Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Lipids ; 2024: 4530255, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38623278

RESUMO

Gangliosides, sialic acid-containing glycosphingolipids, are abundant in cell membranes and primarily involved in controlling cell signaling and cell communication. The altered ganglioside pattern has been demonstrated in several neurodegenerative diseases, characterized during early-onset or infancy, emphasizing the significance of gangliosides in the brain. Enzymes required for the biosynthesis of gangliosides are linked to several devastating neurological disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), hereditary spastic paraplegia (HSP). In this review, we summarized not only the critical roles of biosynthetic enzymes and their inhibitors in ganglioside metabolism but also the efficacy of treatment strategies of ganglioside to address their significance in those diseases.

2.
Mol Genet Metab ; 141(3): 108140, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38262289

RESUMO

Tay-Sachs disease is a rare lysosomal storage disorder (LSD) caused by a mutation in the HexA gene coding ß-hexosaminidase A enzyme. The disruption of the HexA gene causes the accumulation of GM2 ganglioside resulting in progressive neurodegeneration in humans. Surprisingly, Hexa-/- mice did not show neurological phenotypes. Our group recently generated a murine model of Tay-Sachs disease exhibiting excessive GM2 accumulation and severe neuropathological abnormalities mimicking Tay-Sachs patients. Previously, we reported impaired autophagic flux in the brain of Hexa/-Neu3-/- mice. However, regulation of autophagic flux using inducers has not been clarified in Tay-Sachs disease cells. Here, we evaluated the effects of lithium treatment on dysfunctional autophagic flux using LC3 and p62 in the fibroblast and neuroglia of Hexa-/-Neu3-/- mice and Tay-Sachs patients. We discovered the clearance of accumulating autophagosomes, aggregate-prone metabolites, and GM2 ganglioside under lithium-induced conditions. Our data suggest that targeting autophagic flux with an autophagy inducer might be a rational therapeutic strategy for the treatment of Tay-Sachs disease.


Assuntos
Doença de Tay-Sachs , Humanos , Camundongos , Animais , Doença de Tay-Sachs/tratamento farmacológico , Doença de Tay-Sachs/genética , Lítio/farmacologia , Lítio/uso terapêutico , Gangliosídeo G(M2) , Autofagia , Compostos de Lítio/uso terapêutico , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo , beta-N-Acetil-Hexosaminidases/uso terapêutico
3.
Mol Genet Metab ; 140(3): 107648, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37598508

RESUMO

Lysosomal storage diseases (LSDs) are caused by monogenic mutations in genes encoding for proteins related to the lysosomal function. Lysosome plays critical roles in molecule degradation and cell signaling through interplay with many other cell organelles, such as mitochondria, endoplasmic reticulum, and peroxisomes. Even though several strategies (i.e., protein replacement and gene therapy) have been attempted for LSDs with promising results, there are still some challenges when hard-to-treat tissues such as bone (i.e., cartilages, ligaments, meniscus, etc.), the central nervous system (mostly neurons), and the eye (i.e., cornea, retina) are affected. Consistently, searching for novel strategies to reach those tissues remains a priority. Molecular Trojan Horses have been well-recognized as a potential alternative in several pathological scenarios for drug delivery, including LSDs. Even though molecular Trojan Horses refer to genetically engineered proteins to overcome the blood-brain barrier, such strategy can be extended to strategies able to transport and deliver drugs to specific tissues or cells using cell-penetrating peptides, monoclonal antibodies, vesicles, extracellular vesicles, and patient-derived cells. Only some of those platforms have been attempted in LSDs. In this paper, we review the most recent efforts to develop molecular Trojan Horses and discuss how this strategy could be implemented to enhance the current efficacy of strategies such as protein replacement and gene therapy in the context of LSDs.


Assuntos
Barreira Hematoencefálica , Doenças por Armazenamento dos Lisossomos , Humanos , Barreira Hematoencefálica/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/terapia , Sistema Nervoso Central , Terapia Genética/métodos
4.
PLoS One ; 18(3): e0280650, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36928510

RESUMO

Tay-Sachs disease is a lethal lysosomal storage disorder caused by mutations in the HexA gene encoding the α subunit of the lysosomal ß-hexosaminidase enzyme (HEXA). Abnormal GM2 ganglioside accumulation causes progressive deterioration in the central nervous system in Tay-Sachs patients. Hexa-/- mouse model failed to display abnormal phenotype. Recently, our group generated Hexa-/-Neu3-/- mouse showed severe neuropathological indications similar to Tay-Sachs patients. Despite excessive GM2 ganglioside accumulation in the brain and visceral organs, the regulation of autophagy has not been clarified yet in the Tay-Sachs disease mouse model. Therefore, we investigated distinct steps of autophagic flux using markers including LC3 and p62 in four different brain regions from the Hexa-/-Neu3-/- mice model of Tay-Sachs disease. Our data revealed accumulated autophagosomes and autophagolysosomes indicating impairment in autophagic flux in the brain. We suggest that autophagy might be a new therapeutic target for the treatment of devastating Tay-Sachs disease.


Assuntos
Autofagia , Doença de Tay-Sachs , Animais , Camundongos , Autofagia/fisiologia , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Gangliosídeo G(M2)/uso terapêutico , Hexosaminidase A/metabolismo , Doença de Tay-Sachs/metabolismo , Doença de Tay-Sachs/patologia , Modelos Animais de Doenças
5.
Front Mol Biosci ; 9: 892248, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36003081

RESUMO

Introduction: Tay-Sachs disease is an autosomal recessively inherited lysosomal storage disease that results from loss-of-function mutations in the HEXA gene coding ß-hexosaminidase A. HEXA gene deficiency affects the central nervous system owing to GM2 ganglioside accumulation in lysosomes resulting in progressive neurodegeneration in patients. We recently generated a novel mice model with a combined deficiency of ß-hexosaminidase A and neuraminidase 3 (Hexa-/-Neu3-/-) that mimics both the neuropathological and clinical abnormalities of early-onset Tay-Sachs disease. Here, we aimed to explore the secondary accumulation of lipids in the brain of Hexa-/-Neu3-/- mice. Materials and Methods: In the cortex and hippocampus of five-month-old WT, Hexa-/-, Neu3-/-, and Hexa-/-Neu3-/- mice, lipid levels belonging to glycerolipids, glycerophospholipids, and sterol lipids were evaluated using a shotgun lipidomics approach. The levels of myelin were also assessed by luxol fast blue staining and immunohistochemistry using antibodies against myelin basic protein. We further examined glycoconjugate and cholesterol levels by periodic acid-Schiff and filipin staining, respectively. Toluidine blue staining was also performed to display axonal degeneration. Results: Among glycerophospholipids, we demonstrated elevated levels of phosphatidylcholine-ether and lysophosphatidylcholine while decreased levels of phosphatidylcholine and phosphatidylserine in both cortex and hippocampus of Hexa-/-Neu3-/- mice. In the glycerolipid class, we showed an alleviated level of sphingomyelin in both cortex and hippocampus, but the higher levels of diacylglycerol and triacylglycerol were detected in only the hippocampus of Hexa-/-Neu3-/- mice. The lower level of sterol was also detected in the cortex of Hexa-/-Neu3-/- mice but not in the hippocampus. Histochemical studies showed a decrease in the myelin level and axonal degeneration indicating neuronal pathology in the brain of Hexa-/-Neu3-/- mice. Although glycoconjugate accumulation was evident both in the cortex and hippocampus, we did not detect any changes in the level of cholesterol. Conclusion: Our results indicate that alterations in lipid metabolism and neuropathology, such as demyelination and axonal degeneration, might be related to the dysfunctionality of lipid-related cellular pathways like autophagy. Understanding of brain-specific lipid alterations contributes to evaluating the effectiveness of treatments in Hexa-/-Neu3-/- mice in future studies.

6.
Glycoconj J ; 38(6): 649-667, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34686927

RESUMO

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


Assuntos
Mucolipidoses , Neuraminidase/metabolismo , Animais , Gangliosídeos/metabolismo , Lisossomos/metabolismo , Camundongos , Mucolipidoses/metabolismo , Neuraminidase/química , Doenças Neuroinflamatórias , Especificidade por Substrato
7.
J Neuroinflammation ; 17(1): 277, 2020 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-32951593

RESUMO

BACKGROUND: Tay-Sachs disease (TSD), a type of GM2-gangliosidosis, is a progressive neurodegenerative lysosomal storage disorder caused by mutations in the α subunit of the lysosomal ß-hexosaminidase enzyme. This disease is characterized by excessive accumulation of GM2 ganglioside, predominantly in the central nervous system. Although Tay-Sachs patients appear normal at birth, the progressive accumulation of undegraded GM2 gangliosides in neurons leads to death. Recently, an early onset Tay-Sachs disease mouse model, with genotype Hexa-/-Neu3-/-, was generated. Progressive accumulation of GM2 led to premature death of the double KO mice. Importantly, this double-deficient mouse model displays typical features of Tay-Sachs patients, such as cytoplasmic vacuolization of nerve cells, deterioration of Purkinje cells, neuronal death, deceleration in movement, ataxia, and tremors. GM2-gangliosidosis is characterized by acute neurodegeneration preceded by activated microglia expansion, macrophage, and astrocyte activation, along with the production of inflammatory mediators. However, the mechanism of disease progression in Hexa-/-Neu3-/- mice, relevant to neuroinflammation is poorly understood. METHOD: In this study, we investigated the onset and progression of neuroinflammatory changes in the cortex, cerebellum, and retina of Hexa-/-Neu3-/- mice and control littermates by using a combination of molecular genetics and immunochemical procedures. RESULTS: We found elevated levels of pro-inflammatory cytokine and chemokine transcripts, such as Ccl2, Ccl3, Ccl4, and Cxcl10 and also extensive microglial and astrocyte activation and proliferation, accompanied by peripheral blood mononuclear cell infiltration in the vicinity of neurons and oligodendrocytes. Behavioral tests demonstrated a high level of anxiety, and age-dependent loss in both spatial learning and fear memory in Hexa-/-Neu3-/- mice compared with that in the controls. CONCLUSION: Altogether, our data suggest that Hexa-/-Neu3-/- mice display a phenotype similar to Tay-Sachs patients suffering from chronic neuroinflammation triggered by GM2 accumulation. Furthermore, our work contributes to better understanding of the neuropathology in a mouse model of early onset Tay-Sachs disease.


Assuntos
Encéfalo/metabolismo , Modelos Animais de Doenças , Gangliosídeo G(M2)/metabolismo , Mediadores da Inflamação/metabolismo , Retina/metabolismo , Doença de Tay-Sachs/metabolismo , Animais , Encéfalo/patologia , Gangliosídeo G(M2)/genética , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Retina/patologia , Doença de Tay-Sachs/genética , Doença de Tay-Sachs/patologia
8.
JIMD Rep ; 44: 43-54, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29995202

RESUMO

Krabbe disease is a lysosomal storage disease caused by galactosylceramidase deficiency, resulting in neurodegeneration with a rapid clinical downhill course within the first months of life in the classic infantile form. This process may be triggered by the accumulation of galactosylceramide (GalCer) in nervous tissues. Both the enzyme galactosylceramidase and its in vivo activator molecule, saposin A, are essential during GalCer degradation. A clinical manifestation almost identical to Krabbe disease is observed when, instead of the galactosylceramidase protein, the saposin A molecule is defective. Saposin A results from posttranslational processing of the precursor molecule, prosaposin, encoded by the PSAP gene. Clinical and neuroimaging findings in a 7-month-old child strongly suggested Krabbe disease, but this condition was excluded by enzymatic and genetic testing. However, at whole exome sequencing, the previously undescribed homozygous, obviously pathogenic PSAP gene NM_002778.3:c.209T>G(p.Val70Gly) variant was determined in the saposin A domain of the PSAP gene. Fibroblast studies showed GalCer accumulation and the activation of autophagy for the first time in a case of human saposin A deficiency. Our patient represents the second known case in the literature and provides new information concerning the pathophysiology of saposin A deficiency and its intralysosomal effects.

9.
Exp Neurol ; 299(Pt A): 26-41, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28974375

RESUMO

Tay-Sachs disease is a severe lysosomal storage disorder caused by mutations in Hexa, the gene that encodes for the α subunit of lysosomal ß-hexosaminidase A (HEXA), which converts GM2 to GM3 ganglioside. Unexpectedly, Hexa-/- mice have a normal lifespan and show no obvious neurological impairment until at least one year of age. These mice catabolize stored GM2 ganglioside using sialidase(s) to remove sialic acid and form the glycolipid GA2, which is further processed by ß-hexosaminidase B. Therefore, the presence of the sialidase (s) allows the consequences of the Hexa defect to be bypassed. To determine if the sialidase NEU3 contributes to GM2 ganglioside degradation, we generated a mouse model with combined deficiencies of HEXA and NEU3. The Hexa-/-Neu3-/- mice were healthy at birth, but died at 1.5 to 4.5months of age. Thin-layer chromatography and mass spectrometric analysis of the brains of Hexa-/-Neu3-/- mice revealed the abnormal accumulation of GM2 ganglioside. Histological and immunohistochemical analysis demonstrated cytoplasmic vacuolation in the neurons. Electron microscopic examination of the brain, kidneys and testes revealed pleomorphic inclusions of many small vesicles and complex lamellar structures. The Hexa-/-Neu3-/- mice exhibited progressive neurodegeneration with neuronal loss, Purkinje cell depletion, and astrogliosis. Slow movement, ataxia, and tremors were the prominent neurological abnormalities observed in these mice. Furthermore, radiographs revealed abnormalities in the skeletal bones of the Hexa-/-Neu3-/- mice. Thus, the Hexa-/-Neu3-/- mice mimic the neuropathological and clinical abnormalities of the classical early-onset Tay-Sachs patients, and provide a suitable model for the future pre-clinical testing of potential treatments for this condition.


Assuntos
Gangliosidoses GM2/genética , Hexosaminidase B/genética , Neuraminidase/genética , Doença de Tay-Sachs/genética , Animais , Química Encefálica/genética , Vesículas Citoplasmáticas/patologia , Gangliosidoses GM2/metabolismo , Gliose/genética , Gliose/patologia , Glicoesfingolipídeos/metabolismo , Coxeadura Animal/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuraminidase/deficiência , Neurônios/patologia , Células de Purkinje/patologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Doença de Tay-Sachs/patologia
10.
Eur J Orthod ; 39(3): 235-242, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27733487

RESUMO

Aim: The aim of this study is to evaluate and compare therapeutic effects of mesenchymal stem cell (MSCs) and osteoprotegerin (OPG) gene transfer applications on inhibition and/or repair of orthodontically induced inflammatory root resorption (OIIRR). Materials and methods: Thirty Wistar rats were divided into four groups as untreated group (negative control), treated with orthodontic appliance group (positive control), MSCs injection group, and OPG transfected MSCs [gene therapy (GT) group]. About 100g of orthodontic force was applied to upper first molar teeth of rats for 14 days. MSCs and transfected MSC injections were performed at 1st, 6th, and 11th days to the MSC and GT group rats. At the end of experiment, upper first molar teeth were prepared for genetical, scanning electron microscopy (SEM), fluorescent microscopy, and haematoxylin eosin-tartrate resistant acid phosphatase staining histological analyses. Number of total cells, number of osteoclastic cells, number of resorption lacunae, resorption area ratio, SEM resorption ratio, OPG, RANKL, Cox-2 gene expression levels at the periodontal ligament (PDL) were calculated. Paired t-test, Kruskal-Wallis, and chi-square tests were performed. Results: Transferred MSCs showed marked fluorescence in PDL. The results revealed that number of osteoclastic cells, resorption lacunae, resorption area ratio, RANKL, and Cox-2 were reduced after single MSC injections significantly (P < 0.05). GT group showed the lowest number of osteoclastic cells (P < 0.01), number of resorption lacunae, resorption area ratio, and highest OPG expression (P < 0.001). Conclusions: Taken together all these results, MSCs and GT showed marked inhibition and/or repair effects on OIIRR during orthodontic treatment on rats.


Assuntos
Terapia Genética/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Osteoprotegerina/genética , Reabsorção da Raiz/terapia , Técnicas de Movimentação Dentária/efeitos adversos , Animais , Reabsorção Óssea/etiologia , Reabsorção Óssea/patologia , Reabsorção Óssea/terapia , Técnicas de Transferência de Genes , Masculino , Microscopia Eletrônica , Dente Molar/ultraestrutura , Osteoclastos/patologia , Osteoprotegerina/metabolismo , Ligamento Periodontal/metabolismo , Ratos , Ratos Wistar , Reabsorção da Raiz/etiologia , Reabsorção da Raiz/patologia , Técnicas de Movimentação Dentária/métodos
11.
Front Mol Biosci ; 3: 68, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27826550

RESUMO

Lysosomal serine carboxypeptidase Cathepsin A (CTSA) is a multifunctional enzyme with distinct protective and catalytic function. CTSA present in the lysosomal multienzyme complex to facilitate the correct lysosomal routing, stability and activation of with beta-galactosidase and alpha-neuraminidase. Beside CTSA has role in inactivation of bioactive peptides including bradykinin, substances P, oxytocin, angiotensin I and endothelin-I by cleavage of 1 or 2 amino acid(s) from C-terminal ends. In this study, we aimed to elucidate the regulatory role of CTSA on bioactive peptides in knock-in mice model of CTSAS190A . We investigated the level of bradykinin, substances P, oxytocin, angiotensin I and endothelin-I in the kidney, liver, lung, brain and serum from CTSAS190A mouse model at 3- and 6-months of age. Our results suggest CTSA selectively contributes to processing of bioactive peptides in different tissues from CTSAS190A mice compared to age matched WT mice.

12.
PLoS Genet ; 10(2): e1004146, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24586188

RESUMO

The potent vasoconstrictor peptides, endothelin 1 (ET-1) and angiotensin II control adaptation of blood vessels to fluctuations of blood pressure. Previously we have shown that the circulating level of ET-1 is regulated through its proteolytic cleavage by secreted serine carboxypeptidase, cathepsin A (CathA). However, genetically-modified mouse expressing catalytically inactive CathA S190A mutant retained about 10-15% of the carboxypeptidase activity against ET-1 in its tissues suggesting a presence of parallel/redundant catabolic pathway(s). In the current work we provide direct evidence that the enzyme, which complements CathA action towards ET-1 is a retinoid-inducible lysosomal serine carboxypeptidase 1 (Scpep1), a CathA homolog with previously unknown biological function. We generated a mouse strain devoid of both CathA and Scpep1 activities (DD mice) and found that in response to high-salt diet and systemic injections of ET-1 these animals showed significantly increased blood pressure as compared to wild type mice or those with single deficiencies of CathA or Scpep1. We also found that the reactivity of mesenteric arteries from DD mice towards ET-1 was significantly higher than that for all other groups of mice. The DD mice had a reduced degradation rate of ET-1 in the blood whereas their cultured arterial vascular smooth muscle cells showed increased ET-1-dependent phosphorylation of myosin light chain 2. Together, our results define the biological role of mammalian serine carboxypeptidase Scpep1 and suggest that Scpep1 and CathA together participate in the control of ET-1 regulation of vascular tone and hemodynamics.


Assuntos
Carboxipeptidases/metabolismo , Catepsina A/metabolismo , Endotelina-1/metabolismo , Hipertensão/genética , Angiotensina II/genética , Angiotensina II/metabolismo , Animais , Pressão Sanguínea/genética , Carboxipeptidases/genética , Catepsina A/genética , Células Cultivadas , Endotelina-1/genética , Hemodinâmica/genética , Humanos , Hipertensão/patologia , Camundongos , Vasoconstrição/genética
13.
Diabetes ; 62(7): 2338-46, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23520133

RESUMO

Neuraminidases (sialidases) catalyze the removal of sialic acid residues from sialylated glycoconjugates. We now report that mammalian neuraminidase 1 (Neu1), in addition to its catabolic function in lysosomes, is transported to the cell surface where it is involved in the regulation of insulin signaling. Insulin binding to its receptor rapidly induces interaction of the receptor with Neu1, which hydrolyzes sialic acid residues in the glycan chains of the receptor and, consequently, induces its activation. Cells from sialidosis patients with a genetic deficiency of Neu1 show impairment of insulin-induced phosphorylation of downstream protein kinase AKT, and treatment of these cells with purified Neu1 restores signaling. Genetically modified mice with ∼10% of the normal Neu1 activity exposed to a high-fat diet develop hyperglycemia and insulin resistance twice as fast as their wild-type counterparts. Together, these studies identify Neu1 as a novel component of the signaling pathways of energy metabolism and glucose uptake.


Assuntos
Insulina/metabolismo , Mucolipidoses/metabolismo , Neuraminidase/metabolismo , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Dieta Hiperlipídica , Metabolismo Energético/fisiologia , Fibroblastos/metabolismo , Teste de Tolerância a Glucose , Células HEK293 , Humanos , Insulina/genética , Camundongos , Camundongos Knockout , Mucolipidoses/genética , Neuraminidase/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
14.
J Biol Chem ; 287(20): 16689-97, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22451654

RESUMO

Hyaluronan (HA), a member of the glycosaminoglycan (GAG) family, is a critical component of the extracellular matrix. A model for HA degradation that invokes the activity of both hyaluronidases and exoglycosidases has been advanced. However, no in vivo studies have been done to determine the extent to which these enzymes contribute to HA breakdown. Herein, we used mouse models to investigate the contributions of the endoglycosidase HYAL1 and the exoglycosidase ß-hexosaminidase to the lysosomal degradation of HA. We employed histochemistry and fluorophore-assisted carbohydrate electrophoresis to determine the degree of HA accumulation in mice deficient in one or both enzyme activities. Global HA accumulation was present in mice deficient in both enzymes, with the highest levels found in the lymph node and liver. Chondroitin, a GAG similar in structure to HA, also broadly accumulated in mice deficient in both enzymes. Accumulation of chondroitin sulfate derivatives was detected in mice deficient in both enzymes, as well as in ß-hexosaminidase-deficient mice, indicating that both enzymes play a significant role in chondroitin sulfate breakdown. Extensive accumulation of HA and chondroitin when both enzymes are lacking was not observed in mice deficient in only one of these enzymes, suggesting that HYAL1 and ß-hexosaminidase are functionally redundant in HA and chondroitin breakdown. Furthermore, accumulation of sulfated chondroitin in tissues provides in vivo evidence that both HYAL1 and ß-hexosaminidase cleave chondroitin sulfate, but it is a preferred substrate for ß-hexosaminidase. These studies provide in vivo evidence to support and extend existing knowledge of GAG breakdown.


Assuntos
Sulfatos de Condroitina/metabolismo , Ácido Hialurônico/metabolismo , Hialuronoglucosaminidase/metabolismo , Lisossomos/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Animais , Sulfatos de Condroitina/genética , Ácido Hialurônico/genética , Hialuronoglucosaminidase/genética , Fígado/metabolismo , Linfonodos/metabolismo , Lisossomos/genética , Camundongos , Camundongos Knockout , beta-N-Acetil-Hexosaminidases/genética
15.
PLoS Genet ; 6(9): e1001118, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20862357

RESUMO

Tay-Sachs disease is a severe lysosomal disorder caused by mutations in the HexA gene coding for the α-subunit of lysosomal ß-hexosaminidase A, which converts G(M2) to G(M3) ganglioside. Hexa(-/-) mice, depleted of ß-hexosaminidase A, remain asymptomatic to 1 year of age, because they catabolise G(M2) ganglioside via a lysosomal sialidase into glycolipid G(A2), which is further processed by ß-hexosaminidase B to lactosyl-ceramide, thereby bypassing the ß-hexosaminidase A defect. Since this bypass is not effective in humans, infantile Tay-Sachs disease is fatal in the first years of life. Previously, we identified a novel ganglioside metabolizing sialidase, Neu4, abundantly expressed in mouse brain neurons. Now we demonstrate that mice with targeted disruption of both Neu4 and Hexa genes (Neu4(-/-);Hexa(-/-)) show epileptic seizures with 40% penetrance correlating with polyspike discharges on the cortical electrodes of the electroencephalogram. Single knockout Hexa(-/-) or Neu4(-/-) siblings do not show such symptoms. Further, double-knockout but not single-knockout mice have multiple degenerating neurons in the cortex and hippocampus and multiple layers of cortical neurons accumulating G(M2) ganglioside. Together, our data suggest that the Neu4 block exacerbates the disease in Hexa(-/-) mice, indicating that Neu4 is a modifier gene in the mouse model of Tay-Sachs disease, reducing the disease severity through the metabolic bypass. However, while disease severity in the double mutant is increased, it is not profound suggesting that Neu4 is not the only sialidase contributing to the metabolic bypass in Hexa(-/-) mice.


Assuntos
Epilepsia/enzimologia , Epilepsia/patologia , Lisossomos/enzimologia , Neuraminidase/deficiência , Neurônios/enzimologia , Neurônios/patologia , Cadeia alfa da beta-Hexosaminidase/metabolismo , Animais , Comportamento Animal , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Córtex Cerebral/ultraestrutura , Eletroencefalografia , Epilepsia/fisiopatologia , Gangliosídeo G(M2)/metabolismo , Técnicas de Inativação de Genes , Hipocampo/enzimologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Hipocampo/ultraestrutura , Aprendizagem/fisiologia , Lisossomos/patologia , Lisossomos/ultraestrutura , Camundongos , Atividade Motora/fisiologia , Neuraminidase/metabolismo , Neurônios/ultraestrutura
16.
Cell Signal ; 22(2): 314-24, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19796680

RESUMO

The ectodomain of TOLL-like receptors (TLR) is highly glycosylated with several N-linked gylcosylation sites located in the inner concave surface. The precise role of these sugar N-glycans in TLR receptor activation is unknown. Recently, we have shown that Neu1 sialidase and not Neu2, -3 and -4 forms a complex with TLR-2, -3 and -4 receptors on the cell-surface membrane of naïve and activated macrophage cells (Glycoconj J DOI 10.1007/s10719-009-9239-8). Activation of Neu1 is induced by TLR ligands binding to their respective receptors. Here, we show that endotoxin lipopolysaccharide (LPS)-induced MyD88/TLR4 complex formation and subsequent NFkappaB activation is dependent on the removal of alpha-2,3-sialyl residue linked to beta-galactoside of TLR4 by the Neu1 activity associated with LPS-stimulated live primary macrophage cells, macrophage and dendritic cell lines but not with primary Neu1-deficient macrophage cells. Exogenous alpha-2,3 sialyl specific neuraminidase (Streptoccocus pneumoniae) and wild-type T. cruzi trans-sialidase (TS) but not the catalytically inactive mutant TSAsp98-Glu mediate TLR4 dimerization to facilitate MyD88/TLR4 complex formation and NFkappaB activation similar to those responses seen with LPS. These same TLR ligand-induced NFkappaB responses are not observed in TLR deficient HEK293 cells, but are re-established in HEK293 cells stably transfected with TLR4/MD2, and are significantly inhibited by alpha-2,3-sialyl specific Maackia amurensis (MAL-2) lectin, alpha-2,3-sialyl specific galectin-1 and neuraminidase inhibitor Tamiflu but not by alpha-2,6-sialyl specific Sambucus nigra lectin (SNA). Taken together, the findings suggest that Neu1 desialylation of alpha-2,3-sialyl residues of TLR receptors enables in removing a steric hinderance to receptor association for TLR activation and cellular signaling.


Assuntos
Neuraminidase/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Glicoproteínas/farmacologia , Glicosilação , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , NF-kappa B/metabolismo , Neuraminidase/farmacologia , Oseltamivir/farmacologia , Fosforilação , Receptor 4 Toll-Like/química
17.
J Biol Chem ; 285(1): 206-15, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19889639

RESUMO

The differentiation of monocytes into macrophages and dendritic cells is accompanied by induction of cell-surface neuraminidase 1 (Neu1) and cathepsin A (CathA), the latter forming a complex with and activating Neu1. To clarify the biological importance of this phenomenon we have developed the gene-targeted mouse models of a CathA deficiency (CathA(S190A)) and a double CathA/Neu1 deficiency (CathA(S190A-Neo)). Macrophages of CathA(S190A-Neo) mice and their immature dendritic cells showed a significantly reduced capacity to engulf Gram-positive and Gram-negative bacteria and positively and negatively charged polymer beads as well as IgG-opsonized beads and erythrocytes. Properties of the cells derived from CathA(S190A) mice were indistinguishable from those of wild-type controls, suggesting that the absence of Neu1, which results in the increased sialylation of the cell surface proteins, probably affects multiple receptors for phagocytosis. Indeed, treatment of the cells with purified mouse Neu1 reduced surface sialylation and restored phagocytosis. Because Neu1-deficient cells showed reduced internalization of IgG-opsonized sheep erythrocytes whereas binding of the erythrocytes to the cells at 4 degrees C persisted, we speculate that the absence of Neu1 in particular affected transduction of signals from the Fc receptors for immunoglobulin G (FcgammaR). Indeed the macrophages from the Neu1-deficient mice showed increased sialylation and impaired phosphorylation of FcgammaR as well as markedly reduced phosphorylation of Syk kinase in response to treatment with IgG-opsonized beads. Altogether our data suggest that the cell surface Neu1 activates the phagocytosis in macrophages and dendritic cells through desialylation of surface receptors, thus, contributing to their functional integrity.


Assuntos
Macrófagos/citologia , Macrófagos/enzimologia , Neuraminidase/metabolismo , Fagocitose , Animais , Catepsina A/metabolismo , Diferenciação Celular , Membrana Celular/enzimologia , Células Dendríticas/citologia , Eritrócitos/citologia , Eritrócitos/metabolismo , Lectinas/metabolismo , Macrófagos/microbiologia , Camundongos , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/deficiência , Proteínas Opsonizantes/imunologia , Receptores de IgG/imunologia , Ovinos , Transdução de Sinais , Coloração e Rotulagem
18.
Glycoconj J ; 26(9): 1197-212, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19430901

RESUMO

The signaling pathways of mammalian Toll-like receptors (TLR) are well characterized, but the initial molecular mechanisms activated following ligand interactions with the receptors remain poorly defined. Here, we show a membrane controlling mechanism that is initiated by ligand binding to TLR-2, -3 and-4 to induce Neu1 sialidase activity within minutes in live primary bone marrow (BM) macrophage cells and macrophage and dendritic cell lines. Central to this process is that Neu1 and not Neu2,-3 and-4 forms a complex with TLR-2,-3 and-4 on the cell surface of naïve macrophage cells. Neuraminidase inhibitors BCX1827, 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (DANA), zanamivir and oseltamivir carboxylate have a limited significant inhibition of the LPS-induced sialidase activity in live BMC-2 macrophage cells but Tamiflu (oseltamivir phosphate) completely blocks this activity. Tamiflu inhibits LPS-induced sialidase activity in live BMC-2 cells with an IC(50) of 1.2 microM compared to an IC(50) of 1015 microM for its hydrolytic metabolite oseltamivir carboxylate. Tamiflu blockage of LPS-induced Neu1 sialidase activity is not affected in BMC-2 cells pretreated with anticarboxylesterase agent clopidogrel. Endotoxin LPS binding to TLR4 induces Neu1 with subsequent activation of NFkappaB and the production of nitric oxide and pro-inflammatory IL-6 and TNFalpha cytokines in primary and macrophage cell lines. Hypomorphic cathepsin A mice with a secondary Neu1 deficiency respond poorly to LPS-induced pro-inflammatory cytokines compared to the wild-type or hypomorphic cathepsin A with normal Neu1 mice. Our findings establish an unprecedented mechanism for pathogen molecule-induced TLR activation and cell function, which is critically dependent on Neu1 sialidase activity associated with TLR ligand treated live primary macrophage cells and macrophage and dendritic cell lines.


Assuntos
Células Dendríticas/enzimologia , Macrófagos/enzimologia , Neuraminidase/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Receptores Toll-Like/imunologia , Animais , Células da Medula Óssea/citologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/sangue , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Imunoprecipitação , Mediadores da Inflamação/metabolismo , Ligantes , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Camundongos , Neuraminidase/antagonistas & inibidores , Neuraminidase/deficiência , Óxido Nítrico/biossíntese , Oseltamivir/farmacologia
19.
Circulation ; 117(15): 1973-81, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18391110

RESUMO

BACKGROUND: Lysosomal carboxypeptidase, cathepsin A (protective protein, CathA), is a component of the lysosomal multienzyme complex along with beta-galactosidase (GAL) and sialidase Neu1, where it activates Neu1 and protects GAL and Neu1 against the rapid proteolytic degradation. On the cell surface, CathA, Neu1, and the enzymatically inactive splice variant of GAL form the elastin-binding protein complex. In humans, genetic defects of CathA cause galactosialidosis, a metabolic disease characterized by combined deficiency of CathA, GAL, and Neu1 and a lysosomal storage of sialylated glycoconjugates. However, several phenotypic features of galactosialidosis patients, including hypertension and cardiomyopathies, cannot be explained by the lysosomal storage. These observations suggest that CathA may be involved in hemodynamic functions that go beyond its protective activity in the lysosome. METHODS AND RESULTS: We generated a gene-targeted mouse in which the active CathA was replaced with a mutant enzyme carrying a Ser190Ala substitution in the active site. These animals expressed physiological amounts of catalytically inactive CathA protein, capable of forming lysosomal multienzyme complex, and did not develop secondary deficiency of Neu1 and GAL. Conversely, the mice showed a reduced degradation rate of the vasoconstrictor peptide, endothelin-1, and significantly increased arterial blood pressure. CathA-deficient mice also displayed scarcity of elastic fibers in lungs, aortic adventitia, and skin. CONCLUSIONS: Our results provide the first evidence that CathA acts in vivo as an endothelin-1-inactivating enzyme and strongly confirm a crucial role of this enzyme in effective elastic fiber formation.


Assuntos
Catepsina A/fisiologia , Tecido Elástico/metabolismo , Endotelina-1/antagonistas & inibidores , Hipertensão/genética , Animais , Pressão Sanguínea/efeitos dos fármacos , Catepsina A/genética , Células Cultivadas/enzimologia , Células Cultivadas/ultraestrutura , Tecido Elástico/ultraestrutura , Elastina/metabolismo , Endotelina-1/farmacologia , Endotelina-1/fisiologia , Ativação Enzimática , Fibroblastos/enzimologia , Fibroblastos/ultraestrutura , Genes Sintéticos , Hipertensão/enzimologia , Hipertensão/patologia , Lisossomos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multienzimáticos , Neuraminidase/metabolismo , Neurônios/enzimologia , Neurônios/ultraestrutura , Especificidade de Órgãos , RNA Mensageiro/biossíntese , Cloreto de Sódio na Dieta/efeitos adversos , beta-Galactosidase/metabolismo
20.
Hum Mol Genet ; 17(11): 1556-68, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18270209

RESUMO

Mammalian sialidase Neu4, ubiquitously expressed in human tissues, is located in the lysosomal and mitochondrial lumen and has broad substrate specificity against sialylated glycoconjugates. To investigate whether Neu4 is involved in ganglioside catabolism, we transfected beta-hexosaminidase-deficient neuroglia cells from a Tay-Sachs patient with a Neu4-expressing plasmid and demonstrated the correction of storage due to the clearance of accumulated GM2 ganglioside. To further clarify the biological role of Neu4, we have generated a stable loss-of-function phenotype in cultured HeLa cells and in mice with targeted disruption of the Neu4 gene. The silenced HeLa cells showed reduced activity against gangliosides and had large heterogeneous lysosomes containing lamellar structures. Neu4(-/-) mice were viable, fertile and lacked gross morphological abnormalities, but showed a marked vacuolization and lysosomal storage in lung and spleen cells. Lysosomal storage bodies were also present in cultured macrophages preloaded with gangliosides. Thin-layer chromatography showed increased relative level of GD1a ganglioside and a markedly decreased level of GM1 ganglioside in brain of Neu4(-/-) mice suggesting that Neu4 may be important for desialylation of brain gangliosides and consistent with the in situ hybridization data. Increased levels of cholesterol, ceramide and polyunsaturated fatty acids were also detected in the lungs and spleen of Neu4(-/-) mice by high-resolution NMR spectroscopy. Together, our data suggest that Neu4 is a functional component of the ganglioside-metabolizing system, contributing to the postnatal development of the brain and other vital organs.


Assuntos
Gangliosídeos/metabolismo , Lisossomos/metabolismo , Neuraminidase/genética , Neuraminidase/fisiologia , Animais , Comportamento Animal , Encéfalo/enzimologia , Encéfalo/fisiologia , Encéfalo/ultraestrutura , Catálise , Gangliosídeo G(M1)/análise , Gangliosídeo G(M1)/metabolismo , Gangliosídeo G(M2)/análise , Gangliosídeo G(M2)/metabolismo , Gangliosídeos/análise , Células HeLa , Humanos , Pulmão/enzimologia , Pulmão/ultraestrutura , Camundongos , Camundongos Knockout , Neuraminidase/metabolismo , Interferência de RNA , Baço/enzimologia , Baço/ultraestrutura , Distribuição Tecidual , beta-N-Acetil-Hexosaminidases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...