Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Hum Mol Genet ; 31(6): 914-928, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-34617103

RESUMEN

Glycogen Storage Disease Type I (GSDI) is an inherited disease caused by glucose-6 phosphatase (G6Pase) deficiency, leading to a loss of endogenous glucose production and severe hypoglycemia. Moreover, most GSDI patients develop a chronic kidney disease (CKD) due to lipid accumulation in the kidney. Similar to diabetic CKD, activation of renin-angiotensin system (RAS) promotes renal fibrosis in GSDI. Here, we investigated the physiological and molecular effects of RAS blockers in GSDI patients and mice. A retrospective analysis of renal function was performed in 21 GSDI patients treated with RAS blockers. Cellular and metabolic impacts of RAS blockade were analyzed in K.G6pc-/- mice characterized by G6pc1 deletion in kidneys. GSDI patients started RAS blocker treatment at a median age of 21 years and long-term treatment reduced the progression of CKD in about 50% of patients. However, CKD progressed to kidney failure in 20% of treated patients, requiring renal transplantation. In K.G6pc-/- mice, CKD was associated with an impairment of autophagy and ER stress. RAS blockade resulted in a rescue of autophagy and decreased ER stress, concomitantly with decreased fibrosis and improved renal function, but without impact on glycogen and lipid contents. In conclusion, these data confirm the partial beneficial effect of RAS blockers in the prevention of CKD in GSDI. Mechanistically, we show that these effects are linked to a reduction of cell stress, without affecting metabolism.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo I , Insuficiencia Renal Crónica , Animales , Femenino , Glucosa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/complicaciones , Enfermedad del Almacenamiento de Glucógeno Tipo I/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Humanos , Lípidos , Masculino , Ratones , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/genética , Sistema Renina-Angiotensina/genética , Estudios Retrospectivos
2.
J Hepatol ; 69(5): 1074-1087, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30193922

RESUMEN

BACKGROUND & AIMS: Glycogen storage disease type Ia (GSDIa) is a rare genetic disease associated with glycogen accumulation in hepatocytes and steatosis. With age, most adult patients with GSDIa develop hepatocellular adenomas (HCA), which can progress to hepatocellular carcinomas (HCC). In this study, we characterized metabolic reprogramming and cellular defense alterations during tumorigenesis in the liver of hepatocyte-specific G6pc deficient (L.G6pc-/-) mice, which develop all the hepatic hallmarks of GSDIa. METHODS: Liver metabolism and cellular defenses were assessed at pretumoral (four months) and tumoral (nine months) stages in L.G6pc-/- mice fed a high fat/high sucrose (HF/HS) diet. RESULTS: In response to HF/HS diet, hepatocarcinogenesis was highly accelerated since 85% of L.G6pc-/- mice developed multiple hepatic tumors after nine months, with 70% classified as HCA and 30% as HCC. Tumor development was associated with high expression of malignancy markers of HCC, i.e. alpha-fetoprotein, glypican 3 and ß-catenin. In addition, L.G6pc-/- livers exhibited loss of tumor suppressors. Interestingly, L.G6pc-/- steatosis exhibited a low-inflammatory state and was less pronounced than in wild-type livers. This was associated with an absence of epithelial-mesenchymal transition and fibrosis, while HCA/HCC showed a partial epithelial-mesenchymal transition in the absence of TGF-ß1 increase. In HCA/HCC, glycolysis was characterized by a marked expression of PK-M2, decreased mitochondrial OXPHOS and a decrease of pyruvate entry in the mitochondria, confirming a "Warburg-like" phenotype. These metabolic alterations led to a decrease in antioxidant defenses and autophagy and chronic endoplasmic reticulum stress in L.G6pc-/- livers and tumors. Interestingly, autophagy was reactivated in HCA/HCC. CONCLUSION: The metabolic remodeling in L.G6pc-/- liver generates a preneoplastic status and leads to a loss of cellular defenses and tumor suppressors that facilitates tumor development in GSDI. LAY SUMMARY: Glycogen storage disease type Ia (GSD1a) is a rare metabolic disease characterized by hypoglycemia, steatosis, excessive glycogen accumulation and tumor development in the liver. In this study, we have observed that GSDIa livers reprogram their metabolism in a similar way to cancer cells, which facilitates tumor formation and progression, in the absence of hepatic fibrosis. Moreover, hepatic burden due to overload of glycogen and lipids in the cells leads to a decrease in cellular defenses, such as autophagy, which could further promote tumorigenesis in the case of GSDI.


Asunto(s)
Carcinoma Hepatocelular/etiología , Enfermedad del Almacenamiento de Glucógeno Tipo I/complicaciones , Neoplasias Hepáticas/etiología , Hígado/metabolismo , Animales , Autofagia , Dieta Alta en Grasa , Estrés del Retículo Endoplásmico , Transición Epitelial-Mesenquimal , Glucosa/metabolismo , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Sacarosa/administración & dosificación
3.
J Inherit Metab Dis ; 41(6): 955-963, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29869165

RESUMEN

Glycogen storage disease type I (GSDI) is a rare genetic pathology characterized by glucose-6 phosphatase (G6Pase) deficiency, translating in hypoglycemia during short fasts. Besides metabolic perturbations, GSDI patients develop long-term complications, especially chronic kidney disease (CKD). In GSDI patients, CKD is characterized by an accumulation of glycogen and lipids in kidneys, leading to a gradual decline in renal function. At a molecular level, the activation of the renin-angiotensin system is responsible for the development of renal fibrosis, eventually leading to renal failure. The same CKD phenotype was observed in a mouse model with a kidney-specific G6Pase deficiency (K.G6pc-/- mice). Furthermore, GSDI patients and mice develop frequently renal cysts at late stages of the nephropathy, classifying GSDI as a potential polycystic kidney disease (PKD). PKDs are genetic disorders characterized by multiple renal cyst formation, frequently caused by the loss of expression of polycystic kidney genes, such as PKD1/2 and PKHD1. Interestingly, these genes are deregulated in K.G6pc-/- kidneys, suggesting their possible role in GSDI cystogenesis. Finally, renal cysts are known to predispose to renal malignancy development. In addition, HNF1B loss is a malignancy prediction factor. Interestingly, Hnf1b expression was decreased in K.G6pc-/- kidneys. While a single case of renal cancer has been reported in a GSDI patient, a clear cell renal carcinoma was recently observed in one K.G6pc-/- mouse (out of 36 studied mice) at a later stage of the disease. This finding highlights the need to further analyze renal cyst development in GSDI patients in order to evaluate the possible associated risk of carcinogenesis, even if the risk might be limited.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo I/complicaciones , Enfermedades Renales Poliquísticas/etiología , Insuficiencia Renal Crónica/etiología , Animales , Modelos Animales de Enfermedad , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Factor Nuclear 1-beta del Hepatocito/genética , Humanos , Neoplasias Renales/etiología , Neoplasias Renales/patología , Ratones , Ratones Noqueados , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Insuficiencia Renal Crónica/patología
4.
Hum Gene Ther ; 30(10): 1263-1273, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31319709

RESUMEN

Glycogen storage diseases (GSDs) type I (GSDI) and type III (GSDIII), the most frequent hepatic GSDs, are due to defects in glycogen metabolism, mainly in the liver. In addition to hypoglycemia and liver pathology, renal, myeloid, or muscle complications affect GSDI and GSDIII patients. Currently, patient management is based on dietary treatment preventing severe hypoglycemia and increasing the lifespan of patients. However, most of the patients develop long-term pathologies. In the past years, gene therapy for GSDI has generated proof of concept for hepatic GSDs. This resulted in a recent clinical trial of adeno-associated virus (AAV)-based gene replacement for GSDIa. However, the current limitations of AAV-mediated gene transfer still represent a challenge for successful gene therapy in GSDI and GSDIII. Indeed, transgene loss over time was observed in GSDI liver, possibly due to the degeneration of hepatocytes underlying the physiopathology of both GSDI and GSDIII and leading to hepatic tumor development. Moreover, multitissue targeting requires high vector doses to target nonpermissive tissues such as muscle and kidney. Interestingly, recent pharmacological interventions or dietary regimen aiming at the amelioration of the hepatocyte abnormalities before the administration of gene therapy demonstrated improved efficacy in GSDs. In this review, we describe the advances in gene therapy and the limitations to be overcome to achieve efficient and safe gene transfer in GSDs.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/terapia , Enfermedad del Almacenamiento de Glucógeno Tipo I/terapia , Hipoglucemia/terapia , Animales , Ensayos Clínicos como Asunto , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Glucógeno/biosíntesis , Enfermedad del Almacenamiento de Glucógeno Tipo I/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/patología , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/patología , Hepatocitos/enzimología , Hepatocitos/patología , Humanos , Hipoglucemia/enzimología , Hipoglucemia/genética , Hipoglucemia/patología , Hígado/enzimología , Hígado/patología , Transgenes
5.
Mol Metab ; 16: 100-115, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30100243

RESUMEN

OBJECTIVE: Ectopic lipid accumulation in the liver and kidneys is a hallmark of metabolic diseases leading to non-alcoholic fatty liver disease (NAFLD) and chronic kidney disease (CKD). Moreover, recent data have highlighted a strong correlation between NAFLD and CKD incidences. In this study, we use two mouse models of hepatic steatosis or CKD, each initiated independently of the other upon the suppression of glucose production specifically in the liver or kidneys, to elucidate the mechanisms underlying the development of CKD in the context of NAFLD-like pathology. METHODS: Mice with a deletion of G6pc, encoding glucose-6 phosphatase catalytic subunit, specifically in the liver (L.G6pc-/- mice) or the kidneys (K.G6pc-/- mice), were fed with either a standard diet or a high fat/high sucrose (HF/HS) diet during 9 months. These mice represent two original models of a rare metabolic disease named Glycogen Storage Disease Type Ia (GSDIa) that is characterized by both NAFLD-like pathology and CKD. Two other groups of L.G6pc-/- and K.G6pc-/- mice were fed a standard diet for 6 months and then treated with fenofibrate for 3 months. Lipid and glucose metabolisms were characterized, and NAFLD-like and CKD damages were evaluated. RESULTS: Lipid depot exacerbation upon high-calorie diet strongly accelerated hepatic and renal pathologies induced by the G6pc-deficiency. In L.G6pc-/- mice, HF/HS diet increased liver injuries, characterized by higher levels of plasmatic transaminases and increased hepatic tumor incidence. In K.G6pc-/- mice, HF/HS diet increased urinary albumin and lipocalin 2 excretion and aggravated renal fibrosis. In both cases, the worsening of NAFLD-like injuries and CKD was independent of glycogen content. Furthermore, fenofibrate, via the activation of lipid oxidation significantly decreased the hepatic or renal lipid accumulations and prevented liver or kidney damages in L.G6pc-/- and K.G6pc-/- mice, respectively. Finally, we show that L.G6pc-/- mice and K.G6pc-/- mice developed NAFLD-like pathology and CKD independently. CONCLUSIONS: This study highlights the crucial role that lipids play in the independent development of both NAFLD and CKD and demonstrates the importance of lipid-lowering treatments in various metabolic diseases featured by lipid load, from the "rare" GSDIa to the "epidemic" morbid obesity or type 2 diabetes.


Asunto(s)
Glucosa-6-Fosfatasa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Metabolismo de los Lípidos/fisiología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Glucosa/metabolismo , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/fisiopatología , Espacio Intracelular , Riñón/metabolismo , Producto de la Acumulación de Lípidos/fisiología , Metabolismo de los Lípidos/genética , Lípidos/fisiología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA