Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Mol Ther ; 29(1): 3, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33321097
2.
PLoS One ; 11(9): e0162145, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27657498

RESUMO

The stepwise degradation of glycosaminoglycans (GAGs) is accomplished by twelve lysosomal enzymes. Deficiency in any of these enzymes will result in the accumulation of the intermediate substrates on the pathway to the complete turnover of GAGs. The accumulation of these undegraded substrates in almost any tissue is a hallmark of all Mucopolysaccharidoses (MPS). Present therapeutics based on enzyme replacement therapy and bone marrow transplantation have low effectiveness for the treatment of MPS with neurological complications since enzymes used in these therapies are unable to cross the blood brain barrier. Small molecule-based approaches are more promising in addressing neurological manifestations. In this report we identify a target for developing a substrate reduction therapy (SRT) for six MPS resulting from the abnormal degradation of heparan sulfate (HS). Using the minimal promoter of NDST1, one of the first modifying enzymes of HS precursors, we established a luciferase based reporter gene assay capable of identifying small molecules that could potentially reduce HS maturation and therefore lessen HS accumulation in certain MPS. From the screen of 1,200 compounds comprising the Prestwick Chemical library we identified SAHA, a histone deacetylase inhibitor, as the drug that produced the highest inhibitory effects in the reporter assay. More importantly SAHA treated fibroblasts expressed lower levels of endogenous NDST1 and accumulated less 35S GAGs in patient cells. Thus, by using our simple reporter gene assay we have demonstrated that by inhibiting the transcription of NDST1 with small molecules, identified by high throughput screening, we can also reduce the level of sulfated HS substrate in MPS patient cells, potentially leading to SRT.

3.
Hum Gene Ther ; 27(7): 509-21, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27197548

RESUMO

GM2 gangliosidosis is a family of three genetic neurodegenerative disorders caused by the accumulation of GM2 ganglioside (GM2) in neuronal tissue. Two of these are due to the deficiency of the heterodimeric (α-ß), "A" isoenzyme of lysosomal ß-hexosaminidase (HexA). Mutations in the α-subunit (encoded by HEXA) lead to Tay-Sachs disease (TSD), whereas mutations in the ß-subunit (encoded by HEXB) lead to Sandhoff disease (SD). The third form results from a deficiency of the GM2 activator protein (GM2AP), a substrate-specific cofactor for HexA. In their infantile, acute forms, these diseases rapidly progress with mental and psychomotor deterioration resulting in death by approximately 4 years of age. After gene transfer that overexpresses one of the deficient subunits, the amount of HexA heterodimer formed would empirically be limited by the availability of the other endogenous Hex subunit. The present study used a new variant of the human HexA α-subunit, µ, incorporating critical sequences from the ß-subunit that produce a stable homodimer (HexM) and promote functional interactions with the GM2AP- GM2 complex. We report the design of a compact adeno-associated viral (AAV) genome using a synthetic promoter-intron combination to allow self-complementary (sc) packaging of the HEXM gene. Also, a previously published capsid mutant, AAV9.47, was used to deliver the gene to brain and spinal cord while having restricted biodistribution to the liver. The novel capsid and cassette design combination was characterized in vivo in TSD mice for its ability to efficiently transduce cells in the central nervous system when delivered intravenously in both adult and neonatal mice. This study demonstrates that the modified HexM is capable of degrading long-standing GM2 storage in mice, and it further demonstrates the potential of this novel scAAV vector design to facilitate widespread distribution of the HEXM gene or potentially other similar-sized genes to the nervous system.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Hexosaminidases/genética , Mutação/genética , Doença de Tay-Sachs/terapia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Gangliosídeo G(M2)/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Doença de Tay-Sachs/genética
4.
Hum Gene Ther ; 27(7): 497-508, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27199088

RESUMO

GM2 gangliosidosis is a group of neurodegenerative diseases caused by ß-hexosaminidase A (HexA) enzyme deficiency. There is currently no cure. HexA is composed of two similar, nonidentical subunits, α and ß, which must interact with the GM2 activator protein (GM2AP), a substrate-specific cofactor, to hydrolyze GM2 ganglioside. Mutations in either subunit or the activator can result in the accumulation of GM2 ganglioside within neurons throughout the central nervous system. The resulting neuronal cell death induces the primary symptoms of the disease: motor impairment, seizures, and sensory impairments. This study assesses the long-term effects of gene transfer in a Sandhoff (ß-subunit knockout) mouse model. The study utilized a modified human ß-hexosaminidase α-subunit (µ-subunit) that contains critical sequences from the ß-subunit that enables formation of a stable homodimer (HexM) and interaction with GM2AP to hydrolyze GM2 ganglioside. We investigated a self-complementary adeno-associated viral (scAAV) vector expressing HexM, through intravenous injections of the neonatal mice. We monitored one cohort for 8 weeks and another cohort long-term for survival benefit, behavioral, biochemical, and molecular analyses. Untreated Sandhoff disease (SD) control mice reached a humane endpoint at approximately 15 weeks, whereas treated mice had a median survival age of 40 weeks, an approximate 2.5-fold survival advantage. On behavioral tests, the treated mice outperformed their knockout age-matched controls and perform similarly to the heterozygous controls. Through the enzymatic and GM2 ganglioside analyses, we observed a significant decrease in the GM2 ganglioside level, even though the enzyme levels were not significantly increased. Molecular analyses revealed a global distribution of the vector between brain and spinal cord regions. In conclusion, the neonatal delivery of a novel viral vector expressing the human HexM enzyme is effective in ameliorating the SD mouse phenotype for long-term. Our data could have implications not only for treatment of SD but also for Tay-Sachs disease (α-subunit deficiency) and similar brain disorders.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Hexosaminidases/genética , Mutação/genética , Doença de Sandhoff/terapia , Animais , Animais Recém-Nascidos , Comportamento Animal , Modelos Animais de Doenças , Camundongos , Fenótipo , Doença de Sandhoff/enzimologia , Doença de Sandhoff/genética
5.
Mol Ther Methods Clin Dev ; 3: 15057, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26966698

RESUMO

Tay-Sachs or Sandhoff disease result from mutations in either the evolutionarily related HEXA or HEXB genes encoding respectively, the α- or ß-subunits of ß-hexosaminidase A (HexA). Of the three Hex isozymes, only HexA can interact with its cofactor, the GM2 activator protein (GM2AP), and hydrolyze GM2 ganglioside. A major impediment to establishing gene or enzyme replacement therapy based on HexA is the need to synthesize both subunits. Thus, we combined the critical features of both α- and ß-subunits into a single hybrid µ-subunit that contains the α-subunit active site, the stable ß-subunit interface and unique areas in each subunit needed to interact with GM2AP. To facilitate intracellular analysis and the purification of the µ-homodimer (HexM), CRISPR-based genome editing was used to disrupt the HEXA and HEXB genes in a Human Embryonic Kidney 293 cell line stably expressing the µ-subunit. In association with GM2AP, HexM was shown to hydrolyze a fluorescent GM2 ganglioside derivative both in cellulo and in vitro. Gene transfer studies in both Tay-Sachs and Sandhoff mouse models demonstrated that HexM expression reduced brain GM2 ganglioside levels.

6.
J Med Chem ; 58(11): 4483-93, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-25984755

RESUMO

In order to identify structural features of pyrimethamine (5-(4-chlorophenyl)-6-ethylpyrimidine-2,4-diamine) that contribute to its inhibitory activity (IC50 value) and chaperoning efficacy toward ß-N-acetylhexosaminidase, derivatives of the compound were synthesized that differ at the positions bearing the amino, ethyl, and chloro groups. Whereas the amino groups proved to be critical to its inhibitory activity, a variety of substitutions at the chloro position only increased its IC50 by 2-3-fold. Replacing the ethyl group at the 6-position with butyl or methyl groups increased IC50 more than 10-fold. Surprisingly, despite its higher IC50, a derivative lacking the chlorine atom in the para-position was found to enhance enzyme activity in live patient cells a further 25% at concentrations >100 µM, while showing less toxicity. These findings demonstrate the importance of the phenyl group in modulating the chaperoning efficacy and toxicity profile of the derivatives.


Assuntos
Proteínas Mutantes/metabolismo , Mutação/genética , Pirimetamina/química , Pirimetamina/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/patologia , Humanos , Modelos Moleculares , Estrutura Molecular , Proteínas Mutantes/genética , Relação Estrutura-Atividade
7.
Ann Clin Transl Neurol ; 1(6): 379-389, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24999486

RESUMO

OBJECTIVE: Hereditary spastic paraplegias (HSPs) are among the most genetically diverse inherited neurological disorders, with over 70 disease loci identified (SPG1-71) to date. SPG15 and SPG11 are clinically similar, autosomal recessive disorders characterized by progressive spastic paraplegia along with thin corpus callosum, white matter abnormalities, cognitive impairment, and ophthalmologic abnormalities. Furthermore, both have been linked to early-onset parkinsonism. METHODS: We describe two new cases of SPG15 and investigate cellular changes in SPG15 and SPG11 patient-derived fibroblasts, seeking to identify shared pathogenic themes. Cells were evaluated for any abnormalities in cell division, DNA repair, endoplasmic reticulum, endosomes, and lysosomes. RESULTS: Fibroblasts prepared from patients with SPG15 have selective enlargement of LAMP1-positive structures, and they consistently exhibited abnormal lysosomal storage by electron microscopy. A similar enlargement of LAMP1-positive structures was also observed in cells from multiple SPG11 patients, though prominent abnormal lysosomal storage was not evident. The stabilities of the SPG15 protein spastizin/ZFYVE26 and the SPG11 protein spatacsin were interdependent. INTERPRETATION: Emerging studies implicating these two proteins in interactions with the late endosomal/lysosomal adaptor protein complex AP-5 are consistent with shared abnormalities in lysosomes, supporting a converging mechanism for these two disorders. Recent work with Zfyve26-/- mice revealed a similar phenotype to human SPG15, and cells in these mice had endolysosomal abnormalities. SPG15 and SPG11 are particularly notable among HSPs because they can also present with juvenile parkinsonism, and this lysosomal trafficking or storage defect may be relevant for other forms of parkinsonism associated with lysosomal dysfunction.

8.
Anal Biochem ; 458: 20-6, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24769373

RESUMO

GM2 gangliosidosis is a group of inherited neurodegenerative disorders resulting primarily from the excessive accumulation of GM2 gangliosides (GM2) in neuronal cells. As biomarkers for categorising patients and monitoring the effectiveness of developing therapies are lacking for this group of disorders, we sought to develop methodology to quantify GM2 levels in more readily attainable patient samples such as plasma, leukocytes, and cultured skin fibroblasts. Following organic extraction, gangliosides were partitioned into the aqueous phase and isolated using C18 solid-phase extraction columns. Relative quantification of three species of GM2 was achieved using LC/ESI-MS/MS with d35GM1 18:1/18:0 as an internal standard. The assay was linear over the biological range, and all GM2 gangliosidosis patients were demarcated from controls by elevated GM2 in cultured skin fibroblast extracts. However, in leukocytes only some molecular species could be used for differentiation and in plasma only one was informative. A reduction in GM2 was easily detected in patient skin fibroblasts after a short treatment with media from normal cells enriched in secreted ß-hexosaminidase. This method may show promise for measuring the effectiveness of experimental therapies for GM2 gangliosidosis by allowing quantification of a reduction in the primary storage burden.


Assuntos
Cromatografia Líquida de Alta Pressão , Gangliosídeo G(M2)/análise , Espectrometria de Massas em Tandem , Linhagem Celular , Fibroblastos/química , Gangliosídeo G(M2)/sangue , Gangliosídeo G(M2)/isolamento & purificação , Humanos , Leucócitos/química , Extração em Fase Sólida , Espectrometria de Massas por Ionização por Electrospray , beta-N-Acetil-Hexosaminidases/metabolismo
9.
J Org Chem ; 79(10): 4398-404, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24735108

RESUMO

We report herein a newly developed domino reaction that facilitates the synthesis of new 1,5-dideoxy-1,5-iminoribitol iminosugar C-glycosides 7a-e and 8. The key intermediate in this approach is a six-membered cyclic sugar nitrone that is generated in situ and trapped by an alkene dipolarophile via a [2 + 3] cycloaddition reaction to give the corresponding isooxazolidines 10a-e in a "one-pot" protocol. The iminoribitol C-glycosides 7a-e and 8 were found to be modest ß-galactosidase (bGal) inhibitors. However, compounds 7c and 7e showed "pharmacological chaperone" activity for mutant lysosomal bGal activity and facilitated its recovery in GM1 gangliosidosis patient fibroblasts by 2-6-fold.


Assuntos
Alcenos/química , Fibroblastos/química , Gangliosidose GM1/tratamento farmacológico , Lisossomos/química , Chaperonas Moleculares/farmacologia , Chaperonas Moleculares/uso terapêutico , Monossacarídeos/síntese química , Óxidos de Nitrogênio/química , beta-Galactosidase/antagonistas & inibidores , beta-Galactosidase/química , Reação de Cicloadição , Gangliosidose GM1/enzimologia , Gangliosidose GM1/metabolismo , Glicosídeos , Humanos , Lisossomos/metabolismo , Monossacarídeos/química
10.
PLoS One ; 8(6): e66139, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23776622

RESUMO

Sidt2 was identified as a novel integral lysosomal membrane protein recently. We generated global Sidt2 knockout mice by gene targeting. These mice have a comparatively higher random and fasting glucose concentration. Intraperitoneal and oral glucose tolerance tests in Sidt2 knockout mice indicated glucose intolerance and decreased serum insulin level. Notably, the Sidt2(-/-) mice had hypertrophic islets compared with control mice. By Western blot and immunofluorescence, Sidt2(-/-) mouse islets were shown to have increased insulin protein, which actually contained more insulin secretory granules than their controls, demonstrated by electromicroscopy. Consistent with the in vivo study, isolated islet culture from the Sidt2(-/-) mice produced less insulin when stimulated by a high concentration of glucose or a depolarizing concentration of KCl. Under electromicroscope less empty vesicles and more mature ones in Sidt2(-/-) mice islets were observed, supporting impaired insulin secretory granule release. In conclusion, Sidt2 may play a critical role in the regulation of mouse insulin secretory granule secretion.


Assuntos
Intolerância à Glucose/metabolismo , Insulina/metabolismo , Proteínas de Membrana/deficiência , Animais , Western Blotting , Imunofluorescência , Intolerância à Glucose/genética , Ilhotas Pancreáticas/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas de Transporte de Nucleotídeos , Vesículas Secretórias/metabolismo
11.
PLoS One ; 8(3): e57908, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23483939

RESUMO

The hydrolysis in lysosomes of GM2 ganglioside to GM3 ganglioside requires the correct synthesis, intracellular assembly and transport of three separate gene products; i.e., the alpha and beta subunits of heterodimeric beta-hexosaminidase A, E.C. # 3.2.1.52 (encoded by the HEXA and HEXB genes, respectively), and the GM2-activator protein (GM2AP, encoded by the GM2A gene). Mutations in any one of these genes can result in one of three neurodegenerative diseases collectively known as GM2 gangliosidosis (HEXA, Tay-Sachs disease, MIM # 272800; HEXB, Sandhoff disease, MIM # 268800; and GM2A, AB-variant form, MIM # 272750). Elements of both of the hexosaminidase A subunits are needed to productively interact with the GM2 ganglioside-GM2AP complex in the lysosome. Some of these elements have been predicted from the crystal structures of hexosaminidase and the activator. Recently a hybrid of the two subunits has been constructed and reported to be capable of forming homodimers that can perform this reaction in vivo, which could greatly simplify vector-mediated gene transfer approaches for Tay-Sachs or Sandhoff diseases. A cDNA encoding a hybrid hexosaminidase subunit capable of dimerizing and hydrolyzing GM2 ganglioside could be incorporated into a single vector, whereas packaging both subunits of hexosaminidase A into vectors, such as adeno-associated virus, would be impractical due to size constraints. In this report we examine the previously published hybrid construct (H1) and a new more extensive hybrid (H2), with our documented in cellulo (live cell- based) assay utilizing a fluorescent GM2 ganglioside derivative. Unfortunately when Tay-Sachs cells were transfected with either the H1 or H2 hybrid construct and then were fed the GM2 derivative, no significant increase in its turnover was detected. In vitro assays with the isolated H1 or H2 homodimers confirmed that neither was capable of human GM2AP-dependent hydrolysis of GM2 ganglioside.


Assuntos
Proteína Ativadora de G(M2)/metabolismo , Gangliosídeo G(M2)/metabolismo , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Substituição de Aminoácidos/genética , Animais , Western Blotting , Gatos , Cromatografia por Troca Iônica , Cromatografia em Camada Fina , Humanos , Hidrólise , Camundongos , Ligação Proteica , Doença de Sandhoff/metabolismo , Doença de Sandhoff/patologia , Doença de Tay-Sachs/metabolismo , Doença de Tay-Sachs/patologia , Transfecção
12.
Acta Neuropathol ; 125(3): 439-57, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23315026

RESUMO

X-linked Myopathy with Excessive Autophagy (XMEA) is a childhood onset disease characterized by progressive vacuolation and atrophy of skeletal muscle. We show that XMEA is caused by hypomorphic alleles of the VMA21 gene, that VMA21 is the diverged human ortholog of the yeast Vma21p protein, and that like Vma21p, VMA21 is an essential assembly chaperone of the vacuolar ATPase (V-ATPase), the principal mammalian proton pump complex. Decreased VMA21 raises lysosomal pH which reduces lysosomal degradative ability and blocks autophagy. This reduces cellular free amino acids which leads to downregulation of the mTORC1 pathway, and consequent increased macroautophagy resulting in proliferation of large and ineffective autolysosomes that engulf sections of cytoplasm, merge, and vacuolate the cell. Our results uncover a novel mechanism of disease, namely macroautophagic overcompensation leading to cell vacuolation and tissue atrophy.


Assuntos
Adenosina Trifosfatases/metabolismo , Autofagia/genética , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/prevenção & controle , Doenças Musculares/genética , Doenças Musculares/prevenção & controle , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética , Animais , Células Cultivadas , Humanos , Concentração de Íons de Hidrogênio , Leucina/metabolismo , Doenças por Armazenamento dos Lisossomos/patologia , Lisossomos/genética , Lisossomos/metabolismo , Masculino , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Doenças Musculares/patologia , Mutação/genética , Interferência de RNA/fisiologia , RNA Mensageiro/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Frações Subcelulares/metabolismo , Frações Subcelulares/patologia , Fatores de Tempo , Vacúolos/metabolismo
13.
Mol Genet Metab ; 108(1): 65-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23158871

RESUMO

A 12 year-old female presented with a seven-year history of progressive muscle weakness, atrophy, tremor and fasciculations. Cognition was normal. Rectal biopsy revealed intracellular storage material and biochemical testing indicated low hexosaminidase activity consistent with juvenile-onset G(M2)-gangliosidosis. Genetic evaluation revealed compound heterozygosity with two novel mutations in the hexosaminidase ß-subunit (c.512-3 C>A and c.1613+15_1613+18dup). Protein analysis was consistent with biochemical findings and indicated only a small portion of ß-subunits were properly processed. These results provide additional insight into juvenile-onset G(M2)-gangliosidoses and further expand the number of ß-hexosaminidase mutations associated with motor neuron disease.


Assuntos
Doença dos Neurônios Motores/genética , Mutação , beta-N-Acetil-Hexosaminidases/genética , Idade de Início , Criança , Feminino , Humanos , Doença dos Neurônios Motores/psicologia
14.
Mol Genet Metab ; 107(1-2): 203-12, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22784478

RESUMO

Deficiencies of lysosomal ß-D-galactosidase can result in GM1 gangliosidosis, a severe neurodegenerative disease characterized by massive neuronal storage of GM1 ganglioside in the brain. Currently there are no available therapies that can even slow the progression of this disease. Enzyme enhancement therapy utilizes small molecules that can often cross the blood brain barrier, but are also often competitive inhibitors of their target enzyme. It is a promising new approach for treating diseases, often caused by missense mutations, associated with dramatically reduced levels of functionally folded enzyme. Despite a number of positive reports based on assays performed with patient cells, skepticism persists that an inhibitor-based treatment can increase mutant enzyme activity in vivo. To date no appropriate animal model, i.e., one that recapitulates a responsive human genotype and clinical phenotype, has been reported that could be used to validate enzyme enhancement therapy. In this report, we identify a novel enzyme enhancement-agent, N-nonyl-deoxygalactonojirimycin, that enhances the mutant ß-galactosidase activity in the lysosomes of a number of patient cell lines containing a variety of missense mutations. We then demonstrate that treatment of cells from a previously described, naturally occurring feline model (that biochemically, clinically and molecularly closely mimics GM1 gangliosidosis in humans) with this molecule, results in a robust enhancement of their mutant lysosomal ß-galactosidase activity. These data indicate that the feline model could be used to validate this therapeutic approach and determine the relationship between the disease stage at which this therapy is initiated and the maximum clinical benefits obtainable.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Terapia de Reposição de Enzimas , Gangliosidose GM1/metabolismo , Proteínas Mutantes/metabolismo , beta-Galactosidase/metabolismo , 1-Desoxinojirimicina/administração & dosagem , 1-Desoxinojirimicina/farmacologia , Animais , Gatos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Gangliosidose GM1/tratamento farmacológico , Gangliosidose GM1/genética , Temperatura Alta , Humanos , Concentração de Íons de Hidrogênio , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/química , Mutação , Desnaturação Proteica/efeitos dos fármacos , Resultado do Tratamento , beta-Galactosidase/antagonistas & inibidores , beta-Galactosidase/química
15.
Mol Genet Metab ; 106(3): 323-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22592100

RESUMO

Gaucher disease is a prevalent lysosomal storage disease in which affected individuals inherit mutations in the gene (GBA1) encoding lysosomal acid ß-glucosidase (glucocerebrosidase, GCase, EC 3.2.1.45). One of the most prevalent disease-causing mutations in humans is a N370S missense mutation in the GCase protein. As part of a larger endeavor to study the fate of mutant human proteins expressed in plant cells, the N370S mutant protein along with the wild-type- (WT)-GCase, both equipped with a signal peptide, were synthesized in transgenic tobacco BY2 cells, which do not possess lysosomes. The enzymatic activity of plant-recombinant N370S GCase lines was significantly lower (by 81-95%) than that of the WT-GCase lines. In contrast to the WT-GCase protein, which was efficiently secreted from tobacco BY2 cells, and detected in large amounts in the culture medium, only a small proportion of the N370S GCase was secreted. Pharmacological chaperones such as N-(n-nonyl) deoxynojirimycin and ambroxol increased the steady-state mutant protein levels both inside the plant cells and in the culture medium. These findings contradict the assertion that small molecule chaperones increase N370S GCase activity (as assayed in treated patient cell lysates) by stabilizing the enzyme in the lysosome, and suggest that the mutant protein is impaired in its ability to obtain its functional folded conformation, which is a requirement for exiting the lumen of the ER.


Assuntos
Retículo Endoplasmático/metabolismo , Glucosilceramidase/genética , Chaperonas Moleculares/metabolismo , Transporte Biológico , Domínio Catalítico , Células Cultivadas , Doença de Gaucher/enzimologia , Doença de Gaucher/genética , Glucosilceramidase/metabolismo , Humanos , Chaperonas Moleculares/genética , Mutação , Células Vegetais/metabolismo , Plantas Geneticamente Modificadas , Dobramento de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
J Med Chem ; 55(6): 2737-45, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-22360565

RESUMO

A highly divergent route to lipophilic iminosugars that utilizes the thiol-ene reaction was developed to enable the rapid synthesis of a collection of 16 dideoxyiminoxylitols bearing various different lipophilic substituents. Enzyme kinetic analyses revealed that a number of these products are potent, low-nanomolar inhibitors of human glucocerebrosidase that stabilize the enzyme to thermal denaturation by up to 20 K. Cell based assays conducted on Gaucher disease patient derived fibroblasts demonstrated that administration of the compounds can increase lysosomal glucocerebrosidase activity levels by therapeutically relevant amounts, as much as 3.2-fold in cells homozygous for the p.N370S mutation and 1.4-fold in cells homozygous for the p.L444P mutation. Several compounds elicited this increase in enzyme activity over a relatively wide dosage range. The data assembled here illustrate how the lipophilic moiety common to many glucocerebrosidase inhibitors might be used to optimize a lead compound's ability to chaperone the protein in cellulo. The flexibility of this synthetic strategy makes it an attractive approach to the rapid optimization of glycosidase inhibitor potency and pharmacokinetic behavior.


Assuntos
Alilamina/análogos & derivados , Alilamina/síntese química , Carboidratos/síntese química , Doença de Gaucher/tratamento farmacológico , Glucosilceramidase/antagonistas & inibidores , Iminas/síntese química , Xilitol/análogos & derivados , Xilitol/síntese química , Alilamina/farmacologia , Carboidratos/farmacologia , Ensaios Enzimáticos , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/patologia , Doença de Gaucher/enzimologia , Doença de Gaucher/patologia , Glucosilceramidase/genética , Humanos , Iminas/farmacologia , Isomerismo , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Mutação , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade , Xilitol/farmacologia
17.
Glycobiology ; 22(4): 492-503, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22061999

RESUMO

There is a clear need for efficient methods to produce protein therapeutics requiring mannose-termination for therapeutic efficacy. Here we report on a unique system for production of active human lysosomal acid ß-glucosidase (glucocerebrosidase, GCase, EC 3.2.1.45) using seeds of the Arabidopsis thaliana complex-glycan-deficient (cgl) mutant, which are deficient in the activity of N-acetylglucosaminyl transferase I (EC 2.4.1.101). Gaucher disease is a prevalent lysosomal storage disease in which affected individuals inherit mutations in the gene (GBA1) encoding GCase. A gene cassette optimized for seed expression was used to generate the human enzyme in seeds of the cgl (C5) mutant, and the recombinant GCase was mainly accumulated in the apoplast. Importantly, the enzymatic properties including kinetic parameters, half-maximal inhibitory concentration of isofagomine and thermal stability of the cgl-derived GCase were comparable with those of imiglucerase, a commercially available recombinant human GCase used for enzyme replacement therapy in Gaucher patients. N-glycan structural analyses of recombinant cgl-GCase showed that the majority of the N-glycans (97%) were mannose terminated. Additional purification was required to remove ∼15% of the plant-derived recombinant GCase that possessed potentially immunogenic (xylose- and/or fucose-containing) N-glycans. Uptake of cgl-derived GCase by mouse macrophages was similar to that of imiglucerase. The cgl seed system requires no addition of foreign (non-native) amino acids to the mature recombinant GCase protein, and the dry transgenic seeds represent a stable repository of the therapeutic protein. Other strategies that may completely prevent plant-like complex N-glycans are discussed, including the use of a null cgl mutant.


Assuntos
Arabidopsis/genética , Glucosilceramidase/biossíntese , Plantas Geneticamente Modificadas , Sementes/genética , Animais , Arabidopsis/citologia , Configuração de Carboidratos , Sequência de Carboidratos , Células Cultivadas , Estabilidade Enzimática , Glucosilceramidase/isolamento & purificação , Glucosilceramidase/metabolismo , Glicosilação , Humanos , Cinética , Macrófagos/metabolismo , Manose , Camundongos , Dados de Sequência Molecular , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Sementes/citologia
18.
PLoS One ; 6(9): e24951, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21957468

RESUMO

Heparin acetyl-CoA:alpha-glucosaminide N-acetyltransferase (N-acetyltransferase, EC 2.3.1.78) is an integral lysosomal membrane protein containing 11 transmembrane domains, encoded by the HGSNAT gene. Deficiencies of N-acetyltransferase lead to mucopolysaccharidosis IIIC. We demonstrate that contrary to a previous report, the N-acetyltransferase signal peptide is co-translationally cleaved and that this event is required for its intracellular transport to the lysosome. While we confirm that the N-acetyltransferase precursor polypeptide is processed in the lysosome into a small amino-terminal alpha- and a larger ß- chain, we further characterize this event by identifying the mature amino-terminus of each chain. We also demonstrate this processing step(s) is not, as previously reported, needed to produce a functional transferase, i.e., the precursor is active. We next optimize the biochemical assay procedure so that it remains linear as N-acetyltransferase is purified or protein-extracts containing N-acetyltransferase are diluted, by the inclusion of negatively charged lipids. We then use this assay to demonstrate that the purified single N-acetyltransferase protein is both necessary and sufficient to express transferase activity, and that N-acetyltransferase functions as a monomer. Finally, the kinetic mechanism of action of purified N-acetyltransferase was evaluated and found to be a random sequential mechanism involving the formation of a ternary complex with its two substrates; i.e., N-acetyltransferase does not operate through a ping-pong mechanism as previously reported. We confirm this conclusion by demonstrating experimentally that no acetylated enzyme intermediate is formed during the reaction.


Assuntos
Acetiltransferases/biossíntese , Acetiltransferases/metabolismo , Mucopolissacaridose III/enzimologia , Processamento de Proteína Pós-Traducional , Acetiltransferases/deficiência , Acetiltransferases/isolamento & purificação , Western Blotting , Detergentes/química , Retículo Endoplasmático/metabolismo , Ensaios Enzimáticos , Células HeLa , Humanos , Interações Hidrofóbicas e Hidrofílicas , Cinética , Transporte Proteico , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...