Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Diabetologia ; 60(2): 314-323, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27807598

RESUMEN

AIMS/HYPOTHESIS: Salt-inducible kinases (SIKs) are related to the metabolic regulator AMP-activated protein kinase (AMPK). SIK2 is abundant in adipose tissue. The aims of this study were to investigate the expression of SIKs in relation to human obesity and insulin resistance, and to evaluate whether changes in the expression of SIKs might play a causal role in the development of disturbed glucose uptake in human adipocytes. METHODS: SIK mRNA and protein was determined in human adipose tissue or adipocytes, and correlated to clinical variables. SIK2 and SIK3 expression and phosphorylation were analysed in adipocytes treated with TNF-α. Glucose uptake, GLUT protein levels and localisation, phosphorylation of protein kinase B (PKB/Akt) and the SIK substrate histone deacetylase 4 (HDAC4) were analysed after the SIKs had been silenced using small interfering RNA (siRNA) or inhibited using a pan-SIK-inhibitor (HG-9-91-01). RESULTS: We demonstrate that SIK2 and SIK3 mRNA are downregulated in adipose tissue from obese individuals and that the expression is regulated by weight change. SIK2 is also negatively associated with in vivo insulin resistance (HOMA-IR), independently of BMI and age. Moreover, SIK2 protein levels and specific kinase activity display a negative correlation to BMI in human adipocytes. Furthermore, SIK2 and SIK3 are downregulated by TNF-α in adipocytes. Silencing or inhibiting SIK1-3 in adipocytes results in reduced phosphorylation of HDAC4 and PKB/Akt, less GLUT4 at the plasma membrane, and lower basal and insulin-stimulated glucose uptake in adipocytes. CONCLUSION/INTERPRETATION: This is the first study to describe the expression and function of SIKs in human adipocytes. Our data suggest that SIKs might be protective in the development of obesity-induced insulin resistance, with implications for future treatment strategies.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adulto , Anciano , Animales , Western Blotting , Femenino , Humanos , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Masculino , Ratones , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/farmacología
2.
Diabetologia ; 58(9): 2115-23, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26115698

RESUMEN

AIMS/HYPOTHESIS: Dysregulated expression of metabolic and inflammatory genes is a prominent consequence of obesity causing insulin resistance and type 2 diabetes. Finding causative factors is essential to understanding progression of these pathologies and discovering new therapeutic targets. The transcription factor V-maf musculoaponeurotic fibrosarcoma oncogene homologue B (MAFB) is highly expressed in human white adipose tissue (WAT). However, its role in the regulation of WAT function is elusive. We aimed to characterise MAFB expression and function in human WAT in the context of obesity and insulin resistance. METHODS: MAFB mRNA expression was evaluated in human WAT from seven cohorts with large inter-individual variation in BMI and metabolic features. Insulin-induced adipocyte lipogenesis and lipolysis were measured and correlated with MAFB expression. MAFB regulation during adipogenesis and the effects of MAFB suppression in human adipocytes was investigated. MAFB regulation by TNF-α was examined in human primary adipocytes and THP-1 monocytes/macrophages. RESULTS: MAFB expression in human adipocytes is upregulated during adipogenesis, increases with BMI in WAT, correlates with adverse metabolic features and is decreased after weight loss. MAFB downregulation decreases proinflammatory gene expression in adipocytes and interferes with TNF-α effects. Interestingly, MAFB is differentially regulated by TNF-α in adipocytes (suppressed) and THP-1 cells (upregulated). Further, MAFB is primarily expressed in WAT macrophages/monocytes and its expression correlates with macrophage and inflammatory markers. CONCLUSIONS/INTERPRETATION: Our findings indicate that MAFB is a regulator and a marker of adipose tissue inflammation, a process that subsequently causes insulin resistance.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Regulación de la Expresión Génica , Inflamación/metabolismo , Factor de Transcripción MafB/metabolismo , Adipocitos/citología , Tejido Adiposo Blanco/patología , Índice de Masa Corporal , Diferenciación Celular , Estudios de Cohortes , Humanos , Resistencia a la Insulina , Lipogénesis , Lipólisis , Macrófagos/citología , Células Madre Mesenquimatosas/citología , Monocitos/citología , Obesidad/metabolismo , Análisis de Regresión , Factor de Necrosis Tumoral alfa/metabolismo
3.
Endocrinology ; 162(7)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33963396

RESUMEN

CONTEXT: Healthy hyperplasic (many but smaller fat cells) white adipose tissue (WAT) expansion is mediated by recruitment, proliferation and/or differentiation of new fat cells. This process (adipogenesis) is controlled by transcriptional programs that have been mostly identified in rodents. OBJECTIVE: A systemic investigation of adipogenic human transcription factors (TFs) that are relevant for metabolic conditions has not been revealed previously. METHODS: TFs regulated in WAT by obesity, adipose morphology, cancer cachexia, and insulin resistance were selected from microarrays. Their role in differentiation of human adipose tissue-derived stem cells (hASC) was investigated by RNA interference (RNAi) screen. Lipid accumulation, cell number, and lipolysis were measured for all screened factors (148 TFs). RNA (RNAseq), protein (Western blot) expression, insulin, and catecholamine responsiveness were examined in hASC following siRNA treatment of selected target TFs. RESULTS: Analysis of TFs regulated by metabolic conditions in human WAT revealed that many of them belong to adipogenesis-regulating pathways. The RNAi screen identified 39 genes that affected fat cell differentiation in vitro, where 11 genes were novel. Of the latter JARID2 stood out as being necessary for formation of healthy fat cell metabolic phenotype by regulating expression of multiple fat cell phenotype-specific genes. CONCLUSION: This comprehensive RNAi screening in hASC suggests that a large proportion of WAT TFs that are impacted by metabolic conditions might be important for hyperplastic adipose tissue expansion. The screen also identified JARID2 as a novel TF essential for the development of functional adipocytes.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/genética , Complejo Represivo Polycomb 2/genética , Interferencia de ARN/fisiología , Factores de Transcripción/análisis , Factores de Transcripción/genética , Adipocitos/química , Adipocitos/patología , Tejido Adiposo Blanco/química , Tejido Adiposo Blanco/patología , Adolescente , Secuencia de Bases , Diferenciación Celular/genética , Células Cultivadas , Femenino , Neoplasias Gastrointestinales , Regulación de la Expresión Génica , Humanos , Hiperplasia/genética , Resistencia a la Insulina/genética , Masculino , Obesidad/genética , Complejo Represivo Polycomb 2/fisiología , Células Madre/química , Factores de Transcripción/fisiología
4.
Sci Data ; 4: 170164, 2017 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-29087381

RESUMEN

Obesity affects gene expression and metabolism of white adipose tissue (WAT), which results in insulin resistance (IR) and type 2 diabetes. However, WAT is a heterogeneous organ containing many cell types that might respond differently to obesity-induced changes. We performed flow cytometry sorting and RNA expression profiling by microarray of major WAT cell types (adipocytes, CD45-/CD31-/CD34+ progenitors, CD45+/CD14+ monocytes/ macrophages, CD45+/CD14- leukocytes), which allowed us to identify genes enriched in specific cell fractions. Additionally, we included adipocytes and adipocyte progenitor cells obtained from lean and obese individuals. Taken together, we provide a detailed gene expression atlas of major human adipose tissue resident cell types for clinical/basic research and using this dataset provide lists of cell-type specific genes that are of interest for metabolic research.


Asunto(s)
Adipocitos , Células Madre , Transcriptoma , Tejido Adiposo , Humanos , Obesidad/patología , Especificidad de Órganos
5.
Diabetes ; 66(1): 218-230, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27803022

RESUMEN

White adipose tissue (WAT) can develop into several phenotypes with different pathophysiological impact on type 2 diabetes. To better understand the adipogenic process, the transcriptional events that occur during in vitro differentiation of human adipocytes were investigated and the findings linked to WAT phenotypes. Single-molecule transcriptional profiling provided a detailed map of the expressional changes of genes, enhancers, and long noncoding RNAs, where different types of transcripts share common dynamics during differentiation. Common signatures include early downregulated, transient, and late induced transcripts, all of which are linked to distinct developmental processes during adipogenesis. Enhancers expressed during adipogenesis overlap significantly with genetic variants associated with WAT distribution. Transiently expressed and late induced genes are associated with hypertrophic WAT (few but large fat cells), a phenotype closely linked to insulin resistance and type 2 diabetes. Transcription factors that are expressed early or transiently affect differentiation and adipocyte function and are controlled by several well-known upstream regulators such as glucocorticosteroids, insulin, cAMP, and thyroid hormones. Taken together, our results suggest a complex but highly coordinated regulation of adipogenesis.


Asunto(s)
Adipogénesis/fisiología , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/genética , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Adiposidad/genética , Adiposidad/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Resistencia a la Insulina/fisiología , Lipólisis/genética , Lipólisis/fisiología , Análisis de Componente Principal , ARN Interferente Pequeño/genética , Factores de Transcripción/metabolismo
6.
Nutr Metab (Lond) ; 13: 4, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26788115

RESUMEN

BACKGROUND: Previous studies suggest that intake of specific bioactive compounds may have beneficial clinical effects on adipose tissue partly due to their anti-inflammatory and insulin-sensitizing properties. With the overall aim to contribute to better understanding of the mechanisms of selected bioactive nutrients on fat metabolism, we investigated their role on human white adipocyte function. METHODS: The influence of the omega-3-fatty acid docosahexaenoic acid (DHA), the anthocyanin (AC) cyanidin-3-glucoside and its metabolite protocatechuic acid, and the beta-glucan metabolite propionic acid (PI) on adipokine secretion, fatty acid metabolism (lipolysis/lipogenesis) and adipocyte differentiation (lipid accumulation) was studied in human fat cells differentiated in vitro. To investigate possible synergistic, additive or antagonistic effects, DHA was also combined with AC or PI. RESULTS: Each compound, alone or together with DHA, suppressed basal adipocyte lipolysis compared to control treated cells. DHA alone attenuated the secretion of pro-inflammatory adipokines such as chemerin, interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1/CCL2), whereas AC suppressed only the latter two. Treatment with PI decreased IL-6, tumour necrosis factor alpha (TNFα) and adiponectin secretion. A combination of DHA and AC decreased TNFα secretion and increased insulin-stimulated lipogenesis. No effect was found on adipocyte differentiation. At the selected concentrations, none of the compounds was found to be cytotoxic. CONCLUSION: The studied bioactive food compounds or their metabolites have beneficial effects in human primary fat cells measured as decreased basal lipolytic activity and secretion of inflammatory markers. A minor effect was also observed on insulin-stimulated glucose uptake albeit only with the combination of DHA and AC. Taken together, our results may link the reported health benefits of the selected bioactives on metabolic disorders such as insulin resistance, hypertension and dyslipidemia to effects on white adipocytes.

7.
Reprod Toxicol ; 35: 144-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23117151

RESUMEN

Increased incidence of prostate cancer has been reported in men exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). TCDD acts through the aryl hydrocarbon receptor (AhR), which interacts with the androgen receptor (AR). The AR gene contains a polymorphic CAG repeat that influences its transcriptional activity. We investigated the influence of TCDD on prostate cancer cells (PC-3) and non-tumor prostate cells (PNT1A) on 5α-dihydrotestosterone-activated ARs containing CAG repeats within normal length range (16, 22, and 28). The AhR target gene CYP1A1 mRNA expression was induced by TCDD, but was not affected by the AR CAG length. TCDD had no effect on AR activity in PC-3 cells, whereas the shortest AR variant was induced by TCDD in PNT1A cells. In conclusion, the CAG length dependent effect of TCDD on AR activity in PNT1A, but not in PC-3 cells, indicates as a cell-specific effect of TCDD on AR activity.


Asunto(s)
Contaminantes Ambientales/toxicidad , Dibenzodioxinas Policloradas/toxicidad , Receptores Androgénicos/genética , Línea Celular , Línea Celular Tumoral , Citocromo P-450 CYP1A1/genética , Humanos , Masculino , Próstata/citología , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos
8.
Reprod Toxicol ; 32(3): 293-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21729750

RESUMEN

Recently, the effect of exposure to persistent organic pollutants (POPs) on sperm concentration was only seen in men with a short androgen receptor (AR) gene CAG repeat. In order to investigate whether these effects could be observed also in vitro, we tested the impact of 2,2',4,4',5,5'-hexachlorobiphenyl (CB-153) and 1,1-bis-(4-chlorophenyl)-2,2-dichloroethene (4,4'-DDE) on 5α-dihydrotestosterone activated ARs containing 16, 22 and 28 CAG repeats, respectively. Single exposure to 4,4'-DDE had the most pronounced effect on the AR activity containing 16 CAG repeats, whereas 28 CAG was the most sensitive variant when a mixture of the two compounds was added. Thus, our in vitro results have confirmed the in vivo data indicating a CAG repeat length dependent effect of endocrine disrupters on the AR activity.


Asunto(s)
Diclorodifenil Dicloroetileno/toxicidad , Contaminantes Ambientales/toxicidad , Bifenilos Policlorados/toxicidad , Receptores Androgénicos/genética , Repeticiones de Trinucleótidos , Andrógenos/farmacología , Animales , Células COS , Chlorocebus aethiops , Dihidrotestosterona/farmacología , Receptores Androgénicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA