Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pharmacol Res ; 178: 106155, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35248699

RESUMEN

The XELOX chemotherapy protocol that includes capecitabine and oxaliplatin is the routine treatment for colorectal cancer (CRC), but it can cause chemotherapy-related adverse events such as thrombocytopenia (TCP). To identify predictive biomarkers and clarify the mechanism of TCP susceptibility, we conducted integrative analysis using normal colorectal tissue (CRT), plasma, and urine samples collected before CRC patients received adjuvant XELOX chemotherapy. RNA-sequencing and DNA methylation arrays were performed on CRT samples, while liquid chromatography-mass spectrometry was performed on CRT, plasma, and urine samples. Differentially expressed features (DEFs) from each uni-omics analysis were then subjected to integrative analysis using Multi-Omics Factor Analysis (MOFA). Choline-deficiency in plasma and CRT was found as the most critical TCP-related feature. Based on bioinformatic analysis and literature research, we further concluded that choline-deficiency was the possible reason for most of the other TCP-related multi-omics DEFs, including metabolites representing reduced sphingolipid de novo synthesis and elevated solute carrier-mediated transmembrane transportation in CRT and plasma, DNA hypermethylation and elevated expression of genes involved in neuronal system genes. In terms of thrombocytopoiesis, these TCP-related DEFs may cause atypical maintenance and differentiation of megakaryocyte, resulting a suppressed ability of thrombocytopoiesis, making patients more susceptible to chemotherapy-induced TCP. At last, prediction models were developed and validated with reasonably good discrimination. The area under curves (AUCs) of training sets were all > 0.9, while validation sets had AUCs between 0.778 and 0.926. In conclusion, our results produced reliable marker systems for predicting TCP and promising target for developing precision treatment to prevent TCP.


Asunto(s)
Antineoplásicos , Deficiencia de Colina , Neoplasias Colorrectales , Leucopenia , Trombocitopenia , Antineoplásicos/efectos adversos , Colina , Deficiencia de Colina/inducido químicamente , Deficiencia de Colina/tratamiento farmacológico , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Fluorouracilo/uso terapéutico , Humanos , Leucopenia/inducido químicamente , Trombocitopenia/inducido químicamente
2.
FASEB J ; 33(8): 9334-9349, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31120771

RESUMEN

Methyl-donor deficiency is a risk factor for neurodegenerative diseases. Dietary deficiency of the methyl-donors methionine and choline [methionine-choline-deficient (MCD) diet] is a well-established model of nonalcoholic steatohepatitis (NASH), yet brain metabolism has not been studied in this model. We hypothesized that supplemental betaine would protect both the liver and brain in this model and that any benefit to the brain would be due to improved liver metabolism because betaine is a methyl-donor in liver methylation but is not metabolically active in the brain. We fed male Sprague-Dawley rats a control diet, MCD diet, or betaine-supplemented MCD (MCD+B) diet for 8 wk and collected blood and tissue. As expected, betaine prevented MCD diet-induced NASH. However, contrary to our prediction, it did not appear to do so by stimulating methylation; the MCD+B diet worsened hyperhomocysteinemia and depressed liver methylation potential 8-fold compared with the MCD diet. Instead, it significantly increased the expression of genes involved in ß-oxidation: fibroblast growth factor 21 and peroxisome proliferator-activated receptor α. In contrast to that of the liver, brain methylation potential was unaffected by diet. Nevertheless, several phospholipid (PL) subclasses involved in stabilizing brain membranes were decreased by the MCD diet, and these improved modestly with betaine. The protective effect of betaine is likely due to the stimulation of ß-oxidation in liver and the effects on PL metabolism in brain.-Abu Ahmad, N., Raizman, M., Weizmann, N., Wasek, B., Arning, E., Bottiglieri, T., Tirosh, O., Troen, A. M. Betaine attenuates pathology by stimulating lipid oxidation in liver and regulating phospholipid metabolism in brain of methionine-choline-deficient rats.


Asunto(s)
Betaína/uso terapéutico , Deficiencia de Colina/tratamiento farmacológico , Deficiencia de Colina/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Metionina/deficiencia , Metionina/metabolismo , Fosfolípidos/metabolismo , Animales , Western Blotting , Masculino , Aprendizaje por Laberinto , Ratas , Ratas Sprague-Dawley
3.
Mar Drugs ; 17(2)2019 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-30744124

RESUMEN

Nonalcoholic steatohepatitis (NASH) progresses from nonalcoholic fatty liver disease (NAFLD); however, efficacious drugs for NASH treatment are lacking. Sodium alginate (SA), a soluble dietary fiber extracted from brown algae, could protect the small intestine from enterobacterial invasion. NASH pathogenesis has been suggested to be associated with enterobacterial invasion, so we examined the effect of SA on methionine- and choline-deficient (MCD) diet-induced steatohepatitis in mice (the most widely-used model of NASH). The mice (n = 31) were divided into three groups (mice fed with regular chow, MCD diet, and MCD diet premixed with 5% SA) for 4 and 8 weeks. The MCD diet increased lipid accumulation and inflammation in the liver, the NAFLD Activity Score and hepatic mRNA expression of tumor necrosis factor- and collagen 11, and induced macrophage infiltration. Villus shortening, disruption of zonula occludens-1 localization and depletion of mucus production were observed in the small intestine of the MCD-group mice. SA administration improved lipid accumulation and inflammation in the liver, and impaired barrier function in the small intestine. Collectively, these results suggest that SA is useful for NASH treatment because it can prevent hepatic inflammation and fatty degeneration by maintaining intestinal barrier function.


Asunto(s)
Alginatos/farmacología , Hígado Graso/tratamiento farmacológico , Metionina/deficiencia , Animales , Deficiencia de Colina/tratamiento farmacológico , Deficiencia de Colina/metabolismo , Deficiencia de Colina/patología , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/patología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Intestino Delgado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
4.
Biochem Pharmacol ; 222: 116073, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38395263

RESUMEN

Stem cells from the apical papilla(SCAPs) exhibit remarkable tissue repair capabilities, demonstrate anti-inflammatory and pro-angiogenic effects, positioning them as promising assets in the realm of regenerative medicine. Recently, the focus has shifted towards exosomes derived from stem cells, perceived as safer alternatives while retaining comparable physiological functions. This study delves into the therapeutic implications of exosomes derived from SCAPs in the methionine-choline-deficient (MCD) diet-induced mice non-alcoholic steatohepatitis (NASH) model. We extracted exosomes from SCAPs. During the last two weeks of the MCD diet, mice were intravenously administered SCAPs-derived exosomes at two distinct concentrations (50 µg/mouse and 100 µg/mouse) biweekly. Thorough examinations of physiological and biochemical indicators were performed to meticulously evaluate the impact of exosomes derived from SCAPs on the advancement of NASH in mice induced by MCD diet. This findings revealed significant reductions in body weight loss and liver damage induced by the MCD diet following exosomes treatment. Moreover, hepatic fat accumulation was notably alleviated. Mechanistically, the treatment with exosomes led to an upregulation of phosphorylated adenosine monophosphate-activated protein kinase (p-AMPK) levels in the liver, enhancing hepatic fatty acid oxidation and transporter gene expression while inhibiting genes associated with fatty acid synthesis. Additionally, exosomes treatment increased the transcription levels of key liver mitochondrial marker proteins and the essential mitochondrial biogenesis factor. Furthermore, the levels of serum inflammatory factors and hepatic tissue inflammatory factor mRNA expression were significantly reduced, likely due to the anti-inflammatory phenotype induced by exosomes in macrophages. The above conclusion suggests that SCAPs-exosomes can improve NASH.


Asunto(s)
Deficiencia de Colina , Exosomas , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Metionina/metabolismo , Colina/metabolismo , Metabolismo de los Lípidos , Exosomas/metabolismo , Deficiencia de Colina/complicaciones , Deficiencia de Colina/tratamiento farmacológico , Deficiencia de Colina/metabolismo , Hígado/metabolismo , Inflamación/metabolismo , Racemetionina/metabolismo , Racemetionina/farmacología , Antiinflamatorios/farmacología , Dieta , Ácidos Grasos/metabolismo , Ratones Endogámicos C57BL
5.
Toxicol Appl Pharmacol ; 268(3): 264-77, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23481610

RESUMEN

Nonalcoholic steatohepatitis (NASH) is characterized by combined pathology of steatosis, lobular inflammation, fibrosis, and hepatocellular degeneration, with systemic symptoms of diabetes or hyperlipidemia, all in the absence of alcohol abuse. Given the therapeutic importance and conflicting findings regarding the potential for healing the histopathologic features of NASH in humans, particularly fibrosis, we investigated the reversibility of NASH-related findings in Wistar rats fed a choline-deficient and iron-supplemented l-amino acid-defined (CDAA) diet for 12weeks, with a recovery period of 7weeks, during which the diets were switched to a choline-sufficient and iron-supplemented l-amino acid-defined (CSAA) one. Analysis showed that steatosis and inflammation were significantly resolved by the end of the recovery period, along with decreases in AST and ALT activities within 4weeks. In contrast, fibrosis remained even after the recovery period, to an extent similar to that in continuously CDAA-fed animals. Real-time reverse transcriptase-polymerase chain reaction, Western blot, and immunohistochemical investigations revealed that expression of some factors indicating oxidative stress (CYP2E1, 4-HNE, and iNOS) were elevated, whereas catalase and SOD1 were decreased, and a hypoxic state and CD34-positive neovascularization were evident even after the recovery period, although the fibrogenesis pathway by activated α-SMA-positive hepatic stellate cells via TGF-ß and TIMPs decreased to the CSAA group level. In conclusion, persistent fibrosis was noted after the recovery period of 7weeks, possibly due to sustained hypoxia and oxidative stress supposedly caused by capillarization. Otherwise, histopathological features of steatosis and inflammation, as well as serum AST and ALT activities, were recovered.


Asunto(s)
Aminoácidos/administración & dosificación , Deficiencia de Colina/metabolismo , Colina/administración & dosificación , Hígado Graso/metabolismo , Hierro/administración & dosificación , Hierro/toxicidad , Cirrosis Hepática/metabolismo , Aminoácidos/toxicidad , Animales , Deficiencia de Colina/tratamiento farmacológico , Suplementos Dietéticos , Hígado Graso/inducido químicamente , Hígado Graso/patología , Cirrosis Hepática/patología , Masculino , Ratas , Ratas Wistar
6.
J Hepatol ; 54(5): 1002-10, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21145825

RESUMEN

BACKGROUND & AIMS: Liver steatosis enhances ischemia/reperfusion (I/R) injury and is considered a primary factor in graft failure after liver transplantation. Although previous reports have shown a role for qualitative steatosis (macrovesicular vs. microvesicular) in hepatic I/R injury, no studies have compared side by side the specific contribution of individual lipids accumulating in fatty liver to I/R damage. METHODS: We used nutritional and genetic models of micro and macrovesicular fatty livers exhibiting specific lipid profiles to assess their susceptibility to normothermic I/R injury. RESULTS: Unlike choline-deficient (CD) diet-fed mice, characterized by predominant liver triglycerides/free fatty acids (TG/FFA) accumulation, mice fed a cholesterol-enriched (HC) diet, which exhibited enhanced hepatic cholesterol loading in mitochondria, were highly sensitive to I/R-induced liver injury. In vivo two-photon confocal imaging revealed enhanced mitochondrial depolarization and generation of reactive oxygen species following hepatic I/R in HC-fed but not in CD-fed mice, consistent with decreased mitochondrial GSH (mGSH) observed in HC-fed mice. Moreover, ob/ob mice, characterized by increased hepatic TG, FFA, and cholesterol levels, were as sensitive to I/R-mediated liver injury as mice fed the HC diet. Livers from ob/ob mice displayed increased StAR expression and mitochondrial cholesterol accumulation, resulting in mGSH depletion. Interestingly, atorvastatin therapy or squalene synthase inhibition in vivo attenuated StAR overexpression, mitochondrial cholesterol loading, and mGSH depletion, protecting ob/ob mice from I/R-mediated liver injury. CONCLUSIONS: Cholesterol accumulation, particularly in mitochondria, sensitizes to hepatic I/R injury, and thus represents a novel target to prevent the enhanced damage of steatotic livers to I/R-mediated damage.


Asunto(s)
Anticolesterolemiantes/farmacología , Colesterol en la Dieta/farmacocinética , Hígado Graso/tratamiento farmacológico , Ácidos Heptanoicos/farmacología , Ácido Mevalónico/metabolismo , Pirroles/farmacología , Daño por Reperfusión/prevención & control , Animales , Atorvastatina , Colina/farmacología , Deficiencia de Colina/tratamiento farmacológico , Deficiencia de Colina/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Inhibidores Enzimáticos/farmacología , Farnesil Difosfato Farnesil Transferasa/antagonistas & inhibidores , Hígado Graso/metabolismo , Hígado Graso/patología , Glutatión/metabolismo , Lipotrópicos/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Obesidad/metabolismo , Obesidad/patología , Quinuclidinas/farmacología , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo
7.
Liver Int ; 29(7): 988-96, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19386026

RESUMEN

BACKGROUND/AIMS: Non-alcoholic fatty liver disease and non-alcoholic steatohepatitis (NASH) are the hepatic manifestation of metabolic syndrome. However, its therapeutic strategy has not been established. Recently, an angiotensin II type 1 receptor blocker, telmisartan (Tel), has received a great deal of attention as a therapeutic tool for metabolic syndrome. The aim of this study was to investigate the efficacy and mechanisms of Tel on a murine NASH model. METHODS: C57BL/6 mice were fed a methionine- and choline-deficient high-fat diet (MCDHF) or a standard diet with/without the administration of Tel (10 mg/kg/day) for 8 weeks. RESULTS: MCDHF feeding induced marked steatohepatitis with macrophage infiltration. Tel attenuated liver steatosis with decreased hepatic triglycerides (P<0.05) and fibrogenesis with decreased type I collagen and transforming growth factor-beta1 mRNA expressions (P<0.05). Tel also suppressed the infiltration of macrophages into the liver and decreased hepatic monocyte chemoattractant protein-1 and its receptor (CC-chemokine receptor 2; CCR2) mRNA expressions, especially CCR2. In vitro, Tel suppressed CCR2 expression, which was induced by low-density lipoprotein. The size of adipocyte in visceral fat tissue was reduced with an increased serum adiponectin concentration in the Tel group. CONCLUSIONS: In this study, we revealed that Tel attenuated steatohepatitis progression by suppressing the macrophage infiltration into the liver. Tel also affected the reduction of adipocyte size and elevation of serum adiponectin. Tel might serve as a new therapeutic strategy for NASH.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bencimidazoles/farmacología , Benzoatos/farmacología , Hígado Graso/prevención & control , Grasa Intraabdominal/efectos de los fármacos , Cirrosis Hepática/prevención & control , Hígado/efectos de los fármacos , Macrófagos/efectos de los fármacos , Adipocitos/efectos de los fármacos , Adipocitos/patología , Adiponectina/sangre , Animales , Presión Sanguínea/efectos de los fármacos , Línea Celular , Tamaño de la Célula , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Deficiencia de Colina/complicaciones , Deficiencia de Colina/tratamiento farmacológico , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/patología , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/metabolismo , Receptores CCR2/genética , Receptores CCR2/metabolismo , Telmisartán , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Triglicéridos/metabolismo
8.
J Pineal Res ; 46(4): 401-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19552763

RESUMEN

Nonalcoholic steatohepatitis (NASH) may progress to advanced fibrosis and cirrhosis. Mainly, oxidative stress and excessive hepatocyte apoptosis are implicated in the pathogenesis of progressive NASH. Melatonin is not only a powerful antioxidant but also an anti-inflammatory and anti-apoptotic agent. We aimed to evaluate the effects of melatonin on methionine- and choline-deficient diet (MCDD)-induced NASH in rats. Thirty-two male Wistar rats were divided into four groups. Two groups were fed with MCDD while the other two groups were fed a control diet, pair-fed. One of the MCDD groups and one of the control diet groups were administered melatonin 50 mg/kg/day intraperitoneally, and the controls were given a vehicle. After 1 month the liver tissue oxidative stress markers, proinflammatory cytokines and hepatocyte apoptosis were studied by commercially available kits. For grading and staging histological lesions, Brunt et al.'s system was used. Melatonin decreased oxidative stress, proinflammatory cytokines and hepatocyte apoptosis. The drug ameliorated the grade of NASH. The present study suggests that melatonin functions as a potent antioxidant, anti-inflammatory and antiapoptotic agent in NASH and may be a therapeutic option.


Asunto(s)
Deficiencia de Colina/metabolismo , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Melatonina/farmacología , Metionina/deficiencia , Animales , Apoptosis/efectos de los fármacos , Biomarcadores , Colina/metabolismo , Deficiencia de Colina/sangre , Deficiencia de Colina/tratamiento farmacológico , Citocinas/sangre , Dieta , Hígado Graso/sangre , Glutatión/metabolismo , Histocitoquímica , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Malondialdehído/metabolismo , Metionina/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Estadísticas no Paramétricas , Superóxido Dismutasa/metabolismo
9.
Nutrients ; 11(3)2019 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-30889905

RESUMEN

BACKGROUND: Choline is essential for the synthesis of liver phosphatidylcholine (PC), parenchymal maintenance, bile formation, and lipoprotein assembly to secrete triglycerides. In choline deficiency, the liver accretes choline/PC at the expense of lung tissue, thereby impairing pulmonary PC homoeostasis. In cystic fibrosis (CF), exocrine pancreas insufficiency results in impaired cleavage of bile PC and subsequent fecal choline loss. In these patients, the plasma choline concentration is low and correlates with lung function. We therefore investigated the effect of choline supplementation on plasma choline/PC concentration and metabolism, lung function, and liver fat. METHODS: 10 adult male CF patients were recruited (11/2014⁻1/2016), and orally supplemented with 3 × 1 g choline chloride for 84 (84⁻91) days. Pre-/post-supplementation, patients were spiked with 3.6 mg/kg [methyl-D9]choline chloride to assess choline/PC metabolism. Mass spectrometry, spirometry, and hepatic nuclear resonance spectrometry served for analysis. RESULTS: Supplementation increased plasma choline from 4.8 (4.1⁻6.2) µmol/L to 10.5 (8.5⁻15.5) µmol/L at d84 (p < 0.01). Whereas plasma PC concentration remained unchanged, D9-labeled PC was decreased (12.2 [10.5⁻18.3] µmol/L vs. 17.7 [15.5⁻22.4] µmol/L, p < 0.01), indicating D9-tracer dilution due to higher choline pools. Supplementation increased Forced Expiratory Volume in 1 second percent of predicted (ppFEV1) from 70.0 (50.9⁻74.8)% to 78.3 (60.1⁻83.9)% (p < 0.05), and decreased liver fat from 1.58 (0.37⁻8.82)% to 0.84 (0.56⁻1.17)% (p < 0.01). Plasma choline returned to baseline concentration within 60 h. CONCLUSIONS: Choline supplementation normalized plasma choline concentration and increased choline-containing PC precursor pools in adult CF patients. Improved lung function and decreased liver fat suggest that in CF correcting choline deficiency is clinically important. Choline supplementation of CF patients should be further investigated in randomized, placebo-controlled trials.


Asunto(s)
Deficiencia de Colina/tratamiento farmacológico , Colina/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Volumen Espiratorio Forzado/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Adolescente , Adulto , Colina/sangre , Colina/farmacología , Deficiencia de Colina/sangre , Deficiencia de Colina/complicaciones , Fibrosis Quística/sangre , Fibrosis Quística/patología , Fibrosis Quística/fisiopatología , Suplementos Dietéticos , Insuficiencia Pancreática Exocrina/sangre , Insuficiencia Pancreática Exocrina/complicaciones , Insuficiencia Pancreática Exocrina/tratamiento farmacológico , Hígado Graso/sangre , Hígado Graso/etiología , Hígado Graso/prevención & control , Humanos , Hígado/metabolismo , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Fosfatidilcolinas/sangre , Triglicéridos/sangre , Adulto Joven
10.
J Gastroenterol Hepatol ; 23(12): 1909-16, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18422963

RESUMEN

BACKGROUND AND AIM: The effect of polaprezinc, a zinc-carnosine chelate compound, on the development of non-alcoholic steatohepatitis (NASH) was investigated in dietary methionine and choline deficient (MCD) mice. METHODS: Mice were fed the MCD diet with or without polaprezinc (2.2 g/kg diet) for 10 weeks. Liver histopathology, triglyceride and lipid peroxide levels, and the expression of genes linked to fibrosis were then assessed. RESULTS: MCD mice developed steatohepatitis accompanied by mild fibrosis with an increase in lipid peroxidation, hepatic stellate cell (HSC) activation, and the augmented mRNA expression of tumor necrosis factor-alpha, transforming growth factor-beta1 and procollagen alpha1(I). The mRNA expression levels of matrix metalloproteinase (MMP)-2 and tissue inhibitors of metalloproteinase (TIMP)-1 and TIMP-2 were also enhanced. Histopathologically, polaprezinc supplementation did not influence the development of steatosis but it apparently attenuated fibrosis. Polaprezinc slightly reduced lipid peroxidation and suppressed HSC activation as well as the mRNA expression of pro-inflammatory cytokines. Polaprezinc affected the MCD diet-enhanced expression of TIMP-1 even when administered relatively late. CONCLUSION: These results suggest that polaprezinc attenuates fibrosis in NASH by reducing inflammation and lipid peroxidation and, during a later phase, promoting fibrolysis via the inhibition of TIMP expression in the liver. Further investigation is required to clarify the clinical efficacy of polaprezinc in patients with NASH.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Carnosina/análogos & derivados , Hígado Graso/tratamiento farmacológico , Células Estrelladas Hepáticas/efectos de los fármacos , Cirrosis Hepática/prevención & control , Hígado/efectos de los fármacos , Compuestos Organometálicos/farmacología , Alanina Transaminasa/sangre , Animales , Peso Corporal/efectos de los fármacos , Carnosina/farmacología , Deficiencia de Colina/complicaciones , Deficiencia de Colina/tratamiento farmacológico , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/patología , Regulación de la Expresión Génica/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Peroxidación de Lípido/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Compuestos de Zinc/farmacología
11.
Inflammation ; 31(2): 91-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18066656

RESUMEN

To assess the effects of anti-TNF-alpha antibody (infliximab) in experimental steatohepatitis induced by methionine- and choline-deficient (MCD) diet. The study included thirty rats. One group received normal rat food, and two groups received MCD diet. The treatment group received a single dose intra-peritoneal infliximab (4 mg/kg), at week 8. MCD diet increased levels of AST, ALT, TNF-alpha, TGF-beta(1), tissue and plasma MDA (p < 0.05 for each). Moreover, it led to steatosis, ballooning degeneration, inflammation, fibrosis and increased actin expression, histopathologically (p < 0.05 for each). In this experimental steatohepatitis anti-TNF-alpha antibody decreased the levels of AST, ALT, TGF-beta(1) and plasma and tissue MDA (p < 0.05 for each). Moreover, inflammation, necrosis, actin expression and fibrosis decreased in anti-TNF-alpha group compared to placebo group (p < 0.05 for each). This study indicates that anti-TNF-alpha antibody is effective on necrosis, inflammation and fibrosis in the experimental model of non-alcoholic steatohepatitis, induced by MCD diet.


Asunto(s)
Antiinflamatorios/farmacología , Anticuerpos Monoclonales/farmacología , Deficiencia de Colina/tratamiento farmacológico , Hígado Graso/prevención & control , Hepatitis/prevención & control , Cirrosis Hepática/prevención & control , Metionina/deficiencia , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Actinas/metabolismo , Alanina Transaminasa/sangre , Animales , Antiinflamatorios/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Aspartato Aminotransferasas/sangre , Deficiencia de Colina/complicaciones , Deficiencia de Colina/patología , Dieta , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/patología , Hepatitis/etiología , Hepatitis/patología , Infliximab , Inyecciones Intraperitoneales , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Masculino , Malondialdehído/metabolismo , Necrosis , Ratas , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
12.
Biofactors ; 44(3): 289-298, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29672963

RESUMEN

One of the worldwide metabolic health dilemma is nonalcoholic fatty liver diseases (NAFLD). Researchers are searching effective drug to manage NAFLD patients. One of the best way to manage the metabolic imperfection is through natural principal isolated from different sources. Butein, a natural compound known to have numerous pharmacological application. In the current study we assessed the therapeutic effect of butein administration on liver function tests, oxidative stress, antioxidants, lipid abnormalities, serum inflammatory cytokines, and mitochondrial reactive oxygen species levels, in rats with methionine-choline deficient (MCD) diet induced NAFLD. Male Wistar rats were treated with MCD diet with/without butein (200 mg/kg body wt. orally) for 6 weeks. The protective effect of butein, were evident from decreased transaminase activities, restoration of albumin, globulin, albumin/globulin ratio, and oxidants in serum (P < 0.01), further it improved liver antioxidant status (P < 0.01). Butein significantly lowered lipid profile parameters (P < 0.01), suppressed inflammatory cytokines (P < 0.01), and improved liver histology. Further to understand the possible mechanism behind the hepatoprotective and lipid lowering effect of butein, the activities of heme oxygenase (HO1), myeloperoxidase (MPO), and mitochondrial reactive oxygen species (ROS) were measured. We found that butein supplementation significantly decreased the activity of HO1 (P < 0.001), and increased the activity of MPO (P < 0.001). Furthermore butein attenuated mitochondrial ROS produced in NAFLD condition. Present study shows that butein supplementation restore liver function by altering liver oxidative stress, inflammatory markers, vital defensive enzyme activities, and mitochondrial ROS. In summary, butein has remarkable potential to develop effective hepato-protective drug. © 2018 BioFactors, 44(3):289-298, 2018.


Asunto(s)
Chalconas/farmacología , Deficiencia de Colina/tratamiento farmacológico , Dieta/efectos adversos , Hipolipemiantes/farmacología , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Administración Oral , Albúminas/metabolismo , Animales , Colina/metabolismo , Deficiencia de Colina/etiología , Deficiencia de Colina/metabolismo , Deficiencia de Colina/patología , Globulinas/metabolismo , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Metionina/deficiencia , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Peroxidasa/antagonistas & inhibidores , Peroxidasa/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transaminasas/antagonistas & inhibidores , Transaminasas/metabolismo
13.
Life Sci ; 192: 46-54, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29158052

RESUMEN

AIMS: Baicalin (BA), an active flavonoid compound originating from the herb of Scutellaria baicalensis Georgi, has been previously shown to exert anti-inflammation and anti-oxidant effects in liver diseases. However, the potential role of BA in the regulation of non-alcoholic steatohepatitis (NASH) remains elusive. In this study, we newly explored the hepatoprotective effects of BA in MCD diet-induced NASH by ameliorating hepatic steatosis, inflammation, fibrosis and apoptosis. MAIN METHODS: NASH was induced in mice fed a methionine and choline-deficient (MCD) diet for 4weeks. The mice were simultaneously treated with or without BA for 4weeks. Serum liver functional markers and inflammatory indicators were assessed by biochemical and ELISA methods, respectively. The livers were histologically examined using H&E, Oil Red O and Masson's trichrome staining methods. The qRT-PCR, IHC and Western blotting assays were applied to analyze mechanisms underlying BA protection. KEY FINDINGS: BA treatment significantly attenuated MCD diet-induced hepatic lipid accumulation partly through regulating the expression of SREBP-1c, FASN, PPARα and CPT1a. BA treatment dramatically suppressed MCD diet-induced hepatic inflammation, which was associated with decrease in serum TNF-α, IL-1ß and MCP-1 production, macrophage influx and suppression of nuclear factor-κB activation. Additionally, BA was proved to prevent liver fibrosis, which appears to be mediated by inhibition of α-SMA, TGF-ß1 and Col1A1. Furthermore, BA markedly inhibited hepatocyte apoptosis and cleaved caspase-3 protein expression in MCD diet-induced mice. SIGNIFICANCE: These results provide a possible basis of the underlying mechanism for the application of BA in the treatment of NASH.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Flavonoides/uso terapéutico , Metabolismo de los Lípidos/efectos de los fármacos , Cirrosis Hepática/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Animales , Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/genética , Deficiencia de Colina/tratamiento farmacológico , Deficiencia de Colina/patología , Dieta , Flavonoides/farmacología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/patología , Pruebas de Función Hepática , Masculino , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
14.
Biochem Biophys Res Commun ; 364(4): 801-7, 2007 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-17963695

RESUMEN

Rennin-angiotensin system is involved in liver fibrogenesis through activating hepatic stellate cells (HSCs). Telmisartan (Tel) is an angiotensin II type 1 receptor antagonist, could function as a selective peroxisome proliferator-activated receptor gamma activator. Here we studied the effect of Tel on liver fibrosis, pre-neoplastic lesions in vivo and primary HSCs in vitro. In vivo study, we used the choline-deficient L-amino acid-defined (CDAA)-diet induced rat NASH model. The rats were fed the CDAA diet for 8 weeks to induce liver fibrosis and pre-neoplastic lesions, and then co-administrated with Tel for another 10 weeks. Tel prevented liver fibrogenesis and pre-neoplastic lesions by down-regulating TGFbeta1 and TIMP-1, 2 and increasing MMP-13 expression. Tel inhibited HSCs activation and proliferation. These results suggested that Tel could be a promising drug for NASH related liver fibrosis.


Asunto(s)
Bencimidazoles/administración & dosificación , Benzoatos/administración & dosificación , Deficiencia de Colina/tratamiento farmacológico , L-Aminoácido Oxidasa/deficiencia , Cirrosis Hepática/prevención & control , Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Animales , Deficiencia de Colina/complicaciones , Deficiencia de Colina/diagnóstico , Relación Dosis-Respuesta a Droga , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/etiología , Masculino , Ratas , Ratas Wistar , Telmisartán , Resultado del Tratamiento
15.
J Agric Food Chem ; 64(27): 5598-606, 2016 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-27321734

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) refers to hepatic pathologies, including simple fatty liver (SFL), nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis, that may progress to hepatocellular carcinoma. These liver disease states may affect the activity and expression levels of drug-metabolizing enzymes, potentially resulting in an alteration in the pharmacokinetics, therapeutic efficacy, and safety of drugs. This study investigated the hepatic cytochrome P450 (CYP) 2B1-modulating effect of a specific NAFLD state in dietary rat models. Sprague-Dawley rats were given a methionine/choline-deficient (MCD) or high-fat (HF) diet to induce NASH and SFL, respectively. The induction of these disease states was confirmed by plasma chemistry and liver histological analysis. Both the protein and mRNA levels of hepatic CYP2B1 were considerably reduced in MCD diet-fed rats; however, they were similar between the HF diet-fed and control rats. Consistently, the enzyme-kinetic and pharmacokinetic parameters for CYP2B1-mediated bupropion metabolism were considerably reduced in MCD diet-fed rats; however, they were also similar between the HF diet-fed and control rats. These results may promote a better understanding of the influence of NAFLD on CYP2B1-mediated metabolism, which could have important implications for the safety and pharmacokinetics of drug substrates for the CYP2B subfamily in patients with NAFLD.


Asunto(s)
Bupropión/administración & dosificación , Deficiencia de Colina/tratamiento farmacológico , Metionina/deficiencia , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Animales , Colina/metabolismo , Deficiencia de Colina/enzimología , Deficiencia de Colina/genética , Deficiencia de Colina/metabolismo , Citocromo P-450 CYP2B1/genética , Citocromo P-450 CYP2B1/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Fundam Clin Pharmacol ; 30(1): 47-57, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26501493

RESUMEN

Choline has been identified as an essential nutrient with crucial role in many vital biological functions. Recent studies have demonstrated that heart dysfunction can develop in the setting of choline deprivation even in the absence of underlying heart disease. Matrix metalloproteinases (MMPs) are responsible for extracellular matrix degradation, and the dysregulation of MMP-2 and MMP-9 has been involved in the pathogenesis of various cardiovascular disorders. The aim of the study was to investigate the role of MMPs and their inhibitors (TIMPs), in the pathogenesis of choline deficiency-induced cardiomyopathy, and the way they are affected by carnitine supplementation. Male Wistar Albino adult rats were divided into four groups and received standard or choline-deficient diet with or without L-carnitine in drinking water (0.15% w/v) for 1 month. Heart tissue immunohistochemistry for MMP-2, MMP-9, TIMP-1, and TIMP-2 was performed. Choline deficiency was associated with suppressed immunohistochemical expression of MMP-2 and an increased expression of TIMP-2 compared to control, while it had no impact on TIMP-1. MMP-9 expression was decreased without, however, reaching statistical significance. Carnitine did not affect MMP-2, MMP-9, TIMP-1 or TIMP-2 expression. The pattern of TIMP and MMP modulation observed in a choline deficiency setting appears to promote fibrosis. Carnitine, although shown to suppress fibrosis, does not seem to affect MMP-2, MMP-9, TIMP-1 or TIMP-2 expression. Further studies will be required to identify the mechanism underlying the beneficial effects of carnitine.


Asunto(s)
Cardiomiopatías/prevención & control , Carnitina/uso terapéutico , Deficiencia de Colina/tratamiento farmacológico , Matriz Extracelular/metabolismo , Miocardio/metabolismo , Administración Oral , Animales , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Carnitina/administración & dosificación , Deficiencia de Colina/complicaciones , Deficiencia de Colina/metabolismo , Deficiencia de Colina/patología , Modelos Animales de Enfermedad , Matriz Extracelular/patología , Fibrosis , Inmunohistoquímica , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Miocardio/patología , Ratas Wistar , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Inhibidor Tisular de Metaloproteinasa-2/biosíntesis
17.
J Agric Food Chem ; 63(2): 552-61, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25536170

RESUMEN

The berries of bilberry and black currant are a rich source of anthocyanins, which are thought to have favorable effects on nonalcoholic steatohepatitis (NASH). This study was designed to examine whether purified anthocyanins from bilberry and black currant are able to limit the disorders related to NASH induced by a methionine-choline-deficient (MCD) diet in mice. The results showed that treatment with anthocyanins not only alleviated inflammation, oxidative stress, steatosis, and even fibrosis but also improved depletion of mitochondrial content and damage of mitochondrial biogenesis and electron transfer chain developed concomitantly in the liver of mice fed the MCD diet. Furthermore, anthocyanins treatment promoted activation of AMP-activated protein kinase (AMPK) and expression of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α). These data provide evidence that anthocyanins possess significant protective effects against NASH and mitochondrial defects in response to a MCD diet, with a mechanism maybe through affecting the AMPK/PGC-1α signaling pathways.


Asunto(s)
Antocianinas/administración & dosificación , Deficiencia de Colina/tratamiento farmacológico , Hígado Graso/tratamiento farmacológico , Hígado/efectos de los fármacos , Metionina/deficiencia , Mitocondrias/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Ribes/química , Vaccinium myrtillus/química , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Antocianinas/aislamiento & purificación , Deficiencia de Colina/enzimología , Deficiencia de Colina/genética , Deficiencia de Colina/metabolismo , Hígado Graso/enzimología , Hígado Graso/genética , Hígado Graso/metabolismo , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Extractos Vegetales/aislamiento & purificación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
J Interferon Cytokine Res ; 35(6): 464-73, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25715168

RESUMEN

The results of clinical and experimental studies suggest that type I interferons (IFNs) may have direct antifibrotic activity in addition to their antiviral properties. However, the mechanisms are still unclear; in particular, little is known about the antifibrotic activity of IFN-ß and how its activity is distinct from that of IFN-α. Using DNA microarrays, we demonstrated that gene expression in TWNT-4 cells, an activated human hepatic stellate cell line, was remarkably altered by IFN-ß more than by IFN-α. Integrated pathway enrichment analyses revealed that a variety of IFN-ß-mediated signaling pathways are uniquely regulated in TWNT-4 cells, including those related to cell cycle and Toll-like receptor 4 (TLR4) signaling. To investigate the antifibrotic activity of IFN-ß and the involvement of TLR4 signaling in vivo, we used mice fed a choline-deficient l-amino acid-defined diet as a model of nonalcoholic steatohepatitis-related hepatic fibrosis. In this model, the administration of IFN-ß significantly attenuated augmentation of the area of liver fibrosis, with accompanying transcriptional downregulation of the TLR4 adaptor molecule MyD88. Our results provide important clues for understanding the mechanisms of the preferential antifibrotic activity of IFN-ß and suggest that IFN-ß itself, as well as being a modulator of its unique signaling pathway, may be a potential treatment for patients with hepatic fibrosis in a pathogenesis-independent manner.


Asunto(s)
Deficiencia de Colina/tratamiento farmacológico , Interferón beta/farmacología , Cirrosis Hepática/tratamiento farmacológico , Hígado/efectos de los fármacos , Factor 88 de Diferenciación Mieloide/genética , Receptor Toll-Like 4/genética , Animales , Ciclo Celular/efectos de los fármacos , Colina/metabolismo , Deficiencia de Colina/metabolismo , Deficiencia de Colina/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Alimentos Formulados , Regulación de la Expresión Génica , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Humanos , Interferón-alfa/metabolismo , Interferón-alfa/farmacología , Interferón beta/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal , Receptor Toll-Like 4/metabolismo
19.
Eur J Pharmacol ; 754: 19-24, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25701721

RESUMEN

Ipragliflozin is a selective sodium glucose cotransporter 2 (SGLT2) inhibitor that increases urinary glucose excretion by inhibiting renal glucose reabsorption and thereby causes a subsequent antihyperglycemic effect. As nonalcoholic fatty liver disease (NAFLD), including nonalcoholic steatohepatitis (NASH), is closely linked to metabolic diseases such as obesity and diabetes, we investigated the effect of ipragliflozin on NAFLD in rats fed a choline-deficient l-amino acid-defined (CDAA) diet. Five weeks after starting the CDAA diet, rats exhibited hepatic triglyceride (TG) accumulation, fibrosis, and mild inflammation. Repeated oral administration of ipragliflozin (3mg/g, once daily for 5 weeks) prevented both hepatic TG accumulation (188 vs.290 mg/g tissue vehicle-treated group; P<0.001) and large lipid droplet formation. Further, ipragliflozin exerted a prophylactic effect on liver fibrosis, as indicated by a marked decrease in hydroxyproline content and fibrosis score. Pioglitazone, which is known to be effective on hepatic fibrosis in CDAA diet-fed rats as well as NASH patients with type 2 diabetes mellitus (T2DM), also exerted a mild prophylactic effect on fibrosis, but not on hepatic TG accumulation or inflammation. In conclusion, ipragliflozin prevented hepatic TG accumulation and fibrosis in CDAA-diet rats. These findings suggest the therapeutic potential of ipragliflozin for patients with NAFLD.


Asunto(s)
Deficiencia de Colina/tratamiento farmacológico , Alimentos Formulados/efectos adversos , Glucósidos/farmacología , Cirrosis Hepática/prevención & control , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Tiofenos/farmacología , Aminoácidos/farmacología , Animales , Deficiencia de Colina/complicaciones , Deficiencia de Colina/patología , Glucósidos/uso terapéutico , Hidroxiprolina/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Inflamación/inducido químicamente , Inflamación/complicaciones , Inflamación/prevención & control , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/complicaciones , Cirrosis Hepática/patología , Masculino , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Pioglitazona , Ratas , Tiazolidinedionas/uso terapéutico , Tiofenos/uso terapéutico , Triglicéridos/metabolismo
20.
J Ethnopharmacol ; 67(3): 297-305, 1999 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-10617064

RESUMEN

Shao-Yin-Ren Shi-Quang-Da-Bu-Tang (SDT) has been used traditionally to improve the systemic blood circulation and biological energy production in the body. The object of this study is to determine the effect of SDT extract on the decline of cerebral adenosine triphosphate (ATP) and choline content associated with learning and memory impairments in senescence-accelerated mice prone 8 (SAM P8). Twenty-four-week old mice were orally treated with SDT at 400 mg/kg body weight per day, and continued for 12 consecutive weeks. At the termination of the treatment, the body weight of SAM P8 was markedly lower than that of the equal aged senescence-resistant prone 1 (SAM R1), but this was conspicuously recovered to the level of SAM R1 by SDT treatment. SDT also significantly reduced the decline of cerebral weight (P < 0.05). By comparison with normal mice, a spontaneous decrease of cerebral ATP was observed in the SAM P8. Two- and 6-fold increases of cerebral ATP content were found in SAM R1 and SAM P8 by SDT administration, respectively. The cerebral choline content was significantly different between SAM R1 and SAM P8 aged 36-week old (P < 0.01). SDT remarkably restored the decrease of cerebral choline content in SAM P8 (P < 0.01). Taken together, these results demonstrate that SDT can reduce the decrease of cerebral weight, and restore the decline of cerebral ATP and choline content associated with an alteration of neuronal metabolism in SAM P8 brain. This suggests that pharmacological properties of SDT may participate in improvement of declined cerebral energy production and cholinergic neurotransmitter synthesis in senile dementia.


Asunto(s)
Adenosina Trifosfato/deficiencia , Reacción de Prevención/efectos de los fármacos , Encéfalo/efectos de los fármacos , Deficiencia de Colina/tratamiento farmacológico , Fitoterapia , Animales , Peso Corporal/efectos de los fármacos , Encéfalo/metabolismo , Colina/metabolismo , Glucosa/metabolismo , Masculino , Ratones , Tamaño de los Órganos/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA