Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
Ecotoxicol Environ Saf ; 244: 114043, 2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-36087468

RESUMO

Silicosis is one of the most important occupational diseases worldwide, caused by inhalation of silica particles or free crystalline silicon dioxide. As a disease with high mortality, it has no effective treatment and new therapeutic targets are urgently needed. Recent studies have identified FCER1A, encoding α-subunit of the immunoglobulin E (IgE) receptor FcεRI, as a candidate gene involved in the biological pathways leading to respiratory symptoms. FcεRI is known to be important in allergic asthma, but its role in silicosis remains unclear. In this study, serum IgE concentrations and FcεRI expression were assessed in pneumoconiosis patients and silica-exposed mice. The role of FcεRI was explored in a silica-induced mouse model using wild-type and FcεRI-deficient mice. The results showed that serum IgE concentrations were significantly elevated in both pneumoconiosis patients and mice exposed to silica compared with controls. The mRNA and protein expression of FcεRI were also significantly increased in the lung tissue of patients and silica-exposed mice. FcεRI deficiency significantly attenuated the changes in lung function caused by silica exposure. Silica-induced elevations of IL-1ß, IL-6, and TNF-α were significantly attenuated in the lung tissue and bronchoalveolar lavage fluid (BALF) of FcεRI-deficient mice compared with wild-type controls. Additionally, FcεRI-deficient mice showed a significantly lower score of pulmonary fibrosis than wild-type mice following exposure to silica, with significantly lower hydroxyproline content and expression of fibrotic genes Col1a1 and Fn1. Immunofluorescent staining suggested FcεRI mainly on mast cells. Mast cell degranulation took place after silica exposure, as shown by increased serum histamine levels and ß-hexosaminidase activity, which were significantly reduced in FcεRI-deficient mice compared with wild-type controls. Together, these data showed that FcεRI deficiency had a significant protective effect against silica-induced pulmonary inflammation and fibrosis. Our findings provide new insights into the pathophysiological mechanisms of silica-induced pulmonary fibrosis and a potential target for the treatment of silicosis.


Assuntos
Pneumonia , Fibrose Pulmonar , Silicose , Animais , Fibrose , Histamina/metabolismo , Histamina/toxicidade , Hidroxiprolina/metabolismo , Hidroxiprolina/farmacologia , Hidroxiprolina/uso terapêutico , Imunoglobulina E , Interleucina-6/metabolismo , Pulmão , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , RNA Mensageiro/metabolismo , Receptores de IgE/genética , Receptores de IgE/metabolismo , Receptores de IgE/uso terapêutico , Dióxido de Silício/toxicidade , Silicose/genética , Silicose/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , beta-N-Acetil-Hexosaminidases/farmacologia , beta-N-Acetil-Hexosaminidases/uso terapêutico
3.
Bioengineered ; 13(5): 13341-13351, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35635016

RESUMO

miR-205-5p plays a vital role in the inflammation of allergic rhinitis (AR). The study is designed to investigate the effects and mechanism of miR-205-5p in AR in vivo and in vitro. An OVA-induced mice model and anti-DNP IgE-induced RBL-2H3 cell model were established. The pathological alterations in the nasal mucosa were evaluated by hematoxylin-eosin (HE) staining. IgE and histamine levels were detected by corresponding kits and the expressions of PEBP1, High mobility group box-1 (HMGB1) and Toll-like receptor 4 (TLR4) were detected by western blot. The association of miR-205-5p and PEBP1 was determined by dual-luciferase reported assay. ß-hexosaminidase activity was to evaluate the degranulation of RBL-2H3 cell. The pathological injury of nasal mucosa was significantly improved by miR-205-5p inhibition compared to AR mice. Following the treatment of miR-205-5p inhibitor, the levels of helper T cell (Th1) cytokines, interleukin (IL)-2 and interferon-γ (IFN-γ) were increased, while the levels of Th2 cytokines, IL-4 and IL-13, as well as the levels of IgE and histamine were markedly decreased in AR mice. We further found that miR-205-5P inhibition induced increased expression of PEBP1 and decreased expressions of HMGB1and TLR4. In vitro, miR-205-5P was verified to bind to PEBP1. PEBP1 silencing led to the reverse of miR-205-5p effects on decreasing the levels of ß-hexosaminidase activity and histamine, as well as the expressions of HMGB1 and TLR4 on anti-DNP IgE-induced RBL-2H3 cells. Our results indicate that miR-205-5P inhibition may ameliorate pathological injury via PEBP1. MiR-205-5P/ PEBP1 could be potential drug targets in AR.


Assuntos
Proteína HMGB1 , MicroRNAs , Rinite Alérgica , Animais , Citocinas/metabolismo , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Histamina/uso terapêutico , Liberação de Histamina , Imunoglobulina E/metabolismo , Imunoglobulina E/uso terapêutico , Camundongos , MicroRNAs/metabolismo , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Proteína de Ligação a Fosfatidiletanolamina/uso terapêutico , Rinite Alérgica/tratamento farmacológico , Rinite Alérgica/genética , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo , beta-N-Acetil-Hexosaminidases/uso terapêutico
4.
Food Funct ; 13(4): 2216-2227, 2022 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-35133390

RESUMO

Inflammatory bowel disease (IBD) is associated with the microbial composition of the gut and its metabolites. Akkermansia muciniphila is a probiotic that exerts a significant alleviative or therapeutic effect on host enteritis. This study was designed to determine the protective effect and potential mechanism underlying the secretion of ß-acetylaminohexosidase (Amuc_2109) by A. muciniphila against dextran sulfate sodium (DSS)-induced colitis in mice. C57BL/6 mice were gavaged with Amuc_2109 for 21 days, and during the last seven days of treatment, they drank DSS dissolved in their drinking water to induce colitis. Our results showed that supplementation with Amuc_2109 improved DSS-induced colitis as evidenced by lowered disease activity index (DAI) scores, reduced weight loss, increased colon length, and inhibited oxidative stress. In addition, Amuc_2109 inhibited the overexpression of inflammatory cytokines (TNF-α, IL-1ß, IL-6) and the NLR family pyrin domain containing 3 (NLRP3) inflammasome in DSS-induced colitis. Furthermore, supplementation with Amuc_2109 also restored the mRNA expression of tight junction proteins (ZO-1, occludin, claudin-1). Further analysis of fecal microbial 16S rRNA sequences showed that Amuc_2109 reshaped the intestinal microbiota. While the anti-inflammatory effects of Amuc_2109 were only manifested with the wild-type protein, the anti-inflammatory effects were completely lost after the mutation of its key catalytic amino acids rendered Amuc_2109 inactive. In summary, these findings demonstrate the potential of Amuc_2109, as a therapeutic agent for ulcerative colitis. We posit that it will provide additional assistance in the prevention and treatment of mucus layer-related diseases such as ulcerative colitis.


Assuntos
Colite Ulcerativa/prevenção & controle , Substâncias Protetoras/uso terapêutico , beta-N-Acetil-Hexosaminidases/uso terapêutico , Akkermansia , Animais , Colite Ulcerativa/induzido quimicamente , Colo/efeitos dos fármacos , Sulfato de Dextrana , Modelos Animais de Doenças , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Substâncias Protetoras/farmacologia , beta-N-Acetil-Hexosaminidases/farmacologia
5.
Cell Chem Biol ; 25(10): 1255-1267.e8, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30146240

RESUMO

In this study we developed an efficient method to prepare glycoengineered ß-N-acetylhexosaminidase containing multiple mannose-6-phosphates (M6Ps) by combining genetic code expansion with bioorthogonal ligation techniques. We found that multiple M6P-conjugated enzymes were produced with a high efficiency by using combined techniques. Importantly, glycoengineered enzymes entered lysosomes of patient-derived primary cells, which lack endogenous lysosomal ß-N-acetylhexosaminidase, more readily than commercialized human ß-hexosaminidase. Moreover, glycoengineered enzymes successfully removed GM2-ganglioside stored in lysosomes of diseased cells, indicating that its activity is restored in diseased cells. We also synthesized and applied a lysosome-targeting fluorogenic substrate to monitor endogenous and supplemental glycoengineered ß-N-acetylhexosaminidase activities in lysosomes. The results of this study indicate that the present strategy, which relies on genetic code expansion and bioorthogonal ligation techniques, is highly attractive to generate multi-M6P-containing lysosomal enzymes that can be used to study lysosomal storage disorders associated with lysosomal enzyme deficiencies.


Assuntos
Gangliosídeo G(M2)/metabolismo , Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Lisossomos/enzimologia , Manosefosfatos/uso terapêutico , Engenharia de Proteínas/métodos , beta-N-Acetil-Hexosaminidases/uso terapêutico , Animais , Linhagem Celular , Células Cultivadas , Terapia Enzimática , Feminino , Células HEK293 , Humanos , Doenças por Armazenamento dos Lisossomos/enzimologia , Doenças por Armazenamento dos Lisossomos/metabolismo , Doenças por Armazenamento dos Lisossomos/patologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/patologia , Masculino , Manosefosfatos/química , Manosefosfatos/genética , Camundongos , Modelos Moleculares , Células NIH 3T3 , beta-N-Acetil-Hexosaminidases/química , beta-N-Acetil-Hexosaminidases/genética
6.
Mol Genet Metab ; 116(1-2): 80-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25971245

RESUMO

Sandhoff disease (SD) is a fatal neurodegenerative disease caused by a mutation in the enzyme ß-N-acetylhexosaminidase. Children with infantile onset SD develop seizures, loss of motor tone and swallowing problems, eventually reaching a vegetative state with death typically by 4years of age. Other symptoms include vertebral gibbus and cardiac abnormalities strikingly similar to those of the mucopolysaccharidoses. Isolated fibroblasts from SD patients have impaired catabolism of glycosaminoglycans (GAGs). To evaluate mucopolysaccharidosis-like features of the feline SD model, we utilized radiography, MRI, echocardiography, histopathology and GAG quantification of both central nervous system and peripheral tissues/fluids. The feline SD model exhibits cardiac valvular and structural abnormalities, skeletal changes and spinal cord compression that are consistent with accumulation of GAGs, but are much less prominent than the severe neurologic disease that defines the humane endpoint (4.5±0.5months). Sixteen weeks after intracranial AAV gene therapy, GAG storage was cleared in the SD cat cerebral cortex and liver, but not in the heart, lung, skeletal muscle, kidney, spleen, pancreas, small intestine, skin, or urine. GAG storage worsens with time and therefore may become a significant source of pathology in humans whose lives are substantially lengthened by gene therapy or other novel treatments for the primary, neurologic disease.


Assuntos
Terapia Genética , Doença de Sandhoff/genética , Doença de Sandhoff/terapia , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/uso terapêutico , Adenoviridae/genética , Estruturas Animais/patologia , Animais , Gatos , Modelos Animais de Doenças , Vetores Genéticos , Humanos , Mucopolissacaridoses/genética , Mucopolissacaridoses/patologia , Mucopolissacaridoses/terapia , Fenótipo , Doença de Sandhoff/fisiopatologia , Doença de Sandhoff/urina
7.
Bull Exp Biol Med ; 153(1): 94-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22808503

RESUMO

Hypoglycemic effect of hyaluronate-endo-ß-N-acetylhexosaminidase immobilized by electron-beam synthesis nanotechnology (imHEA-HA) was studied in experimental insulin-dependent and insulin-independent diabetes mellitus. The drug exhibited a hypoglycemic effect of its own and potentiated the pharmacological effect of exogenous insulin injected in vivo. Studies on liver cell culture demonstrated an increase of cell sensitivity to insulin after treatment with imHEA-HA.


Assuntos
Enzimas Imobilizadas/uso terapêutico , Glicosaminoglicanos/química , Hipoglicemiantes/uso terapêutico , Nanotecnologia/métodos , beta-N-Acetil-Hexosaminidases/química , beta-N-Acetil-Hexosaminidases/uso terapêutico , Animais , Diabetes Mellitus/tratamento farmacológico , Hialuronoglucosaminidase/metabolismo , Hipoglicemiantes/química , Camundongos
8.
Mol Ther ; 18(8): 1519-26, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20571546

RESUMO

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.


Assuntos
Polissacarídeos/metabolismo , Doença de Sandhoff/metabolismo , Cadeia alfa da beta-Hexosaminidase/metabolismo , beta-N-Acetil-Hexosaminidases/química , beta-N-Acetil-Hexosaminidases/uso terapêutico , Animais , Células CHO , Células Cultivadas , Cromatografia em Camada Delgada , Cricetinae , Cricetulus , Gangliosídeo G(M2)/metabolismo , Glicosilação , Humanos , Immunoblotting , Camundongos , Polissacarídeos/química , Doença de Sandhoff/tratamento farmacológico , Doença de Sandhoff/genética , Cadeia alfa da beta-Hexosaminidase/química , Cadeia alfa da beta-Hexosaminidase/genética , Cadeia alfa da beta-Hexosaminidase/uso terapêutico , Cadeia beta da beta-Hexosaminidase/genética , Cadeia beta da beta-Hexosaminidase/metabolismo , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo
9.
Appl Environ Microbiol ; 73(15): 4805-12, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17557860

RESUMO

Human beta-hexosaminidase A (HexA) is a heterodimeric glycoprotein composed of alpha- and beta-subunits that degrades GM2 gangliosides in lysosomes. GM2 gangliosidosis is a lysosomal storage disease in which an inherited deficiency of HexA causes the accumulation of GM2 gangliosides. In order to prepare a large amount of HexA for a treatment based on enzyme replacement therapy (ERT), recombinant HexA was produced in the methylotrophic yeast Ogataea minuta instead of in mammalian cells, which are commonly used to produce recombinant enzymes for ERT. The problem of antigenicity due to differences in N-glycan structures between mammalian and yeast glycoproteins was potentially resolved by using alpha-1,6-mannosyltransferase-deficient (och1Delta) yeast as the host. Genes encoding the alpha- and beta-subunits of HexA were integrated into the yeast cell, and the heterodimer was expressed together with its isozymes HexS (alphaalpha) and HexB (betabeta). A total of 57 mg of beta-hexosaminidase isozymes, of which 13 mg was HexA (alphabeta), was produced per liter of medium. HexA was purified with immobilized metal affinity column for the His tag attached to the beta-subunit. The purified HexA was treated with alpha-mannosidase to expose mannose-6-phosphate (M6P) residues on the N-glycans. The specific activities of HexA and M6P-exposed HexA (M6PHexA) for the artificial substrate 4MU-GlcNAc were 1.2 +/- 0.1 and 1.7 +/- 0.3 mmol/h/mg, respectively. The sodium dodecyl sulfate-polyacrylamide gel electrophoresis pattern suggested a C-terminal truncation in the beta-subunit of the recombinant protein. M6PHexA was incorporated dose dependently into GM2 gangliosidosis patient-derived fibroblasts via M6P receptors on the cell surface, and degradation of accumulated GM2 ganglioside was observed.


Assuntos
Proteínas Recombinantes/biossíntese , Saccharomycetales/enzimologia , Doença de Sandhoff/enzimologia , Doença de Tay-Sachs/enzimologia , beta-N-Acetil-Hexosaminidases/biossíntese , Sequência de Aminoácidos , Biotecnologia/métodos , Células Cultivadas , Fibroblastos/enzimologia , Hexosaminidase A , Hexosaminidase B , Humanos , Dados de Sequência Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapêutico , Saccharomycetales/genética , Doença de Sandhoff/terapia , Doença de Tay-Sachs/terapia , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/uso terapêutico
10.
Clin Chim Acta ; 378(1-2): 38-41, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17196574

RESUMO

BACKGROUND: GM2 gangliosidoses, including Tay-Sachs disease, Sandhoff disease and the AB variant, comprise deficiencies of beta-hexosaminidase isozymes and GM2 ganglioside activator protein associated with accumulation of GM2 ganglioside (GM2) in lysosomes and neurosomatic clinical manifestations. A simple assay system for intracellular quantification of GM2 is required to evaluate the therapeutic effects on GM2-gangliosidoses. METHODS: We newly established a cell-ELISA system involving anti-GM2 monoclonal antibodies for measuring GM2 storage in fibroblasts from Tay-Sachs and Sandhoff disease patients. RESULTS: We succeeded in detecting the corrective effect of enzyme replacement on elimination of GM2 in the cells with this ELISA system. CONCLUSIONS: This simple and sensitive system should be useful as additional diagnosis tool as well as therapeutic evaluation of GM2 gangliosidoses.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Ensaio de Imunoadsorção Enzimática/métodos , Gangliosídeo G(M2)/análise , Gangliosidoses GM2/terapia , Células Cultivadas , Fibroblastos/química , Gangliosídeo G(M2)/imunologia , Hexosaminidase B , Humanos , Doença de Sandhoff/terapia , Doença de Tay-Sachs/terapia , Cadeia beta da beta-Hexosaminidase , beta-N-Acetil-Hexosaminidases/uso terapêutico
12.
Mol Med Today ; 4(4): 158-65, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9572057

RESUMO

The GM2 gangliosidoses are a group of heritable neurodegenerative disorders caused by excessive accumulation of the ganglioside GM2 owing to deficiency in beta-hexosaminidase activity. Tay-Sachs and Sandhoff diseases have similar clinical phenotypes resulting from a deficiency in human hexosaminidase alpha and beta subunits, respectively. The lack of treatment for GM2 gangliosidoses stimulated interest in developing animal models to understand the molecular mechanisms underlying the various forms of this disease and to test new potential therapies. In this review, we discuss the molecular biology of GM2 gangliosidoses and the different strategies that have been tested in animal models for the treatment of this genetic disorder, including gene transfer and cell engraftment of neural stem cells engineered to express the hexosaminidase isoenzymes.


Assuntos
Gangliosídeo G(M2)/metabolismo , Doença de Sandhoff/metabolismo , Doença de Tay-Sachs/metabolismo , beta-N-Acetil-Hexosaminidases/deficiência , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/uso terapêutico , Adolescente , Adulto , Animais , Transplante de Medula Óssea , Gatos , Transplante de Células , Criança , Modelos Animais de Doenças , Cães , Terapia Genética , Vetores Genéticos/genética , Glicolipídeos/metabolismo , HIV/genética , Humanos , Lactente , Lisossomos/enzimologia , Camundongos , Camundongos Knockout , Neurônios/transplante , Fenótipo , Mutação Puntual , Ratos , Doença de Sandhoff/genética , Doença de Sandhoff/terapia , Suínos , Doença de Tay-Sachs/etnologia , Doença de Tay-Sachs/genética , Doença de Tay-Sachs/terapia , Transplante Homólogo , beta-N-Acetil-Hexosaminidases/administração & dosagem , beta-N-Acetil-Hexosaminidases/química , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/uso terapêutico
13.
Artigo em Inglês | MEDLINE | ID: mdl-2956216

RESUMO

The therapeutic potential of enzyme replacement in lysosomal storage disorders has remained largely unfulfilled, perhaps because of negative reactions to the initial disappointing results. Despite the existence of several animal models that can be utilized to explore solutions to the problems of exogenous enzyme targeting, the interest in ERT prevalent during the 1970's seems to have subsided to be replaced by active interest in bone marrow transplantation (BMT, Krivit and Paul [1986]). This is a logical approach to enzyme replacement in storage disorders of the RE system, and indeed some encouraging results have been obtained. However, in addition to having high morbidity and mortality, in the ultimate analysis BMT presents the same targeting problems as conventional ERT. In our opinion, these problems can be solved more easily in the case of ERT by exploiting the existing cellular uptake mechanisms and infusing enzymes whose structure has been suitably modified by simple biochemical manipulations. Accordingly, we have explored a methodology that takes advantage of negative charges on the cell surface to obtain nonspecific but effective membrane binding of beta-hex coupled to the highly positively charged PLL, followed by internalization and routing to the lysosomes. This system increases uptake of exogenous enzyme by some neurons in vitro and possibly in vivo, but its efficiency depends on the cells' endocytic activity that, in the case of neuronal soma, apparently is low. Thus, we have chosen as recognition marker for specific neuronal uptake a nontoxic fragment of TTx that is efficiently taken up by these cells. The initial results are encouraging; they support our contention that effective enzyme replacement methodologies can be devised, and encourage us to continue our work in this direction. Finally, recombinant DNA techniques are now being applied to a number of LSD, and the genes for several of the pertinent enzymes have been or are being isolated. In addition to representing a first step towards gene replacement therapy, the results of this work will permit the generation of large amounts of human enzymes from bacteria by recombinant DNA methods, thus obviating the problem of enzyme supply for ERT. Since human lysosomal enzymes obtained from bacteria will be nonglycosylated, to obtain cell uptake it will be necessary to resort to the type of modifications that we are trying to develop at this time, i.e., covalent linkage to moieties that allow non-glycosyl-mediated cellular uptake. Thus, our work on beta-hex may provide a model for biochemical manipulations of bacterially produced enzymes applicable to several LSD.


Assuntos
Gangliosidoses/terapia , beta-N-Acetil-Hexosaminidases/uso terapêutico , Células Cultivadas , Humanos , Lisossomos/enzimologia , Neurônios/metabolismo , Toxina Tetânica , beta-N-Acetil-Hexosaminidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...