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

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cancer Gene Ther ; 31(1): 158-173, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37990063

RESUMEN

MSCs (mesenchymal stem cells), responsible for tissue repair, rarely undergo cell fusion with somatic cells. Here, we show that ~5% of bladder cancer cells (UMUC-3) fuses with bone marrow-derived MSC (BM-MSC) in co-culture and maintains high tumorigenicity. In eleven fusion cell clones that have been established, Mb-scale deletions carried by the bladder cancer cells are mostly absent in the fusion cells, but copy number gains contributed by the cancer cells have stayed. Fusion cells exhibit increased populations of mitotic cells with 3-polar spindles, indicative of genomic instability. They grow faster in vitro and exhibit higher colony formation in anchorage-independent growth assay in soft agar than the parent UMUC-3 does. Fusion cells develop tumors, after 4 weeks of time lag, as efficiently as the parent UMUC-3 does in xenograft experiments. 264 genes are identified whose expression is specifically altered in the fusion cells. Many of them are interferon-stimulated genes (ISG), but are activated in a manner independent of interferon. Among them, we show that PD-L1 is induced in fusion cells, and its knockout decreases tumorigenesis in a xenograft model. PD-L1 is induced in a manner independent of STAT1 known to regulate PD-L1 expression, but is regulated by histone modification, and is likely to inhibit phagocytosis by PD1-expressing macrophages, thus protecting cancer cells from immunological attacks. The fusion cells overexpress multiple cytokines including CCL2 that cause tumor progression by converting infiltrating macrophages to tumor-associated-macrophage (TAM). The results present mechanisms of how cell fusion promotes tumorigenesis, revealing a novel link between cell fusion and PD-L1, and underscore the efficacy of cancer immunotherapy.


Asunto(s)
Antígeno B7-H1 , Neoplasias de la Vejiga Urinaria , Humanos , Fusión Celular , Monitorización Inmunológica , Interferones , Carcinogénesis , Línea Celular Tumoral
2.
Chem Biomed Imaging ; 1(2): 157-167, 2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37235189

RESUMEN

With the aim of developing more stable Gd(III)-porphyrin complexes, two types of ligands 1 and 2 with carboxylic acid anchors were synthesized. Due to the N-substituted pyridyl cation attached to the porphyrin core, these porphyrin ligands were highly water-soluble and formed the corresponding Gd(III) chelates, Gd-1 and Gd-2. Gd-1 was sufficiently stable in neutral buffer, presumably due to the preferred conformation of the carboxylate-terminated anchors connected to nitrogen in the meta position of the pyridyl group helping to stabilize Gd(III) complexation by the porphyrin center. 1H NMRD (nuclear magnetic relaxation dispersion) measurements on Gd-1 revealed high longitudinal water proton relaxivity (r1 = 21.2 mM-1 s-1 at 60 MHz and 25 °C), which originates from slow rotational motion resulting from aggregation in aqueous solution. Under visible light irradiation, Gd-1 showed extensive photoinduced DNA cleavage in line with efficient photoinduced singlet oxygen generation. Cell-based assays revealed no significant dark cytotoxicity of Gd-1, while it showed sufficient photocytotoxicity on cancer cell lines under visible light irradiation. These results indicate the potential of this Gd(III)-porphyrin complex (Gd-1) as a core for the development of bifunctional systems acting as an efficient photodynamic therapy photosensitizer (PDT-PS) with magnetic resonance imaging (MRI) detection capabilities.

3.
Am J Hum Genet ; 85(5): 569-80, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19853240

RESUMEN

A modified alpha-N-acetylgalactosaminidase (NAGA) with alpha-galactosidase A (GLA)-like substrate specificity was designed on the basis of structural studies and was produced in Chinese hamster ovary cells. The enzyme acquired the ability to catalyze the degradation of 4-methylumbelliferyl-alpha-D-galactopyranoside. It retained the original NAGA's stability in plasma and N-glycans containing many mannose 6-phosphate (M6P) residues, which are advantageous for uptake by cells via M6P receptors. There was no immunological cross-reactivity between the modified NAGA and GLA, and the modified NAGA did not react to serum from a patient with Fabry disease recurrently treated with a recombinant GLA. The enzyme cleaved globotriaosylceramide (Gb3) accumulated in cultured fibroblasts from a patient with Fabry disease. Furthermore, like recombinant GLA proteins presently used for enzyme replacement therapy (ERT) for Fabry disease, the enzyme intravenously injected into Fabry model mice prevented Gb3 storage in the liver, kidneys, and heart and improved the pathological changes in these organs. Because this modified NAGA is hardly expected to cause an allergic reaction in Fabry disease patients, it is highly promising as a new and safe enzyme for ERT for Fabry disease.


Asunto(s)
Terapia de Reemplazo Enzimático/métodos , Enfermedad de Fabry/tratamiento farmacológico , alfa-N-Acetilgalactosaminidasa/química , alfa-N-Acetilgalactosaminidasa/uso terapéutico , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células CHO , Catálisis , Células Cultivadas , Cricetinae , Cricetulus , Medios de Cultivo Condicionados/química , ADN Complementario/metabolismo , Modelos Animales de Enfermedad , Estabilidad de Medicamentos , Enfermedad de Fabry/enzimología , Enfermedad de Fabry/metabolismo , Fibroblastos/efectos de los fármacos , Colorantes Fluorescentes/metabolismo , Galactósidos/metabolismo , Vectores Genéticos , Humanos , Concentración de Iones de Hidrógeno , Himecromona/análogos & derivados , Himecromona/metabolismo , Inmunohistoquímica , Riñón/efectos de los fármacos , Riñón/patología , Riñón/ultraestructura , Hígado/efectos de los fármacos , Hígado/patología , Hígado/ultraestructura , Ratones , Ratones Noqueados , Modelos Moleculares , Peso Molecular , Miocardio/patología , Miocardio/ultraestructura , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/uso terapéutico , Retroviridae/genética , Transfección , Trihexosilceramidas/metabolismo , alfa-N-Acetilgalactosaminidasa/genética , alfa-N-Acetilgalactosaminidasa/aislamiento & purificación
4.
Hum Mol Genet ; 18(4): 621-31, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19017726

RESUMEN

Hypoglycosylation and reduced laminin-binding activity of alpha-dystroglycan are common characteristics of dystroglycanopathy, which is a group of congenital and limb-girdle muscular dystrophies. Fukuyama-type congenital muscular dystrophy (FCMD), caused by a mutation in the fukutin gene, is a severe form of dystroglycanopathy. A retrotransposal insertion in fukutin is seen in almost all cases of FCMD. To better understand the molecular pathogenesis of dystroglycanopathies and to explore therapeutic strategies, we generated knock-in mice carrying the retrotransposal insertion in the mouse fukutin ortholog. Knock-in mice exhibited hypoglycosylated alpha-dystroglycan; however, no signs of muscular dystrophy were observed. More sensitive methods detected minor levels of intact alpha-dystroglycan, and solid-phase assays determined laminin binding levels to be approximately 50% of normal. In contrast, intact alpha-dystroglycan is undetectable in the dystrophic Large(myd) mouse, and laminin-binding activity is markedly reduced. These data indicate that a small amount of intact alpha-dystroglycan is sufficient to maintain muscle cell integrity in knock-in mice, suggesting that the treatment of dystroglycanopathies might not require the full recovery of glycosylation. To examine whether glycosylation defects can be restored in vivo, we performed mouse gene transfer experiments. Transfer of fukutin into knock-in mice restored glycosylation of alpha-dystroglycan. In addition, transfer of LARGE produced laminin-binding forms of alpha-dystroglycan in both knock-in mice and the POMGnT1 mutant mouse, which is another model of dystroglycanopathy. Overall, these data suggest that even partial restoration of alpha-dystroglycan glycosylation and laminin-binding activity by replacing or augmenting glycosylation-related genes might effectively deter dystroglycanopathy progression and thus provide therapeutic benefits.


Asunto(s)
Distroglicanos/metabolismo , Laminina/metabolismo , Distrofias Musculares/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Glicosilación , Humanos , Laminina/genética , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/terapia , Mutagénesis Insercional , N-Acetilglucosaminiltransferasas/genética , Unión Proteica , Proteínas/genética , Proteínas/metabolismo , Transferasas
5.
Mol Genet Metab ; 103(1): 26-32, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21353612

RESUMEN

Small molecules including imino sugars are expected to act as chaperones for a mutant α-galactosidase A (GLA), which will be useful for pharmacological chaperone therapy for Fabry disease. However, there is little detailed information about the molecular mechanism. We paid attention to an M51I mutant GLA which had been reported to strongly react to an imino sugar. The predicted structural change caused by this amino acid substitution is very small and located on the surface of the molecule. We produced the mutant enzyme in yeast, and determined its enzymological characteristics. The enzymological parameter values are almost the same as those of the wild-type GLA, although the mutant enzyme is unstable not only under neutral pH conditions but also under acidic ones. Then, we directly examined the effect of imino sugars including 1-deoxygalactonojirimycin and galactostatin bisulfite on the purified mutant enzyme. The imino sugars apparently improved the stability of the mutant enzyme under both neutral and acidic pH conditions. The results of surface plasmon resonance biosensor assaying suggested that the imino sugars retained their binding activity as to the mutant enzyme under both neutral and acidic pH conditions. This information will facilitate improvement of pharmacological chaperone therapy for Fabry disease.


Asunto(s)
Sustitución de Aminoácidos/genética , Iminoazúcares/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , alfa-Galactosidasa/química , alfa-Galactosidasa/metabolismo , Animales , Células COS , Chlorocebus aethiops , Activación Enzimática/efectos de los fármacos , Estabilidad de Enzimas/efectos de los fármacos , Estabilidad de Enzimas/genética , Iminoazúcares/farmacología , Modelos Moleculares , Proteínas Mutantes/genética , Unión Proteica/efectos de los fármacos , alfa-Galactosidasa/genética
6.
J Hum Genet ; 56(6): 440-6, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21471980

RESUMEN

Recently, pharmacological chaperone therapy for Pompe disease with small molecules such as imino sugars has attracted interest. But mutant acid α-glucosidase (GAA) species responsive to imino sugars are limited. To elucidate the characteristics of a mutant GAA responsive to imino sugars, we performed biochemical and structural analyses. Among cultured fibroblast cell lines derived from Japanese Pompe patients, only one carrying p.S529V/p.S619R amino acid substitutions responded to 1-deoxynojirimycin (DNJ), and an expression study revealed that DNJ, N-butyl-deoxynojirimycin and nojirimycin-1-sulfonic acid increased the enzyme activity of the S529V mutant GAA expressed in Chinese hamster ovary cells. The results of western blotting analysis suggested that these imino sugars facilitated the intracellular transportation of the mutant GAA and stabilized it. Among these imino sugars, DNJ exhibited the strongest action on the mutant GAA. Structural analysis revealed that DNJ almost completely occupied the active site pocket, and interacted with amino acid residues comprising it through van der Waals contacts and hydrogen bonds. This information will be useful for improvement of pharmacological chaperone therapy for Pompe disease.


Asunto(s)
Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , alfa-Glucosidasas/química , alfa-Glucosidasas/metabolismo , Sustitución de Aminoácidos/genética , Animales , Células CHO , Dominio Catalítico/genética , Células Cultivadas , Cricetinae , Cricetulus , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Humanos , Iminoazúcares/metabolismo , Iminoazúcares/farmacología , Modelos Moleculares , Unión Proteica , Conformación Proteica , alfa-Glucosidasas/genética
7.
Mol Genet Metab ; 96(4): 233-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19181556

RESUMEN

Enzyme enhancement therapy (EET) for Fabry disease involving imino sugars has been developed and attracted interest. It is thought that imino sugars act as pharmacological chaperones for wild-type and mutant alpha-galactosidases (GLAs) in cells, but the mechanisms underlying the molecular interactions between the imino sugars and the enzyme have not been clarified yet. We examined various kinds of imino sugars and found that galactostatin bisulfite (GBS) inhibited GLA in vitro and increased the enzyme activity in cultured Fabry fibroblasts as in the case of 1-deoxygalactonojirimycin (DGJ). Then, we analyzed the molecular interactions between the imino sugars and recombinant human GLA by means of isothermal titration calorimetry and surface plasmon resonance biosensor assays, and first determined the thermodynamic and binding-kinetics parameters of imino sugar and GLA complex formation. The results revealed that DGJ bound to the enzyme more strongly than GBS, the binding of DGJ to the enzyme protein being enthalpy-driven. In the case of GBS, the reaction was mainly enthalpy-driven, but there was a possibility that entropy-driven factors were involved in the binding. Structural analysis in silico revealed that both the chemicals fit into the active-site pocket and undergo hydrogen bonding with residues comprising the active-site pocket including the catalytic ones. The side chain of GBS was oriented towards the entrance of the active-site pocket, and thus it could be in contact with residues comprising the wall of the active-site pocket. Thermodynamic, kinetic and structural studies should provide us with a lot of information for improving EET for Fabry disease.


Asunto(s)
Enfermedad de Fabry/enzimología , Iminoazúcares/farmacología , alfa-Galactosidasa/antagonistas & inhibidores , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/farmacología , Animales , Células CHO , Dominio Catalítico , Células Cultivadas , Cricetinae , Cricetulus , Enfermedad de Fabry/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/patología , Galactosamina/análogos & derivados , Galactosamina/química , Galactosamina/farmacología , Humanos , Iminoazúcares/química , Cinética , Modelos Moleculares , Termodinámica , alfa-Galactosidasa/metabolismo
8.
J Hum Genet ; 54(6): 324-30, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19343043

RESUMEN

To elucidate the mechanism underlying transport and processing defects from the viewpoint of enzyme folding, we constructed three-dimensional models of human acid alpha-glucosidase encompassing 27 relevant amino acid substitutions by means of homology modeling. Then, we determined in each separate case the number of affected atoms, the root-mean-square distance value and the solvent-accessible surface area value. The analysis revealed that the amino acid substitutions causing a processing or transport defect responsible for Pompe disease were widely spread over all of the five domains comprising the acid alpha-glucosidase. They were distributed from the core to the surface of the enzyme molecule, and the predicted structural changes varied from large to very small. Among the structural changes, we paid particular attention to G377R and G483R. These two substitutions are predicted to cause electrostatic changes in neighboring small regions on the molecular surface. The quality control system of the endoplasmic reticulum apparently detects these very small structural changes and degrades the mutant enzyme precursor (G377R), but also the cellular sorting system might be misled by these minor changes whereby the precursor is secreted instead of being transported to lysosomes (G483R).


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Proteínas Mutantes/química , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , alfa-Glucosidasas/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Retículo Endoplásmico/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Humanos , Lisosomas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Conformación Proteica , Homología de Secuencia de Aminoácido , alfa-Glucosidasas/metabolismo
9.
Clin Chim Acta ; 391(1-2): 68-73, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18328816

RESUMEN

BACKGROUND: Recently, enzyme enhancement therapy (EET) for Pompe disease involving imino sugars, which act as potential inhibitors of acid alpha-glucosidases in vitro, to improve the stability and/or transportation of mutant acid alpha-glucosidases in cells was studied and attracted interest. However, the mechanism underlying the molecular interaction between the imino sugars and the enzyme has not been clarified yet. METHODS: We examined the inhibitory and binding effects of four imino sugars on a recombinant human acid alpha-glucosidase, alglucosidase alfa, by means of inhibition assaying and isothermal titration calorimetry (ITC). Furthermore, we built structural models of complexes of the catalytic domain of the enzyme with the imino sugars bound to its active site by homology modeling, and examined the molecular interaction between them. RESULTS: All of the imino sugars examined exhibited a competitive inhibitory action against the enzyme, 1-deoxynojirimycin (DNJ) exhibiting the strongest action among them. ITC revealed that one compound molecule binds to one enzyme molecule and that DNJ most strongly binds to the enzyme among them. Structural analysis revealed that the active site of the enzyme is almost completely occupied by DNJ. CONCLUSION: These biochemical and structural analyses increased our understanding of the molecular interaction between a human acid alpha-glucosidase and imino sugars.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Iminoazúcares/metabolismo , alfa-Glucosidasas/metabolismo , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/metabolismo , 1-Desoxinojirimicina/farmacología , Sitios de Unión , Dominio Catalítico , Interacciones Farmacológicas , Inhibidores de Glicósido Hidrolasas , Humanos , Iminoazúcares/química , Iminoazúcares/farmacología , Modelos Moleculares , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Termodinámica
10.
FEBS Open Bio ; 8(8): 1188-1201, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30087825

RESUMEN

Mammalian eukaryotic translation initiation factor 3 (eIF3) is the largest complex of the translation initiation factors. The eIF3 complex is comprised of thirteen subunits, which are named eIF3a to eIF3 m in most multicellular organisms. The eIF3e gene locus is one of the most frequent integration sites of mouse mammary tumor virus (MMTV), which induces mammary tumors in mice. MMTV-integration events result in the expression of C-terminal-truncated eIF3e proteins, leading to mammary tumor formation. We have shown that tumor formation can be partly caused by activation of hypoxia-inducible factor 2α. To investigate the function of eIF3e in mammals, we generated eIF3e-deficient mice. These eIF3e-/- mice are embryonically lethal, while eIF3e+/- mice are much smaller than wild-type mice. In addition, eIF3e+/- mouse embryonic fibroblasts (MEFs) contained reduced levels of eIF3a and eIF3c subunits and exhibited reduced cellular proliferation. These results suggest that eIF3e is essential for embryonic development in mice and plays a role in maintaining eIF3 integrity.

11.
Clin Chim Acta ; 378(1-2): 142-6, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17204262

RESUMEN

BACKGROUND: In cultured fibroblasts from I-cell disease patients the transport of many lysosomal enzymes is defective, and affected cells contain inclusion bodies filled with undegraded substrates. However, the contents of these inclusion bodies have not been well characterized yet. We attempted to identify accumulated substances in cultured I-cell disease fibroblasts cytochemically. METHODS: Cultured fibroblasts from I-cell disease patients were double-stained with a monoclonal antibody to lysosome-associated membrane protein-1 (LAMP-1) and that to GM2 ganglioside, or a series of lectins that specifically bind to sugar moieties. RESULTS: The patients' cells were granularly stained with the antibody to GM2 ganglioside and the lectins including Maakia amurensis, Datura stramonium, and concanavalin A. Their localization was coincident with that of LAMP-1. CONCLUSIONS: GM2 ganglioside and various kinds of glycoconjugates having sialic acidalpha2-3galactose, galactosebeta1-4N-acetylglucosamine and mannose residues accumulate in enlarged lysosomes in I-cell disease fibroblasts.


Asunto(s)
Fibroblastos/química , Gangliósido G(M2)/análisis , Proteínas de Membrana de los Lisosomas/análisis , Mucolipidosis/metabolismo , Células Cultivadas , Femenino , Histocitoquímica , Humanos , Lactante , Lectinas/análisis , Lisosomas/enzimología , Microscopía Electrónica , Mucolipidosis/patología , Piel/ultraestructura
12.
Clin Chim Acta ; 372(1-2): 154-7, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16753138

RESUMEN

BACKGROUND: A unique adult male patient who developed cardiomyopathy was first suspected to have cardiac Fabry disease based on the pathological findings in heart tissues obtained on biopsy, but the alpha-galactosidase activity in his leukocytes was normal and no mutation was detected in the coding region of the alpha-galactosidase gene. We identified accumulated materials in the myocardium of this patient. METHODS: Pathological and biochemical analyses were performed using the autopsied heart tissues as samples. RESULTS: Although numerous lamellar and concentric inclusion bodies were ultrastructurally found in the autopsied myocardium, the alpha-galactosidase activity in the heart tissues was not decreased. Lipid analysis revealed the accumulation of phospholipids including phosphatidylethanolamine, phosphatidylcholine, and phosphatidylinositol, but not globotriaosylcereamide or gangliosides. CONCLUSIONS: We found that a large amount of phospholipids accumulated in the myocardium of a patient with idiopathic cardiomyopathy, and electron microscopic findings of lamellar and concentric inclusion bodies in cardiomyocytes. A cardiac phospholipid storage disorder should be considered as an important candidate disease on differential diagnosis of myocardiac disorders including cardiac Fabry disease.


Asunto(s)
Cardiomiopatías/metabolismo , Fosfolípidos/metabolismo , Autopsia , Cardiomiopatías/enzimología , Cardiomiopatías/patología , Cromatografía en Capa Delgada , Humanos , Masculino , Persona de Mediana Edad , alfa-Galactosidasa/metabolismo
13.
Biochem Biophys Rep ; 7: 157-163, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28955902

RESUMEN

GM2 gangliosidoses are autosomal recessive lysosomal storage diseases (LSDs) caused by mutations in the HEXA, HEXB and GM2A genes, which encode the human lysosomal ß-hexosaminidase (Hex) α- and ß-subunits, and GM2 activator protein (GM2A), respectively. These diseases are associated with excessive accumulation of GM2 ganglioside (GM2) in the brains of patients with neurological symptoms. Here we established a CHO cell line overexpressing human GM2A, and purified GM2A from the conditioned medium, which was taken up by fibroblasts derived from a patient with GM2A deficiency, and had the therapeutic effects of reducing the GM2 accumulated in fibroblasts when added to the culture medium. We also demonstrated for the first time that recombinant GM2A could enhance the replacement effect of human modified HexB (modB) with GM2-degrading activity, which is composed of homodimeric altered ß-subunits containing a partial amino acid sequence of the α-subunit, including the GSEP loop necessary for binding to GM2A, on reduction of the GM2 accumulated in fibroblasts derived from a patient with Tay-Sachs disease, a HexA (αß heterodimer) deficiency, caused by HEXA mutations. We predicted the same manner of binding of GM2A to the GSEP loop located in the modified HexB ß-subunit to that in the native HexA α-subunit on the basis of the x-ray crystal structures. These findings suggest the effectiveness of combinational replacement therapy involving the human modified HexB and GM2A for GM2 gangliosidoses.

14.
Anat Embryol (Berl) ; 209(6): 485-94, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15891907

RESUMEN

MFG-E8, a secreted integrin-binding protein, consists of two EGF domains containing a RGD motif and two discoidin domains. In mouse embryogenesis, MFG-E8 is highly expressed in gonadal stromal cells near mesonephros at 11.5-12.5 dpc, but its function in gonadogenesis has not been characterized. To clarify a possible role of MFG-E8 in developing gonads, we analyzed the adhesion activity of 10.5-15.5 dpc gonadal cells to recombinant proteins of EGF or discoidin domains of MFG-E8. In EGF-coated wells, the gonadal cells at 11.5-12.5 dpc revealed a significantly higher adhesion activity as compared to those at 10.5 and 15.5 dpc, while discoidin domains showed a constant number of the adhered cells throughout these stages. To identify the adhesive cells of 11.5-dpc gonads, immunohistochemistry with anti-SF1/Ad4Bp antibody (a specific marker for supporting, steroidogenic, and coelomic epithelial cells) and staining for alkaline phosphatase (a germ cell marker) were carried out. As a result, EGF domains, as well as discoidin domains, were capable of binding to all three groups of SF1/Ad4Bp-positive and negative somatic cells, and germ cells of 11.5-dpc gonads. These findings therefore suggest that MFG-E8 mediates the cell-to-cell interaction among several somatic cell types and germ cells in mouse early gonadogenesis.


Asunto(s)
Antígenos de Superficie/metabolismo , Biomarcadores/metabolismo , Adhesión Celular/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Gónadas/citología , Proteínas de la Leche/metabolismo , Organogénesis , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Mitogénicos/metabolismo , Animales , Línea Celular , Receptores con Dominio Discoidina , Gónadas/embriología , Gónadas/metabolismo , Humanos , Ratones , Estructura Terciaria de Proteína/fisiología
15.
Biol Pharm Bull ; 31(9): 1691-5, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18758061

RESUMEN

To examine the uptake of a recombinant human alpha-L-iduronidase (laronidase) by cultured fibroblasts from a patient with mucopolysaccharidosis I (MPS I) and its effect on the cleavage of accumulated substrates, we performed enzymological, Western blotting, immunocytochemical and morphological studies. Laronidase was incorporated into the MPS I cells dose-dependently mainly via mannose 6-phosphate (M6P) receptors. Then the incorporated enzyme was transported to lysosomes and processed to the mature form, the pathological changes of the cells being improved. Furthermore, we compared the uptake of laronidase by cultured mouse osteoblasts with that by cultured mouse fibroblasts. The enzyme was incorporated into the cultured mouse osteoblasts mainly via M6P receptors, although mannose (Man) receptors were partially involved in the uptake of the enzyme, as in the cultured fibroblasts. But the uptake by the former was apparently lower than that by the latter. The administration of a high dose of the enzyme or development of a recombinant alpha-L-iduronidase containing many M6P residues is required for further improvement of enzyme replacement therapy for skeletal disorders caused by MPS I.


Asunto(s)
Fibroblastos/metabolismo , Iduronidasa/metabolismo , Osteoblastos/metabolismo , Animales , Células Cultivadas , Fibroblastos/ultraestructura , Humanos , Cinética , Ratones , Mucopolisacaridosis I/metabolismo , Osteoblastos/ultraestructura , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes/metabolismo
16.
J Hum Genet ; 52(12): 1018-1025, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17965825

RESUMEN

Peripheral neuropathy is one of the important manifestations of Fabry disease. Enzyme replacement therapy with presently available recombinant alpha-galactosidases does not always improve the Fabry neuropathy. But the reason has not been determined yet. We established a Schwann cell line from Fabry mice, characterized it, and then examined the uptake of alpha-galactosidase by cells and its effect on the degradation of accumulated substrate. The cells exhibited a distinct Schwann cell morphology and biochemical phenotype (alpha-Galactosidase activity was deficient, and numerous cytoplasmic inclusion bodies were present in the cells). A recombinant alpha-galactosidase added to the culture medium was incorporated into the cultured Fabry Schwann cells dose dependently. But the increase in cell-associated enzyme activity was less than that in the cases of human and mouse Fabry fibroblasts. The administration of a high dose of the enzyme improved the pathological changes in cells, although a low dose of it did not. Cellular uptake of the enzyme was strongly inhibited in the presence of mannose 6-phosphate. This suggests that the enzyme is incorporated via cation-independent mannose 6-phosphate receptors in Schwann cells. The low expression of cation-independent mannose 6-phosphate receptors in Schwann cells must be one of the reasons their uptake of the present enzymes was low. The administration of a high dose of the enzyme or the development of an enzyme containing many mannose 6-phosphate residues is required to improve Fabry neuropathy.


Asunto(s)
Enfermedad de Fabry/tratamiento farmacológico , Células de Schwann/metabolismo , alfa-Galactosidasa/farmacocinética , Animales , Transporte Biológico , Relación Dosis-Respuesta a Droga , Enfermedad de Fabry/enzimología , Enfermedad de Fabry/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Receptor IGF Tipo 2 , Proteínas Recombinantes , Células de Schwann/efectos de los fármacos , alfa-Galactosidasa/antagonistas & inhibidores
17.
Appl Environ Microbiol ; 73(15): 4805-12, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17557860

RESUMEN

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.


Asunto(s)
Proteínas Recombinantes/biosíntesis , Saccharomycetales/enzimología , Enfermedad de Sandhoff/enzimología , Enfermedad de Tay-Sachs/enzimología , beta-N-Acetilhexosaminidasas/biosíntesis , Secuencia de Aminoácidos , Biotecnología/métodos , Células Cultivadas , Fibroblastos/enzimología , Hexosaminidasa A , Hexosaminidasa B , Humanos , Datos de Secuencia Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , Saccharomycetales/genética , Enfermedad de Sandhoff/terapia , Enfermedad de Tay-Sachs/terapia , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/uso terapéutico
18.
J Hum Genet ; 52(11): 898-906, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17805474

RESUMEN

We constructed structural models of the catalytic domain and the surrounding region of human wild-type acid alpha-glucosidase and the enzyme with amino acid substitutions by means of homology modeling, and examined whether the amino acid replacements caused structural and biochemical changes in the enzyme proteins. Missense mutations including p.R600C, p.S619R and p.R437C are predicted to cause apparent structural changes. Nonsense mutation of p.C103X terminates the translation of acid alpha-glucosidase halfway through its biosynthesis and is deduced not to allow formation of the active site pocket. The mutant proteins resulting from these missense and nonsense mutations found in patients with Pompe disease are predictably unstable and degraded quickly in cells. The structural change caused by p.G576S is predicted to be small, and cells from a subject homozygous for this amino acid substitution exhibited 15 and 11% of the normal enzyme activity levels for an artificial substrate and glycogen, respectively, and corresponding amounts of the enzyme protein on Western blotting. No accumulation of glycogen was found in organs including skeletal muscle in the subject, and thus the residual enzyme activity could protect cells from glycogen storage. On the other hand, p.E689K, which is known as a neutral polymorphism, little affected the three-dimensional structure of acid alpha-glucosidase. Structural study on a mutant acid alpha-glucosidase in silico combined with biochemical investigation is useful for understanding the molecular pathology of Pompe disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , alfa-Glucosidasas/genética , Secuencia de Aminoácidos , Western Blotting , Células Cultivadas , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , alfa-Glucosidasas/química , alfa-Glucosidasas/metabolismo
19.
Hum Mol Genet ; 15(8): 1279-89, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16531417

RESUMEN

Recent studies have revealed an association between post-translational modification of alpha-dystroglycan (alpha-DG) and certain congenital muscular dystrophies known as secondary alpha-dystroglycanopathies (alpha-DGpathies). Fukuyama-type congenital muscular dystrophy (FCMD) is classified as a secondary alpha-DGpathy because the responsible gene, fukutin, is a putative glycosyltransferase for alpha-DG. To investigate the pathophysiology of secondary alpha-DGpathies, we profiled gene expression in skeletal muscle from FCMD patients. cDNA microarray analysis and quantitative real-time polymerase chain reaction showed that expression of developmentally regulated genes, including myosin heavy chain (MYH) and myogenic transcription factors (MRF4, myogenin and MyoD), in FCMD muscle fibers is inconsistent with dystrophy and active muscle regeneration, instead more of implicating maturational arrest. FCMD skeletal muscle contained mainly immature type 2C fibers positive for immature-type MYH. These characteristics are distinct from Duchenne muscular dystrophy, suggesting that another mechanism in addition to dystrophy accounts for the FCMD skeletal muscle lesion. Immunohistochemical analysis revealed morphologically aberrant neuromuscular junctions (NMJs) lacking MRF4 co-localization. Hypoglycosylated alpha-DG indicated a lack of aggregation, and acetylcholine receptor (AChR) clustering was compromised in FCMD and the myodystrophy mouse, another model of secondary alpha-DGpathy. Electron microscopy showed aberrant NMJs and neural terminals, as well as myotubes with maturational defects. Functional analysis of NMJs of alpha-DGpathy showed decreased miniature endplate potential and higher sensitivities to d-Tubocurarine, suggesting aberrant or collapsed formation of NMJs. Because alpha-DG aggregation and subsequent clustering of AChR are crucial for NMJ formation, hypoglycosylation of alpha-DG results in aberrant NMJ formation and delayed muscle terminal maturation in secondary alpha-DGpathies. Although severe necrotic degeneration or wasting of skeletal muscle fibers is the main cause of congenital muscular dystrophies, maturational delay of muscle fibers also underlies the etiology of secondary alpha-DGpathies.


Asunto(s)
Distroglicanos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofias Musculares/metabolismo , Unión Neuromuscular/metabolismo , Animales , Niño , Preescolar , Perfilación de la Expresión Génica , Glicosilación , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Ratones , Microscopía Electrónica , Fibras Musculares Esqueléticas/fisiología , Unión Neuromuscular/ultraestructura , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo
20.
J Hum Genet ; 51(3): 180-188, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16372133

RESUMEN

We compared two recombinant alpha-galactosidases developed for enzyme replacement therapy for Fabry disease, agalsidase alfa and agalsidase beta, as to specific alpha-galactosidase activity, stability in plasma, mannose 6-phosphate (M6P) residue content, and effects on cultured human Fabry fibroblasts and Fabry mice. The specific enzyme activities of agalsidase alfa and agalsidase beta were 1.70 and 3.24 mmol h(-1) mg protein(-1), respectively, and there was no difference in stability in plasma between them. The M6P content of agalsidase beta (3.6 mol/mol protein) was higher than that of agalsidase alfa (1.3 mol/mol protein). The administration of both enzymes resulted in marked increases in alpha-galactosidase activity in cultured human Fabry fibroblasts, and Fabry mouse kidneys, heart, spleen and liver. However, the increase in enzyme activity in cultured fibroblasts, kidneys, heart and spleen was higher when agalsidase beta was used. An immunocytochemical analysis revealed that the incorporated recombinant enzyme degraded the globotriaosyl ceramide accumulated in cultured Fabry fibroblasts in a dose-dependent manner, with the effect being maintained for at least 7 days. Repeated administration of agalsidase beta apparently decreased the number of accumulated lamellar inclusion bodies in renal tubular cells of Fabry mice.


Asunto(s)
Enfermedad de Fabry/tratamiento farmacológico , Isoenzimas/uso terapéutico , alfa-Galactosidasa/uso terapéutico , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Enfermedad de Fabry/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/ultraestructura , Humanos , Isoenzimas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Proteínas Recombinantes , alfa-Galactosidasa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA