Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Adv Exp Med Biol ; 960: 161-196, 2017.
Article in English | MEDLINE | ID: mdl-28585199

ABSTRACT

The adipose tissue organ is organised as distinct anatomical depots located all along the body axis and it is constituted of three different types of adipocytes : white, beige and brown which are integrated with vascular, immune, neural and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concert action of the three type of adipocytes/tissues has been reported to ensure an optimal metabolic status in rodents. However, when one or multiple of these adipose depots become dysfunctional as a consequence of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity counteracts obesity and its associated lipotoxic metabolic effects. The development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition and expandability capacity as well as molecular and metabolic signatures in both physiological and pathophysiological conditions.


Subject(s)
Adipose Tissue/physiology , Metabolic Syndrome/physiopathology , Adipocytes/metabolism , Adipocytes/physiology , Adipose Tissue/metabolism , Animals , Energy Metabolism/physiology , Humans , Lipid Metabolism/physiology , Metabolic Syndrome/metabolism , Obesity/metabolism , Obesity/physiopathology
2.
Diabetologia ; 59(6): 1075-88, 2016 06.
Article in English | MEDLINE | ID: mdl-27039901

ABSTRACT

White adipose tissue (WAT) has key metabolic and endocrine functions and plays a role in regulating energy homeostasis and insulin sensitivity. WAT is characterised by its capacity to adapt and expand in response to surplus energy through processes of adipocyte hypertrophy and/or recruitment and proliferation of precursor cells in combination with vascular and extracellular matrix remodelling. However, in the context of sustained obesity, WAT undergoes fibro-inflammation, which compromises its functionality, contributing to increased risk of type 2 diabetes and cardiovascular diseases. Conversely, brown adipose tissue (BAT) and browning of WAT represent potential therapeutic approaches, since dysfunctional white adipocyte-induced lipid overspill can be halted by BAT/browning-mediated oxidative anti-lipotoxic effects. Better understanding of the cellular and molecular pathophysiological mechanisms regulating adipocyte size, number and depot-dependent expansion has become a focus of interest over recent decades. Here, we summarise the mechanisms contributing to adipose tissue (AT) plasticity and function including characteristics and cellular complexity of the various adipose depots and we discuss recent insights into AT origins, identification of adipose precursors, pathophysiological regulation of adipogenesis and its relation to WAT/BAT expandability in obesity and its associated comorbidities.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipogenesis/physiology , Adipose Tissue/cytology , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fibrosis/metabolism , Fibrosis/pathology , Humans , Obesity/metabolism , Obesity/pathology
3.
Cell Rep ; 42(6): 112640, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37318951

ABSTRACT

The relevance of extracellular matrix (ECM) remodeling is reported in white adipose tissue (AT) and obesity-related dysfunctions, but little is known about the importance of ECM remodeling in brown AT (BAT) function. Here, we show that a time course of high-fat diet (HFD) feeding progressively impairs diet-induced thermogenesis concomitantly with the development of fibro-inflammation in BAT. Higher markers of fibro-inflammation are associated with lower cold-induced BAT activity in humans. Similarly, when mice are housed at thermoneutrality, inactivated BAT features fibro-inflammation. We validate the pathophysiological relevance of BAT ECM remodeling in response to temperature challenges and HFD using a model of a primary defect in the collagen turnover mediated by partial ablation of the Pepd prolidase. Pepd-heterozygous mice display exacerbated dysfunction and BAT fibro-inflammation at thermoneutrality and in HFD. Our findings show the relevance of ECM remodeling in BAT activation and provide a mechanism for BAT dysfunction in obesity.


Subject(s)
Adipose Tissue, Brown , Obesity , Humans , Animals , Mice , Adipose Tissue, Brown/metabolism , Obesity/metabolism , Diet, High-Fat , Inflammation/metabolism , Adipose Tissue, White/metabolism , Extracellular Matrix , Thermogenesis , Energy Metabolism , Mice, Inbred C57BL
4.
Nat Commun ; 9(1): 4974, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30478315

ABSTRACT

Activation of brown adipose tissue-mediated thermogenesis is a strategy for tackling obesity and promoting metabolic health. BMP8b is secreted by brown/beige adipocytes and enhances energy dissipation. Here we show that adipocyte-secreted BMP8b contributes to adrenergic-induced remodeling of the neuro-vascular network in adipose tissue (AT). Overexpression of bmp8b in AT enhances browning of the subcutaneous depot and maximal thermogenic capacity. Moreover, BMP8b-induced browning, increased sympathetic innervation and vascularization of AT were maintained at 28 °C, a condition of low adrenergic output. This reinforces the local trophic effect of BMP8b. Innervation and vascular remodeling effects required BMP8b signaling through the adipocytes to 1) secrete neuregulin-4 (NRG4), which promotes sympathetic axon growth and branching in vitro, and 2) induce a pro-angiogenic transcriptional and secretory profile that promotes vascular sprouting. Thus, BMP8b and NRG4 can be considered as interconnected regulators of neuro-vascular remodeling in AT and are potential therapeutic targets in obesity.


Subject(s)
Adipocytes, Brown/metabolism , Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/innervation , Adrenergic Agents/pharmacology , Bone Morphogenetic Proteins/metabolism , 3T3-L1 Cells , Adipose Tissue, Brown/metabolism , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Neovascularization, Physiologic , Neuregulins/genetics , Neuregulins/metabolism , Proteomics , Signal Transduction , Subcutaneous Fat/metabolism , Thermogenesis , Vascular Endothelial Growth Factor A/metabolism
5.
Diabetes ; 64(9): 3121-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25695947

ABSTRACT

Inflammation and lipid accumulation are hallmarks of muscular pathologies resulting from metabolic diseases such as obesity and type 2 diabetes. During obesity, the hypertrophy of visceral adipose tissue (VAT) contributes to muscle dysfunction, particularly through the dysregulated production of adipokines. We have investigated the cross talk between human adipocytes and skeletal muscle cells to identify mechanisms linking adiposity and muscular dysfunctions. First, we demonstrated that the secretome of obese adipocytes decreased the expression of contractile proteins in myotubes, consequently inducing atrophy. Using a three-dimensional coculture of human myotubes and VAT adipocytes, we showed the decreased expression of genes corresponding to skeletal muscle contractility complex and myogenesis. We demonstrated an increased secretion by cocultured cells of cytokines and chemokines with interleukin (IL)-6 and IL-1ß as key contributors. Moreover, we gathered evidence showing that obese subcutaneous adipocytes were less potent than VAT adipocytes in inducing these myotube dysfunctions. Interestingly, the atrophy induced by visceral adipocytes was corrected by IGF-II/insulin growth factor binding protein-5. Finally, we observed that the skeletal muscle of obese mice displayed decreased expression of muscular markers in correlation with VAT hypertrophy and abnormal distribution of the muscle fiber size. In summary, we show the negative impact of obese adipocytes on muscle phenotype, which could contribute to muscle wasting associated with metabolic disorders.


Subject(s)
Adipocytes/metabolism , Contractile Proteins/metabolism , Intra-Abdominal Fat/cytology , Muscle Fibers, Skeletal/metabolism , Obesity, Morbid/metabolism , Adipocytes/immunology , Adult , Animals , Atrophy/immunology , Atrophy/metabolism , Coculture Techniques , Cytokines/immunology , Female , Gene Expression Regulation , Humans , Inflammation , Insulin-Like Growth Factor Binding Protein 5/pharmacology , Insulin-Like Growth Factor II/pharmacology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Obese , Muscle Fibers, Skeletal/immunology , Muscle Fibers, Skeletal/pathology , Obesity, Morbid/immunology , Subcutaneous Fat/cytology , Subcutaneous Fat/immunology , Subcutaneous Fat/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
6.
Diabetes ; 64(4): 1180-92, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25352638

ABSTRACT

Adipose tissue dysfunction is an important determinant of obesity-associated, lipid-induced metabolic complications. Ceramides are well-known mediators of lipid-induced insulin resistance in peripheral organs such as muscle. DEGS1 is the desaturase catalyzing the last step in the main ceramide biosynthetic pathway. Functional suppression of DEGS1 activity results in substantial changes in ceramide species likely to affect fundamental biological functions such as oxidative stress, cell survival, and proliferation. Here, we show that degs1 expression is specifically decreased in the adipose tissue of obese patients and murine models of genetic and nutritional obesity. Moreover, loss-of-function experiments using pharmacological or genetic ablation of DEGS1 in preadipocytes prevented adipogenesis and decreased lipid accumulation. This was associated with elevated oxidative stress, cellular death, and blockage of the cell cycle. These effects were coupled with increased dihydroceramide content. Finally, we validated in vivo that pharmacological inhibition of DEGS1 impairs adipocyte differentiation. These data identify DEGS1 as a new potential target to restore adipose tissue function and prevent obesity-associated metabolic disturbances.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Ceramides/metabolism , Fatty Acid Desaturases/metabolism , Obesity/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipogenesis/drug effects , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adult , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Death/drug effects , Cell Death/physiology , Ceramides/pharmacology , Fatty Acid Desaturases/antagonists & inhibitors , Fatty Acid Desaturases/genetics , Female , Humans , Insulin/metabolism , Lipolysis/drug effects , Lipolysis/physiology , Male , Mice , Middle Aged , Oxidative Stress/drug effects , Oxidative Stress/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Diabetes ; 63(2): 535-49, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24130331

ABSTRACT

During obesity, chronic inflammation of human white adipose tissue (WAT) is associated with metabolic and vascular alterations. Endothelial cells from visceral WAT (VAT-ECs) exhibit a proinflammatory and senescent phenotype and could alter adipocyte functions. We aimed to determine the contribution of VAT-ECs to adipocyte dysfunction related to inflammation and to rescue these alterations by anti-inflammatory strategies. We developed an original three-dimensional setting allowing maintenance of unilocular adipocyte functions. Coculture experiments demonstrated that VAT-ECs provoked a decrease in the lipolytic activity, adipokine secretion, and insulin sensitivity of adipocytes from obese subjects, as well as an increased production of several inflammatory molecules. Interleukin (IL)-6 and IL-1ß were identified as potential actors in these adipocyte alterations. The inflammatory burst was not observed in cocultured cells from lean subjects. Interestingly, pericytes, in functional interactions with ECs, exhibited a proinflammatory phenotype with diminished angiopoietin-1 (Ang-1) secretion in WAT from obese subjects. Using the anti-inflammatory Ang-1, we corrected some deleterious effects of WAT-ECs on adipocytes, improving lipolytic activity and insulin sensitivity and reducing the secretion of proinflammatory molecules. In conclusion, we identified a negative impact of VAT-ECs on adipocyte functions during human obesity. Therapeutic options targeting EC inflammation could prevent adipocyte alterations that contribute to obesity comorbidities.


Subject(s)
Adipocytes/physiology , Adipose Tissue/cytology , Angiopoietin-1/pharmacology , Endothelial Cells/physiology , Animals , Cell Culture Techniques , Humans , Inflammation , Insulin , Lipolysis/physiology , Obesity/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Endocrinology ; 154(3): 1069-79, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23372021

ABSTRACT

The hypertrophied white adipose tissue (WAT) during human obesity produces inflammatory mediators, including cytokines (IL-6 and TNFα) and chemokines ([C-C motif] chemokine ligand 2 and IL-8). These inflammatory factors are preferentially produced by the nonadipose cells, particularly the adipose tissue infiltrating macrophages. We identified the chemokine (C-X-C motif) ligand 2 (CXCL2) by a transcriptomic approach. Because CXCL2 could represent a WAT-produced chemokine, we explored its role in obesity-associated inflammation. CXCL2 levels in serum and mRNA in WAT were higher in obese subjects compared with lean ones. CXCL2 secretions were higher in sc and visceral (vis) WAT from obese compared with lean subjects. In vis WAT, CXCL2 mRNA expression was higher in macrophages compared with other WAT cells and positively correlated with the inflammatory macrophage markers TNFα and IL-6. CXCL2 triggered the in vitro adhesion of the neutrophils, its selective cell targets, to endothelial cells (ECs) of vis WAT (vis WAT-ECs). Immunohistological analysis indicated that activated neutrophils were adherent to the endothelium of vis WAT from human obese subjects. Blood neutrophils from obese subjects released high levels of proinflammatory mediators (IL-8, chemokine motif ligand 2 [CCL2], matrix metalloproteinase [MMP] 9, and myeloperoxidase [MPO]). Visceral WAT-ECs, treated by neutrophil-conditioned media prepared from obese subjects, displayed an increase of the expression of inflammatory molecules associated with senescence and angiogenic capacities. To conclude, CXCL2, a WAT-produced chemokine being up-regulated in obesity, stimulates neutrophil adhesion to vis WAT-ECs. Activated neutrophils in obesity may influence vis WAT-ECs functions and contribute to WAT inflammation.


Subject(s)
Adipose Tissue, White/physiopathology , Chemokine CXCL2/physiology , Neutrophils/physiology , Obesity, Morbid/physiopathology , Adipose Tissue, White/pathology , Adult , Case-Control Studies , Cell Adhesion/physiology , Chemokine CXCL2/genetics , Chemokines/physiology , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , Humans , Inflammation/etiology , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/physiology , Neutrophil Activation/physiology , Neutrophils/pathology , Obesity, Morbid/genetics , Obesity, Morbid/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL