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











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 12(1): 20535, 2022 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-36446811

RESUMEN

The regulated translocation of the glucose transporter, GLUT4, to the surface of adipocytes and muscle is a key action of insulin. This is underpinned by the delivery and fusion of GLUT4-containing vesicles with the plasma membrane. Recent studies have revealed that a further action of insulin is to mediate the dispersal of GLUT4 molecules away from the site of GLUT4 vesicle fusion with the plasma membrane. Although shown in adipocytes, whether insulin-stimulated dispersal occurs in other cells and/or is exhibited by other proteins remains a matter of debate. Here we show that insulin stimulates GLUT4 dispersal in the plasma membrane of adipocytes, induced pluripotent stem cell-derived cardiomyocytes and HeLa cells, suggesting that this phenomenon is specific to GLUT4 expressed in all cell types. By contrast, insulin-stimulated dispersal of TfR was not observed in HeLa cells, suggesting that the mechanism may be unique to GLUT4. Consistent with dispersal being an important physiological mechanism, we observed that insulin-stimulated GLUT4 dispersal is reduced under conditions of insulin resistance. Adipocytes of different sizes have been shown to exhibit distinct metabolic properties: larger adipocytes exhibit reduced insulin-stimulated glucose transport compared to smaller cells. Here we show that both GLUT4 delivery to the plasma membrane and GLUT4 dispersal are reduced in larger adipocytes, supporting the hypothesis that larger adipocytes are refractory to insulin challenge compared to their smaller counterparts, even within a supposedly homogeneous population of cells.


Asunto(s)
Adipocitos , Insulina , Humanos , Células HeLa , Tamaño de la Célula , Insulina/farmacología , Translocación Genética , Miocitos Cardíacos
2.
PeerJ ; 9: e11485, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34113491

RESUMEN

The global incidence, associated mortality rates and economic burden of diabetes are now such that it is considered one of the most pressing worldwide public health challenges. Considerable research is now devoted to better understanding the mechanisms underlying the onset and progression of this disease, with an ultimate aim of improving the array of available preventive and therapeutic interventions. One area of particular unmet clinical need is the significantly elevated rate of cardiomyopathy in diabetic patients, which in part contributes to cardiovascular disease being the primary cause of premature death in this population. This review will first consider the role of metabolism and more specifically the insulin sensitive glucose transporter GLUT4 in diabetic cardiac disease, before addressing how we may use exercise to intervene in order to beneficially impact key functional clinical outcomes.

3.
PLoS One ; 14(7): e0217885, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31344028

RESUMEN

Induced pluripotent stem cell derived cardiomyocytes (iPSC-CM) have the potential to transform regenerative cardiac medicine and the modelling of cardiac disease. This is of particular importance in the context of diabetic cardiomyopathy where diabetic individuals exhibit reduced cardiac diastolic contractile performance in the absence of vascular disease, significantly contributing towards high cardiovascular morbidity. In this study, the capacity of iPSC-CM to act as a novel cellular model of cardiomyocytes was assessed. The diabetic phenotype is characterised by insulin resistance, therefore there was a specific focus upon metabolic parameters. Despite expressing crucial insulin signalling intermediates and relevant trafficking proteins, it was identified that iPSC-CM do not exhibit insulin-stimulated glucose uptake. iPSC-CM are spontaneously contractile however contraction mediated uptake was not found to mask any insulin response. The fundamental limitation identified in these cells was a critical lack of expression of the insulin sensitive glucose transporter GLUT4. Using comparative immunoblot analysis and the GLUT-selective inhibitor BAY-876 to quantify expression of these transporters, we show that iPSC-CM express high levels of GLUT1 and low levels of GLUT4 compared to primary cardiomyocytes and cultured adipocytes. Interventions to overcome this limitation were unsuccessful. We suggest that the utility of iPSC-CMs to study cardiac metabolic disorders may be limited by their apparent foetal-like phenotype.


Asunto(s)
Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4/biosíntesis , Glucosa/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Transporte Biológico Activo/efectos de los fármacos , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Transportador de Glucosa de Tipo 4/antagonistas & inhibidores , Humanos , Células Madre Pluripotentes Inducidas/patología , Resistencia a la Insulina , Ratones , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/patología , Pirazoles/farmacología , Quinolinas/farmacología , Conejos
4.
Artículo en Inglés | MEDLINE | ID: mdl-31920989

RESUMEN

SNARE proteins are integral to intracellular vesicular trafficking, which in turn is the process underlying the regulated expression of substrate transporters such as the glucose transporter GLUT4 at the cell surface of insulin target tissues. Impaired insulin stimulated GLUT4 trafficking is associated with reduced cardiac function in many disease states, most notably diabetes. Despite this, our understanding of the expression and regulation of SNARE proteins in cardiac tissue and how these may change in diabetes is limited. Here we characterize the array of SNARE proteins expressed in cardiac tissue, and quantify the levels of expression of VAMP2, SNAP23, and Syntaxin4-key proteins involved in insulin-stimulated GLUT4 translocation. We examined SNARE protein levels in cardiac tissue from two rodent models of insulin resistance, db/db mice and high-fat fed mice, and show alterations in patterns of expression are evident. Such changes may have implications for cardiac function.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA