Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Mol Metab ; 74: 101749, 2023 08.
Article in English | MEDLINE | ID: mdl-37271337

ABSTRACT

OBJECTIVE: Maresin 1 (MaR1) is a docosahexaenoic acid-derived proresolving lipid mediator with insulin-sensitizing and anti-steatosis properties. Here, we aim to unravel MaR1 actions on brown adipose tissue (BAT) activation and white adipose tissue (WAT) browning. METHODS: MaR1 actions were tested in cultured murine brown adipocytes and in human mesenchymal stem cells (hMSC)-derived adipocytes. In vivo effects of MaR1 were tested in diet-induced obese (DIO) mice and lean WT and Il6 knockout (Il6-/-) mice. RESULTS: In cultured differentiated murine brown adipocytes, MaR1 reduces the expression of inflammatory genes, while stimulates glucose uptake, fatty acid utilization and oxygen consumption rate, along with the upregulation of mitochondrial mass and genes involved in mitochondrial biogenesis and function and the thermogenic program. In Leucine Rich Repeat Containing G Protein-Coupled Receptor 6 (LGR6)-depleted brown adipocytes using siRNA, the stimulatory effect of MaR1 on thermogenic genes was abrogated. In DIO mice, MaR1 promotes BAT remodeling, characterized by higher expression of genes encoding for master regulators of mitochondrial biogenesis and function and iBAT thermogenic activation, together with increased M2 macrophage markers. In addition, MaR1-treated DIO mice exhibit a better response to cold-induced BAT activation. Moreover, MaR1 induces a beige adipocyte signature in inguinal WAT of DIO mice and in hMSC-derived adipocytes. MaR1 potentiates Il6 expression in brown adipocytes and BAT of cold exposed lean WT mice. Interestingly, the thermogenic properties of MaR1 were abrogated in Il6-/- mice. CONCLUSIONS: These data reveal MaR1 as a novel agent that promotes BAT activation and WAT browning by regulating thermogenic program in adipocytes and M2 polarization of macrophages. Moreover, our data suggest that LGR6 receptor is mediating MaR1 actions on brown adipocytes, and that IL-6 is required for the thermogenic effects of MaR1.


Subject(s)
Adipose Tissue, Brown , Docosahexaenoic Acids , Mice , Humans , Animals , Adipose Tissue, Brown/metabolism , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/metabolism , Interleukin-6/metabolism , Adipose Tissue, White/metabolism , Adipocytes, Brown/metabolism
2.
J Physiol Biochem ; 79(2): 451-465, 2023 May.
Article in English | MEDLINE | ID: mdl-37204588

ABSTRACT

Obesity exacerbates aging-induced adipose tissue dysfunction. This study aimed to investigate the effects of long-term exercise on inguinal white adipose tissue (iWAT) and interscapular brown adipose tissue (iBAT) of aged obese mice. Two-month-old female mice received a high-fat diet for 4 months. Then, six-month-old diet-induced obese animals were allocated to sedentarism (DIO) or to a long-term treadmill training (DIOEX) up to 18 months of age. In exercised mice, iWAT depot revealed more adaptability, with an increase in the expression of fatty acid oxidation genes (Cpt1a, Acox1), and an amelioration of the inflammatory status, with a favorable modulation of pro/antiinflammatory genes and lower macrophage infiltration. Additionally, iWAT of trained animals showed an increment in the expression of mitochondrial biogenesis (Pgc1a, Tfam, Nrf1), thermogenesis (Ucp1), and beige adipocytes genes (Cd137, Tbx1). In contrast, iBAT of aged obese mice was less responsive to exercise. Indeed, although an increase in functional brown adipocytes genes and proteins (Pgc1a, Prdm16 and UCP1) was observed, few changes were found on inflammation-related and fatty acid metabolism genes. The remodeling of iWAT and iBAT depots occurred along with an improvement in the HOMA index for insulin resistance and in glucose tolerance. In conclusion, long-term exercise effectively prevented the loss of iWAT and iBAT thermogenic properties during aging and obesity. In iWAT, the long-term exercise program also reduced the inflammatory status and stimulated a fat-oxidative gene profile. These exercise-induced adipose tissue adaptations could contribute to the beneficial effects on glucose homeostasis in aged obese mice.


Subject(s)
Adipose Tissue, Brown , Adipose Tissue, White , Female , Mice , Animals , Adipose Tissue, Brown/metabolism , Mice, Obese , Adipose Tissue, White/metabolism , Obesity/therapy , Obesity/metabolism , Glucose/metabolism , Fatty Acids/metabolism , Thermogenesis/genetics , Mice, Inbred C57BL
3.
J. physiol. biochem ; 79(2)may. 2023. graf
Article in English | IBECS | ID: ibc-222555

ABSTRACT

Obesity exacerbates aging-induced adipose tissue dysfunction. This study aimed to investigate the effects of long-term exercise on inguinal white adipose tissue (iWAT) and interscapular brown adipose tissue (iBAT) of aged obese mice. Two-month-old female mice received a high-fat diet for 4 months. Then, six-month-old diet-induced obese animals were allocated to sedentarism (DIO) or to a long-term treadmill training (DIOEX) up to 18 months of age. In exercised mice, iWAT depot revealed more adaptability, with an increase in the expression of fatty acid oxidation genes (Cpt1a, Acox1), and an amelioration of the inflammatory status, with a favorable modulation of pro/antiinflammatory genes and lower macrophage infiltration. Additionally, iWAT of trained animals showed an increment in the expression of mitochondrial biogenesis (Pgc1a, Tfam, Nrf1), thermogenesis (Ucp1), and beige adipocytes genes (Cd137, Tbx1). In contrast, iBAT of aged obese mice was less responsive to exercise. Indeed, although an increase in functional brown adipocytes genes and proteins (Pgc1a, Prdm16 and UCP1) was observed, few changes were found on inflammation-related and fatty acid metabolism genes. The remodeling of iWAT and iBAT depots occurred along with an improvement in the HOMA index for insulin resistance and in glucose tolerance. In conclusion, long-term exercise effectively prevented the loss of iWAT and iBAT thermogenic properties during aging and obesity. In iWAT, the long-term exercise program also reduced the inflammatory status and stimulated a fat-oxidative gene profile. These exercise-induced adipose tissue adaptations could contribute to the beneficial effects on glucose homeostasis in aged obese mice. (AU)


Subject(s)
Animals , Mice , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Obesity/therapy , Thermogenesis/genetics
4.
J Nutr Biochem ; 111: 109153, 2023 01.
Article in English | MEDLINE | ID: mdl-36150680

ABSTRACT

This study aimed to characterize the potential beneficial effects of chronic docosahexaenoic acid (DHA) supplementation on restoring subcutaneous white adipose tissue (scWAT) plasticity in obese aged female mice. Two-month-old female C57BL/6J mice received a control (CT) or a high fat diet (HFD) for 4 months. Then, 6-month-old diet-induced obese (DIO) mice were distributed into the DIO and the DIOMEG group (fed with a DHA-enriched HFD) up to 18 months. In scWAT, the DHA-enriched diet reduced the mean adipocyte size and reversed the upregulation of lipogenic genes induced by the HFD, reaching values even lower than those observed in CT animals. DIO mice exhibited an up-regulation of lipolytic and fatty oxidation gene expressions that was reversed in DHA-supplemented mice except for Cpt1a mRNA levels, which were higher in DIOMEG as compared to CT mice. DHA restored the increase of proinflammatory genes observed in scWAT of DIO mice. While no changes were observed in total macrophage F4/80+/CD11b+ content, the DHA treatment switched scWAT macrophages profile by reducing the M1 marker Cd11c and increasing the M2 marker CD206. These events occurred alongside with a stimulation of beige adipocyte specific genes, the restoration of UCP1 and pAKT/AKT ratio, and a recovery of the HFD-induced Fgf21 upregulation. In summary, DHA supplementation induced a metabolic remodeling of scWAT to a healthier phenotype in aged obese mice by modulating genes controlling lipid accumulation in adipocytes, reducing the inflammatory status, and inducing beige adipocyte markers in obese aged mice.


Subject(s)
Docosahexaenoic Acids , Obesity , Female , Mice , Animals , Mice, Obese , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/metabolism , Obesity/metabolism , Mice, Inbred C57BL , Adipose Tissue, White/metabolism , Diet, High-Fat/adverse effects , Subcutaneous Fat/metabolism , Dietary Supplements , Adipose Tissue/metabolism
5.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769201

ABSTRACT

Aging usually comes associated with increased visceral fat accumulation, reaching even an obesity state, and favoring its associated comorbidities. One of the processes involved in aging is cellular senescence, which is highly dependent on the activity of the regulators of the cell cycle. The aim of this study was to analyze the changes in the expression of p27 and cdk2 in different adipose tissue depots during aging, as well as their regulation by obesity in mice. Changes in the expression of p27 and CDK2 in visceral and subcutaneous white adipose tissue (WAT) biopsies were also analyzed in a human cohort of obesity and type 2 diabetes. p27, but not cdk2, exhibits a lower expression in subcutaneous than in visceral WAT in mice and humans. p27 is drastically downregulated by aging in subcutaneous WAT (scWAT), but not in gonadal WAT, of female mice. Obesity upregulates p27 and cdk2 expression in scWAT, but not in other fat depots of aged mice. In humans, a significant upregulation of p27 was observed in visceral WAT of subjects with obesity. Taken together, these results show a differential adipose depot-dependent regulation of p27 and cdk2 in aging and obesity, suggesting that p27 and cdk2 could contribute to the adipose-tissue depot's metabolic differences. Further studies are necessary to fully corroborate this hypothesis.


Subject(s)
Adipose Tissue, White/metabolism , Aging/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Obesity/metabolism , Animals , Female , Mice
6.
FASEB J ; 35(6): e21592, 2021 06.
Article in English | MEDLINE | ID: mdl-33960028

ABSTRACT

Brown adipose tissue (BAT) dysfunction in aging and obesity has been related to chronic unresolved inflammation, which could be mediated by an impaired production of specialized proresolving lipid mediators (SPMs), such as Lipoxins-LXs, Resolvins-Rvs, Protectins-PDs, and Maresins-MaRs. Our aim was to characterize the changes in BAT SPMs signatures and their association with BAT dysfunction during aging, especially under obesogenic conditions, and their modulation by a docosahexaenoic acid (DHA)-rich diet. Lipidomic, functional, and molecular studies were performed in BAT of 2- and 18-month-old lean (CT) female mice and in 18-month-old diet-induced obese (DIO) mice fed with a high-fat diet (HFD), or a DHA-enriched HFD. Aging downregulated Prdm16 and UCP1 levels, especially in DIO mice, while DHA partially restored them. Arachidonic acid (AA)-derived LXs and DHA-derived MaRs and PDs were the most abundant SPMs in BAT of young CT mice. Interestingly, the sum of LXs and of PDs were significantly lower in aged DIO mice compared to young CT mice. Some of the SPMs most significantly reduced in obese-aged mice included LXB4 , MaR2, 4S,14S-diHDHA, 10S,17S-diHDHA (a.k.a. PDX), and RvD6. In contrast, DHA increased DHA-derived SPMs, without modifying LXs. However, MicroPET studies showed that DHA was not able to counteract the impaired cold exposure response in BAT of obese-aged mice. Our data suggest that a defective SPMs production could underlie the decrease of BAT activity observed in obese-aged mice, and highlight the relevance to further characterize the physiological role and therapeutic potential of specific SPMs on BAT development and function.


Subject(s)
Adipose Tissue, Brown/metabolism , Aging/pathology , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Lipids/analysis , Obesity/physiopathology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/pathology , Animals , Diet, High-Fat , Female , Lipid Metabolism , Lipidomics , Male , Mice , Mice, Inbred C57BL
7.
Article in English | MEDLINE | ID: mdl-33631309

ABSTRACT

Scavenger receptor class B type 1 (SR-B1) is a membrane lipoprotein receptor/lipid transporter involved in the pathogenesis of atherosclerosis, but its role in obesity and fatty liver development is unclear. Here, we determined the effects of SR-B1 deficiency on plasma metabolic and inflammatory parameters as well as fat deposition in adipose tissue and liver during obesity. To induce obesity, we performed high-fat diet (HFD) exposure for 12 weeks in male SR-B1 knock-out (SR-B1-/-, n = 14) and wild-type (WT, n = 12) mice. Compared to HFD-fed WT mice, plasma from HFD-fed SR-B1-/- animals exhibited increased total cholesterol, triglycerides (TG) and tumor necrosis factor-α (TNF-α) levels. In addition, hypertrophied adipocytes and macrophage-containing crown-like structures (CLS) were observed in adipose tissue from HFD-fed SR-B1 deficient mice. Remarkably, liver from obese SR-B1-/- mice showed attenuated TG content, dysregulation in hepatic peroxisome proliferator-activated receptors (PPARs) expression, increased hepatic TG secretion, and altered hepatic fatty acid (FA) composition. In conclusion, we show that SR-B1 deficiency alters the metabolic environment of obese mice through modulation of liver and adipose tissue lipid accumulation. Our findings provide the basis for further elucidation of SR-B1's role in obesity and fatty liver, two major public health issues that increase the risk of advanced chronic diseases and overall mortality.


Subject(s)
Adipose Tissue/pathology , CD36 Antigens/deficiency , Diet, High-Fat/adverse effects , Fatty Liver/complications , Fatty Liver/metabolism , Obesity/complications , Obesity/etiology , Animals , Disease Susceptibility , Fatty Acids/metabolism , Fatty Liver/pathology , Inflammation/complications , Liver/metabolism , Male , Mice , Triglycerides/metabolism
8.
Nutrients ; 13(2)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546405

ABSTRACT

Obesity and aging are associated to non-alcoholic fatty liver disease (NAFLD) development. Here, we investigate whether long-term feeding with a docosahexaenoic acid (DHA)-enriched diet and aerobic exercise, alone or in combination, are effective in ameliorating NAFLD in aged obese mice. Two-month-old female C57BL/6J mice received control or high fat diet (HFD) for 4 months. Then, the diet-induced obese (DIO) mice were distributed into four groups: DIO, DIO + DHA (15% dietary lipids replaced by a DHA-rich concentrate), DIO + EX (treadmill running), and DIO + DHA + EX up to 18 months. The DHA-rich diet reduced liver steatosis in DIO mice, decreasing lipogenic genes (Dgat2, Scd1, Srebp1c), and upregulated lipid catabolism genes (Hsl/Acox) expression. A similar pattern was observed in the DIO + EX group. The combination of DHA + exercise potentiated an increase in Cpt1a and Ppara genes, and AMPK activation, key regulators of fatty acid oxidation. Exercise, alone or in combination with DHA, significantly reversed the induction of proinflammatory genes (Mcp1, Il6, Tnfα, Tlr4) in DIO mice. DHA supplementation was effective in preventing the alterations induced by the HFD in endoplasmic reticulum stress-related genes (Ern1/Xbp1) and autophagy markers (LC3II/I ratio, p62, Atg7). In summary, long-term DHA supplementation and/or exercise could be helpful to delay NAFLD progression during aging in obesity.


Subject(s)
Aging/physiology , Docosahexaenoic Acids/administration & dosage , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/complications , Physical Conditioning, Animal/physiology , Animals , Autophagy/genetics , Autophagy/physiology , Diet, High-Fat , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Inflammation/genetics , Lipid Metabolism , Lipogenesis/genetics , Liver/chemistry , Liver/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Obesity/etiology , RNA, Messenger/analysis
9.
Cancer Discov ; 11(5): 1268-1285, 2021 05.
Article in English | MEDLINE | ID: mdl-33355179

ABSTRACT

For millions of years, endogenous retroelements have remained transcriptionally silent within mammalian genomes by epigenetic mechanisms. Modern anticancer therapies targeting the epigenetic machinery awaken retroelement expression, inducing antiviral responses that eliminate tumors through mechanisms not completely understood. Here, we find that massive binding of epigenetically activated retroelements by RIG-I and MDA5 viral sensors promotes ATP hydrolysis and depletes intracellular energy, driving tumor killing independently of immune signaling. Energy depletion boosts compensatory ATP production by switching glycolysis to mitochondrial oxidative phosphorylation, thereby reversing the Warburg effect. However, hyperfunctional succinate dehydrogenase in mitochondrial electron transport chain generates excessive oxidative stress that unleashes RIP1-mediated necroptosis. To maintain ATP generation, hyperactive mitochondrial membrane blocks intrinsic apoptosis by increasing BCL2 dependency. Accordingly, drugs targeting BCL2 family proteins and epigenetic inhibitors yield synergistic responses in multiple cancer types. Thus, epigenetic therapy kills cancer cells by rewiring mitochondrial metabolism upon retroelement activation, which primes mitochondria to apoptosis by BH3-mimetics. SIGNIFICANCE: The state of viral mimicry induced by epigenetic therapies in cancer cells remodels mitochondrial metabolism and drives caspase-independent tumor cell death, which sensitizes to BCL2 inhibitor drugs. This novel mechanism underlies clinical efficacy of hypomethylating agents and venetoclax in acute myeloid leukemia, suggesting similar combination therapies for other incurable cancers.This article is highlighted in the In This Issue feature, p. 995.


Subject(s)
Antineoplastic Agents/pharmacology , Epigenesis, Genetic/drug effects , Mitochondria/drug effects , Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Humans
10.
J Physiol Biochem ; 76(2): 251-267, 2020 May.
Article in English | MEDLINE | ID: mdl-31853728

ABSTRACT

Adipose tissue dysfunction represents the hallmark of obesity. Brown/beige adipose tissues play a crucial role in maintaining energy homeostasis through non-shivering thermogenesis. Brown adipose tissue (BAT) activity has been inversely related to body fatness, suggesting that BAT activation is protective against obesity. BAT plays also a key role in the control of triglyceride clearance, glucose homeostasis, and insulin sensitivity. Therefore, BAT/beige activation has been proposed as a strategy to prevent or ameliorate obesity development and associated commorbidities. In the last few years, a variety of preclinical studies have proposed n-3 polyunsaturated fatty acids (n-3 PUFAs) as novel inducers of BAT activity and white adipose tissue browning. Here, we review the in vitro and in vivo available evidences of the thermogenic properties of n-3 PUFAs, especially focusing on the molecular and cellular physiological mechanisms involved. Finally, we also discuss the challenges and future perspectives to better characterize the therapeutic potential of n-3 PUFAs as browning agents, especially in humans.


Subject(s)
Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Fatty Acids, Omega-3 , Obesity , Animals , Cells, Cultured , Energy Metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/physiology , Humans , Mice , Obesity/drug therapy , Obesity/metabolism , Rats , Thermogenesis
11.
Nutrients ; 11(4)2019 Apr 18.
Article in English | MEDLINE | ID: mdl-31003450

ABSTRACT

Aging is a complex phenomenon characterized by the progressive loss of tissue and organ function. The oxidative-stress theory of aging postulates that age-associated functional losses are due to the accumulation of ROS-induced damage. Liver function impairment and non-alcoholic fatty liver disease (NAFLD) are common among the elderly. NAFLD can progress to non-alcoholic steatohepatitis (NASH) and evolve to hepatic cirrhosis or hepatic carcinoma. Oxidative stress, lipotoxicity, and inflammation play a key role in the progression of NAFLD. A growing body of evidence supports the therapeutic potential of omega-3 polyunsaturated fatty acids (n-3 PUFA), mainly docosahaexenoic (DHA) and eicosapentaenoic acid (EPA), on metabolic diseases based on their antioxidant and anti-inflammatory properties. Here, we performed a systematic review of clinical trials analyzing the efficacy of n-3 PUFA on both systemic oxidative stress and on NAFLD/NASH features in adults. As a matter of fact, it remains controversial whether n-3 PUFA are effective to counteract oxidative stress. On the other hand, data suggest that n-3 PUFA supplementation may be effective in the early stages of NAFLD, but not in patients with more severe NAFLD or NASH. Future perspectives and relevant aspects that should be considered when planning new randomized controlled trials are also discussed.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Oxidative Stress/drug effects , Aging , Humans
12.
Biol Rev Camb Philos Soc ; 93(2): 1145-1164, 2018 05.
Article in English | MEDLINE | ID: mdl-29230933

ABSTRACT

The discovery of metabolically active brown adipose tissue (BAT) in adult humans has fuelled the research of diverse aspects of this previously neglected tissue. BAT is solely present in mammals and its clearest physiological role is non-shivering thermogenesis, owing to the capacity of brown adipocytes to dissipate metabolic energy as heat. Recently, a number of other possible functions have been proposed, including direct regulation of glucose and lipid homeostasis and the secretion of a number of factors with diverse regulatory actions. Herein, we review recent advances in general biological knowledge of BAT and discuss the possible implications of this tissue in human metabolic health. In particular, we confront the claimed thermogenic potential of BAT for human energy balance and body mass regulation, mostly based on animal studies, with the most recent quantifications of human BAT.


Subject(s)
Adipose Tissue, Brown/physiology , Obesity/pathology , Animals , Humans , Obesity/metabolism , Obesity/prevention & control
13.
Biofactors ; 43(1): 117-131, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-27507611

ABSTRACT

In obesity, the increment of adiposity levels disrupts the whole body homeostasis, promoting an over production of oxidants and inflammatory mediators. The current study aimed to characterize the transcriptomic changes promoted by supplementation with eicosapentaenoic acid (EPA, 1.3 g/day), α-lipoic acid (0.3 g/day), or both (EPA + α-lipoic acid, 1.3 g/day + 0.3 g/day) in subcutaneous abdominal adipose tissue from overweight/obese healthy women, who followed a hypocaloric diet (30% of total energy expenditure) during ten weeks, by using a microarray approach. At the end of the intervention, a total of 33,297 genes were analyzed using Affymetrix GeneChip arrays. EPA promoted changes in extracellular matrix remodeling gene expression, besides a rise of genes associated with either chemotaxis or wound repair. α-Lipoic acid decreased expression of genes related with cell adhesion and inflammation. Furthermore, α-lipoic acid, especially in combination with EPA, upregulated the expression of genes associated with lipid catabolism while downregulated genes involved in lipids storage. Together, all these data suggest that some of the metabolic effects of EPA and α-lipoic acid could be related to their regulatory actions on adipose tissue metabolism. © 2016 BioFactors, 43(1):117-131, 2017.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Caloric Restriction , Eicosapentaenoic Acid/administration & dosage , Obesity/diet therapy , Thioctic Acid/administration & dosage , Transcriptome , Abdominal Fat/metabolism , Administration, Oral , Adult , Dietary Supplements , Female , Humans , Metabolic Networks and Pathways , Middle Aged , Obesity/metabolism
14.
Biochim Biophys Acta ; 1861(3): 260-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26721419

ABSTRACT

Chemerin is a novel adipokine associated with obesity and insulin resistance. α-Lipoic acid (α-LA) has shown beneficial properties on diabetes and obesity. The aim of this study was to examine the effects of α-LA on chemerin production in adipocytes in absence or presence of TNF-α, insulin and AICAR. The potential signaling pathways involved in α-LA effects on chemerin were also analyzed. α-LA actions on chemerin were tested in differentiated 3T3-L1 adipocytes and in some cases in human subcutaneous and omental adipocytes. Chemerin mRNA levels were measured by RT-PCR and the amount of chemerin secreted to culture media was determined by ELISA. α-LA induced a concentration-dependent inhibition on both chemerin secretion and mRNA levels in 3T3-L1 adipocytes. The AMPK activator AICAR and the PI3K inhibitor LY294002 dramatically abrogated both chemerin secretion and gene expression, and further potentiated the inhibitory effect of α-LA on chemerin secretion. Insulin was able to partially reverse the inhibitory action of α-LA on chemerin secretion. α-LA also reduced basal chemerin secretion in both subcutaneous and omental adipocytes from overweight/obese subjects. Moreover, α-LA was able to abolish the stimulatory effects of the pro-inflammatory cytokine TNF-α on chemerin secretion. Our data demonstrated the ability of α-LA to inhibit chemerin production, an adipokine associated to obesity and metabolic syndrome, suggesting that the reduction of chemerin could contribute to the antiobesity/antidiabetic properties described for α-LA.


Subject(s)
Adipocytes/drug effects , Anti-Obesity Agents/pharmacology , Chemokines/metabolism , Hypoglycemic Agents/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Thioctic Acid/pharmacology , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Chemokines/blood , Chemokines/genetics , Dose-Response Relationship, Drug , Down-Regulation , Humans , Insulin/pharmacology , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , PPAR gamma/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats, Wistar , Ribonucleotides/pharmacology , Signal Transduction/drug effects , Transfection , Tumor Necrosis Factor-alpha/pharmacology
15.
Biochem Biophys Res Commun ; 467(1): 39-45, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26417690

ABSTRACT

AIMS: Mutations in 1-acylglycerol-3-phosphate O-acyltransferase 2 (AGPAT2) result in lipodystrophy, insulin resistance and diabetes. Autophagy is required for normal adipogenesis and adipose tissue development. The aim of this study was to determine whether impaired autophagy or excessive cell death underlie the adipogenic inability of Agpat2(-/-) mice preadipocytes. METHODS: Preadipocytes were isolated from interscapular brown adipose tissue (BAT) of Agpat2(-/-) and Agpat2(+/+) newborn mice and cultured/differentiated in vitro. Intracellular lipids were quantified by oil red O staining. Cell death was assessed by lactate dehydrogenase (LDH) activity. Apoptosis and autophagy regulatory factors were determined at the mRNA and protein level with Real-time PCR, immunoblot and immunofluorescence. RESULTS: Adipogenically induced Agpat2(-/-) preadipocytes had fewer lipid-loaded cells and lower levels of adipocyte markers than wild type preadipocytes. Before adipogenic differentiation, autophagy-related proteins (ATGs) ATG3, ATG5-ATG12 complex, ATG7 and LC3II were increased but autophagic flux was reduced, as suggested by increased p62 levels, in Agpat2(-/-) preadipocytes. Adipogenic induction increased LDH levels in the culture media in Agpat2(-/-) preadipocytes but no differences were observed in the activation of Caspase 3 or in markers of autophagic flux. CONCLUSIONS: AGPAT2 is required for in vitro adipogenesis of mouse preadipocytes. Autophagy defects or apoptosis are not involved in the adipogenic failure of Agpat2(-/-) preadipocytes.


Subject(s)
Acyltransferases/deficiency , Adipocytes, Brown/cytology , Adipocytes, Brown/enzymology , Adipogenesis/physiology , Acyltransferases/genetics , Adipogenesis/genetics , Animals , Apoptosis , Autophagy , Cell Differentiation , Cells, Cultured , Lipid Metabolism , Mice , Mice, Knockout , Stem Cells/cytology , Stem Cells/metabolism
16.
J. physiol. biochem ; 71(3): 471-478, sept. 2015. tab
Article in English | IBECS | ID: ibc-142443

ABSTRACT

Lipodystrophy encompass a group of heterogeneous disorders consisting in marked reduction, absence, and/or the redistribution of adipose tissue. Lipodystrophy is frequently complicated with severe insulin resistance, diabetes, hyperlipidemia, and fatty liver. Anatomically, lipodystrophies can be partial or generalized. Etiologically, they can be congenital or acquired. Lipodystrophy diagnosis can be challenging, and it has been suggested that partial forms can be easily misdiagnosed as common central obesity with associated metabolic syndrome. Conventional insulin-sensitizing approaches usually fail to fully ameliorate insulin resistance in lipodystrophic patients. Leptin replacement is an approved therapy for the metabolic complications of congenital generalized lipodystrophy. Novel nutritional interventions are promising complementary approaches for treating lipodystrophy metabolic complications


Subject(s)
Humans , Lipodystrophy/physiopathology , Metabolic Diseases/physiopathology , Adipose Tissue/metabolism , Insulin Resistance , Hypertriglyceridemia/physiopathology , Hypertriglyceridemic Waist/physiopathology , Leptin
17.
J Physiol Biochem ; 71(3): 471-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25833179

ABSTRACT

Lipodystrophy encompass a group of heterogeneous disorders consisting in marked reduction, absence, and/or the redistribution of adipose tissue. Lipodystrophy is frequently complicated with severe insulin resistance, diabetes, hyperlipidemia, and fatty liver. Anatomically, lipodystrophies can be partial or generalized. Etiologically, they can be congenital or acquired. Lipodystrophy diagnosis can be challenging, and it has been suggested that partial forms can be easily misdiagnosed as common central obesity with associated metabolic syndrome. Conventional insulin-sensitizing approaches usually fail to fully ameliorate insulin resistance in lipodystrophic patients. Leptin replacement is an approved therapy for the metabolic complications of congenital generalized lipodystrophy. Novel nutritional interventions are promising complementary approaches for treating lipodystrophy metabolic complications.


Subject(s)
Adipose Tissue/metabolism , Lipodystrophy/metabolism , Adipose Tissue/pathology , Animals , Humans , Insulin Resistance , Lipodystrophy/drug therapy , Lipodystrophy/etiology
18.
Biochim Biophys Acta ; 1851(3): 273-81, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25542506

ABSTRACT

α-Lipoic acid (α-Lip) is a natural occurring antioxidant with beneficial anti-obesity properties. The aim of this study was to investigate the putative effects of α-Lip on mitochondrial biogenesis and the acquirement of brown-like characteristics by subcutaneous adipocytes from overweight/obese subjects. Thus, fully differentiated human subcutaneous adipocytes were treated with α-Lip (100 and 250µM) for 24h for studies on mitochondrial content and morphology, mitochondrial DNA (mtDNA) copy number, fatty acid oxidation enzymes and brown/beige characteristic genes. The involvement of the Sirtuin1/Peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (SIRT1/PGC-1α) pathway was also evaluated. Our results showed that α-Lip increased mitochondrial content in cultured human adipocytes as revealed by electron microscopy and by mitotracker green labeling. Moreover, an enhancement in mtDNA content was observed. This increase was accompanied by an up-regulation of SIRT1 protein levels, a decrease in PGC-1α acetylation and up-regulation of Nuclear respiratory factor 1 (Nrf1) and Mitochondrial transcription factor (Tfam) transcription factors. Enhanced oxygen consumption and fatty acid oxidation enzymes, Carnitine palmitoyl transferase 1 and Acyl-coenzyme A oxidase (CPT-1 and ACOX) were also observed. Mitochondria from α-Lip-treated adipocytes exhibited some morphological characteristics of brown mitochondria, and α-Lip also induced up-regulation of some brown/beige adipocytes markers such as cell death-inducing DFFA-like effector a (Cidea) and T-box 1 (Tbx1). Moreover, α-Lip up-regulated PR domain containing 16 (Prdm16) mRNA levels in treated adipocytes. Therefore, our study suggests the ability of α-Lip to promote mitochondrial biogenesis and brown-like remodeling in cultured white subcutaneous adipocytes from overweight/obese donors.


Subject(s)
Adipocytes, Brown/drug effects , Antioxidants/pharmacology , Mitochondria/drug effects , Mitochondrial Turnover/drug effects , Thioctic Acid/pharmacology , Acetylation/drug effects , Adipocytes, Brown/metabolism , Adipocytes, Brown/pathology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Differentiation , DNA Copy Number Variations/drug effects , DNA, Mitochondrial/agonists , DNA, Mitochondrial/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Obesity/metabolism , Obesity/pathology , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxygen Consumption/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Primary Cell Culture , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , Subcutaneous Fat/pathology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
J Lipid Res ; 55(12): 2634-43, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25351614

ABSTRACT

Cardiotrophin-1 (CT-1) is a cytokine with antiobesity properties and with a role in lipid metabolism regulation and adipose tissue function. The aim of this study was to analyze the molecular mechanisms involved in the lipolytic actions of CT-1 in adipocytes. Recombinant CT-1 (rCT-1) effects on the main proteins and signaling pathways involved in the regulation of lipolysis were evaluated in 3T3-L1 adipocytes and in mice. rCT-1 treatment stimulated basal glycerol release in a concentration- and time-dependent manner in 3T3-L1 adipocytes. rCT-1 (20 ng/ml for 24 h) raised cAMP levels, and in parallel increased protein kinase (PK)A-mediated phosphorylation of perilipin and hormone sensitive lipase (HSL) at Ser660. siRNA knock-down of HSL or PKA, as well as pretreatment with the PKA inhibitor H89, blunted the CT-1-induced lipolysis, suggesting that the lipolytic action of CT-1 in adipocytes is mainly mediated by activation of HSL through the PKA pathway. In ob/ob mice, acute rCT-1 treatment also promoted PKA-mediated phosphorylation of perilipin and HSL at Ser660 and Ser563, and increased adipose triglyceride lipase (desnutrin) content in adipose tissue. These results showed that the ability of CT-1 to regulate the activity of the main lipases underlies the lipolytic action of this cytokine in vitro and in vivo, and could contribute to CT-1 antiobesity effects.


Subject(s)
Adipocytes, White/metabolism , Carrier Proteins/metabolism , Cytokines/metabolism , Lipase/metabolism , Lipolysis , Phosphoproteins/metabolism , Sterol Esterase/metabolism , Up-Regulation , 3T3-L1 Cells , Adipocytes, White/drug effects , Adipocytes, White/enzymology , Animals , Carrier Proteins/biosynthesis , Cell Cycle Proteins/agonists , Cell Cycle Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/genetics , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Gene Silencing , Lipase/antagonists & inhibitors , Lipase/chemistry , Lipolysis/drug effects , Male , Mice , Mice, Mutant Strains , Perilipin-1 , Phosphoproteins/biosynthesis , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Sterol Esterase/antagonists & inhibitors , Sterol Esterase/chemistry , Sterol Esterase/genetics , Up-Regulation/drug effects
20.
Obesity (Silver Spring) ; 22(10): 2210-5, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25045030

ABSTRACT

OBJECTIVE: α-Lipoic acid (α-LA) is a natural occurring antioxidant with beneficial effects on obesity. The aim of this study was to investigate the putative effects of α-LA on triglyceride accumulation and lipogenesis in subcutaneous adipocytes from overweight/obese subjects and to determine the potential mechanisms involved. METHODS: Fully differentiated human subcutaneous adipocytes were treated with α-LA (100 and 250 µM) during 24 h for studying triglyceride content, de novo lipogenesis, and levels of key lipogenic enzymes. The involvement of AMP-activated protein kinase (AMPK) activation was also evaluated. RESULTS: α-LA down-regulated triglyceride content by inhibiting fatty acid esterification and de novo lipogenesis. These effects were mediated by reduction in fatty acid synthase (FAS), stearoyl-coenzyme A desaturase 1, and diacylglycerol O-acyltransferase 1 protein levels. Interestingly, α-LA increased AMPK and acetyl CoA carboxylase phosphorylation, while the presence of the AMPK inhibitor Compound C reversed the inhibition observed on FAS protein levels. CONCLUSIONS: α-LA down-regulates key lipogenic enzymes, inhibiting lipogenesis and reducing triglyceride accumulation through the activation of AMPK signaling pathway in human subcutaneous adipocytes from overweight/obese subjects.


Subject(s)
Adipocytes/drug effects , Fatty Acids/metabolism , Lipid Metabolism/drug effects , Lipogenesis/drug effects , Obesity/metabolism , Overweight/metabolism , Thioctic Acid/pharmacology , Adipocytes/metabolism , Adipocytes/pathology , Cells, Cultured , Down-Regulation/drug effects , Esterification/drug effects , Female , Humans , Obesity/pathology , Overweight/pathology , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , Subcutaneous Fat/pathology , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...