Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Am Chem Soc ; 144(2): 832-844, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-34985906

RESUMEN

Owing to its roles in human health and disease, the modification of nuclear, cytoplasmic, and mitochondrial proteins with O-linked N-acetylglucosamine residues (O-GlcNAc) has emerged as a topic of great interest. Despite the presence of O-GlcNAc on hundreds of proteins within cells, only two enzymes regulate this modification. One of these enzymes is O-GlcNAcase (OGA), a dimeric glycoside hydrolase that has a deep active site cleft in which diverse substrates are accommodated. Chemical tools to control OGA are emerging as essential resources for helping to decode the biochemical and cellular functions of the O-GlcNAc pathway. Here we describe rationally designed bicyclic thiazolidine inhibitors that exhibit superb selectivity and picomolar inhibition of human OGA. Structures of these inhibitors in complex with human OGA reveal the basis for their exceptional potency and show that they extend out of the enzyme active site cleft. Leveraging this structure, we create a high affinity chemoproteomic probe that enables simple one-step purification of endogenous OGA from brain and targeted proteomic mapping of its post-translational modifications. These data uncover a range of new modifications, including some that are less-known, such as O-ubiquitination and N-formylation. We expect that these inhibitors and chemoproteomics probes will prove useful as fundamental tools to decipher the mechanisms by which OGA is regulated and directed to its diverse cellular substrates. Moreover, the inhibitors and structures described here lay out a blueprint that will enable the creation of chemical probes and tools to interrogate OGA and other carbohydrate active enzymes.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Compuestos Bicíclicos con Puentes/química , Inhibidores Enzimáticos/química , Histona Acetiltransferasas/metabolismo , Hialuronoglucosaminidasa/metabolismo , Secuencia de Aminoácidos , Encéfalo/metabolismo , Compuestos Bicíclicos con Puentes/metabolismo , Dominio Catalítico , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos/metabolismo , Histona Acetiltransferasas/antagonistas & inhibidores , Humanos , Hialuronoglucosaminidasa/antagonistas & inhibidores , Espectrometría de Masas , Péptidos/análisis , Péptidos/química , Procesamiento Proteico-Postraduccional , Proteómica/métodos , Relación Estructura-Actividad , Tiazolidinas/química , Tiazolidinas/metabolismo , Cadena alfa de beta-Hexosaminidasa/antagonistas & inhibidores , Cadena alfa de beta-Hexosaminidasa/metabolismo
3.
Proteins ; 89(11): 1587-1601, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34288098

RESUMEN

ß-hexosaminidase A (HexA) protein is responsible for the degradation of GM2 gangliosides in the central and peripheral nervous systems. Tay-Sachs disease occurs when HexA within Hexosaminidase does not properly function and harmful GM2 gangliosides begin to build up within the neurons. In this study, in silico methods such as SIFT, PolyPhen-2, PhD-SNP, and MutPred were utilized to analyze the effects of nonsynonymous single nucleotide polymorphisms (nsSNPs) on HexA in order to identify possible pathogenetic and deleterious variants. Molecular dynamics (MD) simulations showed that two mutants, P25S and W485R, experienced an increase in structural flexibility compared to the native protein. Particularly, there was a decrease in the overall number and frequencies of hydrogen bonds for the mutants compared to the wildtype. MM/GBSA calculations were performed to help assess the change in binding affinity between the wildtype and mutant structures and a mechanism-based inhibitor, NGT, which is known to help increase the residual activity of HexA. Both of the mutants experienced a decrease in the binding affinity from -23.8 kcal/mol in wildtype to -20.9 and -18.7 kcal/mol for the P25S and W485R variants of HexA, respectively.


Asunto(s)
Gangliósido G(M2)/química , Simulación de Dinámica Molecular , Mutación Puntual , Polimorfismo de Nucleótido Simple , Enfermedad de Tay-Sachs/genética , Cadena alfa de beta-Hexosaminidasa/química , Acetilglucosamina/análogos & derivados , Acetilglucosamina/química , Acetilglucosamina/farmacología , Sitios de Unión , Sistema Nervioso Central/enzimología , Sistema Nervioso Central/patología , Gangliósido G(M2)/metabolismo , Expresión Génica , Humanos , Enlace de Hidrógeno , Neuronas/enzimología , Neuronas/patología , Sistema Nervioso Periférico/enzimología , Sistema Nervioso Periférico/patología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Enfermedad de Tay-Sachs/enzimología , Enfermedad de Tay-Sachs/patología , Termodinámica , Tiazoles/química , Tiazoles/farmacología , Cadena alfa de beta-Hexosaminidasa/genética , Cadena alfa de beta-Hexosaminidasa/metabolismo
4.
Neurobiol Dis ; 134: 104667, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31682993

RESUMEN

The favorable outcome of in vivo and ex vivo gene therapy approaches in several Lysosomal Storage Diseases suggests that these treatment strategies might equally benefit GM2 gangliosidosis. Tay-Sachs and Sandhoff disease (the main forms of GM2 gangliosidosis) result from mutations in either the HEXA or HEXB genes encoding, respectively, the α- or ß-subunits of the lysosomal ß-Hexosaminidase enzyme. In physiological conditions, α- and ß-subunits combine to generate ß-Hexosaminidase A (HexA, αß) and ß-Hexosaminidase B (HexB, ßß). A major impairment to establishing in vivo or ex vivo gene therapy for GM2 gangliosidosis is the need to synthesize the α- and ß-subunits at high levels and with the correct stoichiometric ratio, and to safely deliver the therapeutic products to all affected tissues/organs. Here, we report the generation and in vitro validation of novel bicistronic lentiviral vectors (LVs) encoding for both the murine and human codon optimized Hexa and Hexb genes. We show that these LVs drive the safe and coordinate expression of the α- and ß-subunits, leading to supranormal levels of ß-Hexosaminidase activity with prevalent formation of a functional HexA in SD murine neurons and glia, murine bone marrow-derived hematopoietic stem/progenitor cells (HSPCs), and human SD fibroblasts. The restoration/overexpression of ß-Hexosaminidase leads to the reduction of intracellular GM2 ganglioside storage in transduced and in cross-corrected SD murine neural progeny, indicating that the transgenic enzyme is secreted and functional. Importantly, bicistronic LVs safely and efficiently transduce human neurons/glia and CD34+ HSPCs, which are target and effector cells, respectively, in prospective in vivo and ex vivo GT approaches. We anticipate that these bicistronic LVs may overcome the current requirement of two vectors co-delivering the α- or ß-subunits genes. Careful assessment of the safety and therapeutic potential of these bicistronic LVs in the SD murine model will pave the way to the clinical development of LV-based gene therapy for GM2 gangliosidosis.


Asunto(s)
Gangliosidosis GM2/metabolismo , Terapia Genética/métodos , Células Madre Hematopoyéticas/metabolismo , Células-Madre Neurales/metabolismo , Cadena alfa de beta-Hexosaminidasa/metabolismo , Cadena beta de beta-Hexosaminidasa/metabolismo , Animales , Gangliosidosis GM2/genética , Vectores Genéticos , Humanos , Lentivirus , Ratones , Cadena alfa de beta-Hexosaminidasa/genética , Cadena beta de beta-Hexosaminidasa/genética
5.
Methods Mol Biol ; 1885: 233-250, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30506202

RESUMEN

Tay-Sachs disease (TSD) is an autosomal recessive lysosomal storage disorder caused by mutations of the HEXA gene resulting in the deficiency of hexosaminidase A (Hex A) and subsequent neuronal accumulation of GM2 gangliosides. Infantile TSD is a devastating and fetal neurodegenerative disease with death before the age of 3-5 years. A small proportion of TSD patients carry milder mutations and may present juvenile or adult onset milder disease. TSD is more prevalent among Ashkenazi Jewish (AJ) individuals and some other genetically isolated populations with carrier frequencies of approximately ~1:27 which is much higher than that of 1:300 in the general population. Carrier screening and prenatal testing for TSD are effective in preventing the birth of affected fetuses greatly diminishing the incidence of TSD. Testing of targeted HEXA mutations by genotyping or sequencing can detect 98% of carriers in AJ individuals; however, the detection rate is much lower for most other ethnic groups. When combined with enzyme analysis, above 98% of carriers can be reliably identified regardless of ethnic background. Multiplex PCR followed by allele-specific primer extension is one method to test for known and common mutations. Sanger sequencing or other sequencing methods are useful to identify private mutations. For prenatal testing, only predefined parental mutations need to be tested. In the event of unknown mutational status or the presence of variants of unknown significance (VUS), enzyme analysis must be performed in conjunction with DNA-based assays to enhance the diagnostic accuracy. Enzymatic assays involve the use of synthetic substrates 4-methylumbelliferyl-N-acetyl-ß-glucosamine (4-MUG) and 4-methylumbelliferyl-6-sulfo-2-acetamido-2-deoxy-ß-D-glucopyranoside (4-MUGS) to measure the percentage Hex A activity (Hex A%) and specific Hex A activity respectively. These biochemical and molecular tests can be performed in both direct specimens and cultured cells from chorionic villi sampling or amniocentesis.


Asunto(s)
Pruebas Genéticas , Diagnóstico Prenatal/métodos , Enfermedad de Tay-Sachs/diagnóstico , Enfermedad de Tay-Sachs/genética , Alelos , Contaminación de ADN , Análisis Mutacional de ADN , Electroforesis Capilar , Pruebas Genéticas/métodos , Pruebas Genéticas/normas , Genotipo , Humanos , Mutación , Reacción en Cadena de la Polimerasa , Diagnóstico Prenatal/normas , Enfermedad de Tay-Sachs/metabolismo , Cadena alfa de beta-Hexosaminidasa/genética , Cadena alfa de beta-Hexosaminidasa/metabolismo
6.
BMC Med Genet ; 19(1): 162, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30208878

RESUMEN

BACKGROUND: Mucolipidosis alpha/beta is an inborn error of metabolism characterized by deficiency of GlcNAc-1-phosphotransferase, in which essential alpha/beta subunits are encoded by the GNPTAB gene. The autosomal recessive condition is due to disruptions of hydrolase mannose 6-phosphate marker generation, defective lysosomal targeting and subsequent intracellular accumulation of non-degraded material. Clinical severity depends on residual GlcNAc-1-phosphotransferase activity, which distinguishes between the milder type III disease and the severe, neonatal onset type II disease. CASE PRESENTATION: We report the clinical, biochemical and genetic diagnosis of mucolipidosis III alpha/beta in a two-year-old Chinese boy who initially presented with poor weight gain, microcephaly and increased tone. He was confirmed to harbor the common splice site mutation c.2715 + 1G > A and the nonsense variant c.2404C > T (p.Q802*). Clinically, the patient had multiple phenotypic features typical of mucopolysaccharidosis including joint contractures, coarse facial features, kypho-lordosis, pectus carinatum and umbilical hernia. However, the relatively mild developmental delay compared to severe type I and type II mucopolysaccharidosis and the absence of macrocephaly raised the possibility of the less commonly diagnosed mucolipidosis alpha/beta. Critical roles of lysosomal enzyme activity assay, which showed elevated α-iduronidase, iduronate sulfatase, galactose-6-sulphate sulphatase, arylsulfatase B and α-hexosaminidase activities; and genetic study, which confirmed the parental origin of both mutations, were highlighted. CONCLUSIONS: The recently reported nonsense variant c.2404C > T in the GNPTAB gene is further recognized and this contributes to the genotype-phenotype spectrum of mucolipidosis alpha/beta.


Asunto(s)
Codón sin Sentido , Mucolipidosis/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Preescolar , Condroitinsulfatasas/genética , Condroitinsulfatasas/metabolismo , Regulación de la Expresión Génica , Genes Recesivos , Humanos , Iduronato Sulfatasa/genética , Iduronato Sulfatasa/metabolismo , Iduronidasa/genética , Iduronidasa/metabolismo , Lisosomas/enzimología , Lisosomas/patología , Masculino , Mucolipidosis/diagnóstico , Mucolipidosis/enzimología , Mucolipidosis/patología , N-Acetilgalactosamina-4-Sulfatasa/genética , N-Acetilgalactosamina-4-Sulfatasa/metabolismo , Linaje , Transferasas (Grupos de Otros Fosfatos Sustitutos)/deficiencia , Cadena alfa de beta-Hexosaminidasa/genética , Cadena alfa de beta-Hexosaminidasa/metabolismo
7.
Genet Test Mol Biomarkers ; 20(9): 504-9, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27362553

RESUMEN

BACKGROUND AND AIMS: Carrier screening for Tay-Sachs disease is performed by sequence analysis of the HEXA gene and/or hexosaminidase A enzymatic activity testing. Enzymatic analysis (EA) has been suggested as the optimal carrier screening method, especially in non-Ashkenazi Jewish (non-AJ) individuals, but its utilization and efficacy have not been fully evaluated in the general population. This study assesses the reliability of EA in comparison with HEXA sequence analysis in non-AJ populations. METHODS: Five hundred eight Hispanic and African American patients (516 samples) had EA of their leukocytes performed and 12 of these patients who tested positive by EA ("carriers") had subsequent HEXA gene sequencing performed. RESULTS: Of the 508 patients, 25 (4.9%) were EA positive and 40 (7.9%) were inconclusive. Of the 12 patients who were sequenced, 11 did not carry a pathogenic variant and one carried a likely deleterious mutation (NM_000520.4(HEXA):c.1510C>T). CONCLUSIONS: High inconclusive rates and poor correlation between positive/inconclusive enzyme results and identification of pathogenic mutations suggest that ethnic-specific recalibration of reference ranges for EA may be necessary. Alternatively, HEXA gene sequencing could be performed.


Asunto(s)
Pruebas de Enzimas/métodos , Tamización de Portadores Genéticos/métodos , Enfermedad de Tay-Sachs/enzimología , Enfermedad de Tay-Sachs/genética , Cadena alfa de beta-Hexosaminidasa/genética , Cadena alfa de beta-Hexosaminidasa/metabolismo , Negro o Afroamericano/genética , Etnicidad/genética , Pruebas Genéticas/métodos , Heterocigoto , Hispánicos o Latinos/genética , Humanos , Judíos/genética , Mutación , Ciudad de Nueva York/epidemiología , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Enfermedad de Tay-Sachs/diagnóstico , Enfermedad de Tay-Sachs/epidemiología
8.
J Lipid Res ; 56(9): 1747-61, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26175473

RESUMEN

Ganglioside GM2 is the major lysosomal storage compound of Tay-Sachs disease. It also accumulates in Niemann-Pick disease types A and B with primary storage of SM and with cholesterol in type C. Reconstitution of GM2 catabolism with ß-hexosaminidase A and GM2 activator protein (GM2AP) at uncharged liposomal surfaces carrying GM2 as substrate generated only a physiologically irrelevant catabolic rate, even at pH 4.2. However, incorporation of anionic phospholipids into the GM2 carrying liposomes stimulated GM2 hydrolysis more than 10-fold, while the incorporation of plasma membrane stabilizing lipids (SM and cholesterol) generated a strong inhibition of GM2 hydrolysis, even in the presence of anionic phospholipids. Mobilization of membrane lipids by GM2AP was also inhibited in the presence of cholesterol or SM, as revealed by surface plasmon resonance studies. These lipids also reduced the interliposomal transfer rate of 2-NBD-GM1 by GM2AP, as observed in assays using Förster resonance energy transfer. Our data raise major concerns about the usage of recombinant His-tagged GM2AP compared with untagged protein. The former binds more strongly to anionic GM2-carrying liposomal surfaces, increases GM2 hydrolysis, and accelerates intermembrane transfer of 2-NBD-GM1, but does not mobilize membrane lipids.


Asunto(s)
Proteína Activadora de G (M2)/metabolismo , Gangliósido G(M2)/metabolismo , Liposomas/metabolismo , Lípidos de la Membrana/metabolismo , Ceramidas/metabolismo , Colesterol/genética , Colesterol/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Proteína Activadora de G (M2)/genética , Células HEK293 , Humanos , Hidrólisis/efectos de los fármacos , Lisofosfolípidos/administración & dosificación , Lípidos de la Membrana/genética , Monoglicéridos/administración & dosificación , Enfermedades de Niemann-Pick/genética , Enfermedades de Niemann-Pick/metabolismo , Enfermedades de Niemann-Pick/patología , Esfingomielinas/metabolismo , Resonancia por Plasmón de Superficie , Enfermedad de Tay-Sachs/genética , Enfermedad de Tay-Sachs/metabolismo , Enfermedad de Tay-Sachs/patología , Cadena alfa de beta-Hexosaminidasa/metabolismo
9.
Biochim Biophys Acta ; 1849(8): 1104-15, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25934542

RESUMEN

Nonsense-mediated mRNA decay (NMD), an mRNA surveillance mechanism, eliminates premature termination codon-containing (PTC⁺) transcripts. For instance, it maintains the homeostasis of splicing factors and degrades aberrant transcripts of human genetic disease genes. Here we examine the inhibitory effect on the NMD pathway and consequent increase of PTC+ transcripts by the dietary compound curcumin. We have found that several PTC⁺ transcripts including that of serine/arginine-rich splicing factor 1 (SRSF1) were specifically increased in cells by curcumin. We also observed a similar curcumin effect on the PTC⁺ mutant transcript from a Tay-Sachs-causing HEXA allele or from a beta-globin reporter gene. The curcumin effect was accompanied by significantly reduced expression of the NMD factors UPF1, 2, 3A and 3B. Consistently, in chromatin immunoprecipitation assays, curcumin specifically reduced the occupancy of acetyl-histone H3 and RNA polymerase II at the promoter region (-376 to -247nt) of human UPF1, in a time- and dosage-dependent way. Importantly, knocking down UPF1 abolished or substantially reduced the difference of PTC(+) transcript levels between control and curcumin-treated cells. The disrupted curcumin effect was efficiently rescued by expression of exogenous Myc-UPF1 in the knockdown cells. Together, our data demonstrate that a group of PTC⁺ transcripts are stabilized by a dietary compound curcumin through the inhibition of UPF factor expression and the NMD pathway.


Asunto(s)
Codón sin Sentido/genética , Curcumina/farmacología , Degradación de ARNm Mediada por Codón sin Sentido/efectos de los fármacos , ARN Mensajero/metabolismo , Terminación de la Transcripción Genética/efectos de los fármacos , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Proteínas Nucleares/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Factores de Empalme Serina-Arginina , Enfermedad de Tay-Sachs/genética , Enfermedad de Tay-Sachs/metabolismo , Cadena alfa de beta-Hexosaminidasa/genética , Cadena alfa de beta-Hexosaminidasa/metabolismo
10.
Biochimie ; 101: 75-82, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24389457

RESUMEN

ß-Hexosaminidase, involved in degradation of glycoproteins and glycosphingolipids, is altered in several tumours leading to enhanced migration capacity. To date, the expression of the ß-hexosaminidase isoenzymes in prostate cancer cells has not been elucidated. By using PC3, LNCaP, DUCaP, MDAPCa 2b, and hyperplasic prostate (BPH-1) cell lines, we analysed the ß-hexosaminidase activity in each cell line and determined ß-hexosaminidase α subunit gene expression in PC3, LNCaP, and BPH-1. We then investigated the methylation status of the gene promoter and determined the cellular responses of PC3 and LNCaP after transfection with ß-hexosaminidase α subunit. We found that each prostate cancer cell line had a decrease in total hexosaminidase activity and that the lack of hexosaminidase A activity, observed in PC3 and LNCaP cells, was associated with mRNA disappearance. The HEXA promoter region in LNCaP and PC3 cell lines had methylated CpG islands, as confirmed by 5'-Aza-2'-deoxycitidine treatment, in PC3 cells, used as cell cancer model. We also tested, the involvement of hexosaminidase A in the migration capacity by migration assay using Hex α subunit-transfected PC3. Finally, we found that, after Hex α subunit transfection, both PC3 and LNCaP were less susceptible to exogenous ceramide treatment. Results indicate a likely contribution of the lysosomal enzyme to the acquisition of cancerous features.


Asunto(s)
Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Cadena alfa de beta-Hexosaminidasa/genética , Línea Celular Tumoral , Regulación hacia Abajo , Represión Enzimática , Epigénesis Genética , Humanos , Masculino , Regiones Promotoras Genéticas , Neoplasias de la Próstata , Esfingosina/análogos & derivados , Esfingosina/farmacología , Cadena alfa de beta-Hexosaminidasa/metabolismo
11.
J Comp Pathol ; 148(2-3): 243-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22878054

RESUMEN

A 1.5-year-old neutered male rabbit was presented with chronic nasal discharge and ataxia. Rapid progression of neurological signs was noted subsequent to general anaesthesia and the rabbit was humanely destroyed due to the poor prognosis. At necropsy examination there were no gross changes affecting the brain or spinal cord. Microscopical examination revealed that the perikarya of numerous neurons in the brain and spinal cord were distended by the intracytoplasmic accumulation of pale, finely granular to vacuolar material. Transmission electron microscopy showed this to be composed of concentric membranous cytoplasmic bodies. Thin layer chromatography revealed elevation of GM2 ganglioside in the brain of this rabbit compared with that of an unaffected control rabbit. Enzymatically, there was markedly reduced activity of tissue ß-hexosaminidase A in brain and liver tissue from the rabbit. This was a result of an almost complete absence of the enzymatic activity of the α-subunit of that enzyme. These findings are consistent with sphingolipidosis comparable with human GM2 gangliosidosis variant B1.


Asunto(s)
Gangliosidosis GM2/metabolismo , Gangliosidosis GM2/veterinaria , Neuronas/metabolismo , Conejos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Gangliosidosis GM2/diagnóstico , Gangliosidosis GM2/patología , Cuerpos de Inclusión/ultraestructura , Masculino , Neuronas/patología , Vacuolas/ultraestructura , Cadena alfa de beta-Hexosaminidasa/metabolismo
12.
Mol Cell Biochem ; 363(1-2): 109-18, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22147196

RESUMEN

Lysosomes are not only degrading organelles but also involved in other critical cellular processes. In addition, active lysosomal glycohydrolases have been detected in an extra-lysosomal compartment: the presence of glycohydrolases on the plasma membrane (PM) has been widely demonstrated, and a possible role on the modification of the cell surface glycosphingolipids (GSL) participating in the modulation of cell functions such as cell-to-cell interactions and signal transduction pathways has been proposed. On this basis, the coordinated expression of lysosomal glycohydrolases and their translocation to the PM appear to be crucial for many cellular events. In this paper, we report evidence for the existence of a coordinated mechanism regulating the expression/activity of both lysosomal and PM-associated glycohydrolases. We show that the over-expression of the acidic glycohydrolase ß-hexosaminidase α-subunit in mouse NIH/3T3 fibroblasts induces the increased expression of the Hex ß-subunit necessary to form the active isoenzyme dimers as well as of other glycohydrolases participating in the GSL catabolism, such as ß-galactosidase and ß-glucocerebrosidase. More interestingly, this regulatory effect was also extended to the PM-associated hydrolases. In addition, transfected cells displayed a rearrangement of the GSL expression pattern that cannot be simply explained by the increased activity of a single enzyme. These observations clearly indicate that the expression level of metabolically related glycohydrolases is regulated in a coordinated manner and this regulation mechanism also involves the PM-associated isoforms.


Asunto(s)
Membrana Celular/enzimología , Fibroblastos/enzimología , Glicósido Hidrolasas/metabolismo , Glicoesfingolípidos/metabolismo , Lisosomas/enzimología , Cadena alfa de beta-Hexosaminidasa/metabolismo , Animales , Exocitosis , Humanos , Ratones , Células 3T3 NIH , Transfección , Cadena alfa de beta-Hexosaminidasa/genética
13.
Cell Biochem Funct ; 30(1): 61-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21997228

RESUMEN

In an attempt to investigate whether the genetic defect in the HEXA and HEXB genes (which causes the absence of the lysosomal ß-N-acetyl-hexosaminidase), are related to the wide inflammation in GM2 gangliosidoses (Tay-Sachs and Sandhoff disease), we have chosen the dendritic cells (DCs) as a study model. Using the RNA interference approach, we generated an in vitro model of HEXs knock-down immunogenic DCs (i-DCs) from CD34(+)-haemopoietic stem cells (CD34(+)-HSCs), thus mimicking the Tay-Sachs (HEXA-/-) and Sandhoff (HEXB-/-) cells. We showed that the absence of ß-N-acetyl-hexosaminidase activity does not alter the differentiation of i-DCs from HSCs, but it is critical for the activation of CD4(+)T cells because knock-down of HEXA or HEXB gene causes a loss of function of i-DCs. Notably, the silencing of the HEXA gene had a stronger immune inhibitory effect, thereby indicating a major involvement of ß-N-acetyl-hexosaminidase A isoenzyme within this mechanism.


Asunto(s)
Células Dendríticas/inmunología , Células Madre/inmunología , Cadena alfa de beta-Hexosaminidasa/genética , Cadena beta de beta-Hexosaminidasa/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Gangliosidosis GM2/inmunología , Gangliosidosis GM2/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Células Madre/citología , Células Madre/metabolismo , Cadena alfa de beta-Hexosaminidasa/metabolismo , Cadena beta de beta-Hexosaminidasa/metabolismo
14.
PLoS Genet ; 6(9): e1001118, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20862357

RESUMEN

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.


Asunto(s)
Epilepsia/enzimología , Epilepsia/patología , Lisosomas/enzimología , Neuraminidasa/deficiencia , Neuronas/enzimología , Neuronas/patología , Cadena alfa de beta-Hexosaminidasa/metabolismo , Animales , Conducta Animal , Corteza Cerebral/enzimología , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Corteza Cerebral/ultraestructura , Electroencefalografía , Epilepsia/fisiopatología , Gangliósido G(M2)/metabolismo , Técnicas de Inactivación de Genes , Hipocampo/enzimología , Hipocampo/patología , Hipocampo/fisiopatología , Hipocampo/ultraestructura , Aprendizaje/fisiología , Lisosomas/patología , Lisosomas/ultraestructura , Ratones , Actividad Motora/fisiología , Neuraminidasa/metabolismo , Neuronas/ultraestructura
15.
Mol Ther ; 18(8): 1519-26, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20571546

RESUMEN

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.


Asunto(s)
Polisacáridos/metabolismo , Enfermedad de Sandhoff/metabolismo , Cadena alfa de beta-Hexosaminidasa/metabolismo , beta-N-Acetilhexosaminidasas/química , beta-N-Acetilhexosaminidasas/uso terapéutico , Animales , Células CHO , Células Cultivadas , Cromatografía en Capa Delgada , Cricetinae , Cricetulus , Gangliósido G(M2)/metabolismo , Glicosilación , Humanos , Immunoblotting , Ratones , Polisacáridos/química , Enfermedad de Sandhoff/tratamiento farmacológico , Enfermedad de Sandhoff/genética , Cadena alfa de beta-Hexosaminidasa/química , Cadena alfa de beta-Hexosaminidasa/genética , Cadena alfa de beta-Hexosaminidasa/uso terapéutico , Cadena beta de beta-Hexosaminidasa/genética , Cadena beta de beta-Hexosaminidasa/metabolismo , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/metabolismo
16.
Methods Mol Biol ; 444: 147-59, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18425478

RESUMEN

Tay-Sachs disease is an autosomal recessive storage disease caused by the impaired activity of the lysosomal enzyme hexosaminidase A. In this fatal disease, the sphingolipid GM2 ganglioside accumulates in the neurons. Due to high carrier rates and the severity of the disease, population screening and prenatal diagnosis of Tay-Sachs disease are routinely carried out in Israel. Laboratory diagnosis of Tay-Sachs is carried out with biochemical and DNA-based methods in peripheral and umbilical cord blood, amniotic fluid, and chorionic villi samples. The assay of hexosaminidase A (Hex A) activity is carried out with synthetic substrates, 4-methylumbelliferyl-6-sulfo-N-acetyl-beta-glucosaminide (4-MUGS) and 4-methylumbelliferil-N-acetyl-beta-glucosamine (4-MUG), and the DNA-based analysis involves testing for the presence of specific known mutations in the alpha-subunit gene of Hex A. Prenatal diagnosis of Tay-Sachs disease is accomplished within 24-48 h from sampling. The preferred strategy is to simultaneously carry out enzymatic analysis in the amniotic fluid supernatant or in chorionic villi and molecular DNA-based testing in an amniotic fluid cell-pellet or in chorionic villi.


Asunto(s)
Bioensayo , Análisis Mutacional de ADN , Pruebas Genéticas , Diagnóstico Prenatal/métodos , Enfermedad de Tay-Sachs/diagnóstico , Cadena alfa de beta-Hexosaminidasa , Acetilglucosamina/análogos & derivados , Acetilglucosamina/metabolismo , Amniocentesis , Líquido Amniótico/enzimología , Vellosidades Coriónicas/enzimología , Muestra de la Vellosidad Coriónica , Femenino , Sangre Fetal/enzimología , Regulación del Desarrollo de la Expresión Génica , Humanos , Himecromona/análogos & derivados , Himecromona/metabolismo , Mutación , Reacción en Cadena de la Polimerasa , Valor Predictivo de las Pruebas , Embarazo , Reproducibilidad de los Resultados , Especificidad por Sustrato , Enfermedad de Tay-Sachs/enzimología , Enfermedad de Tay-Sachs/genética , Factores de Tiempo , Cadena alfa de beta-Hexosaminidasa/genética , Cadena alfa de beta-Hexosaminidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...