Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
JIMD Rep ; 64(6): 460-467, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37927484

RESUMEN

Fabry disease (FD, OMIM 301500) is a rare X-linked inherited lysosomal storage disorder associated with reduced activities of α-galactosidase A (aGal, EC 3.2.1.22). The current standard of care for FD is based on enzyme replacement therapy (ERT), in which a recombinantly produced version of αGal is intravenously (iv) applied to Fabry patients in biweekly intervals. Though the iv application is clinically efficacious, periodical infusions are inconvenient, time- and resource-consuming and they negatively impact the patients' quality of life. Subcutaneous (sc) injection, in contrast, is an established route of administration for treatment of chronic conditions. It opens the beneficial option of self-administration, thereby improving patients' quality of life and at the same time reducing treatment costs. We have previously shown that Moss-α-Galactosidase (moss-aGal), recombinantly produced in the moss Physcomitrium patens, is efficient in degrading accumulated Gb3 in target organs of murine model of FD and in the phase I clinical study, we obtained first efficacy evidence in human patients following single iv infusion. Here, we tested the efficacy of subcutaneous administration of moss-aGal and compared it with the results observed following iv infusion in Fabry mice. The obtained findings demonstrate that subcutaneously applied moss-aGal is correctly transported to target organs and efficacious in degrading Gb3 deposits there and thus suggest the possibility of using this route of administration for therapy of Fabry disease.

2.
Sci Rep ; 13(1): 4748, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36959353

RESUMEN

Fabry disease is caused by a deficiency of α-galactosidase A (GLA) leading to the lysosomal accumulation of globotriaosylceramide (Gb3) and other glycosphingolipids. Fabry patients experience significant damage to the heart, kidney, and blood vessels that can be fatal. Here we apply directed evolution to generate more stable GLA variants as potential next generation treatments for Fabry disease. GLAv05 and GLAv09 were identified after screening more than 12,000 GLA variants through 8 rounds of directed evolution. Both GLAv05 and GLAv09 exhibit increased stability at both lysosomal and blood pH, stability to serum, and elevated enzyme activity in treated Fabry fibroblasts (19-fold) and GLA-/- podocytes (10-fold). GLAv05 and GLAv09 show improved pharmacokinetics in mouse and non-human primates. In a Fabry mouse model, the optimized variants showed prolonged half-lives in serum and relevant tissues, and a decrease of accumulated Gb3 in heart and kidney. To explore the possibility of diminishing the immunogenic potential of rhGLA, amino acid residues in sequences predicted to bind MHC II were targeted in late rounds of GLAv09 directed evolution. An MHC II-associated peptide proteomics assay confirmed a reduction in displayed peptides for GLAv09. Collectively, our findings highlight the promise of using directed evolution to generate enzyme variants for more effective treatment of lysosomal storage diseases.


Asunto(s)
Enfermedad de Fabry , Humanos , Ratones , Animales , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/genética , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo , Riñón/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo
3.
Mol Genet Metab Rep ; 33: 100919, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36186841

RESUMEN

Fabry disease is an X-linked lysosomal storage disorder caused by a deficiency of α-galactosidase A and subsequent accumulation of glycosphingolipids with terminal α-D-galactosyl residues. The molecular process through which this abnormal metabolism of glycosphingolipids causes multisystem dysfunction in Fabry disease is not fully understood. We sought to determine whether dysregulated DNA methylation plays a role in the development of this disease. In the present study, using isogenic cellular models derived from Fabry patient endothelial cells, we tested whether manipulation of α-galactosidase A activity and glycosphingolipid metabolism affects DNA methylation. Bisulfite pyrosequencing revealed that changes in α-galactosidase A activity were associated with significantly altered DNA methylation in the androgen receptor promoter, and this effect was highly CpG loci-specific. Methylation array studies showed that α-galactosidase A activity and glycosphingolipid levels were associated with differential methylation of numerous CpG sites throughout the genome. We identified 15 signaling pathways that may be susceptible to methylation alterations in Fabry disease. By incorporating RNA sequencing data, we identified 21 genes that have both differential mRNA expression and methylation. Upregulated expression of collagen type IV alpha 1 and alpha 2 genes correlated with decreased methylation of these two genes. Methionine levels were elevated in Fabry patient cells and Fabry mouse tissues, suggesting that a perturbed methionine cycle contributes to the observed dysregulated methylation patterns. In conclusion, this study provides evidence that α-galactosidase A deficiency and glycosphingolipid storage may affect DNA methylation homeostasis and highlights the importance of epigenetics in the pathogenesis of Fabry disease and, possibly, of other lysosomal storage disorders.

4.
Clin Pharmacol Drug Dev ; 10(9): 1075-1088, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33876577

RESUMEN

Approved therapies for Fabry disease (FD) include migalastat, an oral pharmacological chaperone, and agalsidase beta and agalsidase alfa, 2 forms of enzyme replacement therapy. Broad tissue distribution may be beneficial for clinical efficacy in FD, which has severe manifestations in multiple organs. Here, migalastat and agalsidase beta biodistribution were assessed in mice and modeled using physiologically based pharmacokinetic (PBPK) analysis, and migalastat biodistribution was subsequently extrapolated to humans. In mice, migalastat concentration was highest in kidneys and the small intestine, 2 FD-relevant organs. Agalsidase beta was predominantly sequestered in the liver and spleen (organs unaffected in FD). PBPK modeling predicted that migalastat 123 mg every other day resulted in concentrations exceeding the in vitro half-maximal effective concentration in kidneys, small intestine, skin, heart, and liver in human subjects. However, extrapolation of mouse agalsidase beta concentrations to humans was unsuccessful. In conclusion, migalastat may distribute to tissues that are inaccessible to intravenous agalsidase beta in mice, and extrapolation of mouse migalastat concentrations to humans showed adequate tissue penetration, particularly in FD-relevant organs.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Isoenzimas/farmacocinética , Modelos Biológicos , alfa-Galactosidasa/farmacocinética , 1-Desoxinojirimicina/farmacocinética , Adulto , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Especificidad de la Especie , Distribución Tisular , Adulto Joven , alfa-Galactosidasa/genética
5.
J Lipid Res ; 61(11): 1410-1423, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32868283

RESUMEN

Fabry disease is caused by deficient activity of α-galactosidase A, an enzyme that hydrolyzes the terminal α-galactosyl moieties from glycolipids and glycoproteins, and subsequent accumulation of glycosphingolipids, mainly globotriaosylceramide (Gb3), globotriaosylsphingosine (lyso-Gb3), and galabiosylceramide. However, there is no known link between these compounds and disease severity. In this study, we compared Gb3 isoforms (various fatty acids) and lyso-Gb3 analogs (various sphingosine modifications) in two strains of Fabry disease mouse models: a pure C57BL/6 (B6) background or a B6/129 mixed background, with the latter exhibiting more prominent cardiac and renal hypertrophy and thermosensation deficits. Total Gb3 and lyso-Gb3 levels in the heart, kidney, and dorsal root ganglion (DRG) were similar in the two strains. However, levels of the C20-fatty acid isoform of Gb3 and particular lyso-Gb3 analogs (+18, +34) were significantly higher in Fabry-B6/129 heart tissue when compared with Fabry-B6. By contrast, there was no difference in Gb3 and lyso-Gb3 isoforms/analogs in the kidneys and DRG between the two strains. Furthermore, using immunohistochemistry, we found that Gb3 massively accumulated in DRG mechanoreceptors, a sensory neuron subpopulation with preserved function in Fabry disease. However, Gb3 accumulation was not observed in nonpeptidergic nociceptors, the disease-relevant subpopulation that has remarkably increased isolectin-B4 (the marker of nonpeptidergic nociceptors) binding and enlarged cell size. These findings suggest that specific species of Gb3 or lyso-Gb3 may play major roles in the pathogenesis of Fabry disease, and that Gb3 and lyso-Gb3 are not responsible for the pathology in all tissues or cell types.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Fabry/metabolismo , Glicoesfingolípidos/metabolismo , Animales , Enfermedad de Fabry/genética , Femenino , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Índice de Severidad de la Enfermedad
6.
Nat Commun ; 10(1): 1785, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31040271

RESUMEN

Lysosomal replacement enzymes are essential therapeutic options for rare congenital lysosomal enzyme deficiencies, but enzymes in clinical use are only partially effective due to short circulatory half-life and inefficient biodistribution. Replacement enzymes are primarily taken up by cell surface glycan receptors, and glycan structures influence uptake, biodistribution, and circulation time. It has not been possible to design and systematically study effects of different glycan features. Here we present a comprehensive gene engineering screen in Chinese hamster ovary cells that enables production of lysosomal enzymes with N-glycans custom designed to affect key glycan features guiding cellular uptake and circulation. We demonstrate distinct circulation time and organ distribution of selected glycoforms of α-galactosidase A in a Fabry disease mouse model, and find that an α2-3 sialylated glycoform designed to eliminate uptake by the mannose 6-phosphate and mannose receptors exhibits improved circulation time and targeting to hard-to-reach organs such as heart. The developed design matrix and engineered CHO cell lines enables systematic studies towards improving enzyme replacement therapeutics.


Asunto(s)
Lisosomas/enzimología , Animales , Células CHO , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/enzimología , Enfermedad de Fabry/metabolismo , Glicosilación , Masculino , Ratones , Ratones Noqueados , Proteínas Recombinantes/uso terapéutico , alfa-Galactosidasa/uso terapéutico
7.
Gene Ther ; 25(7): 497-509, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30072816

RESUMEN

Here we present our progress in inducing an ectopic brown adipose tissue (BAT) phenotype in skeletal muscle (SKM) as a potential gene therapy for obesity and its comorbidities. We used ultrasound-targeted microbubble destruction (UTMD), a novel targeted, non-viral approach to gene therapy, to deliver genes in the BAT differentiation pathway into rodent SKM to engineer a thermogenic BAT phenotype with ectopic mUCP-1 overexpression. In parallel, we performed a second protocol using wild-type Ucp-1-null knockout mice to test whether the effects of the gene therapy are UCP-1 dependent. Our main findings were a robust cellular presence of mUCP-1 immunostaining (IHC), significantly higher expression levels of mUCP-1 measured by qRT-PCR, and highest temperature elevation measured by infrared thermography in the treated thigh, achieved in rats after delivering the UTMD-PRDM16/PGC-1a/BMP7/hyPB gene cocktail. Interestingly, the weight loss obtained in the treated rats with the triple gene delivery, never recovered the levels observed in the controls in spite of food intake recovery. Our results establish the feasibility of minimally invasive UTMD gene-based therapy administration in SKM, to induce overexpression of ectopic mUCP-1 after delivery of the thermogenic BAT gene program, and describe systemic effects of this intervention on food intake, weight loss, and thermogenesis.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Terapia Genética , Obesidad/terapia , Proteína Desacopladora 1/genética , Tejido Adiposo Pardo/trasplante , Animales , Ingestión de Alimentos/genética , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Ratas , Termogénesis/genética , Proteína Desacopladora 1/administración & dosificación
8.
J Inherit Metab Dis ; 41(2): 231-238, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29110178

RESUMEN

Fabry disease is a glycosphingolipidosis caused by deficient activity of α-galactosidase A; it is one of a few diseases that are associated with priapism, an abnormal prolonged erection of the penis. The goal of this study was to investigate the pathogenesis of Fabry disease-associated priapism in a mouse model of the disease. We found that Fabry mice develop late-onset priapism. Neuronal nitric oxide synthase (nNOS), which was predominantly present as the 120-kDa N-terminus-truncated form, was significantly upregulated in the penis of 18-month-old Fabry mice compared to wild type controls (~fivefold). Endothelial NOS (eNOS) was also upregulated (~twofold). NO level in penile tissues of Fabry mice was significantly higher than wild type controls at 18 months. Gene transfer-mediated enzyme replacement therapy reversed abnormal nNOS expression in the Fabry mouse penis. The penile nNOS level was restored by antiandrogen treatment, suggesting that hyperactive androgen receptor signaling in Fabry mice may contribute to nNOS upregulation. However, the phosphodiesterase-5A expression level and the adenosine content in the penis, which are known to play roles in the development of priapism in other etiologies, were unchanged in Fabry mice. In conclusion, these data suggested that increased nNOS (and probably eNOS) content and the consequential elevated NO production and high arterial blood flow in the penis may be the underlying mechanism of priapism in Fabry mice. Furthermore, in combination with previous findings, this study suggested that regulation of NOS expression is susceptible to α-galactosidase A deficiency, and this may represent a general pathogenic mechanism of Fabry vasculopathy.


Asunto(s)
Enfermedad de Fabry/complicaciones , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Erección Peniana , Pene/enzimología , Priapismo/etiología , Animales , Modelos Animales de Enfermedad , Terapia de Reemplazo Enzimático/métodos , Enfermedad de Fabry/enzimología , Enfermedad de Fabry/fisiopatología , Enfermedad de Fabry/terapia , Terapia Genética/métodos , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Pene/fisiopatología , Priapismo/enzimología , Priapismo/fisiopatología , Priapismo/terapia , Flujo Sanguíneo Regional , Transducción de Señal , Regulación hacia Arriba , alfa-Galactosidasa/biosíntesis , alfa-Galactosidasa/genética
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 37(10): 1345-1350, 2017 Oct 20.
Artículo en Chino | MEDLINE | ID: mdl-29070464

RESUMEN

OBJECTIVE: To elucidate the mechanisms of up regulated expression of cytoplasmic phospholipase A2 (CPLA2) induced by one lung ventilation (OLV) by investigating the interactions between nuclear factor kappaB (NF-κB) and C-PLA2. METHODS: Forty-eight healthy Japanese white rabbits were randomized into control group, solvent treatment group (group S), NF-κB inhibitor (PDTC)/solvent treatment group ( group PS), C-PLA2 inhibitor (AACOCF3)/solvent treatment group (group AS), OLV group (group O), solvent treatment plus OLV group (SO group), NFκB inhibitor (PDTC)/solvent treatment plus OLV group (group PSO) and CPLA2 inhibitor (AACOCF3)/solvent treatment plus OLV group (group ASO). ELISA was used to detect arachidonic acid (AA) content in the lung tissues, and NFκB and CPLA2 expressions were detected by Western blotting and quantitative PCR. Lung injuries were assessed based on the lung histological score, and the polymorphonuclear leukocyte count in the bronchial alveolar lavage fluid, myeloperoxidase (MPO) content in the lung tissues, and lung wet/dry weight (W/D) raito were determined. RESULTS: Treatment of the rabbits with the solvent did not produce any adverse effects. OLV caused obvious lung injury in the rabbits and up regulated the expressions of CPLA2 and NFκB in the lung tissues (P<0.05). In rabbits without OLV, treatment with AACOCF3 or PDTC significantly down regulated both CPLA2 and NFκB expressions without affecting the other parameters. In rabbits with OLV, treatment with AACOCF3 or PDTC obviously lowered CPLA2 and NFκB expressions and lessened the OLV-induced lung injuries. CONCLUSION: Both C-PLA2 and NF-κB play important roles and show interactions in OLV-induced lung injury in rabbits.


Asunto(s)
Lesión Pulmonar/metabolismo , FN-kappa B/aislamiento & purificación , Ventilación Unipulmonar/efectos adversos , Fosfolipasas A2/metabolismo , Animales , Citoplasma/metabolismo , Regulación de la Expresión Génica , Pulmón , Conejos , Distribución Aleatoria
10.
Hum Mol Genet ; 26(6): 1182-1192, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28158561

RESUMEN

Fabry disease is caused by deficient activity of α-galactosidase A and subsequent accumulation of glycosphingolipids (mainly globotriaosylceramide, Gb3), leading to multisystem organ dysfunction. Oxidative stress and nitric oxide synthase (NOS) uncoupling are thought to contribute to Fabry cardiovascular diseases. We hypothesized that decreased tetrahydrobiopterin (BH4) plays a role in the pathogenesis of Fabry disease. We found that BH4 was decreased in the heart and kidney but not in the liver and aorta of Fabry mice. BH4 was also decreased in the plasma of female Fabry patients, which was not corrected by enzyme replacement therapy (ERT). Gb3 levels were inversely correlated with BH4 levels in animal tissues and cultured patient cells. To investigate the role of BH4 deficiency in disease phenotypes, 12-month-old Fabry mice were treated with gene transfer-mediated ERT or substrate reduction therapy (SRT) for 6 months. In the Fabry mice receiving SRT but not ERT, BH4 deficiency was restored, concomitant with ameliorated cardiac and renal hypertrophy. Additionally, glutathione levels were decreased in Fabry mouse tissues in a sex-dependent manner. Renal BH4 levels were closely correlated with glutathione levels and inversely correlated with cardiac and kidney weight. In conclusion, this study showed that BH4 deficiency occurs in Fabry disease and may contribute to the pathogenesis of the disease through oxidative stress associated with a reduced antioxidant capacity of cells and NOS uncoupling. This study also suggested dissimilar efficacy of ERT and SRT in correcting pre-existing pathologies in Fabry disease.


Asunto(s)
Biopterinas/análogos & derivados , Terapia de Reemplazo Enzimático , Enfermedad de Fabry/genética , alfa-Galactosidasa/genética , Animales , Biopterinas/deficiencia , Biopterinas/genética , Biopterinas/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Fabry/mortalidad , Enfermedad de Fabry/fisiopatología , Femenino , Glutatión/metabolismo , Glicoesfingolípidos/metabolismo , Humanos , Riñón/metabolismo , Riñón/patología , Ratones , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Estrés Oxidativo/genética , alfa-Galactosidasa/biosíntesis , alfa-Galactosidasa/metabolismo
11.
Oncotarget ; 7(49): 80391-80403, 2016 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-27823982

RESUMEN

Established adriamycin cardiomyopathy is a lethal disease. When congestive heart failure develops, mortality is approximately 50% in a year. It has been known that ANGPTLs has various functions in lipid metabolism, inflammation, cancer cell invasion, hematopoietic stem activity and diabetes. We hypothesized that ANGPTL8 is capable of maintaining heart function by stimulating adult cardiac progenitor cells to initiate myocardial regeneration. We employed UTMD to deliver piggybac transposon plasmids with the human ANGPTL8 gene to the liver of rats with adriamycin cardiomyopathy. After ANGPTL8 gene liver delivery, overexpression of transgenic human ANGPTL8 was found in rat liver cells and blood. UTMD- ANGPTL8 gene therapy restored LV mass, fractional shortening index, and LV posterior wall diameter to nearly normal. Our results also showed that ANGPTL8 reversed established ADM cardiomyopathy. This was associated with activation of ISL-1 positive cardiac progenitor cells in the epicardium. A time-course experiment shown that ISL-1 cardiac progenitor cells proliferated and formed a niche in the epicardial layer and then migrated into sub-epicardium. The observed myocardial regeneration accompanying reversal of adriamycin cardiomyopathy was associated with upregulation of PirB expression on the cell membrane of cardiac muscle cells or progenitor cells stimulated by ANGPTL8.


Asunto(s)
Proteínas Similares a la Angiopoyetina/biosíntesis , Cardiomiopatías/terapia , Doxorrubicina , Terapia Genética/métodos , Hígado/metabolismo , Miocitos Cardíacos/metabolismo , Hormonas Peptídicas/biosíntesis , Células Madre/metabolismo , Proteína 8 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina/sangre , Proteínas Similares a la Angiopoyetina/genética , Animales , Cardiomiopatías/inducido químicamente , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiotoxicidad , Línea Celular , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Humanos , Proteínas con Homeodominio LIM/metabolismo , Masculino , Microburbujas , Contracción Miocárdica , Miocitos Cardíacos/patología , Hormonas Peptídicas/sangre , Hormonas Peptídicas/genética , Ratas Sprague-Dawley , Receptores Inmunológicos/metabolismo , Recuperación de la Función , Regeneración , Nicho de Células Madre , Células Madre/patología , Factores de Tiempo , Factores de Transcripción/metabolismo , Ultrasonido , Función Ventricular Izquierda , Remodelación Ventricular
12.
J Inherit Metab Dis ; 39(3): 447-455, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26960552

RESUMEN

Fabry disease is caused by deficient activity of α-galactosidase A and subsequent intracellular accumulation of glycosphingolipids, mainly globotriaosylceramide (Gb3). Vascular endothelial cells may play important roles in disease pathogenesis, and are one of the main target cell types in therapeutic interventions. In this study, we generated immortalized aortic endothelial cell lines from a mouse model of Fabry disease. These cells retained endothelial cell-specific markers and functions. Gb3 expression level in one of these clones (referred to as FMEC2) was highly susceptible to culture media, and appeared to be regulated by glucosylceramide synthase. Results also showed that Gb3 could be upregulated by hydrocortisone. FMEC2 express the mannose 6-phosphate receptor and sortilin but not the mannose receptor. Uptake studies suggested that sortilin plays a role in the binding and internalization of mammalian cell-produced α-galactosidase A. Moss-aGal (a plant-made enzyme) was endocytosed by FMEC2 via a receptor other than the aforementioned receptors. In conclusion, this study suggests that glucosylceramide synthase and hydrocortisone may play important roles in modulating Gb3 levels in Fabry mouse aortic endothelial cells, and that endocytosis of recombinant α-galactosidase A involves a combination of multiple receptors depending on the properties of the enzyme.


Asunto(s)
Aorta/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Enfermedad de Fabry/enzimología , Enfermedad de Fabry/metabolismo , Trihexosilceramidas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Endocitosis/fisiología , Endotelio Vascular/enzimología , Glucosiltransferasas/metabolismo , Glicoesfingolípidos/metabolismo , Lectinas Tipo C/metabolismo , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptor IGF Tipo 2/metabolismo , Receptores de Superficie Celular/metabolismo , alfa-Galactosidasa/metabolismo
13.
J Inherit Metab Dis ; 39(2): 293-303, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26310963

RESUMEN

Enzyme replacement therapy (ERT) is an effective treatment for several lysosomal storage disorders (LSDs). Intravenously infused enzymes are taken up by tissues through either the mannose 6-phosphate receptor (M6PR) or the mannose receptor (MR). It is generally believed that M6PR-mediated endocytosis is a key mechanism for ERT in treating LSDs that affect the non-macrophage cells of visceral organs. However, the therapeutic efficacy of MR-mediated delivery of mannose-terminated enzymes in these diseases has not been fully evaluated. We tested the effectiveness of a non-phosphorylated α-galactosidase A produced from moss (referred to as moss-aGal) in vitro and in a mouse model of Fabry disease. Endocytosis of moss-aGal was MR-dependent. Compared to agalsidase alfa, a phosphorylated form of α-galactosidase A, moss-aGal was more preferentially targeted to the kidney. Cellular localization of moss-aGal and agalsidase alfa in the heart and kidney was essentially identical. A single injection of moss-aGal led to clearance of accumulated substrate in the heart and kidney to an extent comparable to that achieved by agalsidase alfa. This study suggested that mannose-terminated enzymes may be sufficiently effective for some LSDs in which non-macrophage cells are affected, and that M6P residues may not always be a prerequisite for ERT as previously considered.


Asunto(s)
Enfermedad de Fabry/enzimología , Enfermedad de Fabry/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Manosafosfatos/metabolismo , Receptores de Superficie Celular/metabolismo , alfa-Galactosidasa/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Terapia de Reemplazo Enzimático/métodos , Femenino , Humanos , Isoenzimas/metabolismo , Riñón/metabolismo , Enfermedades por Almacenamiento Lisosomal/enzimología , Enfermedades por Almacenamiento Lisosomal/metabolismo , Masculino , Receptor de Manosa , Ratones , Ratones Endogámicos C57BL , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes
14.
Diabetologia ; 58(5): 1036-44, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25720603

RESUMEN

AIMS/HYPOTHESIS: ANGPTL8 is a circulatory hormone secreted from liver and adipose tissue that promotes pancreatic beta cell proliferation and interferes with triacylglycerol metabolism in mice. The clinical significance of its effects on inducing beta cell proliferation is limited because it causes severe hypertriacylglycerolaemia. METHODS: We employed ultrasound-targeted microbubble destruction (UTMD) to deliver human ANGPTL8 gene plasmids to the pancreas, liver and skeletal muscle of normal adult rats. RESULTS: Human ANGPTL8 was consistently detected in the circulation 1 month after UTMD. ANGPTL8 gene delivery promoted the proliferation of adult and aged beta cells, expanded the beta cell mass, improved glucose tolerance and increased the fasting blood insulin level after UTMD treatment without causing severe hypertriacylglycerolaemia. ANGPTL8 gene therapy significantly alleviated but did not totally reverse STZ-induced diabetes in a rat model. CONCLUSIONS/INTERPRETATION: ANGPTL8 induced adult and aged beta cell regeneration in a rat model.


Asunto(s)
Angiopoyetinas/genética , Glucemia/metabolismo , Diabetes Mellitus Experimental/terapia , Técnicas de Transferencia de Gen , Células Secretoras de Insulina/metabolismo , Regeneración/genética , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Terapia Genética , Insulina/sangre , Microburbujas , Páncreas/metabolismo , Ratas
15.
Biochem Biophys Res Commun ; 458(4): 823-9, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25701791

RESUMEN

UNLABELLED: Recently GLP-1 was found to have cardioprotective effects independent of those attributable to tight glycemic control. METHODS AND RESULTS: We employed ultrasound targeted microbubble destruction (UTMD) to deliver piggybac transposon plasmids encoding the GLP-1 gene with a nuclear localizing signal to rat hearts with adriamycin cardiomyopathy. After a single UTMD treatment, overexpression of transgenic GLP-1 was found in nuclei of rat heart cells with evidence that transfected cardiac cells had undergone proliferation. UTMD-GLP-1 gene therapy restored LV mass, fractional shortening index, and LV posterior wall diameter to nearly normal. Nuclear overexpression of GLP-1 by inducing phosphorylation of FoxO1-S256 and translocation of FoxO1 from the nucleus to the cytoplasm significantly inactivated FoxO1 and activated the expression of cyclin D1 in nuclei of cardiac muscle cells. Reversal of adriamycin cardiomyopathy appeared to be mediated by dedifferentiation and proliferation of nuclear FoxO1-positive cardiac muscle cells with evidence of embryonic stem cell markers (OCT4, Nanog, SOX2 and c-kit), cardiac early differentiation markers (NKX2.5 and ISL-1) and cellular proliferation markers (BrdU and PHH3) after UTMD with GLP-1 gene therapy. CONCLUSIONS: Intranuclear myocardial delivery of the GLP-1gene can reverse established adriamycin cardiomyopathy by stimulating myocardial regeneration.


Asunto(s)
Cardiomiopatías/inducido químicamente , Cardiomiopatías/terapia , Doxorrubicina , Péptido 1 Similar al Glucagón/genética , Miocardio/citología , Miocardio/patología , Plásmidos/uso terapéutico , Animales , Cardiomiopatías/genética , Cardiomiopatías/patología , Diferenciación Celular , Proliferación Celular , Ciclina D1/genética , Factores de Transcripción Forkhead/metabolismo , Técnicas de Transferencia de Gen/instrumentación , Terapia Genética , Péptido 1 Similar al Glucagón/metabolismo , Microburbujas , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas del Tejido Nervioso/metabolismo , Plásmidos/genética , Ratas , Ultrasonografía/instrumentación , Regulación hacia Arriba
16.
Hum Mol Genet ; 24(11): 3181-91, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25701874

RESUMEN

Fabry disease is caused by deficient activity of lysosomal enzyme α-galactosidase A. The enzyme deficiency results in intracellular accumulation of glycosphingolipids, leading to a variety of clinical manifestations including hypertrophic cardiomyopathy and renal insufficiency. The mechanism through which glycosphingolipid accumulation causes these manifestations remains unclear. Current treatment, especially when initiated at later stage of the disease, does not produce completely satisfactory results. Elucidation of the pathogenesis of Fabry disease is therefore crucial to developing new treatments. We found increased activity of androgen receptor (AR) signaling in Fabry disease. We subsequently also found that blockade of AR signaling either through castration or AR-antagonist prevented and reversed cardiac and kidney hypertrophic phenotype in a mouse model of Fabry disease. Our findings implicate abnormal AR pathway in the pathogenesis of Fabry disease and suggest blocking AR signaling as a novel therapeutic approach.


Asunto(s)
Antagonistas de Receptores Androgénicos/farmacología , Enfermedad de Fabry/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Enfermedades Renales/metabolismo , Receptores Androgénicos/metabolismo , Animales , Enfermedad de Fabry/tratamiento farmacológico , Femenino , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/tratamiento farmacológico , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Androgénicos/genética , Transducción de Señal , Transcripción Genética
17.
Mol Ther Nucleic Acids ; 2: e130, 2013 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-24150577

RESUMEN

Krabbe disease is a devastating neurodegenerative lysosomal storage disorder caused by a deficiency of ß-galactocerebrosidase (GALC). Gene therapy is a promising therapeutic approach for Krabbe disease. As the human brain is large and it is difficult to achieve global gene transduction, the efficacy of cross-correction is a critical determinant of the outcome of gene therapy for this disease. We investigated whether HIV Tat protein transduction domain (PTD) can improve the cross-correction of GALC. Tat-PTD significantly increased (~6-fold) cross-correction of GALC through enhanced secretion and uptake in a cell-culture model system. The effects of Tat-PTD were gene and flanking amino acids dependent. Tat-fusion increased the secretion of α-galactosidase A (α-gal A), but this did not improve its cross-correction. Tat-fusion did not change either secretion or uptake of ß-glucocerebrosidase (GC). Tat-PTD increased GALC protein synthesis, abolished reactivity of GC to the 8E4 antibody, and likely reduced mannose phosphorylation in all these lysosomal enzymes. This study demonstrated that Tat-PTD can be useful for increasing cross-correction efficiency of lysosomal enzymes. However, Tat-PTD is not a mere adhesive motif but possesses a variety of biological functions. Therefore, the potential beneficial effect of Tat-PTD should be assessed individually on each lysosomal enzyme.Molecular Therapy-Nucleic Acids (2013) 2, e130; doi:10.1038/mtna.2013.57; published online 22 October 2013.

18.
J Lipid Res ; 52(9): 1742-6, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21747096

RESUMEN

The aim of our study was to measure globotriaosylceramide (Gb(3)) and lyso-Gb(3) levels by tandem mass spectrometry in the urine and kidney in Fabry (gla knockout) mice and wild-type controls. We found that urine Gb(3) of male and female Fabry mice was higher than wild-type mice of the same sex but also significantly higher in male mice compared with females of the same genotype. In kidney tissue, sex and genotype-dependent differences in Gb(3) levels paralleled those in the urine. Isoforms C16, C22:1, and C24OHA were particularly higher in males compared with females in both wild-type and Fabry mice. Similarly, kidney lyso-Gb(3) concentrations were significantly higher in 12-month-old male Fabry mice than in their homozygous female counterparts. However, lyso-Gb(3) was undetectable in wild-type mice of both sexes. α-Galactosidase A activity and mRNA levels in kidney were significantly lower in male wild-type mice compared with female mice. This study shows the sex differences in kidney and urine Gb(3) and kidney lyso-Gb(3) levels in both wild-type and Fabry mice, and it suggests that these male-female differences should be taken into consideration when using murine models for Fabry disease.


Asunto(s)
Enfermedad de Fabry/orina , Riñón/química , Caracteres Sexuales , Trihexosilceramidas/análisis , Trihexosilceramidas/orina , Animales , Biomarcadores/análisis , Biomarcadores/orina , Modelos Animales de Enfermedad , Enfermedad de Fabry/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Espectrometría de Masas en Tándem/métodos , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo
19.
Proc Natl Acad Sci U S A ; 107(17): 7886-91, 2010 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-20385825

RESUMEN

Most lysosomal storage diseases (LSDs) are life-threatening genetic diseases. The pathogenesis of these diseases is poorly understood. Induced pluripotent stem (iPS) cell technology offers new opportunities for both mechanistic studies and development of stem cell- based therapies. Here we report the generation of disease-specific iPS cells from mouse models of Fabry disease, globoid cell leukodystrophy (GLD), and mucopolysaccharidosis VII (MPSVII). These mouse model-derived iPS cells showed defects in disease-specific enzyme activities and significant accumulation of substrates for these enzymes. In the lineage-directed differentiation studies, Fabry-iPS and GLD-iPS cells were efficiently differentiated into disease-relevant cell types, such as cardiomyocytes and neural stem cells, which might be useful in mechanistic and therapeutic studies. Notably, MPSVII-iPS cells demonstrated a markedly impaired ability to form embryoid bodies (EBs) in vitro. MPSVII-EBs exibited elevated levels of hyaluronan and its receptor CD44, and markedly reduced expression levels of E-cadherin and cell-proliferating marker. Partial correction of enzyme deficiency in MSPVII-iPS cells led to improved EB formation and reversal of aberrant protein expression. These data indicate a potential mechanism for the partial lethality of MPSVII mice in utero, and suggest a possible abnormality of embryonic development in MPSVII patients. Thus, our study demonstrates the unique promise of iPS cells for studying the pathogenesis and treatment of LSDs.


Asunto(s)
Línea Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Enfermedad de Fabry/fisiopatología , Células Madre Pluripotentes Inducidas/citología , Leucodistrofia de Células Globoides/fisiopatología , Mucopolisacaridosis VII/fisiopatología , Fosfatasa Alcalina , Animales , Western Blotting , Cadherinas/metabolismo , Diferenciación Celular/fisiología , Cromatografía Líquida de Alta Presión , Cromatografía en Capa Delgada , Enfermedad de Fabry/metabolismo , Enfermedad de Fabry/terapia , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Leucodistrofia de Células Globoides/metabolismo , Leucodistrofia de Células Globoides/terapia , Ratones , Mucopolisacaridosis VII/metabolismo , Mucopolisacaridosis VII/terapia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
In Vivo ; 22(4): 409-13, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18712165

RESUMEN

Although tenascin-C (TN) is highly up-regulated during the proliferation of reactive astrocytes, little is known about the function of TN at injury sites in the central nervous system (CNS). Here, the function of TN-expressing astrocytes in the injured brain was investigated by analyzing TN-deficient mice with stab-wound injuries of the cerebral cortex. Glial fibrillary acid protein expression after injury was down-regulated earlier in TN-deficient mice than in wild-type (WT) mice. To evaluate immune responses in the injured CNS in the absence of TN, inflammatory cytokine production was examined after unilateral stab injuries of the cerebral cortex in TN-deficient and WT mice. The expression of interleukin (IL)-1beta, tumor necrosis factor-a and IL-6 was higher in TN-deficient mice, whereas levels of IL-4 and granulocyte colony-stimulating factor were lower in TN-deficient mice than WT mice. Our findings suggest that TN helps to regulate production of inflammatory cytokines in the injured brain.


Asunto(s)
Sistema Nervioso Central/metabolismo , Citocinas/biosíntesis , Inflamación , Tenascina/deficiencia , Animales , Astrocitos/citología , Lesiones Encefálicas/metabolismo , Sistema Nervioso Central/lesiones , Corteza Cerebral/metabolismo , Regulación hacia Abajo , Proteína Ácida Fibrilar de la Glía/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA