Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Endocrinology ; 156(3): 789-801, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25521582

ABSTRACT

During obesity, a hypoxic state develops within the adipose tissue, resulting in insulin resistance. To understand the underlying mechanism, we analyzed the involvement of caveolae because they play a crucial role in the activation of insulin receptors. In the present study, we demonstrate that in 3T3-L1 adipocytes, hypoxia induces the disappearance of caveolae and inhibits the expression of Cavin-1 and Cavin-2, two proteins necessary for the formation of caveolae. In mice, hypoxia induced by the ligature of the spermatic artery results in the decrease of cavin-1 and cavin-2 expression in the epididymal adipose tissue. Down-regulation of the expression of cavins in response to hypoxia is dependent on hypoxia-inducible factor-1. Indeed, the inhibition of hypoxia-inducible factor-1 restores the expression of cavins and caveolae formation. Expression of cavins regulates insulin signaling because the silencing of cavin-1 and cavin-2 impairs insulin signaling pathway. In human, cavin-1 and cavin-2 are decreased in the sc adipose tissue of obese diabetic patients compared with lean subjects. Moreover, the expression of cavin-2 correlates negatively with the homeostatic model assessment index of insulin resistance and glycated hemoglobin level. In conclusion, we propose a new mechanism in which hypoxia inhibits cavin-1 and cavin-2 expression, resulting in the disappearance of caveolae. This leads to the inhibition of insulin signaling and the establishment of insulin resistance.


Subject(s)
Adipocytes/drug effects , Caveolae/physiology , Membrane Proteins/metabolism , Oxygen/pharmacology , RNA-Binding Proteins/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Down-Regulation , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Obesity , Phosphate-Binding Proteins , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins/genetics , Signal Transduction
2.
Sci Transl Med ; 5(203): 203ra124, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24048522

ABSTRACT

Achondroplasia is a rare genetic disease characterized by abnormal bone development, resulting in short stature. It is caused by a single point mutation in the gene coding for fibroblast growth factor receptor 3 (FGFR3), which leads to prolonged activation upon ligand binding. To prevent excessive intracellular signaling and rescue the symptoms of achondroplasia, we have developed a recombinant protein therapeutic approach using a soluble form of human FGFR3 (sFGFR3), which acts as a decoy receptor and prevents FGF from binding to mutant FGFR3. sFGFR3 was injected subcutaneously to newborn Fgfr3(ach/+) mice-the mouse model of achondroplasia-twice per week throughout the growth period during 3 weeks. Effective maturation of growth plate chondrocytes was restored in bones of treated mice, with a dose-dependent enhancement of skeletal growth in Fgfr3(ach/+) mice. This resulted in normal stature and a significant decrease in mortality and associated complications, without any evidence of toxicity. These results describe a new approach for restoring bone growth and suggest that sFGFR3 could be a potential therapy for children with achondroplasia and related disorders.


Subject(s)
Achondroplasia/drug therapy , Bone Development/drug effects , Receptor, Fibroblast Growth Factor, Type 3/therapeutic use , Animals , Female , Humans , Male , Mice , Signal Transduction/drug effects
3.
PLoS One ; 7(12): e52154, 2012.
Article in English | MEDLINE | ID: mdl-23272222

ABSTRACT

REDD1 (Regulated in development and DNA damage response 1) is a hypoxia and stress response gene and is a negative regulator of mTORC1. Since mTORC1 is involved in the negative feedback loop of insulin signaling, we have studied the role of REDD1 on insulin signaling pathway and its regulation by insulin. In human and murine adipocytes, insulin transiently stimulates REDD1 expression through a MEK dependent pathway. In HEK-293 cells, expression of a constitutive active form of MEK stabilizes REDD1 and protects REDD1 from proteasomal degradation mediated by CUL4A-DDB1 ubiquitin ligase complex. In 3T3-L1 adipocytes, silencing of REDD1 with siRNA induces an increase of mTORC1 activity as well as an inhibition of insulin signaling pathway and lipogenesis. Rapamycin, a mTORC1 inhibitor, restores the insulin signaling after downregulation of REDD1 expression. This observation suggests that REDD1 positively regulates insulin signaling through the inhibition of mTORC1 activity. In conclusion, our results demonstrate that insulin increases REDD1 expression, and that REDD1 participates in the biological response to insulin.


Subject(s)
Adipocytes/metabolism , Insulin/metabolism , Signal Transduction , Transcription Factors/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Enzyme Activation , HEK293 Cells , Humans , Insulin/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction/drug effects
4.
Biochem J ; 445(2): 265-73, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22545627

ABSTRACT

GLUT4 (glucose transporter 4) is responsible for the insulin-induced uptake of glucose by muscle and fat cells. In non-stimulated (basal) cells, GLUT4 is retained intracellularly, whereas insulin stimulation leads to its translocation from storage compartments towards the cell surface. How GLUT4 is retained intracellularly is largely unknown. Previously, aberrant GLUT4 N-glycosylation has been linked to increased basal cell-surface levels, while N-glycosylation-deficient GLUT4 was found to be quickly degraded. As recycling and degradation of GLUT4 are positively correlated, we hypothesized that incorrect N-glycosylation of GLUT4 might reduce its intracellular retention, resulting in an increased cell-surface recycling, in increased basal cell-surface levels, and in enhanced GLUT4 degradation. In the present study, we have investigated N-glycosylation-deficient GLUT4 in detail in 3T3-L1 preadipocytes, 3T3-L1 adipocytes and L6 myoblasts. We have found no alterations in retention, insulin response, internalization or glucose transport activity. Degradation of the mutant molecule was increased, although once present at the cell surface, its degradation was identical with that of wild-type GLUT4. Our findings indicate that N-glycosylation is important for efficient trafficking of GLUT4 to its proper compartments, but once the transporter has arrived there, N-glycosylation plays no further major role in its intracellular trafficking, nor in its functional activity.


Subject(s)
Adipocytes/metabolism , Cell Membrane/metabolism , Glucose Transporter Type 4/metabolism , Mutant Proteins/metabolism , Myoblasts/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , Biological Transport , Glucose/metabolism , Glucose Transporter Type 4/genetics , Glycosylation , Immunoblotting , Mice , Mutant Proteins/genetics , Mutation/genetics , Myoblasts/cytology , Proteolysis
5.
PLoS One ; 7(4): e35612, 2012.
Article in English | MEDLINE | ID: mdl-22530059

ABSTRACT

BACKGROUND: Osteopontin (OPN) plays an important role in the progression of chronic liver diseases. We aimed to quantify the liver, adipose tissue and serum levels of OPN in heavy alcohol drinkers and to compare them with the histological severity of hepatic inflammation and fibrosis. METHODOLOGY/PRINCIPAL FINDINGS: OPN was evaluated in the serum of a retrospective and prospective group of 109 and 95 heavy alcohol drinkers, respectively, in the liver of 34 patients from the retrospective group, and in the liver and adipose tissue from an additional group of 38 heavy alcohol drinkers. Serum levels of OPN increased slightly with hepatic inflammation and progressively with the severity of hepatic fibrosis. Hepatic OPN expression correlated with hepatic inflammation, fibrosis, TGFß expression, neutrophils accumulation and with the serum OPN level. Interestingly, adipose tissue OPN expression also correlated with hepatic fibrosis even after 7 days of alcohol abstinence. The elevated serum OPN level was an independent risk factor in estimating significant (F ≥ 2) fibrosis in a model combining alkaline phosphatase, albumin, hemoglobin, OPN and FibroMeter® levels. OPN had an area under the receiving operator curve that estimated significant fibrosis of 0.89 and 0.88 in the retrospective and prospective groups, respectively. OPN, Hyaluronate (AUROC: 0.88), total Cytokeratin 18 (AUROC: 0.83) and FibroMeter® (AUROC: 0.90) estimated significance to the same extent in the retrospective group. Finally, the serum OPN levels also correlated with hepatic fibrosis and estimated significant (F ≥ 2) fibrosis in 86 patients with chronic hepatitis C, which suggested that its elevated level could be a general response to chronic liver injury. CONCLUSION/SIGNIFICANCE: OPN increased in the liver, adipose tissue and serum with liver fibrosis in alcoholic patients. Further, OPN is a new relevant biomarker for significant liver fibrosis. OPN could thus be an important actor in the pathogenesis of this chronic liver disease.


Subject(s)
Adipose Tissue/metabolism , Liver Cirrhosis, Alcoholic/metabolism , Liver Cirrhosis, Alcoholic/pathology , Liver/metabolism , Osteopontin/metabolism , Adult , Female , Fibrosis , Hepatitis C, Chronic/complications , Humans , Liver/pathology , Liver Cirrhosis, Alcoholic/complications , Male , Middle Aged , Osteopontin/blood , Prognosis , ROC Curve , Risk Factors , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism
6.
Curr Opin Oncol ; 24(1): 103-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22123231

ABSTRACT

PURPOSE OF REVIEW: To focus on the potential role of metformin, a widely used antidiabetic drug, in cancer treatment. RECENT FINDINGS: Epidemiological, preclinical and cellular studies have shown in the last 6 years that metformin exerts antitumoral properties. Here, we review the very last findings concerning metformin action in cancer. The results of the first clinical trials as well as the combined action of metformin and chemotherapeutics agents in vitro and in vivo will be discussed. Recent studies show that metformin could also regulate inflammation and, therefore, may play a role in tumor microenvironment. Finally, we will present the latest publications concerning the molecular mechanisms implicated in metformin action, especially the AMP-activated kinase-independent pathways. SUMMARY: The numerous in-vitro and in-vivo studies warrant the ongoing clinical trials, which should definitively help us to determine if metformin could be used in cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Neoplasms/drug therapy , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Humans , Mice , Neoplasms/metabolism , Neoplasms/prevention & control
7.
Cancer Res ; 71(13): 4366-72, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21540236

ABSTRACT

Metformin is a widely prescribed antidiabetic drug associated with a reduced risk of cancer. Many studies show that metformin inhibits cancer cell viability through the inhibition of mTOR. We recently showed that antiproliferative action of metformin in prostate cancer cell lines is not mediated by AMP-activated protein kinase (AMPK). We identified REDD1 (also known as DDIT4 and RTP801), a negative regulator of mTOR, as a new molecular target of metformin. We show that metformin increases REDD1 expression in a p53-dependent manner. REDD1 invalidation, using siRNA or REDD1(-/-) cells, abrogates metformin inhibition of mTOR. Importantly, inhibition of REDD1 reverses metformin-induced cell-cycle arrest and significantly protects from the deleterious effects of metformin on cell transformation. Finally, we show the contribution of p53 in mediating metformin action in prostate cancer cells. These results highlight the p53/REDD1 axis as a new molecular target in anticancer therapy in response to metformin treatment.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Metformin/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transcription Factors/biosynthesis , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , Cyclin D1/metabolism , Humans , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p53/metabolism
8.
PLoS One ; 6(3): e17599, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21445263

ABSTRACT

BACKGROUND: Quantification of hepatocyte death is useful to evaluate the progression of alcoholic liver diseases. Our aims were to quantify and correlate the circulating levels of Cytokeratin 18 (CK18) and its caspases-generated fragment to disease severity in heavy alcoholics. METHODOLOGY/PRINCIPAL FINDINGS: CK18 and CK18-fragment were evaluated in the serum of 143 heavy alcoholics. Serum levels of markers of hepatocyte death (CK18), apoptosis (CK18 fragment) and necrosis (CK18 -CK18 fragment) increased in patients with severe fibrosis compared to patients with mild fibrosis. These markers strongly correlated with Mallory-Denk bodies, hepatocyte ballooning, fibrosis and with hepatic TNFα and TGFß assessed in the liver of 24 patients. Elevated levels of serum hepatocyte death and apoptotic markers were independent risk factors in predicting severe fibrosis in a model combining alkaline phosphatase, bilirubin, prothrombin index, hyaluronate, hepatocyte death and apoptotic markers. The level of markers of hepatocyte death and apoptosis had an area under the receiving operator curve that predicted severe fibrosis of 0.84 and 0.76, respectively. CONCLUSION/SIGNIFICANCE: Death of hepatocytes can be easily evaluated with serum markers and correlated with severe fibrosis in heavy alcohol drinkers. These biomarkers could be useful to rapidly evaluate liver injuries and the efficacy of therapies.


Subject(s)
Apoptosis , Biomarkers/blood , Hepatocytes/pathology , Liver Cirrhosis/blood , Liver Diseases, Alcoholic/blood , Adult , Female , Humans , Liver Cirrhosis/pathology , Liver Diseases, Alcoholic/pathology , Male , Middle Aged , Polymerase Chain Reaction , Transforming Growth Factor beta/blood , Tumor Necrosis Factor-alpha/blood
9.
Biochimie ; 93(4): 697-709, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21195125

ABSTRACT

Insulin increases muscle and fat cell glucose uptake by inducing the translocation of glucose transporter GLUT4 from intracellular compartments to the plasma membrane. Here, we have demonstrated that in 3T3-L1 adipocytes, DMSO at concentrations higher than 7.5% augmented cell surface GLUT4 levels in the absence and presence of insulin, but that at lower concentrations, DMSO only enhanced GLUT4 levels in insulin-stimulated cells. At a 5% concentration, DMSO also increased cell surface levels of the transferrin receptor and GLUT1. Glucose uptake experiments indicated that while DMSO enhanced cell surface glucose transporter levels, it also inhibited glucose transporter activity. Our studies further demonstrated that DMSO did not sensitize the adipocytes for insulin and that its effect on GLUT4 was readily reversible (t1/2∼12 min) and maintained in insulin-resistant adipocytes. An enhancement of insulin-induced GLUT4 translocation was not observed in 3T3-L1 preadipocytes and L6 myotubes, indicating cell specificity. DMSO did not enhance insulin signaling nor exocytosis of GLUT4 vesicles, but inhibited GLUT4 internalization. While other chemical chaperones (glycerol and 4-phenyl butyric acid) also acutely enhanced insulin-induced GLUT4 translocation, these effects were not mediated via changes in GLUT4 endocytosis. We conclude that DMSO is the first molecule to be described that instantaneously enhances insulin-induced increases in cell surface GLUT4 levels in adipocytes, at least in part through a reduction in GLUT4 endocytosis.


Subject(s)
Adipocytes/drug effects , Dimethyl Sulfoxide/pharmacology , Endocytosis/drug effects , Glucose Transporter Type 4/metabolism , Glucose/metabolism , Insulin/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Exocytosis , Mice , Molecular Chaperones/metabolism , Protein Transport/drug effects
10.
J Hepatol ; 55(3): 660-665, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21238518

ABSTRACT

BACKGROUND & AIMS: Non-alcoholic fatty liver disease (NAFLD) is a major hepatic consequence of obesity. It has been suggested that the high sensitivity C-reactive protein (hs-CRP) is an obesity-independent surrogate marker of severity of NAFLD, especially development of non-alcoholic steato-hepatitis (NASH), but this remains controversial. We aimed to investigate whether associations between various features of NAFLD and hs-CRP are independent of body mass index (BMI) in its broad range among obese patients. METHODS: A total of 627 obese adults (80% females), representing three cohorts from France and Belgium, had information on liver histology obtained from liver biopsies and measures of hs-CRP and BMI. We investigated whether the different features of NAFLD and BMI were associated with hs-CRP, with and without mutual adjustments using linear regression. RESULTS: BMI and hs-CRP were strongly associated. Per every 10% increase in BMI the hs-CRP level increased by 19-20% (p<0.001), and adjustment for NAFLD-stage (including no-NAFLD) did not influence the association. We found no BMI-independent association between NASH and hs-CRP. However, a positive association between degree of steatosis and hs-CRP was observed (p<0.05) and this effect remained significant after adjusting for BMI, lobular inflammation, hepatocyte ballooning, and fibrosis. We found no significant associations between the other features of NAFLD and hs-CRP. CONCLUSIONS: This study indicates that it is the accumulation of fat -both in the adipose tissue and in liver steatosis- that leads to increased hs-CRP levels among obese patients. Thus, hs-CRP may be a marker of steatosis, but not of severity of NAFLD, in obese patients.


Subject(s)
Body Mass Index , C-Reactive Protein/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Obesity/metabolism , Adiposity , Adolescent , Adult , Aged , Biomarkers/metabolism , Fatty Liver/complications , Female , Humans , Linear Models , Male , Middle Aged , Non-alcoholic Fatty Liver Disease , Obesity/complications , Severity of Illness Index , Young Adult
11.
PLoS One ; 5(12): e15560, 2010 Dec 20.
Article in English | MEDLINE | ID: mdl-21187969

ABSTRACT

In response to insulin, glucose transporter GLUT4 translocates from intracellular compartments towards the plasma membrane where it enhances cellular glucose uptake. Here, we show that sera from various species contain a factor that dose-dependently induces GLUT4 translocation and glucose uptake in 3T3-L1 adipocytes, human adipocytes, myoblasts and myotubes. Notably, the effect of this factor on GLUT4 is fully maintained in insulin-resistant cells. Our studies demonstrate that the serum-induced increase in cell surface GLUT4 levels is not due to inhibition of its internalization and is not mediated by insulin, PDGF, IGF-1, or HGF. Similarly to insulin, serum also augments cell surface levels of GLUT1 and TfR. Remarkably, the acute effect of serum on GLUT4 is largely additive to that of insulin, while it also sensitizes the cells to insulin. In accordance with these findings, serum does not appear to activate the same repertoire of downstream signaling molecules that are implicated in insulin-induced GLUT4 translocation. We conclude that in addition to insulin, at least one other biological proteinaceous factor exists that contributes to GLUT4 regulation and still functions in insulin resistance. The challenge now is to identify this factor.


Subject(s)
Gene Expression Regulation , Glucose Transporter Type 4/metabolism , Insulin/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , Glucose/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Insulin Resistance , Insulin-Like Growth Factor I/metabolism , Mice , Platelet-Derived Growth Factor/metabolism , Protein Transport , Receptors, Transferrin/metabolism
12.
PLoS One ; 5(10): e13577, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-21042596

ABSTRACT

BACKGROUND: Obesity modulates inflammation and activation of immune pathways which can lead to liver complications. We aimed at identifying expression patterns of inflammatory and immune response genes specifically associated with obesity and NASH in the liver of morbidly obese patients. METHODOLOGY/PRINCIPAL FINDINGS: Expression of 222 genes was evaluated by quantitative RT-PCR in the liver of morbidly obese patients with histologically normal liver (n = 6), or with severe steatosis without (n = 6) or with NASH (n = 6), and in lean controls (n = 5). Hepatic expression of 58 out of 222 inflammatory and immune response genes was upregulated in NASH patients. The most notable changes occurred in genes encoding chemokines and chemokine receptors involved in leukocyte recruitment, CD and cytokines involved in the T cell activation towards a Th1 phenotype, and immune semaphorins. This regulation seems to be specific for the liver since visceral adipose tissue expression and serum levels of MCP1, IP10, TNFα and IL6 were not modified. Importantly, 47 other genes were already upregulated in histologically normal liver (e.g. CRP, Toll-like receptor (TLR) pathway). Interestingly, serum palmitate, known to activate the TLR pathway, was increased with steatosis. CONCLUSION/SIGNIFICANCE: The liver of obese patients without histological abnormalities already displayed a low-grade inflammation and could be more responsive to activators of the TLR pathway. NASH was then characterized by a specific gene signature. These findings help to identify new potential actors of the pathogenesis of NAFLD.


Subject(s)
Gene Expression Profiling , Inflammation/genetics , Liver/metabolism , Obesity, Morbid/genetics , Adult , Case-Control Studies , Fatty Liver/genetics , Female , Humans , Male , Non-alcoholic Fatty Liver Disease , Obesity, Morbid/immunology , Reverse Transcriptase Polymerase Chain Reaction
13.
J Proteome Res ; 9(9): 4501-12, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20684516

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease in most western countries. Current NAFLD diagnosis methods (e.g., liver biopsy analysis or imaging techniques) are poorly suited as tests for such a prevalent condition, from both a clinical and financial point of view. The present work aims to demonstrate the potential utility of serum metabolic profiling in defining phenotypic biomarkers that could be useful in NAFLD management. A parallel animal model/human NAFLD exploratory metabolomics approach was employed, using ultra performance liquid chromatography-mass spectrometry (UPLC-MS) to analyze 42 serum samples collected from nondiabetic, morbidly obese, biopsy-proven NAFLD patients, and 17 animals belonging to the glycine N-methyltransferase knockout (GNMT-KO) NAFLD mouse model. Multivariate statistical analysis of the data revealed a series of common biomarkers that were significantly altered in the NAFLD (GNMT-KO) subjects in comparison to their normal liver counterparts (WT). Many of the compounds observed could be associated with biochemical perturbations associated with liver dysfunction (e.g., reduced Creatine) and inflammation (e.g., eicosanoid signaling). This differential metabolic phenotyping approach may have a future role as a supplement for clinical decision making in NAFLD and in the adaption to more individualized treatment protocols.


Subject(s)
Chromatography, Liquid/methods , Mass Spectrometry/methods , Metabolomics/methods , Animals , Biomarkers/blood , Disease Models, Animal , Disease Progression , Fatty Liver/blood , Glycine N-Methyltransferase/genetics , Humans , Lipid Metabolism , Male , Mice , Mice, Knockout , Multivariate Analysis , Non-alcoholic Fatty Liver Disease , Principal Component Analysis
14.
Mol Cancer Ther ; 9(5): 1092-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20442309

ABSTRACT

Metformin is the most widely used antidiabetic drug in the world, and there is increasing evidence of a potential efficacy of this agent as an anticancer drug. First, epidemiological studies show a decrease in cancer incidence in metformin-treated patients. Second, metformin decreases insulin resistance and indirectly reduces insulin level, a beneficial effect because insulin promotes cancer cell growth. Third, several reports outline a direct inhibitory effect of metformin on cancer cell growth and an antitumoral action. Finally, metformin activates the AMP activated protein kinase (AMPK) pathway, a major sensor of the energetic status of the cell, which has been proposed as a promising therapeutic target in cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Neoplasms/drug therapy , Adenylate Kinase/metabolism , Adenylate Kinase/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Humans , Hypoglycemic Agents/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Medical Oncology/trends , Metformin/pharmacology , Models, Biological , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , TOR Serine-Threonine Kinases
15.
Cancer Res ; 70(6): 2465-75, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215500

ABSTRACT

Targeting cancer cell metabolism is a new promising strategy to fight cancer. Metformin, a widely used antidiabetic agent, exerts antitumoral and antiproliferative action. In this study, the addition of metformin to 2-deoxyglucose (2DG) inhibited mitochondrial respiration and glycolysis in prostate cancer cells leading to a severe depletion in ATP. The combination of the two drugs was much more harmful for cancer cells than the treatment with metformin or 2DG alone, leading to 96% inhibition of cell viability in LNCaP prostate cancer cells. In contrast, a moderate effect on cell viability was observed in normal prostate epithelial cells. At the cellular level, the combination of metformin and 2DG induced p53-dependent apoptosis via the energy sensor pathway AMP kinase, and the reexpression of a functional p53 in p53-deficient prostate cancer cells restored caspase-3 activity. In addition to apoptosis, the combination of metformin and 2DG arrested prostate cancer cells in G(2)-M. This G(2)-M arrest was independent of p53 and correlated with a stronger decrease in cell viability than obtained with either drug. Finally, metformin inhibited 2DG-induced autophagy, decreased beclin 1 expression, and triggered a switch from a survival process to cell death. Our study reinforces the growing interest of metabolic perturbators in cancer therapy and highlights the potential use of the combination of metformin and 2DG as an anticancerous treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Deoxyglucose/pharmacology , Metformin/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Adenosine Triphosphate/deficiency , Adenosine Triphosphate/metabolism , Adenylate Kinase/metabolism , Autophagy/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Deoxyglucose/administration & dosage , Deoxyglucose/antagonists & inhibitors , Drug Synergism , Humans , Male , Metformin/administration & dosage , Prostatic Neoplasms/pathology
16.
FEBS Lett ; 584(3): 537-42, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19968987

ABSTRACT

Glucose transporter 4 (GLUT4) is efficiently retained intracellularly. Here, we investigated the insulin-induced reduction of retention. While increasing insulin concentrations led to gradual increases in both the amount of recycling GLUT4 molecules and cell surface GLUT4 levels, the kinetics of the increase in time was independent of insulin concentration. To determine whether there are GLUT4 subpools that have a distinct insulin sensitivity, adipocytes were consecutively stimulated twice with a low concentration of insulin while recycling GLUT4 molecules were continuously labeled. This revealed that not the same pool of GLUT4 molecules was mobilized twice and thus that upon insulin stimulation, GLUT4 is likely to be recruited at random for insertion within the plasma membrane.


Subject(s)
Cell Membrane/metabolism , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Protein Transport/drug effects , 3T3-L1 Cells , Animals , Dose-Response Relationship, Drug , Hypoglycemic Agents/pharmacology , Mice
17.
Diabetes ; 59(1): 61-70, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19808894

ABSTRACT

OBJECTIVE: Activation of extracellular signal-regulated kinase-(ERK)-1/2 by cytokines in adipocytes is involved in the alterations of adipose tissue functions participating in insulin resistance. This study aims at identifying proteins regulating ERK1/2 activity, specifically in response to inflammatory cytokines, to provide new insights into mechanisms leading to abnormal adipose tissue function. RESEARCH DESIGN AND METHODS: Kinase activities were inhibited with pharmacological inhibitors or siRNA. Lipolysis was monitored through glycerol production. Gene expression in adipocytes and adipose tissue of obese mice and subjects was measured by real-time PCR. RESULTS: IkappaB kinase-(IKK)-beta inhibition prevented mitogen-activated protein (MAP) kinase kinase (MEK)/ERK1/2 activation in response to interleukin (IL)-1beta and tumor necrosis factor (TNF)-alpha but not insulin in 3T3-L1 and human adipocytes, suggesting that IKKbeta regulated a MAP kinase kinase kinase (MAP3K) involved in ERK1/2 activation induced by inflammatory cytokines. We show that the MAP3K8 called Tpl2 was expressed in adipocytes and that IL-1beta and TNF-alpha activated Tpl2 and regulated its expression through an IKKbeta pathway. Pharmacological inhibition or silencing of Tpl2 prevented MEK/ERK1/2 activation by these cytokines but not by insulin, demonstrating its involvement in ERK1/2 activation specifically in response to inflammatory stimuli. Importantly, Tpl2 was implicated in cytokine-induced lipolysis and in insulin receptor substrate-1 serine phosphorylation. Tpl2 mRNA expression was upregulated in adipose tissue of obese mice and patients and correlated with TNF-alpha expression. CONCLUSIONS: Tpl2 is selectively involved in inflammatory cytokine-induced ERK1/2 activation in adipocytes and is implicated in their deleterious effects on adipocyte functions. The deregulated expression of Tpl2 in adipose tissue suggests that Tpl2 may be a new actor in adipose tissue dysfunction in obesity.


Subject(s)
3T3-L1 Cells/cytology , Adipocytes/cytology , Interleukin-1beta/pharmacology , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins/genetics , Tumor Necrosis Factor-alpha/pharmacology , Adipocytes/physiology , Animals , Cell Differentiation , Enzyme Activation , Fasting , Humans , I-kappa B Kinase/metabolism , Lipolysis , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Obese , Mitogen-Activated Protein Kinase 3/drug effects , Obesity, Morbid/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Thinness , Up-Regulation
18.
Biochem Pharmacol ; 79(9): 1300-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20026082

ABSTRACT

Insulin induces a translocation of the glucose transporter GLUT4 from intracellular storage compartments towards the cell surface in adipocytes and skeletal muscle cells, allowing the cells to take up glucose. In type 2 diabetes-associated insulin resistance, the efficiency of this process is reduced. The thiazolidinediones, widely prescribed as anti-diabetic therapy, are generally regarded as insulin-sensitizers. The aim of this study was to evaluate the effect of the thiazolidinedione rosiglitazone (BRL 49653) on GLUT4 in adipocytes. When applied during differentiation, rosiglitazone dose dependently augmented GLUT4 expression along with the formation of lipid droplets. Intriguingly, its presence during differentiation led to increases in both cell surface GLUT4 levels and insulin sensitivity of GLUT4 translocation in mature adipocytes. Treatment of fully differentiated adipocytes with rosiglitazone also led to increases in GLUT4 at the plasma membrane. Rosiglitazone similarly affected cell surface levels of the endosomal transferrin receptor, but did not alter the GLUT4 internalization rate. The augmentation in cell surface GLUT4 levels was maintained in adipocytes that were rendered insulin-resistant in vitro by a 24h insulin treatment and moreover in these cells rosiglitazone also fully restored insulin-induced GLUT4 translocation. We conclude that in adipocytes, rosiglitazone increases cell surface GLUT4 levels by increasing its endosomal recycling and restores insulin-induced GLUT4 translocation in insulin resistance. These results implicate novel modes of action on GLUT4 that are all likely to contribute to the insulin-sensitizing effect of rosiglitazone in type 2 diabetes.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Endosomes/metabolism , Glucose Transporter Type 4/metabolism , Thiazolidinediones/pharmacology , 3T3-L1 Cells , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin Resistance , Mice , Rosiglitazone , Time Factors
19.
J Biol Chem ; 285(8): 5157-64, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-19996311

ABSTRACT

REDD1 (regulated in development and DNA damage responses) is essential for the inhibition of mTORC1 (mammalian target of rapamycin complex) signaling pathway in response to hypoxia. REDD1 expression is regulated by many stresses such as hypoxia, oxidative stress, and energy depletion. However, the regulation of REDD1 expression in response to insulin remains unknown. In the present study, we demonstrate that in murine and in human adipocytes, insulin stimulates REDD1 expression. Insulin-induced REDD1 expression occurs through phosphoinositide 3-kinase/mTOR-dependent pathways. Moreover, using echinomycin, a hypoxia-inducible factor 1 (HIF-1) inhibitor, and HIF-1alpha small interfering RNA, we demonstrate that insulin stimulates REDD1 expression only through the transcription factor HIF-1. In conclusion, our study shows that insulin stimulates REDD1 expression in adipocytes.


Subject(s)
Adipocytes/metabolism , Gene Expression Regulation/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin/metabolism , Transcription Factors/metabolism , 3T3-L1 Cells , Animals , Anti-Bacterial Agents/pharmacology , Echinomycin/pharmacology , Gene Expression Regulation/drug effects , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Insulin/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/pharmacology , TOR Serine-Threonine Kinases
20.
J Cell Biol ; 187(6): 859-74, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-20008564

ABSTRACT

Mammalian target of rapamycin (mTOR) is a key regulator of cell growth that associates with raptor and rictor to form the mTOR complex 1 (mTORC1) and mTORC2, respectively. Raptor is required for oxidative muscle integrity, whereas rictor is dispensable. In this study, we show that muscle-specific inactivation of mTOR leads to severe myopathy, resulting in premature death. mTOR-deficient muscles display metabolic changes similar to those observed in muscles lacking raptor, including impaired oxidative metabolism, altered mitochondrial regulation, and glycogen accumulation associated with protein kinase B/Akt hyperactivation. In addition, mTOR-deficient muscles exhibit increased basal glucose uptake, whereas whole body glucose homeostasis is essentially maintained. Importantly, loss of mTOR exacerbates the myopathic features in both slow oxidative and fast glycolytic muscles. Moreover, mTOR but not raptor and rictor deficiency leads to reduced muscle dystrophin content. We provide evidence that mTOR controls dystrophin transcription in a cell-autonomous, rapamycin-resistant, and kinase-independent manner. Collectively, our results demonstrate that mTOR acts mainly via mTORC1, whereas regulation of dystrophin is raptor and rictor independent.


Subject(s)
Carrier Proteins/metabolism , Dystrophin/metabolism , Muscle, Skeletal/enzymology , Muscular Dystrophy, Animal/enzymology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adaptor Proteins, Signal Transducing , Age Factors , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cells, Cultured , Dystrophin/genetics , Electroporation , Energy Metabolism , Enzyme Activation , Female , Glucose/metabolism , Glycogen/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Muscle/enzymology , Muscle Contraction , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Mutation , Oxidation-Reduction , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Rats , Regulatory-Associated Protein of mTOR , Severity of Illness Index , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transduction, Genetic , Utrophin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...