Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Ann Endocrinol (Paris) ; 85(3): 220-225, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38871505

ABSTRACT

In addition to the major subcutaneous and visceral adipose tissues (AT), other adipose depots are dispersed throughout the body and are found in close interaction with proximal organs such as mammary and periprostatic AT (MAT and PPAT respectively). These ATs have an effect on proximal organ function during physiological processes and diseases such as cancer. We highlighted here some of their most distinctive features in terms of tissular organization and responses to external stimuli and discussed how obesity affects them based on our current knowledge.


Subject(s)
Adipose Tissue , Obesity , Humans , Adipose Tissue/physiology , Female , Obesity/physiopathology , Neoplasms/pathology , Animals , Breast/physiology , Breast/pathology , Breast Neoplasms/pathology , Mammary Glands, Human/physiology , Mammary Glands, Human/pathology , Intra-Abdominal Fat , Subcutaneous Fat/physiology , Subcutaneous Fat/pathology
2.
Sci Rep ; 13(1): 4707, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36949082

ABSTRACT

Obesity is a negative prognosis factor for breast cancer. Yet, the biological mechanisms underlying this effect are still largely unknown. An emerging hypothesis is that the transfer of free fatty acids (FFA) between adipocytes and tumor cells might be altered under obese conditions, contributing to tumor progression. Currently there is a paucity of models to study human mammary adipocytes (M-Ads)-cancer crosstalk. As for other types of isolated white adipocytes, herein, we showed that human M-Ads die within 2-3 days by necrosis when grown in 2D. As an alternative, M-Ads were grown in a fibrin matrix, a 3D model that preserve their distribution, integrity and metabolic function for up to 5 days at physiological glucose concentrations (5 mM). Higher glucose concentrations frequently used in in vitro models promote lipogenesis during M-Ads culture, impairing their lipolytic function. Using transwell inserts, the matrix embedded adipocytes were cocultured with breast cancer cells. FFA transfer between M-Ads and cancer cells was observed, and this event was amplified by obesity. Together these data show that our 3D model is a new tool for studying the effect of M-Ads on tumor cells and beyond with all the components of the tumor microenvironment including the immune cells.


Subject(s)
Adipocytes , Breast Neoplasms , Fatty Acids, Nonesterified , Mammary Glands, Human , Obesity , Thinness , Cell Culture Techniques, Three Dimensional , Adipocytes/metabolism , Adipocytes/pathology , Primary Cell Culture , Mammary Glands, Human/pathology , Breast Neoplasms/pathology , Obesity/metabolism , Obesity/pathology , Thinness/metabolism , Thinness/pathology , Humans , MDA-MB-231 Cells , Fatty Acids, Nonesterified/metabolism , Prognosis
3.
Cancer Metastasis Rev ; 41(3): 589-605, 2022 09.
Article in English | MEDLINE | ID: mdl-35708800

ABSTRACT

Bone marrow adipose tissues (BMATs) and their main cellular component, bone marrow adipocytes (BMAds), are found within the bone marrow (BM), which is a niche for the development of hematological malignancies as well as bone metastasis from solid tumors such as breast and prostate cancers. In humans, BMAds are present within the hematopoietic or "red" BMAT and in the "yellow" BMAT where they are more densely packed. BMAds are emerging as new actors in tumor progression; however, there are many outstanding questions regarding their precise role. In this review, we summarized our current knowledge regarding the development, distribution, and regulation by external stimuli of the BMATs in mice and humans and addressed how obesity could affect these traits. We then discussed the specific metabolic phenotype of BMAds that appear to be different from "classical" white adipocytes, since they are devoid of lipolytic function. According to this characterization, we presented how tumor cells affect the in vitro and in vivo phenotype of BMAds and the signals emanating from BMAds that are susceptible to modulate tumor behavior with a specific emphasis on their metabolic crosstalk with cancer cells. Finally, we discussed how obesity could affect this crosstalk. Deciphering the role of BMAds in tumor progression would certainly lead to the identification of new targets in oncology in the near future.


Subject(s)
Adipocytes , Bone Marrow Cells , Neoplasms , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Humans , Male , Mice , Neoplasms/pathology , Obesity/complications
4.
Front Endocrinol (Lausanne) ; 12: 744527, 2021.
Article in English | MEDLINE | ID: mdl-34646237

ABSTRACT

Over the last two decades, increased interest of scientists to study bone marrow adiposity (BMA) in relation to bone and adipose tissue physiology has expanded the number of publications using different sources of bone marrow adipose tissue (BMAT). However, each source of BMAT has its limitations in the number of downstream analyses for which it can be used. Based on this increased scientific demand, the International Bone Marrow Adiposity Society (BMAS) established a Biobanking Working Group to identify the challenges of biobanking for human BMA-related samples and to develop guidelines to advance establishment of biobanks for BMA research. BMA is a young, growing field with increased interest among many diverse scientific communities. These bring new perspectives and important biological questions on how to improve and build an international community with biobank databases that can be used and shared all over the world. However, to create internationally accessible biobanks, several practical and legislative issues must be addressed to create a general ethical protocol used in all institutes, to allow for exchange of biological material internationally. In this position paper, the BMAS Biobanking Working Group describes similarities and differences of patient information (PIF) and consent forms from different institutes and addresses a possibility to create uniform documents for BMA biobanking purposes. Further, based on discussion among Working Group members, we report an overview of the current isolation protocols for human bone marrow adipocytes (BMAds) and bone marrow stromal cells (BMSCs, formerly mesenchymal), highlighting the specific points crucial for effective isolation. Although we remain far from a unified BMAd isolation protocol and PIF, we have summarized all of these important aspects, which are needed to build a BMA biobank. In conclusion, we believe that harmonizing isolation protocols and PIF globally will help to build international collaborations and improve the quality and interpretation of BMA research outcomes.


Subject(s)
Adipose Tissue , Bone Marrow , Tissue Banks/organization & administration , Adiposity , Biological Specimen Banks , Humans
5.
STAR Protoc ; 2(3): 100629, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34235494

ABSTRACT

Primary human bone marrow adipocytes (BM-Ads) display a specific metabolism that is not recapitulated by in vitro differentiated bone marrow mesenchymal stromal cells. These findings highlight the need for using primary BM-Ads in studies of the metabolic impact of BM-Ads on surrounding cells. Here, we present a protocol for isolating human BM-Ads from bone marrow aspirates and verifying adipocyte suspension purity. These isolated and purified BM-Ads can be used for functional assays or frozen for molecular analyses. For complete details on the use and execution of this protocol, please refer to Attane et al. (2020).


Subject(s)
Adipocytes/cytology , Bone Marrow Cells/cytology , Femur/cytology , Fluorescent Antibody Technique , Humans , Reproducibility of Results
6.
Int J Mol Sci ; 22(4)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671469

ABSTRACT

Bone metastasis remains the most frequent and the deadliest complication of prostate cancer (PCa). Mechanisms leading to the homing of tumor cells to bone remain poorly characterized. Role of chemokines in providing navigational cues to migrating cancer cells bearing specific receptors is well established. Bone is an adipocyte-rich organ since 50 to 70% of the adult bone marrow (BM) volume comprise bone marrow adipocytes (BM-Ads), which are likely to produce chemokines within the bone microenvironment. Using in vitro migration assays, we demonstrated that soluble factors released by human primary BM-Ads are able to support the directed migration of PCa cells in a CCR3-dependent manner. In addition, we showed that CCL7, a chemokine previously involved in the CCR3-dependent migration of PCa cells outside of the prostate gland, is released by human BM-Ads. These effects are amplified by obesity and ageing, two clinical conditions known to promote aggressive and metastatic PCa. In human tumors, we found an enrichment of CCR3 in bone metastasis vs. primary tumors at mRNA levels using Oncomine microarray database. In addition, immunohistochemistry experiments demonstrated overexpression of CCR3 in bone versus visceral metastases. These results underline the potential importance of BM-Ads in the bone metastatic process and imply a CCR3/CCL7 axis whose pharmacological interest needs to be evaluated.


Subject(s)
Adipocytes/metabolism , Adipocytes/pathology , Bone Marrow/pathology , Bone and Bones/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, CCR3/metabolism , Aging/pathology , Bone Marrow/drug effects , Bone and Bones/drug effects , Cell Line, Tumor , Chemokine CCL7/metabolism , Chemotaxis/drug effects , Culture Media, Conditioned/pharmacology , Humans , Male , Neoplasm Metastasis , Obesity/complications , Prostatic Neoplasms/complications
7.
JCI Insight ; 5(24)2020 12 17.
Article in English | MEDLINE | ID: mdl-33201859

ABSTRACT

Enhanced energy expenditure in brown (BAT) and white adipose tissues (WAT) can be therapeutic against metabolic diseases. We examined the thermogenic role of adipose α/ß-hydrolase domain 6 (ABHD6), which hydrolyzes monoacylglycerol (MAG), by employing adipose-specific ABHD6-KO mice. Control and KO mice showed similar phenotypes at room temperature and thermoneutral conditions. However, KO mice were resistant to hypothermia, which can be accounted for by the simultaneously increased lipolysis and lipogenesis of the thermogenic glycerolipid/free fatty acid (GL/FFA) cycle in visceral fat, despite unaltered uncoupling protein 1 expression. Upon cold stress, nuclear 2-MAG levels increased in visceral WAT of the KO mice. Evidence is provided that 2-MAG causes activation of PPARα in white adipocytes, leading to elevated expression and activity of GL/FFA cycle enzymes. In the ABHD6-ablated BAT, glucose and oxidative metabolism were elevated upon cold induction, without changes in GL/FFA cycle and lipid turnover. Moreover, response to in vivo ß3-adrenergic stimulation was comparable between KO and control mice. Our data reveal a MAG/PPARα/GL/FFA cycling metabolic signaling network in visceral adipose tissue, which contributes to cold tolerance, and that adipose ABHD6 is a negative modulator of adaptive thermogenesis.


Subject(s)
Monoacylglycerol Lipases/metabolism , Thermogenesis/genetics , Thermotolerance/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Cold Temperature , Energy Metabolism , Female , Hydrolases/metabolism , Male , Mice , Mice, Inbred C57BL , Monoacylglycerol Lipases/genetics , Monoglycerides/metabolism , Obesity/metabolism , PPAR alpha/metabolism , Uncoupling Protein 1/metabolism
8.
Trends Cancer ; 6(7): 593-604, 2020 07.
Article in English | MEDLINE | ID: mdl-32610069

ABSTRACT

Over the past decade, it has become apparent that metabolic reprogramming is a key event in tumor progression. The tumor microenvironment (TME) is a source of metabolites for tumor cells. Lipid-filled mature adipocytes are frequently found in proximity to invasive human tumors and release free fatty acids (FFAs) through lipolysis. These FFAs are taken up by tumor cells and used to promote tumor progression by mechanisms that include mitochondrial fatty acid oxidation (FAO). This review discusses recent advances in our understanding of this metabolic symbiosis between adipocytes and cancer cells and underlines the differences in this metabolic crosstalk between the various types of cancer and their localization.


Subject(s)
Adipocytes/metabolism , Fatty Acids, Nonesterified/metabolism , Lipolysis , Neoplasms/pathology , Tumor Microenvironment , Adipocytes/cytology , Disease Progression , Energy Metabolism , Extracellular Vesicles/metabolism , Humans , Lipase/metabolism , Lipid Droplets/metabolism , Mitochondria/metabolism , Oxidation-Reduction
9.
EMBO J ; 39(3): e102525, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31919869

ABSTRACT

Extracellular vesicles are emerging key actors in adipocyte communication. Notably, small extracellular vesicles shed by adipocytes stimulate fatty acid oxidation and migration in melanoma cells and these effects are enhanced in obesity. However, the vesicular actors and cellular processes involved remain largely unknown. Here, we elucidate the mechanisms linking adipocyte extracellular vesicles to metabolic remodeling and cell migration. We show that adipocyte vesicles stimulate melanoma fatty acid oxidation by providing both enzymes and substrates. In obesity, the heightened effect of extracellular vesicles depends on increased transport of fatty acids, not fatty acid oxidation-related enzymes. These fatty acids, stored within lipid droplets in cancer cells, drive fatty acid oxidation upon being released by lipophagy. This increase in mitochondrial activity redistributes mitochondria to membrane protrusions of migrating cells, which is necessary to increase cell migration in the presence of adipocyte vesicles. Our results provide key insights into the role of extracellular vesicles in the metabolic cooperation that takes place between adipocytes and tumors with particular relevance to obesity.


Subject(s)
Adipocytes/cytology , Extracellular Vesicles/metabolism , Fatty Acids/metabolism , Melanoma/metabolism , Obesity/complications , 3T3 Cells , Adipocytes/metabolism , Animals , Autophagy , Cell Line, Tumor , Cell Movement , Humans , Lipid Metabolism , Male , Mice , Mitochondria/metabolism , Mitochondrial Dynamics , Obesity/metabolism , Oxidation-Reduction
10.
Cell Rep ; 30(4): 949-958.e6, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31995765

ABSTRACT

Under caloric restriction, bone marrow adipocytes (BM-Ads) do not decrease in size compared to white adipocytes, suggesting they harbor unique metabolic properties. We compare human primary BM-Ads with paired subcutaneous adipocytes (SC-Ads) using proteomic and lipidomic approaches. We find that, although SC-Ads and BM-Ads share similar morphological features, they possess distinct lipid metabolism. Although BM-Ad shows enrichment in proteins involved in cholesterol metabolism, correlating with increased free cholesterol content, proteins involved in lipolysis were downregulated. In particular, monoacylglycerol lipase expression is strongly reduced in BM-Ads, leading to monoacylglycerol accumulation. Consequently, basal and induced lipolytic responses are absent in BM-Ads, affirming their differences in metabolic fitness upon caloric restriction. These specific metabolic features are not recapitulated in vitro using common protocols to differentiate bone marrow mesenchymal stem cells. Thus, contrary to classical SC-Ads, BM-Ads display a specific lipid metabolism, as they are devoid of lipolytic activity and exhibit a cholesterol-orientated metabolism.


Subject(s)
Adipocytes/metabolism , Bone Marrow/metabolism , Lipid Metabolism , Proteome/metabolism , Adipocytes/cytology , Adipocytes/enzymology , Adipocytes/ultrastructure , Animals , Bone Marrow/enzymology , Caloric Restriction , Cell Line , Cells, Cultured , Cholesterol/metabolism , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Lipolysis/physiology , Mice , Microscopy, Electron, Transmission , Monoacylglycerol Lipases/genetics , Monoacylglycerol Lipases/metabolism , Protein Interaction Maps/genetics , Protein Interaction Maps/physiology , Proteome/genetics , Proteomics
11.
Curr Med Chem ; 27(24): 3984-4001, 2020.
Article in English | MEDLINE | ID: mdl-29708068

ABSTRACT

Metabolic reprogramming represents an important hallmark of cancer cells. Besides de novo fatty acid synthesis, it is now clear that cancer cells can acquire Fatty Acids (FA) from tumor-surrounding adipocytes to increase their invasive capacities. Indeed, adipocytes release FA in response to tumor secreted factors that are transferred to tumor cells to be either stored as triglycerides and other complex lipids or oxidized in mitochondria. Like all cells, FA can be released over time from triglyceride stores through lipolysis and then oxidized in mitochondria in cancer cells. This metabolic interaction results in specific metabolic remodeling in cancer cells, and underpins adipocyte stimulated tumor progression. Lipolysis and fatty acid oxidation therefore represent novel targets of interest in the treatment of cancer. In this review, we summarize the recent advances in our understanding of the metabolic reprogramming induced by adipocytes, with a focus on breast cancer. Then, we recapitulate recent reports studying the effect of lipolysis and fatty acid oxidation inhibitors on tumor cells and discuss the interest to target these metabolic pathways as new therapeutic approaches for cancer.


Subject(s)
Adipocytes , Lipolysis , Fatty Acids , Lipid Metabolism , Triglycerides
12.
Mol Cancer Res ; 17(3): 821-835, 2019 03.
Article in English | MEDLINE | ID: mdl-30606769

ABSTRACT

Prostate gland is surrounded by periprostatic adipose tissue (PPAT), which is increasingly believed to play a paracrine role in prostate cancer progression. Our previous work demonstrates that adipocytes promote homing of prostate cancer cells to PPAT and that this effect is upregulated by obesity. Here, we show that once tumor cells have invaded PPAT (mimicked by an in vitro model of coculture), they establish a bidirectional crosstalk with adipocytes, which promotes tumor cell invasion. Indeed, tumor cells induce adipocyte lipolysis and the free fatty acids (FFA) released are taken up and stored by tumor cells. Incubation with exogenous lipids also stimulates tumor cell invasion, underlining the importance of lipid transfer in prostate cancer aggressiveness. Transferred FFAs (after coculture or exogenous lipid treatment) stimulate the expression of one isoform of the pro-oxidant enzyme NADPH oxidase, NOX5. NOX5 increases intracellular reactive oxygen species (ROS) that, in turn, activate a HIF1/MMP14 pathway, which is responsible for the increased tumor cell invasion. In obesity, tumor-surrounding adipocytes are more prone to activate the depicted signaling pathway and to induce tumor invasion. Finally, the expression of NOX5 and MMP14 is upregulated at the invasive front of human tumors where cancer cells are in close proximity to adipocytes and this process is amplified in obese patients, underlining the clinical relevance of our results. IMPLICATIONS: Our work emphasizes the key role of adjacent PPAT in prostate cancer dissemination and proposes new molecular targets for the treatment of obese patients exhibiting aggressive diseases.


Subject(s)
Adipose Tissue/physiopathology , Obesity/complications , Prostatic Neoplasms/etiology , Animals , Cell Culture Techniques , Humans , Male , Mice , Oxidative Stress , Prostatic Neoplasms/pathology , Transfection
13.
J Lipid Res ; 59(10): 1793-1804, 2018 10.
Article in English | MEDLINE | ID: mdl-29678957

ABSTRACT

Cancer cells must adapt their metabolism in order to meet the energy requirements for cell proliferation, survival in nutrient-deprived environments, and dissemination. In particular, FA metabolism is emerging as a critical process for tumors. FA metabolism can be modulated through intrinsic changes in gene expression or signaling between tumor cells and also in response to signals from the surrounding microenvironment. Among these signals, extracellular vesicles (EVs) could play an important role in FA metabolism remodeling. In this review, we will present the role of EVs in tumor progression and especially in metabolic reprogramming. Particular attention will be granted to adipocytes. These cells, which are specialized in storing and releasing FAs, are able to shift tumor metabolism toward the use of FAs and, subsequently, increase tumor aggressiveness. Recent work demonstrates the involvement of EVs in this metabolic symbiosis.


Subject(s)
Extracellular Vesicles/metabolism , Neoplasms/pathology , Adipocytes/metabolism , Adipocytes/pathology , Animals , Carcinogenesis , Disease Progression , Humans , Neoplasms/metabolism
14.
JCI Insight ; 2(4): e87489, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28239646

ABSTRACT

In breast cancer, a key feature of peritumoral adipocytes is their loss of lipid content observed both in vitro and in human tumors. The free fatty acids (FFAs), released by adipocytes after lipolysis induced by tumor secretions, are transferred and stored in tumor cells as triglycerides in lipid droplets. In tumor cell lines, we demonstrate that FFAs can be released over time from lipid droplets through an adipose triglyceride lipase-dependent (ATGL-dependent) lipolytic pathway. In vivo, ATGL is expressed in human tumors where its expression correlates with tumor aggressiveness and is upregulated by contact with adipocytes. The released FFAs are then used for fatty acid ß-oxidation (FAO), an active process in cancer but not normal breast epithelial cells, and regulated by coculture with adipocytes. However, in cocultivated cells, FAO is uncoupled from ATP production, leading to AMPK/acetyl-CoA carboxylase activation, a circle that maintains this state of metabolic remodeling. The increased invasive capacities of tumor cells induced by coculture are completely abrogated by inhibition of the coupled ATGL-dependent lipolysis/FAO pathways. These results show a complex metabolic symbiosis between tumor-surrounding adipocytes and cancer cells that stimulate their invasiveness, highlighting ATGL as a potential therapeutic target to impede breast cancer progression.


Subject(s)
Adipocytes/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/metabolism , Fatty Acids, Nonesterified/metabolism , Lipase/metabolism , Lipolysis , Acetyl-CoA Carboxylase/metabolism , Adenosine Triphosphate/metabolism , Adenylate Kinase/metabolism , Aged , Animals , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/pathology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Female , Humans , Mice , Middle Aged , Neoplasm Invasiveness , Oxidation-Reduction , Triglycerides/metabolism
15.
Diabetologia ; 59(12): 2654-2663, 2016 12.
Article in English | MEDLINE | ID: mdl-27677764

ABSTRACT

AIMS/HYPOTHESIS: To directly assess the role of beta cell lipolysis in insulin secretion and whole-body energy homeostasis, inducible beta cell-specific adipose triglyceride lipase (ATGL)-deficient (B-Atgl-KO) mice were studied under normal diet (ND) and high-fat diet (HFD) conditions. METHODS: Atgl flox/flox mice were cross-bred with Mip-Cre-ERT mice to generate Mip-Cre-ERT/+;Atgl flox/flox mice. At 8 weeks of age, these mice were injected with tamoxifen to induce deletion of beta cell-specific Atgl (also known as Pnpla2), and the mice were fed an ND or HFD. RESULTS: ND-fed male B-Atgl-KO mice showed decreased insulinaemia and glucose-induced insulin secretion (GSIS) in vivo. Changes in GSIS correlated with the islet content of long-chain saturated monoacylglycerol (MAG) species that have been proposed to be metabolic coupling factors for insulin secretion. Exogenous MAGs restored GSIS in B-Atgl-KO islets. B-Atgl-KO male mice fed an HFD showed reduced insulinaemia, glycaemia in the fasted and fed states and after glucose challenge, as well as enhanced insulin sensitivity. Moreover, decreased insulinaemia in B-Atgl-KO mice was associated with increased energy expenditure, and lipid metabolism in brown (BAT) and white (WAT) adipose tissues, leading to reduced fat mass and body weight. CONCLUSIONS/INTERPRETATION: ATGL in beta cells regulates insulin secretion via the production of signalling MAGs. Decreased insulinaemia due to lowered GSIS protects B-Atgl-KO mice from diet-induced obesity, improves insulin sensitivity, increases lipid mobilisation from WAT and causes BAT activation. The results support the concept that fuel excess can drive obesity and diabetes via hyperinsulinaemia, and that an islet beta cell ATGL-lipolysis/adipose tissue axis controls energy homeostasis and body weight via insulin secretion.


Subject(s)
Adipose Tissue/metabolism , Body Weight/physiology , Energy Metabolism/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Adipose Tissue/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Blotting, Western , Calcium/metabolism , Diet, High-Fat/adverse effects , Female , Homeostasis/drug effects , Homeostasis/physiology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Lipase/metabolism , Lipid Metabolism/drug effects , Lipolysis/drug effects , Lipolysis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology , Tandem Mass Spectrometry
16.
J Clin Invest ; 126(9): 3598-612, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27525435

ABSTRACT

Disorders of glucose homeostasis are common in chronic kidney disease (CKD) and are associated with increased mortality, but the mechanisms of impaired insulin secretion in this disease remain unclear. Here, we tested the hypothesis that defective insulin secretion in CKD is caused by a direct effect of urea on pancreatic ß cells. In a murine model in which CKD is induced by 5/6 nephrectomy (CKD mice), we observed defects in glucose-stimulated insulin secretion in vivo and in isolated islets. Similarly, insulin secretion was impaired in normal mouse and human islets that were cultured with disease-relevant concentrations of urea and in islets from normal mice treated orally with urea for 3 weeks. In CKD mouse islets as well as urea-exposed normal islets, we observed an increase in oxidative stress and protein O-GlcNAcylation. Protein O-GlcNAcylation was also observed in pancreatic sections from CKD patients. Impairment of insulin secretion in both CKD mouse and urea-exposed islets was associated with reduced glucose utilization and activity of phosphofructokinase 1 (PFK-1), which could be reversed by inhibiting O-GlcNAcylation. Inhibition of O-GlcNAcylation also restored insulin secretion in both mouse models. These results suggest that insulin secretory defects associated with CKD arise from elevated circulating levels of urea that increase islet protein O-GlcNAcylation and impair glycolysis.


Subject(s)
Glycolysis , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Kidney Failure, Chronic/metabolism , Urea/chemistry , Animals , Antioxidants/metabolism , Cyanates/chemistry , Disease Models, Animal , Exocytosis , Glucokinase/metabolism , Glucose/metabolism , Glucose Tolerance Test , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Phosphofructokinase-1/metabolism , Reactive Oxygen Species/metabolism , Uremia/metabolism
17.
PLoS One ; 11(7): e0159165, 2016.
Article in English | MEDLINE | ID: mdl-27403868

ABSTRACT

Many metabolic studies employ tissue-specific gene knockout mice, which requires breeding of floxed gene mice, available mostly on C57BL/6N (NN) genetic background, with cre or Flp recombinase-expressing mice, available on C57BL/6J (JJ) background, resulting in the generation of mixed C57BL/6NJ (NJ) genetic background mice. Recent awareness of many genetic differences between NN and JJ strains including the deletion of nicotinamide nucleotide transhydrogenase (nnt), necessitates examination of the consequence of mixed NJ background on glucose tolerance, beta cell function and other metabolic parameters. Male mice with NN and NJ genetic background were fed with normal or high fat diets (HFD) for 12 weeks and glucose and insulin homeostasis were studied. Genotype had no effect on body weight and food intake in mice fed normal or high fat diets. Insulinemia in the fed and fasted states and after a glucose challenge was lower in HFD-fed NJ mice, even though their glycemia and insulin sensitivity were similar to NN mice. NJ mice showed mild glucose intolerance. Moreover, glucose- but not KCl-stimulated insulin secretion in isolated islets was decreased in HFD-fed NJ vs NN mice without changes in insulin content and beta cell mass. Under normal diet, besides reduced fed insulinemia, NN and NJ mice presented similar metabolic parameters. However, HFD-fed NJ mice displayed lower fed and fasted insulinemia and glucose-induced insulin secretion in vivo and ex vivo, as compared to NN mice. These results strongly caution against using unmatched mixed genetic background C57BL/6 mice for comparisons, particularly under HFD conditions.


Subject(s)
Diet, High-Fat/adverse effects , Genetic Background , Insulin/metabolism , Animals , Genotype , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Insulin Resistance/genetics , Insulin Secretion , Male , Mice , Mice, Inbred C57BL
18.
PLoS One ; 11(4): e0153017, 2016.
Article in English | MEDLINE | ID: mdl-27043434

ABSTRACT

Diet induced obese (DIO) mice can be stratified according to their weight gain in response to high fat diet as low responders (LDR) and high responders (HDR). This allows the study of ß-cell failure and the transitions to prediabetes (LDR) and early diabetes (HDR). C57BL/6N mice were fed for 8 weeks with a normal chow diet (ND) or a high fat diet and stratified as LDR and HDR. Freshly isolated islets from ND, LDR and HDR mice were studied ex-vivo for mitochondrial metabolism, AMPK activity and signalling, the expression and activity of key enzymes of energy metabolism, cholesterol synthesis, and mRNA profiling. Severely compromised glucose-induced insulin secretion in HDR islets, as compared to ND and LDR islets, was associated with suppressed AMP-kinase activity. HDR islets also showed reduced acetyl-CoA carboxylase activity and enhanced activity of 3-hydroxy-3-methylglutaryl-CoA reductase, which led respectively to elevated fatty acid oxidation and increased cholesterol biosynthesis. HDR islets also displayed mitochondrial membrane hyperpolarization and reduced ATP turnover in the presence of elevated glucose. Expression of protein kinase Cε, which reduces both lipolysis and production of signals for insulin secretion, was elevated in DIO islets. Genes whose expression increased or decreased by more than 1.2-fold were minor between LDR and ND islets (17 differentially expressed), but were prominent between HDR and ND islets (1508 differentially expressed). In HDR islets, particularly affected genes were related to cell cycle and proliferation, AMPK signaling, mitochondrial metabolism and cholesterol metabolism. In conclusion, chronically reduced AMPK activity, mitochondrial dysfunction, elevated cholesterol biosynthesis in islets, and substantial alterations in gene expression accompany ß-cell failure in HDR islets. The ß-cell compensation process in the prediabetic state (LDR) is largely independent of transcriptional adaptive changes, whereas the transition to early diabetes (HDR) is associated with major alterations in gene expression.


Subject(s)
Diet/adverse effects , Insulin-Secreting Cells/metabolism , Obesity/etiology , Obesity/metabolism , Adenylate Kinase/metabolism , Animals , Cells, Cultured , Cholesterol/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Insulin/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mitochondria/genetics , Mitochondria/metabolism , Oxygen Consumption , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Transcriptome
19.
Cell Rep ; 14(12): 2872-88, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26997277

ABSTRACT

Suppression of α/ß-domain hydrolase-6 (ABHD6), a monoacylglycerol (MAG) hydrolase, promotes glucose-stimulated insulin secretion by pancreatic ß cells. We report here that high-fat-diet-fed ABHD6-KO mice show modestly reduced food intake, decreased body weight gain and glycemia, improved glucose tolerance and insulin sensitivity, and enhanced locomotor activity. ABHD6-KO mice also show increased energy expenditure, cold-induced thermogenesis, brown adipose UCP1 expression, fatty acid oxidation, and white adipose browning. Adipose browning and cold-induced thermogenesis are replicated by the ABHD6 inhibitor WWL70 and by antisense oligonucleotides targeting ABHD6. Our evidence suggests that one mechanism by which the lipolysis derived 1-MAG signals intrinsic and cell-autonomous adipose browning is via PPARα and PPARγ activation, and that ABHD6 regulates adipose browning by controlling signal competent 1-MAG levels. Thus, ABHD6 regulates energy homeostasis, brown adipose function, and white adipose browning and is a potential therapeutic target for obesity and type 2 diabetes.


Subject(s)
Adipose Tissue, Brown/metabolism , Diabetes Mellitus, Type 2/genetics , Monoacylglycerol Lipases/metabolism , Obesity/genetics , 3T3-L1 Cells , Animals , Biphenyl Compounds/pharmacology , Carbamates/pharmacology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/prevention & control , Diet, High-Fat , Diglycerides/pharmacology , Energy Metabolism/drug effects , Female , HEK293 Cells , Humans , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoacylglycerol Lipases/antagonists & inhibitors , Monoacylglycerol Lipases/genetics , Motor Activity/drug effects , Obesity/etiology , Obesity/metabolism , Obesity/prevention & control , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , PPAR gamma/metabolism , Thermogenesis , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
20.
Proc Natl Acad Sci U S A ; 113(4): E430-9, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26755581

ABSTRACT

Obesity, and the associated disturbed glycerolipid/fatty acid (GL/FA) cycle, contribute to insulin resistance, islet ß-cell failure, and type 2 diabetes. Flux through the GL/FA cycle is regulated by the availability of glycerol-3-phosphate (Gro3P) and fatty acyl-CoA. We describe here a mammalian Gro3P phosphatase (G3PP), which was not known to exist in mammalian cells, that can directly hydrolyze Gro3P to glycerol. We identified that mammalian phosphoglycolate phosphatase, with an uncertain function, acts in fact as a G3PP. We found that G3PP, by controlling Gro3P levels, regulates glycolysis and glucose oxidation, cellular redox and ATP production, gluconeogenesis, glycerolipid synthesis, and fatty acid oxidation in pancreatic islet ß-cells and hepatocytes, and that glucose stimulated insulin secretion and the response to metabolic stress, e.g., glucolipotoxicity, in ß-cells. In vivo overexpression of G3PP in rat liver lowers body weight gain and hepatic glucose production from glycerol and elevates plasma HDL levels. G3PP is expressed at various levels in different tissues, and its expression varies according to the nutritional state in some tissues. As Gro3P lies at the crossroads of glucose, lipid, and energy metabolism, control of its availability by G3PP adds a key level of metabolic regulation in mammalian cells, and G3PP offers a potential target for type 2 diabetes and cardiometabolic disorders.


Subject(s)
Carbohydrate Metabolism/physiology , Glycerophosphates/metabolism , Hepatocytes/enzymology , Insulin-Secreting Cells/enzymology , Lipid Metabolism/physiology , Phosphoric Monoester Hydrolases/physiology , Signal Transduction/physiology , Amino Acid Sequence , Animals , Cell Line , Fatty Acids/metabolism , Glycerol/metabolism , Hydrolysis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Lactones/pharmacology , Male , Mice , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Nutritional Status , Orlistat , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , RNA Interference , Rats , Sequence Homology, Amino Acid , Stress, Physiological/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...