Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.639
Filter
1.
Sci Rep ; 14(1): 10789, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38734719

ABSTRACT

Brown adipocytes are potential therapeutic targets for the prevention of obesity-associated metabolic diseases because they consume circulating glucose and fatty acids for heat production. Angiotensin II (Ang II) peptide is involved in the pathogenesis of obesity- and cold-induced hypertension; however, the mechanism underlying the direct effects of Ang II on human brown adipocytes remains unclear. Our transcriptome analysis of chemical compound-induced brown adipocytes (ciBAs) showed that the Ang II type 1 receptor (AGTR1), but not AGTR2 and MAS1 receptors, was expressed. The Ang II/AGTR1 axis downregulated the expression of mitochondrial uncoupling protein 1 (UCP1). The simultaneous treatment with ß-adrenergic receptor agonists and Ang II attenuated UCP1 expression, triglyceride lipolysis, and cAMP levels, although cAMP response element-binding protein (CREB) phosphorylation was enhanced by Ang II mainly through the protein kinase C pathway. Despite reduced lipolysis, both coupled and uncoupled mitochondrial respiration was enhanced in Ang II-treated ciBAs. Instead, glycolysis and glucose uptake were robustly activated upon treatment with Ang II without a comprehensive transcriptional change in glucose metabolic genes. Elevated mitochondrial energy status induced by Ang II was likely associated with UCP1 repression. Our findings suggest that the Ang II/AGTR1 axis participates in mitochondrial thermogenic functions via glycolysis.


Subject(s)
Adipocytes, Brown , Angiotensin II , Glycolysis , Mitochondria , Thermogenesis , Uncoupling Protein 1 , Humans , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , Glycolysis/drug effects , Angiotensin II/pharmacology , Angiotensin II/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Lipolysis/drug effects , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/genetics , Glucose/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism
2.
J Diabetes Res ; 2024: 5511454, 2024.
Article in English | MEDLINE | ID: mdl-38736904

ABSTRACT

Adipose tissue dysfunction is seen among obese and type 2 diabetic individuals. Adipocyte proliferation and hypertrophy are the root causes of adipose tissue expansion. Solute carrier family 25 member 28 (SLC25A28) is an iron transporter in the inner mitochondrial membrane. This study is aimed at validating the involvement of SLC25A28 in adipose accumulation by tail vein injection of adenovirus (Ad)-SLC25A28 and Ad-green fluorescent protein viral particles into C57BL/6J mice. After 16 weeks, the body weight of the mice was measured. Subsequently, morphological analysis was performed to establish a high-fat diet (HFD)-induced model. SLC25A28 overexpression accelerated lipid accumulation in white and brown adipose tissue (BAT), enhanced body weight, reduced serum triglyceride (TG), and impaired serum glucose tolerance. The protein expression level of lipogenesis, lipolysis, and serum adipose secretion hormone was evaluated by western blotting. The results showed that adipose TG lipase (ATGL) protein expression was reduced significantly in white and BAT after overexpression SLC25A28 compared to the control group. Moreover, SLC25A28 overexpression inhibited the BAT formation by downregulating UCP-1 and the mitochondrial biosynthesis marker PGC-1α. Serum adiponectin protein expression was unregulated, which was consistent with the expression in inguinal white adipose tissue (iWAT). Remarkably, serum fibroblast growth factor (FGF21) protein expression was negatively related to the expansion of adipose tissue after administrated by Ad-SLC25A28. Data from the current study indicate that SLC25A28 overexpression promotes diet-induced obesity and accelerates lipid accumulation by regulating hormone secretion and inhibiting lipolysis in adipose tissue.


Subject(s)
Adipogenesis , Adipose Tissue, Brown , Adipose Tissue, White , Diet, High-Fat , Lipase , Mice, Inbred C57BL , Animals , Mice , Male , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Lipase/metabolism , Lipase/genetics , Obesity/metabolism , Lipolysis , Uncoupling Protein 1/metabolism , Fibroblast Growth Factors/metabolism , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Adipocytes/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Lipogenesis , Acyltransferases
3.
Food Res Int ; 186: 114350, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729698

ABSTRACT

In this study, three types of ß-sitosterol-based oleogels (ß-sitosterol + Î³-oryzanol oleogels, ß-sitosterol + lecithin, oleogels and ß-sitosterol + monostearate oleogels), loaded with astaxanthin, were employed as the oil phase to create oleogel-based emulsions (SO, SL, and SM) using high-pressure homogenization. The microstructure revealed that fine-scale crystals were dispersed within the oil phase of the droplets in the ß-sitosterol oleogel-based emulsion. The bioaccessibility of astaxanthin was found to be 58.13 %, 51.24 %, 36.57 %, and 45.72 % for SM, SL, SO, and the control group, respectively. Interestingly, the release of fatty acids was positively correlated with the availability of astaxanthin (P = 0.981). Further analysis of FFAs release and kinetics indicated that the structural strength of the oil-phase in the emulsions influenced the degree and rate of lipolysis. Additionally, the micellar fraction analysis suggested that the nature and composition of the oleogelators in SM and SL also impacted lipolysis and the bioaccessibility of astaxanthin. Furthermore, interfacial binding of lipase and isothermal titration calorimetry (ITC) measurements revealed that the oleogel network within the oil phase of the emulsion acted as a physical barrier, hindering the interaction between lipase and lipid. Overall, ß-sitosterol oleogel-based emulsions offer a versatile platform for delivering hydrophobic molecules, enhancing the bioavailability of active compounds, and achieving sustained release.


Subject(s)
Emulsions , Organic Chemicals , Sitosterols , Xanthophylls , Sitosterols/chemistry , Xanthophylls/chemistry , Organic Chemicals/chemistry , Biological Availability , Lipolysis , Lecithins/chemistry , Fatty Acids/chemistry , Phenylpropionates
4.
Food Res Int ; 186: 114376, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729723

ABSTRACT

Commercial beef burgers and vegan analogues were purchased and, after a microwave treatment, they were submitted to an in vitro digestion (INFOGEST). Vegan cooked burgers showed similar protein content (16-17 %) but lower amounts of total peptides than beef burgers. The protein digestibility was higher in beef burgers. Peptide amounts increased during in vitro digestion, reaching similar amounts in both types of products in the micellar phase (bioaccessible fraction). The fat content in cooked vegan burgers was significantly lower than in beef burgers (16.7 and 21.2 %, respectively), with a higher amount of PUFAs and being the lipolysis activity, measure by FFA, less intense both after cooking and after the gastrointestinal process. Both types of cooked samples showed high carbonyl amounts (34.18 and 25.51 nmol/mg protein in beef and vegan samples, respectively), that decreased during in vitro digestion. On the contrary, lipid oxidation increased during gastrointestinal digestion, particularly in vegan samples. The antioxidant capacity (ABTS and DPPH) showed higher values for vegan products in cooked samples, but significantly decreased during digestion, reaching similar values for both types of products.


Subject(s)
Cooking , Digestion , Microwaves , Red Meat , Cooking/methods , Red Meat/analysis , Animals , Cattle , Antioxidants/analysis , Meat Products/analysis , Lipolysis , Diet, Vegan
5.
Food Res Int ; 187: 114421, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763671

ABSTRACT

This study focused on the protein-stabilised triglyceride (TG)/water interfaces and oil-in-water emulsions, and explored the influence of varying molar ratios of bile salts (BSs) and phospholipids (PLs) on the intestinal lipolysis of TGs. The presence of these two major groups of biosurfactants delivered with human bile to the physiological environment of intestinal digestion was replicated in our experiments by using mixtures of individual BSs and PLs under in vitro small intestinal lipolysis conditions. Conducted initially, retrospective analysis of available scientific literature revealed that an average molar ratio of 9:4 for BSs to PLs (BS/PL) can be considered physiological in the postprandial adult human small intestine. Our experimental data showed that combining BSs and PLs synergistically enhanced interfacial activity, substantially reducing oil-water interfacial tension (IFT) during interfacial lipolysis experiments with pancreatic lipase, especially at the BS/PL-9:4 ratio. Other BS/PL molar proportions (BS/PL-6.5:6.5 and BS/PL-4:9) and an equimolar amount of BSs (BS-13) followed in IFT reduction efficiency, while using PLs alone as biosurfactants was the least efficient. In the following emulsion lipolysis experiments, BS/PL-9:4 outperformed other BS/PL mixtures in terms of enhancing the TG digestion extent. The degree of TG conversion and the desorption efficiency of interfacial material post-lipolysis correlated directly with the BS/PL ratio, decreasing as the PL proportion increased. In conclusion, this study highlights the crucial role of biliary PLs, alongside BSs, in replicating the physiological function of bile in intestinal lipolysis of emulsified TGs. Our results showed different contributions of PLs and BSs to lipolysis, strongly suggesting that any future in vitro studies aiming to simulate the human digestion conditions should take into account the impact of biliary PLs - not just BSs - to accurately mimic the physiological role of bile in intestinal lipolysis. This is particularly crucial given the fact that existing in vitro digestion protocols typically focus solely on applying specific concentrations and/or compositions of BSs to simulate the action of human bile during intestinal digestion, while overlooking the presence and concentration of biliary PLs under physiological gut conditions.


Subject(s)
Bile Acids and Salts , Digestion , Emulsions , Lipolysis , Phospholipids , Triglycerides , Emulsions/chemistry , Triglycerides/metabolism , Triglycerides/chemistry , Bile Acids and Salts/metabolism , Humans , Phospholipids/chemistry , Phospholipids/metabolism , Digestion/physiology , Lipase/metabolism , Intestine, Small/metabolism , Surface-Active Agents/chemistry
6.
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
7.
PLoS Pathog ; 20(4): e1012123, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38607975

ABSTRACT

RAB GTPases (RABs) control intracellular membrane trafficking with high precision. In the present study, we carried out a short hairpin RNA (shRNA) screen focused on a library of 62 RABs during infection with porcine reproductive and respiratory syndrome virus 2 (PRRSV-2), a member of the family Arteriviridae. We found that 13 RABs negatively affect the yield of PRRSV-2 progeny virus, whereas 29 RABs have a positive impact on the yield of PRRSV-2 progeny virus. Further analysis revealed that PRRSV-2 infection transcriptionally regulated RAB18 through RIG-I/MAVS-mediated canonical NF-κB activation. Disrupting RAB18 expression led to the accumulation of lipid droplets (LDs), impaired LDs catabolism, and flawed viral replication and assembly. We also discovered that PRRSV-2 co-opts chaperone-mediated autophagy (CMA) for lipolysis via RAB18, as indicated by the enhanced associations between RAB18 and perlipin 2 (PLIN2), CMA-specific lysosomal associated membrane protein 2A (LAMP2A), and heat shock protein family A (Hsp70) member 8 (HSPA8/HSC70) during PRRSV-2 infection. Knockdown of HSPA8 and LAMP2A impacted on the yield of PRRSV-2 progeny virus, implying that the virus utilizes RAB18 to promote CMA-mediated lipolysis. Importantly, we determined that the C-terminal domain (CTD) of HSPA8 could bind to the switch II domain of RAB18, and the CTD of PLIN2 was capable of associating with HSPA8, suggesting that HSPA8 facilitates the interaction between RAB18 and PLIN2 in the CMA process. In summary, our findings elucidate how PRRSV-2 hijacks CMA-mediated lipid metabolism through innate immune activation to enhance the yield of progeny virus, offering novel insights for the development of anti-PRRSV-2 treatments.


Subject(s)
Chaperone-Mediated Autophagy , Porcine respiratory and reproductive syndrome virus , Swine , Animals , Lipolysis , Up-Regulation , rab GTP-Binding Proteins/genetics , Lysosomal Membrane Proteins , RNA, Small Interfering
8.
JCI Insight ; 9(9)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38602778

ABSTRACT

BACKGROUND: Upper-body obesity (UBO) results in insulin resistance with regards to free fatty acid (FFA) release; how this differs by fat depot and sex between adults with UBO and lean adults is unknown. We tested the hypothesis that insulin suppression of FFA release from the splanchnic bed, leg fat, and upper-body nonsplanchnic (UBNS) adipose tissue would be impaired in UBO. METHODS: Fourteen volunteers with UBO (7 men and 7 women) and 14 healthy volunteers with normal weight (7 men and 7 women) participated in studies that included femoral artery, femoral vein, and hepatic vein catheterization. We then measured leg and splanchnic plasma flow as well as FFA kinetics (using isotopic tracers) under overnight fasting as well as low- and high-dose insulin infusion using the insulin clamp technique. RESULTS: We found the expected insulin resistance in UBO; the most quantitatively important difference between adults with UBO and lean adults was greater FFA release from UBNS adipose tissue when plasma insulin concentrations were in the postprandial, physiological range. There were obesity, but not sex, differences in the regulation of splanchnic FFA release and sex differences in the regulation of leg FFA release. CONCLUSION: Reversing the defects in insulin-regulated UBNS adipose tissue FFA release would have the greatest effect on systemic FFA abnormalities in UBO. FUNDING: These studies were supported by the US Public Health Service (grants DK45343 and DK40484), the Novo Nordic Foundation (grant NNF18OC0031804 and NNF16OC0021406), and the Independent Research Fund Denmark (grant 8020-00420B).


Subject(s)
Adipose Tissue , Fatty Acids, Nonesterified , Insulin Resistance , Insulin , Lipolysis , Obesity , Adult , Female , Humans , Male , Young Adult , Adipose Tissue/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Nonesterified/blood , Insulin/metabolism , Obesity/metabolism , Postprandial Period , Thinness/metabolism
9.
Biochim Biophys Acta Rev Cancer ; 1879(3): 189103, 2024 May.
Article in English | MEDLINE | ID: mdl-38679401

ABSTRACT

Cancer-associated cachexia (CAC) is a complex multiple organ syndrome that significantly contributes to reduced quality of life and increased mortality among many cancer patients. Its multifactorial nature makes its early diagnosis and effective therapeutic interventions challenging. Adipose tissue is particularly impacted by cachexia, typically through increased lipolysis, browning and thermogenesis, mainly at the onset of the disease. These processes lead to depletion of fat mass and contribute to the dysfunction of other organs. The ß-adrenergic signalling pathways are classical players in the regulation of adipose tissue metabolism. They are activated upon sympathetic stimulation inducing lipolysis, browning and thermogenesis, therefore contributing to energy expenditure. Despite accumulating evidence suggesting that ß3-adrenergic receptor stimulation may be crucial to the adipose tissue remodelling during cachexia, the literature remains controversial. Moreover, there is limited knowledge regarding sexual dimorphism of adipose tissue in the context of cachexia. This review paper aims to present the current knowledge regarding adipose tissue wasting during CAC, with a specific focus on the role of the ß3-adrenergic receptor, placing it as a potential therapeutic target against cachexia.


Subject(s)
Adipose Tissue , Cachexia , Lipolysis , Neoplasms , Receptors, Adrenergic, beta-3 , Signal Transduction , Cachexia/metabolism , Cachexia/pathology , Cachexia/etiology , Humans , Neoplasms/complications , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Adrenergic, beta-3/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Energy Metabolism , Thermogenesis , Animals
10.
J Endocrinol ; 261(3)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38579777

ABSTRACT

Adipose tissue was once known as a reservoir for energy storage but is now considered a crucial organ for hormone and energy flux with important effects on health and disease. Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone secreted from the small intestinal K cells, responsible for augmenting insulin release, and has gained attention for its independent and amicable effects with glucagon-like peptide 1 (GLP-1), another incretin hormone secreted from the small intestinal L cells. The GIP receptor (GIPR) is found in whole adipose tissue, whereas the GLP-1 receptor (GLP-1R) is not, and some studies suggest that GIPR action lowers body weight and plays a role in lipolysis, glucose/lipid uptake/disposal, adipose tissue blood flow, lipid oxidation, and free-fatty acid (FFA) re-esterification, which may or may not be influenced by other hormones such as insulin. This review summarizes the research on the effects of GIP in adipose tissue (distinct depots of white and brown) using cellular, rodent, and human models. In doing so, we explore the mechanisms of GIPR-based medications for treating metabolic disorders, such as type 2 diabetes and obesity, and how GIPR agonism and antagonism contribute to improvements in metabolic health outcomes, potentially through actions in adipose tissues.


Subject(s)
Adipose Tissue , Gastric Inhibitory Polypeptide , Receptors, Gastrointestinal Hormone , Humans , Gastric Inhibitory Polypeptide/metabolism , Animals , Adipose Tissue/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Glucose/metabolism , Lipolysis , Obesity/metabolism
11.
Cells ; 13(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667324

ABSTRACT

After exposure to cold stress, animals enhance the production of beige adipocytes and expedite thermogenesis, leading to improved metabolic health. Although brown adipose tissue in rodents is primarily induced by ß3-adrenergic receptor (ADRB3) stimulation, the activation of major ß-adrenergic receptors (ADRBs) in pigs has been a topic of debate. To address this, we developed overexpression vectors for ADRB1, ADRB2, and ADRB3 and silenced the expression of these receptors to observe their effects on the adipogenic differentiation stages of porcine preadipocytes. Our investigation revealed that cold stress triggers the transformation of subcutaneous white adipose tissue to beige adipose tissue in pigs by modulating adrenergic receptor levels. Meanwhile, we found that ADRB3 promotes the transformation of white adipocytes into beige adipocytes. Notably, ADRB3 enhances the expression of beige adipose tissue marker genes, consequently influencing cellular respiration and metabolism by regulating lipolysis and mitochondrial expression. Therefore, ADRB3 may serve as a pivotal gene in animal husbandry and contribute to the improvement of cold intolerance in piglets.


Subject(s)
Adipocytes, Beige , Cold Temperature , Receptors, Adrenergic, beta-3 , Animals , Receptors, Adrenergic, beta-3/metabolism , Receptors, Adrenergic, beta-3/genetics , Adipocytes, Beige/metabolism , Swine , Adipogenesis/genetics , Lipolysis , Thermogenesis/genetics , Cell Differentiation , Mitochondria/metabolism
12.
Environ Sci Pollut Res Int ; 31(19): 28632-28643, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38558334

ABSTRACT

Lipases represent versatile biocatalysts extensively employed in transesterification reactions for ester production. Ethyl oleate holds significance in biodiesel production, serving as a sustainable alternative to petroleum-derived diesel. In this study, our goal was to prospect lipase and assess its efficacy as a biocatalyst for ethyl oleate synthesis. For quantitative analysis, a base medium supplemented with Rhodamine B, olive oil, and Tween 80 was used. Solid-state fermentation utilized crambe seeds of varying particle sizes and humidity levels as substrates. In the synthesis of ethyl oleate, molar ratios of 1:3, 1:6, and 1:9, along with a total enzymatic activity of 60 U in n-heptane, were utilized at temperatures of 30 °C, 37 °C, and 44 °C. Reactions were conducted in a shaker at 200 rpm for 60 min. As a result, we first identified Penicillium polonicum and employed the method of solid-state fermentation using crambe seeds as a substrate to produce lipase. Our findings revealed heightened lipolytic activity (22.5 Ug-1) after 96 h of fermentation using crambe cake as the substrate. Optimal results were achieved with crambe seeds at a granulometry of 0.6 mm and a fermentation medium humidity of 60%. Additionally, electron microscopy suggested the immobilization of lipase in the substrate, enabling enzyme reuse for up to 4 cycles with 100% enzymatic activity. Subsequently, we conducted applicability tests of biocatalysts for ethyl oleate synthesis, optimizing parameters such as the acid/alcohol molar ratio, temperature, and reaction time. We attained 100% conversion within 30 min at 37 °C, and our results indicated that the molar ratio proportion did not significantly influence the outcome. These findings provide a methodological alternative for the utilization of biocatalysts in ethyl oleate synthesis.


Subject(s)
Fermentation , Lipase , Oleic Acids , Penicillium , Oleic Acids/biosynthesis , Oleic Acids/metabolism , Penicillium/metabolism , Lipase/metabolism , Esterification , Biocatalysis , Lipolysis
13.
Proc Natl Acad Sci U S A ; 121(15): e2321255121, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38564632

ABSTRACT

Omega-3 polyunsaturated fatty acids (PUFA) found primarily in fish oil have been a popular supplement for cardiovascular health because they can substantially reduce circulating triglyceride levels in the bloodstream to prevent atherosclerosis. Beyond this established extracellular activity, here, we report a mode of action of PUFA, regulating intracellular triglyceride metabolism and lipid droplet (LD) dynamics. Real-time imaging of the subtle and highly dynamic changes of intracellular lipid metabolism was enabled by a fluorescence lifetime probe that addressed the limitations of intensity-based fluorescence quantifications. Surprisingly, we found that among omega-3 PUFA, only docosahexaenoic acid (DHA) promoted the lipolysis in LDs and reduced the overall fat content by approximately 50%, and consequently helped suppress macrophage differentiation into foam cells, one of the early steps responsible for atherosclerosis. Eicosapentaenoic acid, another omega-3 FA in fish oil, however, counteracted the beneficial effects of DHA on lipolysis promotion and cell foaming prevention. These in vitro findings warrant future validation in vivo.


Subject(s)
Atherosclerosis , Fatty Acids, Omega-3 , Humans , Lipolysis , Fluorescence , Fatty Acids, Omega-3/metabolism , Fish Oils/pharmacology , Docosahexaenoic Acids/metabolism , Macrophages/metabolism , Triglycerides
14.
Food Res Int ; 184: 114243, 2024 May.
Article in English | MEDLINE | ID: mdl-38609222

ABSTRACT

Recent explorations into rice bran oil (RBO) have highlighted its potential, owing to an advantageous fatty acid profile in the context of health and nutrition. Despite this, the susceptibility of rice bran lipids to oxidative degradation during storage remains a critical concern. This study focuses on the evolution of lipid degradation in RBO during storage, examining the increase in free fatty acids (FFAs), the formation of oxylipids, and the generation of volatile secondary oxidation products. Our findings reveal a substantial rise in FFA levels, from 109.55 to 354.06 mg/g, after 14 days of storage, highlighting significant lipid deterioration. Notably, key oxylipids, including 9,10-EpOME, 12,13(9,10)-DiHOME, and 13-oxoODE, were identified, with a demonstrated positive correlation between total oxylipids and free polyunsaturated fatty acids (PUFAs), specifically linoleic acid (LA) and α-linolenic acid (ALA). Furthermore, the study provides a detailed analysis of primary volatile secondary oxidation products. The insights gained from this study not only sheds light on the underlying mechanisms of lipid rancidity in rice bran but also offers significant implications for extending the shelf life and preserving the nutritional quality of RBO, aligning with the increasing global interest in this high-quality oil.


Subject(s)
Lipidomics , Lipolysis , Fatty Acids , Fatty Acids, Nonesterified , Linoleic Acid , Rice Bran Oil
15.
Food Res Int ; 184: 114255, 2024 May.
Article in English | MEDLINE | ID: mdl-38609233

ABSTRACT

Bile Salts (BS) are responsible for stimulating lipid digestion in our organism. Gut microbiota are responsible for the deconjugation process of primary conjugated to secondary unconjugated BS. We use two structurally distinct BS and characterize the rate of lipolysis as a compound parameter. A static in-vitro digestion model as well as meta-analysis of literature data has been performed to determine the most influential factors affecting the lipid digestion process. The results demonstrate that lipolysis of emulsions using conjugated BS (NaTC, FFA = 60.0 %, CMC in SIF = 5.58 mM, MSR of linoleic acid = 0.21, rate of adsorption = -0.057 mN/m.s) enhances the release of FFA compared to deconjugated BS (NaDC, FFA = 49.5 %, CMC in SIF = 2.49 mM, MSR of linoleic acid = 0.16 rate of adsorption = -0.064 mN/m.s). These results indicate that conjugation plays an important role in controlling the rate of lipolysis in our organism which can be in turn, tuned by the microflora composition of our gut, ultimately controlling the rate of deconjugation of the BS.


Subject(s)
Bile , Linoleic Acid , Emulsions , Lipolysis , Chemical Phenomena , Bile Acids and Salts
16.
Nat Cell Biol ; 26(4): 552-566, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38561547

ABSTRACT

Metabolic crosstalk of the major nutrients glucose, amino acids and fatty acids (FAs) ensures systemic metabolic homeostasis. The coordination between the supply of glucose and FAs to meet various physiological demands is especially important as improper nutrient levels lead to metabolic disorders, such as diabetes and metabolic dysfunction-associated steatohepatitis (MASH). In response to the oscillations in blood glucose levels, lipolysis is thought to be mainly regulated hormonally to control FA liberation from lipid droplets by insulin, catecholamine and glucagon. However, whether general cell-intrinsic mechanisms exist to directly modulate lipolysis via glucose sensing remains largely unknown. Here we report the identification of such an intrinsic mechanism, which involves Golgi PtdIns4P-mediated regulation of adipose triglyceride lipase (ATGL)-driven lipolysis via intracellular glucose sensing. Mechanistically, depletion of intracellular glucose results in lower Golgi PtdIns4P levels, and thus reduced assembly of the E3 ligase complex CUL7FBXW8 in the Golgi apparatus. Decreased levels of the E3 ligase complex lead to reduced polyubiquitylation of ATGL in the Golgi and enhancement of ATGL-driven lipolysis. This cell-intrinsic mechanism regulates both the pool of intracellular FAs and their extracellular release to meet physiological demands during fasting and glucose deprivation. Moreover, genetic and pharmacological manipulation of the Golgi PtdIns4P-CUL7FBXW8-ATGL axis in mouse models of simple hepatic steatosis and MASH, as well as during ex vivo perfusion of a human steatotic liver graft leads to the amelioration of steatosis, suggesting that this pathway might be a promising target for metabolic dysfunction-associated steatotic liver disease and possibly MASH.


Subject(s)
Blood Glucose , Lipolysis , Phosphatidylinositol Phosphates , Animals , Humans , Mice , Fatty Acids/metabolism , Glucose , Lipase/genetics , Lipase/metabolism , Lipolysis/genetics , Ubiquitin-Protein Ligases/metabolism
17.
Mol Metab ; 83: 101930, 2024 May.
Article in English | MEDLINE | ID: mdl-38570069

ABSTRACT

OBJECTIVE: Tumour progression drives profound alterations in host metabolism, such as adipose tissue depletion, an early event of cancer cachexia. As fatty acid consumption by cancer cells increases upon acidosis of the tumour microenvironment, we reasoned that fatty acids derived from distant adipose lipolysis may sustain tumour fatty acid craving, leading to the adipose tissue loss observed in cancer cachexia. METHODS: To evaluate the pro-lipolytic capacities of acid-exposed cancer cells, primary mouse adipocytes from subcutaneous and visceral adipose tissue were exposed to pH-matched conditioned medium from human and murine acid-exposed cancer cells (pH 6.5), compared to naive cancer cells (pH 7.4). To further address the role of tumoral acidosis on adipose tissue loss, a pH-low insertion peptide was injected into tumour-bearing mice, and tumoral acidosis was neutralised with a sodium bicarbonate buffer. Prolipolytic mediators were identified by transcriptomic approaches and validated on murine and human adipocytes. RESULTS: Here, we reveal that acid-exposed cancer cells promote lipolysis from subcutaneous and visceral adipocytes and that dampening acidosis in vivo inhibits adipose tissue depletion. We further found a set of well-known prolipolytic factors enhanced upon acidosis adaptation and unravelled a role for ß-glucuronidase (GUSB) as a promising new actor in adipocyte lipolysis. CONCLUSIONS: Tumoral acidosis promotes the mobilization of fatty acids derived from adipocytes via the release of soluble factors by cancer cells. Our work paves the way for therapeutic approaches aimed at tackling cachexia by targeting the tumour acidic compartment.


Subject(s)
Acidosis , Adipocytes , Adipose Tissue , Cachexia , Lipolysis , Animals , Mice , Acidosis/metabolism , Adipocytes/metabolism , Humans , Adipose Tissue/metabolism , Cachexia/metabolism , Male , Tumor Microenvironment , Cell Line, Tumor , Mice, Inbred C57BL , Fatty Acids/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Female , Glucuronidase/metabolism , Hydrogen-Ion Concentration
18.
Mol Cell Endocrinol ; 588: 112225, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38570133

ABSTRACT

Although Liraglutide (Lira) increases serum irisin levels in type 2 diabetes mellitus (T2DM), it is unclear whether it induces expression of uncoupling protein 1 (UCP1) of adipocytes via promoting irisin secretion from skeletal muscle. Male T2DM rats were treated with 0.4 mg/kg/d Lira twice a day for 8 weeks, and the protein expression of phosphorylated AMP kinase (p-AMPK), phosphorylated acetyl-CoA carboxylase 1 (p-ACC1) and UCP1 in white adipose tissues were detected. Differentiated C2C12 cells were treated with palmitic acid (PA) and Lira to detect the secretion of irisin. Differentiated 3T3-L1 cells were treated with irisin, supernatant from Lira-treated C2C12 cells, Compound C or siAMPKα1, the triglyceride (TG) content and the related gene expression were measured. The transcriptome in irisin-treated differentiated 3T3-L1 cells was analyzed. Lira elevated serum irisin levels, decreased the adipocyte size and increased the protein expression of UCP1, p-AMPK and p-ACC1 in WAT. Moreover, it promoted the expression of PGC1α and FNDC5, the secretion of irisin in PA-treated differentiated C2C12 cells. The irisin and supernatant decreased TG synthesis and promoted the expression of browning- and lipolysis-related genes in differentiated 3T3-L1 cells. While Compound C and siAMPKα1 blocked AMPK activities and expression, irisin partly reversed the pathway. Finally, the transcriptome analysis indicated that differently expressed genes are mainly involved in browning and lipid metabolism. Overall, our findings showed that Lira modulated muscle-to-adipose signaling pathways in diabetes via irisin-mediated AMPKα/ACC1/UCP1/PPARα pathway. Our results suggest a new mechanism for the treatment of T2DM by Lira.


Subject(s)
3T3-L1 Cells , Adipocytes , Fibronectins , Lipolysis , Liraglutide , Uncoupling Protein 1 , Animals , Fibronectins/metabolism , Fibronectins/genetics , Mice , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Male , Adipocytes/metabolism , Adipocytes/drug effects , Lipolysis/drug effects , Liraglutide/pharmacology , Rats , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/drug therapy , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , AMP-Activated Protein Kinases/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Rats, Sprague-Dawley , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/drug effects
19.
Int Immunopharmacol ; 132: 111906, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38593501

ABSTRACT

BACKGROUND: Age-related visceral obesity could contribute to the development of cardiometabolic complications. The pathogenesis of visceral fat mass accumulation during the aging process remains complex and largely unknown. Interleukin-6 (IL-6) has emerged as one of the prominent inflammaging markers which are elevated in circulation during aging. However, the precise role of IL-6 in regulating age-related visceral adipose tissue accumulation remains uncertain. RESULTS: A cross-sectional study including 77 older adults (≥65 years of age) was initially conducted. There was a significant positive association between serum IL-6 levels and visceral fat mass. We subsequently validated a modest but significant elevation in serum IL-6 levels in aged mice. Furthermore, we demonstrated that compared to wildtype control, IL-6 deficiency (IL-6 KO) significantly attenuated the accumulation of visceral adipose tissue during aging. Further metabolic characterization suggested that IL-6 deficiency resulted in improved lipid metabolism parameters and energy expenditure in aged mice. Moreover, histological examinations of adipose depots revealed that the absence of IL-6 ameliorated adipocyte hypertrophy in visceral adipose tissue of aged mice. Mechanically, the ablation of IL-6 could promote the PKA-mediated lipolysis and consequently mitigate lipid accumulation in adipose tissue in aged mice. CONCLUSION: Our findings identify a detrimental role of IL-6 during the aging process by promoting visceral adipose tissue accumulation through inhibition of lipolysis. Therefore, strategies aimed at preventing or reducing IL-6 levels may potentially ameliorate age-related obesity and improve metabolism during aging.


Subject(s)
Aging , Interleukin-6 , Intra-Abdominal Fat , Lipolysis , Mice, Knockout , Animals , Interleukin-6/metabolism , Intra-Abdominal Fat/metabolism , Aging/metabolism , Aged , Male , Humans , Mice , Female , Mice, Inbred C57BL , Cross-Sectional Studies , Adipocytes/metabolism
20.
Cell Rep ; 43(3): 113967, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38492219

ABSTRACT

Non-canonical lipolysis induced by inflammatory cytokines or Toll-like receptor ligands is required for the regulation of inflammation during endotoxemia and sepsis. Canonical lipolysis induced by catecholamines declines during aging due to factors including an expansion of lymphocytes, pro-inflammatory macrophage polarization, and an increase in chronic low-grade inflammation; however, the extent to which the non-canonical pathway of lipolysis is active and impacted by immune cells during aging remains unclear. Therefore, we aimed to define the extent to which immune cells from old mice influence non-canonical lipolysis during sepsis. We identified age-associated impairments of non-canonical lipolysis and an accumulation of dysfunctional B1 B cells in the visceral white adipose tissue (vWAT) of old mice. Lifelong deficiency of B cells results in restored non-canonical lipolysis and reductions in pro-inflammatory macrophage populations. Our study suggests that targeting the B cell-macrophage signaling axis may resolve metabolic dysfunction in aged vWAT and attenuate septic severity in older individuals.


Subject(s)
Lipolysis , Sepsis , Animals , Mice , Adipose Tissue/metabolism , Inflammation/metabolism , Macrophages/metabolism , Sepsis/metabolism , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...