Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Sci Rep ; 14(1): 563, 2024 01 04.
Article En | MEDLINE | ID: mdl-38177175

Polycystic ovary syndrome (PCOS) is associated with symptoms of moderate to severe anxiety and depression. Hyperandrogenism is a key feature together with lower levels of the adipocyte hormone adiponectin. Androgen exposure leads to anxiety-like behavior in female offspring while adiponectin is reported to be anxiolytic. Here we test the hypothesis that elevated adiponectin levels protect against the development of androgen-induced anxiety-like behavior. Pregnant mice overexpressing adiponectin (APNtg) and wildtypes were injected with vehicle or dihydrotestosterone to induce prenatal androgenization (PNA) in the offspring. Metabolic profiling and behavioral tests were performed in 4-month-old female offspring. PNA offspring spent more time in the closed arms of the elevated plus maze, indicating anxiety-like behavior. Intriguingly, neither maternal nor offspring adiponectin overexpression prevented an anxiety-like behavior in PNA-exposed offspring. However, adiponectin overexpression in dams had metabolic imprinting effects, shown as lower fat mass and glucose levels in their offspring. While serum adiponectin levels were elevated in APNtg mice, cerebrospinal fluid levels were similar between genotypes. Adiponectin overexpression improved metabolic functions but did not elicit anxiolytic effects in PNA-exposed offspring. These observations might be attributed to increased circulating but unchanged cerebrospinal fluid adiponectin levels in APNtg mice. Thus, increased adiponectin levels in the brain are likely needed to stimulate anxiolytic effects.


Anti-Anxiety Agents , Polycystic Ovary Syndrome , Prenatal Exposure Delayed Effects , Pregnancy , Humans , Mice , Female , Animals , Polycystic Ovary Syndrome/metabolism , Androgens/adverse effects , Adiponectin , Anti-Anxiety Agents/adverse effects , Anxiety/metabolism , Prenatal Exposure Delayed Effects/chemically induced
2.
Mol Med ; 29(1): 138, 2023 Oct 20.
Article En | MEDLINE | ID: mdl-37864157

OBJECTIVE: Non-alcoholic fatty liver disease (NAFLD), the primary hepatic consequence of obesity, is affecting about 25% of the global adult population. The aim of this study was to examine the in vivo role of STE20-type protein kinase TAOK3, which has been previously reported to regulate hepatocellular lipotoxicity in vitro, in the development of NAFLD and systemic insulin resistance in the context of obesity. METHODS: Taok3 knockout mice and wild-type littermates were challenged with a high-fat diet. Various in vivo tests were performed to characterize the whole-body metabolism. NAFLD progression in the liver, and lipotoxic damage in adipose tissue, kidney, and skeletal muscle were compared between the genotypes by histological assessment, immunofluorescence microscopy, protein and gene expression profiling, and biochemical assays. Intracellular lipid accumulation and oxidative/ER stress were analyzed in cultured human and mouse hepatocytes where TAOK3 was knocked down by small interfering RNA. The expression of TAOK3-related STE20-type kinases was quantified in different organs from high-fat diet-fed Taok3-/- and wild-type mice. RESULTS: TAOK3 deficiency had no impact on body weight or composition, food consumption, locomotor activity, or systemic glucose or insulin homeostasis in obese mice. Consistently, Taok3-/- mice and wild-type littermates developed a similar degree of high-fat diet-induced liver steatosis, inflammation, and fibrosis, and we detected no difference in lipotoxic damage of adipose tissue, kidney, or skeletal muscle when comparing the two genotypes. In contrast, the silencing of TAOK3 in vitro markedly suppressed ectopic lipid accumulation and metabolic stress in mouse and human hepatocytes. Interestingly, the hepatic mRNA abundance of several TAOK3-related kinases, which have been previously implicated to increase the risk of NAFLD susceptibility, was significantly elevated in Taok3-/- vs. wild-type mice. CONCLUSIONS: In contrast to the in vitro observations, genetic deficiency of TAOK3 in mice failed to mitigate the detrimental metabolic consequences of chronic exposure to dietary lipids, which may be partly attributable to the activation of liver-specific compensation response for the genetic loss of TAOK3 by related STE20-type kinases.


Non-alcoholic Fatty Liver Disease , Adult , Animals , Humans , Mice , Diet, High-Fat/adverse effects , Lipids , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/etiology , Obesity/metabolism
3.
J Cell Biochem ; 124(3): 382-395, 2023 03.
Article En | MEDLINE | ID: mdl-36715685

Brown adipose tissue (BAT) consists of highly metabolically active adipocytes that catabolize nutrients to produce heat. Playing an active role in triacylglycerol (TAG) clearance, research has shown that dietary fatty acids can modulate the TAG chemistry deposition in BAT after weeks-long dietary intervention, similar to what has been shown in white adipose tissue (WAT). Our objective was to compare the influence of sustained, nonchronic dietary intervention (a 1-week interval) on WAT and interscapular BAT lipid metabolism and deposition in situ. We use quantitative, label-free chemical microscopy to show that 1 week of high fat diet (HFD) intervention results in dramatically larger lipid droplet (LD) growth in BAT (and liver) compared to LD growth in inguinal WAT (IWAT). Moreover, BAT showed lipid remodeling as increased unsaturated TAGs in LDs, resembling the dietary lipid composition, while WAT (and liver) did not show lipid remodeling on this time scale. Concurrently, expression of genes involved in lipid metabolism, particularly desaturases, was reduced in BAT and liver from HFD-fed mice after 1 week. Our data show that BAT lipid chemistry remodels exceptionally fast to dietary lipid intervention compared WAT, which further points towards a role in TAG clearance.


Adipose Tissue, Brown , Diet, High-Fat , Mice , Animals , Adipose Tissue, Brown/metabolism , Diet, High-Fat/adverse effects , Microscopy , Adipose Tissue, White/metabolism , Liver/metabolism , Dietary Fats , Adipose Tissue , Mice, Inbred C57BL
4.
Mol Metab ; 39: 101010, 2020 09.
Article En | MEDLINE | ID: mdl-32408016

OBJECTIVE: Obesity-induced insulin resistance is closely associated with chronic subclinical inflammation in white adipose tissue. However, the mechanistic involvement of adipocyte-derived inflammation under these disease conditions remains unclear. Our aim was to investigate the relative inflammation-related contributions of adipocytes and macrophages to insulin sensitivity. METHODS: RIDα/ß is an adenoviral protein complex that inhibits several inflammatory pathways, including TLR4, TNFα, and IL1ß signaling. We generated novel mouse models with adipocyte-specific and macrophage-specific doxycycline (dox)-inducible RIDα/ß-transgenic mice (RIDad and RIDmac mice, respectively). RESULTS: RIDα/ß induction significantly reduced LPS-stimulated inflammatory markers, such as Tnf, Il1b, and Saa3 in adipose tissues. Surprisingly, RIDad mice had elevated levels of postprandial glucose and insulin and exhibited glucose intolerance and insulin resistance, even under chow-fed conditions. Moreover, the RIDad mice displayed further insulin resistance under obesogenic (high-fat diet, HFD) conditions despite reduced weight gain. In addition, under pre-existing obese and inflamed conditions on an HFD, subsequent induction of RIDα/ß in RIDad mice reduced body weight gain, further exacerbating glucose tolerance, enhancing insulin resistance and fatty liver, and reducing adiponectin levels. This occurred despite effective suppression of the inflammatory pathways (including TNFα and IL1ß). In contrast, RIDmac mice, upon HFD feeding, displayed similar weight gain, comparable adiponectin levels, and insulin sensitivity, suggesting that the inflammatory properties of macrophages did not exert a negative impact on metabolic readouts. RIDα/ß expression and the ensuing suppression of inflammation in adipocytes enhanced adipose tissue fibrosis and reduced vascularization. CONCLUSION: Our novel findings further corroborate our previous observations suggesting that suppressing adipocyte inflammation impairs adipose tissue function and promotes insulin resistance, despite beneficial effects on weight gain.


Adipocytes/metabolism , Adipose Tissue/metabolism , Inflammation/metabolism , Insulin Resistance , Insulin/metabolism , Animals , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Glucose/metabolism , Glucose Intolerance/metabolism , Immunohistochemistry , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Inflammation Mediators/metabolism , Macrophages/metabolism , Mice , Mice, Transgenic , Obesity/etiology , Obesity/metabolism , Obesity/pathology
5.
Nat Commun ; 10(1): 139, 2019 01 11.
Article En | MEDLINE | ID: mdl-30635569

Hypoglycaemia (low plasma glucose) is a serious and potentially fatal complication of insulin-treated diabetes. In healthy individuals, hypoglycaemia triggers glucagon secretion, which restores normal plasma glucose levels by stimulation of hepatic glucose production. This counterregulatory mechanism is impaired in diabetes. Here we show in mice that therapeutic concentrations of insulin inhibit glucagon secretion by an indirect (paracrine) mechanism mediated by stimulation of intra-islet somatostatin release. Insulin's capacity to inhibit glucagon secretion is lost following genetic ablation of insulin receptors in the somatostatin-secreting δ-cells, when insulin-induced somatostatin secretion is suppressed by dapagliflozin (an inhibitor of sodium-glucose co-tranporter-2; SGLT2) or when the action of secreted somatostatin is prevented by somatostatin receptor (SSTR) antagonists. Administration of these compounds in vivo antagonises insulin's hypoglycaemic effect. We extend these data to isolated human islets. We propose that SSTR or SGLT2 antagonists should be considered as adjuncts to insulin in diabetes therapy.


Diabetes Mellitus/pathology , Glucagon/metabolism , Hypoglycemia/pathology , Insulin/metabolism , Sodium-Glucose Transporter 2/metabolism , Somatostatin/metabolism , Animals , Benzhydryl Compounds/pharmacology , Blood Glucose/analysis , Diabetes Mellitus/drug therapy , Female , Glucagon-Secreting Cells/drug effects , Glucosides/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Insulin/genetics , Receptors, Somatostatin/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
6.
Nat Metab ; 1(12): 1243-1257, 2019 12.
Article En | MEDLINE | ID: mdl-31984308

Mitochondrial function in white adipose tissue (WAT) is an important yet understudied aspect in adipocyte biology. Here, we report a role for amyloid precursor protein (APP) in compromising WAT mitochondrial function through a high-fat diet (HFD)-induced, unconventional mis-localization to mitochondria that further promotes obesity. In humans and mice, obese conditions significantly induce APP production in WAT and its enrichment in mitochondria. Mechanistically, a HFD-induced dysregulation of signal recognition particle subunit 54c is responsible for the mis-targeting of APP to adipocyte mitochondria. Mis-localized APP blocks the protein import machinery, leading to mitochondrial dysfunction in WAT. Adipocyte-specific and mitochondria-targeted APP overexpressing mice display increased body mass and reduced insulin sensitivity, along with dysfunctional WAT due to a dramatic hypertrophic program in adipocytes. Elimination of adipocyte APP rescues HFD-impaired mitochondrial function with significant protection from weight gain and systemic metabolic deficiency. Our data highlights an important role of APP in modulating WAT mitochondrial function and obesity-associated metabolic dysfunction.


Adipose Tissue/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Mitochondria/metabolism , Obesity/genetics , Obesity/metabolism , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Adult , Animals , Body Weight , Cell Size , Diet, High-Fat , Fatty Liver/metabolism , Female , HEK293 Cells , Humans , Insulin Resistance/genetics , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Weight Gain
8.
J Lipid Res ; 53(7): 1254-67, 2012 Jul.
Article En | MEDLINE | ID: mdl-22504909

Subclinical inflammation is frequently associated with obesity. Here, we aim to better define the acute inflammatory response during fasting. To do so, we analyzed representatives of immune-related proteins in circulation and in tissues as potential markers for adipose tissue inflammation and modulation of the immune system. Lipopolysaccharide treatment or high-fat diet led to an increase in circulating serum amyloid (SAA) and α1-acid glycoprotein (AGP), whereas adipsin levels were reduced. Mouse models that are protected against diet-induced challenges, such as adiponectin-overexpressing animals or mice treated with PPARγ agonists, displayed lower SAA levels and higher adip-sin levels. An oral lipid gavage, as well as prolonged fasting, increased circulating SAA concurrent with the elevation of free FA levels. Moreover, prolonged fasting was associated with an increased number of Mac2-positive crown-like structures, an increased capillary permeability, and an increase in several M2-type macrophage markers in adipose tissue. This fasting-induced increase in SAA and M2-type macrophage markers was impaired in metabolically challenged animals. These data suggest that metabolic inflexibility is associated with a lack of "immunological fitness."


Fasting/metabolism , Amyloid/blood , Animals , Biomarkers/blood , Diet, High-Fat/adverse effects , Fasting/physiology , Inflammation , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred Strains , Orosomucoid/analysis
9.
Exp Physiol ; 97(11): 1224-35, 2012 Nov.
Article En | MEDLINE | ID: mdl-22523382

Interleukin-6 (IL-6) is released from working skeletal muscle during exercise. We investigated the acute and the long-term beneficial effects of IL-6 on exercise-induced glucose uptake in skeletal muscle and insulin sensitivity. The acute effect on exercise-induced glucose uptake was measured in IL-6-deficient (IL-6(-/-)) mice and wild-type control animals using a tracer technique. There was no difference in serum disappearance of (3)[H]2-deoxyglucose after a single bout of exercise between IL-6(-/-) and wild-type mice (13565 ± 426 versus 14343 ± 1309 d.p.m. min ml(-1), P = 0.5). The glucose uptake rate in the extensor digitorum longus muscle was, however, lower in IL-6(-/-) compared with wild-type mice (398 ± 44 versus 657 ± 41 nmol g(-1) min(-1), P < 0.01). In a long-term study, we monitored insulin sensitivity, serum retinol-binding protein-4 (RBP-4) levels, running activity, food intake, body weight and body composition in IL-6(-/-) and wild-type mice on a high-fat diet (HFD), with or without access to running wheels. In sedentary IL-6(-/-) and wild-type mice, the HFD decreased insulin sensitivity (glucose area under the concentration-time curve increased about 20% during an insulin tolerance test, P < 0.05 for both genotypes versus baseline) and led to a 30% increase in serum RBP-4 levels (P < 0.01 for both genotypes versus baseline). Wild-type mice with access to running wheels were protected against these effects of the HFD and maintained their baseline insulin sensitivity and serum RBP-4 levels. In contrast, IL-6(-/-) mice did not benefit from running to the same extent as wild-type animals. The IL-6(-/-) mice with access to running wheels had a similar decrease in insulin sensitivity to their sedentary littermates (glucose area under the concentration-time curve during an insulin tolerance test in runners versus sedentary IL-6(-/-) HFD mice, 312 ± 14 versus 340 ± 22 mmol min l(-1), P = 0.4) and displayed a 14% increase in serum RBP-4 compared with baseline levels (P < 0.01). Our results indicate that endogenous IL-6 contributes to the exercise-induced increase in insulin sensitivity, but plays only a minor role for glucose uptake into skeletal muscle during exercise.


Insulin Resistance/physiology , Insulin/metabolism , Interleukin-6/metabolism , Physical Conditioning, Animal/physiology , Animals , Body Composition/physiology , Body Weight/physiology , Diet, High-Fat/methods , Eating/physiology , Female , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Retinol-Binding Proteins, Plasma/metabolism , Running/physiology
10.
Cell Metab ; 15(2): 171-85, 2012 Feb 08.
Article En | MEDLINE | ID: mdl-22326219

Caveolin-1 is a major structural component of raft structures within the plasma membrane and has been implicated as a regulator of cellular signal transduction with prominent expression in adipocytes. Here, we embarked on a comprehensive characterization of the metabolic pathways dysregulated in caveolin-1 null mice. We found that these mice display decreased circulating levels of total and high molecular weight adiponectin and a reduced ability to change substrate use in response to feeding/fasting conditions. Caveolin-1 null mice are extremely lean but retain muscle mass despite lipodystrophy and massive metabolic dysfunction. Hepatic gluconeogenesis is chronically elevated, while hepatic steatosis is reduced. Our data suggest that the complex phenotype of the caveolin-1 null mouse is caused by altered metabolic and mitochondrial function in adipose tissue with a subsequent compensatory response driven mostly by the liver. This mouse model highlights the central contributions of adipose tissue for system-wide preservation of metabolic flexibility.


Adipose Tissue/metabolism , Caveolin 1/deficiency , Liver/metabolism , Metabolic Networks and Pathways/physiology , Mitochondria/metabolism , Adiponectin/blood , Animals , Fatty Acids, Nonesterified/metabolism , Fatty Liver/diagnostic imaging , Gluconeogenesis/physiology , Histological Techniques , Mice , Tomography, X-Ray Computed
11.
Endocrinology ; 152(8): 3074-81, 2011 Aug.
Article En | MEDLINE | ID: mdl-21693678

Obesity is frequently associated with an infiltration of macrophages into adipose tissue. Adipocyte dysfunction causes a phenotypic switch of macrophages from an alternatively activated M2-like phenotype towards a proinflammatory M1 phenotype. The cross talk between adipocytes and infiltrating immune cells, in particular macrophages, is thought to contribute to local and eventually systemic inflammation. Here, we tested the phenotypic impact of a lack of adipocytes on the inflammatory status of macrophages. We took advantage of the fat apoptosis through targeted activation of caspase-8 (FAT-ATTAC) mouse model that allows for the inducible system-wide elimination of adipocytes through a proapoptotic mechanism and followed the degree and type of inflammatory response upon ablation of live adipocytes. Analysis of depots 2 wk after elimination of adipocytes resulted in markedly reduced levels of adipose tissue and a robust down-regulation of circulating adipokines. Quantitative PCR and immunohistochemistry on epididymal and inguinal fat depots revealed an increase of the macrophage markers F4/80 and CD11c. Using polychromatic flow cytometry, we observed an up-regulation of alternatively activated M2 macrophage markers (CD206 and CD301) on the majority of F4/80 positive cells. Apoptosis of adipocytes is sufficient to initiate a large influx of macrophages into the remnant fat pads. However, these macrophages are alternatively activated, antiinflammatory M2 macrophages and not M1 cells. We conclude that adipocyte death is sufficient to initiate macrophage infiltration, and live adipocytes are required to initiate and/or sustain a proinflammatory response within the infiltrating macrophages in adipose tissue.


Adipocytes/physiology , Adipose Tissue/physiology , Apoptosis , Macrophage Activation , Macrophages/physiology , Animals , Antigens, Differentiation/analysis , Antigens, Differentiation/genetics , Asialoglycoproteins/analysis , CD11c Antigen/analysis , CD11c Antigen/genetics , Cell Polarity , Inflammation/etiology , Lectins, C-Type/analysis , Male , Mannose Receptor , Mannose-Binding Lectins/analysis , Membrane Proteins/analysis , Mice , Receptors, Cell Surface/analysis
12.
Endocrinology ; 152(4): 1336-46, 2011 Apr.
Article En | MEDLINE | ID: mdl-21285312

The perinatal environment appears important in establishing metabolic phenotypes in adulthood. Mice deficient in IL-6 (IL-6(-/-)) tend to develop mature-onset obesity, but it is unknown whether perinatal exposure to IL-6 produced by the dam influences the metabolism of adult offspring. To address this issue, we monitored IL-6(-/-) offspring of IL-6(-/-) or IL-6(+/-) dams, as well as wild-type (WT) mice. At adult age, IL-6(-/-) mice weighed significantly more and had more body fat than WT mice, regardless of maternal genotype, and had lower insulin sensitivity. This phenotype was more pronounced in IL-6(-/-) offspring of IL-6(-/-) dams, because they gained weight significantly faster than IL-6(-/-) offspring of IL-6(+/-) dams and had more body fat and higher serum leptin levels at an earlier age. The leptin content was 2-fold higher in milk from IL-6(-/-) than WT dams. However, cross-fostering IL-6(-/-) mice with WT dams did not alter body weight, body composition, or adipocyte size at adult age compared with IL-6(-/-) mice fostered by IL-6(-/-) dams. Conversely, WT mice fostered by IL-6(-/-) dams weighed significantly more than those fostered by WT dams and had more body fat, larger adipocytes, and altered hypothalamic gene expression. We conclude that body fat of adult mice can be increased by perinatal exposure to factors affected by lack of maternal IL-6.


Adiposity/physiology , Body Composition/physiology , Interleukin-6/deficiency , Interleukin-6/physiology , Adipocytes/cytology , Adipocytes/metabolism , Adiposity/genetics , Animals , Body Composition/genetics , Body Weight/genetics , Body Weight/physiology , Enzyme-Linked Immunosorbent Assay , Female , Glucose Clamp Technique , Hypothalamus/metabolism , Interleukin-6/genetics , Leptin/blood , Male , Mice , Mice, Mutant Strains , Milk/chemistry , Pregnancy , RNA, Messenger
13.
Dis Model Mech ; 3(9-10): 567-80, 2010.
Article En | MEDLINE | ID: mdl-20616094

Diabetes is characterized by the loss, or gradual dysfunction, of insulin-producing pancreatic beta-cells. Although beta-cells can replicate in younger adults, the available diabetes therapies do not specifically target beta-cell regeneration. Novel approaches are needed to discover new therapeutics and to understand the contributions of endocrine progenitors and beta-cell regeneration during islet expansion. Here, we show that the regulators of G protein signaling Rgs16 and Rgs8 are expressed in pancreatic progenitor and endocrine cells during development, then extinguished in adults, but reactivated in models of both type 1 and type 2 diabetes. Exendin-4, a glucagon-like peptide 1 (Glp-1)/incretin mimetic that stimulates beta-cell expansion, insulin secretion and normalization of blood glucose levels in diabetics, also promoted re-expression of Rgs16::GFP within a few days in pancreatic ductal-associated cells and islet beta-cells. These findings show that Rgs16::GFP and Rgs8::GFP are novel and early reporters of G protein-coupled receptor (GPCR)-stimulated beta-cell expansion after therapeutic treatment and in diabetes models. Rgs16 and Rgs8 are likely to control aspects of islet progenitor cell activation, differentiation and beta-cell expansion in embryos and metabolically stressed adults.


Diabetes Mellitus, Type 1/embryology , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/embryology , Islets of Langerhans/pathology , RGS Proteins/metabolism , Aging/drug effects , Aging/pathology , Animals , Animals, Newborn , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Disease Models, Animal , Exenatide , Female , Gene Expression Regulation, Developmental/drug effects , Green Fluorescent Proteins/metabolism , Hyperglycemia/complications , Hyperglycemia/pathology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mice, Obese , Peptides/pharmacology , Pregnancy , RGS Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regeneration/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Venoms/pharmacology
14.
Am J Pathol ; 176(3): 1364-76, 2010 Mar.
Article En | MEDLINE | ID: mdl-20093494

Metabolically healthy individuals effectively adapt to changes in nutritional state. Here, we focus on the effects of the adipocyte-derived secretory molecule adiponectin on adipose tissue in mouse models with genetically altered adiponectin levels. We found that higher adiponectin levels increased sensitivity to the lipolytic effects of adrenergic receptor agonists. In parallel, adiponectin-overexpressing mice also display enhanced clearance of circulating fatty acids and increased expansion of subcutaneous adipose tissue with chronic high fat diet (HFD) feeding. These adaptive changes to the HFD were associated with increased mitochondrial density in adipocytes, smaller adipocyte size, and a general transcriptional up-regulation of factors involved in lipid storage through efficient esterification of free fatty acids. The physiological response to adiponectin overexpression resembles in many ways the effects of chronic exposure to beta3-adrenergic agonist treatment, which also results in improvements in insulin sensitivity. In addition, using a novel computed tomography-based method for measurements of hepatic lipids, we resolved the temporal events taking place in the liver in response to acute HFD exposure in both wild-type and adiponectin-overexpressing mice. Increased levels of adiponectin potently protect against HFD-induced hepatic lipid accumulation and preserve insulin sensitivity. Given these profound effects of adiponectin, we propose that adiponectin is a factor that increases the metabolic flexibility of adipose tissue, enhancing its ability to maintain proper function under metabolically challenging conditions.


Adiponectin/metabolism , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Cell Respiration , Cyclic AMP Response Element-Binding Protein/metabolism , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Fatty Acids, Nonesterified/blood , Fatty Liver/blood , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Gene Expression Regulation/drug effects , Glycerol/blood , Insulin/blood , Liver/diagnostic imaging , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Size/drug effects , Phosphorylation/drug effects , Receptors, Adrenergic, beta-3/metabolism , Tomography, X-Ray Computed , Triglycerides/metabolism , Weight Gain/drug effects
15.
Drug Discov Today Dis Models ; 4(1): 17-24, 2007.
Article En | MEDLINE | ID: mdl-18193096

Both the disproportionate loss of adipose tissue in the case of lipodystrophies and the disproportionate gain of adipose tissue in obesity are frequently associated with an increase in insulin resistance and its complications. Leptin replacement is a very promising therapeutic approach for the management of the complications of lipodystrophy. In contrast, leptin treatment for the reversal of obesity-related metabolic disorders has not proven to be successful. There is a need to better understand both of these phenomena. Mouse models of lipodystrophy may provide us with new pharmaceutical targets for the treatment and prevention of metabolic disturbances related to dysfunctional adipose tissue both in the context of lipodystrophy and obesity.

...