39 results on '"human adipocytes"'
Search Results
2. Arsenic induces metabolome remodeling in mature human adipocytes.
- Author
-
Gasser M, Lenglet S, Bararpour N, Sajic T, Vaucher J, Wiskott K, Augsburger M, Fracasso T, Gilardi F, and Thomas A
- Subjects
- Humans, Adipose Tissue metabolism, Adipocytes, Insulin metabolism, Metabolome, Arsenic metabolism, Diabetes Mellitus, Type 2, Insulin Resistance
- Abstract
Human lifetime exposure to arsenic through drinking water, food supply or industrial pollution leads to its accumulation in many organs such as liver, kidneys, lungs or pancreas but also adipose tissue. Recently, population-based studies revealed the association between arsenic exposure and the development of metabolic diseases such as obesity and type 2 diabetes. To shed light on the molecular bases of such association, we determined the concentration that inhibited 17% of cell viability and investigated the effects of arsenic acute exposure on adipose-derived human mesenchymal stem cells differentiated in vitro into mature adipocytes and treated with sodium arsenite (NaAsO
2 , 10 nM to 10 µM). Untargeted metabolomics and gene expression analyses revealed a strong dose-dependent inhibition of lipogenesis and lipolysis induction, reducing the cellular ability to store lipids. These dysregulations were emphasized by the inhibition of the cellular response to insulin, as shown by the perturbation of several genes and metabolites involved in the mentioned biological pathways. Our study highlighted the activation of an adaptive oxidative stress response with the strong induction of metallothioneins and increased glutathione levels in response to arsenic accumulation that could exacerbate the decreased insulin sensitivity of the adipocytes. Arsenic exposure strongly affected the expression of arsenic transporters, responsible for arsenic influx and efflux, and induced a pro-inflammatory state in adipocytes by enhancing the expression of the inflammatory interleukin 6 (IL6). Collectively, our data showed that an acute exposure to low levels of arsenic concentrations alters key adipocyte functions, highlighting its contribution to the development of insulin resistance and the pathogenesis of metabolic disorders., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
3. Trifuhalol A, a phlorotannin from the brown algae Agarum cribrosum, reduces adipogenesis of human primary adipocytes through Wnt/β-catenin and AMPK-dependent pathways.
- Author
-
Kim AT and Park Y
- Abstract
Trifuhalol A, a fucol-type phlorotannin, was extracted and identified from the brown algae Agarum cribrosum . The total yield and purity of trifuhalol A from A. cribrosum were 0.98% and 86%, respectively. Trifuhalol A at 22 and 44 μM inhibited lipid accumulation in human primary adipocytes. Consistently trifuhalol A suppressed the expression of adipogenesis-related genes, such as proliferator-activated receptor-gamma (PPAR-γ), CCAAT/enhancer-binding protein-alpha (C/EBP-α), fatty acid synthase (FAS), and sterol regulatory element-binding protein-1 (SREBP-1), in a dose-dependent manner. Trifuhalol A increased the level of proteins such as wingless/integrated (Wnt)10b, nuclear-β-catenin, total-β-catenin, phospho-AMP-activated protein kinase ( p AMPK), and phospho-liver kinase B1 ( p LKB1) as well as the expression of genes such as Wnt10b, Frizzled 1, and low-density lipoprotein receptor-related protein 6 (LRP6). Additionally, trifuhalol A decreased the expression of the glycogen synthase kinase-3beta (GSK3β) gene. These results suggest that trifuhalol A reduces fat accumulation in human adipocytes via the Wnt/β-catenin- and AMPK-dependent pathways., Competing Interests: The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Given her role as the Associate Editor at the time of submission, Dr. Yeonhwa Park was not involved in the peer review of this article and had no access to information regarding its peer review. Full responsibility for the editorial process for this article was delegated to another editor, as per the Journal guidelines. The authors declare that they have no other known competing financial interests that could have appeared to influence the work reported in this paper., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
4. Non-aromatizable androgens modulate the lipopolysaccharide induced expression of the P2X7 receptor in human adipocytes.
- Author
-
Di Vincenzo A, Granzotto M, Crescenzi M, Vettor R, and Rossato M
- Abstract
Introduction: The activation of the P2X7 receptor subtype (P2X7R) has a main role in orchestrating the cellular inflammatory response in many different tissues. Obesity is characterized by dysfunctional fat deposition leading to a tissue-specific and systemic low-grade inflammation. Androgens and estrogens contribute to the whole adipose tissue inflammatory state, but the involvement of sex steroids in the purinergic signaling modulation in adipocytes is still unknown. Methods: We performed an in vitro study to evaluate the possible role of sex hormones on the P2X7R gene expression in human adipocytes, at baseline and after stimulation with bacterial lipopolysaccharide (LPS). We evaluated P2X7R gene expression during in vitro differentiation of human adipocytes, in the absence and presence of testosterone (T) and 17β-estradiol (E2) in the presence and absence of LPS. Furthermore, we analyzed the effects of incubation with dihydrotestosterone (DHT), a non-aromatizable androgen, using the co-incubation of isolated human adipocytes with T alone or in combination with anastrozole, an inhibitor of aromatase, the enzyme responsible of T conversion to E2. Results: At baseline, incubation of adipocytes with T or E2 did not significantly affect P2X7R gene expression. On the contrary, the incubation with DHT was associated with a significant reduction of P2X7R gene expression. LPS incubation significantly increased gene expression of P2X7R with respect to baseline. Interestingly, after LPS stimulation, DHT exposure showed an additional effect, markedly increasing the P2X7R gene expression. This amplificatory effect was confirmed by the incubation of adipocytes to both anastrozole and testosterone. In these experimental conditions, while no effect was observed at baseline, an amplification of the expression of the P2X7R mRNA was observed after stimulation with LPS. Discussion: The purinergic system is involved in the inflammatory response of adipocytes, and androgens may modulate its activity. In particular DHT, a non-aromatizable androgen, amplifies the LPS-induced P2X7R gene expression in human adipocytes thus showing a gender regulated response of the expression of this purinergic receptor strongly involved in the inflammatory response in adipose tissue., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Di Vincenzo, Granzotto, Crescenzi, Vettor and Rossato.)
- Published
- 2023
- Full Text
- View/download PDF
5. Regulation of Human Sortilin Alternative Splicing by Glucagon-like Peptide-1 (GLP1) in Adipocytes.
- Author
-
Lui A, Patel RS, Krause-Hauch M, Sparks RP, and Patel NA
- Subjects
- Humans, Adipocytes, Glucagon-Like Peptide 1 genetics, Glucose, Alternative Splicing, Diabetes Mellitus, Type 2
- Abstract
Type 2 diabetes mellitus is a chronic metabolic disease with no cure. Adipose tissue is a major site of systemic insulin resistance. Sortilin is a central component of the glucose transporter -Glut4 storage vesicles (GSV) which translocate to the plasma membrane to uptake glucose from circulation. Here, using human adipocytes we demonstrate the presence of the alternatively spliced, truncated sortilin variant (Sort_T) whose expression is significantly increased in diabetic adipose tissue. Artificial-intelligence-based modeling, molecular dynamics, intrinsically disordered region analysis, and co-immunoprecipitation demonstrated association of Sort_T with Glut4 and decreased glucose uptake in adipocytes. The results show that glucagon-like peptide-1 (GLP1) hormone decreases Sort_T. We deciphered the molecular mechanism underlying GLP1 regulation of alternative splicing of human sortilin. Using splicing minigenes and RNA-immunoprecipitation assays, the results show that GLP1 regulates Sort_T alternative splicing via the splice factor, TRA2B. We demonstrate that targeted antisense oligonucleotide morpholinos reduces Sort_T levels and improves glucose uptake in diabetic adipocytes. Thus, we demonstrate that GLP1 regulates alternative splicing of sortilin in human diabetic adipocytes.
- Published
- 2023
- Full Text
- View/download PDF
6. Availability of abundant thiamine determines efficiency of thermogenic activation in human neck area derived adipocytes.
- Author
-
Arianti R, Vinnai BÁ, Győry F, Guba A, Csősz É, Kristóf E, and Fésüs L
- Subjects
- Humans, Adipose Tissue, Brown, Membrane Transport Proteins, Cell Differentiation, Thermogenesis genetics, Uncoupling Protein 1 genetics, Thiamine, Adipocytes, Brown
- Abstract
Brown/beige adipocytes express uncoupling protein-1 (UCP1) that enables them to dissipate energy as heat. Systematic activation of this process can alleviate obesity. Human brown adipose tissues are interspersed in distinct anatomical regions including deep neck. We found that UCP1 enriched adipocytes differentiated from precursors of this depot highly expressed ThTr2 transporter of thiamine and consumed thiamine during thermogenic activation of these adipocytes by cAMP which mimics adrenergic stimulation. Inhibition of ThTr2 led to lower thiamine consumption with decreased proton leak respiration reflecting reduced uncoupling. In the absence of thiamine, cAMP-induced uncoupling was diminished but restored by thiamine addition reaching the highest levels at thiamine concentrations larger than present in human blood plasma. Thiamine is converted to thiamine pyrophosphate (TPP) in cells; the addition of TPP to permeabilized adipocytes increased uncoupling fueled by TPP-dependent pyruvate dehydrogenase. ThTr2 inhibition also hampered cAMP-dependent induction of UCP1, PGC1a, and other browning marker genes, and thermogenic induction of these genes was potentiated by thiamine in a concentration-dependent manner. Our study reveals the importance of amply supplied thiamine during thermogenic activation in human adipocytes which provides TPP for TPP-dependent enzymes not fully saturated with this cofactor and by potentiating the induction of thermogenic genes., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Endre Kristóf and László Fésüs reports financial support was provided by National, Research, Development and Innovation Office. László Fésüs reports financial support was provided by European Regional Development Fund. Endre Kristóf and László Fésüs reports a relationship with National, Research, Development and Innovation Office that includes: funding grants. László Fésüs reports a relationship with European Regional Development Fund that includes: funding grants., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
7. The impact of metabolic endotoxaemia on the browning process in human adipocytes.
- Author
-
Omran F, Murphy AM, Younis AZ, Kyrou I, Vrbikova J, Hainer V, Sramkova P, Fried M, Ball G, Tripathi G, Kumar S, McTernan PG, and Christian M
- Subjects
- Humans, Adipocytes metabolism, Obesity metabolism, Lipopolysaccharides, Endotoxins metabolism, Diabetes Mellitus, Type 2, Endotoxemia metabolism
- Abstract
Background: Dysfunctional adipose tissue (AT) is known to contribute to the pathophysiology of metabolic disease, including type 2 diabetes mellitus (T2DM). This dysfunction may occur, in part, as a consequence of gut-derived endotoxaemia inducing changes in adipocyte mitochondrial function and reducing the proportion of BRITE (brown-in-white) adipocytes. Therefore, the present study investigated whether endotoxin (lipopolysaccharide; LPS) directly contributes to impaired human adipocyte mitochondrial function and browning in human adipocytes, and the relevant impact of obesity status pre and post bariatric surgery., Methods: Human differentiated abdominal subcutaneous (AbdSc) adipocytes from participants with obesity and normal-weight participants were treated with endotoxin to assess in vitro changes in mitochondrial function and BRITE phenotype. Ex vivo human AbdSc AT from different groups of participants (normal-weight, obesity, pre- and 6 months post-bariatric surgery) were assessed for similar analyses including circulating endotoxin levels., Results: Ex vivo AT analysis (lean & obese, weight loss post-bariatric surgery) identified that systemic endotoxin negatively correlated with BAT gene expression (p < 0.05). In vitro endotoxin treatment of AbdSc adipocytes (lean & obese) reduced mitochondrial dynamics (74.6% reduction; p < 0.0001), biogenesis (81.2% reduction; p < 0.0001) and the BRITE phenotype (93.8% reduction; p < 0.0001). Lean AbdSc adipocytes were more responsive to adrenergic signalling than obese AbdSc adipocytes; although endotoxin mitigated this response (92.6% reduction; p < 0.0001)., Conclusions: Taken together, these data suggest that systemic gut-derived endotoxaemia contributes to both individual adipocyte dysfunction and reduced browning capacity of the adipocyte cell population, exacerbating metabolic consequences. As bariatric surgery reduces endotoxin levels and is associated with improving adipocyte functionality, this may provide further evidence regarding the metabolic benefits of such surgical interventions., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
8. RNA-seq reveals that anti-obesity irisin and triiodothyronine (T3) hormones differentially affect the purinergic signaling transcriptomics in differentiated human adipocytes.
- Author
-
Mathias LS, Herman-de-Sousa C, Cury SS, Nogueira CR, Correia-de-Sá P, and de Oliveira M
- Subjects
- Humans, Adipocytes drug effects, Adipocytes metabolism, Obesity genetics, Obesity metabolism, RNA-Seq, Fibronectins genetics, Fibronectins metabolism, Transcriptome, Triiodothyronine pharmacology, Triiodothyronine metabolism
- Abstract
The anti-obesity thyroid hormone, triiodothyronine (T3), and irisin, an exercise- and/or cold-induced myokine, stimulate thermogenesis and energy consumption while decreasing lipid accumulation. The involvement of ATP signaling in adipocyte cell function and obesity has attracted increasing attention, but the crosstalk between the purinergic signaling cascade and anti-obesity hormones lacks experimental evidence. In this study, we investigated the effects of T3 and irisin in the transcriptomics of membrane-bound purinoceptors, ectonucleotidase enzymes and nucleoside transporters participating in the purinergic signaling in cultured human adipocytes. The RNA-seq analysis revealed that differentiated adipocytes express high amounts of ADORA1, P2RY11, P2RY12, and P2RX6 gene transcripts, along with abundant levels of transcriptional products encoding to purine metabolizing enzymes (ENPP2, ENPP1, NT5E, ADA and ADK) and transporters (SLC29A1, SCL29A2). The transcriptomics of purinergic signaling markers changed in parallel to the upsurge of "browning" adipocyte markers, like UCP1 and P2RX5, after treatment with T3 and irisin. Upregulation of ADORA1, ADORA2A and P2RX4 gene transcription was obtained with irisin, whereas T3 preferentially upregulated NT5E, SLC29A2 and P2RY11 genes. Irisin was more powerful than T3 towards inhibition of the leptin gene transcription, the SCL29A1 gene encoding for the ENT1 transporter, the E-NPP2 (autotaxin) gene, and genes that encode for two ADP-sensitive P2Y receptors, P2RY1 and P2RY12. These findings indicate that anti-obesity irisin and T3 hormones differentially affect the purinergic signaling transcriptomics, which might point towards new directions for the treatment of obesity and related metabolic disorders that are worth to be pursued in future functional studies., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
9. Human visceral and subcutaneous adipose stem and progenitor cells retain depot-specific adipogenic properties during obesity.
- Author
-
Mathur N, Severinsen MCK, Jensen ME, Naver L, Schrölkamp M, Laye MJ, Watt MJ, Nielsen S, Krogh-Madsen R, Pedersen BK, and Scheele C
- Abstract
Abdominal obesity associates with cardiometabolic disease and an accumulation of lipids in the visceral adipose depot, whereas lipid accumulation in the subcutaneous depot is more benign. We aimed to further investigate whether the adipogenic properties where cell-intrinsic, or dependent on a depot-specific or obesity-produced microenvironment. We obtained visceral and subcutaneous biopsies from non-obese women ( n = 14) or women living with morbid obesity ( n = 14) and isolated adipose stem and progenitor cells (ASPCs) from the stromal vascular fraction of non-obese ( n = 13) and obese ( n = 13). Following in vitro differentiation into mature adipocytes, we observed a contrasting pattern with a lower gene expression of adipogenic markers and a higher gene expression of immunogenic markers in the visceral compared to the subcutaneous adipocytes. We identified the immunogenic factor BST2 as a marker for visceral ASPCs. The effect of obesity and insulin resistance on adipogenic and immunogenic markers in the in vitro differentiated cells was minor. In contrast, differentiation with exogenous Tumor necrosis factor resulted in increased immunogenic signatures, including increased expression of BST2 , and decreased adipogenic signatures in cells from both depots. Our data, from 26 women, underscore the intrinsic differences between human visceral and subcutaneous adipose stem and progenitor cells, suggest that dysregulation of adipocytes in obesity mainly occurs at a post-progenitor stage, and highlight an inflammatory microenvironment as a major constraint of human adipogenesis., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Mathur, Severinsen, Jensen, Naver, Schrölkamp, Laye, Watt, Nielsen, Krogh-Madsen, Pedersen and Scheele.)
- Published
- 2022
- Full Text
- View/download PDF
10. Effects of Chemical Structures Interacting with Amine Oxidases on Glucose, Lipid and Hydrogen Peroxide Handling by Human Adipocytes.
- Author
-
Carpéné C, Viana P, Iffiú-Soltesz Z, Tapolcsányi P, Földi AÁ, Mátyus P, and Dunkel P
- Subjects
- Adipocytes, Benzylamines metabolism, Benzylamines pharmacology, Glucose metabolism, Hexoses pharmacology, Humans, Hydrogen Peroxide pharmacology, Insulin metabolism, Lipids pharmacology, Monoamine Oxidase metabolism, Monoamine Oxidase Inhibitors pharmacology, Triglycerides metabolism, Amine Oxidase (Copper-Containing) metabolism
- Abstract
Benzylamine is a natural molecule present in food and edible plants, capable of activating hexose uptake and inhibiting lipolysis in human fat cells. These effects are dependent on its oxidation by amine oxidases present in adipocytes, and on the subsequent hydrogen peroxide production, known to exhibit insulin-like actions. Virtually, other substrates interacting with such hydrogen peroxide-releasing enzymes potentially can modulate lipid accumulation in adipose tissue. Inhibition of such enzymes has also been reported to influence lipid deposition. We have therefore studied in human adipocytes the lipolytic and lipogenic activities of pharmacological entities designed to interact with amine oxidases highly expressed in this cell type: the semicarbazide-sensitive amine oxidase (SSAO also known as PrAO or VAP-1) and the monoamine oxidases (MAO). The results showed that SZV-2016 and SZV-2017 behaved as better substrates than benzylamine, releasing hydrogen peroxide once oxidized, and reproduced or even exceeded its insulin-like metabolic effects in fat cells. Additionally, several novel SSAO inhibitors, such as SZV-2007 and SZV-1398, have been evidenced and shown to inhibit benzylamine metabolic actions. Taken as a whole, our findings reinforce the list of molecules that influence the regulation of triacylglycerol assembly/breakdown, at least in vitro in human adipocytes. The novel compounds deserve deeper investigation of their mechanisms of interaction with SSAO or MAO, and constitute potential candidates for therapeutic use in obesity and diabetes.
- Published
- 2022
- Full Text
- View/download PDF
11. CYP1A1, VEGFA and Adipokine Responses of Human Adipocytes Co-exposed to PCB126 and Hypoxia.
- Author
-
Amine ZE, Mauger JF, and Imbeault P
- Subjects
- Adipocytes metabolism, Adipokines metabolism, Adiponectin metabolism, Aryl Hydrocarbon Receptor Nuclear Translocator, Cytochrome P-450 CYP1A1 genetics, Cytochrome P-450 CYP1A1 metabolism, Humans, Hypoxia, Interleukin-6 metabolism, Interleukin-8 metabolism, Leptin metabolism, Vascular Endothelial Growth Factor A metabolism, Environmental Pollutants toxicity, Polychlorinated Biphenyls toxicity
- Abstract
It is increasingly recognized that hypoxia may develop in adipose tissue as its mass expands. Adipose tissue is also the main reservoir of lipophilic pollutants, including polychlorinated biphenyls (PCBs). Both hypoxia and PCBs have been shown to alter adipose tissue functions. The signaling pathways induced by hypoxia and pollutants may crosstalk, as they share a common transcription factor: aryl hydrocarbon receptor nuclear translocator (ARNT). Whether hypoxia and PCBs crosstalk and affect adipokine secretion in human adipocytes remains to be explored. Using primary human adipocytes acutely co-exposed to different levels of hypoxia (24 h) and PCB126 (48 h), we observed that hypoxia significantly inhibits the PCB126 induction of cytochrome P450 (CYP1A1) transcription in a dose-response manner, and that Acriflavine (ACF)-an HIF1α inhibitor-partially restores the PCB126 induction of CYP1A1 under hypoxia. On the other hand, exposure to PCB126 did not affect the transcription of the vascular endothelial growth factor-A (VEGFA) under hypoxia. Exposure to hypoxia increased leptin and interleukin-6 (IL-6), and decreased adiponectin levels dose-dependently, while PCB126 increased IL-6 and IL-8 secretion in a dose-dependent manner. Co-exposure to PCB126 and hypoxia did not alter the adipokine secretion pattern observed under hypoxia and PCB126 exposure alone. In conclusion, our results indicate that (1) hypoxia inhibits PCB126-induced CYP1A1 expression at least partly through ARNT-dependent means, suggesting that hypoxia could affect PCB metabolism and toxicity in adipose tissue, and (2) hypoxia and PCB126 affect leptin, adiponectin, IL-6 and IL-8 secretion differently, with no apparent crosstalk between the two factors.
- Published
- 2022
- Full Text
- View/download PDF
12. Palmitate induces DNA damage and senescence in human adipocytes in vitro that can be alleviated by oleic acid but not inorganic nitrate.
- Author
-
Ishaq A, Tchkonia T, Kirkland JL, Siervo M, and Saretzki G
- Subjects
- Adipocytes, DNA Damage, Humans, Hypertrophy, Inflammation, Palmitates pharmacology, Nitrates pharmacology, Oleic Acid pharmacology
- Abstract
Hypertrophy in white adipose tissue (WAT) can result in sustained systemic inflammation, hyperlipidaemia, insulin resistance, and onset of senescence in adipocytes. Inflammation and hypertrophy can be induced in vitro using palmitic acid (PA). WAT adipocytes have innately low β-oxidation capacity, while inorganic nitrate can promote a beiging phenotype, with promotion of β-oxidation when cells are exposed to nitrate during differentiation. We hypothesized that treatment of human adipocytes with PA in vitro can induce senescence, which might be attenuated by nitrate treatment through stimulation of β-oxidation to remove accumulated lipids. Differentiated subcutaneous and omental adipocytes were treated with PA and nitrate and senescence markers were analyzed. PA induced DNA damage and increased p16
INK4a levels in both human subcutaneous and omental adipocytes in vitro. However, lipid accumulation and lipid droplet size increased after PA treatment only in subcutaneous adipocytes. Thus, hypertrophy and senescence seem not to be causally associated. Contrary to our expectations, subsequent treatment of PA-induced adipocytes with nitrate did not attenuate PA-induced lipid accumulation or senescence. Instead, we found a significantly beneficial effect of oleic acid (OA) on human subcutaneous adipocytes when applied together with PA, which reduced the DNA damage caused by PA treatment., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
13. The Combined Partial Knockdown of CBS and MPST Genes Induces Inflammation, Impairs Adipocyte Function-Related Gene Expression and Disrupts Protein Persulfidation in Human Adipocytes.
- Author
-
Latorre J, Aroca A, Fernández-Real JM, Romero LC, and Moreno-Navarrete JM
- Abstract
Recent studies in mice and humans demonstrated the relevance of H
2 S synthesising enzymes, such as CTH, CBS, and MPST, in the physiology of adipose tissue and the differentiation of preadipocyte into adipocytes. Here, our objective was to investigate the combined role of CTH, CBS, and MPST in the preservation of adipocyte protein persulfidation and adipogenesis. Combined partial CTH , CBS , and MPST gene knockdown was achieved treating fully human adipocytes with siRNAs against these transcripts (siRNA_MIX). Adipocyte protein persulfidation was analyzed using label-free quantitative mass spectrometry coupled with a dimedone-switch method for protein labeling and purification. Proteomic analysis quantified 216 proteins with statistically different levels of persulfidation in KD cells compared to control adipocytes. In fully differentiated adipocytes, CBS and MPST mRNA and protein levels were abundant, while CTH expression was very low. It is noteworthy that siRNA_MIX administration resulted in a significant decrease in CBS and MPST expression, without impacting on CTH. The combined partial knockdown of the CBS and MPST genes resulted in reduced cellular sulfide levels in parallel to decreased expression of relevant genes for adipocyte biology, including adipogenesis, mitochondrial biogenesis, and lipogenesis, but increased proinflammatory- and senescence-related genes. It should be noted that the combined partial knockdown of CBS and MPST genes also led to a significant disruption in the persulfidation pattern of the adipocyte proteins. Although among the less persulfidated proteins, we identified several relevant proteins for adipocyte adipogenesis and function, among the most persulfidated, key mediators of adipocyte inflammation and dysfunction as well as some proteins that might play a positive role in adipogenesis were found. In conclusion, the current study indicates that the combined partial elimination of CBS and MPST (but not CTH) in adipocytes affects the expression of genes related to the maintenance of adipocyte function and promotes inflammation, possibly by altering the pattern of protein persulfidation in these cells, suggesting that these enzymes were required for the functional maintenance of adipocytes.- Published
- 2022
- Full Text
- View/download PDF
14. Cadmium acute exposure induces metabolic and transcriptomic perturbations in human mature adipocytes.
- Author
-
Gasser M, Lenglet S, Bararpour N, Sajic T, Wiskott K, Augsburger M, Fracasso T, Gilardi F, and Thomas A
- Subjects
- Adipocytes metabolism, Adipose Tissue metabolism, Cadmium toxicity, Humans, Insulin metabolism, Obesity chemically induced, Obesity genetics, Transcriptome, Zinc metabolism, Diabetes Mellitus, Type 2 genetics, Metabolic Diseases
- Abstract
Obesity is considered as a major public health concern with strong economic and social burdens. Exposure to pollutants such as heavy metals can contribute to the development of obesity and its associated metabolic disorders, including type 2 diabetes and cardiovascular diseases. Adipose tissue is an endocrine and paracrine organ that plays a key role in the development of these diseases and is one of the main target of heavy metal accumulation. In this study, we determined by inductively coupled plasma mass spectrometry cadmium concentrations in human subcutaneous and visceral adipose tissues, ranging between 2.5 nM and 2.5 µM. We found a positive correlation between cadmium levels and age, sex and smoking status and a negative correlation between cadmium and body mass index. Based on cadmium adipose tissue concentrations found in humans, we investigated the effects of cadmium exposure, at concentrations between 1 nM and 10 µM, on adipose-derived human mesenchymal stem cells differentiated into mature adipocytes in vitro. Transcriptomic analysis highlighted that such exposure altered the expression of genes involved in trace element homeostasis and heavy metal detoxification, such as Solute Carrier Family transporters and metallothioneins. This effect correlated with zinc level alteration in cells and cellular media. Interestingly, dysregulation of zinc homeostasis and transporters has been particularly associated with the development of obesity and type 2 diabetes. Moreover, we found that cadmium exposure induces the pro-inflammatory state of the adipocytes by enhancing the expression of genes such as IL-6, IL-1B and CCL2, cytokines also induced in obesity. Finally, cadmium modulates various adipocyte functions such as the insulin response signaling pathway and lipid homeostasis. Collectively, our data identified some of the cellular mechanisms by which cadmium alters adipocyte functions, thus highlighting new facets of its potential contribution to the progression of metabolic disorders., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
15. Aquaglyceroporins and orthodox aquaporins in human adipocytes.
- Author
-
Huang P, Hansen JS, Saba KH, Bergman A, Negoita F, Gourdon P, Hagström-Andersson A, and Lindkvist-Petersson K
- Subjects
- Adipocytes metabolism, Aquaglyceroporins genetics, Aquaglyceroporins metabolism, Aquaporin 3 metabolism, Aquaporins metabolism, Gene Expression Regulation genetics, Glycerol metabolism, Homeostasis genetics, Humans, Hyperglycemia metabolism, Hyperglycemia pathology, Transcriptome genetics, Water metabolism, Aquaporin 1 genetics, Aquaporin 3 genetics, Aquaporins genetics, Hyperglycemia genetics, Perilipin-1 genetics
- Abstract
Aquaporins play a crucial role in water homeostasis in the human body, and recently the physiological importance of aquaporins as glycerol channels have been demonstrated. The aquaglyceroporins (AQP3, AQP7, AQP9 and AQP10) represent key glycerol channels, enabling glycerol flux across the membranes of cells. Adipocytes are the major source of glycerol and during lipolysis, glycerol is released to be metabolized by other tissues through a well-orchestrated process. Here we show that both AQP3 and AQP7 bind to the lipid droplet protein perilipin 1 (PLIN1), suggesting that PLIN1 is involved in the coordination of the subcellular translocation of aquaglyceroporins in human adipocytes. Moreover, in addition to aquaglyceroporins, we discovered by transcriptome sequencing that AQP1 is expressed in human primary adipocytes. AQP1 is mainly a water channel and thus is thought to be involved in the response to hyper-osmotic stress by efflux of water during hyperglycemia. Thus, this data suggests a contribution of both orthodox aquaporin and aquaglyceroporin in human adipocytes to maintain the homeostasis of glycerol and water during fasting and feeding., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
16. High doses of catecholamines activate glucose transport in human adipocytes independently from adrenoceptor stimulation or vanadium addition.
- Author
-
Carpéné C, Boulet N, Grolleau JL, and Morin N
- Abstract
Background: When combined with vanadium salts, catecholamines strongly activate glucose uptake in rat and mouse adipocytes., Aim: To test whether catecholamines activate glucose transport in human adipocytes., Methods: The uptake of 2-deoxyglucose (2-DG) was measured in adipocytes isolated from pieces of abdominal subcutaneous tissue removed from women undergoing reconstructive surgery. Pharmacological approaches with amine oxidase inhibitors, adrenoreceptor agonists and antioxidants were performed to unravel the mechanisms of action of noradrenaline or adrenaline (also named epinephrine)., Results: In human adipocytes, 45-min incubation with 100 µmol/L adrenaline or noradrenaline activated 2-DG uptake up to more than one-third of the maximal response to insulin. This stimulation was not reproduced with millimolar doses of dopamine or serotonin and was not enhanced by addition of vanadate to the incubation medium. Among various natural amines and adrenergic agonists tested, no other molecule was more efficient than adrenaline and noradrenaline in stimulating 2-DG uptake. The effect of the catecholamines was not impaired by pargyline and semicarbazide, contrarily to that of benzylamine or methylamine, which are recognized substrates of semicarbazide-sensitive amine oxidase. Hydrogen peroxide at 1 mmol/L activated hexose uptake but not pyrocatechol or benzoquinone, and only the former was potentiated by vanadate. Catalase and the phosphoinositide 3-kinase inhibitor wortmannin inhibited adrenaline-induced activation of 2-DG uptake., Conclusion: High doses of catecholamines exert insulin-like actions on glucose transport in human adipocytes. At submillimolar doses, vanadium did not enhance this catecholamine activation of glucose transport. Consequently, this dismantles our previous suggestion to combine the metal ion with catecholamines to improve the benefit/risk ratio of vanadium-based antidiabetic approaches., Competing Interests: Conflict-of-interest statement: The authors declare no competing financial interests., (©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
17. Sustained Activation of TNFα-Induced DNA Damage Response in Newly Differentiated Adipocytes.
- Author
-
Valverde M and Sánchez-Brito A
- Subjects
- Adipocytes cytology, Adipocytes drug effects, Adipose Tissue cytology, Adipose Tissue metabolism, Cell Cycle drug effects, Cell Survival drug effects, Cells, Cultured, Comet Assay methods, Humans, Membrane Potential, Mitochondrial drug effects, Reactive Oxygen Species metabolism, Tumor Necrosis Factor-alpha pharmacology, Adipocytes metabolism, Cell Differentiation, DNA Damage, Tumor Necrosis Factor-alpha metabolism
- Abstract
The response to DNA damage is the mechanism that allows the interaction between stress signals, inflammatory secretions, DNA repair, and maintenance of cell and tissue homeostasis. Adipocyte dysfunction is the cellular trigger for various disease states such as insulin resistance, diabetes, and obesity, among many others. Previously, our group demonstrated that adipogenesis per se, from mesenchymal/stromal stem cells derived from human adipose tissue (hASCs), involves an accumulation of DNA damage and a gradual loss of the repair capacity of oxidative DNA damage. Therefore, our objective was to identify whether healthy adipocytes differentiated for the first time from hASCs, when receiving inflammatory signals induced with TNFα, were able to persistently activate the DNA Damage Response and thus trigger adipocyte dysfunction. We found that TNFα at similar levels circulating in obese humans induce a sustained response to DNA damage response as part of the Senescence-Associated Secretory Phenotype. This mechanism shows the impact of inflammatory environment early affect adipocyte function, independently of aging.
- Published
- 2021
- Full Text
- View/download PDF
18. Epigallocatechin gallate decreases plasma triglyceride, blood pressure, and serum kisspeptin in obese human subjects.
- Author
-
Chatree S, Sitticharoon C, Maikaew P, Pongwattanapakin K, Keadkraichaiwat I, Churintaraphan M, Sripong C, Sririwichitchai R, and Tapechum S
- Subjects
- Adipocytes, Brown drug effects, Adipocytes, Brown metabolism, Adipocytes, White drug effects, Adipocytes, White metabolism, Adiponectin blood, Adult, Blood Glucose metabolism, Catechin pharmacology, Humans, Kidney drug effects, Kidney physiopathology, Leptin blood, Lipolysis, Liver drug effects, Liver physiopathology, Middle Aged, Obesity genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Uncoupling Protein 1 genetics, Uncoupling Protein 1 metabolism, Blood Pressure drug effects, Catechin analogs & derivatives, Kisspeptins blood, Obesity blood, Obesity physiopathology, Triglycerides blood
- Abstract
Obesity is one of major risk factors increasing chronic diseases including type II diabetes, cardiovascular diseases, and hypertension. The effects of epigallocatechin gallate (EGCG), the major active compound in green tea, on reduced obesity and improved metabolic profiles are still controversial. Furthermore, the effects of EGCG on human adipocyte lipolysis and browning of white adipocytes have not been elucidated. This study aimed to investigate the effects of EGCG on obesity, lipolysis, and browning of human white adipocytes. The results showed that, when compared to the baseline values, EGCG significantly decreased fasting plasma triglyceride levels ( P < 0.05), systolic blood pressure ( P < 0.05), diastolic blood pressure ( P < 0.05), and serum kisspeptin levels ( P < 0.05) after 8 weeks of supplement. On the other hand, supplement of EGCG in obese human subjects for 4 or 8 weeks did not decrease body weight, body mass index, waist and hip circumferences, nor total body fat mass or percentage when compared to their baseline values. The study in human adipocytes showed that EGCG did not increase the glycerol release when compared to vehicle, suggesting that it had no lipolytic effect. Furthermore, treatment of EGCG did not enhance uncoupling protein 1 ( UCP1 ) mRNA expression in human white adipocytes when compared with treatment of pioglitazone, the peroxisome proliferator-activated receptor γ (PPAR-γ) agonist, suggesting that EGCG did not augment the browning effect of PPAR-γ on white adipocytes. This study revealed that EGCG reduced 2 metabolic risk factors which are triglyceride and blood pressure in the human experiment. We also showed a novel evidence that EGCG decreased kisspeptin levels. However, EGCG had no effects on obesity reduction in humans, lipolysis, nor browning of human white adipocytes.
- Published
- 2021
- Full Text
- View/download PDF
19. Studying Brown Adipose Tissue in a Human in vitro Context.
- Author
-
Samuelson I and Vidal-Puig A
- Subjects
- Humans, In Vitro Techniques, Adipocytes, Brown physiology, Adipose Tissue, Brown physiology, Energy Metabolism physiology, Thermogenesis physiology
- Abstract
New treatments for obesity and associated metabolic disease are increasingly warranted with the growth of the obesity pandemic. Brown adipose tissue (BAT) may represent a promising therapeutic target to treat obesity, as this tissue has been shown to regulate energy expenditure through non-shivering thermogenesis. Three different strategies could be employed for therapeutic targeting of human thermogenic adipocytes: increasing BAT mass through stimulation of BAT progenitors, increasing BAT function through regulatory pathways, and increasing WAT browning through promotion of beige adipocyte formation. However, these strategies require deeper understanding of human brown and beige adipocytes. While murine studies have greatly increased our understanding of BAT, it is becoming clear that human BAT does not exactly resemble that of the mouse, highlighting the need for human in vitro models of brown adipocytes. Several different human brown adipocyte models will be discussed here, along with the potential to improve brown adipocyte culture through recreation of the BAT microenvironment., (Copyright © 2020 Samuelson and Vidal-Puig.)
- Published
- 2020
- Full Text
- View/download PDF
20. Tunicamycin-Induced Endoplasmic Reticulum Stress Mediates Mitochondrial Dysfunction in Human Adipocytes.
- Author
-
Jackisch L, Murphy AM, Kumar S, Randeva H, Tripathi G, and McTernan PG
- Subjects
- Adipocytes metabolism, Adipocytes pathology, Adipose Tissue drug effects, Adipose Tissue metabolism, Adipose Tissue pathology, Adult, Cell Differentiation drug effects, Cell Differentiation genetics, Cells, Cultured, Cohort Studies, Endoplasmic Reticulum Stress physiology, Female, Humans, Mitochondria physiology, Obesity genetics, Obesity metabolism, Obesity pathology, Young Adult, Adipocytes drug effects, Endoplasmic Reticulum Stress drug effects, Mitochondria drug effects, Tunicamycin pharmacology
- Abstract
Context: Dysfunctional endoplasmic reticulum (ER) and mitochondria are known to contribute to the pathology of metabolic disease. This damage may occur, in part, as a consequence of ER-mitochondria cross-talk in conditions of nutrient excess such as obesity. To date, insight into this dynamic relationship has not been characterized in adipose tissue. Therefore, this study investigated whether ER stress contributes to the development of mitochondrial inefficiency in human adipocytes from lean and obese participants., Methods: Human differentiated adipocytes from Chub-S7 cell line and primary abdominal subcutaneous adipocytes from lean and obese participants were treated with tunicamycin to induce ER stress. Key parameters of mitochondrial function were assessed, including mitochondrial respiration, membrane potential (MMP), and dynamics., Results: ER stress led to increased respiratory capacity in a model adipocyte system (Chub-S7 adipocytes) in a concentration and time dependent manner (24 h: 23%↑; 48 h: 68%↑, P < 0.001; 72 h: 136%↑, P < 0.001). This corresponded with mitochondrial inefficiency and diminished MMP, highlighting the formation of dysfunctional mitochondria. Morphological analysis revealed reorganization of mitochondrial network, specifically mitochondrial fragmentation. Furthermore, p-DRP1, a key protein in fission, significantly increased (P < 0.001). Additionally, adipocytes from obese subjects displayed lower basal respiration (49%↓, P < 0.01) and were unresponsive to tunicamycin in contrast to their lean counterparts, demonstrating inefficient mitochondrial oxidative capacity., Conclusion: These human data suggest that adipocyte mitochondrial inefficiency is driven by ER stress and exacerbated in obesity. Nutrient excess-induced ER stress leads to mitochondrial dysfunction that may therefore shift lipid deposition ectopically and thus have further implications on the development of related metabolic disorders., (© Endocrine Society 2020. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
21. Insulin and β-adrenergic receptors mediate lipolytic and anti-lipolytic signalling that is not altered by type 2 diabetes in human adipocytes.
- Author
-
Jönsson C, Castor Batista AP, Kjølhede P, and Strålfors P
- Subjects
- Adipocytes cytology, Adipose Tissue cytology, Adolescent, Adult, Aged, Aged, 80 and over, Cells, Cultured, Female, Humans, Middle Aged, Obesity metabolism, Young Adult, Adipocytes metabolism, Adipose Tissue metabolism, Diabetes Mellitus, Type 2 metabolism, Insulin pharmacology, Lipolysis drug effects, Lipolysis physiology, Receptors, Adrenergic, beta metabolism
- Abstract
Control of fatty acid storage and release in adipose tissue is fundamental in energy homeostasis and the development of obesity and type 2 diabetes. We here take the whole signalling network into account to identify how insulin and β-adrenergic stimulation in concert controls lipolysis in mature subcutaneous adipocytes obtained from non-diabetic and, in parallel, type 2 diabetic women. We report that, and show how, the anti-lipolytic effect of insulin can be fully explained by protein kinase B (PKB/Akt)-dependent activation of the phosphodiesterase PDE3B. Through the same PKB-dependent pathway β-adrenergic receptor signalling, via cAMP and PI3Kα, is anti-lipolytic and inhibits its own stimulation of lipolysis by 50%. Through this pathway both insulin and β-adrenergic signalling control phosphorylation of FOXO1. The dose-response of lipolysis is bell-shaped, such that insulin is anti-lipolytic at low concentrations, but at higher concentrations of insulin lipolysis was increasingly restored due to inhibition of PDE3B. The control of lipolysis was not altered in adipocytes from diabetic individuals. However, the release of fatty acids was increased by 50% in diabetes due to reduced reesterification of lipolytically liberated fatty acids. In conclusion, our results reveal mechanisms of control by insulin and β-adrenergic stimulation - in human adipocytes - that define a network of checks and balances ensuring robust control to secure uninterrupted supply of fatty acids without reaching concentrations that put cellular integrity at risk. Moreover, our results define how selective insulin resistance leave lipolytic control by insulin unaltered in diabetes, while the fatty acid release is substantially increased., (© 2019 The Author(s).)
- Published
- 2019
- Full Text
- View/download PDF
22. Elevated UCP1 levels are sufficient to improve glucose uptake in human white adipocytes.
- Author
-
Tews D, Pula T, Funcke JB, Jastroch M, Keuper M, Debatin KM, Wabitsch M, and Fischer-Posovszky P
- Subjects
- Adipocytes, Brown metabolism, Animals, Biological Transport, Gene Expression, Glycolysis, Humans, Mice, Mitochondria, Thermogenesis, Uncoupling Protein 1 genetics, Adipocytes, White metabolism, Glucose metabolism, Uncoupling Protein 1 metabolism
- Abstract
Brown adipose tissue (BAT) has been considered beneficial for metabolic health by participating in the regulation of glucose homoeostasis. The browning factors that improve glucose uptake beyond normal levels are still unknown but glucose uptake is not affected in UCP1 knockout mice. Here, we demonstrate in human white adipocytes that basal/resting glucose uptake is improved by solely elevating UCP1 protein levels. Generating human white Simpson-Golabi-Behmel syndrome (SGBS) adipocytes with a stable knockout and overexpression of UCP1, we discovered that UCP1 overexpressing adipocytes significantly improve glucose uptake by 40%. Mechanistically, this is caused by higher glycolytic flux, seen as increased oxygen consumption, extracellular acidification and lactate secretion rates. The improvements in glucose handling are comparable to white-to-brown transitions, as judged by, for the first time, directly comparing in vitro differentiated mouse brown vs white adipocytes. Although no adipogenic, metabolic and mitochondrial gene expressions were significantly altered in SGBS cells, pharmacological inhibition of GLUT1 completely abrogated differences between UCP1+ and control cells, thereby uncovering GLUT1-mediated uptake as permissive gatekeeper. Collectively, our data demonstrate that elevating UCP1 levels is sufficient to improve human white adipocytes as a glucose sink without adverse cellular effects, thus not requiring the adrenergic controlled, complex network of browning which usually hampers translational efforts., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
23. Basolateral presence of the proinflammatory cytokine tumor necrosis factor -α and secretions from adipocytes and macrophages reduce intestinal sugar transport.
- Author
-
Castilla-Madrigal R, Gil-Iturbe E, Sáinz N, Moreno-Aliaga MJ, and Lostao MP
- Subjects
- Animals, Caco-2 Cells, Culture Media, Conditioned metabolism, Disease Models, Animal, Down-Regulation, Glutamine metabolism, Humans, Male, Mice, Inbred C57BL, Obesity metabolism, Secretory Pathway, Signal Transduction, THP-1 Cells, Tumor Necrosis Factor-alpha genetics, Adipocytes metabolism, Intestinal Absorption, Intestinal Mucosa metabolism, Macrophages metabolism, Methylglucosides metabolism, Paracrine Communication, Sodium-Glucose Transporter 1 metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
We have previously demonstrated in Caco-2 cells that tumor necrosis factor-α (TNF-α) inhibits sugar uptake, acting from the apical membrane, by decreasing the expression of the Na
+ -glucose cotransporter SGLT1 in the brush border membrane. The goal was to investigate the hypothesis that TNF-α from abdominal adipose tissue (adipocytes and macrophages) would decrease sugar and amino acid transport acting from the basolateral membrane of the enterocytes. TNF-α placed in the basal compartment of Caco-2 cells decreased α-methyl- d-glucose (αMG) and glutamine uptake. The apical medium derived from these Caco-2 cells apically placed in another set of cells, also reduced sugar and glutamine transport. Reverse-transcription polymerase chain reaction analysis demonstrated upregulation of TNF-α, IL-1β, and MCP1 expression in Caco-2 cells exposed to basal TNF-α. Similarly, αMG uptake was inhibited after Caco-2 cells were incubated, in the basal compartment, with medium from visceral human mesenchymal stem cells-derived adipocytes of overweight individuals. The apical medium collected from those Caco-2 cells, and placed in the upper side of other set of cells, also decreased sugar uptake. Basal presence of medium derived from lipopolysaccharide-activated macrophages and nonactivated macrophages decreased αMG uptake as well. Diet-induced obese mice showed an increase in the visceral adipose tissue surrounding the intestine. In this physiological condition, there was a reduction on αMG uptake in jejunal everted rings. Altogether, these results suggest that basolateral TNF-α, which can be produced by adipocytes and macrophages during obesity, would be able to activate TNF-α and other proinflammatory proteins expression in the small intestine and diminish intestinal sugar and amino acids transport., (© 2018 Wiley Periodicals, Inc.)- Published
- 2019
- Full Text
- View/download PDF
24. Insulin induces Thr484 phosphorylation and stabilization of SIK2 in adipocytes.
- Author
-
Säll J, Negoita F, Hansson B, Kopietz F, Linder W, Pettersson AML, Ekelund M, Laurencikiene J, Degerman E, Stenkula KG, and Göransson O
- Subjects
- Adipocytes cytology, Animals, Cells, Cultured, Humans, Phosphorylation, Rats, Rats, Sprague-Dawley, Adipocytes metabolism, Adipose Tissue metabolism, Insulin metabolism, Insulin Resistance physiology, Protein Serine-Threonine Kinases metabolism
- Abstract
Aims/hypothesis: Salt-inducible kinase 2 (SIK2) is downregulated in adipose tissue from obese or insulin-resistant individuals and inhibition of SIK isoforms results in reduced glucose uptake and insulin signalling in adipocytes. However, the regulation of SIK2 itself in response to insulin in adipocytes has not been studied in detail. The aim of our work was to investigate effects of insulin on various aspects of SIK2 function in adipocytes., Methods: Primary adipocytes were isolated from human subcutaneous and rat epididymal adipose tissue. Insulin-induced phosphorylation of SIK2 and HDAC4 was analyzed using phosphospecific antibodies and changes in the catalytic activity of SIK2 with in vitro kinase assay. SIK2 protein levels were analyzed in primary adipocytes treated with the proteasome inhibitor MG132., Results: We have identified a novel regulatory pathway of SIK2 in adipocytes, which involves insulin-induced phosphorylation at Thr484. This phosphorylation is impaired in individuals with a reduced insulin action. Insulin stimulation does not affect SIK2 catalytic activity or cellular activity towards HDAC4, but is associated with increased SIK2 protein levels in adipocytes., Conclusion/interpretation: Our data suggest that downregulation of SIK2 in the adipose tissue of insulin-resistant individuals can partially be caused by impaired insulin signalling, which might result in defects in SIK2 expression and function., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
25. AMPK activation by A-769662 and 991 does not affect catecholamine-induced lipolysis in human adipocytes.
- Author
-
Kopietz F, Berggreen C, Larsson S, Säll J, Ekelund M, Sakamoto K, Degerman E, Holm C, and Göransson O
- Subjects
- Adipocytes metabolism, Adipose Tissue metabolism, Aminoimidazole Carboxamide analogs & derivatives, Aminoimidazole Carboxamide pharmacology, Animals, Biphenyl Compounds, Female, Humans, Lipolysis physiology, Male, Mice, Phosphorylation drug effects, Rats, Rats, Sprague-Dawley, Ribonucleotides pharmacology, Sterol Esterase metabolism, Adenylate Kinase metabolism, Adipocytes drug effects, Adipose Tissue drug effects, Catecholamines pharmacology, Lipolysis drug effects, Pyrones pharmacology, Thiophenes pharmacology
- Abstract
Activation of AMP-activated protein kinase (AMPK) is considered an attractive strategy for the treatment of type 2 diabetes. Favorable metabolic effects of AMPK activation are mainly observed in skeletal muscle and liver tissue, whereas the effects in human adipose tissue are only poorly understood. Previous studies, which largely employed the AMPK activator 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR), suggest an antilipolytic role of AMPK in adipocytes. The aim of this work was to reinvestigate the role of AMPK in the regulation of lipolysis, using the novel allosteric small-molecule AMPK activators A-769662 and 991, with a focus on human adipocytes. For this purpose, human primary subcutaneous adipocytes were treated with A-769662, 991, or AICAR, as a control, before being stimulated with isoproterenol. AMPK activity status, glycerol release, and the phosphorylation of hormone-sensitive lipase (HSL), a key regulator of lipolysis, were then monitored. Our results show that both A-769662 and 991 activated AMPK to a level that was similar to, or greater than, that induced by AICAR. In contrast to AICAR, which as expected was antilipolytic, neither A-769662 nor 991 affected lipolysis in human adipocytes, although 991 treatment led to altered HSL phosphorylation. Furthermore, we suggest that HSL Ser660 is an important regulator of lipolytic activity in human adipocytes. These data suggest that the antilipolytic effect observed with AICAR in previous studies is, at least to some extent, AMPK independent.
- Published
- 2018
- Full Text
- View/download PDF
26. Spexin: A novel regulator of adipogenesis and fat tissue metabolism.
- Author
-
Kolodziejski PA, Pruszynska-Oszmalek E, Micker M, Skrzypski M, Wojciechowicz T, Szwarckopf P, Skieresz-Szewczyk K, Nowak KW, and Strowski MZ
- Subjects
- 3T3-L1 Cells, Adipocytes cytology, Adipocytes drug effects, Adipocytes metabolism, Animals, Cell Differentiation drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Down-Regulation, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Glucose metabolism, Humans, Insulin pharmacology, Lipolysis, Mice, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Galanin genetics, Receptors, Galanin metabolism, Adipogenesis drug effects, Lipid Metabolism drug effects, Peptide Hormones metabolism
- Abstract
Spexin (SPX, NPQ) is a novel peptide involved in the regulation of energy metabolism. SPX inhibits food intake and reduces body weight. In obese humans, SPX is the most down-regulated gene in fat. Therefore, SPX might be involved in the regulation of lipid metabolism. Here, we study the effects of SPX on lipolysis, lipogenesis, glucose uptake, adipogenesis, cell proliferation and survival in isolated human adipocytes or murine 3T3-L1 cells. SPX and its receptors, GALR2 and GALR3, are present at mRNA and protein levels in murine 3T3-L1 cells and human adipocytes. SPX inhibits adipogenesis and down-regulates mRNA expression of proadipogenic genes such as Pparγ, C/ebpα, C/ebpβ and Fabp4. SPX stimulates lipolysis by increasing the phosphorylation of hormone sensitive lipase (HSL). Simultaneously, SPX inhibits lipogenesis and glucose uptake in human adipocytes and murine 3T3-L1 cells. SPX has no effect on murine 3T3-L1 cell proliferation and viability. Moreover, our research showed that the SPX effect on adipocytes metabolism is mediated via GALR2 and GALR3 receptors. SPX is a novel regulator of lipid metabolism in murine 3T3-L1 and human adipocytes., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. Inhibition of FOXO1 transcription factor in primary human adipocytes mimics the insulin-resistant state of type 2 diabetes.
- Author
-
Rajan MR, Nyman E, Brännmark C, Olofsson CS, and Strålfors P
- Subjects
- Adipocytes metabolism, Adult, Aged, Antigens, CD metabolism, Cells, Cultured, Female, Humans, Middle Aged, Phosphorylation, Receptor, Insulin metabolism, Signal Transduction, TOR Serine-Threonine Kinases metabolism, Adipocytes pathology, Diabetes Mellitus, Type 2 physiopathology, Forkhead Box Protein O1 antagonists & inhibitors, Gene Expression Regulation, Insulin metabolism, Insulin Resistance
- Abstract
Type 2 diabetes is characterized by insulin resistance in the expanding adipose tissue of obesity. The insulin resistance manifests in human adipocytes as system-wide impairment of insulin signalling. An exception is the regulation of transcription factor FOXO1 (forkhead box protein O1), which is phosphorylated downstream of mTORC2 (mammalian/mechanistic target of rapamycin in complex with raptor) and is therefore not exhibiting impaired response to insulin. However, the abundance, and activity, of FOXO1 is reduced by half in adipocytes from patients with diabetes. To elucidate the effect of reduced FOXO1 activity, we here transduced human adipocytes with a dominant-negative construct of FOXO1 (DN-FOXO1). Inhibition of FOXO1 reduced the abundance of insulin receptor, glucose transporter-4, ribosomal protein S6, mTOR and raptor. Functionally, inhibition of FOXO1 induced an insulin-resistant state network-wide, a state that qualitatively and quantitatively mimicked adipocytes from patients with type 2 diabetes. In contrast, and in accordance with these effects of DN-FOXO1, overexpression of wild-type FOXO1 appeared to augment insulin signalling. We combined experimental data with mathematical modelling to show that the impaired insulin signalling in FOXO1-inhibited cells to a large extent can be explained by reduced mTORC1 activity - a mechanism that defines much of the diabetic state in human adipocytes. Our findings demonstrate that FOXO1 is critical for maintaining normal insulin signalling of human adipocytes., (© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2018
- Full Text
- View/download PDF
28. Short-term effects of obestatin on hexose uptake and triacylglycerol breakdown in human subcutaneous adipocytes.
- Author
-
Carpéné C, Les F, Estève D, and Galitzky J
- Abstract
Aim: To study complete dose-dependent effects of obestatin on lipolytic and glucose transport activities in human adipocyte preparations highly responsive to insulin., Methods: Adipocytes were prepared by liberase digestion from subcutaneous abdominal adipose tissue obtained from overweight subjects undergoing plastic surgery. The index of lipolytic activity was the glycerol released in the incubation medium, while glucose transport was assessed by [
3 H]-2-deoxyglucose uptake assay., Results: When tested from 0.1 nmol/L to 1 μmol/L, obestatin did not stimulate glycerol release; it did not inhibit the lipolytic effect of isoprenaline and did not alter the insulin antilipolytic effect. Obestatin hardly activated glucose transport at 1 μmol/L only. Moreover, the obestatin stimulation effect was clearly lower than the threefold increase induced by insulin 100 nmol/L., Conclusion: Low doses of obestatin cannot directly influence lipolysis and glucose uptake in human fat cells., Competing Interests: Conflict-of-interest statement: All authors declare that they do not have any potential conflict of interest in relation to this article.- Published
- 2018
- Full Text
- View/download PDF
29. Activation of IRF1 in Human Adipocytes Leads to Phenotypes Associated with Metabolic Disease.
- Author
-
Friesen M, Camahort R, Lee YK, Xia F, Gerszten RE, Rhee EP, Deo RC, and Cowan CA
- Subjects
- Adipocytes cytology, Animals, Cells, Cultured, Female, Humans, Inflammation genetics, Inflammation metabolism, Interferon Regulatory Factor-1 metabolism, Mesenchymal Stem Cell Transplantation adverse effects, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Mice, Mice, Nude, Obesity genetics, Transcriptome, Up-Regulation, Adipocytes metabolism, Interferon Regulatory Factor-1 genetics, Obesity metabolism, Phenotype
- Abstract
The striking rise of obesity-related metabolic disorders has focused attention on adipocytes as critical mediators of disease phenotypes. To better understand the role played by excess adipose in metabolic dysfunction it is crucial to decipher the transcriptional underpinnings of the low-grade adipose inflammation characteristic of diseases such as type 2 diabetes. Through employing a comparative transcriptomics approach, we identified IRF1 as differentially regulated between primary and in vitro-derived genetically matched adipocytes. This suggests a role as a mediator of adipocyte inflammatory phenotypes, similar to its function in other tissues. Utilizing adipose-derived mesenchymal progenitors we subsequently demonstrated that expression of IRF1 in adipocytes indeed contributes to upregulation of inflammatory processes, both in vitro and in vivo. This highlights IRF1's relevance to obesity-related inflammation and the resultant metabolic dysregulation., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. Eicosapentaenoic acid and arachidonic acid differentially regulate adipogenesis, acquisition of a brite phenotype and mitochondrial function in primary human adipocytes.
- Author
-
Fleckenstein-Elsen M, Dinnies D, Jelenik T, Roden M, Romacho T, and Eckel J
- Subjects
- Adipocytes metabolism, Adipogenesis drug effects, Carnitine O-Palmitoyltransferase genetics, Cell Differentiation drug effects, Cells, Cultured, Citrate (si)-Synthase metabolism, Docosahexaenoic Acids pharmacology, Female, Humans, Mitochondria metabolism, Uncoupling Protein 1 genetics, Adipocytes drug effects, Arachidonic Acid pharmacology, Eicosapentaenoic Acid pharmacology, Mitochondria drug effects
- Abstract
Scope: n-3 and n-6 PUFAs have several opposing biological effects and influence white adipose tissue (WAT) function. The recent discovery of thermogenic UCP1-expressing brite adipocytes within WAT raised the question whether n-3 and n-6 PUFAs exert differential effects on brite adipocyte formation and mitochondrial function., Methods and Results: Primary human preadipocytes were treated with n-3 PUFAs (eicosapentaenoic acid, EPA; docosahexaenoic acid, DHA) or n-6 PUFA (arachidonic acid, ARA) during differentiation, and adipogenesis, white and brite gene expression markers, mitochondrial content and function were analyzed at day 12 of differentiation. Adipogenesis was equally increased by n-3 and n-6 PUFAs. The n-6 PUFA ARA increased lipid droplet size and expression of the white-specific marker TCF21 while decreased mitochondrial protein expression and respiratory function. In contrast, EPA increased expression of the brown adipocyte-related genes UCP1 and CPT1B, and improved mitochondrial function of adipocytes. The opposing effects of EPA and ARA on gene expression and mitochondrial function were also observed in cells treated from day 8 to 12 of adipocyte differentiation., Conclusion: EPA promotes brite adipogenesis and improves parameters of mitochondrial function, such as increased expression of CPTB1, citrate synthase activity and higher maximal respiratory capacity, while ARA reduced mitochondrial spare respiratory capacity in vitro., (© 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2016
- Full Text
- View/download PDF
31. Olanzapine promotes the accumulation of lipid droplets and the expression of multiple perilipins in human adipocytes.
- Author
-
Nimura S, Yamaguchi T, Ueda K, Kadokura K, Aiuchi T, Kato R, Obama T, and Itabe H
- Subjects
- Adipocytes cytology, Adipocytes metabolism, Cell Differentiation, Cells, Cultured, Dose-Response Relationship, Drug, Humans, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells drug effects, Olanzapine, Perilipin-1, RNA, Messenger genetics, Transcription Factors genetics, Adipocytes drug effects, Antipsychotic Agents pharmacology, Benzodiazepines pharmacology, Carrier Proteins metabolism, Lipid Droplets metabolism, Phosphoproteins metabolism
- Abstract
Second generation antipsychotics are useful for the treatment of schizophrenia, but concerns have been raised about the side effects of diabetes mellitus and obesity. Olanzapine, especially, is associated with more weight gain than the others. It has been reported that olanzapine promotes adipocyte-differentiation in rodents both in vivo and in vitro. In this study the effects of antipsychotics on human adipocytes were investigated by using human mesenchymal stem cells (hMSCs). When hMSCs were differentiated and treated with various antipsychotics, olanzapine and clozapine increased intracellular lipids. Olanzapine induced lipid accumulation in a dose-dependent manner. Proteomic analysis revealed that PLIN4 and several enzymes for lipid metabolism were increased in the hMSCs after olanzapine treatment. During adipocyte differentiation, olanzapine increased the protein expression of PLIN1, PLIN2 and PLIN4. These proteins are known to be associated with the initial stage of lipid droplet formation. Immunocytochemistry showed that olanzapine increased and enlarged the lipid droplets coated with PLIN1 and PLIN2 while PLIN4 was largely distributed in the cytosol. mRNA expression of PLIN2, but not PLIN1 or PLIN4, was increased by olanzapine. On the other hand, olanzapine did not alter the mRNA level of transcription regulators involved in adipocyte-differentiation or adipokines. The present study shows that olanzapine induced transient PLIN2 expression in hMSCs that could result in an accumulation of lipid droplets and overexpression of PLIN1 and PLIN4, providing information of possible interest for olanzapine-induced weight gain., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
32. An siRNA-based method for efficient silencing of gene expression in mature brown adipocytes.
- Author
-
Isidor MS, Winther S, Basse AL, Petersen MC, Cannon B, Nedergaard J, and Hansen JB
- Abstract
Brown adipose tissue is a promising therapeutic target for opposing obesity, glucose intolerance and insulin resistance. The ability to modulate gene expression in mature brown adipocytes is important to understand brown adipocyte function and delineate novel regulatory mechanisms of non-shivering thermogenesis. The aim of this study was to optimize a lipofection-based small interfering RNA (siRNA) transfection protocol for efficient silencing of gene expression in mature brown adipocytes. We determined that a critical parameter was to deliver the siRNA to mature adipocytes by reverse transfection, i.e. transfection of non-adherent cells. Using this protocol, we effectively knocked down both high- and low-abundance transcripts in a model of mature brown adipocytes (WT-1) as well as in primary mature mouse brown adipocytes. A functional consequence of the knockdown was confirmed by an attenuated increase in uncoupled respiration (thermogenesis) in response to β-adrenergic stimulation of mature WT-1 brown adipocytes transfected with uncoupling protein 1 siRNA. Efficient gene silencing was also obtained in various mouse and human white adipocyte models (3T3-L1, primary mouse white adipocytes, hMADS) with the ability to undergo "browning." In summary, we report an easy and versatile reverse siRNA transfection protocol to achieve specific silencing of gene expression in various models of mature brown and browning-competent white adipocytes, including primary cells.
- Published
- 2015
- Full Text
- View/download PDF
33. Fructose Alters Intermediary Metabolism of Glucose in Human Adipocytes and Diverts Glucose to Serine Oxidation in the One-Carbon Cycle Energy Producing Pathway.
- Author
-
Varma V, Boros LG, Nolen GT, Chang CW, Wabitsch M, Beger RD, and Kaput J
- Abstract
Increased consumption of sugar and fructose as sweeteners has resulted in the utilization of fructose as an alternative metabolic fuel that may compete with glucose and alter its metabolism. To explore this, human Simpson-Golabi-Behmel Syndrome (SGBS) preadipocytes were differentiated to adipocytes in the presence of 0, 1, 2.5, 5 or 10 mM of fructose added to a medium containing 5 mM of glucose representing the normal blood glucose concentration. Targeted tracer [1,2-13C2]-d-glucose fate association approach was employed to examine the influence of fructose on the intermediary metabolism of glucose. Increasing concentrations of fructose robustly increased the oxidation of [1,2-13C2]-d-glucose to 13CO2 (p < 0.000001). However, glucose-derived 13CO2 negatively correlated with 13C labeled glutamate, 13C palmitate, and M+1 labeled lactate. These are strong markers of limited tricarboxylic acid (TCA) cycle, fatty acid synthesis, pentose cycle fluxes, substrate turnover and NAD+/NADP+ or ATP production from glucose via complete oxidation, indicating diminished mitochondrial energy metabolism. Contrarily, a positive correlation was observed between glucose-derived 13CO2 formed and 13C oleate and doses of fructose which indicate the elongation and desaturation of palmitate to oleate for storage. Collectively, these results suggest that fructose preferentially drives glucose through serine oxidation glycine cleavage (SOGC pathway) one-carbon cycle for NAD+/NADP+ production that is utilized in fructose-induced lipogenesis and storage in adipocytes.
- Published
- 2015
- Full Text
- View/download PDF
34. Phosphatidylinositol 3-kinase (PI3K) signalling regulates insulin-like-growth factor binding protein-2 (IGFBP-2) production in human adipocytes.
- Author
-
Wilhelm F, Kässner F, Schmid G, Kratzsch J, Laner A, Wabitsch M, Körner A, Kiess W, and Garten A
- Subjects
- Adipocytes cytology, Blotting, Western, Cell Proliferation, Cells, Cultured, Enzyme-Linked Immunosorbent Assay, Humans, PTEN Phosphohydrolase antagonists & inhibitors, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, Proto-Oncogene Proteins c-akt metabolism, RNA, Messenger genetics, RNA, Small Interfering genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, TOR Serine-Threonine Kinases metabolism, Adipocytes metabolism, Insulin-Like Growth Factor Binding Protein 2 metabolism, Phosphatidylinositol 3-Kinases metabolism, Signal Transduction
- Abstract
Objective: Insulin-like-growth factor binding protein 2 (IGFBP-2) is thought to be a marker for the phosphatase and tensin homolog (PTEN) status and activity of the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway. We aimed to evaluate whether or not lipoma cells of a patient with a heterozygous deletion in the PTEN gene would produce more IGFBP-2 than PTEN non deficient control cells. Moreover, we analysed the influence of pharmacological inhibitors of the PI3K/AKT/mTOR pathway on IGFBP-2 production., Design: PTEN deficient preadipocytes and control PTEN non deficient preadipocytes were differentiated in vitro and treated with the respective inhibitors. PTEN was transiently down regulated by siRNA in human preadipocytes. IGFBP-2 mRNA and protein expression and IGFBP-2 in culture supernatant were measured., Results: PTEN deficient lipoma cells were found to produce IGFBP-2 during in vitro differentiation in comparable amounts to PTEN non deficient cells. In contrast, acute down regulation of PTEN in preadipocytes resulted in enhanced production of IGFBP-2. Incubation with the PI3K inhibitors LY294002 and wortmannin decreased IGFBP-2 mRNA and protein. Neither the mTOR complex 1 inhibitor rapamycin nor PD98059, an inhibitor of MEK (mitogen-activated protein kinase kinase), showed a significant effect on IGFBP-2 production., Conclusion: IGFBP-2 production in PTEN deficient preadipocytes was not influenced by PTEN deficiency or by inhibition of mTORC1 and MAPK. In contrast, inhibition of PI3K decreased IGFBP-2 expression and secretion., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. Mature adipocyte proteome reveals differentially altered protein abundances between lean, overweight and morbidly obese human subjects.
- Author
-
Benabdelkamel H, Masood A, Almidani GM, Alsadhan AA, Bassas AF, Duncan MW, and Alfadda AA
- Subjects
- Body Mass Index, Female, Gene Expression Regulation, Humans, Lipid Metabolism, Obesity, Morbid pathology, Protein Interaction Maps, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Subcutaneous Fat, Abdominal metabolism, Subcutaneous Fat, Abdominal pathology, Adipocytes metabolism, Obesity, Morbid metabolism, Proteome isolation & purification, Proteomics methods, Subcutaneous Fat, Abdominal cytology
- Abstract
Overweight (OW) and obese individuals are considered to be graded parts of the scale having increasing weight as a common feature. They may not, however, be part of the same continuum and may differ metabolically. In this study we applied an untargeted proteomic approach to compare protein abundances in mature adipocytes derived from the subcutaneous adipose tissue of overweight and morbidly obese female subjects to those of lean age matched controls. Mature adipocytes were isolated from liposuction samples of abdominal subcutaneous adipose tissue collected from both lean (L; n = 7, 23.3 ± 0.4 kg/m(2); mean BMI ± SD), overweight (OW; n = 8, 27.9 ± 0.6 kg/m(2); mean BMI ± SD) and morbidly obese (MOB; n = 7, 44.8 ± 3.8 kg/m(2); mean BMI ± SD) individuals. Total protein extracts were then compared by two-dimensional difference in gel electrophoresis (2D DIGE). One hundred and ten differentially expressed protein spots (i.e., fitting the statistical criteria ANOVA test, p < 0.05; fold-change ≥1.5) were detected, and of these, 89 were identified by MALDI-TOF mass spectrometry. Of these, 66 protein spots were common to both groups whereas 23 were unique to the MOB group. Significant differences were evident in the abundances of key proteins involved in glucose and lipid metabolism, energy regulation, cytoskeletal structure and redox control signaling pathways. Differences in the abundance of some chaperones were also evident. The differentially abundant proteins were investigated using Ingenuity Pathway Analysis (IPA) to establish their associations with known biological functions. The network identified in the OW group with the highest score relates to-: cell-to-cell signaling and interaction; in contrast, in the MOB group the major interacting pathways are associated with lipid metabolism, small molecule biochemistry and cancer. The differences in abundance of the differentially regulated proteins were validated by immunoblotting. These findings provide insights into metabolic differences in OW and MOB individuals., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. Metabolic fate of fructose in human adipocytes: a targeted 13 C tracer fate association study.
- Author
-
Varma V, Boros LG, Nolen GT, Chang CW, Wabitsch M, Beger RD, and Kaput J
- Abstract
The development of obesity is becoming an international problem and the role of fructose is unclear. Studies using liver tissue and hepatocytes have contributed to the understanding of fructose metabolism. Excess fructose consumption also affects extra hepatic tissues including adipose tissue. The effects of fructose on human adipocytes are not yet fully characterized, although in vivo studies have noted increased adiposity and weight gain in response to fructose sweetened-beverages. In order to understand and predict the metabolic responses of adipocytes to fructose, this study examined differentiating and differentiated human adipocytes in culture, exposed to a range of fructose concentrations equivalent to that reported in blood after consuming fructose. A stable isotope based dynamic profiling method using [U-
13 C6 ]-d-fructose tracer was used to examine the metabolism and fate of fructose. A targeted stable isotope tracer fate association method was used to analyze metabolic fluxes and flux surrogates with exposure to escalating fructose concentration. This study demonstrated that fructose stimulates anabolic processes in adipocytes robustly, including glutamate and de novo fatty acid synthesis. Furthermore, fructose also augments the release of free palmitate from fully differentiated adipocytes. These results imply that in the presence of fructose, the metabolic response of adipocytes in culture is altered in a dose dependent manner, particularly favoring increased glutamate and fatty acid synthesis and release, warranting further in vivo studies.- Published
- 2015
- Full Text
- View/download PDF
37. Calorie restriction-induced changes in the secretome of human adipocytes, comparison with resveratrol-induced secretome effects.
- Author
-
Renes J, Rosenow A, Roumans N, Noben JP, and Mariman EC
- Subjects
- Adipocytes drug effects, Adipocytes pathology, Adipokines genetics, Adipokines isolation & purification, Adipokines metabolism, Adipose Tissue, White drug effects, Adipose Tissue, White metabolism, Adipose Tissue, White pathology, Arrhythmias, Cardiac metabolism, Arrhythmias, Cardiac pathology, Cells, Cultured, Electrophoresis, Gel, Two-Dimensional, Gene Expression Regulation drug effects, Genetic Diseases, X-Linked metabolism, Genetic Diseases, X-Linked pathology, Gigantism metabolism, Gigantism pathology, Glucose deficiency, Heart Defects, Congenital metabolism, Heart Defects, Congenital pathology, Humans, Insulin Resistance, Intellectual Disability metabolism, Intellectual Disability pathology, Molecular Sequence Annotation, Obesity metabolism, Obesity pathology, Proteome metabolism, Proteomics, Resveratrol, Sirtuin 1 genetics, Sirtuin 1 isolation & purification, Sirtuin 1 metabolism, Tandem Mass Spectrometry, Adipocytes metabolism, Antioxidants pharmacology, Caloric Restriction, Proteome isolation & purification, Stilbenes pharmacology
- Abstract
Obesity is characterized by dysfunctional white adipose tissue (WAT) that ultimately may lead to metabolic diseases. Calorie restriction (CR) reduces the risk for age and obesity-associated complications. The impact of CR on obesity has been examined with human intervention studies, which showed alterations in circulating adipokines. However, a direct effect of CR on the human adipocyte secretome remains elusive. Therefore, the effect of a 96h low glucose CR on the secretion profile of in vitro cultured mature human SGBS adipocytes was investigated by using proteomics technology. Low-glucose CR decreased the adipocyte triglyceride contents and resulted in an altered secretion profile. Changes in the secretome indicated an improved inflammatory phenotype. In addition, several adipocyte-secreted proteins related to insulin resistance showed a reversed expression after low-glucose CR. Furthermore, 6 novel CR-regulated adipocyte-secreted proteins were identified. Since resveratrol (RSV) mimics CR we compared results from this study with data from our previous RSV study on the SGBS adipocyte secretome. The CR and RSV adipocyte secretomes partly differed from each other, although both treatment strategies lead to secretome changes indicating a less inflammatory phenotype. Furthermore, both treatments induced SIRT1 expression and resulted in a reversed expression of detrimental adipokines associated with metabolic complications., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
38. Interleukin-1β mediates macrophage-induced impairment of insulin signaling in human primary adipocytes.
- Author
-
Gao D, Madi M, Ding C, Fok M, Steele T, Ford C, Hunter L, and Bing C
- Subjects
- Adipocytes, White cytology, Adipocytes, White immunology, Adipocytes, White metabolism, Antibodies, Neutralizing pharmacology, Caspase 1 chemistry, Caspase 1 metabolism, Caspase Inhibitors pharmacology, Cell Communication, Cell Line, Cells, Cultured, Culture Media, Conditioned chemistry, Culture Media, Conditioned metabolism, Gene Expression Regulation drug effects, Humans, Interleukin 1 Receptor Antagonist Protein genetics, Interleukin 1 Receptor Antagonist Protein metabolism, Interleukin-1beta antagonists & inhibitors, Lipolysis drug effects, Macrophages cytology, Macrophages drug effects, Macrophages immunology, Receptors, Interleukin-1 agonists, Receptors, Interleukin-1 antagonists & inhibitors, Receptors, Interleukin-1 metabolism, Recombinant Proteins metabolism, Adipocytes, White drug effects, Hypoglycemic Agents pharmacology, Insulin pharmacology, Insulin Resistance, Interleukin-1beta metabolism, Macrophages metabolism, Signal Transduction
- Abstract
Adipose tissue expansion during obesity is associated with increased macrophage infiltration. Macrophage-derived factors significantly alter adipocyte function, inducing inflammatory responses and decreasing insulin sensitivity. Identification of the major factors that mediate detrimental effects of macrophages on adipocytes may offer potential therapeutic targets. IL-1β, a proinflammatory cytokine, is suggested to be involved in the development of insulin resistance. This study investigated the role of IL-1β in macrophage-adipocyte cross-talk, which affects insulin signaling in human adipocytes. Using macrophage-conditioned (MC) medium and human primary adipocytes, we examined the effect of IL-1β antagonism on the insulin signaling pathway. Gene expression profile and protein abundance of insulin signaling molecules were determined, as was the production of proinflammatory cytokine/chemokines. We also examined whether IL-1β mediates MC medium-induced alteration in adipocyte lipid storage. MC medium and IL-1β significantly reduced gene expression and protein abundance of insulin signaling molecules, including insulin receptor substrate-1, phosphoinositide 3-kinase p85α, and glucose transporter 4 and phosphorylation of Akt. In contrast, the expression and release of the proinflammatory markers, including IL-6, IL-8, monocyte chemotactic protein-1, and chemokine (C-C motif) ligand 5 by adipocytes were markedly increased. These changes were significantly reduced by blocking IL-1β activity, its receptor binding, or its production by macrophages. MC medium-inhibited expression of the adipogenic factors and -stimulated lipolysis was also blunted with IL-1β neutralization. We conclude that IL-1β mediates, at least in part, the effect of macrophages on insulin signaling and proinflammatory response in human adipocytes. Blocking IL-1β could be beneficial for preventing obesity-associated insulin resistance and inflammation in human adipose tissue., (Copyright © 2014 the American Physiological Society.)
- Published
- 2014
- Full Text
- View/download PDF
39. Proteomic analysis of mature adipocytes from obese patients in relation to aging.
- Author
-
Alfadda AA, Benabdelkamel H, Masood A, Moustafa A, Sallam R, Bassas A, and Duncan M
- Subjects
- Actins metabolism, Adipocytes pathology, Adult, Aging pathology, Apoptosis, Female, Humans, Middle Aged, Obesity pathology, Prohibitins, Protein Disulfide-Isomerases metabolism, Protein Interaction Maps, Proteomics, Repressor Proteins metabolism, STAT3 Transcription Factor metabolism, Subcutaneous Fat metabolism, Subcutaneous Fat pathology, Young Adult, Adipocytes metabolism, Aging metabolism, Obesity metabolism
- Abstract
Obesity and aging are interrelated conditions that both cause changes in adipocyte metabolism and affect the distribution of fat in both subcutaneous and visceral depots. In addition, both weight gain and aging can lead to similar clinical outcomes such as insulin resistance, cardiovascular disease, type 2 diabetes mellitus, atherosclerosis and stroke. Our objective was to examine the changes in protein expression within the subcutaneous adipose tissue of obese patients, matched for BMI, in relation to age. Mature adipocytes were isolated from liposuction samples of abdominal subcutaneous adipose tissue collected from both young (26.2±4.3 (mean age±SD); n=7) and old (52.2±4.7 (mean age±SD); n=7) obese individuals. Total protein extracts were then compared by two-dimensional difference in gel electrophoresis (2D DIGE). Thirty differentially expressed protein spots (ANOVA test, p≤0.05; fold-change ≥1.8) were detected, of which, 15 were identified by MALDI-TOF mass spectrometry. These were comprised of a total of thirteen unique protein sequences. Nine proteins were more abundant in the adipocytes isolated from old vs. young individuals. These proteins included prohibitin 1, protein disulphide isomerase A3, beta actin, profilin, aldo-ketoreductase 1 C2, alpha crystallin B and the annexins A1, A5 and A6. Four other proteins were less abundant in the adipocytes from old, obese subjects and these included keratin type 2 cytoskeletal 1, keratin type 2 cytoskeletal 10 and hemoglobins A and B. The differentially abundant proteins were investigated by Ingenuity Pathway Analysis (IPA) to reveal their associations with known biological functions. This analysis identified signal transducer and activator of transcription 3 as the central molecule in the connectivity map and the apoptotic pathway as the pathway with the highest score. Differences in the abundances of several proteins were confirmed by immunoblotting: i.e., prohibitin 1, protein disulphide isomerase A3, beta actin, profilin and signal transducer and activator of transcription 3 proteins. In conclusion, proteomic analysis of subcutaneous adipose tissue reveals differences in the abundance of proteins in adipocytes isolated from young vs. old individuals. These differentially abundant proteins are involved in the regulation of apoptosis, cellular senescence and inflammatory response. All these are common pathologic events in both obesity and aging., (© 2013.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.