Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Mol Genet Metab ; 104(1-2): 123-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21703893

RESUMEN

Our study is the first to demonstrate the ability to generate iPS cells from a mouse model of Pompe disease. Initially, mouse tail tip fibroblasts were harvested from male, 8-week-old (GAA) knockout mice, and three reprogramming factors (Oct3/4, Sox2 and Klf4) were transfected into the isolated donor cells using a retroviral vector. These iPS cells also showed decreased levels of GAA enzymatic activity and strong positive staining with periodic acid-Schiff (indicating the accumulation of glycogen) and acid phosphatase (lysosomal activation marker). Pompe-iPS cells were differentiated into skeletal muscle cells in Matrigel®-coated plates. Spindle-shaped skeletal muscle cells were successfully generated from Pompe-iPS cells and showed spontaneous contraction and positive staining with the myosin heavy chain antibody. Electron microscopic analysis of the skeletal muscle cells showed typical morphological features, including Z-bands, I-bands, A-bands and H-bands, which were visible in wild-type and Pompe cells. Furthermore, Pompe skeletal muscle cells accumulated massive glycogen in lysosomes. This study indicates that the iPS and skeletal muscle cells generated in this study could also be a useful disease model for studies investigating the pathogenesis and treatment of skeletal muscle in Pompe disease.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Células Madre Pluripotentes Inducidas/patología , Células Musculares/patología , Músculo Esquelético/patología , Animales , Forma de la Célula , Modelos Animales de Enfermedad , Células Madre Pluripotentes Inducidas/ultraestructura , Factor 4 Similar a Kruppel , Ratones , Ratones Desnudos , Células Musculares/ultraestructura , Músculo Esquelético/ultraestructura , Fenotipo
11.
Mol Genet Metab ; 95(3): 163-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18707907

RESUMEN

Fabry disease, an X-linked systemic vasculopathy, is caused by a deficiency of alpha-galactosidase A resulting in globotriaosylceramide (Gb(3)) storage in cells. The pathogenic role of Gb(3) in the disease is not known. Based on previous work, we tested the hypothesis that accumulation of Gb(3) in the vascular endothelium of Fabry disease is associated with increased production of reactive oxygen species (ROS) and increased expression of cell adhesion molecules. Gb(3)-loading resulted in increased intracellular ROS production in cultured vascular endothelial cells in a dose-dependent manner. Increased Gb(3) also induced expression of intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin. Reduction of endogenous Gb(3) by treatment of the cells with an inhibitor of glycosphingolipid synthase or alpha-galactosidase A led to decreased expression of adhesion molecules. Plasma from Fabry patients significantly increased ROS generation in endothelial cells when compared with plasma from non-Fabry controls. This effect was not influenced by reduction of intracellular Gb(3). This study provided direct evidence that excess intracellular Gb(3) induces oxidative stress and up-regulates the expression of cellular adhesion molecules in vascular endothelial cells. In addition, other factors in patient's plasma may also contribute to oxidative stress in Fabry vascular endothelial cells.


Asunto(s)
Selectina E/genética , Células Endoteliales/metabolismo , Enfermedad de Fabry/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Estrés Oxidativo , Trihexosilceramidas/metabolismo , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/genética , Células Cultivadas , Selectina E/metabolismo , Enfermedad de Fabry/genética , Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Plasma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
Endocrinology ; 148(12): 5865-73, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17761763

RESUMEN

Human chorionic gonadotropin (hCG) and LH play an important role in reproductive physiology. Both hCG and LH bind to the same LH/choriogonadotropin receptor (LH/CG-R). Recent reports documented the temporal and spatial expression of LH/CG-R in the developing and mature mammalian brain. Administration of hCG promoted nerve regeneration in vivo and neurite outgrowth and survival of primary neurons in vitro. The function of hCG/LH and LH/CG-R in the nervous system remains unclear. In this study, we report that hCG/LH induced distinct morphological and biochemical changes, characteristic of neuronal differentiation, in PC12 cells stably expressing LH/CG-R and that the differentiation effect is ligand dose and time dependent. Western blot analysis revealed that both the ERKs and p38 MAPK are activated after hCG treatment. Inhibitor studies showed both the ERK and p38 MAPK signal transduction pathways are required for this differentiation process, which is cAMP dependent and protein kinase A independent. These findings imply a potential role for hCG/LH and LH/CG-R in the development, maintenance, and regeneration of the mammalian nervous system, and in the neuropathogenesis of genetic diseases caused by a mutated LH/CG-R.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Gonadotropina Coriónica/farmacología , Neuronas/efectos de los fármacos , Receptores de HL/fisiología , Animales , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , MAP Quinasa Quinasa 4/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células PC12 , Fosfatidilinositol 3-Quinasas/metabolismo , Plásmidos/genética , Ratas , Receptores de HL/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transfección , Fosfolipasas de Tipo C/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
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
14.
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.

15.
Mol Genet Metab ; 92(1-2): 137-44, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17644384

RESUMEN

Fabry disease is an inborn error of glycosphingolipid catabolism resulting from a deficiency of lysosomal enzyme alpha-galactosidase A. The major clinical manifestations of the disease, such as stroke, cardiac dysfunction, and renal impairment, are thought to be caused by vasculopathy due to progressive accumulation of globotriaosylceramide in vascular endothelial cells. The pathogenesis of the vasculopathy has not been elucidated. Since in vitro studies using primary endothelial cells are hampered by the limited lifespan of these cells, the availability of cultured endothelial cells with an extended lifespan is critical for the study of the vasculopathy of Fabry disease. We therefore generated an endothelial cell line from a Fabry hemizygote by introduction of human telomerase reverse transcriptase gene. The cell line has markedly extended lifespan compared to parental primary cells. The cells stably express many key markers of endothelial cells such as von Willebrand factor, CD31, CD34, and endothelial nitric oxide synthase (eNOS) and retain functional characteristics such as uptake of acetylated low-density lipoprotein, responsiveness to angiogenic growth factors, up-regulation of eNOS production upon extracellular stimuli, and formation of tube-like structures on Matrigel basement membrane matrix. The cells show significantly reduced activity of alpha-galactosidase A compared with primary endothelial cells from normal individuals and accumulate globotriaosylceramide in lysosomes. This cell line will provide a useful in vitro model of Fabry disease and will facilitate systematic studies to investigate pathogenic mechanisms and explore new therapeutic approaches for Fabry disease.


Asunto(s)
Endotelio Vascular/patología , Enfermedad de Fabry/patología , Acetilación , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , División Celular , Línea Celular Transformada , Endotelio Vascular/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Lisosomas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Reacción en Cadena de la Polimerasa , Telomerasa/metabolismo , Trihexosilceramidas/metabolismo , Factor de von Willebrand/metabolismo
16.
Mol Genet Metab ; 84(4): 332-43, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15781194

RESUMEN

Globoid cell leukodystrophy (GLD, Krabbe disease) is a severe demyelinating disease caused by a genetic defect of beta-galactocerebrosidase (GALC). To date treatment to GLD is limited to hematopoietic stem cell transplantation. Experimental approaches by means of gene therapy in twitcher mouse, an authentic murine model of human GLD, showed significant but only marginal improvements of the disease. To clarify whether the introduction of GALC could provide beneficial effects on the oligodendrocytes in GLD, we transduced twitcher oligodendrocytes by stereotactically injecting recombinant retrovirus encoding GALC-myc-tag fusion gene into the forebrain subventricular zone of neonatal twitcher mouse. In vivo effects of exogenous GALC on twitcher oligodendrocytes were studied histologically by combined immunostaining for the myc-epitope and the oligodendroglial specific marker, pi form of glutathione-S-transferase, at around 40 days of age. We show here that GALC transduction led to dramatic morphological improvement of the twitcher oligodendrocytes comparing with those in untreated twitcher controls. This study provided direct in vivo evidence that GALC transduction could prevent or correct aberrant morphology of oligodendrocytes in GLD which may be closely related to the dysfunction and/or degeneration of oligodendrocytes and the demyelination in this disease.


Asunto(s)
Galactosilceramidasa/genética , Terapia Genética/métodos , Leucodistrofia de Células Globoides/patología , Oligodendroglía/citología , Animales , Encéfalo/citología , Encéfalo/patología , Encéfalo/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Epítopos/genética , Galactosilceramidasa/metabolismo , Técnicas de Transferencia de Gen , Genes myc , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Humanos , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/terapia , Ratones , Ratones Endogámicos BALB C , Oligodendroglía/fisiología , Retroviridae/genética , Transducción Genética
17.
J Gene Med ; 7(5): 540-51, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15685691

RESUMEN

BACKGROUND: Brain-directed prenatal gene therapy may benefit some lysosomal storage diseases that affect the central nervous system (CNS) before birth. Our previous study showed that intrauterine introduction of recombinant adenoviruses into cerebral ventricles results in efficient gene transfer to the CNS in the mouse. However, transgene expression decreased with time due to the non-integrative property of adenoviral vectors. In this study, in order to obtain permanent gene transduction, we investigated the feasibility of retrovirus-mediated in utero gene transduction. METHODS: Concentrated retrovirus encoding the LacZ gene was injected into the cerebral ventricles of the embryos of normal and twitcher mice (a murine model of Krabbe disease) at embryonic day 12. The distribution and maintenance of the transgene expression in the recipient brain were analyzed histochemically, biochemically and by the quantitative polymerase chain reaction method pre- and postnatally. RESULTS: Efficient and highly persistent gene transduction to the brain was achieved both in normal and the twitcher mouse. Transduced neurons, astrocytes and oligodendrocytes were distributed throughout the brain. The transduced LacZ gene, its transcript and protein expression in the brain were maintained for 14 months without decrement. In addition, gene transduction to multiple tissues other than the brain was also detected at low levels. CONCLUSIONS: This study suggests that brain-directed in utero gene transfer using retrovirus vector may be beneficial to the treatment of lysosomal storage diseases with severe brain damage early in life, such as Krabbe disease.


Asunto(s)
Encéfalo/metabolismo , Embrión de Mamíferos/metabolismo , Terapia Genética/métodos , Vectores Genéticos , Leucodistrofia de Células Globoides/terapia , Retroviridae/genética , Transducción Genética/métodos , Animales , Estudios de Factibilidad , Femenino , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes Neurológicos , Neuronas/metabolismo , Plásmidos , Transgenes , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
18.
J Gene Med ; 6(11): 1206-15, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15459963

RESUMEN

BACKGROUND: In some lysosomal storage diseases, considerable alterations of the central nervous system (CNS) occur prior to birth and neurodegeneration progresses rapidly soon after birth causing early death in patients. No effective treatment is available after birth. Treatment may need to be initiated before birth to prevent the onset or progression of neurological changes and thereby irreversible brain damage. The aim of this study is to investigate the feasibility and effectiveness of brain-directed prenatal gene therapy for lysosomal storage diseases. METHODS: Recombinant adenovirus encoding the lacZ gene was injected into the lateral ventricles of mouse embryos and the pattern of gene transduction to the CNS was investigated. In the therapeutic experiment, adenovirus expressing beta-glucuronidase was injected into the cerebral ventricles of the embryos of mucopolysaccharidosis VII mice and the therapeutic effects on the brain were evaluated. RESULTS: Injection of adenoviral vectors to the cerebral ventricles of mouse embryos led to widespread gene transduction throughout the brain and the spinal cord and transgene expression persisted over 10 months in those surviving the procedure. The prenatal transduction of the therapeutic gene to the brain of the mucopolysaccharidosis VII mouse efficiently prevented lysosomal storage in most brain cells before birth until 4 months after birth. CONCLUSIONS: Brain-directed in utero gene therapy through an intra-ventricular route would be an effective strategy to treat some lysosomal storage diseases with early and severe CNS alterations.


Asunto(s)
Adenoviridae/genética , Sistema Nervioso Central/metabolismo , Terapias Fetales/métodos , Terapia Genética , Mucopolisacaridosis VII/embriología , Mucopolisacaridosis VII/terapia , Transducción Genética , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Sistema Nervioso Central/patología , Femenino , Genes Reporteros , Vectores Genéticos/administración & dosificación , Humanos , Inyecciones Intraventriculares , Operón Lac , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis VII/genética , Embarazo , Médula Espinal/embriología , Médula Espinal/metabolismo , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
19.
J Neurosci Res ; 68(5): 588-94, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12111848

RESUMEN

The twitcher mouse is a murine model of human globoid cell leukodystrophy (GLD; Krabbe disease) caused by a genetic defect in the activity of galactosylceramidase (GALC). An accumulation of cytotoxic metabolite, galactosylsphingosine (psychosine), in myelin forming cells (oligodendrocytes and Schwann cells) of the twitcher mouse as well as patients with GLD has been suggested to cause dysfunction of these cells and subsequent demyelination in the central and peripheral nervous system. To investigate further the cellular pathomechanism of GLD, we established spontaneously immortalized Schwann cell lines from the twitcher mouse. Long-term cultures of Schwann cells derived from dorsal root ganglia and consecutive peripheral nerves of 3-week-old twitcher mice were maintained for 6 months, and spontaneously developed colonies were expanded further and characterized. One of the cell lines, designated TwS1, showed distinct Schwann cell phenotypes, was passaged twice a week and maintained for over 10 months without phenotypic alterations. The TwS1 cells had a nonsense mutation in the GALC genome, and showed markedly reduced GALC activity and elevated psychosine levels. Ultrastructurally, varieties of cytoplasmic inclusions were demonstrated in TwS1 cells. When TwS1 cells were infected with a retrovirus vector encoding GALC, GALC activity was markedly increased and psychosine levels were significantly decreased. These immortalized Schwann cells can be useful in studies on the nervous system lesions in GLD.


Asunto(s)
Línea Celular Transformada/citología , Leucodistrofia de Células Globoides/genética , Células de Schwann/citología , Adenoviridae/genética , Animales , Línea Celular Transformada/enzimología , Galactosilceramidasa/genética , Galactosilceramidasa/metabolismo , Galactosilceramidas/metabolismo , Cuerpos de Inclusión/ultraestructura , Ratones Mutantes Neurológicos , Microscopía Electrónica , Fenotipo , Psicosina/metabolismo
20.
J Neurosci Res ; 74(2): 266-77, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14515356

RESUMEN

In the present study, we investigated the feasibility of using human neural stem cells (NSCs) in the treatment of diffuse central nervous system (CNS) alterations in a murine model of mucopolysaccharidosis VII (MPS VII), a lysosomal storage disease caused by a genetic defect in the beta-glucuronidase gene. An immortalized NSC line derived from human fetal telencephalon was genetically engineered to overexpress beta-glucuronidase and transplanted into the cerebral ventricles of neonatal MPS VII mouse. Transplanted human NSCs were found to integrate and migrate in the host brain and to produce large amount of beta-glucuronidase. Brain contents of the substrates of beta-glucuronidase were reduced to nearly normal levels, and widespread clearing of lysosomal storage was observed in the MPS VII mouse brain at 25 days posttransplantation. The number of engrafted cells decreased markedly after the transplantation, and it appears that the major cause of the cell death was not the immune response of the host but apoptotic cell death of grafted human NSCs. Results showed that human NSCs would serve as a useful gene transfer vehicle for the treatment of diffuse CNS lesions in human lysosomal storage diseases and are potentially applicable in the treatment of patients suffering from neurological disorders.


Asunto(s)
Trasplante de Tejido Encefálico/métodos , Ingeniería Genética/métodos , Glucuronidasa/deficiencia , Mucopolisacaridosis VII/terapia , Trasplante de Células Madre/métodos , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Encéfalo/patología , Encéfalo/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Feto , Vectores Genéticos/uso terapéutico , Glucuronidasa/biosíntesis , Glucuronidasa/genética , Supervivencia de Injerto/fisiología , Humanos , Lisosomas/metabolismo , Ratones , Ratones Mutantes Neurológicos , Mucopolisacaridosis VII/metabolismo , Mucopolisacaridosis VII/fisiopatología , Retroviridae/genética , Trasplante Heterólogo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA