Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
FASEB J ; 34(9): 12521-12532, 2020 09.
Article in English | MEDLINE | ID: mdl-32744782

ABSTRACT

Class Ia phosphoinositide 3-kinases (PI3K) are critical mediators of insulin and growth factor action. We have demonstrated that the p85α regulatory subunit of PI3K modulates the unfolded protein response (UPR) by interacting with and regulating the nuclear translocation of XBP-1s, a transcription factor essential for the UPR. We now show that PI3K activity is required for full activation of the UPR. Pharmacological inhibition of PI3K in cells blunts the ER stress-dependent phosphorylation of IRE1α and PERK, decreases induction of ATF4, CHOP, and XBP-1 and upregulates UPR target genes. Cells expressing a human p85α mutant (R649W) previously shown to inhibit PI3K, exhibit decreased activation of IRE1α and PERK and reduced induction of CHOP and ATF4. Pharmacological inhibition of PI3K, overexpression of a mutant of p85α that lacks the ability to interact with the p110α catalytic subunit (∆p85α) or expression of mutant p85α (R649W) in vivo, decreased UPR-dependent induction of ER stress response genes. Acute tunicamycin treatment of R649W+/- mice revealed reduced induction of UPR target genes in adipose tissue, whereas chronic tunicamycin exposure caused sustained increases in UPR target genes in adipose tissue. Finally, R649W+/- cells exhibited a dramatic resistance to ER stress-dependent apoptosis. These data suggest that PI3K pathway dysfunction causes ER stress that may drive the pathogenesis of several diseases including Type 2 diabetes and various cancers.


Subject(s)
Adipose Tissue/metabolism , Apoptosis , Class Ia Phosphatidylinositol 3-Kinase/physiology , Endoplasmic Reticulum Stress , Unfolded Protein Response , Adipose Tissue/cytology , Animals , Cell Line , Male , Mice , Mice, Inbred C57BL , X-Box Binding Protein 1/metabolism
2.
Diabetes ; 67(7): 1297-1309, 2018 07.
Article in English | MEDLINE | ID: mdl-29724723

ABSTRACT

Phosphatidylinositol 3-kinase (PI3K) plays a central role in insulin signaling, glucose metabolism, cell growth, cell development, and apoptosis. A heterozygous missense mutation (R649W) in the p85α regulatory subunit gene of PI3K (PIK3R1) has been identified in patients with SHORT (Short stature, Hyperextensibility/Hernia, Ocular depression, Rieger anomaly, and Teething delay) syndrome, a disorder characterized by postnatal growth retardation, insulin resistance, and partial lipodystrophy. Knock-in mice with the same heterozygous mutation mirror the human phenotype. In this study, we show that Pik3r1 R649W knock-in mice fed a high-fat diet (HFD) have reduced weight gain and adipose accumulation. This is accompanied by reduced expression of several genes involved in lipid metabolism. Interestingly, despite the lower level of adiposity, the HFD knock-in mice are more hyperglycemic and more insulin-resistant than HFD-fed control mice. Likewise, when crossed with genetically obese ob/ob mice, the ob/ob mice carrying the heterozygous R649W mutation were protected from obesity and hepatic steatosis but developed a severe diabetic state. Together, our data demonstrate a central role of PI3K in development of obesity and fatty liver disease, separating these effects from the role of PI3K in insulin resistance and the resultant hyperglycemia.


Subject(s)
Diabetes Mellitus/genetics , Fatty Liver/genetics , Growth Disorders/genetics , Hypercalcemia/genetics , Metabolic Diseases/genetics , Nephrocalcinosis/genetics , Obesity/genetics , Phosphatidylinositol 3-Kinases/genetics , Amino Acid Substitution , Animals , Arginine/genetics , Class Ia Phosphatidylinositol 3-Kinase , Diabetes Mellitus/pathology , Fatty Liver/pathology , Female , Gene Knock-In Techniques , Genes, Dominant , Genetic Predisposition to Disease , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Mutation, Missense , Obesity/pathology , Tryptophan/genetics
3.
Invest Ophthalmol Vis Sci ; 58(7): 3100-3106, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28632845

ABSTRACT

Purpose: To determine the ocular consequences of a dominant-negative mutation in the p85α subunit of phosphatidylinositol 3-kinase (PIK3R1) using a knock-in mouse model of SHORT syndrome, a syndrome associated with short stature, lipodystrophy, diabetes, and Rieger anomaly in humans. Methods: We investigated knock-in mice heterozygous for the SHORT syndrome mutation changing arginine 649 to tryptophan in p85α (PIK3R1) using physical examination, optical coherence tomography (OCT), tonometry, and histopathologic sections from paraffin-embedded eyes, and compared the findings to similar investigations in two human subjects with SHORT syndrome heterozygous for the same mutation. Results: While overall eye development was normal with clear cornea and lens, normal anterior chamber volume, normal intraocular pressure, and no changes in the retinal structure, OCT images of the knock-in mouse eyes revealed a significant decrease in thickness and width of the iris resulting in increased pupil area and irregularity of shape. Both human subjects had Rieger anomaly with similar defects including thin irides and irregular pupils, as well as a prominent ring of Schwalbe, goniosynechiae, early cataract formation, and glaucoma. Although the two subjects had had diabetes for more than 30 years, there were no signs of diabetic retinopathy. Conclusions: A dominant-negative mutation in the p85α regulatory subunit of PI3K affects development of the iris, and contributes to changes consistent with anterior segment dysgenesis in both humans and mice.


Subject(s)
Anterior Eye Segment/abnormalities , DNA/genetics , Eye Abnormalities/genetics , Iris/abnormalities , Mutation , Phosphatidylinositol 3-Kinases/genetics , Animals , Anterior Eye Segment/diagnostic imaging , Anterior Eye Segment/enzymology , Class Ia Phosphatidylinositol 3-Kinase , DNA Mutational Analysis , Disease Models, Animal , Eye Abnormalities/diagnosis , Eye Abnormalities/enzymology , Eye Diseases, Hereditary , Female , Humans , Intraocular Pressure , Iris/diagnostic imaging , Male , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Tomography, Optical Coherence
5.
Nat Commun ; 8: 14892, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28345670

ABSTRACT

Despite a high degree of homology, insulin receptor (IR) and IGF-1 receptor (IGF1R) mediate distinct cellular and physiological functions. Here, we demonstrate how domain differences between IR and IGF1R contribute to the distinct functions of these receptors using chimeric and site-mutated receptors. Receptors with the intracellular domain of IGF1R show increased activation of Shc and Gab-1 and more potent regulation of genes involved in proliferation, corresponding to their higher mitogenic activity. Conversely, receptors with the intracellular domain of IR display higher IRS-1 phosphorylation, stronger regulation of genes in metabolic pathways and more dramatic glycolytic responses to hormonal stimulation. Strikingly, replacement of leucine973 in the juxtamembrane region of IR to phenylalanine, which is present in IGF1R, mimics many of these signalling and gene expression responses. Overall, we show that the distinct activities of the closely related IR and IGF1R are mediated by their intracellular juxtamembrane region and substrate binding to this region.


Subject(s)
Gene Expression , Insulin/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Amino Acid Substitution , Animals , Binding Sites , Cell Proliferation , Gene Expression Regulation , Glycolysis , Leucine/chemistry , Mice , Mutagenesis, Site-Directed , Phenylalanine/chemistry , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/genetics
6.
Nature ; 542(7642): 450-455, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28199304

ABSTRACT

Adipose tissue is a major site of energy storage and has a role in the regulation of metabolism through the release of adipokines. Here we show that mice with an adipose-tissue-specific knockout of the microRNA (miRNA)-processing enzyme Dicer (ADicerKO), as well as humans with lipodystrophy, exhibit a substantial decrease in levels of circulating exosomal miRNAs. Transplantation of both white and brown adipose tissue-brown especially-into ADicerKO mice restores the level of numerous circulating miRNAs that are associated with an improvement in glucose tolerance and a reduction in hepatic Fgf21 mRNA and circulating FGF21. This gene regulation can be mimicked by the administration of normal, but not ADicerKO, serum exosomes. Expression of a human-specific miRNA in the brown adipose tissue of one mouse in vivo can also regulate its 3' UTR reporter in the liver of another mouse through serum exosomal transfer. Thus, adipose tissue constitutes an important source of circulating exosomal miRNAs, which can regulate gene expression in distant tissues and thereby serve as a previously undescribed form of adipokine.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , MicroRNAs/blood , MicroRNAs/metabolism , Paracrine Communication , 3' Untranslated Regions/genetics , Adipokines/metabolism , Adipose Tissue/transplantation , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/transplantation , Adipose Tissue, White/metabolism , Adipose Tissue, White/transplantation , Animals , Exosomes/genetics , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Genes, Reporter/genetics , Glucose Tolerance Test , Liver/metabolism , Male , Mice , MicroRNAs/genetics , Models, Biological , Organ Specificity/genetics , RNA, Messenger/genetics , Ribonuclease III/deficiency , Ribonuclease III/genetics , Transcription, Genetic
7.
Cell Metab ; 25(2): 448-462, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28065828

ABSTRACT

Insulin and IGF1 signaling are important for adipose tissue development and function; however, their role in mature adipocytes is unclear. Mice with a tamoxifen-inducible knockout of insulin and/or IGF1 receptors (IR/IGF1R) demonstrate a rapid loss of white and brown fat due to increased lipolysis and adipocyte apoptosis. This results in insulin resistance, glucose intolerance, hepatosteatosis, islet hyperplasia with hyperinsulinemia, and cold intolerance. This phenotype, however, resolves over 10-30 days due to a proliferation of preadipocytes and rapid regeneration of both brown and white adipocytes as identified by mTmG lineage tracing. This cycle can be repeated with a second round of receptor inactivation. Leptin administration prior to tamoxifen treatment blocks development of the metabolic syndrome without affecting adipocyte loss or regeneration. Thus, IR is critical in adipocyte maintenance, and this loss of adipose tissue stimulates regeneration of brown/white fat and reversal of metabolic syndrome associated with fat loss.


Subject(s)
Adipocytes/metabolism , Gene Deletion , Metabolic Syndrome/metabolism , Receptor, Insulin/metabolism , Adipocytes/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fatty Liver/complications , Fatty Liver/metabolism , Fatty Liver/pathology , Glucose Intolerance/complications , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Hyperglycemia/complications , Hyperglycemia/metabolism , Hyperglycemia/pathology , Insulin/metabolism , Insulin Resistance , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Leptin/pharmacology , Lipodystrophy/complications , Lipodystrophy/metabolism , Lipodystrophy/pathology , Metabolic Syndrome/complications , Mice , Organ Specificity/drug effects , Receptor, IGF Type 1/metabolism , Regeneration/drug effects , Tamoxifen/pharmacology
9.
J Clin Invest ; 126(4): 1401-12, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26974159

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) signaling pathway is central to the action of insulin and many growth factors. Heterozygous mutations in the gene encoding the p85α regulatory subunit of PI3K (PIK3R1) have been identified in patients with SHORT syndrome - a disorder characterized by short stature, partial lipodystrophy, and insulin resistance. Here, we evaluated whether SHORT syndrome-associated PIK3R1 mutations account for the pathophysiology that underlies the abnormalities by generating knockin mice that are heterozygous for the Pik3r1Arg649Trp mutation, which is homologous to the mutation found in the majority of affected individuals. Similar to the patients, mutant mice exhibited a reduction in body weight and length, partial lipodystrophy, and systemic insulin resistance. These derangements were associated with a reduced capacity of insulin and other growth factors to activate PI3K in liver, muscle, and fat; marked insulin resistance in liver and fat of mutation-harboring animals; and insulin resistance in vitro in cells derived from these mice. In addition, mutant mice displayed defective insulin secretion and GLP-1 action on islets in vivo and in vitro. These data demonstrate the ability of this heterozygous mutation to alter PI3K activity in vivo and the central role of PI3K in insulin/growth factor action, adipocyte function, and glucose metabolism.


Subject(s)
Growth Hormone , Insulin Resistance/genetics , Liver/enzymology , Mutation, Missense , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Substitution , Animals , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Lipodystrophy/enzymology , Lipodystrophy/genetics , Lipodystrophy/pathology , Liver/pathology , Mice , Mice, Mutant Strains
10.
Proc Natl Acad Sci U S A ; 111(3): 1192-7, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395790

ABSTRACT

In insulin resistant states such as type 2 diabetes, there is a high demand on the ß-cell to synthesize and secrete insulin, which challenges the ability of the endoplasmic reticulum (ER) to synthesize and fold nascent proteins. This creates a state of ER stress that triggers a coordinated program referred to as the unfolded protein response (UPR) that attempts to restore ER homeostasis. We identified a role for the p85α regulatory subunit of PI3K to modulate the UPR by promoting the nuclear localization of X-box binding protein 1, a transcription factor central to the UPR. In the present study we demonstrate that reducing p85α expression in ß-cells can markedly delay the onset and severity of the diabetic phenotype observed in Akita(+/-) mice, which express a mutant insulin molecule. This is due to a decrease in activation of ER stress-dependent apoptotic pathways and a preservation of ß-cell mass and function. These data demonstrate that modulation of p85α can protect pancreatic ß-cells from ER stress, pointing to a potentially therapeutic target in diabetic states.


Subject(s)
Apoptosis , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Insulin-Secreting Cells/cytology , Alleles , Animals , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation, Enzymologic , Genotype , Glucose/metabolism , Hyperglycemia/metabolism , Insulin/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Oxidative Stress , Pancreas/physiology , Phenotype , Protein Denaturation , Protein Folding , Time Factors
11.
Proc Natl Acad Sci U S A ; 110(28): 11409-14, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23798383

ABSTRACT

Visceral and s.c. fat exhibit different intrinsic properties, including rates of lipolysis, and are associated with differential risk for the development of type 2 diabetes. These effects are in part related to cell autonomous differences in gene expression. In the present study, we show that expression of Shox2 (Short stature homeobox 2) is higher in s.c. than visceral fat in both rodents and humans and that levels are further increased in humans with visceral obesity. Fat-specific disruption of Shox2 in male mice results in protection from high fat diet-induced obesity, with a preferential loss of s.c. fat. The reduced adipocyte size is secondary to a twofold increase in the expression of ß3 adrenergic receptor (Adrb3) at both the mRNA and protein level and a parallel increase in lipolytic rate. These effects are mimicked by knockdown of Shox2 in C3H10T1/2 cells. Conversely, overexpression of Shox2 leads to a repression of Adrb3 expression and decrease lipolytic rate. Shox2 does not affect differentiation but directly interacts with CCAAT/enhancer binding protein alpha and attenuates its transcriptional activity of the Adrb3 promoter. Thus, Shox2 can regulate the expression of Adrb3 and control the rate of lipolysis and, in this way, exerts control of the phenotypic differences between visceral and s.c. adipocytes.


Subject(s)
Adipocytes/cytology , Homeodomain Proteins/physiology , Animals , Diet , Homeodomain Proteins/genetics , Insulin Resistance , Lipolysis , Mice , Mice, Knockout , Obesity/etiology , Obesity/genetics
12.
J Biol Chem ; 287(15): 12016-26, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22337886

ABSTRACT

Disturbed Wnt signaling has been implicated in numerous diseases, including type 2 diabetes and the metabolic syndrome. In the present study, we have investigated cross-talk between insulin and Wnt signaling pathways using preadipocytes with and without knockdown of the Wnt co-receptors LRP5 and LRP6 and with and without knock-out of insulin and IGF-1 receptors. We find that Wnt stimulation leads to phosphorylation of insulin signaling key mediators, including Akt, GSK3ß, and ERK1/2, although with a lower fold stimulation and slower time course than observed for insulin. These Wnt effects are insulin/IGF-1 receptor-dependent and are lost in insulin/IGF-1 receptor double knock-out cells. Conversely, in LRP5 knockdown preadipocytes, insulin-induced phosphorylation of IRS1, Akt, GSK3ß, and ERK1/2 is highly reduced. This effect is specific to insulin, as compared with IGF-1, stimulation and appears to be due to an inducible interaction between LRP5 and the insulin receptor as demonstrated by co-immunoprecipitation. These data demonstrate that Wnt and insulin signaling pathways exhibit cross-talk at multiple levels. Wnt induces phosphorylation of Akt, ERK1/2, and GSK3ß, and this is dependent on insulin/IGF-1 receptors. Insulin signaling also involves the Wnt co-receptor LRP5, which has a positive effect on insulin signaling. Thus, altered Wnt and LRP5 activity can serve as modifiers of insulin action and insulin resistance in the pathophysiology of diabetes and metabolic syndrome.


Subject(s)
Adipocytes/metabolism , Insulin/physiology , Low Density Lipoprotein Receptor-Related Protein-5/physiology , Receptor Cross-Talk , Wnt Signaling Pathway , 3T3-L1 Cells , Animals , Gene Expression Regulation , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Immunoprecipitation , Insulin/metabolism , Kinetics , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , MAP Kinase Signaling System , Mice , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Wnt3A Protein/physiology , beta Catenin/metabolism
13.
Methods Enzymol ; 490: 147-58, 2011.
Article in English | MEDLINE | ID: mdl-21266249

ABSTRACT

The endoplasmic reticulum (ER) consists of an interconnected, membranous network that is the major site for the synthesis and folding of integral membrane and secretory proteins. Within the ER lumen, protein folding is facilitated by molecular chaperones and a variety of enzymes that ensure that polypeptides obtain their appropriate, tertiary conformation (Dobson, C. M. (2004). Principles of protein folding, misfolding and aggregation. Semin. Cell Dev. Biol. 15, 3-16; Ni, M., and Lee, A. S. (2007). ER chaperones in mammalian development and human diseases. FEBS Lett. 581, 3641-3651.). Physiological conditions that increase protein synthesis or stimuli that disturb the processes by which proteins obtain their native conformation, create an imbalance between the protein-folding demand and capacity of the ER. This results in the accumulation of unfolded or improperly folded proteins in the ER lumen and a state of ER stress. The cellular response, referred to as the unfolded protein response (UPR), results in activation of three linked signal transduction pathways: PKR-like kinase (PERK), inositol requiring 1 α (IRE1α), and activating transcription factor 6α (ATF6α) (Ron, D., and Walter, P. (2007). Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell. Biol. 8, 519-529; Schroder, M., and Kaufman, R. (2005). ER stress and the unfolded protein response. Mutat. Res./Fundam. Mol. Mech. Mutagen. 569, 29-63.). Collectively, the combined actions of these signaling cascades serve to reduce ER stress through attenuation of translation to reduce protein synthesis and through activation of transcriptional programs that ultimately serve to increase ER protein-folding capacity. Recently, we and Park et al. have characterized a novel function for the p85α and p85ß subunits as modulators of the UPR by virtue of their ability to facilitate the nuclear entry of XBP-1s following induction of ER stress (Park, S. W., Zhou, Y., Lee, J., Lu, A., Sun, C., Chung, J., Ueki, K., and Ozcan, U. (2010). Regulatory subunits of PI3K, p85alpha and p85 beta, interact with XBP1 and increase its nuclear translocation. Nat. Med. 16, 429-437; Winnay, J. N., Boucher, J., Mori, M. A., Ueki, K., and Kahn, C. R. (2010). A regulatory subunit of phosphoinositide 3-kinase increases the nuclear accumulation of X-box-binding protein-1 to modulate the unfolded protein response. Nat. Med. 16, 438-445.). This chapter describes the recently elucidated role for the regulatory subunits of PI 3-kinase as modulators of the UPR and provides methods to measure UPR pathway activation.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Protein Subunits/metabolism , Unfolded Protein Response/physiology , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/physiology , Endoribonucleases/genetics , Endoribonucleases/metabolism , Humans , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Regulatory Factor X Transcription Factors , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , X-Box Binding Protein 1 , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
14.
Nat Med ; 16(4): 438-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20348923

ABSTRACT

Class Ia phosphoinositide 3-kinase (PI3K), an essential mediator of the metabolic actions of insulin, is composed of a catalytic (p110alpha or p110beta) and regulatory (p85alphaalpha, p85betaalpha or p55alpha) subunit. Here we show that p85alphaalpha interacts with X-box-binding protein-1 (XBP-1), a transcriptional mediator of the unfolded protein response (UPR), in an endoplasmic reticulum (ER) stress-dependent manner. Cell lines with knockout or knockdown of p85alphaalpha show marked alterations in the UPR, including reduced ER stress-dependent accumulation of nuclear XBP-1, decreased induction of UPR target genes and increased rates of apoptosis. This is associated with a decreased activation of inositol-requiring protein-1alpha (IRE1alpha) and activating transcription factor-6alphaalpha (ATF6alpha). Mice with deletion of p85alpha in liver (L-Pik3r1(-/-)) show a similar attenuated UPR after tunicamycin administration, leading to an increased inflammatory response. Thus, p85alphaalpha forms a previously unrecognized link between the PI3K pathway, which is central to insulin action, and the regulation of the cellular response to ER stress, a state that when unresolved leads to insulin resistance.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Transcription Factors/physiology , Unfolded Protein Response/physiology , Activating Transcription Factor 6 , Animals , Apoptosis/physiology , Cell Line , Cell Nucleus/physiology , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Endoribonucleases/physiology , Gene Knockdown Techniques , Insulin/physiology , Liver/metabolism , Liver/physiology , Membrane Proteins/physiology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Regulatory Factor X Transcription Factors , Stress, Physiological/physiology , Trans-Activators/physiology , Transcription Factors/metabolism , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects , X-Box Binding Protein 1
15.
Mol Cell Biol ; 29(11): 3219-28, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19273608

ABSTRACT

Insulin and insulin-like growth factor I (IGF-I) are ubiquitous hormones that regulate growth and metabolism of most mammalian cells, including pancreatic beta-cells. In addition to being an insulin secretagogue, glucose regulates proliferation and survival of beta-cells. However, it is unclear whether the latter effects of glucose occur secondary to autocrine activation of insulin signaling proteins by secreted insulin. To examine this possibility we studied the effects of exogenous glucose or insulin in beta-cell lines completely lacking either insulin receptors (betaIRKO) or insulin receptor substrate 2 (betaIRS2KO). Exogenous addition of either insulin or glucose activated proteins in the insulin signaling pathway in control beta-cell lines with the effects of insulin peaking earlier than glucose. Insulin stimulation of betaIRKO and betaIRS2KO cells led to blunted activation of phosphatidylinositol 3-kinase and Akt kinase, while surprisingly, glucose failed to activate either kinase but phosphorylated extracellular signal-regulated kinase. Control beta-cells exhibited low expression of IGF-1 receptors compared to compensatory upregulation in betaIRKO cells. The signaling data support the slow growth and reduced DNA and protein synthesis in betaIRKO and betaIRS2KO cells in response to glucose stimulation. Together, these studies provide compelling evidence that the growth and survival effects of glucose on beta-cells require activation of proteins in the insulin signaling pathway.


Subject(s)
Glucose/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Receptor, Insulin/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucose/metabolism , Insulin/pharmacology , Insulin-Secreting Cells/enzymology , Mice , Mice, Knockout , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects
16.
Nature ; 454(7207): 1000-4, 2008 Aug 21.
Article in English | MEDLINE | ID: mdl-18719589

ABSTRACT

Adipose tissue is central to the regulation of energy balance. Two functionally different types of fat are present in mammals: white adipose tissue, the primary site of triglyceride storage, and brown adipose tissue, which is specialized in energy expenditure and can counteract obesity. Factors that specify the developmental fate and function of white and brown adipose tissue remain poorly understood. Here we demonstrate that whereas some members of the family of bone morphogenetic proteins (BMPs) support white adipocyte differentiation, BMP7 singularly promotes differentiation of brown preadipocytes even in the absence of the normally required hormonal induction cocktail. BMP7 activates a full program of brown adipogenesis including induction of early regulators of brown fat fate PRDM16 (PR-domain-containing 16; ref. 4) and PGC-1alpha (peroxisome proliferator-activated receptor-gamma (PPARgamma) coactivator-1alpha; ref. 5), increased expression of the brown-fat-defining marker uncoupling protein 1 (UCP1) and adipogenic transcription factors PPARgamma and CCAAT/enhancer-binding proteins (C/EBPs), and induction of mitochondrial biogenesis via p38 mitogen-activated protein (MAP) kinase-(also known as Mapk14) and PGC-1-dependent pathways. Moreover, BMP7 triggers commitment of mesenchymal progenitor cells to a brown adipocyte lineage, and implantation of these cells into nude mice results in development of adipose tissue containing mostly brown adipocytes. Bmp7 knockout embryos show a marked paucity of brown fat and an almost complete absence of UCP1. Adenoviral-mediated expression of BMP7 in mice results in a significant increase in brown, but not white, fat mass and leads to an increase in energy expenditure and a reduction in weight gain. These data reveal an important role of BMP7 in promoting brown adipocyte differentiation and thermogenesis in vivo and in vitro, and provide a potential new therapeutic approach for the treatment of obesity.


Subject(s)
Adipogenesis , Adipose Tissue, Brown/growth & development , Adipose Tissue, Brown/metabolism , Bone Morphogenetic Proteins/metabolism , Energy Metabolism , Transforming Growth Factor beta/metabolism , 3T3-L1 Cells , Adipose Tissue, White/growth & development , Animals , Bone Morphogenetic Protein 7 , Cell Line , Energy Metabolism/genetics , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Nude , Mitochondria/physiology , Thermogenesis , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Mol Endocrinol ; 20(1): 147-66, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16109736

ABSTRACT

Steroidogenic factor 1 (SF-1) is an orphan nuclear receptor that has emerged as a critical mediator of endocrine function at multiple levels of the hypothalamic-pituitary-steroidogenic axis. Within the adrenal cortex, ACTH-dependent transcriptional responses, including transcriptional activation of several key steroidogenic enzymes within the steroid biosynthetic pathway, are largely dependent upon SF-1 action. The absence of a bona fide endogenous eukaryotic ligand for SF-1 suggests that signaling pathway activation downstream of the melanocortin 2 receptor (Mc2r) modulates this transcriptional response. We have used the chromatin immunoprecipitation assay to examine the temporal formation of ACTH-dependent transcription complexes on the Mc2r gene promoter. In parallel, ACTH-dependent signaling events were examined in an attempt to correlate transcriptional events with the upstream activation of signaling pathways. Our results demonstrate that ACTH-dependent signaling cascades modulate the temporal dynamics of SF-1-dependent complex assembly on the Mc2r promoter. Strikingly, the pattern of SF-1 recruitment and the subsequent attainment of active rounds of transcription support a kinetic model of SF-1 transcriptional activation, a model originally established in the context of ligand-dependent transcription by several classical nuclear hormone receptors. An assessment of the major ACTH-dependent signaling pathways highlights pivotal roles for the MAPK as well as the cAMP-dependent protein kinase A pathway in the entrainment of SF-1-mediated transcriptional events. In addition, the current study demonstrates that specific enzymatic activities are capable of regulating distinct facets of a highly ordered transcriptional response.


Subject(s)
Adrenocorticotropic Hormone/physiology , Homeodomain Proteins/metabolism , Receptor, Melanocortin, Type 2/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Acetylation , Adrenal Cortex/cytology , Animals , Cell Line , Chromatin Immunoprecipitation , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Histone Deacetylases/physiology , Histones/metabolism , Homeodomain Proteins/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Phosphoprotein Phosphatases/physiology , Phosphoric Monoester Hydrolases/metabolism , Promoter Regions, Genetic , Receptor, Melanocortin, Type 2/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Steroidogenic Factor 1 , Transcription Factors/genetics , Up-Regulation
18.
Endocrinology ; 147(3): 1322-32, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16339206

ABSTRACT

Steroidogenic factor-1 (SF-1), has emerged as a critical nuclear receptor regulating development and differentiation at several levels of the hypothalamic-pituitary-steroidogenic axis. Although many coregulatory factors have been shown to physically and functionally interact with SF-1, the relative importance of these interactions in SF-1 target tissues has not been thoroughly established. In this study we assessed roles of steroid receptor coactivator-1 (SRC-1) in hypothalamic-pituitary-adrenal (HPA) axis function using SRC-1-deficient (SRC-1-/-) mice in the absence or presence of SF-1 haploinsufficiency. Surprisingly, SRC-1 deficiency did not alter baseline HPA axis function or the acute rise in corticosterone after ACTH administration and failed to exacerbate adrenocortical dysfunction in SF-1+/- mice. However, after exposure to paradigms of acute and chronic stress, SRC-1-/- mice exhibited an elevation in serum corticosterone despite normal (nonsuppressed) ACTH, suggesting an increase in adrenal sensitivity as well as a concomitant defect in glucocorticoid-mediated feedback inhibition of the HPA axis. An examination of potential compensatory mechanism(s) revealed an increase in adrenal weight, selective elevation of melanocortin 2 receptor mRNA, and a coincident increase in SRC-2 and SRC-3 expression in SRC-1-/- adrenals. A reduction in blood glucose was observed in SRC-1-/- mice after chronic stress, consistent with a generalized state of glucocorticoid resistance. Dexamethasone suppression tests confirmed a weakened ability of glucocorticoids to 1) elevate serum glucose levels and induce hepatic phosphoenolpyruvate carboxykinase transcription and 2) suppress pituitary proopiomelanocortin transcript levels in SRC-1-/- animals. Collectively, these data are consistent with an indispensable role for SRC-1 in mediating actions of glucocorticoids in pituitary and liver.


Subject(s)
Homeodomain Proteins/physiology , Hypothalamus/metabolism , Pituitary Gland/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/metabolism , Animals , Blood Glucose/metabolism , Chromatin Immunoprecipitation , DNA Primers/chemistry , Dexamethasone/pharmacology , Female , Histone Acetyltransferases , Homeodomain Proteins/metabolism , Immunohistochemistry , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Transgenic , Nuclear Receptor Coactivator 1 , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Pro-Opiomelanocortin/biosynthesis , Protein Binding , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Restraint, Physical , Reverse Transcriptase Polymerase Chain Reaction , Steroidogenic Factor 1 , Time Factors , Transcription Factors/metabolism , Transcription, Genetic
19.
J Clin Invest ; 114(6): 828-36, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15372107

ABSTRACT

Inadequate compensatory beta cell hyperplasia in insulin-resistant states triggers the development of overt diabetes. The mechanisms that underlie this crucial adaptive response are not fully defined. Here we show that the compensatory islet-growth response to insulin resistance in 2 models--insulin receptor (IR)/IR substrate-1 (IRS-1) double heterozygous mice and liver-specific IR KO (LIRKO) mice--is severely restricted by PDX-1 heterozygosity. Six-month-old IR/IRS-1 and LIRKO mice both showed up to a 10-fold increase in beta cell mass, which involved epithelial-to-mesenchymal transition. In both models, superimposition of PDX-1 haploinsufficiency upon the background of insulin resistance completely abrogated the adaptive islet hyperplastic response, and instead the beta cells showed apoptosis resulting in premature death of the mice. This study shows that, in postdevelopmental states of beta cell growth, PDX-1 is a critical regulator of beta cell replication and is required for the compensatory response to insulin resistance.


Subject(s)
Insulin Resistance/physiology , Islets of Langerhans/pathology , Trans-Activators/deficiency , Aging/physiology , Animals , Blood Glucose/metabolism , C-Peptide/blood , Cell Division , Homeodomain Proteins/genetics , Hyperplasia , Insulin Receptor Substrate Proteins , Insulin Resistance/genetics , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Mice , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/genetics , Trans-Activators/genetics
20.
Mol Cell Biol ; 24(2): 675-86, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14701740

ABSTRACT

CCAAT/enhancer-binding protein alpha (C/EBPalpha) is one of the key transcription factors that mediate lineage specification and differentiation of multipotent myeloid progenitors into mature granulocytes. Although C/EBPalpha is known to induce granulopoiesis while suppressing monocyte differentiation, it is unclear how C/EBPalpha regulates this cell fate choice at the mechanistic level. Here we report that inducers of monocyte differentiation inhibit the alternate cell fate choice, that of granulopoiesis, through inhibition of C/EBPalpha. This inhibition is mediated by extracellular signal-regulated kinases 1 and/or 2 (ERK1/2), which interact with C/EBPalpha through an FXFP docking site and phosphorylate serine 21. As a consequence of C/EBPalpha phosphorylation, induction of granulocyte differentiation by C/EBPalpha or retinoic acid is inhibited. Our analysis of C/EBPalpha by fluorescent resonance energy transfer revealed that phosphorylation induces conformational changes in C/EBPalpha, increasing the distance between the amino termini of C/EBPalpha dimers. Thus, myeloid development is partly regulated by an ERK1/2-mediated change in the conformation of C/EBPalpha that favors monocyte differentiation by blocking granulopoiesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/metabolism , Granulocytes/cytology , Granulocytes/metabolism , Leukopoiesis/physiology , 3T3-L1 Cells , Amino Acid Sequence , Animals , Binding Sites/genetics , CCAAT-Enhancer-Binding Protein-alpha/chemistry , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Granulocytes/drug effects , Humans , K562 Cells , Leukopoiesis/drug effects , Mice , Models, Molecular , Molecular Sequence Data , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Phosphorylation , Serine/chemistry , Tretinoin/pharmacology , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...