Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
2.
J Biol Chem ; 288(8): 5682-93, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23275337

ABSTRACT

Selective serotonin reuptake inhibitors (SSRIs) are antidepressants used for the treatment of mood and anxiety disorders. Here, we demonstrate that incubation (2 h) of murine islets or Min6 ß cell line with the SSRIs paroxetine, fluoxetine, or sertraline inhibited insulin-induced Tyr phosphorylation of insulin receptor substrate (IRS)-2 protein and the activation of its downstream targets Akt and the ribosomal protein S6 kinase-1 (S6K1). Inhibition was dose-dependent with half-maximal effects at ∼15-20 µM. It correlated with a rapid dephosphorylation and activation of the IRS kinase GSK3ß. Introduction of GSK3ß siRNAs eliminated the inhibitory effects of the SSRIs. Inhibition of IRS-2 action by 30 µM SSRI was associated with a marked inhibition of glucose-stimulated insulin secretion from murine and human pancreatic islets. Secretion induced by basic secretagogues (KCl and Arg) was not affected by these drugs. Prolonged treatment (16 h) of Min6 cells with sertraline resulted in the induction of inducible nitric oxide synthase; activation of endoplasmic reticulum stress, and the initiation of the unfolded protein response, manifested by enhanced transcription of ATF4 and C/EBP homologous protein. This triggered an apoptotic process, manifested by enhanced caspase 3/7 activity, which resulted in ß cell death. These findings implicate SSRIs as inhibitors of IRS protein function and insulin action through the activation of GSK3ß. They further suggest that SSRIs inhibit insulin secretion; induce the unfolded protein response; activate an apoptotic process, and trigger ß cell death. Given that SSRIs promote insulin resistance while inhibiting insulin secretion, these drugs might accelerate the transition from an insulin-resistant state to overt diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Apoptosis , Cell Death , Cell Line , Diabetes Mellitus/chemically induced , Diabetes Mellitus/metabolism , Fluoxetine/pharmacology , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Inbred C57BL , Paroxetine/pharmacology , Sertraline/pharmacology , Signal Transduction , Unfolded Protein Response
3.
Nanotoxicology ; 6: 804-12, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22007682

ABSTRACT

Upon exposure, TiO(2) nanoparticles (NPs) have been recovered in internal organs such as the liver, and are proposed to cause cellular/organ dysfunction, particularly in the liver and lungs. We hypothesized that despite being considered "inert" as bulk material, TiO(2) NPs may impair insulin responses in liver-derived cells, either indirectly by inflammatory activation of macrophages, and/or by directly interfering with insulin signaling. Using qRT-PCR and conditioned medium (CM) approaches, we show that exposure to TiO(2) NPs activates macrophages' expression of TNF-α, IL-6, IL-8, IL-1α and IL-1ß and the resulting CM induces insulin resistance in Fao cells. Furthermore, direct exposure of Fao cells to TiO(2) results in activation of the stress kinases JNK and p38MAP kinase, and in induction of insulin resistance at the signaling and metabolic levels. Collectively, our findings provide a proof-of-concept for the ability of man-made NPs to induce insulin resistance in liver-derived cells, an endocrine abnormality underlying some of the most common human diseases.


Subject(s)
Insulin Resistance , Liver/drug effects , Macrophage Activation , Metal Nanoparticles/toxicity , Titanium/toxicity , Animals , Blotting, Western , Cell Line, Tumor , Culture Media, Conditioned , Glycogen/biosynthesis , Insulin/metabolism , Rats , Real-Time Polymerase Chain Reaction , Signal Transduction
4.
Mol Endocrinol ; 24(11): 2179-92, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20843941

ABSTRACT

Insulin receptor substrate-1 (IRS-1) plays a pivotal role in insulin signaling, therefore its degradation is exquisitely regulated. Here, we show that insulin-stimulated degradation of IRS-1 requires the presence of a highly conserved Ser/Thr-rich domain that we named domain involved in degradation of IRS-1 (DIDI). DIDI (amino acids 386-430 of IRS-1) was identified by comparing the intracellular degradation rate of several truncated forms of IRS-1 transfected into CHO cells. The isolated DIDI domain underwent insulin-stimulated Ser/Thr phosphorylation, suggesting that it serves as a target for IRS-1 kinases. The effects of deletion of DIDI were studied in Fao rat hepatoma and in CHO cells expressing Myc-IRS-1(WT) or Myc-IRS-1(Δ386-430). Deletion of DIDI maintained the ability of IRS-1(Δ386-434) to undergo ubiquitination while rendering it insensitive to insulin-induced proteasomal degradation, which affected IRS-1(WT) (80% at 8 h). Consequently, IRS-1(Δ386-434) mediated insulin signaling (activation of Akt and glycogen synthesis) better than IRS-1(WT). IRS-1(Δ386-434) exhibited a significant greater preference for nuclear localization, compared with IRS-1(WT). Higher nuclear localization was also observed when cells expressing IRS-1(WT) were incubated with the proteasome inhibitor MG-132. The sequence of DIDI is conserved more than 93% across species, from fish to mammals, as opposed to approximately 40% homology of the entire IRS-1. These findings implicate DIDI as a novel, highly conserved domain of IRS-1, which mediates its cellular localization, rate of degradation, and biological activity, with a direct impact on insulin signal transduction.


Subject(s)
Insulin Receptor Substrate Proteins/chemistry , Insulin Receptor Substrate Proteins/metabolism , Insulin/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational/drug effects , Animals , Apoptosis/drug effects , CHO Cells , Cricetinae , Cricetulus , Cytoprotection/drug effects , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Structure, Tertiary , Protein Transport/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Rats , Sequence Deletion , Signal Transduction/drug effects , Structure-Activity Relationship , Ubiquitination/drug effects
5.
Diabetes ; 59(9): 2188-97, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20547979

ABSTRACT

OBJECTIVE: Cellular stress and proinflammatory cytokines induce phosphorylation of insulin receptor substrate (IRS) proteins at Ser sites that inhibit insulin and IGF-1 signaling. Here, we examined the role of Ser phosphorylation of IRS-2 in mediating the inhibitory effects of proinflammatory cytokines and cellular stress on beta-cell function. RESEARCH DESIGN AND METHODS: Five potential inhibitory Ser sites located proximally to the P-Tyr binding domain of IRS-2 were mutated to Ala. These IRS-2 mutants, denoted IRS-2(5A), and their wild-type controls (IRS-2(WT)) were introduced into adenoviral constructs that were infected into Min6 cells or into cultured murine islets. RESULTS: When expressed in cultured mouse islets, IRS-2(5A) was better than IRS-2(WT) in protecting beta-cells from apoptosis induced by a combination of IL-1beta, IFN-gamma, TNF-alpha, and Fas ligand. Cytokine-treated islets expressing IRS2(5A) secreted significantly more insulin in response to glucose than did islets expressing IRS-2(WT). This could be attributed to the higher transcription of Pdx1 in cytokine-treated islets that expressed IRS-2(5A). Accordingly, transplantation of 200 islets expressing IRS2(5A) into STZ-induced diabetic mice restored their ability to respond to a glucose load similar to naïve mice. In contrast, mice transplanted with islets expressing IRS2(WT) maintained sustained hyperglycemia 3 days after transplantation. CONCLUSIONS: Elimination of a physiological negative feedback control mechanism along the insulin-signaling pathway that involves Ser/Thr phosphorylation of IRS-2 affords protection against the adverse effects of proinflammatory cytokines and improves beta-cell function under stress. Genetic approaches that promote IRS2(5A) expression in pancreatic beta-cells, therefore, could be considered a rational treatment against beta-cell failure after islet transplantation.


Subject(s)
Insulin Receptor Substrate Proteins/physiology , Insulin-Secreting Cells/physiology , Insulin/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blood Glucose/metabolism , CHO Cells , Caspases/metabolism , Cricetinae , Cricetulus , Cytokines/pharmacology , Diabetes Mellitus, Experimental/surgery , Glucose/pharmacology , Glucose Tolerance Test , Homeodomain Proteins/genetics , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans Transplantation/physiology , Male , Mice , Mice, Inbred C57BL , Serine/physiology , Signal Transduction , Trans-Activators/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...