Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2233: 203-222, 2021.
Article in English | MEDLINE | ID: mdl-33222137

ABSTRACT

Exocytosis, the fusion of vesicles with the plasma membrane, can be measured with the patch-clamp technique as increases in membrane capacitance. Here we provide detailed information on how to monitor white adipocyte exocytosis using this method. We describe how to isolate the stromal vascular fraction of cells (SVF) within adipose tissue and how to differentiate SVF and cultured 3T3-L1 cells into adipocytes suitable for patch-clamp studies. We also give detailed protocols of how to record and analyze exocytosis in the differentiated cells.


Subject(s)
Adipocytes, White/ultrastructure , Cell Membrane/genetics , Exocytosis/genetics , Patch-Clamp Techniques/methods , 3T3-L1 Cells , Adipocytes, White/metabolism , Animals , Cell Membrane/ultrastructure , Mice
2.
Am J Physiol Endocrinol Metab ; 318(6): E866-E877, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32315212

ABSTRACT

The receptor activator of nuclear factor-κB (NF-κB) (RANK), its ligand (RANKL), and the decoy receptor osteoprotegerin (OPG) are a triad of proteins that regulate bone metabolism, and serum OPG is considered a biomarker for cardiovascular diseases and Type 2 diabetes; however, the implications of OPG in adipose tissue metabolism remains elusive. In this study, we investigate RANK-RANKL-OPG signaling in white adipose tissue browning. Histological analysis of osteoprotegerin knockout (OPG-/-) mice showed subcutaneous white adipose tissue (sWAT) browning, resistance for high-fat diet-induced weight gain, and preserved glucose metabolism compared with wild-type (WT) mice. Stromal vascular fraction (SVF) cells from sWAT of OPG-/- mice showed multilocular morphology and higher expression of brown adipocyte marker genes compared with those from the WT group. Infusion of RANKL induced browning and elevated respiratory rates in sWAT, along with increased whole body oxygen consumption in mice measured by indirect calorimetry. Subcutaneous WAT-derived SVF and 3T3-L1 cells, but not mature white adipocytes, differentiated into beige adipose tissue in the presence of RANKL. Moreover, SVF cells, even under white adipocyte differentiation, showed multilocular lipid droplet, lower lipid content, and increased expression of beige adipocyte markers with RANKL stimulation. In this study, we show for the first time the contribution of RANKL to increase energy expenditure by inducing beige adipocyte differentiation in preadipocytes.


Subject(s)
Adipocytes, Beige/metabolism , Adipogenesis/genetics , Adipose Tissue, White/metabolism , Obesity/metabolism , Osteoprotegerin/genetics , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , 3T3-L1 Cells , Adipocytes, Beige/cytology , Adipocytes, Beige/ultrastructure , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipocytes, White/ultrastructure , Adipose Tissue, Beige/cytology , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/cytology , Animals , Calorimetry, Indirect , Diet, High-Fat , Energy Metabolism/drug effects , Energy Metabolism/genetics , Lipid Droplets/ultrastructure , Mice , Mice, Knockout , Osteoprotegerin/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/genetics , RANK Ligand/pharmacology , Signal Transduction , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , Weight Gain/drug effects , Weight Gain/genetics
3.
Adv Nutr ; 11(2): 447-460, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31355852

ABSTRACT

The high prevalence of obesity and its associated metabolic diseases has heightened the importance of understanding control of adipose tissue development and energy metabolism. In mammals, 3 types of adipocytes with different characteristics and origins have been identified: white, brown, and beige. Beige and brown adipocytes contain numerous mitochondria and have the capability to burn energy and counteract obesity, while white adipocytes store energy and are closely associated with metabolic disorders and obesity. Thus, regulation of the development and function of different adipocytes is important for controlling energy balance and combating obesity and related metabolic disorders. Melatonin is a neurohormone, which plays multiple roles in regulating inflammation, blood pressure, insulin actions, and energy metabolism. This article summarizes and discusses the role of melatonin in white, beige, and brown adipocytes, especially in affecting adipogenesis, inducing beige formation or white adipose tissue browning, enhancing brown adipose tissue mass and activities, improving anti-inflammatory and antioxidative effects, regulating adipokine secretion, and preventing body weight gain. Based on the current findings, melatonin is a potential therapeutic agent to control energy metabolism, adipogenesis, fat deposition, adiposity, and related metabolic diseases.


Subject(s)
Adipocytes, Beige/physiology , Adipocytes, Brown/physiology , Adipocytes, White/physiology , Melatonin/physiology , Adipocytes, Beige/ultrastructure , Adipocytes, Brown/ultrastructure , Adipocytes, White/ultrastructure , Adipogenesis/physiology , Adiposity/physiology , Animals , Body Weight/physiology , Cell Differentiation/physiology , Energy Metabolism/physiology , Homeostasis , Humans , Mitochondria/physiology , Obesity/physiopathology
4.
Diabetes ; 67(3): 400-411, 2018 03.
Article in English | MEDLINE | ID: mdl-29298809

ABSTRACT

Sucrose nonfermenting-related kinase (SNRK) is a member of the AMPK-related kinase family, and its physiological role in adipose energy homeostasis and inflammation remains unknown. We previously reported that SNRK is ubiquitously and abundantly expressed in both white adipose tissue (WAT) and brown adipose tissue (BAT), but SNRK expression diminishes in adipose tissue in obesity. In this study we report novel experimental findings from both animal models and human genetics. SNRK is essential for survival; SNRK globally deficient pups die within 24 h after birth. Heterozygous mice are characterized by inflamed WAT and less BAT. Adipocyte-specific ablation of SNRK causes inflammation in WAT, ectopic lipid deposition in liver and muscle, and impaired adaptive thermogenesis in BAT. These metabolic disorders subsequently lead to decreased energy expenditure, higher body weight, and insulin resistance. We further confirm the significant association of common variants of the SNRK gene with obesity risk in humans. Through applying a phosphoproteomic approach, we identified eukaryotic elongation factor 1δ and histone deacetylase 1/2 as potential SNRK substrates. Taking these data together, we conclude that SNRK represses WAT inflammation and is essential to maintain BAT thermogenesis, making it a novel therapeutic target for treating obesity and associated metabolic disorders.


Subject(s)
Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Energy Metabolism , Mitochondria/metabolism , Panniculitis/metabolism , Protein Serine-Threonine Kinases/metabolism , Adipocytes, Brown/immunology , Adipocytes, Brown/pathology , Adipocytes, Brown/ultrastructure , Adipocytes, White/immunology , Adipocytes, White/pathology , Adipocytes, White/ultrastructure , Animals , Body Mass Index , Cells, Cultured , Crosses, Genetic , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria/immunology , Mitochondria/pathology , Mitochondria/ultrastructure , Obesity/genetics , Obesity/physiopathology , Panniculitis/etiology , Panniculitis/immunology , Panniculitis/pathology , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , Thermogenesis
5.
J Cell Physiol ; 232(10): 2887-2899, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27987321

ABSTRACT

White adipocytes are plastic cells able to reversibly transdifferentiate into brown adipocytes and into epithelial glandular cells under physiologic stimuli in vivo. These plastic properties could be used in future for regenerative medicine, but are incompletely explored in their details. Here, we focused on plastic properties of human mature adipocytes (MA) combining gene expression profile through microarray analysis with morphologic data obtained by electron and time lapse microscopy. Primary MA showed the classic morphology and gene expression profile of functional mature adipocytes. Notably, despite their committed status, MA expressed high levels of reprogramming genes. MA from ceiling cultures underwent transdifferentiation toward fibroblast-like cells with a well-differentiated morphology and maintaining stem cell gene signatures. The main morphologic aspect of the transdifferentiation process was the secretion of large lipid droplets and the development of organelles necessary for exocrine secretion further supported the liposecretion process. Of note, electron microscope findings suggesting liposecretion phenomena were found also in explants of human fat and rarely in vivo in fat biopsies from obese patients. In conclusion, both MA and post-liposecretion adipocytes show a well-differentiated phenotype with stem cell properties in line with the extraordinary plasticity of adipocytes in vivo. J. Cell. Physiol. 232: 2887-2899, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipogenesis , Cell Plasticity , Lipid Metabolism , Mesenchymal Stem Cells/metabolism , Obesity/metabolism , Adipocytes, Brown/ultrastructure , Adipocytes, White/ultrastructure , Aged , Aged, 80 and over , Cell Lineage , Cell Shape , Cells, Cultured , Cellular Reprogramming , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Genetic Markers , Humans , Lipid Droplets/metabolism , Mesenchymal Stem Cells/ultrastructure , Microscopy, Confocal , Microscopy, Electron , Microscopy, Video , Middle Aged , Obesity/pathology , Obesity/physiopathology , Oligonucleotide Array Sequence Analysis , Phenotype , Time Factors , Time-Lapse Imaging
6.
Diabetes ; 65(5): 1255-67, 2016 05.
Article in English | MEDLINE | ID: mdl-26956488

ABSTRACT

Coxsackie virus and adenovirus receptor-like membrane protein (CLMP) was identified as the tight junction-associated transmembrane protein of epithelial cells with homophilic binding activities. CLMP is also recognized as adipocyte adhesion molecule (ACAM), and it is upregulated in mature adipocytes in rodents and humans with obesity. Here, we present that aP2 promoter-driven ACAM transgenic mice are protected from obesity and diabetes with the prominent reduction of adipose tissue mass and smaller size of adipocytes. ACAM is abundantly expressed on plasma membrane of mature adipocytes and associated with formation of phalloidin-positive polymerized form of cortical actin (F-actin). By electron microscopy, the structure of zonula adherens with an intercellular space of ∼10-20 nm was observed with strict parallelism of the adjoining cell membranes over distances of 1-20 µm, where ACAM and γ-actin are abundantly expressed. The formation of zonula adherens may increase the mechanical strength, inhibit the adipocyte hypertrophy, and improve the insulin sensitivity.


Subject(s)
Actin Cytoskeleton/metabolism , Adherens Junctions/metabolism , Adipocytes, White/metabolism , Adiposity , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Obesity/prevention & control , Up-Regulation , 3T3-L1 Cells , Actin Cytoskeleton/pathology , Actin Cytoskeleton/ultrastructure , Adherens Junctions/pathology , Adherens Junctions/ultrastructure , Adipocytes, White/cytology , Adipocytes, White/pathology , Adipocytes, White/ultrastructure , Animals , Cell Adhesion , Cell Size , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetes Mellitus/prevention & control , Diet, High-Fat/adverse effects , Dietary Sucrose/adverse effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Recombinant Fusion Proteins/metabolism
7.
Biochim Biophys Acta ; 1861(5): 430-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26923434

ABSTRACT

Mechanistic target of rapamycin complex 1 (mTORC1) loss of function reduces adiposity whereas partial mTORC1 inhibition enhances fat deposition. Herein we evaluated how constitutive mTORC1 activation in adipocytes modulates adiposity in vivo. Mice with constitutive mTORC1 activation in adipocytes induced by tuberous sclerosis complex (Tsc)1 deletion and littermate controls were evaluated for body mass, energy expenditure, glucose and fatty acid metabolism, mitochondrial function, mRNA and protein contents. Adipocyte-specific Tsc1 deletion reduced visceral, but not subcutaneous, fat mass, as well as adipocyte number and diameter, phenotypes that were associated with increased lipolysis, UCP-1 content (browning) and mRNA levels of pro-browning transcriptional factors C/EBPß and ERRα. Adipocyte Tsc1 deletion enhanced mitochondrial oxidative activity, fatty acid oxidation and the expression of PGC-1α and PPARα in both visceral and subcutaneous fat. In brown adipocytes, however, Tsc1 deletion did not affect UCP-1 content and basal respiration. Adipocyte Tsc1 deletion also reduced visceral adiposity and enhanced glucose tolerance, liver and muscle insulin signaling and adiponectin secretion in mice fed with purified low- or high-fat diet. In conclusion, adipocyte-specific Tsc1 deletion enhances mitochondrial activity, induces browning and reduces visceral adiposity in mice.


Subject(s)
Adipocytes, Brown/enzymology , Adipocytes, White/enzymology , Adipose Tissue, Brown/enzymology , Adiposity , Intra-Abdominal Fat/enzymology , Mitochondria/enzymology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adipocytes, Brown/ultrastructure , Adipocytes, White/ultrastructure , Adiponectin/deficiency , Adiponectin/genetics , Adipose Tissue, Brown/ultrastructure , Adiposity/genetics , Animals , Cell Respiration , Diet, Fat-Restricted , Diet, High-Fat , Energy Metabolism , Enzyme Activation , Gene Expression Regulation , Genotype , Glucose/metabolism , Insulin/metabolism , Intra-Abdominal Fat/ultrastructure , Lipolysis , Male , Mechanistic Target of Rapamycin Complex 1 , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Oxidation-Reduction , Phenotype , Signal Transduction , Time Factors , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
8.
Am J Physiol Endocrinol Metab ; 303(8): E1053-60, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22912368

ABSTRACT

The transcription factor Tbx15 is expressed predominantly in brown adipose tissue and in those white adipose depots that are capable of giving rise to brown-in-white ("brite"/"beige") adipocytes. Therefore, we have investigated a possible role here of Tbx15 in brown and brite adipocyte differentiation in vitro. Adipocyte precursors were isolated from interscapular and axilliary brown adipose tissues, inguinal white ("brite") adipose tissue, and epididymal white adipose tissue in 129/Sv mouse pups and differentiated in culture. Differentiation was enhanced by chronic treatment with the PPARγ agonist rosiglitazone plus the sympathetic neurotransmitter norepinephrine. Using short interfering RNAs (siRNA) directed toward Tbx15 in these primary adipocyte cultures, we decreased Tbx15 expression >90%. This resulted in reduced expression levels of adipogenesis markers (PPARγ, aP2). Importantly, Tbx15 knockdown reduced the expression of brown phenotypic marker genes (PRDM16, PGC-1α, Cox8b/Cox4, UCP1) in brown adipocytes and even more markedly in inguinal white adipocytes. In contrast, Tbx15 knockdown had no effect on white adipocytes originating from a depot that is not brite competent in vivo (epididymal). Therefore, Tbx15 may be essential for the development of the adipogenic and thermogenic programs in adipocytes/adipomyocytes capable of developing brown adipocyte features.


Subject(s)
Adipocytes, Brown/physiology , Adipocytes, White/physiology , T-Box Domain Proteins/physiology , Adipocytes, Brown/ultrastructure , Adipocytes, White/ultrastructure , Adipogenesis/physiology , Animals , Blotting, Western , Cell Differentiation/physiology , DNA Primers , Genetic Markers , Hypoglycemic Agents/pharmacology , Mice , PPAR gamma/agonists , Phenotype , RNA/biosynthesis , RNA/isolation & purification , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Rosiglitazone , T-Box Domain Proteins/genetics , Thiazolidinediones/pharmacology
9.
J Nutr Biochem ; 23(9): 1100-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22137261

ABSTRACT

Resveratrol is a natural polyphenolic compound with anti-inflammatory, antioxidant and neuroprotective properties, and it serves as a chemopreventive and chemotherapeutic agent. However, only very limited data have been obtained regarding the effects of resveratrol on preadipocytes, and the mechanisms of these effects remain largely unknown. In this study, murine 3T3-L1 preadipocytes were incubated with resveratrol, and cell apoptosis was investigated. Resveratrol caused S-phase arrest to inhibit cell proliferation and significantly increased the lactate dehydrogenase leaking ratio. Hoechst 33258 staining and transmission electron microscopy revealed the ultrastructural changes in nuclear chromatins of apoptotic cells. Furthermore, resveratrol activated the mitochondrial signaling with decreases in the mitochondrial membrane potential, cytochrome c release and the activation of caspase 9 and caspase 3. Resveratrol treatment also increased the protein level of Sirt1. By using small interfering RNAs of Sirt1, adenosine-monophosphate-activated protein kinase (AMPK) α, survivin and the AMPK agonist (5-aminoimidazole-4-carboxamide 1-ß-D-ribofuranoside) and specific inhibitors for protein kinase B (AKT) or caspases, it was demonstrated that activation of Sirt1 inhibited AKT activation and further decreased the expression of survivin. It could also increase AMPK activation. Both signaling pathways activated mitochondrion-mediated pathway. Our findings clarified the apoptotic effects of resveratrol in 3T3-L1 preadipocytes and revealed the involved pathway including AMPK, AKT and survivin, suggesting its potential therapeutic application in the treatment or prevention of obesity and related metabolic symptoms.


Subject(s)
Adipocytes, White/drug effects , Antioxidants/pharmacology , Apoptosis/drug effects , Inhibitor of Apoptosis Proteins/metabolism , Repressor Proteins/metabolism , Second Messenger Systems/drug effects , Sirtuin 1/metabolism , Stilbenes/pharmacology , 3T3-L1 Cells , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipocytes, White/metabolism , Adipocytes, White/ultrastructure , Animals , Caspase Inhibitors/pharmacology , Caspases/chemistry , Caspases/metabolism , Chromatin Assembly and Disassembly/drug effects , Down-Regulation/drug effects , Gene Silencing , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Membrane Potential, Mitochondrial/drug effects , Mice , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Resveratrol , S Phase/drug effects , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Survivin , Up-Regulation/drug effects
10.
Eur J Histochem ; 55(2): e16, 2011 May 24.
Article in English | MEDLINE | ID: mdl-22193296

ABSTRACT

Technological developments based on the use of autologous white adipose tissue (WAT) attracted attention to minor fat depots as possible sources of adipose tissue. In plastic surgery, the trochanteric fatty pad is one of the most used WAT depots for its location and organoleptic characteristics that make it particularly suitable for reconstructive procedures. Despite its wide use in clinic, the structure of this depot has never been studied in detail and it is not known if structural differences exist among trochanteric fat and other subcutaneous WAT depots. The present study was performed on trochanteric fat pad with the aim to clarify the morphology of its adipocytes, stroma and microcirculation, with particular reference to the stem niches. Histological and ultrastructural studies showed that the main peculiar feature of the trochanteric fat concerns its stromal component, which appears less dense than in the other subcutaneous WATs studied. The intra-parenchymal collagen stroma is poor and the extracellular compartment shows large spaces, filled with electron-light material, in which isolated collagen bundles are present. The adipocytes are wrapped in weak and easily detachable collagen baskets. These connective sheaths are very thin compared to the sheaths in other subcutaneous WAT depots. The capillaries are covered by large, long and thin elements surrounded by an external lamina; these perivascular cells are poor in organelles and mainly contain poly-ribosomes. In conclusion, when compared to other WAT deposits, the trochanteric fatty pad shows structural peculiarities in its stroma and microcirculation suggesting a high regenerative potential. Resistance, dissociability, microvascular weft and high regenerative potential make the trochanteric fatty pad a privileged source for harvesting in autologous WAT-based regenerative procedures.


Subject(s)
Adipocytes, White/ultrastructure , Hip , Subcutaneous Fat/ultrastructure , Female , Humans , Middle Aged , Regenerative Medicine
11.
Am J Physiol Endocrinol Metab ; 298(6): E1244-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20354155

ABSTRACT

The origin of brown adipocytes arising in white adipose tissue (WAT) after cold acclimatization is unclear. Here, we demonstrate that several UCP1-immunoreactive brown adipocytes occurring in WAT after cold acclimatization have a mixed morphology (paucilocular adipocytes). These cells also had a mixed mitochondrioma with classic "brown" and "white" mitochondria, suggesting intermediate steps in the process of direct transformation of white into brown adipocytes (transdifferentiation). Quantitative electron microscopy disclosed that cold exposure (6 degrees C for 10 days) did not induce an increase in WAT preadipocytes. beta(3)-adrenoceptor-knockout mice had a blunted brown adipocyte occurrence upon cold acclimatization. Administration of the beta(3)-adrenoceptor agonist CL316,243 induced the occurrence of brown adipocytes, with the typical morphological features found after cold acclimatization. In contrast, administration of the beta(1)-adrenoceptor agonist xamoterol increased only the number of preadipocytes. These findings indicate that transdifferentiation depends on beta(3)-adrenoceptor activation, whereas preadipocyte recruitment is mediated by beta(1)-adrenoceptor. RT-qPCR experiments disclosed that cold exposure induced enhanced expression of the thermogenic genes and of genes expressed selectively in brown adipose tissue (iBAT) and in both interscapular BAT and WAT. beta(3)-adrenoceptor suppression blunted their expression only in WAT. Furthermore, cold acclimatization induced an increased WAT expression of the gene coding for C/EBPalpha (an antimitotic protein), whereas Ccna1 expression (related to cell proliferation) was unchanged. Overall, our data strongly suggest that the cold-induced emergence of brown adipocytes in WAT predominantly reflects beta(3)-adrenoceptor-mediated transdifferentiation.


Subject(s)
Adipocytes, Brown/physiology , Adipocytes, White/physiology , Adipocytes, Brown/cytology , Adipocytes, Brown/ultrastructure , Adipocytes, White/cytology , Adipocytes, White/ultrastructure , Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/physiology , Cell Differentiation/physiology , Cell Transdifferentiation , Cold Temperature , Cyclin A1/genetics , Cyclin A1/physiology , Dioxoles/pharmacology , Female , Immunohistochemistry , Ion Channels/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Mitochondrial Proteins/physiology , RNA/chemistry , RNA/genetics , Receptors, Adrenergic, beta-3/physiology , Reverse Transcriptase Polymerase Chain Reaction , Uncoupling Protein 1
12.
Am J Physiol Endocrinol Metab ; 297(5): E977-86, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19458063

ABSTRACT

Mammals have two types of adipocytes, white and brown, but their anatomy and physiology is different. White adipocytes store lipids, and brown adipocytes burn them to produce heat. Previous descriptions implied their localization in distinct sites, but we demonstrated that they are mixed in many depots, raising the concept of adipose organ. We explain the reason for their cohabitation with the hypothesis of reversible physiological transdifferentiation; they are able to convert one into each other. If needed, the brown component of the organ could increase at the expense of the white component and vice versa. This plasticity is important because the brown phenotype of the organ associates with resistance to obesity and related disorders. Another example of physiological transdifferetiation of adipocytes is offered by the mammary gland; the pregnancy hormonal stimuli seems to trigger a reversible transdifferentiation of adipocytes into milk-secreting epithelial glands. The obese adipose organ is infiltrated by macrophages inducing chronic inflamation that is widely considered as a causative factor for insulin resistance. We showed that the vast majority of macrophages infiltrating the obese organ are arranged around dead adipocytes, forming characteristic crown-like structures. We recently found that visceral fat is more infiltrated than the subcutaneous fat despite a smaller size of visceral adipocytes. This suggests a different susceptibility of visceral and subcutaneous adipocytes to death, raising the concept of smaller critical death size that could be important to explain the key role of visceral fat for the metabolic disorders associated with obesity.


Subject(s)
Adipocytes/physiology , Adipose Tissue/cytology , Adipose Tissue/physiology , Cell Differentiation/physiology , Acclimatization/physiology , Adipocytes, Brown/physiology , Adipocytes, Brown/ultrastructure , Adipocytes, White/physiology , Adipocytes, White/ultrastructure , Animals , Cell Transdifferentiation , Female , Humans , Ion Channels/physiology , Lactation/physiology , Mitochondrial Proteins/physiology , Obesity/pathology , Overweight/pathology , Pregnancy , Pregnancy Complications/metabolism , Temperature , Uncoupling Protein 1
13.
PLoS One ; 4(12): e8458, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20107496

ABSTRACT

BACKGROUND: Brown adipocytes are specialised in dissipating energy through adaptive thermogenesis, whereas white adipocytes are specialised in energy storage. These essentially opposite functions are possible for two reasons relating to mitochondria, namely expression of uncoupling protein 1 (UCP1) and a remarkably higher mitochondrial abundance in brown adipocytes. METHODOLOGY/PRINCIPAL FINDINGS: Here we report a comprehensive characterisation of gene expression linked to mitochondrial DNA replication, transcription and function during white and brown fat cell differentiation in vitro as well as in white and brown fat, brown adipose tissue fractions and in selected adipose tissues during cold exposure. We find a massive induction of the majority of such genes during brown adipocyte differentiation and recruitment, e.g. of the mitochondrial transcription factors A (Tfam) and B2 (Tfb2m), whereas only a subset of the same genes were induced during white adipose conversion. In addition, PR domain containing 16 (PRDM16) was found to be expressed at substantially higher levels in brown compared to white pre-adipocytes and adipocytes. We demonstrate that forced expression of Tfam but not Tfb2m in brown adipocyte precursor cells promotes mitochondrial DNA replication, and that silencing of PRDM16 expression during brown fat cell differentiation blunts mitochondrial biogenesis and expression of brown fat cell markers. CONCLUSIONS/SIGNIFICANCE: Using both in vitro and in vivo model systems of white and brown fat cell differentiation, we report a detailed characterisation of gene expression linked to mitochondrial biogenesis and function. We find significant differences in differentiating white and brown adipocytes, which might explain the notable increase in mitochondrial content observed during brown adipose conversion. In addition, our data support a key role of PRDM16 in triggering brown adipocyte differentiation, including mitochondrial biogenesis and expression of UCP1.


Subject(s)
Adipocytes, Brown/cytology , Cell Differentiation/genetics , Cell Movement/genetics , DNA Replication/genetics , DNA, Mitochondrial/genetics , Gene Expression Regulation , Transcription, Genetic , Adipocytes, Brown/enzymology , Adipocytes, Brown/ultrastructure , Adipocytes, White/cytology , Adipocytes, White/enzymology , Adipocytes, White/ultrastructure , Animals , Cell Nucleus/metabolism , Cold Temperature , DNA/metabolism , DNA-Binding Proteins/metabolism , Gene Dosage , Gene Knockdown Techniques , High Mobility Group Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/ultrastructure , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Protein Binding , Trans-Activators/metabolism , Transcription Factors/metabolism
14.
Histol Histopathol ; 22(2): 199-210, 2007 02.
Article in English | MEDLINE | ID: mdl-17149693

ABSTRACT

Leptin is a hormone that plays a central role in the regulation of food intake and energy expenditure. Originally discovered in mature white adipocytes, it was subsequently isolated from the gastric mucosa. This tissue contains a large number of epithelial endocrine and exocrine cells secreting leptin in the blood stream and in the gastric lumen, respectively. Light and electron microscopy have shown that adipocytes and gastric epithelial cells contain leptin along their rough endoplasmic reticulum-Golgi-granules secretory pathway. Both tissues synthesize a soluble form of the leptin receptor that is secreted bound to leptin in the blood and into the gastric juice. This soluble receptor protect leptin and enhances its half-life. Despite the similarities in the mechanisms of leptin secretion by adipocytes and gastric epithelial cells, they are in fact radically different. In gastric cells leptin follows a rapid regulated secretion pathway whereas adipocytes secrete leptin in a constitutive slow fashion. These differences can be explained by the specific roles play by leptin originating from these two different tissues. Gastric leptin is involved in the short-term regulation of digestion, including delay of gastric emptying, absorption of nutrients by the intestinal wall and secretion of gastric, intestinal and pancreatic hormones. On the other hand, leptin secreted by white adipocytes acts primarily on the hypothalamus for the long-term regulation of food intake. Therefore, the coordination of adipose and gastric leptins ensures the proper management of food processing and energy storage.


Subject(s)
Adipocytes, White/metabolism , Adipose Tissue, White/metabolism , Gastric Mucosa/metabolism , Leptin/metabolism , Adipocytes, White/ultrastructure , Adipose Tissue, White/cytology , Animals , Humans
15.
Endocrinology ; 148(3): 1009-18, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17138655

ABSTRACT

Whereas the uptake of oxidized lipoproteins by scavenger receptor CD36 in macrophages has been associated with foam cell formation and atherogenesis, little is known about the role of CD36 in regulating lipid metabolism in adipocytes. Here we report that treatment of 3T3-L1 adipocytes with hexarelin, a GH-releasing peptide that interacts with CD36, resulted in a depletion of intracellular lipid content with no significant change in CD36 expression. Microarray analysis revealed an increased pattern in several genes involved in fatty acid mobilization toward the mitochondrial oxidative phosphorylation process in response to hexarelin. Interestingly, many of these up-regulated genes are known targets of peroxisomal proliferator-activated receptor (PPAR)-gamma, such as FATP, CPT-1, and F(1)-ATPase, suggesting that adipocyte response to hexarelin may involve PPARgamma activation. Expression studies also indicate an increase in thermogenic markers PPARgamma coactivator 1alpha and uncoupling protein-1, which are normally expressed in brown adipocytes. Electron microscopy of hexarelin-treated 3T3-L1 adipocytes showed an intense and highly organized cristae formation that spans the entire width of mitochondria, compared with untreated cells, and cytochrome c oxidase activity was enhanced by hexarelin, two features characteristic of highly oxidative tissues. A similar mitochondrial phenotype was detected in epididymal white fat of mice treated with hexarelin, along with an increased expression of thermogenic markers that was lost in treated CD36-null mice, suggesting that the ability of hexarelin to promote a brown fat-like phenotype also occurs in vivo and is dependent on CD36. These results provide a potential role for CD36 to impact the overall metabolic activity of fat usage and mitochondrial biogenesis in adipocytes.


Subject(s)
Adipocytes, White/drug effects , CD36 Antigens/physiology , Mitochondria/drug effects , Oligopeptides/pharmacology , 3T3-L1 Cells , Adipocytes, White/chemistry , Adipocytes, White/metabolism , Adipocytes, White/ultrastructure , Animals , CD36 Antigens/genetics , Electron Transport Complex IV/metabolism , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Lipids/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/physiology , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction/drug effects , Receptors, Scavenger/physiology , Thermogenesis/drug effects , Thermogenesis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...