Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.283
Filter
Add more filters

Publication year range
1.
Nature ; 625(7993): 175-180, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38093006

ABSTRACT

Oxytocin (OXT), a nine-amino-acid peptide produced in the hypothalamus and released by the posterior pituitary, has well-known actions in parturition, lactation and social behaviour1, and has become an intriguing therapeutic target for conditions such as autism and schizophrenia2. Exogenous OXT has also been shown to have effects on body weight, lipid levels and glucose homeostasis1,3, suggesting that it may also have therapeutic potential for metabolic disease1,4. It is unclear, however, whether endogenous OXT participates in metabolic homeostasis. Here we show that OXT is a critical regulator of adipose tissue lipolysis in both mice and humans. In addition, OXT serves to facilitate the ability of ß-adrenergic agonists to fully promote lipolysis. Most surprisingly, the relevant source of OXT in these metabolic actions is a previously unidentified subpopulation of tyrosine hydroxylase-positive sympathetic neurons. Our data reveal that OXT from the peripheral nervous system is an endogenous regulator of adipose and systemic metabolism.


Subject(s)
Adipose Tissue , Lipolysis , Neurons , Oxytocin , Animals , Humans , Mice , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adrenergic beta-Agonists/pharmacology , Lipolysis/drug effects , Neurons/metabolism , Oxytocin/metabolism , Oxytocin/pharmacology , Tyrosine 3-Monooxygenase/metabolism
2.
Nature ; 579(7798): 279-283, 2020 03.
Article in English | MEDLINE | ID: mdl-32132708

ABSTRACT

Although it is well-established that reductions in the ratio of insulin to glucagon in the portal vein have a major role in the dysregulation of hepatic glucose metabolism in type-2 diabetes1-3, the mechanisms by which glucagon affects hepatic glucose production and mitochondrial oxidation are poorly understood. Here we show that glucagon stimulates hepatic gluconeogenesis by increasing the activity of hepatic adipose triglyceride lipase, intrahepatic lipolysis, hepatic acetyl-CoA content and pyruvate carboxylase flux, while also increasing mitochondrial fat oxidation-all of which are mediated by stimulation of the inositol triphosphate receptor 1 (INSP3R1). In rats and mice, chronic physiological increases in plasma glucagon concentrations increased mitochondrial oxidation of fat in the liver and reversed diet-induced hepatic steatosis and insulin resistance. However, these effects of chronic glucagon treatment-reversing hepatic steatosis and glucose intolerance-were abrogated in Insp3r1 (also known as Itpr1)-knockout mice. These results provide insights into glucagon biology and suggest that INSP3R1 may represent a target for therapies that aim to reverse nonalcoholic fatty liver disease and type-2 diabetes.


Subject(s)
Glucagon/pharmacology , Gluconeogenesis/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Liver/drug effects , Acetyl Coenzyme A/metabolism , Adipose Tissue/drug effects , Animals , Diabetes Mellitus, Type 2/physiopathology , Enzyme Activation/drug effects , Glucagon/blood , Inositol 1,4,5-Trisphosphate Receptors/genetics , Lipase/metabolism , Lipolysis/drug effects , Lipolysis/genetics , Mice, Knockout , Mitochondria/drug effects , Non-alcoholic Fatty Liver Disease/physiopathology , Oxidation-Reduction/drug effects
3.
Am J Pathol ; 194(8): 1494-1510, 2024 08.
Article in English | MEDLINE | ID: mdl-38705384

ABSTRACT

Dyslipolysis of adipocytes plays a critical role in various diseases. Adipose triglyceride lipase (ATGL) is a rate-limiting enzyme in adipocyte autonomous lipolysis. However, the degree of adipocyte lipolysis related to the prognoses in acute pancreatitis (AP) and the role of ATGL-mediated lipolysis in the pathogenesis of AP remain elusive. Herein, the visceral adipose tissue consumption rate in the acute stage was measured in both patients with AP and mouse models. Lipolysis levels and ATGL expression were detected in cerulein-induced AP models. CL316,243, a lipolysis stimulator, and adipose tissue-specific ATGL knockout mice were used to further investigate the role of lipolysis in AP. The ATGL-specific inhibitor, atglistatin, was used in C57Bl/6N and ob/ob AP models. This study indicated that increased visceral adipose tissue consumption rate in the acute phase was independently associated with adverse prognoses in patients with AP, which was validated in mouse AP models. Lipolysis of adipocytes was elevated in AP mice. Stimulation of lipolysis aggravated AP. Genetic blockage of ATGL specifically in adipocytes alleviated the damage to AP. The application of atglistatin effectively protected against AP in both lean and obese mice. These findings demonstrated that ATGL-mediated adipocyte lipolysis exacerbates AP and highlighted the therapeutic potential of ATGL as a drug target for AP.


Subject(s)
Adipocytes , Lipase , Lipolysis , Pancreatitis , Animals , Female , Humans , Male , Mice , Acute Disease , Acyltransferases , Adipocytes/metabolism , Adipocytes/pathology , Disease Models, Animal , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Lipase/metabolism , Lipase/genetics , Lipolysis/drug effects , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/pathology , Pancreatitis/metabolism
4.
J Lipid Res ; 65(8): 100598, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39032560

ABSTRACT

All-trans retinoic acid (atRA), a metabolite of vitamin A, reduces hepatic lipid accumulation in liver steatosis model animals. Lipophagy, a new lipolysis pathway, degrades a lipid droplet (LD) via autophagy in adipose tissue and the liver. We recently found that atRA induces lipophagy in adipocytes. However, it remains unclear whether atRA induces lipophagy in hepatocytes. In this study, we investigated the effects of atRA on lipophagy in Hepa1c1c7 cells and the liver of mice fed a high-fat diet (HFD). First, we confirmed that atRA induced autophagy in Hepa1c1c7 cells by Western blotting and the GFP-LC3-mCherry probe. Next, we evaluated the lipolysis in fatty Hepa1c1c7 cells treated with the knockdown of Atg5, an essential gene in autophagy induction. Atg5-knockdown partly suppressed the atRA-induced lipolysis in fatty Hepa1c1c7 cells. We also found that atRA reduced the protein, but not mRNA, expression of Rubicon, a negative regulator of autophagy, in Hepa1c1c7 cells and the liver of HFD-fed mice. Rubicon-knockdown partly inhibited the atRA-induced lipolysis in fatty Hepa1c1c7 cells. In addition, atRA reduced hepatic Rubicon expression in young mice, but the effect of atRA on it diminished in aged mice. Finally, we investigated the mechanism underlying reduced Rubicon protein expression by atRA in hepatocytes. A protein synthesis inhibitor, but not proteasome or lysosomal inhibitors, significantly blocked the reduction of Rubicon protein expression by atRA in Hepa1c1c7 cells. These results suggest that atRA may promote lipophagy in fatty hepatocytes by reducing hepatic Rubicon expression via inhibiting protein synthesis.


Subject(s)
Autophagy , Lipolysis , Tretinoin , Animals , Tretinoin/pharmacology , Mice , Autophagy/drug effects , Lipolysis/drug effects , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/cytology , Male , Mice, Inbred C57BL , Liver/metabolism , Liver/drug effects , Cell Line, Tumor , Diet, High-Fat/adverse effects , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 5/genetics , Intracellular Signaling Peptides and Proteins
5.
J Endocrinol Invest ; 47(9): 2249-2260, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38460092

ABSTRACT

BACKGROUND: The organ-specific effects of gender-affirming sex hormone treatment (GAHT) in transgender women (TW) and transgender men (TM) are insufficiently explored. This study investigated the effects of GAHT on adipose tissue function. METHODS: In a single-center interventional prospective study, 32 adults undergoing GAHT, 15 TW and 17 TM, were examined with anthropometry and abdominal subcutaneous adipose tissue biopsies obtained before initiation of treatment, 1 month after endogenous sex hormone inhibition and three and 11 months after initiated GAHT. Fat cell size, basal/stimulated lipolysis and cytokine secretion in adipose tissue were analyzed. RESULTS: TW displayed an increase in complement component 3a and retinol-binding protein 4 (RBP4) secretion after sex hormone inhibition, which returned to baseline following estradiol treatment. No changes in lipolysis were seen in TW. TM showed downregulation of RBP4 after treatment, but no changes in basal lipolysis. In TM, the estrogen suppression led to higher noradrenaline stimulated (NA) lipolysis that was normalized following testosterone treatment. At 11 months, the ratio of NA/basal lipolysis was lower compared to baseline. There were no significant changes in fat cell size in either TW or TM. CONCLUSION: In TW, gonadal hormone suppression results in transient changes in cytokines and in TM there are some changes in NA-stimulated lipolysis following testosterone treatment. However, despite the known metabolic effects of sex hormones, the overall effects of GAHT on adipose tissue function are small and likely have limited clinical relevance, but larger studies with longer follow-up are needed to confirm these findings. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02518009, Retrospectively registered 7 August 2015.


Subject(s)
Adipokines , Lipolysis , Transgender Persons , Humans , Lipolysis/drug effects , Female , Male , Adult , Prospective Studies , Adipokines/metabolism , Sex Reassignment Procedures/methods , Testosterone , Gonadal Steroid Hormones , Middle Aged , Young Adult , Follow-Up Studies
6.
Mar Drugs ; 22(8)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39195446

ABSTRACT

This study explores the anti-obesity effects of the ethyl acetate extract of Ecklonia cava (EC-ETAC) on 3T3-L1 preadipocytes, focusing on its impact on adipogenesis, lipolysis, and adipose browning via the HO-1/Nrf2 pathway. Western blot analysis revealed that EC-ETAC significantly inhibited adipogenic transcription factors (PPARγ, C/EBPα, SREBP-1) and lipogenesis-related proteins (FAS, LPL). Concurrently, EC-ETAC enhanced lipolytic markers (p-AMPK, p-HSL) and adipose browning-related proteins (UCP-1, PGC-1α), indicating its role in promoting lipolysis and adipose browning. The inhibition of HO-1 by zinc protoporphyrin (ZnPP) significantly reversed these effects, underscoring the critical role of HO-1 in mediating the anti-obesity properties of EC-ETAC. Additionally, fluorescence measurements and Oil Red O staining confirmed the reduction of lipid accumulation and oxidative stress upon EC-ETAC treatment. These findings suggest that EC-ETAC exerts its anti-obesity effects by modulating the HO-1/Nrf2 pathway, which is crucial for regulating adipogenesis, lipolysis, and adipose browning. This study highlights the potential of EC-ETAC as a natural therapeutic agent for obesity management and supports further research into its clinical applications. By targeting the HO-1/Nrf2 pathway, EC-ETAC could offer a novel approach to enhancing energy expenditure and reducing fat mass, thereby improving metabolic health.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Anti-Obesity Agents , Heme Oxygenase-1 , NF-E2-Related Factor 2 , Phaeophyceae , Signal Transduction , Animals , Mice , Adipogenesis/drug effects , NF-E2-Related Factor 2/metabolism , Phaeophyceae/chemistry , Signal Transduction/drug effects , Adipocytes/drug effects , Adipocytes/metabolism , Heme Oxygenase-1/metabolism , Anti-Obesity Agents/pharmacology , Lipolysis/drug effects , Plant Extracts/pharmacology , Oxidative Stress/drug effects , Obesity/drug therapy , Obesity/metabolism , Membrane Proteins
7.
J Dairy Sci ; 107(7): 5104-5121, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38278290

ABSTRACT

Dairy cows with clinical ketosis (CK) exhibit excessive adipose tissue (AT) lipolysis and systemic inflammation. Lipolysis in cows can be induced by the canonical (hormonally induced) and inflammatory lipolytic pathways. Currently, the most common treatment for CK is oral propylene glycol (PG); however, PG does not reduce lipolysis or inflammation. Niacin (NIA) can reduce the activation of canonical lipolysis, whereas cyclooxygenase inhibitors such as flunixin meglumine (FM) can limit inflammation and inhibit the inflammatory lipolytic pathway. The objective of this study was to determine the effects of including NIA and FM in the standard PG treatment for postpartum CK on AT function. Multiparous Jersey cows (n = 18; 7.1 ± 3.8 DIM) were selected from a commercial dairy. Inclusion criteria were CK symptoms (lethargy, depressed appetite, and drop in milk yield) and high blood levels of BHB (≥1.2 mmol/L). Cows with CK were randomly assigned to one of 3 treatments: (1) PG: 310 g administered orally once per day for 5 d, (2) PG+NIA: 24 g administered orally once per day for 3 d, and (3) PG+NIA+FM: 1.1 mg/kg administered IV once per day for 3 d. Healthy control cows (HC; n = 6) matched by lactation and DIM (±2 d) were sampled. Subcutaneous AT explants were collected at d 0 and d 7 relative to enrollment. To assess AT insulin sensitivity, explants were treated with insulin (1 µL/L) during lipolysis stimulation with a ß-adrenergic receptor agonist (isoproterenol, 1 µM). Lipolysis was quantified by glycerol release in the media. Lipid mobilization and inflammatory gene networks were evaluated using quantitative PCR. Protein biomarkers of lipolysis, insulin signaling, and AT inflammation, including hormone-sensitive lipase, protein kinase B (Akt), and ERK1/2, were quantified by capillary immunoassays. Flow cytometry of AT cellular components was used to characterize macrophage inflammatory phenotypes. Statistical significance was determined by a nonparametric t-test when 2 groups (HC vs. CK) were analyzed and an ANOVA test with Tukey adjustment when 3 treatment groups (PG vs. PG+NIA vs. PG+NIA+FM) were evaluated. At d 0, AT from CK cows showed higher mRNA expression of lipolytic enzymes ABHD5, LIPE, and LPL, as well as increased phosphorylation of hormone-sensitive lipase compared with HC. At d 0, insulin reduced lipolysis by 41% ± 8% in AT from HC, but CK cows were unresponsive (-2.9 ± 4%). Adipose tissue from CK cows exhibited reduced Akt phosphorylation compared with HC. Cows with CK had increased AT expression of inflammatory gene markers, including CCL2, IL8, IL10, TLR4, and TNF, along with ERK1/2 phosphorylation. Adipose tissue from CK cows showed increased macrophage infiltration compared with HC. By d 7, AT from PG+NIA+FM cows had a more robust response to insulin, as evidenced by reduced glycerol release (36.5% ± 8% compared with PG at 26.9% ± 7% and PG+NIA at 7.4% ± 8%) and enhanced phosphorylation of Akt. By d 7, PG+NIA+FM cows presented lower inflammatory markers, including ERK1/2 phosphorylation, and reduced macrophage infiltration, compared with PG and PG+NIA. These data suggest that including NIA and FM in CK treatment improves AT insulin sensitivity and reduces AT inflammation and macrophage infiltration.


Subject(s)
Adipose Tissue , Cattle Diseases , Ketosis , Lipolysis , Animals , Cattle , Lipolysis/drug effects , Female , Adipose Tissue/metabolism , Ketosis/veterinary , Ketosis/drug therapy , Cattle Diseases/drug therapy , Cattle Diseases/metabolism , Lactation , Niacin/pharmacology , Niacin/therapeutic use , Postpartum Period
8.
Int J Mol Sci ; 25(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38732183

ABSTRACT

The impact of microplastics (MPs) on the metabolic functions of the liver is currently unclear and not completely understood. To investigate the effects of the administration of MPs on the hepatic metabolism of normal and obese mice, alterations in the lipid, glucose (Glu), and amino acid regulation pathways were analyzed in the liver and adipose tissues of C57BL/6Korl (wild type, WT) or C57BL/6-Lepem1hwl/Korl mice (leptin knockout, Lep KO) orally administered polystyrene (PS) MPs for 9 weeks. Significant alterations in the lipid accumulation, adipogenesis, lipogenesis, and lipolysis pathways were detected in the liver tissue of MP-treated WT and Lep KO mice compared to the vehicle-treated group. These alterations in their liver tissues were accompanied by an upregulation of the serum lipid profile, as well as alterations in the adipogenesis, lipogenesis, and lipolysis pathways in the adipose tissues of MP-treated WT and Lep KO mice. Specifically, the level of leptin was increased in the adipose tissues of MP-treated WT mice without any change in their food intake. Also, MP-induced disruptions in the glycogenolysis, Glu transporter type 4 (GLUT4)-5' AMP-activated protein kinase (AMPK) signaling pathway, levels of lipid intermediates, and the insulin resistance of the liver tissues of WT and Lep KO mice were observed. Furthermore, the levels of seven endogenous metabolites were remarkably changed in the serum of WT and Lep KO mice after MP administrations. Finally, the impact of the MP administration observed in both types of mice was further verified in differentiated 3T3-L1 adipocytes and HepG2 cells. Thus, these results suggest that the oral administration of MPs for 9 weeks may be associated with the disruption of lipid, Glu, and amino acid metabolism in the liver tissue of obese WT and Lep KO mice.


Subject(s)
Amino Acids , Glucose , Lipid Metabolism , Liver , Mice, Inbred C57BL , Mice, Knockout , Microplastics , Polystyrenes , Animals , Liver/metabolism , Liver/drug effects , Mice , Glucose/metabolism , Lipid Metabolism/drug effects , Amino Acids/metabolism , Administration, Oral , Leptin/metabolism , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Adipogenesis/drug effects , Male , Lipogenesis/drug effects , Obesity/metabolism , Obesity/etiology , Obesity/genetics , Humans , Lipolysis/drug effects
9.
AAPS PharmSciTech ; 25(7): 192, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164485

ABSTRACT

Lipid-based formulations (LbFs) have demonstrated success in pharmaceutical applications; however, challenges persist in dissolving entire doses of the drug into defined liquid volumes. In this study, the temperature-induced supersaturation method was employed in LbF to address drug loading and pill burden issues. Supersaturated LbFs (super-LbF) were prepared using the temperature-induced supersaturation method, where the drug load is above its equilibrium solubility. Further, the influence of the drug's physicochemical and thermal characteristics on drug loading and their relevance with an apparent degree of supersaturation (aDS) was studied using two model drugs, ibrutinib and enzalutamide. All the prepared LbFs were evaluated in terms of physical stability, dispersion, and solubilization capacity, as well as pharmacokinetic assessments. Drug re-crystallization was observed in the lipid solution on long-term storage at higher aDS values of 2-2.5. Furthermore, high-throughput lipolysis studies demonstrated a significant decrease in drug concentration across all LbFs (regardless of drug loading) due to a decline in the formulation solvation capacity and subsequent generation of in-situ supersaturation. Further, the in vivo results demonstrated comparable pharmacokinetic parameters between conventional LbF and super-LbF. The short duration of the thermodynamic metastable state limits the potential absorption benefits. However, super-LbFs of Ibr and Enz showed superior profiles, with 1.7-fold and 5.2-fold increased drug exposure compared to their respective crystalline suspensions. In summary, this study emphasizes the potential of temperature-induced supersaturation in LbF for enhancing drug loading and highlights the intricate interplay between drug properties, formulation characteristics, and in vivo performance.


Subject(s)
Adenine , Benzamides , Chemistry, Pharmaceutical , Lipids , Nitriles , Phenylthiohydantoin , Piperidines , Solubility , Temperature , Nitriles/chemistry , Nitriles/administration & dosage , Piperidines/chemistry , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Benzamides/chemistry , Benzamides/pharmacokinetics , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/administration & dosage , Phenylthiohydantoin/pharmacokinetics , Phenylthiohydantoin/administration & dosage , Lipids/chemistry , Animals , Chemistry, Pharmaceutical/methods , Drug Compounding/methods , Male , Pyrimidines/pharmacokinetics , Pyrimidines/chemistry , Pyrimidines/administration & dosage , Drug Stability , Crystallization/methods , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyrazoles/administration & dosage , Lipolysis/drug effects , Rats
10.
EMBO J ; 38(11)2019 06 03.
Article in English | MEDLINE | ID: mdl-31028084

ABSTRACT

Alternatively activated M2 macrophages play an important role in maintenance of tissue homeostasis by scavenging dead cells, cell debris and lipoprotein aggregates via phagocytosis. Using proteomics, we investigated how alternative activation, driven by IL-4, modulated the phagosomal proteome to control macrophage function. Our data indicate that alternative activation enhances homeostatic functions such as proteolysis, lipolysis and nutrient transport. Intriguingly, we identified the enhanced recruitment of the TAK1/MKK7/JNK signalling complex to phagosomes of IL-4-activated macrophages. The recruitment of this signalling complex was mediated through K63 polyubiquitylation of the macrophage scavenger receptor 1 (MSR1). Triggering of MSR1 in IL-4-activated macrophages leads to enhanced JNK activation, thereby promoting a phenotypic switch from an anti-inflammatory to a pro-inflammatory state, which was abolished upon MSR1 deletion or JNK inhibition. Moreover, MSR1 K63 polyubiquitylation correlated with the activation of JNK signalling in ovarian cancer tissue from human patients, suggesting that it may be relevant for macrophage phenotypic shift in vivo Altogether, we identified that MSR1 signals through JNK via K63 polyubiquitylation and provides evidence for the receptor's involvement in macrophage polarization.


Subject(s)
Inflammation , Interleukin-4/pharmacology , JNK Mitogen-Activated Protein Kinases/physiology , Macrophage Activation , Scavenger Receptors, Class A/agonists , Scavenger Receptors, Class A/genetics , Animals , Cell Polarity/drug effects , Cell Polarity/genetics , Cells, Cultured , Female , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/physiology , Lipolysis/drug effects , Lipolysis/genetics , Lipoproteins, LDL/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophages/drug effects , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/drug effects , Phagocytosis/genetics , Polysaccharides/pharmacology , Protein Processing, Post-Translational/genetics , RAW 264.7 Cells , Scavenger Receptors, Class A/chemistry , Scavenger Receptors, Class A/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Ubiquitination/genetics
11.
Nature ; 550(7674): 119-123, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28953873

ABSTRACT

Catecholamine-induced lipolysis, the first step in the generation of energy substrates by the hydrolysis of triglycerides, declines with age. The defect in the mobilization of free fatty acids in the elderly is accompanied by increased visceral adiposity, lower exercise capacity, failure to maintain core body temperature during cold stress, and reduced ability to survive starvation. Although catecholamine signalling in adipocytes is normal in the elderly, how lipolysis is impaired in ageing remains unknown. Here we show that adipose tissue macrophages regulate the age-related reduction in adipocyte lipolysis in mice by lowering the bioavailability of noradrenaline. Unexpectedly, unbiased whole-transcriptome analyses of adipose macrophages revealed that ageing upregulates genes that control catecholamine degradation in an NLRP3 inflammasome-dependent manner. Deletion of NLRP3 in ageing restored catecholamine-induced lipolysis by downregulating growth differentiation factor-3 (GDF3) and monoamine oxidase A (MAOA) that is known to degrade noradrenaline. Consistent with this, deletion of GDF3 in inflammasome-activated macrophages improved lipolysis by decreasing levels of MAOA and caspase-1. Furthermore, inhibition of MAOA reversed the age-related reduction in noradrenaline concentration in adipose tissue, and restored lipolysis with increased levels of the key lipolytic enzymes adipose triglyceride lipase (ATGL) and hormone sensitive lipase (HSL). Our study reveals that targeting neuro-immunometabolic signalling between the sympathetic nervous system and macrophages may offer new approaches to mitigate chronic inflammation-induced metabolic impairment and functional decline.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Aging/metabolism , Catecholamines/metabolism , Inflammasomes/metabolism , Lipolysis , Macrophages/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Aging/drug effects , Aging/genetics , Animals , Caspase 1/metabolism , Catecholamines/pharmacology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Growth Differentiation Factor 3/deficiency , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , Lipase/metabolism , Lipolysis/drug effects , Lipolysis/genetics , Mice , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Norepinephrine/metabolism , Sterol Esterase/metabolism
13.
Nature ; 538(7623): 109-113, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27680705

ABSTRACT

Cancer stem cells (CSCs) may be responsible for tumour dormancy, relapse and the eventual death of most cancer patients. In addition, these cells are usually resistant to cytotoxic conditions. However, very little is known about the biology behind this resistance to therapeutics. Here we investigated stem-cell death in the digestive system of adult Drosophila melanogaster. We found that knockdown of the coat protein complex I (COPI)-Arf79F (also known as Arf1) complex selectively killed normal and transformed stem cells through necrosis, by attenuating the lipolysis pathway, but spared differentiated cells. The dying stem cells were engulfed by neighbouring differentiated cells through a draper-myoblast city-Rac1-basket (also known as JNK)-dependent autophagy pathway. Furthermore, Arf1 inhibitors reduced CSCs in human cancer cell lines. Thus, normal or cancer stem cells may rely primarily on lipid reserves for energy, in such a way that blocking lipolysis starves them to death. This finding may lead to new therapies that could help to eliminate CSCs in human cancers.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Lipolysis/physiology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , ADP-Ribosylation Factor 1/antagonists & inhibitors , ADP-Ribosylation Factor 1/deficiency , Animals , Apoptosis , Autophagy , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Coat Protein Complex I/deficiency , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drug Resistance, Neoplasm/drug effects , Energy Metabolism , Enterocytes/cytology , Female , Gastrointestinal Tract/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lipolysis/drug effects , MAP Kinase Signaling System , Male , Membrane Proteins/metabolism , Necrosis/chemically induced , Neoplastic Stem Cells/drug effects , Phagocytosis , rac GTP-Binding Proteins/metabolism
14.
Proc Natl Acad Sci U S A ; 116(22): 10937-10942, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31085638

ABSTRACT

Mirabegron (Myrbetriq) is a ß3-adrenoreceptor agonist approved for treating overactive bladder syndrome in human patients. This drug can activate brown adipose tissue (BAT) in adult humans and rodents through the ß3-adrenoreceptor-mediated sympathetic activation. However, the effect of the mirabegron, approved by the US Food and Drug Administration, on atherosclerosis-related cardiovascular disease is unknown. Here, we show that the clinical dose of mirabegron-induced BAT activation and browning of white adipose tissue (WAT) exacerbate atherosclerotic plaque development. In apolipoprotein E-/- (ApoE-/-) and low-density lipoprotein (LDL) receptor-/- (Ldlr-/-) mice, oral administration of clinically relevant doses of mirabegron markedly accelerates atherosclerotic plaque growth and instability by a mechanism of increasing plasma levels of both LDL-cholesterol and very LDL-cholesterol remnants. Stimulation of atherosclerotic plaque development by mirabegron is dependent on thermogenesis-triggered lipolysis. Genetic deletion of the critical thermogenesis-dependent protein, uncoupling protein 1, completely abrogates the mirabegron-induced atherosclerosis. Together, our findings suggest that mirabegron may trigger cardiovascular and cerebrovascular diseases in patients who suffer from atherosclerosis.


Subject(s)
Acetanilides/pharmacology , Adipose Tissue, Brown/drug effects , Atherosclerosis/pathology , Lipolysis/drug effects , Thiazoles/pharmacology , Urological Agents/pharmacology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Atherosclerosis/physiopathology , Cholesterol, LDL/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Receptors, LDL/genetics
15.
Int J Mol Sci ; 23(9)2022 Apr 23.
Article in English | MEDLINE | ID: mdl-35563078

ABSTRACT

Oncostatin M (OSM) is an immune cell-derived cytokine that is upregulated in adipose tissue in obesity. Upon binding its receptor (OSMR), OSM induces the phosphorylation of the p66 subunit of Src homology 2 domain-containing transforming protein 1 (SHC1), called p66Shc, and activates the extracellular signal-related kinase (ERK) pathway. Mice with adipocyte-specific OSMR deletion (OsmrFKO) are insulin resistant and exhibit adipose tissue inflammation, suggesting that intact adipocyte OSM-OSMR signaling is necessary for maintaining adipose tissue health. How OSM affects specific adipocyte functions is still unclear. Here, we examined the effects of OSM on adipocyte lipolysis. We treated 3T3-L1 adipocytes with OSM, insulin, and/or inhibitors of SHC1 and ERK and measured glycerol release. We also measured phosphorylation of p66Shc, ERK, and insulin receptor substrate-1 (IRS1) and the expression of lipolysis-associated genes in OSM-exposed 3T3-L1 adipocytes and primary adipocytes from control and OsmrFKO mice. We found that OSM induces adipocyte lipolysis via a p66Shc-ERK pathway and inhibits the suppression of lipolysis by insulin. Further, OSM induces phosphorylation of inhibitory IRS1 residues. We conclude that OSM is a stimulator of lipolysis and inhibits adipocyte insulin response. Future studies will determine how these roles of OSM affect adipose tissue function in health and disease.


Subject(s)
Insulin , Lipolysis , Oncostatin M , 3T3-L1 Cells/metabolism , Adipocytes/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Insulin/metabolism , Insulin, Regular, Human , Lipolysis/drug effects , Mice , Oncostatin M/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism
16.
Int J Mol Sci ; 23(13)2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35806278

ABSTRACT

Casein kinase 2 (CK2) is a ubiquitously expressed serine/threonine kinase and is upregulated in human obesity. CX-4945 (Silmitasertib) is a CK2 inhibitor with anti-cancerous and anti-adipogenic activities. However, the anti-adipogenic and pro-lipolytic effects and the mode of action of CX-4945 in (pre)adipocytes remain elusive. Here, we explored the effects of CX-4945 on adipogenesis and lipolysis in differentiating and differentiated 3T3-L1 cells, a murine preadipocyte cell line. CX-4945 at 15 µM strongly reduced lipid droplet (LD) accumulation and triglyceride (TG) content in differentiating 3T3-L1 cells, indicating the drug's anti-adipogenic effect. Mechanistically, CX-4945 reduced the expression levels of CCAAT/enhancer-binding protein-α (C/EBP-α), peroxisome proliferator-activated receptor-γ (PPAR-γ), fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), and perilipin A in differentiating 3T3-L1 cells. Strikingly, CX-4945 further increased the phosphorylation levels of cAMP-activated protein kinase (AMPK) and liver kinase B-1 (LKB-1) while decreasing the intracellular ATP content in differentiating 3T3-L1 cells. In differentiated 3T3-L1 cells, CX-4945 had abilities to stimulate glycerol release and elevate the phosphorylation levels of hormone-sensitive lipase (HSL), pointing to the drug's pro-lipolytic effect. In addition, CX-4945 induced the activation of extracellular signal-regulated kinase-1/2 (ERK-1/2), and PD98059, an inhibitor of ERK-1/2, attenuated the CX4945-induced glycerol release and HSL phosphorylation in differentiated 3T3-L1 cells, indicating the drug's ERK-1/2-dependent lipolysis. In summary, this investigation shows that CX-4945 has strong anti-adipogenic and pro-lipolytic effects on differentiating and differentiated 3T3-L1 cells, mediated by control of the expression and phosphorylation levels of CK2, C/EBP-α, PPAR-γ, FAS, ACC, perilipin A, AMPK, LKB-1, ERK-1/2, and HSL.


Subject(s)
Adipogenesis , Casein Kinase II , Naphthyridines , Phenazines , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Cell Differentiation/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Glycerol/pharmacology , Humans , Lipolysis/drug effects , Mice , Naphthyridines/pharmacology , PPAR gamma/metabolism , Perilipin-1/metabolism , Phenazines/pharmacology , Sterol Esterase/metabolism
17.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163277

ABSTRACT

Hydrogen sulfide (H2S) and inorganic polysulfides are important signaling molecules; however, little is known about their role in the adipose tissue. We examined the effect of H2S and polysulfides on adipose tissue lipolysis. H2S and polysulfide production by mesenteric adipose tissue explants in rats was measured. The effect of Na2S and Na2S4, the H2S and polysulfide donors, respectively, on lipolysis markers, plasma non-esterified fatty acids (NEFA) and glycerol, was examined. Na2S but not Na2S4 increased plasma NEFA and glycerol in a time- and dose-dependent manner. Na2S increased cyclic AMP but not cyclic GMP concentration in the adipose tissue. The effect of Na2S on NEFA and glycerol was abolished by the specific inhibitor of protein kinase A, KT5720. The effect of Na2S on lipolysis was not abolished by propranolol, suggesting no involvement of ß-adrenergic receptors. In addition, Na2S had no effect on phosphodiesterase activity in the adipose tissue. Obesity induced by feeding rats a highly palatable diet for 1 month was associated with increased plasma NEFA and glycerol concentrations, as well as greater H2S production in the adipose tissue. In conclusion, H2S stimulates lipolysis and may contribute to the enhanced lipolysis associated with obesity.


Subject(s)
Adipose Tissue/metabolism , Hydrogen Sulfide/metabolism , Lipolysis/physiology , Adipose Tissue/drug effects , Animals , Cyclic GMP/metabolism , Fatty Acids, Nonesterified/metabolism , Lipolysis/drug effects , Male , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology , Obesity/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta/metabolism , Sulfides/metabolism
18.
J Biol Chem ; 295(42): 14379-14390, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32796035

ABSTRACT

In a healthy person, the kidney filters nearly 200 g of glucose per day, almost all of which is reabsorbed. The primary transporter responsible for renal glucose reabsorption is sodium-glucose cotransporter-2 (SGLT2). Based on the impact of SGLT2 to prevent renal glucose wasting, SGLT2 inhibitors have been developed to treat diabetes and are the newest class of glucose-lowering agents approved in the United States. By inhibiting glucose reabsorption in the proximal tubule, these agents promote glycosuria, thereby reducing blood glucose concentrations and often resulting in modest weight loss. Recent work in humans and rodents has demonstrated that the clinical utility of these agents may not be limited to diabetes management: SGLT2 inhibitors have also shown therapeutic promise in improving outcomes in heart failure, atrial fibrillation, and, in preclinical studies, certain cancers. Unfortunately, these benefits are not without risk: SGLT2 inhibitors predispose to euglycemic ketoacidosis in those with type 2 diabetes and, largely for this reason, are not approved to treat type 1 diabetes. The mechanism for each of the beneficial and harmful effects of SGLT2 inhibitors-with the exception of their effect to lower plasma glucose concentrations-is an area of active investigation. In this review, we discuss the mechanisms by which these drugs cause euglycemic ketoacidosis and hyperglucagonemia and stimulate hepatic gluconeogenesis as well as their beneficial effects in cardiovascular disease and cancer. In so doing, we aim to highlight the crucial role for selecting patients for SGLT2 inhibitor therapy and highlight several crucial questions that remain unanswered.


Subject(s)
Diabetic Ketoacidosis/etiology , Heart Failure/prevention & control , Neoplasms/prevention & control , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Animals , Cell Proliferation/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Diabetic Ketoacidosis/pathology , Heart Failure/pathology , Humans , Lipolysis/drug effects , Neoplasms/pathology , Risk , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
19.
Am J Physiol Endocrinol Metab ; 320(4): E653-E670, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33522398

ABSTRACT

Insulin inhibits systemic nonesterified fatty acid (NEFA) flux to a greater degree than glucose or any other metabolite. This remarkable effect is mainly due to insulin-mediated inhibition of intracellular triglyceride (TG) lipolysis in adipose tissues and is essential to prevent diabetic ketoacidosis, but also to limit the potential lipotoxic effects of NEFA in lean tissues that contribute to the development of diabetes complications. Insulin also regulates adipose tissue fatty acid esterification, glycerol and TG synthesis, lipogenesis, and possibly oxidation, contributing to the trapping of dietary fatty acids in the postprandial state. Excess NEFA flux at a given insulin level has been used to define in vivo adipose tissue insulin resistance. Adipose tissue insulin resistance defined in this fashion has been associated with several dysmetabolic features and complications of diabetes, but the mechanistic significance of this concept is not fully understood. This review focusses on the in vivo regulation of adipose tissue fatty acid metabolism by insulin and the mechanistic significance of the current definition of adipose tissue insulin resistance. One hundred years after the discovery of insulin and despite decades of investigations, much is still to be understood about the multifaceted in vivo actions of this hormone on adipose tissue fatty acid metabolism.


Subject(s)
Adipose Tissue/drug effects , Insulin/isolation & purification , Insulin/pharmacology , Lipid Metabolism/drug effects , Adipose Tissue/metabolism , Animals , Anniversaries and Special Events , Drug Discovery/history , Endocrinology/history , Endocrinology/trends , Fatty Acids, Nonesterified/metabolism , Glucose/metabolism , History, 20th Century , History, 21st Century , Humans , Insulin/history , Insulin/therapeutic use , Lipogenesis/drug effects , Lipolysis/drug effects
20.
Am J Physiol Gastrointest Liver Physiol ; 321(3): G298-G307, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34259586

ABSTRACT

Drug-induced liver injury is an emerging form of acute and chronic liver disease that may manifest as fatty liver. Amiodarone (AMD), a widely used antiarrhythmic drug, can cause hepatic injury and steatosis by a variety of mechanisms, not all completely understood. We hypothesized that repetitive AMD administration may induce hepatic lipotoxicity not only via effects on the liver but also via effects on adipose tissue. Indeed, repetitive AMD administration induced endoplasmic reticulum (ER) stress in both liver and adipose tissue. In adipose tissue, AMD reduced lipogenesis and increased lipolysis. Moreover, AMD treatment induced ER stress and ER stress-dependent lipolysis in 3T3L1 adipocytes in vitro. In the liver, AMD caused increased expression of genes encoding proteins involved in fatty acid (FA) uptake and transfer (Cd36, Fabp1, and Fabp4), and resulted in increased hepatic accumulation of free FAs, but not of triacylglycerols. In line with this, there was increased expression of hepatic de novo FA synthesis genes. However, AMD significantly reduced the expression of the desaturase Scd1 and elongase Elovl6, detected at mRNA and protein levels. Accordingly, the FA profile of hepatic total lipids revealed increased accumulation of palmitate, an SCD1 and ELOVL6 substrate, and reduced levels of palmitoleate and cis-vaccenate, products of the enzymes. In addition, AMD-treated mice displayed increased hepatic apoptosis. The studies show that repetitive AMD induces ER stress and aggravates lipolysis in adipose tissue while inducing a lipotoxic hepatic lipid environment, suggesting that AMD-induced liver damage is due to compound insult to liver and adipose tissue.NEW & NOTEWORTHY AMD chronic administration induces hepatic lipid accumulation by several mechanisms, including induction of hepatic ER stress, impairment of ß-oxidation, and inhibition of triacylglycerol secretion. Our study shows that repetitive AMD treatment induces not only hepatic ER stress but also adipose tissue ER stress and lipolysis and hepatic accumulation of free fatty acids and enrichment of palmitate in the total lipids. Understanding the toxicity mechanisms of AMD would help devise ways to limit liver damage.


Subject(s)
Amiodarone/pharmacology , Endoplasmic Reticulum Stress/drug effects , Fatty Acids, Nonesterified/metabolism , Fatty Acids/metabolism , Lipolysis/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Amiodarone/metabolism , Animals , Fatty Acid-Binding Proteins/metabolism , Fatty Liver/metabolism , Lipid Metabolism/drug effects , Lipogenesis/physiology , Liver/drug effects , Liver/metabolism , Liver Diseases/metabolism , Mice , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL