Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 3953, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37402735

ABSTRACT

Urocortin 2 (UCN2) acts as a ligand for the G protein-coupled receptor corticotropin-releasing hormone receptor 2 (CRHR2). UCN2 has been reported to improve or worsen insulin sensitivity and glucose tolerance in vivo. Here we show that acute dosing of UCN2 induces systemic insulin resistance in male mice and skeletal muscle. Inversely, chronic elevation of UCN2 by injection with adenovirus encoding UCN2 resolves metabolic complications, improving glucose tolerance. CRHR2 recruits Gs in response to low concentrations of UCN2, as well as Gi and ß-Arrestin at high concentrations of UCN2. Pre-treating cells and skeletal muscle ex vivo with UCN2 leads to internalization of CRHR2, dampened ligand-dependent increases in cAMP, and blunted reductions in insulin signaling. These results provide mechanistic insights into how UCN2 regulates insulin sensitivity and glucose metabolism in skeletal muscle and in vivo. Importantly, a working model was derived from these results that unifies the contradictory metabolic effects of UCN2.


Subject(s)
Insulin Resistance , Animals , Male , Mice , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Glucose/metabolism , Insulin , Ligands , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins/genetics , Urocortins/metabolism
2.
Am J Physiol Gastrointest Liver Physiol ; 322(2): G247-G255, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34935522

ABSTRACT

Growth differentiation factor 15 (GDF15), a TGFß superfamily cytokine, acts through its receptor, cell line-derived neurotrophic factorfamily receptor α-like (GFRAL), to suppress food intake and promote nausea. GDF15 is broadly expressed at low levels but increases in states of disease such as cancer, cachexia, and sepsis. Whether GDF15 is necessary for inducing sepsis-associated anorexia and body weight loss is currently unclear. To test this we used a model of moderate systemic infection in GDF15KO and GFRALKO mice with lipopolysaccharide (LPS) treatment to define the role of GDF15 signaling in infection-mediated physiologic responses. Since physiological responses to LPS depend on housing temperature, we tested the effects of subthermoneutral and thermoneutral conditions on eliciting anorexia and inducing GDF15. Our data demonstrate a conserved LPS-mediated increase in circulating GDF15 levels in mouse, rat, and human. However, we did not detect differences in LPS-induced anorexia between WT and GDF15KO or GFRALKO mice. Furthermore, there were no differences in anorexia or circulating GDF15 levels at either thermoneutral or subthermoneutral housing conditions in LPS-treated mice. These data demonstrate that GDF15 is not necessary to drive food intake suppression in response to moderate doses of LPS.NEW & NOTEWORTHY Although many responses to LPS depend on housing temperature, the anorexic response to LPS does not. LPS results in a potent and rapid increase in circulating levels of GDF15 in mice, rats, and humans. Nevertheless, GDF15 and its receptor (GFRAL) are not required for the anorexic response to systemic LPS administration. The anorexic response to LPS likely involves a myriad of complex physiological alterations.


Subject(s)
Anorexia/metabolism , Growth Differentiation Factor 15/drug effects , Growth Differentiation Factor 15/metabolism , Lipopolysaccharides/pharmacology , Animals , Eating/drug effects , Humans , Mice , Nausea/chemically induced , Rats , Weight Loss/drug effects
3.
iScience ; 24(6): 102554, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34189431

ABSTRACT

Growth differentiation factor 15 (GDF15) causes anorexia and weight loss in animal models, and higher circulating levels are associated with cachexia and reduced survival in cancer and other chronic diseases such as sepsis. To investigate the role of sepsis-induced GDF15, we examined whether GDF15 neutralization via a validated and highly potent monoclonal antibody, mAB2, modulates lipopolysaccharide (LPS)-induced anorexia, weight loss, and mortality in rodents. LPS injection transiently increased circulating GDF15 in wild-type mice, decreased food intake and body weight, and increased illness behavior and mortality at a high dose. GDF15 neutralization with mAB2 did not prevent or exacerbate any of the effects of LPS. Similarly, in GDF15 knockout mice, the LPS effect on appetite and survival was comparable with that observed in wild-type controls. Therefore, effective inhibition of circulating active GDF15 via an antibody or via gene knockout demonstrated that survival in the LPS acute inflammation model was independent of GDF15.

4.
Sci Rep ; 11(1): 8921, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33903632

ABSTRACT

GDF15 is a distant TGF-ß family member that induces anorexia and weight loss. Due to its function, GDF15 has attracted attention as a potential therapeutic for the treatment of obesity and its associated metabolic diseases. However, the pharmacokinetic and physicochemical properties of GDF15 present several challenges for its development as a therapeutic, including a short half-life, high aggregation propensity, and protease susceptibility in serum. Here, we report the design, characterization and optimization of GDF15 in an Fc-fusion protein format with improved therapeutic properties. Using a structure-based engineering approach, we combined knob-into-hole Fc technology and N-linked glycosylation site mutagenesis for half-life extension, improved solubility and protease resistance. In addition, we identified a set of mutations at the receptor binding site of GDF15 that show increased GFRAL binding affinity and led to significant half-life extension. We also identified a single point mutation that increases p-ERK signaling activity and results in improved weight loss efficacy in vivo. Taken together, our findings allowed us to develop GDF15 in a new therapeutic format that demonstrates better efficacy and potential for improved manufacturability.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Growth Differentiation Factor 15/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Recombinant Fusion Proteins/pharmacology , Weight Loss/drug effects , Animals , CHO Cells , Cricetulus , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glycosylation , Humans , Mice , Point Mutation , Protein Engineering
5.
EMBO J ; 37(24)2018 12 14.
Article in English | MEDLINE | ID: mdl-30446598

ABSTRACT

A finely tuned balance of self-renewal, differentiation, proliferation, and survival governs the pool size and regenerative capacity of blood-forming hematopoietic stem and progenitor cells (HSPCs). Here, we report that protein kinase C delta (PKCδ) is a critical regulator of adult HSPC number and function that couples the proliferative and metabolic activities of HSPCs. PKCδ-deficient mice showed a pronounced increase in HSPC numbers, increased competence in reconstituting lethally irradiated recipients, enhanced long-term competitive advantage in serial transplantation studies, and an augmented HSPC recovery during stress. PKCδ-deficient HSPCs also showed accelerated proliferation and reduced apoptosis, but did not exhaust in serial transplant assays or induce leukemia. Using inducible knockout and transplantation models, we further found that PKCδ acts in a hematopoietic cell-intrinsic manner to restrict HSPC number and bone marrow regenerative function. Mechanistically, PKCδ regulates HSPC energy metabolism and coordinately governs multiple regulators within signaling pathways implicated in HSPC homeostasis. Together, these data identify PKCδ as a critical regulator of HSPC signaling and metabolism that acts to limit HSPC expansion in response to physiological and regenerative demands.


Subject(s)
Apoptosis , Bone Marrow/enzymology , Cell Proliferation , Hematopoietic Stem Cells/enzymology , Protein Kinase C-delta/metabolism , Signal Transduction , Animals , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , Protein Kinase C-delta/genetics
6.
Diabetes ; 64(12): 4023-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26307588

ABSTRACT

Protein kinase C (PKC)δ has been shown to be increased in liver in obesity and plays an important role in the development of hepatic insulin resistance in both mice and humans. In the current study, we explored the role of PKCδ in skeletal muscle in the control of insulin sensitivity and glucose metabolism by generating mice in which PKCδ was deleted specifically in muscle using Cre-lox recombination. Deletion of PKCδ in muscle improved insulin signaling in young mice, especially at low insulin doses; however, this did not change glucose tolerance or insulin tolerance tests done with pharmacological levels of insulin. Likewise, in young mice, muscle-specific deletion of PKCδ did not rescue high-fat diet-induced insulin resistance or glucose intolerance. However, with an increase in age, PKCδ levels in muscle increased, and by 6 to 7 months of age, muscle-specific deletion of PKCδ improved whole-body insulin sensitivity and muscle insulin resistance and by 15 months of age improved the age-related decline in whole-body glucose tolerance. At 15 months of age, M-PKCδKO mice also exhibited decreased metabolic rate and lower levels of some proteins of the OXPHOS complex suggesting a role for PKCδ in the regulation of mitochondrial mass at older age. These data indicate an important role of PKCδ in the regulation of insulin sensitivity and mitochondrial homeostasis in skeletal muscle with aging.


Subject(s)
Aging , Energy Metabolism , Enzyme Induction , Insulin Resistance , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Protein Kinase C-delta/metabolism , Adiposity , Animals , Blood Glucose/analysis , Diet, High-Fat/adverse effects , Enzyme Repression , Glucose Intolerance/blood , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Insulin/blood , Mice, Knockout , Mice, Transgenic , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/pathology , Mitochondrial Dynamics , Muscle Development , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Obesity/blood , Obesity/etiology , Obesity/metabolism , Protein Kinase C-delta/genetics , Recombinant Proteins/metabolism
7.
Cell Metab ; 22(3): 516-530, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26299453

ABSTRACT

Obesity, diabetes, and metabolic syndrome result from complex interactions between genetic and environmental factors, including the gut microbiota. To dissect these interactions, we utilized three commonly used inbred strains of mice-obesity/diabetes-prone C57Bl/6J mice, obesity/diabetes-resistant 129S1/SvImJ from Jackson Laboratory, and obesity-prone but diabetes-resistant 129S6/SvEvTac from Taconic-plus three derivative lines generated by breeding these strains in a new, common environment. Analysis of metabolic parameters and gut microbiota in all strains and their environmentally normalized derivatives revealed strong interactions between microbiota, diet, breeding site, and metabolic phenotype. Strain-dependent and strain-independent correlations were found between specific microbiota and phenotypes, some of which could be transferred to germ-free recipient animals by fecal transplantation. Environmental reprogramming of microbiota resulted in 129S6/SvEvTac becoming obesity resistant. Thus, development of obesity/metabolic syndrome is the result of interactions between gut microbiota, host genetics, and diet. In permissive genetic backgrounds, environmental reprograming of microbiota can ameliorate development of metabolic syndrome.


Subject(s)
Diabetes Mellitus/genetics , Diabetes Mellitus/microbiology , Gastrointestinal Microbiome , Metabolic Syndrome/genetics , Metabolic Syndrome/microbiology , Obesity/genetics , Obesity/microbiology , Animals , Diabetes Mellitus/pathology , Diet , Gene-Environment Interaction , Genotype , Insulin Resistance , Male , Metabolic Syndrome/pathology , Mice, Inbred C57BL , Mice, Inbred Strains , Obesity/pathology , Phenotype , Weight Gain
8.
FASEB J ; 28(10): 4408-19, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25005176

ABSTRACT

Mitochondrial dysfunction in adipose tissue occurs in obesity, type 2 diabetes, and some forms of lipodystrophy, but whether this dysfunction contributes to or is the result of these disorders is unknown. To investigate the physiological consequences of severe mitochondrial impairment in adipose tissue, we generated mice deficient in mitochondrial transcription factor A (TFAM) in adipocytes by using mice carrying adiponectin-Cre and TFAM floxed alleles. These adiponectin TFAM-knockout (adipo-TFAM-KO) mice had a 75-81% reduction in TFAM in the subcutaneous and intra-abdominal white adipose tissue (WAT) and interscapular brown adipose tissue (BAT), causing decreased expression and enzymatic activity of proteins in complexes I, III, and IV of the electron transport chain (ETC). This mitochondrial dysfunction led to adipocyte death and inflammation in WAT and a whitening of BAT. As a result, adipo-TFAM-KO mice were resistant to weight gain, but exhibited insulin resistance on both normal chow and high-fat diets. These lipodystrophic mice also developed hypertension, cardiac hypertrophy, and cardiac dysfunction. Thus, isolated mitochondrial dysfunction in adipose tissue can lead a syndrome of lipodystrophy with metabolic syndrome and cardiovascular complications.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Insulin Resistance , Lipodystrophy/metabolism , Mitochondria/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Animals , Cardiomegaly/genetics , Cardiomegaly/metabolism , DNA-Binding Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , High Mobility Group Proteins/genetics , Hypertension/genetics , Hypertension/metabolism , Lipodystrophy/genetics , Lipodystrophy/physiopathology , Male , Mice , Weight Gain
9.
Bull Cancer ; 100(10): 1017-22, 2013 Oct.
Article in French | MEDLINE | ID: mdl-24067889

ABSTRACT

The improper adherence to therapy is an emerging medical and economic issue in oncology which raised with the increasing use of oral anti-cancer treatment. Currently, the average rate of non-adherence to oral anti-cancer therapy is estimated at around 21%. In this study, we use the examples of the imatinib treatment against chronic lymphocytic leukemia and the tamoxifene treatment against breast cancer to assess the negative consequences of the non-adherence to therapy in terms of medical outcome and health care cost. One of the main causes of non-adherence to these oral cancer treatments is depression. Surprisingly, this aspect is still relatively unknown to oncologists, while depression has been taken into account for the treatment of other chronic diseases (e.g. diabetes…). We therefore propose that cancer patients should be screened for depression throughout their treatment to improve the adherence to therapy. Cancer patients should have the opportunity to explain their own perception of their disease and their treatment that are key parameters in the onset of depression. The recent use of oral therapy in cancer treatment should thus be accompanied by the establishment of a global management of cancer patient on a case-by-case basis.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzamides/administration & dosage , Breast Neoplasms/drug therapy , Depressive Disorder/psychology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Medication Adherence/psychology , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Tamoxifen/administration & dosage , Administration, Oral , Antineoplastic Agents/economics , Benzamides/economics , Breast Neoplasms/psychology , Depressive Disorder/diagnosis , Depressive Disorder/therapy , Female , Health Care Costs , Humans , Imatinib Mesylate , Leukemia, Lymphocytic, Chronic, B-Cell/psychology , Medical Oncology , Medication Adherence/statistics & numerical data , Piperazines/economics , Pyrimidines/economics , Tamoxifen/economics , Treatment Outcome
10.
Cell Metab ; 16(6): 765-76, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23168219

ABSTRACT

Obesity and type 2 diabetes are associated with mitochondrial dysfunction in adipose tissue, but the role for adipose tissue mitochondria in the development of these disorders is currently unknown. To understand the impact of adipose tissue mitochondria on whole-body metabolism, we have generated a mouse model with disruption of the mitochondrial transcription factor A (TFAM) specifically in fat. F-TFKO adipose tissue exhibit decreased mtDNA copy number, altered levels of proteins of the electron transport chain, and perturbed mitochondrial function with decreased complex I activity and greater oxygen consumption and uncoupling. As a result, F-TFKO mice exhibit higher energy expenditure and are protected from age- and diet-induced obesity, insulin resistance, and hepatosteatosis, despite a greater food intake. Thus, TFAM deletion in the adipose tissue increases mitochondrial oxidation that has positive metabolic effects, suggesting that regulation of adipose tissue mitochondria may be a potential therapeutic target for the treatment of obesity.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , DNA-Binding Proteins/metabolism , Insulin Resistance , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Obesity/metabolism , Transcription Factors/metabolism , Animals , Cell Line , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Electron Transport Complex I/metabolism , Energy Metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Obesity/pathology , Oxidative Phosphorylation , Oxygen/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
11.
Diabetes ; 61(9): 2289-98, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22751693

ABSTRACT

Obesity, especially visceral obesity, is associated with insulin resistance and metabolic syndrome. We previously identified the cell surface proteoglycan glypican-4 as differentially expressed in subcutaneous versus visceral white fat depots. Here we show that glypican-4 is released from cells and adipose tissue explants of mice, and that circulating glypican-4 levels correlate with BMI and insulin sensitivity in humans. Furthermore, glypican-4 interacts with the insulin receptor, enhances insulin receptor signaling, and enhances adipocyte differentiation. Conversely, depletion of glypican-4 results in reduced activation of the insulin receptor and prevents adipocyte differentiation in vitro by inhibiting insulin-mediated C/EBPß phosphorylation. These functions of glypican-4 are independent of its glycosylphosphatidylinositol membrane anchorage, as a nonmembrane-bound mutant of glypican-4 phenocopies the effects of native glypican-4 overexpression. In summary, glypican-4 is a novel circulating insulin sensitizing adipose-derived factor that, unlike other insulin sensitizers, acts directly on the insulin receptor to enhance signaling.


Subject(s)
Glypicans/pharmacology , Receptor, Insulin/drug effects , Adipocytes , Animals , Cell Differentiation/drug effects , Glypicans/biosynthesis , Humans , Intra-Abdominal Fat/metabolism , Mice , Receptor, Insulin/metabolism
13.
J Clin Invest ; 121(6): 2504-17, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21576825

ABSTRACT

C57BL/6J and 129S6/Sv (B6 and 129) mice differ dramatically in their susceptibility to developing diabetes in response to diet- or genetically induced insulin resistance. A major locus contributing to this difference has been mapped to a region on mouse chromosome 14 that contains the gene encoding PKCδ. Here, we found that PKCδ expression in liver was 2-fold higher in B6 versus 129 mice from birth and was further increased in B6 but not 129 mice in response to a high-fat diet. PRKCD gene expression was also elevated in obese humans and was positively correlated with fasting glucose and circulating triglycerides. Mice with global or liver-specific inactivation of the Prkcd gene displayed increased hepatic insulin signaling and reduced expression of gluconeogenic and lipogenic enzymes. This resulted in increased insulin-induced suppression of hepatic gluconeogenesis, improved glucose tolerance, and reduced hepatosteatosis with aging. Conversely, mice with liver-specific overexpression of PKCδ developed hepatic insulin resistance characterized by decreased insulin signaling, enhanced lipogenic gene expression, and hepatosteatosis. Therefore, changes in the expression and regulation of PKCδ between strains of mice and in obese humans play an important role in the genetic risk of hepatic insulin resistance, glucose intolerance, and hepatosteatosis; and thus PKCδ may be a potential target in the treatment of metabolic syndrome.


Subject(s)
Fatty Liver/enzymology , Insulin Resistance/physiology , Liver/enzymology , Obesity/enzymology , Protein Kinase C-delta/physiology , Aging/metabolism , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/therapy , Dietary Fats/toxicity , Enzyme Induction/drug effects , Fasting/blood , Fatty Liver/etiology , Female , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Glucose Intolerance/enzymology , Glucose Intolerance/genetics , Humans , Insulin/pharmacology , Lipogenesis/drug effects , Lipogenesis/genetics , Male , Metabolic Syndrome/enzymology , Metabolic Syndrome/prevention & control , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Obesity/complications , Protein Kinase C-delta/biosynthesis , Protein Kinase C-delta/deficiency , Protein Kinase C-delta/genetics , Species Specificity , Triglycerides/blood
14.
Proc Natl Acad Sci U S A ; 108(7): 2771-6, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282637

ABSTRACT

Increased intraabdominal (visceral) fat is associated with a high risk of diabetes and metabolic syndrome. We have previously shown that the mesodermal developmental transcription factor Tbx15 is highly differentially expressed between visceral and subcutaneous (s.c.) fat in both humans and rodents, and in humans visceral fat Tbx15 expression is decreased in obesity. Here we show that, in mice, Tbx15 is 260-fold more highly expressed in s.c. preadipocytes than in epididymal preadipocytes. Overexpression of Tbx15 in 3T3-L1 preadipocytes impairs adipocyte differentiation and decreases triglyceride content. This defect in differentiation can be corrected by stimulating cells with the PPARγ agonist rosiglitazone (Rosi). However, triglyceride accumulation remains decreased by ∼50%, due to a decrease in basal lipogenic rate and increase in basal lipolytic rate. 3T3-L1 preadipocytes overexpressing Tbx15 also have a 15% reduction in mitochondrial mass and a 28% reduction in basal mitochondrial respiration (P = 0.004) and ATP turnover (P = 0.02), and a 45% (P = 0.003) reduction in mitochondrial respiratory capacity. Thus, differential expression of Tbx15 between fat depots plays an important role in the interdepot differences in adipocyte differentiation, triglyceride accumulation, and mitochondrial function that may contribute to the risk of diabetes and metabolic disease.


Subject(s)
Adipocytes/physiology , Cell Differentiation/genetics , Cell Respiration/genetics , Mitochondria/physiology , Subcutaneous Fat/metabolism , T-Box Domain Proteins/metabolism , 3T3-L1 Cells , Adenosine Triphosphate/metabolism , Animals , Azo Compounds , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Respiration/physiology , Cloning, Molecular , DNA Primers/genetics , Energy Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Oxygen Consumption/physiology , PPAR gamma/agonists , Polymerase Chain Reaction , Rosiglitazone , T-Box Domain Proteins/genetics , Thiazolidinediones/pharmacology
15.
Sci Signal ; 3(151): ra87, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21139139

ABSTRACT

Insulin and insulin-like growth factor 1 (IGF-1) act as antiapoptotic hormones. We found that, unexpectedly, double-knockout (DKO) cells that lacked both insulin and IGF-1 receptors (IR and IGF1R, respectively) were resistant to apoptosis induced through either the intrinsic or the extrinsic pathway. This resistance to apoptosis was associated with decreased abundance of the proapoptotic protein Bax and increases in abundance of the antiapoptotic proteins Bcl-2, Bcl-xL, XIAP, and Flip. These changes in protein abundance involved primarily posttranscriptional mechanisms. Restoration of IR or IGF1R to DKO cells also restored their sensitivity to apoptosis. Notably, expression of a catalytically inactive mutant form of the IR also restored susceptibility to apoptosis. Thus, IR and IGF1R have bidirectional roles in the control of cell survival and can be viewed as dependence receptors. Insulin and IGF-1 binding stimulates receptor tyrosine kinase activity and blocks apoptosis, whereas unliganded IR and IGF1R, acting through a mechanism independent of their catalytic activity, exert a permissive effect on cell death.


Subject(s)
Apoptosis/physiology , Models, Biological , Receptor, IGF Type 1/physiology , Receptor, Insulin/physiology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Line, Transformed , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Hydrogen Peroxide/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidants/pharmacology , Phosphotransferases/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology
16.
Diabetes ; 59(11): 2960-71, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713682

ABSTRACT

OBJECTIVE: Type 2 diabetes and obesity are increasingly affecting human populations around the world. Our goal was to identify early molecular signatures predicting genetic risk to these metabolic diseases using two strains of mice that differ greatly in disease susceptibility. RESEARCH DESIGN AND METHODS: We integrated metabolic characterization, gene expression, protein-protein interaction networks, RT-PCR, and flow cytometry analyses of adipose, skeletal muscle, and liver tissue of diabetes-prone C57BL/6NTac (B6) mice and diabetes-resistant 129S6/SvEvTac (129) mice at 6 weeks and 6 months of age. RESULTS: At 6 weeks of age, B6 mice were metabolically indistinguishable from 129 mice, however, adipose tissue showed a consistent gene expression signature that differentiated between the strains. In particular, immune system gene networks and inflammatory biomarkers were upregulated in adipose tissue of B6 mice, despite a low normal fat mass. This was accompanied by increased T-cell and macrophage infiltration. The expression of the same networks and biomarkers, particularly those related to T-cells, further increased in adipose tissue of B6 mice, but only minimally in 129 mice, in response to weight gain promoted by age or high-fat diet, further exacerbating the differences between strains. CONCLUSIONS: Insulin resistance in mice with differential susceptibility to diabetes and metabolic syndrome is preceded by differences in the inflammatory response of adipose tissue. This phenomenon may serve as an early indicator of disease and contribute to disease susceptibility and progression.


Subject(s)
Inflammation/physiopathology , Insulin Resistance/physiology , Metabolic Diseases/etiology , Animals , Diabetes Mellitus, Type 2/epidemiology , Disease Progression , Energy Intake , Energy Metabolism , Environment , Flow Cytometry , Genetic Predisposition to Disease , Humans , Inflammation/complications , Inflammation/genetics , Insulin Resistance/genetics , Metabolic Diseases/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Overweight/epidemiology , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , World Health Organization
17.
Nat Med ; 14(7): 778-82, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18587407

ABSTRACT

Despite the well-documented association between gallstones and the metabolic syndrome, the mechanistic links between these two disorders remain unknown. Here we show that mice solely with hepatic insulin resistance, created by liver-specific disruption of the insulin receptor (LIRKO mice) are markedly predisposed toward cholesterol gallstone formation due to at least two distinct mechanisms. Disinhibition of the forkhead transcription factor FoxO1, increases expression of the biliary cholesterol transporters Abcg5 and Abcg8, resulting in an increase in biliary cholesterol secretion. Hepatic insulin resistance also decreases expression of the bile acid synthetic enzymes, particularly Cyp7b1, and produces partial resistance to the farnesoid X receptor, leading to a lithogenic bile salt profile. As a result, after twelve weeks on a lithogenic diet, all of the LIRKO mice develop gallstones. Thus, hepatic insulin resistance provides a crucial link between the metabolic syndrome and increased cholesterol gallstone susceptibility.


Subject(s)
Cholelithiasis/metabolism , Insulin Resistance/genetics , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bile Acids and Salts/biosynthesis , Cholelithiasis/chemically induced , Cholelithiasis/genetics , Cholesterol/metabolism , Cholesterol, Dietary/administration & dosage , Cytochrome P450 Family 7 , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Male , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Animal , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
18.
Mol Cell Biol ; 27(19): 6818-31, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17646392

ABSTRACT

TRB3 has been implicated in the regulation of several biological processes in mammalian cells through its ability to influence Akt and other signaling pathways. In this study, we investigated the role of TRB3 in regulating adipogenesis and the activity of adipogenic transcription factors. We find that TRB3 is expressed in 3T3-L1 preadipocytes, and this expression is transiently suppressed during the initial days of differentiation concomitant with induction of C/EBPbeta. This event appears to be a prerequisite for adipogenesis. Overexpression of TRB3 blocks differentiation of 3T3-L1 cells at a step downstream of C/EBPbeta. Ectopic expression of TRB3 in mouse fibroblasts also inhibits the C/EBPbeta-dependent induction of PPARgamma2 and blocks their differentiation into adipocytes. This inhibition of preadipocyte differentiation by TRB3 appears to be the result of two complementary effects. First, TRB3 inhibits extracellular signal-regulated kinase activity, which prevents the phosphorylation of regulatory sites on C/EBPbeta. Second, TRB3 directly interacts with the DR1 domain of C/EBPbeta in the nucleus, further inhibiting both its ability to bind its response element and its ability to transactivate the C/EBPalpha and a-FABP promoters. Thus, TRB3 is an important negative regulator of adipogenesis that acts at an early step in the differentiation cascade to block the C/EBPbeta proadipogenic function.


Subject(s)
Adipocytes/physiology , Adipogenesis/physiology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation/physiology , Transcription, Genetic , 3T3-L1 Cells , Adipocytes/cytology , Animals , Biomarkers/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Cycle Proteins/genetics , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation , Mice , Signal Transduction/physiology
19.
J Biol Chem ; 280(12): 11432-8, 2005 Mar 25.
Article in English | MEDLINE | ID: mdl-15644333

ABSTRACT

CCAAT/enhancer-binding protein beta (C/EBP beta) is expressed early during the adipocyte differentiation program and plays an important role in this process. In an attempt to identify novel proteins that interact with C/EBP beta, we performed a yeast two-hybrid screen with a preadipocyte cDNA library and identified a new co-regulator, delta-interacting protein A (DIPA). DIPA mRNA is expressed during adipocyte differentiation of clonal cell lines. DIPA interacts with C/EBP beta and -delta proteins in intact cells and inhibits their transcriptional activity but not that of C/EBP alpha. Stable overexpression of DIPA in preadipocytes partially inhibits adipocyte differentiation, whereas its gene silencing enhances this process. DIPA and C/EBP beta co-localize in the nucleus, and overexpression of DIPA in preadipocytes results in a partial inhibition of the mitotic clonal expansion which is critical for differentiation. Thus, DIPA is a novel partner of C/EBP beta that down-regulates early events of adipogenesis.


Subject(s)
Adipocytes/cytology , CCAAT-Enhancer-Binding Protein-beta/antagonists & inhibitors , Carrier Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Differentiation , Cells, Cultured , Gene Silencing , Mice , Mitosis , NIH 3T3 Cells , PPAR gamma/metabolism , Promoter Regions, Genetic , Repressor Proteins , Transcription, Genetic , Two-Hybrid System Techniques
20.
Exp Cell Res ; 293(2): 219-28, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14729459

ABSTRACT

Adipocyte differentiation involves dramatic cell shape alterations that are accompanied by changes in the expression of cytoskeletal and extracellular matrix (ECM) proteins. Aortic carboxypeptidase-like protein (ACLP) is a secreted protein associated with the extracellular matrix whose expression is induced during smooth muscle (SM) differentiation. We analyzed the expression of ACLP gene during adipocyte differentiation of 3T3-F442A, 3T3-L1, and Ob1771 preadipocytes. Our results show that ACLP mRNA and protein are expressed in growing cells and after commitment. Thereafter, their expression levels decrease, as opposed to that of aP2 and PPARgamma2. Consistent with these observations, ACLP mRNA is expressed in the stromal-vascular fraction of adipose tissue but not in the adipocyte fraction. Overexpression of ACLP in 3T3-F442A preadipocytes inhibits adipocyte differentiation at both morphological and molecular level. However, ACLP overexpression promotes transdifferentiation of preadipocytes into smooth muscle-like cells, which express specific markers such as SM22alpha, SM alpha-actin, SM-MHC, and caldesmon. These findings demonstrate that overexpression of a single extracellular matrix protein is sufficient to induce transdifferentiation and that ACLP may modulate the commitment of mesodermal cells into different lineages depending upon its pattern of expression.


Subject(s)
Adipocytes/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Mesenchymal Stem Cells/metabolism , Muscle, Smooth/metabolism , Neoplasm Proteins , Nerve Tissue Proteins , Proteins/metabolism , Actins/metabolism , Adipocytes/cytology , Animals , Biomarkers , Calmodulin-Binding Proteins/metabolism , Carboxypeptidases , Carrier Proteins/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Gene Expression Regulation, Developmental/genetics , Mesenchymal Stem Cells/cytology , Mice , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Smooth/cytology , Myosin Heavy Chains/metabolism , NIH 3T3 Cells , Proteins/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins , Transcription Factors/metabolism , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...