Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Endocrinol ; 258(3)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37578842

RESUMO

Bromodomain-containing protein 7 (BRD7) has emerged as a player in the regulation of glucose homeostasis. Hepatic BRD7 levels are decreased in obese mice, and the reinstatement of hepatic BRD7 in obese mice has been shown to establish euglycemia and improve glucose homeostasis. Of note, the upregulation of hepatic BRD7 levels activates the AKT cascade in response to insulin without enhancing the sensitivity of the insulin receptor (InsR)-insulin receptor substrate (IRS) axis. In this report, we provide evidence for the existence of an alternative insulin signaling pathway that operates independently of IRS proteins and demonstrate the involvement of BRD7 in this pathway. To investigate the involvement of BRD7 as a downstream component of InsR, we utilized liver-specific InsR knockout mice. Additionally, we employed liver-specific IRS1/2 knockout mice to examine the requirement of IRS1/2 for the action of BRD7. Our investigation of glucose metabolism parameters and insulin signaling unveiled the significance of InsR activation in mediating BRD7's effect on glucose homeostasis in the liver. Moreover, we identified an interaction between BRD7 and InsR. Notably, our findings indicate that IRS1/2 is not necessary for BRD7's regulation of glucose metabolism, particularly in the context of obesity. The upregulation of hepatic BRD7 significantly reduces blood glucose levels and restores glucose homeostasis in high-fat diet-challenged liver-specific IRS1/2 knockout mice. These findings highlight the presence of an alternative insulin signaling pathway that operates independently of IRS1/2 and offer novel insights into the mechanisms of a previously unknown insulin signaling in obesity.


Assuntos
Resistência à Insulina , Receptor de Insulina , Animais , Camundongos , Glucose/metabolismo , Homeostase/genética , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Fígado/metabolismo , Camundongos Knockout , Camundongos Obesos , Obesidade/genética , Obesidade/metabolismo , Receptor de Insulina/metabolismo , Fatores de Transcrição/metabolismo
2.
Front Endocrinol (Lausanne) ; 14: 1152579, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38317714

RESUMO

The regulatory subunit of phosphatidylinositol 3-kinase (PI3K), known as p85, is a critical component in the insulin signaling pathway. Extensive research has shed light on the diverse roles played by the two isoforms of p85, namely p85α and p85ß. The gene pik3r1 encodes p85α and its variants, p55α and p50α, while pik3r2 encodes p85ß. These isoforms exhibit various activities depending on tissue types, nutrient availability, and cellular stoichiometry. Whole-body or liver-specific deletion of pik3r1 have shown to display increased insulin sensitivity and improved glucose homeostasis; however, skeletal muscle-specific deletion of p85α does not exhibit any significant effects on glucose homeostasis. On the other hand, whole-body deletion of pik3r2 shows improved insulin sensitivity with no significant impact on glucose tolerance. Meanwhile, liver-specific double knockout of pik3r1 and pik3r2 leads to reduced insulin sensitivity and glucose tolerance. In the context of obesity, upregulation of hepatic p85α or p85ß has been shown to improve glucose homeostasis. However, hepatic overexpression of p85α in the absence of p50α and p55α results in increased insulin resistance in obese mice. p85α and p85ß have distinctive roles in cancer development. p85α acts as a tumor suppressor, but p85ß promotes tumor progression. In the immune system, p85α facilitates B cell development, while p85ß regulates T cell differentiation and maturation. This review provides a comprehensive overview of the distinct functions attributed to p85α and p85ß, highlighting their significance in various physiological processes, including insulin signaling, cancer development, and immune system regulation.


Assuntos
Hiperinsulinismo , Resistência à Insulina , Neoplasias , Camundongos , Animais , Resistência à Insulina/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Camundongos Knockout , Insulina/metabolismo , Glucose , Isoformas de Proteínas
3.
J Mol Cell Biol ; 13(12): 889-901, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-34751372

RESUMO

Bromodomain-containing protein 7 (BRD7) has been shown to interact with the regulatory subunit of phosphatidylinositol 3-kinase (PI3K), p85, in the insulin signaling pathway. Here, we show that upregulation of hepatic BRD7 improves glucose homeostasis even in the absence of either p85 isoform, p85α or p85ß. However, BRD7 leads to differential activation of downstream effector proteins in the insulin signaling pathway depending on which isoform of p85 is present. In the presence of only p85α, BRD7 overexpression increases phosphorylation of insulin receptor (IR) upon insulin stimulation, without increasing the recruitment of p85 to IR substrate. Overexpression of BRD7 also increases activation of Akt in response to insulin, but does not affect basal phosphorylation levels of Akt. Meanwhile, the phosphorylation of glycogen synthase kinase 3ß (GSK3ß) is increased by overexpression of BRD7. On the other hand, in the presence of only p85ß, BRD7 overexpression does not affect phosphorylation levels of IR, and Akt phosphorylation is not affected by insulin stimulation following BRD7 upregulation. However, BRD7 overexpression leads to increased basal phosphorylation levels of Akt and GSK3ß. These data demonstrate that BRD7's action on glucose homeostasis does not require the presence of both p85 isoforms, and p85α and p85ß have unique roles in insulin signaling in the liver.


Assuntos
Insulina , Fosfatidilinositol 3-Quinase , Glucose/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo
4.
Int J Mol Sci ; 21(19)2020 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-32992509

RESUMO

Bromodomain is a conserved structural module found in many chromatin-associated proteins. Bromodomain-containing protein 7 (BRD7) is a member of the bromodomain-containing protein family, and was discovered two decades ago as a protein that is downregulated in nasopharyngeal carcinoma. Since then, BRD7 has been implicated in a variety of cellular processes, including chromatin remodeling, transcriptional regulation, and cell cycle progression. Decreased BRD7 activity underlies the pathophysiological properties of various diseases in different organs. BRD7 plays an important role in the pathogenesis of many cancers and, more recently, its roles in the regulation of metabolism and obesity have also been highlighted. Here, we review the involvement of BRD7 in a variety of pathophysiological conditions, with a focus on glucose homeostasis, obesity, and cancer.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Neoplasias/metabolismo , Obesidade/metabolismo , Animais , Glicemia/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo
5.
Sci Rep ; 9(1): 5327, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30926848

RESUMO

Obesity is a debilitating disease that has become a global epidemic. Although progress is being made, the underlying molecular mechanism by which obesity develops still remains elusive. Recently, we reported that the expression levels of bromodomain-containing protein 7 (BRD7) are significantly reduced in the liver of obese mice. However, it is not clear whether decreased levels of hepatic BRD7 are directly associated with the development of obesity and disturbance in glucose homeostasis. Here, using heterozygous BRD7 knockout and liver-specific BRD7 knockout mouse models, we report that reduced BRD7 levels lead to increased weight gain with little effect on glucose metabolism. On the other hand, upregulating BRD7 in the liver starting at an early age protects mice from gaining excessive weight and developing glucose intolerance and insulin resistance when challenged with a high-fat diet.


Assuntos
Proteínas Cromossômicas não Histona/deficiência , Predisposição Genética para Doença , Hiperglicemia/sangue , Hiperglicemia/genética , Obesidade/sangue , Obesidade/genética , Animais , Biomarcadores , Glicemia , Peso Corporal , Modelos Animais de Doenças , Estudos de Associação Genética , Genótipo , Homeostase , Hiperglicemia/diagnóstico , Insulina/metabolismo , Resistência à Insulina , Fígado/metabolismo , Camundongos , Camundongos Knockout , Obesidade/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...