Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Chemosphere ; 349: 140852, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38048832

RESUMEN

Certain environmental chemicals affect the body's energy balance and are known as metabolism disrupting chemicals (MDCs). MDCs have been implicated in the development of metabolic diseases, such as obesity and type 2 diabetes. In contrast to their well-known impact on developing adipocytes, MDC effects leading to altered energy balance and development of insulin resistance in mature white adipocytes, constituents of adult adipose tissue, are largely unclear. Here, we investigated the effects of six well-established environmental MDCs (bisphenol A (BPA), perfluorooctanoic acid (PFOA), triclosan (TCS), p,p-dichlorodiphenyl-dichloroethylene (ppDDE), tributyltin chloride (TBT) and triphenyl phosphate (TPP)) on mature human white adipocytes derived from mesenchymal stem cells in vitro. We aimed to identify biomarkers and sensitive endpoints of their metabolism disrupting effects. While most of the tested exposures had no effect on adipocyte glucose consumption, lipid storage and assessed gene expression endpoints, the highest concentration of triclosan affected the total lipid storage and adipocyte size, as well as glucose consumption and mRNA expression of the glucose transporter GLUT1, leptin and adiponectin. Additionally, an increased expression of adiponectin was observed with TPP and the positive control PPARγ agonist rosiglitazone. In contrast, the lipidomic analysis of the cell culture medium after a 3-day exposure was extremely sensitive and revealed concentration-dependent changes in the extracellular lipidome of adipocytes exposed to nearly all studied chemicals. While some of the extracellular lipidome changes were specific for the MDC used, some effects were found common to several tested chemicals and included increases in lysophosphatidylcholines, glycerophospholipids and ceramides and a decrease in fatty acids, with possible implications in inflammation, lipid and glucose uptake. This study points to early signs of metabolic disruption and likely systemic effects of mature adipocyte exposure to environmental chemicals, as well as to the need to include lipidomic endpoints in the assessment of adverse effects of MDCs.


Asunto(s)
Diabetes Mellitus Tipo 2 , Triclosán , Humanos , Adipocitos Blancos , Lipidómica , Adiponectina , Triclosán/toxicidad , Glucosa/farmacología
2.
J Pharm Sci ; 108(1): 652-660, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30419273

RESUMEN

Many marketed pharmaceuticals reach extremely high tissue concentrations due to accumulation in lysosomes (lysosomotropism). Quantitative prediction of intracellular concentrations of accumulating drugs is challenging, especially for macrocyclic compounds that mainly do not fit in current in silico models. We tested a unique library of 47 compounds (containing 39 macrocycles) specifically designed to cover the entire range of accumulation intensities observed with pharmaceuticals so far. For the first time, we show that intracellular concentration of compounds measured by liquid chromatography with tandem mass spectrometry correlates with the induction of phospholipidosis and inhibition of autophagy, but the highest correlation was observed with the increase of lysosomal volume (R = 0.95), all measured by high-throughput imaging assays. Based only on imaging data, we developed a 5-class in vitro model for the prediction of compound accumulation with the accuracy of 81%. The measured change of total lysosomal volume can thus be used in high-throughput screening for determination of the actual intensity of intracellular accumulation of new macrocyclic compounds. The models are largely based on macrocycles, greatly improving the screening and prediction of intracellular accumulation of this challenging class. However, all tested nonmacrocyclic compounds fitted well in the models, indicating potential use of the models in broader chemical space.


Asunto(s)
Lisosomas/química , Compuestos Macrocíclicos/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Células Hep G2 , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Ratones , Fosfolípidos/metabolismo , Células RAW 264.7
3.
Eur J Pharmacol ; 829: 44-53, 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29627311

RESUMEN

Some cationic amphiphilic drugs (CADs) have been individually reported to interfere with the differentiation of immune system cells, such as macrophages and dendritic cells. To investigate the possible generic nature of this process, in this study we aimed to see whether these drugs are capable of interfering with the differentiation of adipocytes. Further, we investigated whether this feature might be connected to the lysosomotropic character of these drugs, and their disturbance of intracellular membrane trafficking rather than to the individual pharmacologic properties of each drug. Thus, for the selected set of compounds consisting of seven structurally and pharmacologically diverse CADs and three non-CAD controls we have measured the impact on differentiation of 3T3-L1K murine preadipocytes to adipocytes. We conclude that CADs indeed inhibit adipocyte differentiation, as shown morphologically, at the level of lipid droplet formation and on the expression of genetic markers of adipocytes. Furthermore, the intensity of this inhibitory effect was found to strongly positively correlate with the extent of drug accumulation in adipocytes, with their affinity for phospholipid membranes, as well as with their ability to induce phospholipidosis and inhibit autophagy.


Asunto(s)
Adipocitos/efectos de los fármacos , Autofagia/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Lisosomas/efectos de los fármacos , Fosfolípidos/metabolismo , Células 3T3 , Adipocitos/citología , Adipogénesis/efectos de los fármacos , Animales , Membrana Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Gotas Lipídicas/efectos de los fármacos , Lisosomas/metabolismo , Ratones
4.
Chem Biol Interact ; 281: 1-10, 2018 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-29248446

RESUMEN

Many chemicals accumulate in organisms through a variety of different mechanisms. Cationic amphiphilic drugs (CADs) accumulate in lysosomes and bind to membranes causing phospholipidosis, whereas many lipophilic chemicals target adipose tissue. Perfluoroalkyl substances (PFASs) are widely used as surfactants, but many of them are highly bioaccumulating and persistent in the environment, making them notorious environmental toxicants. Understanding the mechanisms of their bioaccumulation is, therefore, important for their regulation and substitution with new, less harmful chemicals. We compared the highly bioaccumulative perfluorooctanesulfonic acid PFOS to its three less bioaccumulative alternatives perfluorooctanoic acid (PFOA), perfluorohexanoic acid (PFHxA) and perfluorobutane sulfonic acid (PFBS), in their ability to accumulate and remain in lung epithelial cells (NCI-H292) and adipocytes (3T3-L1K) in vitro. As a reference point we tested a set of cationic amphiphilic drugs (CADs), known to highly accumulate in cells and strongly bind to phospholipids, together with their respective non-CAD controls. Finally, all compounds were examined for their ability to bind to neutral lipids and phospholipids in cell-free systems. Cellular accumulation and retention of the test compounds were highly correlated between the lung epithelial cells and adipocytes. Interestingly, although an anion itself, intensities of PFOS accumulation and retention in cells were comparable to those of CAD compounds, but PFOS failed to induce phospholipidosis or alter lysosomal volume. Compared to other lipophilicity measures, phospholipophilicity shows the highest correlation (Rˆ2 = 0.75) to cellular accumulation data in both cell types and best distinguishes between high and low accumulating compounds. This indicates that binding to phospholipids may be the most important component in driving high cellular accumulation in lung epithelial cells, as well as in adipocytes, and for both CADs and bioaccumulating PFASs. Obtained continuous PLS models based on compound's affinity for phospholipids and neutral lipids can be used as good prediction models of cellular accumulation and retention of PFASs and CADs.


Asunto(s)
Ácidos Alcanesulfónicos/metabolismo , Fluorocarburos/metabolismo , Lisosomas/metabolismo , Preparaciones Farmacéuticas/metabolismo , Fosfolípidos/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Ácidos Alcanesulfónicos/química , Animales , Azitromicina/química , Azitromicina/metabolismo , Caproatos/química , Caproatos/metabolismo , Caprilatos/química , Caprilatos/metabolismo , Cationes/química , Línea Celular , Supervivencia Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fluorocarburos/química , Humanos , Análisis de los Mínimos Cuadrados , Modelos Lineales , Lípidos/química , Ratones , Preparaciones Farmacéuticas/química , Fosfolípidos/química , Ácidos Sulfónicos/química , Ácidos Sulfónicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA