

Human lysosomal beta-hexosaminidase A is a heterodimer composed of alpha- and beta-subunits encoded by HEXA and HEXB, respectively. We genetically introduced an additional N-glycosylation sequon into HEXA, which caused amino acid substitutions (S51 to N and A53 to T) at homologous positions to N84 and T86 in the beta-subunit. The mutant HexA (NgHexA) obtained from a Chinese hamster ovary (CHO) cell line co-expressing the mutated HEXA and wild-type HEXB complementary DNAs was demonstrated to contain an additional mannose-6-phosphate (M6P)-type-N-glycan. NgHexA was more efficiently taken up than the wild-type HexA and delivered to lysosomes, where it degraded accumulated substrates including GM2 ganglioside (GM2) when administered to cultured fibroblasts derived from a Sandhoff disease (SD) patient. On intracerebroventricular (i.c.v.) administration of NgHexA to SD model mice, NgHexA more efficiently restored the HexA activity and reduced the GM2 and GA2 (asialoGM2) accumulated in neural cells of the brain parenchyma than the wild-type HexA. These findings indicate that i.c.v. administration of the modified human HexA with an additional M6P-type N-glycan is applicable for enzyme replacement therapy (ERT) involving an M6P-receptor as a molecular target for HexA deficiencies including Tay-Sachs disease and SD.


Subject(s)
Polysaccharides/metabolism , Sandhoff Disease/metabolism , beta-Hexosaminidase alpha Chain/metabolism , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/therapeutic use , Animals , CHO Cells , Cells, Cultured , Chromatography, Thin Layer , Cricetinae , Cricetulus , G(M2) Ganglioside/metabolism , Glycosylation , Humans , Immunoblotting , Mice , Polysaccharides/chemistry , Sandhoff Disease/drug therapy , Sandhoff Disease/genetics , beta-Hexosaminidase alpha Chain/chemistry , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase alpha Chain/therapeutic use , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/metabolism , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism
8.
Clin Chim Acta ; 391(1-2): 68-73, 2008 May.
Article in English | MEDLINE | ID: mdl-18328816

ABSTRACT

BACKGROUND: Recently, enzyme enhancement therapy (EET) for Pompe disease involving imino sugars, which act as potential inhibitors of acid alpha-glucosidases in vitro, to improve the stability and/or transportation of mutant acid alpha-glucosidases in cells was studied and attracted interest. However, the mechanism underlying the molecular interaction between the imino sugars and the enzyme has not been clarified yet. METHODS: We examined the inhibitory and binding effects of four imino sugars on a recombinant human acid alpha-glucosidase, alglucosidase alfa, by means of inhibition assaying and isothermal titration calorimetry (ITC). Furthermore, we built structural models of complexes of the catalytic domain of the enzyme with the imino sugars bound to its active site by homology modeling, and examined the molecular interaction between them. RESULTS: All of the imino sugars examined exhibited a competitive inhibitory action against the enzyme, 1-deoxynojirimycin (DNJ) exhibiting the strongest action among them. ITC revealed that one compound molecule binds to one enzyme molecule and that DNJ most strongly binds to the enzyme among them. Structural analysis revealed that the active site of the enzyme is almost completely occupied by DNJ. CONCLUSION: These biochemical and structural analyses increased our understanding of the molecular interaction between a human acid alpha-glucosidase and imino sugars.


Subject(s)
Glycogen Storage Disease Type II/enzymology , Imino Sugars/metabolism , alpha-Glucosidases/metabolism , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/metabolism , 1-Deoxynojirimycin/pharmacology , Binding Sites , Catalytic Domain , Drug Interactions , Glycoside Hydrolase Inhibitors , Humans , Imino Sugars/chemistry , Imino Sugars/pharmacology , Models, Molecular , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Thermodynamics
9.
Structure ; 15(12): 1591-602, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18073109

ABSTRACT

Phosphorylation of endogenous inhibitor proteins for type-1 Ser/Thr phosphatase (PP1) provides a mechanism for reciprocal coordination of kinase and phosphatase activities. A myosin phosphatase inhibitor protein CPI-17 is phosphorylated at Thr38 through G-protein-mediated signals, resulting in a >1000-fold increase in inhibitory potency. We show here the solution NMR structure of phospho-T38-CPI-17 with rmsd of 0.36 +/- 0.06 A for the backbone secondary structure, which reveals how phosphorylation triggers a conformational change and exposes an inhibitory surface. This active conformation is stabilized by the formation of a hydrophobic core of intercalated side chains, which is not formed in a phospho-mimetic D38 form of CPI-17. Thus, the profound increase in potency of CPI-17 arises from phosphorylation, conformational change, and hydrophobic stabilization of a rigid structure that poses the phosphorylated residue on the protein surface and restricts its hydrolysis by myosin phosphatase. Our results provide structural insights into transduction of kinase signals by PP1 inhibitor proteins.


Subject(s)
Enzyme Inhibitors/metabolism , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Amino Acid Sequence , Animals , Circular Dichroism , Enzyme Inhibitors/chemistry , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Conformation , Sequence Homology, Amino Acid , Swine
10.
J Hum Genet ; 52(11): 898-906, 2007.
Article in English | MEDLINE | ID: mdl-17805474

ABSTRACT

We constructed structural models of the catalytic domain and the surrounding region of human wild-type acid alpha-glucosidase and the enzyme with amino acid substitutions by means of homology modeling, and examined whether the amino acid replacements caused structural and biochemical changes in the enzyme proteins. Missense mutations including p.R600C, p.S619R and p.R437C are predicted to cause apparent structural changes. Nonsense mutation of p.C103X terminates the translation of acid alpha-glucosidase halfway through its biosynthesis and is deduced not to allow formation of the active site pocket. The mutant proteins resulting from these missense and nonsense mutations found in patients with Pompe disease are predictably unstable and degraded quickly in cells. The structural change caused by p.G576S is predicted to be small, and cells from a subject homozygous for this amino acid substitution exhibited 15 and 11% of the normal enzyme activity levels for an artificial substrate and glycogen, respectively, and corresponding amounts of the enzyme protein on Western blotting. No accumulation of glycogen was found in organs including skeletal muscle in the subject, and thus the residual enzyme activity could protect cells from glycogen storage. On the other hand, p.E689K, which is known as a neutral polymorphism, little affected the three-dimensional structure of acid alpha-glucosidase. Structural study on a mutant acid alpha-glucosidase in silico combined with biochemical investigation is useful for understanding the molecular pathology of Pompe disease.


Subject(s)
Glycogen Storage Disease Type II/genetics , alpha-Glucosidases/genetics , Amino Acid Sequence , Blotting, Western , Cells, Cultured , Glycogen Storage Disease Type II/enzymology , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid , alpha-Glucosidases/chemistry , alpha-Glucosidases/metabolism
11.
CNS Neurol Disord Drug Targets ; 5(4): 401-13, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16918392

ABSTRACT

Lysosomal diseases comprise a group of inherited disorders resulting from defects of lysosomal enzymes and their cofactors, and in many of them the nervous system is affected. Recently, enzyme replacement therapy with recombinant lysosomal enzymes has been clinically available for several lysosomal diseases. Such enzyme replacement therapies can improve non-neurological disorders but is not effective for neurological ones. In this review, we discuss the molecular pathologies of lysosomal diseases from the protein structural aspect, current enzyme replacement therapies, and attempts to develop enzyme replacement therapies effective for lysosomal diseases associated with neurological disorders, i.e., production of enzymes, brain-specific delivery and incorporation of lysosomal enzymes into cells.


Subject(s)
Enzyme Therapy , Lysosomal Storage Diseases, Nervous System/enzymology , Lysosomes/enzymology , Recombinant Fusion Proteins/therapeutic use , Animals , Culture Media, Conditioned/pharmacology , Enzymes/chemistry , Enzymes/genetics , G(M2) Ganglioside/metabolism , Humans , Lysosomal Storage Diseases, Nervous System/genetics , Lysosomal Storage Diseases, Nervous System/therapy , Lysosomes/genetics , Lysosomes/pathology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sandhoff Disease/enzymology , Sandhoff Disease/genetics , Sandhoff Disease/physiopathology , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/genetics , Tay-Sachs Disease/physiopathology , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/genetics
12.
Biochim Biophys Acta ; 1764(8): 1372-80, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16870514

ABSTRACT

Cathepsin A (CathA) is a lysosomal serine carboxypeptidase that exhibits homology and structural similarity to the yeast and wheat serine carboxypeptidases (CPY and CPW) belonging to the alpha/beta-hydrolase fold family. Human CathA (hCathA) and CPW have been demonstrated to be inhibited by a proteasome (threonine protease) inhibitor, lactacystin, and its active derivative, omuralide (clasto-lactacystin beta-lactone), as well as chymostatin. A hCathA/omuralide complex model constructed on the basis of the X-ray crystal structures of the CPW/chymostatin complex and the yeast proteasome beta-subunit (beta5/PRE2)/omuralide one predicted that the conformation of omuralide in the active-site cleft of proteasome beta5/PRE2 should be very similar to that of chymostatin at the S1 catalytic subsites in the hCathA- and CPW-complexes. The relative positions of the glycine residues, i.e., Gly57 in hCathA, Gly53 in CPW, and Gly47 in beta5/PRE2, present in the oxyanion hole of each enzyme were also highly conserved. These results suggest that omuralide might inhibit hCathA and CPW at the S1 subsite in their active-site clefts through direct binding to the active serine residue.


Subject(s)
Cathepsin A/antagonists & inhibitors , Cathepsin A/chemistry , Cysteine Endopeptidases/chemistry , Lactones/chemistry , Lactones/pharmacology , Proteasome Endopeptidase Complex/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Catalytic Domain , Humans , In Vitro Techniques , Macromolecular Substances , Models, Molecular , Protein Conformation , Saccharomyces cerevisiae/enzymology
13.
Hum Genet ; 117(4): 317-28, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15924232

ABSTRACT

Fabry disease comprises classic and variant phenotypes. The former needs early enzyme replacement therapy, and galactose infusion is effective for some variant cases. Attempts of early diagnosis before manifestations appear will begin in the near future. However, it is difficult to predict the phenotype, to determine the therapeutic approach, only from genetic information. Thus we attempted structural analysis from a novel viewpoint. We built structural models of mutant alpha-galactosidases resulting from 161 missense mutations (147 classic and 14 variant), and evaluated the influence of each replacement on the structure by calculating the numbers of atoms affected. Among them, 11 mutants, biochemically characterized, were further investigated by color imaging of the influenced atoms. In the variant group, the number of atoms influenced by amino-acid replacement was small, especially in the main chain. In 85% of the cases, less than three atoms in the main chain are influenced. In this group, small structural changes, located apart from the active site, result in destabilization of the mutant enzymes, but galactose can stabilize them. Structural changes caused by classic Fabry mutations are generally large or are located in functionally important regions. In 82% of the cases, three atoms or more in the main chain are affected. The classic group comprises dysfunctional and unstable types, and galactose is not expected to stabilize the mutant enzymes. This study demonstrated the correlation of structural changes, and clinical and biochemical phenotypes. Structural investigation is useful for elucidating the bases of Fabry disease and clinical treatment.


Subject(s)
Fabry Disease/enzymology , Models, Molecular , Phenotype , alpha-Galactosidase/genetics , Amino Acid Substitution/genetics , Fabry Disease/genetics , Humans , Mutation, Missense/genetics , Protein Conformation
14.
J Hum Genet ; 49(1): 1-8, 2004.
Article in English | MEDLINE | ID: mdl-14685826

ABSTRACT

Alpha-N-acetylgalactosaminidase (alpha-NAGA) deficiency (Schindler/Kanzaki disease) is a clinically and pathologically heterogeneous genetic disease with a wide spectrum including an early onset neuroaxonal dystrophy (Schindler disease) and late onset angiokeratoma corporis diffusum (Kanzaki disease). In alpha-NAGA deficiency, there are discrepancies between the genotype and phenotype, and also between urinary excretion products (sialyl glycoconjugates) and a theoretical accumulated material (Tn-antigen; Gal NAcalpha1-O-Ser/Thr) resulting from a defect in alpha-NAGA. As for the former issue, previously reported genetic, biochemical and pathological data raise the question whether or not E325K mutation found in Schindler disease patients really leads to the severe phenotype of alpha-NAGA deficiency. The latter issue leads to the question of whether alpha-NAGA deficiency is associated with the basic pathogenesis of this disease. To clarify the pathogenesis of this disease, we performed structural and immunocytochemical studies. The structure of human alpha-NAGA deduced on homology modeling is composed of two domains, domain I, including the active site, and domain II. R329W/Q, identified in patients with Kanzaki disease have been deduced to cause drastic changes at the interface between domains I and II. The structural change caused by E325K found in patients with Schindler disease is localized on the N-terminal side of the tenth beta-strand in domain II and is smaller than those caused by R329W/Q. Immunocytochemical analysis revealed that the main lysosomal accumulated material in cultured fibroblasts from patients with Kanzaki disease is Tn-antigen. These data suggest that a prototype of alpha-NAGA deficiency in Kanzaki disease and factors other than the defect of alpha-NAGA may contribute to severe neurological disorders, and Kanzaki disease is thought to be caused by a single enzyme deficiency.


Subject(s)
Fabry Disease/genetics , Hexosaminidases/deficiency , Neuroaxonal Dystrophies/genetics , Antigens, CD/metabolism , Antigens, Tumor-Associated, Carbohydrate/metabolism , Crystallography , Fabry Disease/enzymology , Fibroblasts/metabolism , Hexosaminidases/genetics , Hexosaminidases/metabolism , Humans , Immunohistochemistry , Lysosomal Membrane Proteins , Models, Molecular , Protein Structure, Tertiary/genetics , Structural Homology, Protein , alpha-N-Acetylgalactosaminidase
15.
J Hum Genet ; 48(11): 582-9, 2003.
Article in English | MEDLINE | ID: mdl-14577003

ABSTRACT

To study the structural basis of the GM2 gangliosidosis B variant, we constructed the three-dimensional structures of the human beta-hexosaminidase alpha-subunit and the heterodimer of the alpha- and beta-subunits, Hex A, by homology modeling. The alpha-subunit is composed of two domains, domains I and II. Nine mutant models due to specific missense mutations were constructed as well and compared with the wild type to determine structural defects. These nine mutations were divided into five groups according to structural defects. R178H is deduced to affect the active site directly, because R178 is important for binding to the substrate. C458Y and W420C are predicted to cause drastic structural changes in the barrel structure carrying the active site pocket. R504C/H is deduced to introduce a disruption of an essential binding with D494 in the beta-subunit for dimerization. R499C/H, located in an extra-helix, is deduced to disrupt hydrogen bonds with domain I and the barrel. R170W and L484P are deduced to affect the interface between domains I and II, causing destabilization. The structural defects reflect the biochemical abnormalities of the disease.


Subject(s)
Gangliosidoses, GM2/enzymology , beta-N-Acetylhexosaminidases/chemistry , Amino Acid Substitution , Cells, Cultured , Gangliosidoses, GM2/genetics , Genetic Variation , Hexosaminidase A , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Models, Molecular , Mutation , Protein Conformation , beta-N-Acetylhexosaminidases/genetics
16.
J Hum Genet ; 47(4): 176-83, 2002.
Article in English | MEDLINE | ID: mdl-12166653

ABSTRACT

To determine the molecular basis of the GM2 gangliosidosis 0 variant, we constructed a three-dimensional structure of the human beta-hexosaminidase beta-subunit by homology modeling. It is composed of two domains, domains I and II, and has three disulfide bonds. C534 is located on an extra helix in domain II and forms a disulfide bond with C551. The extra helix is structurally located near domain I. C534Y, identified in a patient with the infantile form of the disease, was deduced to cause disruption of the disulfide bond, which results in a large conformational change of the extra helix, stabilizing the two domains. The drastic change in the protein structure results in a deficiency of the mature beta-subunit, and deficient activities of beta-hexosaminidases A (abeta) and B (betabeta), followed by abundant accumulation of GM2 ganglioside in the patient's cells. R505 is located on the eighth helix of domain II. R505Q, found in a patient with the chronic form of the disease, is predicted to influence the surface structure of the beta-subunit, although it does not affect the active site. The amino acid substitution causes a partial processing defect and decreased enzyme activities, which result in moderate accumulation of GM2 ganglioside in the patient's cells. The structural defects well reflect biochemical and phenotypic abnormalities of the disease.


Subject(s)
Gangliosidoses, GM2/genetics , beta-N-Acetylhexosaminidases/genetics , Adult , Amino Acid Substitution , Blotting, Western , Cells, Cultured , Female , Fibroblasts , Humans , Immunohistochemistry , Infant , Male , Models, Molecular , Protein Conformation , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...