Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 321
Filtrar
1.
Medicine (Baltimore) ; 101(1): e28435, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-35029890

RESUMO

RATIONALE: Monosialotetrahexosylganglioside (GM1) gangliosidosis is a rare lysosomal storage disorder caused by the deficiency of ß-galactosidase. Because clinical symptoms of GM1 gangliosidosis overlap with other neurodevelopmental disorders, the diagnosis of this disease is not easy, specifically in late infantile GM1 gangliosidosis. This report described a case of late-infantile GM1 gangliosidosis mistaken for juvenile idiopathic arthritis. PATIENT CONCERNS: A 16-year-old girl was referred to our hospital due to persistent multiple joint deformities and mental retardation, which could not be explained by juvenile idiopathic arthritis. DIAGNOSIS: We made a diagnosis of late infantile GM1 gangliosidosis through enzyme assays and genetic testing after a skeletal survey. INTERVENTIONS: The patient underwent cervical domeplasty and laminectomy for cord compression and received rehabilitation treatment. OUTCOMES: The patient is receiving multidisciplinary care at a tertiary center for variable skeletal disease and conditions associated with GM1 gangliosidosis. LESSONS: Late infantile GM1 gangliosidosis should be considered in the differential diagnosis of progressive neurologic decline and skeletal dysostosis.


Assuntos
Gangliosidose GM1/diagnóstico , beta-Galactosidase/deficiência , Adolescente , Artrite Juvenil , Diagnóstico Diferencial , Feminino , Humanos , Doenças por Armazenamento dos Lisossomos , Imageamento por Ressonância Magnética
2.
Infect Immun ; 84(10): 2922-32, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27481242

RESUMO

Streptococcus pneumoniae is an opportunistic pathogen that colonizes the nasopharynx. Herein we show that carbon availability is distinct between the nasopharynx and bloodstream of adult humans: glucose is absent from the nasopharynx, whereas galactose is abundant. We demonstrate that pneumococcal neuraminidase A (NanA), which cleaves terminal sialic acid residues from host glycoproteins, exposed galactose on the surface of septal epithelial cells, thereby increasing its availability during colonization. We observed that S. pneumoniae mutants deficient in NanA and ß-galactosidase A (BgaA) failed to form biofilms in vivo despite normal biofilm-forming abilities in vitro Subsequently, we observed that glucose, sucrose, and fructose were inhibitory for biofilm formation, whereas galactose, lactose, and low concentrations of sialic acid were permissive. Together these findings suggested that the genes involved in biofilm formation were under some form of carbon catabolite repression (CCR), a regulatory network in which genes involved in the uptake and metabolism of less-preferred sugars are silenced during growth with preferred sugars. Supporting this notion, we observed that a mutant deficient in pyruvate oxidase, which converts pyruvate to acetyl-phosphate under non-CCR-inducing growth conditions, was unable to form biofilms. Subsequent comparative transcriptome sequencing (RNA-seq) analyses of planktonic and biofilm-grown pneumococci showed that metabolic pathways involving the conversion of pyruvate to acetyl-phosphate and subsequently leading to fatty acid biosynthesis were consistently upregulated during diverse biofilm growth conditions. We conclude that carbon availability in the nasopharynx impacts pneumococcal biofilm formation in vivo Additionally, biofilm formation involves metabolic pathways not previously appreciated to play an important role.


Assuntos
Biofilmes/crescimento & desenvolvimento , Metabolismo dos Carboidratos/fisiologia , Carboidratos/farmacologia , Galactose/farmacocinética , Neuraminidase/fisiologia , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/fisiologia , Análise de Variância , Animais , Biofilmes/efeitos dos fármacos , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Galactose/metabolismo , Galactose/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ácido N-Acetilneuramínico/metabolismo , Líquido da Lavagem Nasal/química , Septo Nasal/metabolismo , Septo Nasal/microbiologia , Nasofaringe/metabolismo , Nasofaringe/microbiologia , Neuraminidase/metabolismo , Infecções Pneumocócicas/metabolismo , Streptococcus pneumoniae/efeitos dos fármacos , beta-Galactosidase/deficiência , beta-Galactosidase/metabolismo
3.
Am J Med Genet A ; 170(3): 634-44, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26646981

RESUMO

Background GM1 gangliosidosis is a lysosomal storage disorder caused by mutations in GLB1, encoding ß-galactosidase. The range of severity is from type I infantile disease, lethal in early childhood, to type III adult onset, resulting in gradually progressive neurological symptoms in adulthood. The intermediate group of patients has been recently classified as having type II late infantile subtype with onset of symptoms at one to three years of age or type II juvenile subtype with symptom onset at 2-10 years. To characterize disease severity and progression, six Late infantile and nine juvenile patients were evaluated using magnetic resonance imaging (MRI), and MR spectroscopy (MRS). Since difficulties with ambulation (gross motor function) and speech (expressive language) are often the first reported symptoms in type II GM1, patients were also scored in these domains. Deterioration of expressive language and ambulation was more rapid in the late infantile patients. Fourteen MRI scans in six Late infantile patients identified progressive atrophy in the cerebrum and cerebellum. Twenty-six MRI scans in nine juvenile patients revealed greater variability in extent and progression of atrophy. Quantitative MRS demonstrated a deficit of N-acetylaspartate in both the late infantile and juvenile patients with greater in the late infantile patients. This correlates with clinical measures of ambulation and expressive language. The two subtypes of type II GM1 gangliosidosis have different clinical trajectories. MRI scoring, quantitative MRS and brain volume correlate with clinical disease progression and may serve as important minimally-invasive outcome measures for clinical trials.


Assuntos
Atrofia/diagnóstico , Gangliosidose GM1/diagnóstico , Distúrbios da Fala/diagnóstico , beta-Galactosidase/genética , Adolescente , Idade de Início , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Atrofia/genética , Atrofia/metabolismo , Atrofia/patologia , Cerebelo/metabolismo , Cerebelo/patologia , Cérebro/metabolismo , Cérebro/patologia , Criança , Pré-Escolar , Progressão da Doença , Feminino , Gangliosidose GM1/genética , Gangliosidose GM1/metabolismo , Gangliosidose GM1/patologia , Expressão Gênica , Humanos , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Masculino , Limitação da Mobilidade , Índice de Gravidade de Doença , Fala , Distúrbios da Fala/genética , Distúrbios da Fala/metabolismo , Distúrbios da Fala/patologia , Adulto Jovem , beta-Galactosidase/deficiência
4.
ASN Neuro ; 7(1)2015.
Artigo em Inglês | MEDLINE | ID: mdl-25694553

RESUMO

GM1-gangliosidosis is a glycosphingolipid lysosomal storage disease involving accumulation of GM1 and its asialo form (GA1) primarily in the brain. Thin-layer chromatography and X-ray diffraction were used to analyze the lipid content/composition and the myelin structure of the optic and sciatic nerves from 7- and 10-month old ß-galactosidase (ß-gal) +/? and ß-gal -/- mice, a model of GM1gangliosidosis. Optic nerve weight was lower in the ß-gal -/- mice than in unaffected ß-gal +/? mice, but no difference was seen in sciatic nerve weight. The levels of GM1 and GA1 were significantly increased in both the optic nerve and sciatic nerve of the ß-gal -/- mice. The content of myelin-enriched cerebrosides, sulfatides, and plasmalogen ethanolamines was significantly lower in optic nerve of ß-gal -/- mice than in ß-gal +/? mice; however, cholesteryl esters were enriched in the ß-gal -/- mice. No major abnormalities in these lipids were detected in the sciatic nerve of the ß-gal -/- mice. The abnormalities in GM1 and myelin lipids in optic nerve of ß-gal -/- mice correlated with a reduction in the relative amount of myelin and periodicity in fresh nerve. By contrast, the relative amount of myelin and periodicity in the sciatic nerves from control and ß-gal -/- mice were indistinguishable, suggesting minimal pathological involvement in sciatic nerve. Our results indicate that the greater neurochemical pathology observed in the optic nerve than in the sciatic nerve of ß-gal -/- mice is likely due to the greater glycolipid storage in optic nerve.


Assuntos
Gangliosidose GM1/genética , Gangliosidose GM1/patologia , Bainha de Mielina/patologia , Nervo Óptico/citologia , Nervo Óptico/patologia , Nervo Isquiático/patologia , beta-Galactosidase/deficiência , Animais , Cromatografia em Camada Fina , Densitometria , Genótipo , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Transgênicos , Bainha de Mielina/metabolismo , Difração de Raios X , beta-Galactosidase/genética
5.
Yakugaku Zasshi ; 133(5): 509-17, 2013.
Artigo em Japonês | MEDLINE | ID: mdl-23649392

RESUMO

G(M1)-gangliosidosis and Morquio B are rare lysosomal storage diseases associated with a neurodegenerative disorder or dwarfism and skeletal abnormalities, respectively. These diseases are caused by deficiencies in the lysosomal enzyme human ß-D-galactosidase (h-ß-GAL), which lead to accumulations of the h-ß-GAL substrates, G(M1) ganglioside and keratan sulfate due to mutations in the h-ß-GAL gene. H-ß-GAL is an exoglycosidase that catalyzes the hydrolysis of terminal ß-linked galactose residues. Here, we present the crystal structures of h-ß-GAL in complex with its catalytic product galactose or with its inhibitor 1-deoxygalactonojirimycin. H-ß-GAL showed a novel homodimer structure; each monomer was comprised of a catalytic TIM barrel domain followed by ß-domain 1 and ß-domain 2. The long loop region connecting the TIM barrel domain with ß-domain 1 was responsible for the dimerization. To gain structural insight into the molecular defects of h-ß-GAL in the above diseases, the disease-causing mutations were mapped onto the three-dimensional structure. Finally, the possible causes of the diseases are discussed.


Assuntos
Doenças por Armazenamento dos Lisossomos/genética , Mutação , beta-Galactosidase/química , beta-Galactosidase/genética , Animais , Gangliosídeos/metabolismo , Gangliosidose GM1/genética , Humanos , Sulfato de Queratano/metabolismo , Doenças por Armazenamento dos Lisossomos/metabolismo , Lisossomos/metabolismo , Conformação Molecular , Mucopolissacaridose IV/genética , Estrutura Terciária de Proteína , Especificidade por Substrato , beta-Galactosidase/deficiência
6.
FEBS J ; 277(14): 2970-86, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20546307

RESUMO

Urine, amniotic fluid and ascitic fluid samples of galactosialidosis patients were analyzed and structurally characterized for free oligosaccharides using capillary high-performance anion-exchange chromatography with pulsed amperometric detection and online mass spectrometry. In addition to the expected endo-beta-N-acetylglucosaminidase-cleaved products of complex-type sialylated N-glycans, O-sulfated oligosaccharide moieties were detected. Moreover, novel carbohydrate moieties with reducing-end hexose residues were detected. On the basis of structural features such as a hexose-N-acetylhexosamine-hexose-hexose consensus sequence and di-sialic acid units, these oligosaccharides are thought to represent, at least in part, glycan moieties of glycosphingolipids. In addition, C(1)-oxidized, aldohexonic acid-containing versions of most of these oligosaccharides were observed. These observations suggest an alternative catabolism of glycosphingolipids in galactosialidosis patients: oligosaccharide moieties from glycosphingolipids would be released by a hitherto unknown ceramide glycanase activity. The results show the potential and versatility of the analytical approach for structural characterization of oligosaccharides in various body fluids.


Assuntos
Líquido Amniótico/química , Líquido Ascítico/química , Doenças por Armazenamento dos Lisossomos/metabolismo , Doenças por Armazenamento dos Lisossomos/urina , Neuraminidase/deficiência , Oligossacarídeos/análise , beta-Galactosidase/deficiência , Sequência de Carboidratos , Catepsina A/genética , Cromatografia por Troca Iônica , Feto/metabolismo , Glicosídeo Hidrolases/metabolismo , Glicoesfingolipídeos/metabolismo , Hexoses/análise , Humanos , Lactente , Recém-Nascido/metabolismo , Recém-Nascido/urina , Doenças por Armazenamento dos Lisossomos/genética , Espectrometria de Massas , Dados de Sequência Molecular , Oligossacarídeos/química , Oligossacarídeos/urina , Oligossacarídeos de Cadeias Ramificadas/análise , Oligossacarídeos de Cadeias Ramificadas/química , Oligossacarídeos de Cadeias Ramificadas/urina , Açúcares Ácidos/análise , Urina/química
7.
Appl Biochem Biotechnol ; 162(8): 2294-312, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20549573

RESUMO

Although ß-galactosidases are physiologically a very important enzyme and have may therapeutics applications, very little is known about the stability and the folding aspects of the enzyme. We have used ß-galactosidase from Pisum sativum (PsBGAL) as model system to investigate stability, folding, and function relationship of ß-galactosidases. PsBGAL is a vacuolar protein which has a tendency to multimerize at acidic pH with protein concentration ≥100 µg mL⁻¹ and dissociates into its subunits above neutral pH. It exhibits maximum activity as well as stability under acidic conditions. Further, it has different conformational orientations and core secondary structures at different pH. Substantial predominance of ß-content and interfacial interactions through Trp residues play crucial role in pH-dependent multimerization of enzyme. Equilibrium unfolding of PsBGAL at acidic pH follows four-state model when monitored by changes in the secondary structure with two intermediates: one resembling to molten globule-like state while unfolding seen from activity and tertiary structure of PsBGAL fits to two-state model. Unfolding of PsBGAL at higher pH always follows two-state model. Furthermore, unfolding of PsBGAL reveals that it has at least two domains: α/ß barrel containing catalytic site and the other is rich in ß-content responsible for enzyme multimerization.


Assuntos
Pisum sativum/enzimologia , Multimerização Proteica , Estrutura Quaternária de Proteína , beta-Galactosidase/química , beta-Galactosidase/metabolismo , Animais , Concentração de Íons de Hidrogênio , Hidrólise , Interações Hidrofóbicas e Hidrofílicas , Lactose/metabolismo , Leite/metabolismo , Doenças das Plantas , Dobramento de Proteína , Estrutura Secundária de Proteína , Desdobramento de Proteína , Sementes/enzimologia , beta-Galactosidase/deficiência
8.
Biochim Biophys Acta ; 1792(10): 982-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19607915

RESUMO

beta-galactosidase (GLB1) forms a functional lysosomal multienzyme complex with lysosomal protective protein (PPCA) and neuraminidase 1 (NEU1) which is important for its intracellular processing and activity. Mutations in the beta-galactosidase gene cause the lysosomal storage disease G(M1)-gangliosidosis. In order to identify additional molecular changes associated with the presence of beta-galactosidase mutations, the expression of canine lysosomal multienzyme complex components in GLB1(+/+), GLB1(+/-) and GLB1(-/-) fibroblasts was investigated by quantitative RT-PCR, Western blot and enzymatic assays. Quantitative RT-PCR revealed differential regulation of total beta-galactosidase, beta-galactosidase variants and protective protein for beta-galactosidase gene (PPGB) in GLB1(+/-) and GLB1(-/-) compared to GLB1(+/+) fibroblasts. Furthermore, it was shown that PPGB levels gradually increased with the number of mutant beta-galactosidase alleles while no change in the NEU1 expression was observed. This is the first study that simultaneously examine the effect of GLB1(+/+), GLB1(+/-) and GLB1(-/-) genotypes on the expression of lysosomal multienzyme complex components. The findings reveal a possible adaptive process in GLB1 homozygous mutant and heterozygous individuals that could facilitate the design of efficient therapeutic strategies.


Assuntos
Catepsina A/genética , Lisossomos/enzimologia , Complexos Multienzimáticos/genética , Mutação/genética , Neuraminidase/genética , beta-Galactosidase/genética , Animais , Catepsina A/metabolismo , Cães , Fibroblastos/enzimologia , Regulação Enzimológica da Expressão Gênica , Complexos Multienzimáticos/metabolismo , Neuraminidase/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , beta-Galactosidase/deficiência , beta-Galactosidase/metabolismo
11.
Gene Ther ; 16(2): 303-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18818671

RESUMO

GM1-gangliosidosis is a lysosomal storage disease (LSD) caused by an autosomal recessive deficiency of lysosomal acid beta-galactosidase (betagal). This leads to accumulation of GM1-ganglioside and its asialo derivative GA1 in the central nervous system (CNS), and progressive neurodegeneration. Therapeutic AAV-mediated gene delivery to the brain for LSDs has proven very successful in several animal models. GM1-gangliosidosis is also a prime candidate for AAV-mediated gene therapy in the CNS. As global neuropathology characterizes the most severe forms of this disease, therapeutic interventions need to achieve distribution of betagal throughout the entire CNS. Therefore, careful consideration of routes of administration and target structures from where metabolically active enzyme can be produced, released and distributed throughout the CNS, is necessary. The goal of this study was to investigate the pattern and mechanism of distribution of betagal in the adult GM1-gangliosidosis mouse brain upon hippocampal injection of an AAV vector-encoding betagal. We found evidence that three different mechanisms contribute to its distribution in the brain: (1) diffusion; (2) axonal transport within neurons from the site of production; (3) CSF flow in the perivascular space of Virchow-Robin. In addition, we found evidence of axonal transport of vector-encoded mRNA.


Assuntos
Encéfalo/enzimologia , Gangliosidose GM1/enzimologia , Terapia Genética/métodos , beta-Galactosidase/genética , Animais , Transporte Axonal , Dependovirus/genética , Modelos Animais de Doenças , Gangliosidose GM1/terapia , Vetores Genéticos/farmacocinética , Hipocampo/enzimologia , Camundongos , Camundongos Knockout , Neurônios/fisiologia , RNA Mensageiro/genética , Distribuição Tecidual , beta-Galactosidase/biossíntese , beta-Galactosidase/deficiência , beta-Galactosidase/farmacocinética
12.
Diabetes ; 57(12): 3307-14, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18835939

RESUMO

OBJECTIVE: Insulin resistance is an independent risk factor for the development of cardiovascular atherosclerosis. A key step in the development of atherosclerosis is endothelial dysfunction, manifest by a reduction in bioactivity of nitric oxide (NO). Insulin resistance is associated with endothelial dysfunction; however, the mechanistic relationship between these abnormalities and the role of impaired endothelial insulin signaling versus global insulin resistance remains unclear. RESEARCH DESIGN AND METHODS: To examine the effects of insulin resistance specific to the endothelium, we generated a transgenic mouse with endothelium-targeted overexpression of a dominant-negative mutant human insulin receptor (ESMIRO). This receptor has a mutation (Ala-Thr(1134)) in its tyrosine kinase domain that disrupts insulin signaling. Humans with the Thr(1134) mutation are insulin resistant. We performed metabolic and vascular characterization of this model. RESULTS: ESMIRO mice had preserved glucose homeostasis and were normotensive. They had significant endothelial dysfunction as evidenced by blunted aortic vasorelaxant responses to acetylcholine (ACh) and calcium ionophore. Furthermore, the vascular action of insulin was lost in ESMIRO mice, and insulin-induced endothelial NO synthase (eNOS) phosphorylation was blunted. Despite this phenotype, ESMIRO mice demonstrate similar levels of eNOS mRNA and protein expression to wild type. ACh-induced relaxation was normalized by the superoxide dismutase mimetic, Mn(III)tetrakis(1-methyl-4-pyridyl) porphyrin pentachloride. Endothelial cells of ESMIRO mice showed increased superoxide generation and increased mRNA expression of the NADPH oxidase isoforms Nox2 and Nox4. CONCLUSIONS: Selective endothelial insulin resistance is sufficient to induce a reduction in NO bioavailability and endothelial dysfunction that is secondary to increased generation of reactive oxygen species. This arises independent of a significant metabolic phenotype.


Assuntos
Endotélio Vascular/fisiologia , Resistência à Insulina/fisiologia , Alanina , Substituição de Aminoácidos , Animais , Glicemia/metabolismo , Clonagem Molecular , Teste de Tolerância a Glucose , Homeostase , Humanos , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Mutação , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo III/genética , Plasmídeos , Reação em Cadeia da Polimerase , Receptor de Insulina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Treonina , beta-Galactosidase/deficiência , beta-Galactosidase/genética
13.
Am J Med Genet A ; 146A(13): 1736-40, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18546276

RESUMO

The patient is a 24-year-old woman who first came for consultation at age 10 years. Based on clinical phenotype and thin-layer chromatography of urinary oligosaccharides, peripheral leukocytes were sent for beta-galactosidase assay. This testing showed a deficiency in enzyme activity, and gene mutation analysis identified a previously reported mutation p.H281Y (875C > T) and a novel mutation p.W273R (817T > C). Unlike previously reported patients, mutant enzymes in this patient's cultured skin fibroblasts did not respond to treatment with a chaperone compound, N-octyl-4-epi-beta-valienamine.


Assuntos
Mucopolissacaridose IV/enzimologia , Mucopolissacaridose IV/genética , beta-Galactosidase/deficiência , beta-Galactosidase/genética , Adulto , Substituição de Aminoácidos , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Hexosaminas/farmacologia , Humanos , Técnicas In Vitro , Mucopolissacaridose IV/patologia , Fenótipo , Mutação Puntual , Pele/efeitos dos fármacos , Pele/enzimologia
14.
J Comp Neurol ; 509(6): 642-60, 2008 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-18551525

RESUMO

Mutations in the coding region of the WFS1 gene cause Wolfram syndrome, a rare multisystem neurodegenerative disorder of autosomal recessive inheritance. Patients with Wolfram syndrome display considerable clinical pleiomorphism, and symptoms such as neurological complications and psychiatric disorders are common. In the present study we have characterized Wfs1 expression pattern in the mouse central nervous system by using a combination of immunohistochemistry on wild-type mice and X-Gal staining of Wfs1 knockout mice with targeted insertion of the lacZ reporter. We identified a robust enrichment of Wfs1 protein in the central extended amygdala and ventral striatum. Prominent Wfs1 expression was seen in the hippocampal CA1 region, parasubiculum, superficial part of the second and third layers of the prefrontal cortex and proisocortical areas, hypothalamic magnocellular neurosecretory system, and central auditory pathway. Wfs1 expression was also detected in numerous brainstem nuclei and in laminae VIII and IX of the spinal cord. Wfs1-positive nerve fibers were found in the medial forebrain bundle, reticular part of the substantia nigra, globus pallidus, posterior caudate putamen, lateral lemniscus, alveus, fimbria, dorsal hippocampal commissure, subiculum, and to a lesser extent in the central sublenticular extended amygdala, compact part of substantia nigra, and ventral tegmental area. The neuroanatomical findings suggest that the lack of Wfs1 protein function can be related to several neurological and psychiatric symptoms found in Wolfram syndrome. Enrichment of Wfs1 protein in the central extended amygdala suggests a role in the modulation of anxiety and fear.


Assuntos
Sistema Nervoso Central/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Síndrome de Wolfram/genética , Animais , Primers do DNA , Genes Reporter , Imuno-Histoquímica , Proteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , beta-Galactosidase/deficiência , beta-Galactosidase/genética
15.
Brain Dev ; 30(9): 595-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18396002

RESUMO

Galactosialidosis is a rare lysosomal storage disease caused by a combined deficiency of lysosomal beta-galactosidase and neuraminidase, due to a primary defect in protective protein/cathepsin A. Three subtypes are recognized: the early infantile type, the late infantile type, and the juvenile/adult type. Here, we report a case of early infantile galactosialidosis in a female who was born at 31 weeks of gestation, after detection of fetal ascites at 21 weeks of gestation and development of fetal hydrops. After birth she received intensive treatment that led to improvement of edema and pleural effusion, but ascites slowly developed. She died of renal failure on day 207. An autopsy showed that all organs contained vacuolated cells, compatible with a storage disease. The patient had decreased activity of beta-galactosidase and undetectable neuraminidase activity in fibroblasts. A single A-G base transition at position 146 of exon 1 (Q49R) in protective protein/cathepsin A gene was found. The mutation has been reported previously in a Japanese patient with different phenotypes. However homozygous Q49R mutation detected in our case was severe prognosis.


Assuntos
Catepsina A/genética , Doenças por Armazenamento dos Lisossomos/genética , Neuraminidase/deficiência , Mutação Puntual , beta-Galactosidase/deficiência , Encéfalo/metabolismo , Encéfalo/patologia , Catepsina A/metabolismo , Análise Mutacional de DNA , Evolução Fatal , Feminino , Humanos , Hidropisia Fetal/etiologia , Hidropisia Fetal/patologia , Lactente , Recém-Nascido , Rim/metabolismo , Rim/patologia , Fígado/metabolismo , Fígado/patologia , Doenças por Armazenamento dos Lisossomos/complicações , Doenças por Armazenamento dos Lisossomos/metabolismo , Doenças por Armazenamento dos Lisossomos/patologia , Miocárdio/metabolismo , Miocárdio/patologia
16.
Pathol Int ; 58(5): 295-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18429828

RESUMO

Galactosialidosis is an autosomal recessive lysosomal disease associated with a deficiency of beta-galactosidase and neuraminidase. Described herein is the case of a young adult who had been diagnosed with galactosialidosis at 8 years of age. At the age of 30 years, proteinuria and hematuria appeared and the patient underwent a renal biopsy 1 year later. Light microscopy of the kidney sections indicated fine granular contents in the cytoplasm of glomerular endothelial and epithelial cells, arteriolar smooth muscles and proximal tubular epithelial cells on periodic acid silver-methenamin (PAM) stain. Electron microscopy of these cells indicated enlarged, smooth endoplasmic reticulum and lysosomes containing 150 nm-wide rods with a fine lattice structure at 66 A periodicity. Moreover, electron-dense deposits were located in the paramesangial area. Immunofluorescence staining indicated diffuse and global anti-human IgA and C3-positive staining as a mesangial pattern. Given these findings this patient was therefore diagnosed with both galactosialidosis and IgA nephropathy. This is the first report to describe light and electron microscopy observations of storage materials in the kidneys in young/adult galactosialidosis.


Assuntos
Glomerulonefrite por IGA/patologia , Rim/patologia , Doenças por Armazenamento dos Lisossomos/patologia , Adulto , Biópsia , Complemento C3/metabolismo , Feminino , Imunofluorescência , Mesângio Glomerular/ultraestrutura , Glomerulonefrite por IGA/complicações , Glomerulonefrite por IGA/metabolismo , Humanos , Imunoglobulina A/metabolismo , Rim/metabolismo , Glomérulos Renais/patologia , Glomérulos Renais/ultraestrutura , Doenças por Armazenamento dos Lisossomos/complicações , Doenças por Armazenamento dos Lisossomos/metabolismo , Organelas/ultraestrutura , beta-Galactosidase/deficiência , beta-Galactosidase/metabolismo
17.
Biochem Biophys Res Commun ; 367(3): 616-22, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18190792

RESUMO

G(M1)-gangliosidosis is an autosomal recessive lysosomal lipid storage disorder, caused by mutations of the lysosomal beta-galactosidase (beta-gal) and results in the accumulation of G(M1). The underlying mechanisms of neurodegeneration are poorly understood. Here we demonstrate increased autophagy in beta-gal-deficient (beta-gal(-/-)) mouse brains as evidenced by elevation of LC3-II and beclin-1 levels. Activation of autophagy in the beta-gal(-/-) brain was found to be accompanied with enhanced Akt-mTOR and Erk signaling. In addition, the mitochondrial cytochrome c oxidase activity was significantly decreased in brains and cultured astrocytes from beta-gal(-/-) mouse. Mitochondria isolated from beta-gal(-/-) astrocytes were morphologically abnormal and had a decreased membrane potential. These cells were more sensitive to oxidative stress than wild type cells and this sensitivity was suppressed by ATP, an autophagy inhibitor 3-methyladenine and a pan-caspase inhibitor z-VAD-fmk. These results suggest activation of autophagy leading to mitochondrial dysfunction in the brain of G(M1)-gangliosidosis.


Assuntos
Autofagia , Encéfalo/patologia , Gangliosidose GM1/patologia , Mitocôndrias/patologia , Adenina/análogos & derivados , Adenina/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Proteínas Reguladoras de Apoptose , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteína Beclina-1 , Encéfalo/ultraestrutura , Células Cultivadas , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Inibidores Enzimáticos/farmacologia , Gangliosídeo G(M1)/metabolismo , Gangliosidose GM1/genética , Lisossomos/metabolismo , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/enzimologia , Paraquat/farmacologia , Proteínas Quinases/metabolismo , Proteínas/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR , beta-Galactosidase/deficiência
18.
Int J Food Microbiol ; 122(1-2): 148-55, 2008 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-18164089

RESUMO

We reported four foodborne disease outbreaks in Taiwan caused by sucrose-nonfermenting and by beta-galactosidase-deficient variants of non-O1, non-O139 Vibrio cholerae. The sucrose-nonfermenting vibrios collected from three outbreaks were biochemically identified to be V. mimicus and the beta-galactosidase-deficient vibrios from an outbreak to be V. alginolyticus. However, molecular methods including DNA-DNA hybridization, fatty acid profile analysis, and sequence analysis of 16S rRNA, oriC, pyrH, recA, and rpoA indicated that these vibrios should be V. cholerae. These V. cholerae variants carried two hemolysin genes, hlyA and hlx, but contained neither cholera toxin gene, ctx, V. mimicus hemolysin gene, vmh, nor thermo-directed hemolysin, tdh. The sucrose-nonfermenting variants of V. cholerae shared a high level of genetic relatedness; they could derive from a common clone. In our record from 1995 to date, this was the first time that V. cholerae variants were discovered as etiologic agents for foodborne disease outbreaks in Taiwan.


Assuntos
Cólera/epidemiologia , Contaminação de Alimentos/análise , Vibrio cholerae , beta-Galactosidase/deficiência , Surtos de Doenças , Fermentação , Microbiologia de Alimentos , Humanos , Epidemiologia Molecular , Reação em Cadeia da Polimerase/métodos , Sacarose/metabolismo , Taiwan/epidemiologia , Vibrio cholerae/classificação , Vibrio cholerae/enzimologia , Vibrio cholerae/patogenicidade , Virulência/genética
19.
J Cell Mol Med ; 12(5A): 1661-71, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18088383

RESUMO

G(M1)-gangliosidosis is a lysosomal storage disorder caused by a deficiency of ss-galactosidase activity. Human GM1-gangliosidosis has been classified into three forms according to the age of clinical onset and specific biochemical parameters. In the present study, a canine model for type II late infantile human GM1-gangliosidosis was investigated 'in vitro' in detail. For a better understanding of the molecular pathogenesis underlying G(M1)-gangliosidosis the study focused on the analysis of the molecular events and subsequent intracellular protein trafficking of beta-galactosidase. In the canine model the genetic defect results in exclusion or inclusion of exon 15 in the mRNA transcripts and to translation of two mutant precursor proteins. Intracellular localization, processing and enzymatic activity of these mutant proteins were investigated. The obtained results suggested that the beta-galactosidase C-terminus encoded by exons 15 and 16 is necessary for correct C-terminal proteolytic processing and enzyme activity but does not affect the correct routing to the lysosomes. Both mutant protein precursors are enzymatically inactive, but are transported to the lysosomes clearly indicating that the amino acid sequences encoded by exons 15 and 16 are necessary for correct folding and association with protective protein/cathepsin A, whereas the routing to the lysosomes is not influenced. Thus, the investigated canine model is an appropriate animal model for the human late infantile form and represents a versatile system to test gene therapeutic approaches for human and canine G(M1)-gangliosidosis.


Assuntos
Gangliosidose GM1/enzimologia , Processamento de Proteína Pós-Traducional , beta-Galactosidase/metabolismo , Animais , Animais Geneticamente Modificados , Células Cultivadas , Modelos Animais de Doenças , Cães , Gangliosidose GM1/genética , Gangliosidose GM1/patologia , Humanos , Mutação/genética , beta-Galactosidase/deficiência , beta-Galactosidase/genética
20.
Int J Food Sci Nutr ; 59(2): 116-22, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17852503

RESUMO

Different types of herbal yogurts were developed by mixing standardized milk with pretreated herbs, namely tulsi leaf (Ocimum sanctum), pudina leaf (Mentha arvensis) and coriander leaf (Coriandrum sativum), with leaves separately and a 1:1 (v/v) mixture of the strains of lactic starter cultures---Lactobacillus acidophilus (NCIM 2903) and Lactobacillus plantarum (NCIM 2083)-followed by incubation at 40 degrees C for 6 h. The beta-galactosidase enzymatic activity of the abovementioned herbal yogurts was determined and interestingly noted to exhibit higher enzymatic activity compared with the control yogurt (without any herbs). Among all herbal yogurts, tulsi yogurt had the maximum beta-galactosidase activity.


Assuntos
Lactobacillus/enzimologia , Preparações de Plantas/farmacologia , Iogurte/análise , beta-Galactosidase/metabolismo , Antioxidantes/análise , Antioxidantes/farmacologia , Coriandrum/química , Fermentação , Temperatura Alta , Lactobacillus/crescimento & desenvolvimento , Lamiaceae/química , Nitrofenóis/metabolismo , beta-Galactosidase/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...