Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurosci Lett ; 762: 136136, 2021 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-34311050

RESUMEN

Sporadic cerebral amyloid angiopathy (CAA), which is characterized by cerebrovascular amyloid ß (Aß) deposits, causes cerebral hemorrhages and dementia in elderly people. Metformin has been used to treat patients with type 2 diabetes mellitus (T2DM), and animal and clinical studies have reported therapeutic effects of metformin in Alzheimer's disease (AD). However, the therapeutic effects of metformin in CAA are unclear. Here, we used a mixed mouse model of CAA and T2DM (APP23-ob/ob) to investigate whether metformin has therapeutic effects on cerebrovascular Aß deposits. We dissolved metformin hydrochloride in water and administered it orally at 350 mg/kg/day. Treatments started when mice were 6 weeks old and continued until they were 15 months old. After we treated APP23-ob/ob mice with metformin, we counted the numbers of vessels with Aß and measured levels of Aß40 and Aß42 (soluble and insoluble), amyloid precursor protein (APP), APP-processing enzymes (α-, ß-, and γ-secretases), and Aß-degrading enzymes (insulin-degrading enzyme [IDE], neprilysin). Metformin significantly reduced cerebrovascular Aß deposits in APP23-ob/ob mice (p < .05). Compared with controls, metformin-treated APP23-ob/ob mice had significantly reduced Aß levels in the cerebral cortex (p < .05) and hippocampus (p < .05) and increased levels of IDE in the hippocampus (p < .01). Our results indicate that metformin attenuates the severity of CAA by enhancing Aß-cleaving IDE expression. The clinical application of metformin may lead to a novel therapeutic strategy in CAA treatment, especially in patients with T2DM.


Asunto(s)
Angiopatía Amiloide Cerebral/patología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/complicaciones , Hipoglucemiantes/farmacología , Insulisina/efectos de los fármacos , Metformina/farmacología , Péptidos beta-Amiloides/metabolismo , Animales , Angiopatía Amiloide Cerebral/complicaciones , Circulación Cerebrovascular/efectos de los fármacos , Ratones
2.
Mol Cells ; 43(4): 373-383, 2020 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-32191993

RESUMEN

Our previous study revealed a novel role of Fas-associated death domain-containing protein (FADD) in islet development and insulin secretion. Insulin-degrading enzyme (IDE) is a zinc metalloprotease that selectively degrades biologically important substrates associated with type 2 diabetes (T2DM). The current study was designed to investigate the effect of FADD phosphorylation on IDE. We found that the mRNA and protein levels of IDE were significantly downregulated in FADD-D mouse livers compared with control mice. Quantitative real-time polymerase chain reaction analysis showed that FADD regulates the expression of IDE at the transcriptional level without affecting the stability of the mRNA in HepG2 cells. Following treatment with cycloheximide, the IDE protein degradation rate was found to be increased in both FADD-D primary hepatocytes and FADD-knockdown HepG2 cells. Additionally, IDE expression levels were reduced in insulin-stimulated primary hepatocytes from FADD-D mice compared to those from control mice. Moreover, FADD phosphorylation promotes nuclear translocation of FoxO1, thus inhibiting the transcriptional activity of the IDE promoter. Together, these findings imply a novel role of FADD in the reduction of protein stability and expression levels of IDE.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Insulisina/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Fosforilación , Transfección
3.
J Food Biochem ; 43(7): e12855, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31353702

RESUMEN

This study was performed to investigate the effects of Artemisia argyi and 4,5-dicaffeyolquinic acid (4,5-diCQA) as a main compound of ethyl acetate fraction from Artemisia argyi (EFAA) on high-fat diet (HFD)-induced cognitive dysfunction. Both EFAA and 4,5-diCQA were effective in improving cognitive function on HFD-induced cognitive dysfunction. In brain tissue analysis, it was confirmed that EFAA and 4,5-diCQA inhibited the reduction of neurotransmitters as well as oxidative stress and mitochondrial dysfunction. In addition, they inhibited amyloid ß (Aß) accumulation by increasing the expression of insulin-degrading enzyme and consequently prevented apoptosis. In conclusion, it is presumed that Artemisia argyi may help to improve the cognitive impairment due to the HFD, and it is considered that this effect is closely related to the physiological activity of 4,5-diCQA. PRACTICAL APPLICATIONS: Artemisia argyi is used in traditional herbal medicine in Asia. Type 2 diabetes mellitus has been proven by a variety of epidemiological studies to be a risk factor for cognitive impairment, such as Alzheimer's disease. This study confirmed that 4,5-diCQA is a bioactive compound of Artemisia argyi on improving HFD-induced cognitive dysfunction. Therefore, this study can provide useful information to the effect of Artemisia argyi and related substance.


Asunto(s)
Artemisia , Disfunción Cognitiva/tratamiento farmacológico , Insulisina/efectos de los fármacos , Extractos Vegetales/farmacología , Ácido Quínico/análogos & derivados , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Apoptosis/efectos de los fármacos , Artemisia/química , Artemisia/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Dieta Alta en Grasa/efectos adversos , Insulisina/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fitoterapia , Plantas Medicinales/metabolismo , Ácido Quínico/farmacología
4.
ACS Chem Neurosci ; 9(12): 3137-3152, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30052418

RESUMEN

The single greatest risk factor for neurodegenerative diseases is aging. Aging of cells such as microglia in the nervous system has an impact not only on the ability of those cells to function but also on cells they interact with. We have developed a model microglia system that recapitulates the dystrophic/senescent phenotype, and we have combined this with the study of ß-amyloid processing. The model is based on the observation that aged microglia have increased iron content. By overloading a human microglial cell line with iron, we were able to change the secretory profile of the microglia. When combining these senescent microglia with SH-SY5Y cells, we noted an increase in extracellular ß-amyloid. The increased levels of ß-amyloid were due to a decrease in the release of insulin-degrading enzyme by the model senescent microglia. Further analysis revealed that the senescent microglia showed both decreased autophagy and increased ER stress. These studies demonstrate the potential impact of an aging microglial population in terms of ß-amyloid produced by neurons, which could play a causal role in diseases like Alzheimer's disease. Our results also further develop the potential utility of an in vitro model of senescent microglia for the study of brain aging and neurodegenerative disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Senescencia Celular/fisiología , Insulisina/metabolismo , Microglía/metabolismo , Envejecimiento , Enfermedad de Alzheimer , Péptidos beta-Amiloides/efectos de los fármacos , Autofagia , Línea Celular Tumoral , Senescencia Celular/efectos de los fármacos , Citocinas/metabolismo , Estrés del Retículo Endoplásmico , Humanos , Insulisina/efectos de los fármacos , Hierro/metabolismo , Hierro/farmacología , Microglía/efectos de los fármacos , Neuronas/metabolismo
5.
Sci Rep ; 7(1): 14876, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29093479

RESUMEN

Disruption of insulin secretion and clearance both contribute to obesity-induced hyperinsulinemia, though reduced insulin clearance seems to be the main factor. The liver is the major site for insulin degradation, a process mainly coordinated by the insulin-degrading enzyme (IDE). The beneficial effects of taurine conjugated bile acid (TUDCA) on insulin secretion as well as insulin sensitivity have been recently described. However, the possible role of TUDCA in insulin clearance had not yet been explored. Here, we demonstrated that 15 days treatment with TUDCA reestablished plasma insulin to physiological concentrations in high fat diet (HFD) mice, a phenomenon associated with increased insulin clearance and liver IDE expression. TUDCA also increased IDE expression in human hepatic cell line HepG2. This effect was not observed in the presence of an inhibitor of the hepatic membrane bile acid receptor, S1PR2, nor when its downstream proteins were inhibited, including IR, PI3K and Akt. These results indicate that treatment with TUDCA may be helpful to counteract obesity-induced hyperinsulinemia through increasing insulin clearance, likely through enhanced liver IDE expression in a mechanism dependent on S1PR2-Insulin pathway activation.


Asunto(s)
Insulina/farmacocinética , Insulisina/efectos de los fármacos , Hígado/enzimología , Ácido Tauroquenodesoxicólico/farmacología , Animales , Dieta Alta en Grasa , Células Hep G2 , Humanos , Hiperinsulinismo/tratamiento farmacológico , Insulisina/metabolismo , Hígado/metabolismo , Ratones , Ratones Obesos
6.
Am J Physiol Endocrinol Metab ; 309(8): E747-58, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26306598

RESUMEN

The improvement of hepatic insulin sensitivity by the cannabinoid receptor 1 (CB1R) antagonist rimonabant (RIM) has been recently been reported to be due to upregulation of adiponectin. Several studies demonstrated that improvement in insulin clearance accompanies the enhancement of hepatic insulin sensitivity. However, the effects of RIM on hepatic insulin clearance (HIC) have not been fully explored. The aim of this study was to explore the molecular mechanism(s) by which RIM affects HIC, specifically to determine whether upregulation of liver adiponectin receptors (ADRs) and other key genes regulated by adiponectin mediate the effects. To induce insulin resistance in skeletal muscle and liver, dogs were fed a hypercaloric high-fat diet (HFD) for 6 wk. Thereafter, while still maintained on a HFD, animals received RIM (HFD+RIM; n = 11) or placebo (HFD+PL; n = 9) for an additional 16 wk. HIC, calculated as the metabolic clearance rate (MCR), was estimated from the euglycemic-hyperinsulinemic clamp. The HFD+PL group showed a decrease in MCR; in contrast, the HFD+RIM group increased MCR. Consistently, the expression of genes involved in HIC, CEACAM-1 and IDE, as well as gene expression of liver ADRs, were increased in the HFD+RIM group, but not in the HFD+PL group. We also found a positive correlation between CEACAM-1 and the insulin-degrading enzyme IDE with ADRs. Interestingly, expression of liver genes regulated by adiponectin and involved in lipid oxidation were increased in the HFD+RIM group. We conclude that in fat-fed dogs RIM enhances HIC, which appears to be linked to an upregulation of the adiponectin pathway.


Asunto(s)
Antagonistas de Receptores de Cannabinoides/farmacología , Dieta Alta en Grasa , Insulina/metabolismo , Hígado/efectos de los fármacos , Piperidinas/farmacología , Pirazoles/farmacología , ARN Mensajero/efectos de los fármacos , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptores de Adiponectina/efectos de los fármacos , Animales , Antígenos CD/efectos de los fármacos , Antígenos CD/metabolismo , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Perros , Técnica de Clampeo de la Glucosa , Resistencia a la Insulina , Insulisina/efectos de los fármacos , Insulisina/metabolismo , Hígado/metabolismo , Masculino , Tasa de Depuración Metabólica , ARN Mensajero/metabolismo , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo , Rimonabant , Regulación hacia Arriba/efectos de los fármacos
7.
Endocrinology ; 156(12): 4592-603, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26305888

RESUMEN

Chronic exposure to elevated levels of glucocorticoids has been linked to age-related cognitive decline and may play a role in Alzheimer's disease. In the brain, 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) amplifies intracellular glucocorticoid levels. We show that short-term treatment of aged, cognitively impaired C57BL/6 mice with the potent and selective 11ß-HSD1 inhibitor UE2316 improves memory, including after intracerebroventricular drug administration to the central nervous system alone. In the Tg2576 mouse model of Alzheimer's disease, UE2316 treatment of mice aged 14 months for 4 weeks also decreased the number of ß-amyloid (Aß) plaques in the cerebral cortex, associated with a selective increase in local insulin-degrading enzyme (involved in Aß breakdown and known to be glucocorticoid regulated). Chronic treatment of young Tg2576 mice with UE2316 for up to 13 months prevented cognitive decline but did not prevent Aß plaque formation. We conclude that reducing glucocorticoid regeneration in the brain improves cognition independently of reduced Aß plaque pathology and that 11ß-HSD1 inhibitors have potential as cognitive enhancers in age-associated memory impairment and Alzheimer's dementia.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Cognición/efectos de los fármacos , Memoria/efectos de los fármacos , Placa Amiloide/patología , Pirazoles/farmacología , Tiofenos/farmacología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal/efectos de los fármacos , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Humanos , Insulisina/efectos de los fármacos , Insulisina/metabolismo , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
8.
Int J Exp Pathol ; 92(4): 272-80, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21507087

RESUMEN

Insulin-degrading enzyme (IDE) has been shown to enhance the binding of androgen and glucocorticoid receptors to DNA in the nuclear compartment. Glucocorticoids cause hyperglycaemia, peripheral resistance to insulin and compensatory hyperinsulinaemia. The aim of the present study was to investigate the effect of dexamethasone (D), testosterone (T) and dexamethasone plus testosterone (D + T) on the regulation of IDE and on the remodelling of rat ventral prostate after castration (C). Castration led to a marked reduction in prostate weight (PW). Body weight was significantly decreased in the castrated animals treated with dexamethasone, and the relative PW was 2.6-fold (±0.2) higher in the D group, 2.8-fold (±0.3) higher in the T group and 6.6-fold (±0.6) higher in the D + T group in comparison with the castrated rats. Ultrastructural alterations in the ventral prostate in response to androgen deprivation were restored after testosterone and dexamethasone plus testosterone treatments and partially restored with dexamethasone alone. The nuclear IDE protein level indicated a 4.3-fold (±0.4) increase in castrated rats treated with D + T when compared with castration alone. Whole-cell IDE protein levels increased approximately 1.5-fold (±0.1), 1.5-fold (±0.1) and 2.9-fold (±0.2) in the D, T and D + T groups, respectively, when compared with castration alone. In conclusion, the present study reports that dexamethasone-induced hyperinsulinaemic condition plus exogenous testosterone treatment leads to synergistic effects of insulin and testosterone in the prostatic growth and in the amount of IDE in the nucleus and whole epithelial cell.


Asunto(s)
Castración , Dexametasona/farmacología , Insulisina/metabolismo , Próstata/metabolismo , Próstata/patología , Testosterona/farmacología , Andrógenos/farmacología , Animales , Peso Corporal/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/patología , Dexametasona/efectos adversos , Glucocorticoides/efectos adversos , Glucocorticoides/farmacología , Hiperinsulinismo/inducido químicamente , Hiperinsulinismo/metabolismo , Insulisina/efectos de los fármacos , Masculino , Modelos Animales , Próstata/efectos de los fármacos , Ratas , Ratas Wistar
9.
Chemistry ; 17(9): 2752-62, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21274957

RESUMEN

Accumulation of neurotoxic amyloid-ß peptide (Aß) and alteration of metal homeostasis (metallostasis) in the brain are two main factors that have been very often associated with neurodegenerative diseases, such as Alzheimer's disease (AD). Aß is constantly produced from the amyloidprecursor-protein APP precursor and immediately catabolized under normal conditions, whereas dysmetabolism of Aß and/or metal ions seems to lead to a pathological deposition. Although insulin-degrading enzyme (IDE) is the main metalloprotease involved in Aß degradation in the brain being up-regulated in some areas of AD brains, the role of IDE for the onset and development of AD is far from being understood. Moreover, the biomolecular mechanisms involved in the recognition and interaction between IDE and its substrates are still obscure. In spite of the important role of metals (such as copper, aluminum, and zinc), which has brought us to propose a "metal hypothesis of AD", a targeted study of the effect of metallostasis on IDE activity has never been carried out. In this work, we have investigated the role that various metal ions (i.e., Cu(2+), Cu(+), Zn(2+), Ag(+), and Al(3+)) play in modulating the interaction between IDE and two Aß peptide fragments, namely Aß(1-16) and Aß(16-28). It was therefore possible to identify the direct effect that such metal ions have on IDE structure and enzymatic activity without interferences caused by metal-induced substrate modifications. Mass spectrometry and kinetic studies revealed that, among all the metal ions tested, only Cu(2+), Cu(+), and Ag(+) have an inhibitory effect on IDE activity. Moreover, the inhibition of copper(II) is reversed by adding zinc(II), whereas the monovalent cations affect the enzyme activity irreversibly. The molecular basis of their action on the enzyme is also discussed on the basis of computational investigations.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/efectos de los fármacos , Cobre/química , Cobre/metabolismo , Insulisina/metabolismo , Algoritmos , Aluminio/química , Aluminio/metabolismo , Simulación por Computador , Cisteína/química , Cisteína/genética , Humanos , Insulisina/efectos de los fármacos , Homología de Secuencia de Aminoácido , Plata/química , Plata/metabolismo , Zinc/química , Zinc/metabolismo
10.
J Neurosci Res ; 83(7): 1252-61, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16511867

RESUMEN

Cerebrolysin is a peptide mixture with neurotrophic effects that might reduce the neurodegenerative pathology in Alzheimer's disease (AD). We have previously shown in an amyloid protein precursor (APP) transgenic (tg) mouse model of AD-like neuropathology that Cerebrolysin ameliorates behavioral deficits, is neuroprotective, and decreases amyloid burden; however, the mechanisms involved are not completely clear. Cerebrolysin might reduce amyloid deposition by regulating amyloid-beta (Abeta) degradation or by modulating APP expression, maturation, or processing. To investigate these possibilities, APP tg mice were treated for 6 months with Cerebrolysin and analyzed in the water maze, followed by RNA, immunoblot, and confocal microscopy analysis of full-length (FL) APP and its fragments, beta-secretase (BACE1), and Abeta-degrading enzymes [neprilysin (Nep) and insulin-degrading enzyme (IDE)]. Consistent with previous studies, Cerebrolysin ameliorated the performance deficits in the spatial learning portion of the water maze and reduced the synaptic pathology and amyloid burden in the brains of APP tg mice. These effects were associated with reduced levels of FL APP and APP C-terminal fragments, but levels of BACE1, Notch1, Nep, and IDE were unchanged. In contrast, levels of active cyclin-dependent kinase-5 (CDK5) and glycogen synthase kinase-3beta [GSK-3beta; but not stress-activated protein kinase-1 (SAPK1)], kinases that phosphorylate APP, were reduced. Furthermore, Cerebrolysin reduced the levels of phosphorylated APP and the accumulation of APP in the neuritic processes. Taken together, these results suggest that Cerebrolysin might reduce AD-like pathology in the APP tg mice by regulating APP maturation and transport to sites where Abeta protein is generated. This study clarifies the mechanisms through which Cerebrolysin might reduce Abeta production and deposition in AD and further supports the importance of this compound in the potential treatment of early AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Aminoácidos/farmacología , Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/efectos de los fármacos , Encéfalo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Aminoácidos/uso terapéutico , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas , Encéfalo/metabolismo , Encéfalo/patología , Quinasa 5 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Endopeptidasas/efectos de los fármacos , Endopeptidasas/genética , Endopeptidasas/metabolismo , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Insulisina/efectos de los fármacos , Insulisina/genética , Insulisina/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Neprilisina/efectos de los fármacos , Neprilisina/genética , Neprilisina/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo
11.
J Neuropathol Exp Neurol ; 64(2): 139-46, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15751228

RESUMEN

Insulin-degrading enzyme (IDE) has been identified as a candidate protease in the clearance of amyloid-delta (Abeta) peptides from the brain. IDE activity and binding to insulin are known to be inhibited by glucocorticoids in vitro. In Alzheimer disease (AD), both a decrease in IDE levels and an increase in peripheral glucocorticoid levels have been documented. Our study investigated the effects of glucocorticoid treatment on IDE expression in vivo in 12 nonhuman primates (Macaca nemestrina). Year-long, high-dose exposure to the glucocorticoid cortisol (hydrocortisone acetate) was associated with reduced IDE protein levels in the inferior frontal cortex and reduced IDE mRNA levels in the dentate gyrus of the hippocampus. We assessed Abeta40 and Abeta42 levels by ELISA in the brain and in plasma, total plaque burden by immunohistochemistry, and relative Abeta1-40 and Abeta1-42 levels in the brain by mass spectrometry. Glucocorticoid treatment increased Abeta42 relative to Abeta40 levels without a change in overall plaque burden within the brain, while Abeta42 levels were decreased in plasma. These findings support the notion that glucocorticoids regulate IDE and provide a mechanism whereby increased glucocorticoid levels may contribute to AD pathology.


Asunto(s)
Envejecimiento , Péptidos beta-Amiloides/efectos de los fármacos , Encéfalo/efectos de los fármacos , Glucocorticoides/farmacología , Insulisina/efectos de los fármacos , Fragmentos de Péptidos/efectos de los fármacos , Péptidos beta-Amiloides/sangre , Animales , Western Blotting , Encéfalo/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunohistoquímica , Hibridación in Situ , Macaca , Masculino , Fragmentos de Péptidos/sangre , ARN Mensajero/análisis , ARN Mensajero/efectos de los fármacos
12.
Hepatobiliary Pancreat Dis Int ; 3(1): 99-101, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14969848

RESUMEN

BACKGROUND: Cordeceps sinensis (CS) is a herb which can inhibit the liver fibrosis. Hyperinsulinemia is common in liver cirrhosis patients. The activity of insulin degrading enzyme could reflect the metabolism of insulin. This study was to detect the dynamical effects and mechanisms of CS on the activity of hepatic insulinase in CCl4 induced liver cirrhosis in rats. METHODS: Rats were randomly allocated into three groups: normal group, model group and CS group. The rats in the normal group were sacrificed at the beginning of experiment, and the other two groups were sacrificed randomly at the end of the third, sixth and ninth weeks. Blood and tissue specimens were taken. Biochemical assays were used to determine the changes of alanine transaminase (ALT), albumin levels in serum. And radioimmunological assays were used to determine the changes of hyaluronic acid (HA), insulin levels in serum and the activity of hepatic insulinase. RESULTS: No significant differences were seen in the serum levels of ALT, albumin, HA between the CS group and the model group at the third and sixth weeks (P>0.05). The serum levels of ALT, HA in the CS group were lower than those in the model group at the ninth week (P<0.05), but the serum level of albumin in the CS group was higher than that in the model group at the ninth week (P<0.05). No significant differences were observed in the serum levels of insulin and the activity of hepatic insulinase between the CS and model groups at the third week and the normal group (P>0.05). The serum levels of insulin in the CS and model groups at the sixth and ninth weeks were higher than those in the normal group (P<0.05). But the activity of hepatic insulinase was lower than that in the normal group (P<0.05 or P<0.01). No significant differences were found in the serum levels of insulin and the activity of hepatic insulinase between the CS and model groups at the third, sixth and ninth weeks (P>0.05). CONCLUSIONS: CS may decrease the damage to hepatocyte by CCl4, and inhibit hepatic fibrogenesis. Six weeks after CCl4 administration, the activity of hepatic insulinase began decreasing. CS could not inhibit the decrease of the activity of hepatic insulinase.


Asunto(s)
Cordyceps , Medicamentos Herbarios Chinos/farmacología , Insulisina/efectos de los fármacos , Insulisina/metabolismo , Cirrosis Hepática Experimental/tratamiento farmacológico , Alanina Transaminasa/análisis , Albúminas/análisis , Animales , Modelos Animales de Enfermedad , Femenino , Ácido Hialurónico/análisis , Cirrosis Hepática Experimental/enzimología , Masculino , Probabilidad , Radioinmunoensayo , Distribución Aleatoria , Ratas , Ratas Wistar , Valores de Referencia , Sensibilidad y Especificidad
13.
Cell Growth Differ ; 7(6): 787-96, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8780892

RESUMEN

Physiologically, the action of insulin-like growth factors (IGFs) is controlled at different levels, from its transcription start by tissue-specific and development-specific transcriptional factors to its degradation by peptidases such as insulin-degrading enzyme (IDE). Since IGF-II is the major autocrine/paracrine growth factor for neuroblastoma cells, we studied the expression and the role of IDE in this system. Here, we show that (a) IDE is expressed in several human neuroectodermal tumor cell lines, including neuroblastoma cell lines; (b) in a neuroblastoma cell line, IDE expression is up-regulated by retinoic acid, a well-known inducer of neuronal differentiation and/or programmed cell death; (c) IDE is probably not the only IGF-degrading enzyme present in these cells, since the activity of a novel thermolysin-like metalloendopeptidase, clearly distinct from IDE, is also detected. The TME activity is inhibited by IGF-I, Des-IGF-I, and IGF-II, and it is down-regulated by retinoic acid. Since retinoic acid plays a relevant role in controlling the growth of these cells and affects the expression of IDE, we have also: (a) identified the retinoic acid receptors (RARs) and retinoid X receptors (RXRs) expressed in these cell lines and (b) by means of synthetic retinoid analogues identified the RAR/RXR isoforms whose activation may be sufficient to induce the expression of the IDE gene. These results provide evidence that complex posttranslational molecular mechanisms participate in the autocrine/paracrine growth control of the IGF-II loop in neuroblastomas involving proteolytic systems.


Asunto(s)
Endopeptidasas/efectos de los fármacos , Insulisina/efectos de los fármacos , Tretinoina/farmacología , Humanos , Estructura Molecular , Neuroblastoma , Tumor Neuroectodérmico Melanótico/enzimología , Tumor Neuroectodérmico Melanótico/metabolismo , Termolisina/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...