Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Neurol ; 19(1): 20, 2019 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-30738426

RESUMO

BACKGROUND: Currently there are no disease-modifying treatments for Parkinson's disease dementia (PDD), a condition linked to aggregation of the protein α-synuclein in subcortical and cortical brain areas. One of the leading genetic risk factors for Parkinson's disease is being a carrier in the gene for ß-Glucocerebrosidase (GCase; gene name GBA1). Studies in cell culture and animal models have shown that raising the levels of GCase can decrease levels of α-synuclein. Ambroxol is a pharmacological chaperone for GCase and is able to raise the levels of GCase and could therefore be a disease-modifying treatment for PDD. The aims of this trial are to determine if Ambroxol is safe and well-tolerated by individuals with PDD and if Ambroxol affects cognitive, biochemical, and neuroimaging measures. METHODS: This is a phase II, single-centre, double-blind, randomized placebo-controlled trial involving 75 individuals with mild to moderate PDD. Participants will be randomized into Ambroxol high-dose (1050 mg/day), low-dose (525 mg/day), or placebo treatment arms. Assessments will be undertaken at baseline, 6-months, and 12-months follow up times. Primary outcome measures will be the Alzheimer's disease Assessment Scale-cognitive subscale (ADAS-Cog) and the ADCS Clinician's Global Impression of Change (CGIC). Secondary measures will include the Parkinson's disease Cognitive Rating Scale, Clinical Dementia Rating, Trail Making Test, Stroop Test, Unified Parkinson's disease Rating Scale, Purdue Pegboard, Timed Up and Go, and gait kinematics. Markers of neurodegeneration will include MRI and CSF measures. Pharmacokinetics and pharmacodynamics of Ambroxol will be examined through plasma levels during dose titration phase and evaluation of GCase activity in lymphocytes. DISCUSSION: If found effective and safe, Ambroxol will be one of the first disease-modifying treatments for PDD. TRIAL REGISTRATION: ClinicalTrials.gov NCT02914366, 26 Sep 2016/retrospectively registered.


Assuntos
Ambroxol/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Projetos de Pesquisa , Idoso , Encéfalo/efeitos dos fármacos , Demência/etiologia , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/psicologia
2.
J Inherit Metab Dis ; 27(6): 747-56, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15505380

RESUMO

GM(2) gangliosidoses are a group of panethnic lysosomal storage diseases in which GM(2) ganglioside accumulates in the lysosome due to a defect in one of three genes, two of which encode the alpha- or beta-subunits of beta- N -acetylhexosaminidase (Hex) A. A small inframe deletion mutation in the catalytic domain of the alpha-subunit of Hex has been found in five Turkish patients with infantile Tay-Sachs disease. To date it has not been detected in other populations and is the only mutation to be found in exon 10. It results in detectable levels of inactive alpha-protein in its precursor form. Because the alpha- and beta-subunits share 60% sequence identity, the Hex A and Hex B genes are believed to have arisen from a common ancestral gene. Thus the subunits must share very similar three-dimensional structures with conserved functional domains. Hex B, the beta-subunit homodimer is more stable than the heterodimeric Hex A, and much more stable than Hex S, the alpha homodimer. Thus, mutations that completely destabilize the alpha-subunit can often be partially rescued if expressed in the aligned positions in the beta-subunit. To better understand the severity of the Turkish HEXA mutation, we reproduced the 12 bp deletion mutation (1267-1278) in the beta-subunit cDNA. Western blot analysis of permanently transfected CHO cells expressing the mutant detected only the pro-form of the beta-subunit coupled with a total lack of detectable Hex B activity. These data indicate that the deletion of the four amino acids severely affects the folding of even the more stable beta-subunit, causing its retention in the endoplasmic reticulum and ultimate degradation.


Assuntos
beta-N-Acetil-Hexosaminidases/genética , Animais , Western Blotting , Células Cultivadas , Clonagem Molecular , Cricetinae , Primers do DNA , DNA Complementar/genética , Deleção de Genes , Hexosaminidase A , Hexosaminidase B , Humanos , Mutação/genética , Dobramento de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Turquia
5.
Biochemistry ; 40(18): 5440-6, 2001 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-11331008

RESUMO

In humans, beta-hexosaminidase A (alphabeta) is required to hydrolyze GM2 ganglioside. A deficiency of either the alpha- or beta-subunit leads to a severe neurological disease, Tay-Sachs or Sandhoff disease, respectively. In mammals beta-hexosaminidase B (betabeta) and S (alphaalpha) are other major and minor isozymes. The primary structures of the alpha- and beta-subunits are 60% identical, but only the alpha-containing isozymes can efficiently hydrolyze beta-linked GlcNAc-6-SO(4) from natural or artificial substrates. Hexosaminidase has been grouped with glycosidases in family 20. A molecular model of the active site of the human hexosaminidase has been generated from the crystal structure of a family 20 bacterial chitobiase. We now use the chitobiase structure to identify residues close to the carbon-6 oxygen of NAG-A, the nonreducing beta-GlcNAc residue of its bound substrate. The chitobiase side chains in the best interactive positions align with alpha-Asn(423)Arg(424) and beta-Asp(453)Leu(454). The change in charge from positive in alpha to negative in beta is consistent with the lower K(m) of hexosaminidase S, and the much higher K(m) and lower pH optimum of hexosaminidase B, toward sulfated versus unsulfated substrates. In vitro mutagenesis, CHO cell expression, and kinetic analyses of an alphaArg(424)Lys hexosaminidase S detected little change in V(max) but a 2-fold increase in K(m) for the sulfated substrate. Its K(m) for the nonsulfated substrate was unaffected. When alphaAsn(423) was converted to Asp, again only the K(m) for the sulfated substrate was changed, increasing by 6-fold. Neutralization of the charge on alphaArg(424) by substituting Gln produced a hexosaminidase S with a K(m) decrease of 3-fold and a V(max) increased by 6-fold for the unsulfated substrate, parameters nearly identical to those of hexosaminidase B at pH 4.2. As well, for the sulfated substrate at pH 4.2 its K(m) was increased 9-fold and its V(max) decreased 1.5-fold, values very similar to those of hexosaminidase B obtained at pH 3.0, where its betaAsp(453) becomes protonated.


Assuntos
Hexosaminidases/metabolismo , Isoenzimas/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Sequência de Aminoácidos , Animais , Arginina/genética , Asparagina/genética , Sítios de Ligação/genética , Células CHO , Cricetinae , Dissacarídeos/metabolismo , Hexosaminidase B , Hexosaminidases/genética , Humanos , Isoenzimas/genética , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Alinhamento de Sequência , Especificidade por Substrato/genética , Transfecção , beta-N-Acetil-Hexosaminidases/genética
6.
Biochemistry ; 40(7): 2201-9, 2001 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-11329289

RESUMO

Human beta-hexosaminidase A (alpha beta) and B (beta beta) are composed of subunits (alpha and beta) that are 60% identical and have been grouped with other evolutionarily related glycosidases into "Family 20". The three-dimensional structure of only one Family 20 member has been elucidated, a bacterial chitobiase. This enzyme shares primary structure homology with both the human subunits only in its active-site region, and even in this restricted area, the level of identity is only 26%. Thus, the validity of the molecular model for the active site of the human enzyme based on chitobiase must be determined experimentally. In this report, we analyze highly purified mutant forms of human hexosaminidase B that have had conservative substitutions made at Glu and Asp residues predicted by the chitobiase model to be part of its active site. Mutation of beta Glu(355) to Gln reduces k(cat) 5000-fold with only a small effect on K(m), while also shifting the pH optimum. These effects are consistent with assignment of this residue as the acid/base catalytic residue. Similarly, mutation of beta Asp(354) to Asn reduced k(cat) 2000-fold while leaving K(m) essentially unaltered, consistent with assignment of this residue as the residue that interacts with the substrate acetamide group to promote its attack on the anomeric center. These data in conjunction with the mutagenesis studies of Asp(241) and Glu(491) indicate that the molecular model is substantially accurate in its identification of catalytically important residues.


Assuntos
Ácido Aspártico/metabolismo , Ácido Glutâmico/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Acetilglucosaminidase/genética , Acetilglucosaminidase/metabolismo , Substituição de Aminoácidos/genética , Animais , Ácido Aspártico/genética , Sítios de Ligação/genética , Células CHO , Catálise , Cricetinae , Ácido Glutâmico/genética , Hexosaminidase B , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Proteínas de Membrana/metabolismo , Mutagênese Sítio-Dirigida , Conformação Proteica , Reprodutibilidade dos Testes , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Tetraspaninas , Transfecção , beta-N-Acetil-Hexosaminidases/genética
8.
Mol Hum Reprod ; 6(8): 699-706, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10908279

RESUMO

Mammalian sperm-zona pellucida (ZP) interaction is mediated by sperm lectin-like proteins and ZP glycoproteins. We have previously reported the participation of binding sites for N-acetylglucosamine (GlcNAc) residues in human sperm function, including sperm interaction with the ZP. Additionally, previous results from our laboratory suggested that some of these events may be mediated by the glycosidase N-acetylglucosaminidase (beta-hexosaminidase, Hex, in mammals). In this study, we report the possible participation of Hex in human sperm-ZP interaction. Human recombinant Hex (hrHex) was obtained by expression in a stable transfected CHO cell line. When the recombinant enzyme was present during hemizona (HZ) assays, the number of sperm bound per HZ was significantly reduced. The same result was obtained when HZ were preincubated with hrHex. Additionally, the presence of a Hex-specific substrate during the HZ assay produced the same inhibitory effect. These results suggest the participation of a sperm Hex in the interaction with human ZP in vitro.


Assuntos
Interações Espermatozoide-Óvulo/fisiologia , Zona Pelúcida/fisiologia , beta-N-Acetil-Hexosaminidases/metabolismo , Acetilglucosamina/análogos & derivados , Acetilglucosamina/metabolismo , Acetilglucosamina/farmacologia , Animais , Células CHO , Cricetinae , Feminino , Humanos , Concentração de Íons de Hidrogênio , Himecromona/análogos & derivados , Himecromona/metabolismo , Himecromona/farmacologia , Masculino , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Interações Espermatozoide-Óvulo/efeitos dos fármacos , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/farmacologia
9.
Biochemistry ; 39(20): 6219-27, 2000 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-10821697

RESUMO

Tay-Sachs or Sandhoff disease results from a deficiency of either the alpha- or the beta-subunits of beta-hexosaminidase A, respectively. These evolutionarily related subunits have been grouped with the "Family 20" glycosidases. Molecular modeling of human hexosaminidase has been carried out on the basis of the three-dimensional structure of a bacterial member of Family 20, Serratia marcescens chitobiase. The primary sequence identity between the two enzymes is only 26% and restricted to their active site regions; therefore, the validity of this model must be determined experimentally. Because human hexosaminidase cannot be functionally expressed in bacteria, characterization of mutagenized hexosaminidase must be carried out using eukaryotic cell expression systems that all produce endogenous hexosaminidase activity. Even small amounts of endogenous enzyme can interfere with accurate K(m) or V(max) determinations. We report the expression, purification, and characterization of a C-terminal His(6)-tag precursor form of hexosaminidase B that is 99.99% free of endogenous enzyme from the host cells. Control experiments are reported confirming that the kinetic parameters of the His(6)-tag precursor are the same as the untagged precursor, which in turn are identical to the mature isoenzyme. Using highly purified wild-type and Arg(211)Lys-substituted hexosaminidase B, we reexamine the role of Arg(211) in the active site. As we previously reported, this very conservative substitution nevertheless reduces k(cat) by 500-fold. However, the removal of all endogenous activity has now allowed us to detect a 10-fold increase in K(m) that was not apparent in our previous study. That this increase in K(m) reflects a decrease in the strength of substrate binding was confirmed by the inability of the mutant isozyme to efficiently bind an immobilized substrate analogue, i.e., a hexosaminidase affinity column. Thus, Arg(211) is involved in substrate binding, as predicted by the chitobiase model, as well as catalysis.


Assuntos
Arginina/química , beta-N-Acetil-Hexosaminidases/química , Animais , Arginina/genética , Sítios de Ligação/genética , Western Blotting , Células CHO , Cromatografia de Afinidade , Dicroísmo Circular , Cricetinae , Precursores Enzimáticos/química , Precursores Enzimáticos/genética , Estabilidade Enzimática , Hexosaminidase B , Histidina/genética , Humanos , Mutagênese Sítio-Dirigida , Sinais Direcionadores de Proteínas/genética , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/isolamento & purificação , Transfecção , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/isolamento & purificação
10.
J Androl ; 20(6): 779-802, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10591618

RESUMO

Beta-hexosaminidase (Hex) is a lysosomal enzyme that exists as two major isoenzymes: Hex A (subunit structure, alphabeta) and Hex B (betabeta). The presence of Hex in the testis and epididymis suggests important roles for the enzyme and its substrates in male fertility and reproductive functions. Disruption of the Hexb gene encoding the beta-subunit of Hex has led to the generation of a mouse model of human Sandhoff disease that survives to adulthood, enabling us to analyze the effects of Hex A and Hex B deficiency on epithelial cellular morphology of the male reproductive tract. At 1 and 3 months of age, the testes, efferent ducts, and epididymides of Hex-deficient (Hexb -/-) and wild-type (Hexb +/+) mice were perfuse fixed and analyzed by routine light and electron microscopy (LM and EM, respectively) as well as with immunocytochemistry employing antibodies to lysosomal proteins. In the testis, the morphological appearance and topographical arrangement of the cell types of the seminiferous epithelium of Hexb -/- mice were similar to those of wild-type animals at both ages. Both Sertoli and germ cells appeared to be unaffected. However, at both ages, myoid cells and macrophages showed an increased number of lysosomes in their cytoplasm as compared with the number seen in controls. The epithelial cells of the efferent ducts also showed an accumulation of lysosomes that increased with age as compared with controls. Principal cells of the entire epididymis revealed an increase in the size and number of lysosomes at 1 month of age as compared with those of controls, and by 3 months, these lysosomes often filled the supranuclear and basal regions of the cells. Narrow cells of the distal initial segment and intermediate zone, normally slender cells showing several lysosomes, became greatly enlarged and entirely filled with lysosomes in Hexb -/- mice. Clear cells of the caput, corpus, and cauda regions also showed a progressive increase in the size and number of lysosomes with age as compared with controls; the clear cells of the mutant mice were often enlarged and at times bulged into the lumen. Some basal cells of each epididymal region in Hexb -/- mice were similar to controls at 1 and 3 months, showing few lysosomes, while others showed an accumulation of lysosomes. Lysosomes of all affected epithelial cells were of varying sizes, but many large ones were present, apparently resulting from lysosomal fusion. Although pale stained, their identification as lysosomes was confirmed by EM immunocytochemistry with anti-cathepsin D and anti-Hex A antibodies. Predominantly in the proximal initial segment, large, pale cellular aggregates were noted in the LM analysis at the base of the epithelium, which by EM analysis were identified as belonging to two different cell types, narrow cells and halo cells. Taken together, these data reveal an increase in the size and number of lysosomes in all epithelial cell types lining the efferent ducts and entire epididymis as well as in myoid cells and macrophages of the testis. In the light of data showing epididymal defects restricted predominantly to the initial segment in Hexa -/- (Hex A-deficient) mice, our data on the Hexb -/- mice demonstrate a major role for Hex that can be fulfilled by either Hex A or Hex B in the epididymis.


Assuntos
Epididimo/anormalidades , Doença de Sandhoff/patologia , Testículo/anormalidades , beta-N-Acetil-Hexosaminidases/deficiência , beta-N-Acetil-Hexosaminidases/genética , Envelhecimento , Animais , Modelos Animais de Doenças , Epididimo/crescimento & desenvolvimento , Hexosaminidase A , Hexosaminidase B , Humanos , Lisossomos/patologia , Lisossomos/ultraestrutura , Masculino , Camundongos , Camundongos Knockout , Valores de Referência , Células de Sertoli/citologia , Espermatozoides/citologia , Testículo/crescimento & desenvolvimento , Testículo/ultraestrutura
11.
Biochim Biophys Acta ; 1455(2-3): 105-38, 1999 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-10571007

RESUMO

The hydrolysis of GM2-ganglioside is unusual in its requirements for the correct synthesis, processing, and ultimate combination of three gene products. Whereas two of these proteins are the alpha- (HEXA gene) and beta- (HEXB) subunits of beta-hexosaminidase A, the third is a small glycolipid transport protein, the GM2 activator protein (GM2A), which acts as a substrate specific co-factor for the enzyme. A deficiency of any one of these proteins leads to storage of the ganglioside, primarily in the lysosomes of neuronal cells, and one of the three forms of GM2-gangliosidosis, Tay-Sachs disease, Sandhoff disease or the AB-variant form. Studies of the biochemical impact of naturally occurring mutations associated with the GM2 gangliosidoses on mRNA splicing and stability, and on the intracellular transport and stability of the affected protein have provided some general insights into these complex cellular mechanisms. However, such studies have revealed little in the way of structure-function information on the proteins. It appears that the detrimental effect of most mutations is not specifically on functional elements of the protein, but rather on the proteins' overall folding and/or intracellular transport. The few exceptions to this generalization are missense mutations at two codons in HEXA, causing the unique biochemical phenotype known as the B1-variant, and one codon in both the HEXB and GM2A genes. Biochemical characterization of these mutations has led to the localization of functional residues and/or domains within each of the encoded proteins.


Assuntos
Mutação , Doença de Sandhoff/genética , Doença de Tay-Sachs/genética , beta-N-Acetil-Hexosaminidases/genética , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sequência de Carboidratos , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Proteína Ativadora de G(M2) , Gangliosídeo G(M2)/metabolismo , Deleção de Genes , Hexosaminidase A , Hexosaminidase B , Humanos , Isoenzimas/genética , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Fenótipo , Proteínas/genética , Proteínas/metabolismo , Splicing de RNA , Cadeia beta da beta-Hexosaminidase , beta-N-Acetil-Hexosaminidases/química
12.
Am J Hum Genet ; 65(1): 77-87, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10364519

RESUMO

Deficiency of the GM2 activator protein, encoded by GM2A, results in the rare AB-variant form of GM2 gangliosidosis. Four mutations have been identified, but the human gene structure has been only partially characterized. We report a new patient from a Laotian deme whose cells are deficient in both GM2-activator mRNA and protein. However, reverse transcription (RT)-PCR detected some normal-sized cDNA and a smaller cDNA species, which was not seen in the RT-PCR products from normal controls. Sequencing revealed that, although the patient's normal-sized cDNA contained a single nonsense mutation in exon 2, his smaller cDNA was the result of an in-frame deletion of exon 2. Long PCR was used to amplify introns 1 and 2 from patient and normal genomic DNA, and no differences in size, in 5' and 3' end sequences, or in restriction-mapping patterns were observed. From these data we developed a set of four PCR primers that can be used to identify GM2A mutations. We use this procedure to demonstrate that the patient is likely homozygous for the nonsense mutation. Other reports have associated nonsense mutations with dramatically reduced levels of mRNA and with an increased level of skipping of the exon containing the mutation, thus reestablishing an open reading frame. However, a recent article has concluded that, in some cases, the latter observation is caused by an artifact of RT-PCR. In support of this conclusion, we demonstrate that, if the competing, normal-sized cDNA is removed from the initial RT-PCR products, from both patient and normal cells, by an exon 2-specific restriction digest; a second round of PCR produces similar amounts of exon 2-deleted cDNA.


Assuntos
Deleção de Genes , Mutação , Proteínas/genética , Sequência de Bases , Pré-Escolar , Éxons , Proteína Ativadora de G(M2) , Humanos , Íntrons , Masculino , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Doença de Tay-Sachs/diagnóstico
13.
Biochem Biophys Res Commun ; 258(2): 256-9, 1999 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-10329374

RESUMO

The GM2 activator protein is required as a substrate-specific cofactor for beta-hexosaminidase A to hydrolyze GM2 ganglioside. The GM2 activator protein reversibly binds and solubilizes individual GM2 ganglioside molecules, making them available as substrate. Although GM2 ganglioside is the strongest binding ligand for the activator protein, it can also bind and transport between membranes a series of other glycolipids, even at neutral pH. Biosynthetic studies have shown that a large portion of newly synthesized GM2 activator molecules are not targeted to the lysosome, but are secreted and can then be recaptured by other cells through a carbohydrate independent mechanism. Thus, the GM2 activator protein may have other in vivo functions. We found that the GM2 activator protein can inhibit, through specific binding, the ability of platelet activating factor (PAF) to stimulate the release of intracellular Ca2+ pools by human neutrophils. PAF is a biologically potent phosphoacylglycerol. Inhibitors for PAF's role in the pathogenesis of inflammatory bowel disease and asthma have been sought as potential therapeutic agents. The inherent stability and protease resistance of the small, monomeric GM2 activator protein, coupled with the ability to produce large quantities of the functional protein in transformed bacteria, suggest it may serve as such an agent.


Assuntos
Fator de Ativação de Plaquetas/antagonistas & inibidores , Proteínas/metabolismo , Cálcio/metabolismo , Proteína Ativadora de G(M2) , Humanos , Fator de Ativação de Plaquetas/metabolismo , Ligação Proteica , Espectrometria de Fluorescência
14.
J Am Soc Nephrol ; 10(3): 435-43, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10073593

RESUMO

This study describes the molecular characterization of an antigen defined by an autoantibody from a woman with habitual abortion as GM2-activator protein. The patient showed no disorder of renal function. Accidentally with routine serum screening for autoantibodies, an immunoreactivity was found in kidney collecting duct intercalated cells. Three distinct patterns of immunostaining of intercalated cells were observed: staining of the apical pole, basolateral pole, and diffuse cytoplasmic labeling. Ultrastructurally, the immunoreactivity was associated with "studs," which represent the cytoplasmic domain of the vacuolar proton pump in intercalated cells. This pump is subjected to a shuttling mechanism from cytoplasmic stores to the cell membrane, which exclusively occurs in intercalated cells. Peptide sequences of a 23-kD protein purified from rat kidney cortex showed complete identity with corresponding sequences of GM2-activator protein. In the brain, GM2-activator protein is required for hexosaminidase A to split a sugar from ganglioside GM2. Because neither ganglioside GM2 nor GM1 (its precursor) is present in significant amounts in the kidney, the previous finding that this tissue contains the highest level of activator protein in the body was confusing. In this study, a novel role for GM2-activator protein in intercalated cells is proposed, and possible roles in the shuttling mechanism are discussed.


Assuntos
Gangliosídeo G(M2)/metabolismo , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/ultraestrutura , Proteínas/metabolismo , Aborto Habitual/imunologia , Animais , Reações Antígeno-Anticorpo , Autoanticorpos/análise , Sequência de Bases , Transporte Biológico , Membrana Celular/ultraestrutura , Células Cultivadas , Citoplasma/ultraestrutura , Feminino , Proteína Ativadora de G(M2) , Gangliosídeo G(M2)/genética , Gangliosídeo G(M2)/isolamento & purificação , Humanos , Imuno-Histoquímica , Túbulos Renais Coletores/química , Túbulos Renais Coletores/citologia , Microscopia Confocal , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Gravidez , Ligação Proteica , Proteínas/genética , Proteínas/isolamento & purificação , Bombas de Próton/ultraestrutura , Ratos , Ratos Sprague-Dawley , Valores de Referência , Sensibilidade e Especificidade
15.
J Biol Chem ; 273(33): 21386-92, 1998 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-9694901

RESUMO

The GM2 gangliosidoses are caused by mutations in the genes encoding the alpha- (Tay-Sachs) or beta- (Sandhoff) subunits of heterodimeric beta-hexosaminidase A (Hex A), or the GM2 activator protein (AB variant), a substrate-specific co-factor for Hex A. Although the active site associated with the hydrolysis of GM2 ganglioside, as well as part of the binding site for the ganglioside-activator complex, is associated with the alpha-subunit, elements of the beta-subunit are also involved. Missense mutations in these genes normally result in the mutant protein being retained in the endoplasmic reticulum and degraded. The mutations associated with the B1-variant of Tay-Sachs are rare exceptions that directly affect residues in the alpha-active site. We have previously reported two sisters with chronic Sandhoff disease who were heterozygous for the common HEXB deletion allele. Cells from these patients had higher than expected levels of mature beta-protein and residual Hex A activity, approximately 20%. We now identify these patients' second mutant allele as a C1510T transition encoding a beta-Pro504 --> Ser substitution. Biochemical characterization of Hex A from both patient cells and cotransfected CHO cells demonstrated that this substitution (a) decreases the level of heterodimer transport out of the endoplasmic reticulum by approximately 45%, (b) lowers its heat stability, (c) does not affect its Km for neutral or charged artificial substrates, and (d) lowers the ratio of units of ganglioside/units of artificial substrate hydrolyzed by a factor of 3. We concluded that the beta-Pro504 --> Ser mutation directly affects the ability of Hex A to hydrolyze its natural substrate but not its artificial substrates. The effect of the mutation on ganglioside hydrolysis, combined with its effect on intracellular transport, produces chronic Sandhoff disease.


Assuntos
Gangliosídeo G(M2)/metabolismo , Prolina/metabolismo , Doença de Sandhoff/metabolismo , Serina/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Substituição de Aminoácidos , Animais , Sequência de Bases , Células CHO , Doença Crônica , Cricetinae , Primers do DNA , Heterozigoto , Hexosaminidase A , Hexosaminidase B , Homozigoto , Humanos , Hidrólise , Prolina/química , Prolina/genética , Doença de Sandhoff/genética , Serina/química , Serina/genética , beta-N-Acetil-Hexosaminidases/química , beta-N-Acetil-Hexosaminidases/genética
16.
Biochim Biophys Acta ; 1393(1): 1-18, 1998 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-9714704

RESUMO

Although there is only one documented function carried out by the GM2 activator protein in the lysosome, new information suggests that other less obvious roles may also be played by this protein in vivo. This information includes data demonstrating that the GM2 activator is a secretory, as well as a lysosomal protein, and that cells possess a carbohydrate-independent mechanism to re-capture the activator, with or without bound lipid, from the extracellular fluid. Additionally the GM2 activator has been shown to bind, solubilize and transport a broad spectrum of lipid molecules, such as glycolipids, gangliosides and at least one phosphoacylglycerol, between liposomes. At pH 7 the GM2 activator's rate of lipid transport is reduced by only 50% from its maximum rate which is achieved at approx. pH 5, suggesting that the GM2 activator may serve as a general intra- and/or inter-cellular lipid transport protein in vivo. Since the late 1970s the lysosomal form of the GM2 activator has been known to act as a substrate-specific co-factor for the hydrolysis of GM2 ganglioside by beta-hexosaminidase A. Gangliosides are a class of negatively charged glycolipids particularly abundant in neuronal cells which have been linked to numerous in vivo functions, such as memory formation and signal transduction events. Deficiency of the GM2 activator protein results in the storage of GM2 ganglioside and severe neurological disease, the AB-variant form of GM2 gangliosidosis, usually culminating in death before the age of 4 years. The exact mode-of-action of the GM2 activator in its role as a co-factor, and its specificity for various glycolipids are currently matters of debate in the literature.


Assuntos
Gangliosídeo G(M2)/metabolismo , Glicolipídeos/metabolismo , Proteínas/metabolismo , Animais , Transporte Biológico , Proteína Ativadora de G(M2) , Gangliosidoses/metabolismo , Humanos , Hidrólise , Lipossomos/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
17.
Hum Mutat ; 11(6): 432-42, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9603435

RESUMO

Mutations in the HEXA gene, encoding the alpha-subunit of beta-hexosaminidase A (Hex A), that abolish Hex A enzyme activity cause Tay-Sachs disease (TSD), the fatal infantile form of G(M2) gangliosidosis, Type 1. Less severe, subacute (juvenile-onset) and chronic (adult-onset) variants are characterized by a broad spectrum of clinical manifestations and are associated with residual levels of Hex A enzyme activity. We identified a 1422 G-->C (amino acid W474C) substitution in the first position of exon 13 of HEXA of a non-Jewish proband who manifested a subacute variant of G(M2) gangliosidosis. On the second maternally inherited allele, we identified the common infantile disease-causing 4-bp insertion, +TATC 1278, in exon 11. Pulse-chase analysis using proband fibroblasts revealed that the W474C-containing alpha-subunit precursor was normally synthesized, but not phosphorylated or secreted, and the mature lysosomal alpha-subunit was not detected. When the W474C-containing alpha-subunit was transiently co-expressed with the beta-subunit to produce Hex A (alphabeta) in COS-7 cells, the mature alpha-subunit was present, but its level was much lower than that from normal alpha-subunit transfections, although higher than in those cells transfected with an alpha-subunit associated with infantile TSD. Furthermore, the precursor level of the W474C alpha-subunit was found to accumulate in comparison to the normal alpha-subunit precursor levels. We conclude that the 1422 G-->C mutation is the cause of Hex A enzyme deficiency in the proband. The resulting W474C substitution clearly interferes with alpha-subunit processing, but because the base substitution falls at the first position of exon 13, aberrant splicing may also contribute to Hex A deficiency in this proband.


Assuntos
Processamento de Proteína Pós-Traducional , Doença de Tay-Sachs/genética , beta-N-Acetil-Hexosaminidases/metabolismo , Adolescente , Idade de Início , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Células COS , Linhagem Celular , Eletroforese em Gel de Campo Pulsado , Éxons , Fibroblastos/enzimologia , Hexosaminidase A , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Fosforilação , Homologia de Sequência de Aminoácidos , Doença de Tay-Sachs/enzimologia , Transfecção , beta-N-Acetil-Hexosaminidases/química , beta-N-Acetil-Hexosaminidases/genética
18.
Biochemistry ; 37(3): 814-21, 1998 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-9454570

RESUMO

The function of the GM2 activator protein is to act as a substrate-specific cofactor in the hydrolysis of GM2 ganglioside by beta-hexosaminidase A. Mutations in the gene encoding it result in the AB variant form of GM2 gangliosidosis. One such mutation, Cys138 Arg, results in the mutant protein being retained and degraded in the endoplasmic reticulum of mammalian cells. In order to characterize the biochemical effects of this substitution, we expressed the mutant protein in transformed bacteria. We first compared the wild-type protein produced by two bacterial expression methods, one requiring protein refolding, with activator purified from the medium of transfected CHO cells. The "activity" and circular dichroism spectrum (alpha-helical content) of all three proteins were similar, justifying the use of refolded activator from transformed bacteria in structure/function studies. Second, the mutant protein was expressed in both bacterial systems and in each retained approximately 2% of the wild type's specific activity. The presence of even this small amount of activity in the mutant protein coupled with a calculated alpha-helical content nearly identical to the wild type, strongly suggest that no major tertiary or secondary structural changes, respectively, had occurred due to the mutation. However, we demonstrate that its heat stability at 60 degrees C is reduced 14-fold, suggesting some localized change in tertiary structure. The loss of a disulfide loop was confirmed by reacting the mutant protein with Ellman's reagent. A kinetic analysis detected a large increase in the apparent K(m) of beta-hexosaminidase A for the mutant; however, there was no apparent change in Vmax. A fluorescence dequenching assay was used to evaluate the ability of the mutant protein to transport lipids and bind GM2 ganglioside. These assays detected no difference between the wild-type and mutant proteins, indicating that the Cys138 Arg substitution has no effect on these functions. We conclude that the mutation specifically affects a domain in the activator protein that is responsible for the recognition of the activator-GM2 ganglioside complex by beta-hexosaminidase A.


Assuntos
Substituição de Aminoácidos/genética , Arginina/genética , Cisteína/genética , Gangliosídeo G(M2)/genética , Gangliosidoses/genética , Proteínas/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Cisteína/fisiologia , Dissulfetos , Escherichia coli/genética , Proteína Ativadora de G(M2) , Gangliosidoses/enzimologia , Variação Genética , Dados de Sequência Molecular , Oligopeptídeos , Peptídeos/genética , Dobramento de Proteína , Proteínas/genética , Proteínas Recombinantes de Fusão/biossíntese
19.
Biochemistry ; 36(27): 8325-31, 1997 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-9204879

RESUMO

The GM2 activator protein is a small monomeric protein containing a single site for Asn-linked glycosylation. Its only proven in vivo function is to act as a substrate specific cofactor for the hydrolysis of GM2 ganglioside by lysosomal beta-hexosaminidase A. However, we and others have shown it can act as a general glycolipid transporter at neutral pH in vitro. Any other possible in vivo functions would require that some of the newly synthesized activator molecules not be targeted to the lysosome. The lysosomal targeting mechanism for the activator has not been conclusively identified. While earlier reports suggested that it is likely through the mannose-6-phosphate receptor, another more recent report demonstrated that deficient human cells could recapture nonglycosylated, bacterially produced activator, suggesting its use of an alternate targeting pathway. Here, we demonstrate that the mannose-6-phosphate pathway is likely the major intracellular, biosynthetic route to the lysosome, as well as a high affinity recapture pathway for the endocytosis of activator protein from extracellular fluids. Additionally, we show that there exists a second lower affinity recapture pathway that requires its native protein structure, is carbohydrate independent, and likely does not involve its ability to bind glycosphingolipids in the plasma membrane. Finally, we document that the pool of newly synthesized precursor activator protein contains a majority of molecules with a complex-type oligosaccharide, which cannot contain a functional mannose-6-phosphate targeting signal. These molecules makeup the secreted forms of the protein in normal human fibroblasts.


Assuntos
Proteínas/metabolismo , Sequência de Aminoácidos , Cloreto de Amônio/farmacologia , Animais , Sítios de Ligação , Células CHO , Cricetinae , Endocitose , Fibroblastos/metabolismo , Proteína Ativadora de G(M2) , Gangliosídeo G(M2)/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Lisossomos/enzimologia , Manosefosfatos/farmacologia , Proteínas/genética , Proteínas Recombinantes de Fusão , Transfecção , beta-N-Acetil-Hexosaminidases/metabolismo
20.
Biochim Biophys Acta ; 1339(2): 192-202, 1997 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-9187239

RESUMO

The G(M2) activator protein is a substrate specific cofactor for degradation of G(M2) ganglioside by lysosomal beta-hexosaminidase A. Mutations in the gene encoding the activator result in the AB-variant form of G(M2) gangliosidosis. The activator protein contains at least three functional elements; a hydrophobic binding pocket, an oligosaccharide binding site(s), and an area that interacts with hexosaminidase A. In this report a fluorescence dequenching assay specific for only the hydrophobic binding pocket is evaluated and optimized. It is shown that various glycolipids inhibit the transport between liposomes of a self-quenching fluorescent lipid probe, octadecylrhodamine, by the activator protein. The level of inhibition produced by each glycolipid is then used to characterize the oligosaccharide-binding specificity of the activator. The fluorescence dequenching assay is also used to evaluate the functionality of a truncated form of the activator protein. Our results indicate that this simple assay can be used to determine structure-function relationships within the normal or mutant forms of the activator. The data suggest that the C-terminus of the activator is required to produce a functional hydrophobic binding pocket.


Assuntos
Gangliosídeo G(M2)/metabolismo , Glicolipídeos/metabolismo , Proteínas/metabolismo , Sítios de Ligação , Sequência de Carboidratos , Clonagem Molecular , Escherichia coli/genética , Fluorescência , Proteína Ativadora de G(M2) , Hexosaminidase A , Dados de Sequência Molecular , Oligossacarídeos/química , Reação em Cadeia da Polimerase , Proteínas/química , Proteínas/genética , Relação Estrutura-Atividade , beta-N-Acetil-Hexosaminidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...