Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Lipids Health Dis ; 20(1): 115, 2021 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-34563222

RESUMEN

BACKGROUNDS: Cancer-associated cachexia (CAC) is a metabolic syndrome characterized by progressive depletion of adipose and muscle tissue that cannot be corrected by conventional nutritional therapy. Adipose tissue, an important form of energy storage, exhibits marked loss in the early stages of CAC, which affects quality of life and efficacy of chemotherapy. MicroRNAs (miRNAs) are a class of noncoding RNAs that widely exist in all kinds of eukaryotic cells and play regulatory roles in various biological processes. However, the role of miRNAs in adipose metabolism in CAC has rarely been reported. This study attempted to identify important miRNAs in adipose metabolism in CAC and explore their mechanism to identify a new predictive marker or therapeutic target for CAC-related adipose tissue loss (CAL). METHODS: In this study, miRNA sequencing was firstly used to identify differentially expressed miRNAs related to CAL and the reliability of the conclusions was verified in large population samples. Furthermore, functional experiments were performed by up and down regulating miR-410-3p in adipocytes. The binding of miR-410-3p to Insulin Receptor Substrate 1 (IRS-1) was verified by Luciferase reporter assay and functional experiments of IRS-1 were performed in adipocytes. Finally, the expression of miR-410-3p in serum exosomes was detected. RESULTS: miR-410-3p was selected as differentially expressed miRNA through screening and validation. Adipogenesis was suppressed in miR-410-3p upregulation experiment and increased in downregulation experiment. Luciferase reporter assay showed that miR-410-3p binds to 3' non-coding region of IRS-1 and represses its expression and ultimately inhibits adipogenesis. miR-410-3p was highly expressed in serum exosomes of CAC patients, which was consistent with results in adipose tissue. CONCLUSIONS: The expression of miR-410-3p was higher in subcutaneous adipose tissues and serum exosomes of CAC patients, which significantly inhibits adipogenesis and lipid accumulation. The study shows that miR-410-3p could downregulate IRS-1 and downstream adipose differentiation factors including C/EBP-a and PPAR-γ by targeting 3' noncoding region.


Asunto(s)
Adipocitos/citología , Caquexia/metabolismo , Proteínas Sustrato del Receptor de Insulina/biosíntesis , MicroARNs/genética , Neoplasias/metabolismo , Regiones no Traducidas 3' , Adipogénesis , Tejido Adiposo/patología , Anciano , Caquexia/complicaciones , Diferenciación Celular , Exosomas/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Lípidos/química , Masculino , Persona de Mediana Edad , Neoplasias/complicaciones
2.
Mol Biol Rep ; 47(10): 7557-7566, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32929654

RESUMEN

The main pathogenesis of type 1 diabetes mellitus (T1DM) is autoimmune-mediated apoptosis of pancreatic islet ß cells. We sought to characterize the function of microRNA-203a (miR-203a) on pancreatic islet ß cell proliferation and apoptosis. In situ hybridization was used to detect the expression of miR-203a in islet ß cells in normal and hyperglycaemic non-obese diabetic (NOD) mice. Cell proliferation was measured by cell counting kit eight and cell apoptosis was detected using flow cytometry. Insulin receptor substrate 2 (IRS2/Irs2) was determined to be a direct target of miR-203a by Luciferase reporter assay. We detected the effects of miR-203a overexpression or inhibition on proliferation and apoptosis of IRS2-overexpressing or IRS2-knockdown MIN6 cells respectively, and preliminarily explored the downstream targets of the IRS2 pathway. NOD mice model was used to detect miR-203a inhibitor treatment for diabetes. Our experiment showed miR-203a was upregulated in pancreatic ß cells of hyperglycaemic NOD mice. Elevated miR-203a expression inhibited the proliferation and promoted the apoptosis of MIN6 cells. IRS2/Irs2 is a novel target gene directly regulated by miR-203a and miR-203a overexpression downregulated the expression of IRS2. Irs2 silencing reduced cell proliferation and increased apoptosis. Irs2 overexpression could abolish the pro-apoptotic and anti-proliferative effects of miR-203a on MIN6 cells. Hyperglycemia in newly hyperglycemic NOD mice was under control after treatment with miR-203a inhibitor. Our study suggests that miR-203a regulates pancreatic ß cell proliferation and apoptosis by targeting IRS2, treatment with miR-203a inhibitors and IRS2 might provide a new therapeutic strategy for T1DM.


Asunto(s)
Apoptosis , Proliferación Celular , Hiperglucemia/metabolismo , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Células Secretoras de Insulina/metabolismo , MicroARNs/metabolismo , Animales , Línea Celular , Femenino , Hiperglucemia/patología , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos NOD
3.
Brain ; 140(5): 1420-1436, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334990

RESUMEN

See Stayte and Vissel (doi:10.1093/awx064) for a scientific commentary on this article. Multiple system atrophy is a fatal sporadic adult-onset neurodegenerative disorder with no symptomatic or disease-modifying treatment available. The cytopathological hallmark of multiple system atrophy is the accumulation of α-synuclein aggregates in oligodendrocytes, forming glial cytoplasmic inclusions. Impaired insulin/insulin-like growth factor-1 signalling (IGF-1) and insulin resistance (i.e. decreased insulin/IGF-1) have been reported in other neurodegenerative disorders such as Alzheimer's disease. Increasing evidence also suggests impaired insulin/IGF-1 signalling in multiple system atrophy, as corroborated by increased insulin and IGF-1 plasma concentrations in multiple system atrophy patients and reduced IGF-1 brain levels in a transgenic mouse model of multiple system atrophy. We here tested the hypothesis that multiple system atrophy is associated with brain insulin resistance and showed increased expression of the key downstream messenger insulin receptor substrate-1 phosphorylated at serine residue 312 in neurons and oligodendrocytes in the putamen of patients with multiple system atrophy. Furthermore, the expression of insulin receptor substrate 1 (IRS-1) phosphorylated at serine residue 312 was more apparent in inclusion bearing oligodendrocytes in the putamen. By contrast, it was not different between both groups in the temporal cortex, a less vulnerable structure compared to the putamen. These findings suggest that insulin resistance may occur in multiple system atrophy in regions where the neurodegenerative process is most severe and point to a possible relation between α-synuclein aggregates and insulin resistance. We also observed insulin resistance in the striatum of transgenic multiple system atrophy mice and further demonstrate that the glucagon-like peptide-1 analogue exendin-4, a well-tolerated and Federal Drug Agency-approved antidiabetic drug, has positive effects on insulin resistance and monomeric α-synuclein load in the striatum, as well as survival of nigral dopamine neurons. Additionally, plasma levels of exosomal neural-derived IRS-1 phosphorylated at serine residue 307 (corresponding to serine residue 312 in humans) negatively correlated with survival of nigral dopamine neurons in multiple system atrophy mice treated with exendin-4. This finding suggests the potential for developing this peripheral biomarker candidate as an objective outcome measure of target engagement for clinical trials with glucagon-like peptide-1 analogues in multiple system atrophy. In conclusion, our observation of brain insulin resistance in multiple system atrophy patients and transgenic mice together with the beneficial effects of the glucagon-like peptide-1 agonist exendin-4 in transgenic mice paves the way for translating this innovative treatment into a clinical trial.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Atrofia de Múltiples Sistemas/metabolismo , Péptidos/farmacología , Ponzoñas/farmacología , Anciano , Anciano de 80 o más Años , Animales , Supervivencia Celular/efectos de los fármacos , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/fisiología , Exenatida , Femenino , Humanos , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/sangre , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Atrofia de Múltiples Sistemas/sangre , Neuronas/metabolismo , Oligodendroglía/metabolismo , Fosforilación , Agregación Patológica de Proteínas/metabolismo , Putamen/metabolismo , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Lóbulo Temporal/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
4.
Tumour Biol ; 39(5): 1010428317701640, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28475006

RESUMEN

Accumulating evidence indicated that aberrantly expressed microRNAs play critical roles in the initiation and progression of human cancers. However, the underlying functions of miR-493 in human melanoma remains unknown. Here, our study found that miR-493 expression was downregulated in human melanoma tissues and cells. Overexpression of miR-493 suppressed cell proliferation and cell cycle in human melanoma cell line A375. IRS4 was defined as a target for downregulation by miR-493 and was confirmed by luciferase assay. We also found that knockdown of IRS4 counteracted the proliferation promotion by miR-493 inhibitor. In summary, these results demonstrated that miR-493 acts as a tumor suppressor and inhibits cell proliferation and cell cycle in human melanoma by directly targeting IRS4.


Asunto(s)
Proliferación Celular/genética , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Melanoma/genética , MicroARNs/biosíntesis , Ciclo Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Proteínas Sustrato del Receptor de Insulina/genética , Masculino , Melanoma/patología , MicroARNs/genética , Transducción de Señal/genética
5.
Med Sci Monit ; 22: 2825-30, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27510725

RESUMEN

BACKGROUND The aim of this study was to investigate the effects of different statins on glucose uptake and to confirm its mechanism in primary cultured rat cardiomyocytes after administration of atorvastatin, pravastatin, and rosuvastatin. MATERIAL AND METHODS Primary cultured rat cardiomyocytes were randomly assigned to 5 groups: normal control group (OB), insulin group (S1), statin 1-µM (S2), 5-µM (S3), and 10-µM (S4) groups for 3 different statins. The 2-[3H]-DG uptake of each group was determined and the mRNA and protein expression levels of glucose transporter type 4 (GLUT4), insulin receptor substrate (IRs), and RhoA were assessed. RESULTS After treatment with different concentrations of statins and insulin, the 2-[3H]-DG uptake showed a significant negative correlation with the concentration of atorvastatin (P<0.05), and no significant correlation with pravastatin and rosuvastatin. The mRNA and protein expression levels of GLUT4 and IRs-1 in primary cultured cardiomyocytes were both significantly reduced by atorvastatin treatment (P<0.05). Pravastatin and rosuvastatin showed no significant effects on GLUT4 and IRs-1 expression. The mRNA and protein expression levels of RhoA both showed no significant difference when treated with the 3 statins. CONCLUSIONS These results confirm that atorvastatin can inhibit insulin-induced glucose uptake in primary cultured rat cardiomyocytes by regulating the PI3K/Akt insulin signal transduction pathway.


Asunto(s)
Glucosa/farmacocinética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Adipocitos/efectos de los fármacos , Animales , Desoxiglucosa/farmacocinética , Femenino , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/biosíntesis , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Tritio
6.
Aviakosm Ekolog Med ; 50(1): 28-34, 2016.
Artículo en Ruso | MEDLINE | ID: mdl-27344854

RESUMEN

The signaling processes initiating proteolytic events in m. soleus of humans during short-term exposure in the non-weight bearing conditions were analyzed. Dry immersion (DI) was used to induce weight deprivation over 3 days. Western blotting was used to define the IRS-1 content, total and phosphorylated neuronal NO-synthase (nNOS), AMP-activated protein kinase (AMPK) that control the anabolic and catabolic pathways, and concentrations of cytoskeletal protein desmin and Ca²âº-activated protease calpin. Already on day-3 of DI calpain-dependent proteolysis manifests itself by reductions in both the total content and level of nNOS phosphorilation. Moreover, AMPK phosphorilation was decreased drastically.


Asunto(s)
Proteínas Quinasas Activadas por AMP/biosíntesis , Músculo Esquelético/metabolismo , Óxido Nítrico Sintasa de Tipo I/biosíntesis , Proteolisis , Calpaína/biosíntesis , Desmina/biosíntesis , Humanos , Inmersión , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Metabolismo/genética , Músculo Esquelético/fisiología
7.
Prostate ; 75(8): 855-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25683382

RESUMEN

BACKGROUND: The downregulation of the tumor suppressor miR200c plays important roles in many malignant tumors. This study aims to show that miR200c is a posttranscriptional regulator of insulin receptor substrate 1 (IRS1) and over-expression of miR200c suppresses prostate cancer cell growth. METHODS: Bioinformatics analysis was used to show potential post-translational regulation of IRS1 by miR200c. Dual reporter gene assays were chosen to test the binding of miR200c to the potential seed sequences in IRS1 3'UTR. RT-PCR, Q-PCR and western blot were applied to determine the regulation effect of miR200c on IRS1. CCK8 assay, soft agar assay, trypan blue exclusion assay and flow cytometric analysis were used to measure the biological effects of miR200c on prostate cancer cell proliferation and apoptosis. RESULTS: The 449-455 nt, 3061-3067 nt, and 3096-3102 nt of the IRS1 3'-UTR were identified as three potential seed sequences for miR200c. MiR200c directly binds to IRS1 through the seed sequences in IRS1 3'-UTR. Artificial overexpression of miR200c significantly downregulated the mRNA and protein levels of IRS1, together with decreased cell proliferation and increased cell death of PC3 and DU145 cells. CONCLUSIONS: Our results suggest that miR200c plays crucial roles in prostate cancer by post-transcriptional regulation of IRS1. The mir200c/IRS1 pathway may be a potential therapeutic target to prevent prostate cancer cell growth.


Asunto(s)
Aumento de la Célula , Proteínas Sustrato del Receptor de Insulina/biosíntesis , MicroARNs/biosíntesis , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/prevención & control , Apoptosis/fisiología , Muerte Celular/fisiología , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de la Próstata/patología , ARN Mensajero/biosíntesis
8.
Tumour Biol ; 36(11): 8389-98, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26018509

RESUMEN

MicroRNAs play critical roles in the development and progression of human cancers. Although it has been reported that miR-106a* is downregulated in follicular lymphoma, its role in renal cell carcinoma (RCC) remains unknown. This study investigated the expression and role of miR-106a* in human RCC. Our results showed that the miR-106a* expression decreased dramatically in clinical RCC tissues and cell lines. In vitro, overexpression of miR-106a* suppressed RCC cell proliferation and S/G2 transition, whereas inhibition of miR-106a* promoted cell proliferation and S/G2 transition. It was also found that miR-106a* expression was inversely correlated with the expression of insulin receptor substrate 2 (IRS-2). IRS-2 was determined to be a direct target of miR-106a* by a luciferase reporter assay. Importantly, silencing IRS-2 resulted in the same biologic effects as those of miR-106a* overexpression in RCC cells, including inhibition of RCC cell proliferation and triggering of S/G2 cell cycle arrest with inhibition of the PI3K/Akt signaling pathway. These results indicate that miR-106a* affects RCC progression by targeting IRS-2 with suppression of the PI3K/Akt signaling pathway in RCC cells. The findings suggest miR-106a* as a novel strategy for RCC treatment.


Asunto(s)
Carcinoma de Células Renales/genética , Proteínas Sustrato del Receptor de Insulina/biosíntesis , MicroARNs/genética , Adulto , Anciano , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Masculino , MicroARNs/biosíntesis , Persona de Mediana Edad , Proteína Oncogénica v-akt/genética , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal/genética
9.
Tumour Biol ; 36(8): 6507-13, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25820822

RESUMEN

AflatoxinB1 (AFB1) is well known as a potent carcinogen. Epidemiological studies have shown an association between AFB1 exposure and lung cancer in humans. AFB1 can induce the mutations of genes such as tumor suppressor p53 through its metabolite AFB1-8,9-exo-epoxide, which acts as a mutagen to react with DNA. In addition, recent study demonstrates AFB1 positively regulates type I insulin-like growth factor receptor (IGF-IR) signaling in hepatoma cells. The current study aims to determine the effects of AFB1 on Src kinase and insulin receptor substrate (IRS) in lung cancer cells and the effects of AFB1 on lung cancer cell migration. To this end, the effects of AFB1 on IRS expression, Src, Akt, and ERK phosphorylation were measured by Western blot analysis. The migration of lung cancer cells was detected by wound-healing assay. AFB1 downregulates IRS1 but paradoxically upregulates IRS2 through positive regulation of the stability of IRS2 and the proteasomal degradation of IRS1 in lung cancer cell lines A549 and SPCA-1. In addition, AFB1 induces Src, Akt, and ERK1/2 phosphorylation. Treatment of lung cancer cells with Src inhibitor saracatinib abrogates AFB1-induced IRS2 accumulation. Moreover, AFB1 stimulates lung cancer cell migration, which can be inhibited by saracatinib. We conclude that AFB1 may upregulate IRS2 and stimulate lung cancer cell migration through Src.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteínas Sustrato del Receptor de Insulina/genética , Neoplasias Pulmonares/genética , Familia-src Quinasas/genética , Aflatoxina B1/toxicidad , Benzodioxoles/administración & dosificación , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/patología , Fosforilación/efectos de los fármacos , Quinazolinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/biosíntesis
10.
Circ Res ; 113(1): 40-51, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23603512

RESUMEN

RATIONALE: Endothelial microRNA-126 (miR-126) modulates vascular development and angiogenesis. However, its role in the regulation of smooth muscle cell (SMC) function is unknown. OBJECTIVE: To elucidate the role of miR-126 secreted by endothelial cells (ECs) in regulating SMC turnover in vitro and in vivo, as well as the effects of shear stress on the regulation. METHODS AND RESULTS: Coculture of SMCs with ECs or treatment of SMCs with conditioned media from static EC monoculture (EC-CM) increased SMC miR-126 level and SMC turnover; these effects were abolished by inhibition of endothelial miR-126 and by the application of laminar shear stress to ECs. SMC miR-126 did not increase when treated with EC-CM from ECs subjected to inhibition of miR biogenesis, or with CM from sheared ECs. Depletion of extracellular/secreted vesicles in EC-CM did not affect the increase of SMC miR-126 by EC-CM. Biotinylated miR-126 or FLAG (DYKDDDDK epitope)-tagged Argonaute2 transfected into ECs was detected in the cocultured or EC-CM-treated SMCs, indicating a direct EC-to-SMC transmission of miR-126 and Argonaute2. Endothelial miR-126 represses forkhead box O3, B-cell lymphoma 2, and insulin receptor substrate 1 mRNAs in the cocultured SMCs, suggesting the functional roles of the transmitted miR-126. Systemic depletion of miR-126 in mice inhibited neointimal lesion formation of carotid arteries induced by cessation of blood flow. Administration of EC-CM or miR-126 mitigated the inhibitory effect. CONCLUSIONS: Endothelial miR-126 acts as a key intercellular mediator to increase SMC turnover, and its release is reduced by atheroprotective laminar shear stress.


Asunto(s)
Células Endoteliales/metabolismo , Regulación de la Expresión Génica , MicroARNs/fisiología , Miocitos del Músculo Liso/citología , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/fisiología , Arteria Carótida Común/patología , Técnicas de Cultivo de Célula/instrumentación , Línea Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Silenciador del Gen , Genes bcl-2 , Hemorreología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Ligadura , Ratones , MicroARNs/genética , MicroARNs/uso terapéutico , Músculo Liso Vascular/citología , Neointima , Comunicación Paracrina , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Recombinantes de Fusión/fisiología , Arterias Umbilicales/citología
11.
Growth Factors ; 32(1): 41-52, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24438070

RESUMEN

The insulin-like growth factor-1 system, including its critical mediator insulin receptor substrate-1 (IRS-1), is involved in regulating osteosarcoma (OS) cell proliferation or differentiation. The aim of this study is to define the role of IRS-1 in OS cells by assessing the contribution of IRS-1 in the differentiation of human and murine OS cell lines and mouse mesenchymal stem cells (MSCs) and found that the basal level of IRS-1 is important for the initiation of differentiation. Both down-regulation and over-expression of IRS-1 inhibited osteoblastic differentiation. In vivo studies showed that OS cells over-expressing IRS-1 have increased metastatic potential and tumor growth. The proteasome inhibitor MG-132 led to an increase in IRS-1 protein level that inhibited osteoblastic differentiation, suggesting a role for proteasomal regulation in maintaining the appropriate expression level of IRS-1. Thus, precise regulation of IRS-1 expression level is critical for determining the differentiating capacity of MSCs and OS cells, and that derangement of IRS-1 levels can be a critical step in OS transformation.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/biosíntesis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Osteosarcoma/patología , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Inhibidores de Cisteína Proteinasa/farmacología , Regulación del Desarrollo de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Leupeptinas/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Osteocalcina/biosíntesis , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/genética , Factor de Transcripción Sp7 , Factores de Transcripción/biosíntesis
12.
Am J Physiol Gastrointest Liver Physiol ; 304(4): G420-7, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23257919

RESUMEN

Obesity, inflammation, and insulin resistance are closely linked. Substance P (SP), via its neurokinin 1 receptor (NK1R), mediates inflammatory and, possibly, neuroendocrine processes. We examined SP effects on lipid storage and cytokine production in 3T3-L1 adipocytes and adipose tissues. 3T3-L1 adipocytes and preadipocytes express NK1R, and 8 days of SP supplementation during differentiation to 3T3-L1 preadipocytes decreased lipid droplet accumulation. SP (10 nM, 24 h) increased lipolysis in primary adipocytes (138 ± 7%, P < 0.05) and reduced fatty acid uptake (-31 ± 7%, P < 0.05) and mRNA expression of the differentiation-related transcription factors peroxisome proliferator-activated receptor-γ type 2 (-64 ± 2%, P < 0.001) and CCAAT enhancer-binding protein (CEBP)-α (-65 ± 2%, P < 0.001) and the lipid storage genes fatty acid-binding protein type 4 (-59 ± 2%, P < 0.001) and diacylglycerol O-acyltransferase-1 (-45 ± 2%, P < 0.01) in 3T3-L1 adipocytes, while CD36, a fatty acid transporter (+82 ± 19%, P < 0.01), was augmented. SP increased secretion of complement C3 (148 ± 15%, P < 0.04), monocyte chemoattractant protein-1 (156 ± 16%, P < 0.03), and keratinocyte-derived chemokine (148 ± 18%, P = 0.045) in 3T3-L1 adipocytes and monocyte chemoattractant protein-1 (496 ± 142%, P < 0.02) and complement C3 (152 ± 25%, P < 0.04) in adipose tissue and primary adipocytes, respectively. These SP effects were accompanied by downregulation of insulin receptor substrate 1 (-82 ± 2%, P < 0.01) and GLUT4 (-76 ± 2%, P < 0.01) mRNA expression, and SP acutely blocked insulin-mediated stimulation of fatty acid uptake and Akt phosphorylation. Although adiponectin secretion was unchanged, mRNA expression was decreased (-86 ± 8%, P < 0.001). In humans, NK1R expression correlates positively with plasma insulin, fatty acid, and complement C3 and negatively with adiponectin, CEBPα, CEBPß, and peroxisome proliferator-activated receptor-γ mRNA expression in omental, but not subcutaneous, adipose tissue. Our results suggest that, beyond its neuroendocrine and inflammatory effects, SP could also be involved in targeting adipose tissue and influencing insulin resistance.


Asunto(s)
Adipocitos/metabolismo , Adipoquinas/biosíntesis , Sustancia P/fisiología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Animales , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Diferenciación Celular/efectos de los fármacos , Quimiocina CCL2 , Complemento C3/metabolismo , Diacilglicerol O-Acetiltransferasa/metabolismo , Proteínas de Unión a Ácidos Grasos/biosíntesis , Ácidos Grasos/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , PPAR gamma/biosíntesis , Receptores de Neuroquinina-1/biosíntesis
13.
Arterioscler Thromb Vasc Biol ; 32(5): 1124-31, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22426129

RESUMEN

OBJECTIVE: To determine the contribution of hyperinsulinemia to atherosclerosis development. METHODS AND RESULTS: Apolipoprotein E (Apoe) null mice that had knockout of a single allele of the insulin receptor (Insr) gene were compared with littermate Apoe null mice with intact insulin receptors. Plasma insulin levels in Insr haploinsufficient/Apoe null mice were 50% higher in the fasting state and up to 69% higher during a glucose tolerance test, but glucose tolerance was not different in the 2 groups. C-peptide levels, insulin sensitivity, and postreceptor insulin signaling in muscle, liver, fat, and aorta were not different between groups, whereas disappearance in plasma of an injected insulin analog was delayed in Insr haploinsufficient/Apoe null mice, indicating that impaired insulin clearance was the primary cause of hyperinsulinemia. No differences were observed in plasma lipids or blood pressure. Despite the hyperinsulinemia, atherosclerotic lesion size was not different between the 2 groups at time points up to 52 weeks of age when measured as en face lesion area in the aorta, cross-sectional plaque area in the aortic sinus, and cholesterol abundance in the brachiocephalic artery. CONCLUSIONS: Hyperinsulinemia, without substantial vascular or whole-body insulin resistance and without changes in plasma lipids or blood pressure, does not change susceptibility to atherosclerosis.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/genética , Hiperinsulinismo/complicaciones , Resistencia a la Insulina , Animales , Apolipoproteínas E/sangre , Aterosclerosis/sangre , Aterosclerosis/etiología , Progresión de la Enfermedad , Femenino , Regulación de la Expresión Génica , Hiperinsulinismo/sangre , Hiperinsulinismo/genética , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
14.
Exp Cell Res ; 318(14): 1745-58, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22652453

RESUMEN

The adapters IRS1 and IRS2 link growth factor receptors to downstream signaling pathways that regulate proliferation and survival. Both suppress factor-withdrawal-induced apoptosis and have been implicated in cancer progression. However, recent studies suggest IRS1 and IRS2 mediate differential functions in cancer pathogenesis. IRS1 promoted breast cancer proliferation, while IRS2 promoted metastasis. The role of IRS1 and IRS2 in controlling cell responses to chemotherapy is unknown. To determine the role of IRS1 and IRS2 in the sensitivity of cells to chemotherapy, we treated 32D cells lacking or expressing IRS proteins with various concentrations of chemotherapeutic agents. We found that expression of IRS1, in contrast to IRS2, enhanced the sensitivity of 32D cells to chemotherapy-induced apoptosis. When IRS2 was expressed with IRS1, the cells no longer showed enhanced sensitivity. Expression of IRS1 did not alter the expression of pro- and anti-apoptotic proteins; however, 32D-IRS1 cells expressed higher levels of Annexin A2. In 32D-IRS1 cells, IRS1 and Annexin A2 were both located in cytoplasmic and membrane fractions. We also found that IRS1 coprecipitated with Annexin A2, while IRS2 did not. Decreasing Annexin A2 levels reduced 32D-IRS1 cell sensitivity to chemotherapy. These results suggest IRS1 enhances sensitivity to chemotherapy in part through Annexin A2.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Animales , Anexina A2/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Relación Estructura-Actividad
15.
J Biol Chem ; 286(33): 29388-29396, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21700708

RESUMEN

Insulin receptor substrate-2 (IRS-2) plays a critical role in the survival and function of pancreatic ß-cells. Gene disruption of IRS-2 results in failure of the ß-cell compensatory mechanism and diabetes. Nonetheless, the regulation of IRS-2 protein expression in ß-cells remains largely unknown. Inducible nitric-oxide synthase (iNOS), a major mediator of inflammation, has been implicated in ß-cell damage in type 1 and type 2 diabetes. The effects of iNOS on IRS-2 expression have not yet been investigated in ß-cells. Here, we show that iNOS and NO donor decreased IRS-2 protein expression in INS-1/832 insulinoma cells and mouse islets, whereas IRS-2 mRNA levels were not altered. Interleukin-1ß (IL-1ß), alone or in combination with interferon-γ (IFN-γ), reduced IRS-2 protein expression in an iNOS-dependent manner without altering IRS-2 mRNA levels. Proteasome inhibitors, MG132 and lactacystin, blocked the NO donor-induced reduction in IRS-2 protein expression. Treatment with NO donor led to activation of glycogen synthase kinase-3ß (GSK-3ß) and c-Jun N-terminal kinase (JNK/SAPK) in ß-cells. Inhibition of GSK-3ß by pharmacological inhibitors or siRNA-mediated knockdown significantly prevented NO donor-induced reduction in IRS-2 expression in ß-cells. In contrast, a JNK inhibitor, SP600125, did not effectively block reduced IRS-2 expression in NO donor-treated ß-cells. These data indicate that iNOS-derived NO reduces IRS-2 expression by promoting protein degradation, at least in part, through a GSK-3ß-dependent mechanism. Our findings suggest that iNOS-mediated decreased IRS-2 expression may contribute to the progression and/or exacerbation of ß-cell failure in diabetes.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Células Secretoras de Insulina/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Animales , Antracenos/farmacología , Línea Celular Tumoral , Inhibidores de Cisteína Proteinasa/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Células Secretoras de Insulina/citología , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leupeptinas/farmacología , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma , Ratas
16.
Int J Cancer ; 130(12): 2771-82, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21805474

RESUMEN

Ursodeoxycholic acid (UDCA) attenuates colon carcinogenesis in humans and in animal models by an unknown mechanism. We investigated UDCA effects on normal intestinal epithelium in vivo and in vitro to identify the potential chemopreventive mechanism. Feeding of mice with 0.4% UDCA reduced cell proliferation to 50% and suppressed several potential proproliferatory genes including insulin receptor substrate 1 (Irs-1). A similar transcriptional response was observed in the rat intestinal cell line IEC-6 which was then used as an in vitro model. UDCA slowed down the proliferation of IEC-6 cells and induced sustained hyperphosphorylation of ERK1/ERK2 kinases which completely inhibited the proproliferatory effects of EGF and IGF-1. The hyperphosphorylation of ERK1 led to a transcriptional suppression of the Irs-1 gene. Both, the hyperphosphorylation of ERK as well as the suppression of Irs-1 were sufficient to inhibit proliferation of IEC-6 cells. ERK1/ERK2 inhibition in vitro or ERK1 elimination in vitro or in vivo abrogated the antiproliferatory effects of UDCA. We show that UDCA inhibits proliferation of nontransformed intestinal epithelial cells by inducing a sustained hyperphosphorylation of ERK1 kinase which slows down the cell cycle and reduces expression of Irs-1 protein. These data extend our understanding of the physiological and potentially chemopreventive effects of UDCA and identify new targets for chemoprevention.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Ácido Ursodesoxicólico/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular , Factor de Crecimiento Epidérmico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Femenino , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Mucosa Intestinal/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Ácido Ursodesoxicólico/metabolismo
17.
Biochem Biophys Res Commun ; 424(3): 482-7, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22776200

RESUMEN

Studies have shown similarities between the histopathological characteristics of NPC and Alzheimer's disease (AD) including amyloid and tau pathologies. While dysfunction in insulin signaling was widely detected in AD brain, the function of insulin signaling proteins has not been examined in NPC disease. In this study, we have examined the expression and phosphorylation of proteins linked to the insulin signaling pathway in the brain of 9 weeks old NPC(nih) mice. Our results showed lower expression of insulin receptor substrate 2 (IRS2) in the NPC(nih) mice, and insulin receptor substrate 1 (IRS1) expression was almost non-detectable in this NPC mouse model. This reduction was associated with the loss of expression for the regulatory p85 subunit of phosphatidylinositol 3-kinase (p85/PI3K). Interestingly, the impairment was observed to link to a greater reduction of Akt phosphorylation at residue T308 than S473. This aberrant Akt phosphorylation could be contributing to lower GSK3ß phosphorylation detected in the NPC(nih) mouse brain. To our knowledge, this is the first report documenting impaired insulin signaling in the brain of a NPC mouse model.


Asunto(s)
Insulina/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Animales , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Enfermedad de Niemann-Pick Tipo C/genética , Fosforilación , Transducción de Señal
18.
Am J Physiol Endocrinol Metab ; 300(3): E536-42, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21189360

RESUMEN

To determine whether serine/threonine ROCK1 is activated by insulin in vivo in humans and whether impaired activation of ROCK1 could play a role in the pathogenesis of insulin resistance, we measured the activity of ROCK1 and the protein content of the Rho family in vastus lateralis muscle of lean, obese nondiabetic, and obese type 2 diabetic subjects. Biopsies were taken after an overnight fast and after a 3-h hyperinsulinemic euglycemic clamp. Insulin-stimulated GDR was reduced 38% in obese nondiabetic subjects compared with lean, 62% in obese diabetic subjects compared with lean, and 39% in obese diabetic compared with obese nondiabetic subjects (all comparisons P < 0.001). Insulin-stimulated IRS-1 tyrosine phosphorylation is impaired 41-48% in diabetic subjects compared with lean or obese subjects. Basal activity of ROCK1 was similar in all groups. Insulin increased ROCK1 activity 2.1-fold in lean and 1.7-fold in obese nondiabetic subjects in muscle. However, ROCK1 activity did not increase in response to insulin in muscle of obese type 2 diabetic subjects without change in ROCK1 protein levels. Importantly, insulin-stimulated ROCK1 activity was positively correlated with insulin-mediated GDR in lean subjects (P < 0.01) but not in obese or type 2 diabetic subjects. Moreover, RhoE GTPase that inhibits the catalytic activity of ROCK1 by binding to the kinase domain of the enzyme is notably increased in obese type 2 diabetic subjects, accounting for defective ROCK1 activity. Thus, these data suggest that ROCK1 may play an important role in the pathogenesis of resistance to insulin action on glucose disposal in muscle of obese type 2 diabetic subjects.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Hipoglucemiantes/farmacología , Insulina/farmacología , Músculo Esquelético/metabolismo , Quinasas Asociadas a rho/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Adulto , Biotransformación/efectos de los fármacos , Western Blotting , Índice de Masa Corporal , Proteínas del Citoesqueleto/metabolismo , Femenino , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Humanos , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Resistencia a la Insulina , Cinética , Masculino , Persona de Mediana Edad , Músculo Esquelético/efectos de los fármacos , Obesidad/metabolismo , Fosforilación , Tirosina/metabolismo , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA/metabolismo
19.
Biochem J ; 430(1): 141-9, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20522023

RESUMEN

A paradoxical but common finding in the obesity clinic is the identification of individuals who can be considered 'inappropriately' healthy for their degree of obesity. We think that studying these obese but metabolically healthy individuals and comparing them with equally obese but insulin-resistant individuals could provide important insights into the mechanistic link between adipose tissue expansion and associated metabolic alterations. In the present study, we investigated whether there are differences in inflammatory and insulin signalling pathways in VAT (visceral adipose tissue) that could account for the metabolic differences exhibited by morbidly obese individuals who are either insulin-resistant (IR-MO) or paradoxically insulin-sensitive (NIR-MO). Our results indicate that there are pathways common to obesity and unrelated to insulin resistance and others that are discriminative for insulin resistance for a similar degree of obesity. For instance, all morbidly obese patients, irrespective of their insulin resistance, showed increased expression of TNFalpha (tumour necrosis factor alpha) and activation of JNK1/2 (c-Jun N-terminal kinase 1/2). However, the IR-MO group showed significantly elevated expression levels of IL (interleukin)-1beta and IL-6 and increased macrophage infiltrates compared with non-obese individuals and NIR-MO. IkappaBalpha [inhibitor of NF-kappaB (nuclear factor kappaB) alpha], the activation of ERK1/2 (extracellular-signal-regulated kinase 1/2) and NF-kappaB were discriminative of the state of insulin resistance and correlated with differential changes in IRS-1 (insulin receptor substrate 1) expression and Akt activation between IR-MO and NIR-MO individuals. Our results support the concept that NIR-MO individuals lack the inflammatory response that characterizes the IR-MO patient and that IL-6, IL-1beta, ERK and NF-kappaB are important effectors that mediate the inflammation effects promoting insulin resistance.


Asunto(s)
Obesidad Mórbida/inmunología , Adulto , Biomarcadores/metabolismo , Femenino , Humanos , Proteínas I-kappa B/biosíntesis , Proteínas I-kappa B/genética , Inflamación/inmunología , Inflamación/metabolismo , Insulina/fisiología , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Resistencia a la Insulina , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-6/biosíntesis , Interleucina-6/genética , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/metabolismo , Macrófagos/inmunología , Masculino , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Proteínas Quinasas Activadas por Mitógenos/genética , Inhibidor NF-kappaB alfa , FN-kappa B/biosíntesis , FN-kappa B/genética , Obesidad Mórbida/metabolismo , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
20.
J Vet Med Sci ; 73(2): 169-75, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20877155

RESUMEN

The Spontaneously Diabetic Torii (SDT) fatty rat, established by introducing the fa allele of the Zucker fatty rat into the SDT rat genome, is a new model of obesity/type 2 diabetes. The present study investigated effects of food restriction on metabolic and endocrinological function in SDT fatty rats. SDT fatty rats were pair-fed with SDT rats from 7 to 21 weeks of age. The SDT fatty rats were already hyperinsulinemic and hyperlipidemic at 7 weeks of age. After 7 weeks of age, SDT fatty rats showed age-dependently increasing serum glucose levels associated with decreasing serum insulin levels. However, in pair-fed SDT fatty rats, hyperglycemia and hyperinsulinemia were attenuated at 9 weeks of age. After 9 weeks of age, the serum insulin levels unexpectedly increased in the pair-fed SDT fatty rats. Glucose tolerance was also improved, and the pancreatic insulin contents were increased in these rats. Pancreatic islets were hypertrophied in pair-fed SDT fatty rats compared with ad lib-fed SDT fatty rats, which were comparable to SDT rats. This study showed that, in SDT fatty rats, calorie restriction by paired-feeding with SDT rats attenuated hyperglycemia and hyperinsulinemia for the first 2 weeks. Thereafter, the serum insulin levels and pancreatic insulin contents were increased, though the restriction was continued. Hypertrophic pancreatic islets were also remarkable, indicating increased beta cell proliferation. The activated pancreatic beta cell functions might be due to rapid food ingestion, a change of feeding behavior resulting form increasing the fasting period, which was indispensable for calorie restriction.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Privación de Alimentos/fisiología , Islotes Pancreáticos/metabolismo , Animales , Peso Corporal/fisiología , Modelos Animales de Enfermedad , Ingestión de Alimentos/fisiología , Glucoquinasa/biosíntesis , Glucoquinasa/genética , Transportador de Glucosa de Tipo 2/biosíntesis , Transportador de Glucosa de Tipo 2/genética , Histocitoquímica , Insulina/sangre , Insulina/genética , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Islotes Pancreáticos/ultraestructura , Masculino , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Zucker , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA