Skip to main content

Assays for Insulin and Insulin-Like Activity Based on Adipocytes

  • Living reference work entry
  • First Online:
Drug Discovery and Evaluation: Pharmacological Assays
  • 726 Accesses

Abstract

Data from the metabolic assays (and signaling assays; see below) are calculated as stimulation factor above basal activity (absence of insulin/compound/drug candidate) for processes stimulated (e.g., lipogenesis, glucose transport, and GLUT4 translocation) or as difference between the basal and insulin/compound/drug candidate-induced values for processes downregulated (e.g., lipolysis). In each case, these data, which reflect the responsiveness of the metabolic effector system studied toward the respective stimulus (insulin/compound/drug candidate), are normalized to the basal (set at 0 %) and maximal insulin action (set at 100 %; elicited by maximally effective concentration of insulin). For characterization of the sensitivity of the metabolic effector system toward the respective stimulus, effective concentrations for the induction of 150 % (or higher) of the basal activity (set at 100 %) can be given. These so-called EC150-values facilitate the insulin-independent comparison of the relative potency of the insulin-like activity between compounds/drug candidates, in general, and in particular for those frequently observed stimuli, which do not elicit the same maximal response in % stimulation or inhibition and/or fail to approach the maximal insulin response.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Institutional subscriptions

References and Further Reading

Method Using CHO/L6 GLUT4myc-HIR cells

  • Basi NS, Thomaskutti KG, Pointer RH (1992) Regulation of glucose transport in isolated adipocytes by levamisole. Can J Physiol Pharmacol 70:1190–1194

    CAS  PubMed  Google Scholar 

  • Drevon CA (2005) Fatty acids and expression of adipokines. Biochim Biophys Acta 1740:287–292

    CAS  PubMed  Google Scholar 

  • Elsing C, Winn Borner U, Stremmel W (1995) Confocal analysis of hepatocellular long-chain fatty acid uptake. Am J Physiol 269:G842–G851

    CAS  PubMed  Google Scholar 

  • Farese RV, Standaert ML, Yamada K, Huang LC, Zhang C, Cooper DR, Wang Z, Yang Y, Suzuki S, Toyota T, Larner J (1994) Insulin-induced activation of glycerol-3-phosphate acyltransferase by chiro-inositol-containing insulin mediator is defective in adipocytes of insulin-resistant, type II diabetic, Goto–Kakizaki rats. Proc Natl Acad Sci U S A 91:11040–11044

    CAS  PubMed Central  PubMed  Google Scholar 

  • Foley JE, Gliemann J (1981) Accumulation of 2-deoxyglucose against its concentration gradient in rat adipocytes. Biochim Biophys Acta 648:100–106

    CAS  PubMed  Google Scholar 

  • Foley JE, Cushman SW, Salans LB (1978) Glucose transport in isolated rat adipocytes with measurement of L-arabinose uptake. Am J Physiol 234:E112–E119

    CAS  PubMed  Google Scholar 

  • Frost SC, Lane MD (1985) Evidence for the involvement of vicinal sulfhydryl groups in insulin-activated hexose transport by 3T3-L1 adipocytes. J Biol Chem 260:2646–2652

    CAS  PubMed  Google Scholar 

  • Gliemann J, Østerlind K, Vinten J, Gammeltoft S (1972) A procedure for measurement of distribution spaces in isolated fat cells. Biochim Biophys Acta 286:1–9

    CAS  PubMed  Google Scholar 

  • Humbel RE (1959) Messung der Serum–Insulin-Aktivität mit epididymalem Fettgewebe in vitro. Experientia (Basel) 15:256–258

    CAS  Google Scholar 

  • Karnieli E, Zarnowski MJ, Hissin PJ, Simpson IA, Salans LB, Cushman SW (1981) Insulin-stimulated translocation of glucose transport systems in the isolated rat adipose cell. J Biol Chem 256:4772–4777

    CAS  PubMed  Google Scholar 

  • Lee Y-H, Chen S-Y, Wiesner RJ, Huang Y-F (2004) Simple flow cytometric method used to assess lipid accumulation in fat cells. J Lipid Res 45:1162–1167

    CAS  PubMed  Google Scholar 

  • Lingsøe J (1961) Determination of the insulin-like activity in serum using rat epididymal adipose tissue. Scand J Clin Lab Invest 13:628–636

    Google Scholar 

  • Manchester JK, Chi MM, Carter JG, Pusateri ME, McDougal DB, Lowry OH (1990) Measurement of 2-deoxyglucose and 2-deoxyglucose 6-phosphate in tissues. Anal Biochem 185:118–124

    CAS  PubMed  Google Scholar 

  • Moody AJ, Stan MA, Stan M (1974) A simple free fat cell bioassay for insulin. Horm Metab Res 6:12–16

    CAS  PubMed  Google Scholar 

  • Müller G, Jordan H, Petry S, Wetekam E-M, Schindler P (1997a) Analysis of lipid metabolism in adipocytes using fluorescent fatty acids I. Insulin stimulation of lipogenesis. Biochim Biophys Acta 1347:23–39

    PubMed  Google Scholar 

  • Perret P, Ghezzi C, Ogier L, Abbadi M, Morin C, Mathieu JP, Fagret D (2004) Biological studies of radiolabeled glucose analogs iodinated in positions 3, 4 or 6. Nucl Med Biol 31:241–250

    CAS  PubMed  Google Scholar 

  • Rajala MW, Scherer PE (2003) Minireview. The adipocyte-at the crossroads of energy homeostasis, inflammation, and atherosclerosis. Endocrinology 144:3765–3773

    CAS  PubMed  Google Scholar 

  • Sasson S, Oron R, Cerasi E (1993) Enzymatic assay for 2-deoxyglucose 6-phosphate for assessing hexose uptake rates in cultured cells. Anal Biochem 215:309–311

    CAS  PubMed  Google Scholar 

  • Schaffer JE, Lodish HF (1994) Expression cloning and characterization of a novel adipocyte long chain fatty acid transport protein. Cell 97:427–436

    Google Scholar 

  • Shigematsu S, Miller SL, Pessin JE (2001) Differentiated 3T3L1 adipocytes are composed of heterogenous cell population with distinct receptor tyrosine kinase signaling properties. J Biol Chem 276:15292–15297

    CAS  PubMed  Google Scholar 

  • Sooranna SR, Saggerson ED (1976a) Interactions of insulin and adrenaline with glycerol phosphate acylation processes in fat-cells from rat. FEBS Lett 64:36–39

    CAS  PubMed  Google Scholar 

  • Still MC, Khan M, Mitra A (1978) J Org Chem 43:2923–2925

    CAS  Google Scholar 

  • Storch J, Lechene C, Kleinfeld AM (1995) Direct determination of free fatty acid transport across the adipocyte plasma membrane using quantitative fluorescence microscopy. J Biol Chem 266:13473–13476

    Google Scholar 

  • Vila M, Farese RV (1991) Insulin rapidly increases glycerol-3-phosphate-acyltransferase activity in rat adipocytes. Arch Biochem Biophys 284:366–368

    CAS  PubMed  Google Scholar 

  • Von Goor H, Gerrits PO, Groud J (1986) The application of lipid-soluble stains in plastic-embedded sections. Histochemistry 85:251–253

    PubMed  Google Scholar 

  • Whitesell RR, Gliemann J (1979) Kinetic parameters of 3-O-methylglucose and glucose in adipocytes. J Biol Chem 254:5276–5283

    CAS  PubMed  Google Scholar 

  • Yamamoto N, Sato T, Kawasaki K, Murosaki S, Yamamoto Y (2006) A nonradioisotope, enzymatic assay for 2-deoxyglucose uptake in L6 skeletal muscle cells cultured in a 96-well microplate. Anal Biochem 351:139–145

    CAS  PubMed  Google Scholar 

  • Zou C, Wang Y, Shen Z (2005) NBDG as a fluorescent indicator for direct glucose uptake measurement. J Biochem Biophys Methods 64:207–215

    CAS  PubMed  Google Scholar 

Analysis by Fluorescent Resonance Energy Transfer (FRET)

  • Abe H, Morimatsu M, Nikami H, Miyashige T, Saito M (1997) Molecular cloning and mRNA expression of the bovine insulin-responsive glucose transporter GLUT4. J Anim Sci 75:182–188

    CAS  PubMed  Google Scholar 

  • Assimacopoulos-Jeannet F, Cusin I, Greco-Perotto RM, Terrettaz J, Rohner-Jeanrenaud F, Zarjevski N, Jeanrenaud B (1991) Glucose transporters: structure, function, and regulation. Biochemie 73:67–70

    CAS  Google Scholar 

  • Bähr M, von Holtey M, Müller G, Eckel J (1995) Direct stimulation of myocardial glucose transport and glucose transporter-1 (GLUT1) and GLUT4 protein expression by the sulfonylurea glimepiride. Endocrinology 136:2547–2553

    Google Scholar 

  • Begum N, Draznin B (1992) The effect of streptozotocin-induced diabetes on GLUT-4 phosphorylation in rat adipocytes. J Clin Invest 90:1254–1262

    CAS  PubMed Central  PubMed  Google Scholar 

  • Cusin I, Terrettaz J, Rohner-Jeanrenaud F, Zarjevski N, Assimacopoulos-Jeannet F, Jeanrenaud B (1990) Hyperinsulinemia increases the amount of GLUT4 mRNA in white adipose tissue and decreases that of muscles: a clue for increased fat depot and insulin resistance. Endocrinology 127:3246–3248

    CAS  PubMed  Google Scholar 

  • Dalen KT, Ulven SM, Bamberg K, Gustafsson J (2003) Expression of the insulin-responsive glucose transporter GLUT4 in adipocytes is dependent in liver X receptor α. J Biol Chem 278:48283–48291

    CAS  PubMed  Google Scholar 

  • Fingar DC, Hausdorff SF, Blenis J, Birnbaum MJ (1993) Dissociation of pp 70 ribosomal protein S6 kinase from insulin-stimulated glucose transport in 3T3-L1 adipocytes. J Biol Chem 268:3005–3008

    CAS  PubMed  Google Scholar 

  • Galante P, Maerker E, Scholz R, Rett K, Herberg L, Mosthaf L, Häring HU (1994) Insulin-induced translocation of GLUT4 in skeletal muscle of insulin-resistant Zucker rats. Diabetologia 37:3–9

    CAS  PubMed  Google Scholar 

  • Garcia de Herreros A, Birnbaum MJ (1989) The acquisition of increased insulin-responsive hexose transport in 3T3-L1 adipocytes correlates with expression of a novel transporter proteins. J Biol Chem 264:19994–19999

    CAS  PubMed  Google Scholar 

  • Gould GW, Holman GD (1993) The glucose transporter family: structure, function and tissue-specific expression. Biochem J 295:329–341

    CAS  PubMed Central  PubMed  Google Scholar 

  • Hashimoto M, Yang J, Holman GD (2001) Cell-surface recognition of biotinylated membrane proteins requires very long spacer arms: an example from glucose-transporter arms. Chembiochem 2:52–59

    CAS  PubMed  Google Scholar 

  • Hofmann C, Lorenz K, Colca JR (1991) Glucose transport deficiency in diabetic animals is corrected by treatment with the oral antihyperglycemic agent pioglitazone. Endocrinology 129:1915–1925

    CAS  PubMed  Google Scholar 

  • Holman GD, Karim AR, Karim B (1988) Photolabeling of erythrocyte and adipocyte hexose transporters using a benzophone derivative of bis(D-mannose). Biochim Biophys Acta 946:75–84

    CAS  PubMed  Google Scholar 

  • Holman GD, Kozka IJ, Clark AE, Flower CJ, Saltis J, Habberfield AD, Simpson IA, Cushman SW (1990) Cell surface labeling of glucose transporter isoform GLUT4 by bismannose photolabel. Correlation with stimulation of glucose transport in rat adipose cells by insulin and phorbol ester. J Biol Chem 265:18172–18179

    CAS  PubMed  Google Scholar 

  • Holman GD, Parkar BA, Midgley PJW (1986) Exofacial photoaffinity labelling of the human erythrocyte sugar transporter. Biochim Biophys Acta 855:115–126

    CAS  PubMed  Google Scholar 

  • Inoue G, Cheatham B, Kahn CR (1999) Development of an in vitro reconstitution assay for glucose transporter 4 translocation. Proc Natl Acad Sci U S A 96:14919–14924

    CAS  PubMed Central  PubMed  Google Scholar 

  • Jacobs DB, Jung CY (1985) Sulfonylurea potentiates insulin-induced recruitment of glucose transport carrier in rat adipocytes. J Biol Chem 260:2593–2596

    CAS  PubMed  Google Scholar 

  • Jacobs DB, Hayes GR, Lockwood DH (1989) In vitro effect of sulfonylurea on glucose transport and translocation of glucose transporters in adipocytes from streptozotocin-induced diabetic rats. Diabetes 38:205–211

    CAS  PubMed  Google Scholar 

  • James DE, Strube M, Mueckler M (1989) Molecular cloning and characterization of an insulin-regulatable glucose transporter. Nature 338:83–87

    CAS  PubMed  Google Scholar 

  • Kanai F, Nishioka Y, Hayashi H, Kamohara S, Todaka M, Ebina Y (1993) Direct demonstration of insulin-induced GLUT4 translocation to the surface of intact cells by insertion of a c-myc epitope into an exofacial GLUT4 domain. J Biol Chem 268:14523–14526

    CAS  PubMed  Google Scholar 

  • Kanda H, Tamori Y, Shinoda H, Yoshikawa M, Sakaue M, Udagawa J, Otani H, Tashiro F, Miyazaki JI, Kasuga M (2005) Adipocytes from Munc18c-null mice show increased sensitivity to insulin-stimulated GLUT4 externalization. J Clin Invest 115:291–301

    CAS  PubMed Central  PubMed  Google Scholar 

  • Kono T (1983) Recycling of insulin-sensitive glucose transporter in rat adipocytes. Methods Enzymol 98:431–444

    CAS  PubMed  Google Scholar 

  • Koumanov F, Jin B, Yang J, Holman GD (2005) Insulin signaling meets vesicle traffic of GLUT4 at a plasma-membrane activated fusion step. Cell Metab 3:179–189

    Google Scholar 

  • Koumanov F, Yang, Jones AE, Hatanaka Y, Holman GD (1998) Cell-surface biotinylation of GLUT4 using bis-mannose photolabels. Biochem J 330:1209–1215

    CAS  PubMed Central  PubMed  Google Scholar 

  • Kozka IJ, Clark AE, Holman GD (1991) Chronic treatment with insulin selectively down-regulates cell-surface GLUT4 glucose transporters in 3T3-L1 adipocytes. J Biol Chem 266:11726–11731

    CAS  PubMed  Google Scholar 

  • Laurie SM, Cain CC, Lienhard GE, Castle JD (1993) The glucose transporter GLUT4 and secretory membrane proteins (SCAMPs) colocalize in rat adipocytes and partially segregate during insulin stimulation. J Biol Chem 268:19110–19117

    CAS  PubMed  Google Scholar 

  • Lawrence JC, Hiken JF, James DE (1990a) Stimulation of glucose transport and glucose transporter phosphorylation by okadaic acid in rat adipocytes. J Biol Chem 265:19768–19776

    CAS  PubMed  Google Scholar 

  • Lawrence JC, Hiken JF, James DE (1990b) Phosphorylation of the glucose transporter in rat adipocytes. Identification of the intracellular domain at the carboxyl terminus as a target for phosphorylation in intact cells and in vitro. J Biol Chem 265:2324–2332

    CAS  PubMed  Google Scholar 

  • Li W-M, McNeill JH (1997) Quantitative methods for measuring the insulin-regulatable glucose transporter (Glut4). J Pharmacol Toxicol Methods 38:1–10

    CAS  PubMed  Google Scholar 

  • Lizunov VA, Matsumoto H, Zimmerberg J, Cushman SW, Frolov VA (2005) Insulin stimulates the halting, tethering, and fusion of mobile GLUT4 vesicles in rat adipose cells. J Cell Biol 169:481–489

    CAS  PubMed Central  PubMed  Google Scholar 

  • Matthei S, Hamann A, Klein HH, Benecke H, Kreymann G, Flier JS, Greten H (1991) Association of metformin’s effect to increase insulin-stimulated glucose transport with potentiation of insulin-induced translocation of glucose transporters from intracellular pool to plasma membrane in rat adipocytes. Diabetes 40:850–857

    Google Scholar 

  • Matthei S, Trost B, Hammann A, Kausch C, Benecke H, Greten H, Höppner W, Klein HH (1995) The effect of in vivo thyroid hormone status on insulin signalling and GLUT1 and GLUT4 glucose transport systems in rat adipocytes. J Endocrinol 144:347–357

    Google Scholar 

  • McKeel DW, Jarett L (1970) Preparation and characterization of a plasma membrane fraction from isolated fat cells. J Cell Biol 44:417–432

    CAS  PubMed Central  PubMed  Google Scholar 

  • Moore MS, Mahafferey DT, Brodsky FM, Anderson RGW (1987) Assembly of clathrin-coated pits onto purified plasma membranes. Science 236:558–563

    CAS  PubMed  Google Scholar 

  • Mueckler M (1994) Facilitative glucose transporters. Eur J Biochem 219:713–725

    CAS  PubMed  Google Scholar 

  • Müller G, Wied S (1993) The sulfonylurea drug, glimepiride, stimulates glucose transport, glucose transporter translocation, and dephosphorylation in insulin-resistant rat adipocytes in vitro. Diabetes 42:1852–1867

    PubMed  Google Scholar 

  • Niwa H, Yamamura K, Miyazaki J (1991) Efficient selection for high-expression transfectants with a novel eukaryotic vector. Gene 108:193–199

    CAS  PubMed  Google Scholar 

  • Rampal AL, Jhun BH, Kim S, Liu H, Manka M, Lachaal M, Spangler RA, Jung CY (1995) Okadaic acid stimulates glucose transport in rat adipocytes by increasing the externalization rate constant of GLUT4 recycling. J Biol Chem 270:3938–3943

    CAS  PubMed  Google Scholar 

  • Ren JM, Marshall BA, Mueckler MM, McCaleb M, Amatruda JM, Shulman GI (1995) Overexpression of Glut4 protein in muscle increases basal and insulin-stimulated whole body glucose disposal in conscious mice. J Clin Invest 95:429–432

    CAS  PubMed Central  PubMed  Google Scholar 

  • Reusch JEB, Sussman KE, Draznin B (1993) Inverse relationship between GLUT-4 phosphorylation and its intrinsic activity. J Biol Chem 268:3348–3351

    CAS  PubMed  Google Scholar 

  • Robinson LJ, Pang S, Harris DS, Heuser J, James DE (1992) Translocation of the glucose transporter (GLUT4) to the cell surface in permeabilized 3T3-L1 adipocytes: effects of ATP, insulin, and GTP gamma S and localization of GLUT4 to clathrin lattices. J Cell Biol 117:1181–1196

    CAS  PubMed  Google Scholar 

  • Ryder JW, Yang J, Galuska D, Rincon J, Björnholm M, Krook A, Lund S, Pedersen O, Wallberg-Henriksson H, Zierath JR, Holman GD (2000) Diabetes 49:647–654

    CAS  PubMed  Google Scholar 

  • Satoh S, Nishimura H, Clark AE, Kozka IJ, Vannucci SJ, Simpson IA, Quon MJ, Cushman SW, Holman GD (1993) Use of bis-Mannose photolabel to elucidate insulin-regulated GLUT4 subcellular trafficking kinetics in rat adipose cells: evidence that exocytosis is a critical site of hormone action. J Biol Chem 268:17820–17829

    CAS  PubMed  Google Scholar 

  • Schürmann A, Rosenthal W, Hinsch KD, Joost HG (1989) Differential sensitivity to guanine nucleotides of basal and insulin-stimulated glucose transporter activity reconstituted from adipocyte membrane fractions. FEBS Lett 255:259–264

    PubMed  Google Scholar 

  • Simpson IA, Yver DR, Hissin PJ, Wardzala LJ, Karnieli E, Salans LB, Cushman SW (1983) Insulin-stimulated translocation of glucose transporters in the isolated rat adipose tissue cells: characterization of subcellular fractions. Biochim Biophys Acta 763:393–407

    CAS  PubMed  Google Scholar 

  • Smith RM, Charron MJ, Shah N, Lodish HF, Jarett L (1991) Immunoelectron microscopic demonstration of insulin-stimulated translocation of glucose transporters to the plasma membrane of isolated rat adipocytes and masking of the carboxyl-terminal epitope of intracellular GLUT4. Proc Natl Acad Sci U S A 88:6893–6897

    CAS  PubMed Central  PubMed  Google Scholar 

  • Terasaki J, Anai M, Funaki M, Shibata T, Inukai K, Ogihara T, Ishihara H, Katagiri H, Onishi Y, Sadoka H, Fukushima Y, Yataki Y, Kikuchi M, Oka Y, Asana T (1998) Role of JTT-501, a new insulin sensitizer, in restoring impaired GLUT4 translocation in adipocytes of rats fed a high fat diet. Diabetologia 41:400–409

    CAS  PubMed  Google Scholar 

  • Towbin H, Staehelin T, Gordon J (1979) Electrophoretic transfer from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc Natl Acad Sci U S A 76:4350–4354

    CAS  PubMed Central  PubMed  Google Scholar 

  • Uphues I, Kolter T, Eckel J (1995) Failure of insulin-regulated recruitment of the glucose transporter GLUT4 in cardiac muscle of obese Zucker rats is associated with alterations of small-molecular-mass GTP-binding proteins. Biochem J 311:161–166

    CAS  PubMed Central  PubMed  Google Scholar 

  • van Dam EM, Govers R, James DE (2005) Akt activation is required at a late stage of insulin-induced GLUT4 translocation to the plasma membrane. Mol Endocrinol 19:1067–1077

    PubMed  Google Scholar 

  • Vannucci SJ, Nishimura H, Satoh S, Cushman SW, Holman GD, Simpson IA (1992) Cell surface accessibility of GLUT4 glucose transporters in insulin-stimulated rat adipose cells. Modulation by isoprenaline and adenosine. Biochem J 288:325–330

    CAS  PubMed Central  PubMed  Google Scholar 

  • Wang Q, Khayat Z, Kishi K, Ebina Y, Klip A (1998a) GLUT4 translocation by insulin in intact muscle cells: detection by a fast and quantitative assay. FEBS Lett 427:193–197

    CAS  PubMed  Google Scholar 

  • Wilson CM, Cushman SW (1994) Insulin stimulation of glucose transport activity in rat skeletal muscle: increase in cell surface GLUT4 as assessed by photolabeling. Biochem J 299:755–759

    CAS  PubMed Central  PubMed  Google Scholar 

  • Yang J, Holman GD (1993) Comparison of GLUT4 and GLUT1 subcellular trafficking in basal and insulin-stimulated 3T3-L1 cells. J Biol Chem 268:4600–4603

    CAS  PubMed  Google Scholar 

  • Zeller K, Vogel J, Rahner-Welsch S, Kuschinsky W (1995) GLUT1 distribution in adult rat brains. Pflügers Arch Eur J Physiol 429:R63/201

    Google Scholar 

Method Based on the Confocal Image Analysis of GLUT4 in Recombinant Rat MyoblastsReading

  • Baus D, Heermeier K, De Hoop M, Metz-Weidmann C, Gassenhuber J, Dittrich W, Welte S, Tennagels N (2008) Identification of a novel AS160 splice variant that regulates GLUT4 translocation and glucose uptake in rat muscle cells. Cell Signal 20:2237–2246

    CAS  PubMed  Google Scholar 

  • Breen DM, Sanli T, Giacca A, Tsiani E (2008) Stimulation of muscle cell glucose uptake by resveratrol through sirtuins and AMPK. Biochem Biophys Res Commun 374:117–122

    CAS  PubMed  Google Scholar 

  • Brozinick JT Jr, Hawkins ED, Strawbridge AB, Elmendorf JS (2004) Disruption of cortical actin in skeletal muscle demonstrates an essential role of the cytoskeleton in glucose transporter 4 translocation in insulin-sensitive tissues. J Biol Chem 279:40699–40706

    CAS  PubMed Central  PubMed  Google Scholar 

  • Bruss MD, Arias EB, Lienhard GE, Cartee GD (2005) Increased phosphorylation of Akt substrate of 160 kDa (AS160) in rat skeletal muscle in response to insulin or contractile activity. Diabetes 54:41–50

    CAS  PubMed  Google Scholar 

  • Deshmukh A, Coffey VG, Zhong Z, Chibalin AV, Hawley JA, Zierath JR (2006) Exercise-induced phosphorylation of the novel Akt substrates AS160 and filamin A in human skeletal muscle. Diabetes 55:1776–1782

    CAS  PubMed  Google Scholar 

  • Hill MM, Clark SF, Tucker DF, Birnbaum MJ, James DE, Macaulay SL (1999) A role for protein kinase B beta/Akt2 in insulin-stimulated GLUT4 translocation in adipocytes. Mol Cell Biol 19:7771–7781

    CAS  PubMed Central  PubMed  Google Scholar 

  • Kane S, Sano H, Liu SC, Asara JM, Lane WS, Garner CC, Lienhard GE (2002) A method to identify serine kinase substrates: Akt phosphorylates a novel adipocyte protein with a Rab GTPase-activating protein (GAP) domain. J Biol Chem 277:22115–22118

    CAS  PubMed  Google Scholar 

  • Karlsson HK, Zierath JR, Kane S, Krook A, Lienhard GE, Wallberg-Henriksson H (2005) Insulin-stimulated phosphorylation of the Akt substrate AS160 is impaired in skeletal muscle of type 2 diabetic subjects. Diabetes 54:1692–1697

    CAS  PubMed  Google Scholar 

  • Kurtzhals P, Schaffer L, Sorensen A, Kristensen C, Jonassen I, Schmid C, Trub T (2000) Correlations of receptor binding and metabolic and mitogenic potencies of insulin analogs designed for clinical use. Diabetes 49:999–1005

    CAS  PubMed  Google Scholar 

  • Laborde E, Manchem VP (2002) Small molecule activators of the insulin receptor: potential new therapeutic agents for the treatment of diabetes mellitus. Curr Med Chem 9:2231–2242

    CAS  PubMed  Google Scholar 

  • Liu F, Dallas-Yang Q, Castriota G, Fischer P, Santini F, Ferrer M et al (2009) Development of a novel GLUT4 translocation assay for identifying potential novel therapeutic targets for insulin sensitization. Biochem J 418:413–420

    CAS  PubMed  Google Scholar 

  • Sano H, Kane S, Sano E, Miinea CP, Asara JM, Lane WS, Garner CW, Lienhard GE (2003) Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation. J Biol Chem 278:14599–14602

    CAS  PubMed  Google Scholar 

  • Treebak JT, Glund S, Deshmukh A, Klein DK, Long YC, Jensen TE, Jorgensen SB, Viollet B, Andersson L, Neumann D, Wallimann T, Richter EA, Chibalin AV, Zierath JR, Wojtaszewski JF (2006) AMPK-mediated AS160 phosphorylation in skeletal muscle is dependent on AMPK catalytic and regulatory subunits. Diabetes 55:2051–2058

    CAS  PubMed  Google Scholar 

  • Wada T, Azegami M, Sugiyama M, Tsuneki H, Sasaoka T (2008) Characteristics of signalling properties mediated by long-acting insulin analogue glargine and detemir in target cells of insulin. Diabetes Res Clin Pract 81:269–277

    CAS  PubMed  Google Scholar 

  • Wang Q, Khayat Z, Kishi K, Ebina Y, Klip A (1998b) GLUT4 translocation by insulin in intact muscle cells: detection by a fast and quantitative assay. FEBS Lett 427:193–197

    CAS  PubMed  Google Scholar 

  • Watson RT, Pessin JE (2006) Bridging the GAP between insulin signaling and GLUT4 translocation. Trends Biochem Sci 31:215–222

    CAS  PubMed  Google Scholar 

  • Webster NJ, Park K, Pirrung MC (2003) Signaling effects of demethylasterriquinone B1, a selective insulin receptor modulator. Chembiochem 4:379–385

    CAS  PubMed  Google Scholar 

  • Yamaguchi S, Katahira H, Ozawa S, Nakamichi Y, Tanaka T, Shimoyama T, Takahashi K, Yoshimoto K, Imaizumi MO, Nagamatsu S, Ishida H (2005) Activators of AMP-activated protein kinase enhance GLUT4 translocation and its glucose transport activity in 3T3-L1 adipocytes. Am J Physiol Endocrinol Metab 289:E643–E649

    CAS  PubMed  Google Scholar 

  • Zhang B, Salituro G, Szalkowski D, Li Z, Zhang Y, Royo I, Vilella D, Diez MT, Pelaez F et al (1999) Discovery of a small molecule insulin mimetic with antidiabetic activity in mice. Science 284:974–977

    CAS  PubMed  Google Scholar 

Method Based on Fluorescent Fatty Acids

  • Chang BH-J, Li L, Paul A, Taniguchi S, Nannegari V, Heird WC, Chan L (2006) Protection against fatty liver but normal adipogenesis in mice lacking adipose differentiation-related protein. Mol Cell Biol 26:1063–1076

    CAS  PubMed Central  PubMed  Google Scholar 

  • DiRusso CC, Connell EJ, Faergeman NJ, Knudsen J, Hansen JK, Black PN (2000) Murine FATP alleviates growth and biochemical deficiencies of yeast fat1Delta strains. Eur J Biochem 267:4422–4433

    CAS  PubMed  Google Scholar 

  • Faergeman NJ, Black PN, Zhao XD, Knudsen J, DiRusso CC (2001) The Acyl-CoA synthetases encoded within FAA1 and FAA4 in Saccharomyces cerevisiae function as components of the fatty acid transport system linking import, activation, and intracellular utilization. J Biol Chem 276:37051–37059

    CAS  PubMed  Google Scholar 

  • Faergeman NJ, DiRusso CC, Elberger A, Knudsen J, Black PN (1997) Disruption of the Saccharomyces cerevisiae homologue to the murine fatty acid transport protein impairs uptake and growth on long-chain fatty acids. J Biol Chem 272:8531–8538

    CAS  PubMed  Google Scholar 

  • Fernandes PB (1998) Technological advances in high-throughput screening. Curr Opin Chem Biol 2:597–603

    CAS  PubMed  Google Scholar 

  • Johnson DR, Knoll LJ, Levin DE, Gordon JI (1994) Saccharomyces cerevisiae contains four fatty acid activation (FAA) genes: an assessment of their role in regulating protein N-myristoylation and cellular lipid metabolism. J Cell Biol 127:751–762

    CAS  PubMed  Google Scholar 

  • Kazantzis M, Stahl A (2012) Fatty acid transport proteins, implications in physiology and disease. Biochim Biophys Acta 1821:852–857

    Google Scholar 

  • Li H, Black PN, DiRusso CC (2005) A live-cell high-throughput screening assay for identification of fatty acid uptake inhibitors. Anal Biochem 336:11–19

    CAS  PubMed  Google Scholar 

  • Pohl J, Ring A, Korkmaz Ü, Ehehalt R, Stremmel W (2005) FAT/CD36-mediated long-chain fatty acid uptake in adipocytes requires plasma membrane rafts. Mol Biol Cell 16:24–31

    CAS  PubMed Central  PubMed  Google Scholar 

  • Stremmel W, Strohmeyer G, Berk PD (1986) Hepatocellular uptake of oleate is energy dependent, sodium linked, and inhibited by an antibody to a hepatocyte plasma membrane fatty acid binding protein. Proc Natl Acad Sci U S A 83:3584–3588

    CAS  PubMed Central  PubMed  Google Scholar 

Analysis of Lipolysis Products

  • Agmon V, Cherbu S, Dagan A, Grace M, Grabowski GA, Gatt S (1993) Synthesis and use of novel fluorescent glycosphingolipids for estimating beta-glucosidase activity in vitro in the absence of detergents and subtyping Gaucher disease variants following administration into intact cells. Biochim Biophys Acta 1170:72–79

    CAS  PubMed  Google Scholar 

  • Badellino K, Jin W, Rader DJ (2004) Endothelial lipase: a novel drug target for HDL and atherosclerosis? In: Müller G, Petry S (eds) Lipases and phospholipases in drug development. Wiley-VCH, Weinheim, pp 139–154

    Google Scholar 

  • Beisson F, Ferte N, Nari J, Noat G, Arondel V, Verger R (1999) Use of naturally fluorescent triacylglycerols from Parinari glaberrimum to detect low lipase activities from Arabidopsis thaliana seedlings. J Lipid Res 40:2313–2321

    CAS  PubMed  Google Scholar 

  • Beisson F, Tiss A, Riviere C, Verger R (2000) Methods for lipase detection and assay: a critical review. Eur J Lipid Sci Technol 1:133–153

    Google Scholar 

  • Bell RM, Coleman RA (1980) Enzymes of glycerolipid synthesis in eukaryotes. Annu Rev Biochem 49:459–487

    CAS  PubMed  Google Scholar 

  • Ben Ali Y, Carriere F, Verger R, Petry S, Müller G, Abousalham A (2005) Continuous monitoring of cholesterol oleate hydrolysis by hormone-sensitive lipase and other cholesterol esterases. J Lipid Res 46:994–1000

    CAS  Google Scholar 

  • Ben Ali Y, Chahinian H, Petry S, Müller G, Carriere F, Verger R, Abousalham A (2004) Might the kinetic behavior of hormone-sensitive lipase reflect the absence of the lid domain? Biochemistry 43:9298–9306

    Google Scholar 

  • Blanchette-Mackie EJ, Dwyer NK, Barber T, Coxey RA, Takeda T, Rondinone CM, Theodorakis JL, Greenberg AS, Londos C (1995) Perilipin is located on the surface layer of intracellular lipid droplets in adipocytes. J Lipid Res 36:1211–1226

    CAS  PubMed  Google Scholar 

  • Brasaemle DL, Barber T, Wolins NE, Serrero G, Blanchette-Mackie EJ, Londos C (1997) Adipose differentiation-related protein is an ubiquitously expressed lipid storage droplet-associated protein. J Lipid Res 38:2249–2263

    CAS  PubMed  Google Scholar 

  • Brasaemle DL, Dolios G, Shapiro L, Wang R (2004) Proteomic analysis of proteins associated with lipid droplets of basal and lipolytically stimulated 3T3-L1 adipocytes. J Biol Chem 279:46835–46842

    CAS  PubMed  Google Scholar 

  • Brasaemle DL, Levin D, Adler-Wailes D, Londos C (2000) The lipolytic stimulation of 3T3-L1 adipocytes promotes the translocation of hormone-sensitive lipase to the surfaces of lipid storage droplets. Biochim Biophys Acta 1483:251–262

    CAS  PubMed  Google Scholar 

  • Brinster RL, Chen HY, Trumbauer M, Senear AW, Warren R, Palmiter RD (1981) Somatic expression of herpes thymidine kinase in mice following injection of a fusion gene into eggs. Cell 27:223–231

    CAS  PubMed  Google Scholar 

  • Brooks B, Arch JRS, Newsholme EA (1982) Effects of hormones on the rate of the triacylglycerol/fatty acid substrate cycle in adipocytes and epididymal fat pads. FEBS Lett 146:327–330

    CAS  PubMed  Google Scholar 

  • Clifford G, Londos C, Kraemer F, Vernon R, Yeaman S (2000) Translocation of hormone-sensitive lipase and perilipin upon lipolytic stimulation of rat adipocytes. J Biol Chem 275:5011–5015

    CAS  PubMed  Google Scholar 

  • Cohen AW, Razani B, Schubert W, Williams TM, Wang XB, Iyengar P, Brasaemle DL, Scherer PE, Lisanti MP (2004) Role of caveolin-1 in the modulation of lipolysis and lipid droplet formation. Diabetes 53:1261–1270

    CAS  PubMed  Google Scholar 

  • Coleman RA, Lee DP (2004) Enzymes of triacylglycerol synthesis and their regulation. Prog Lipid Res 43:134–176

    CAS  PubMed  Google Scholar 

  • Dagan A, Yedgar S (1987) A facile method for direct determination of phospholipase A2 activity in intact cells. Biochem Int 15:801–808

    CAS  PubMed  Google Scholar 

  • DeLany JP, Floyd ZE, Zvonic S, Smith A, Gravois A, Reiners E, Wu X, Kilroy G, Lefevre M, Gimble JM (2005) Proteomic analysis of primary cultures of human adipose-derived stem cells: modulation by adipogenesis. Mol Cell Proteomics 4:731–740

    CAS  PubMed  Google Scholar 

  • Dichek HL, Parrott C, Ronan R, Brunzell JD, Brewer HB, Sanamarina-Fojo S (1993) Functional characterization of a chimeric lipase genetically engineered from human lipoprotein lipase and human hepatic lipase. J Lipid Res 34:1393–1401

    CAS  PubMed  Google Scholar 

  • Dole VP, Meinertz H (1960) Microdetermination of long chain fatty acids in plasma and tissue. J Biol Chem 235:2595–2599

    CAS  PubMed  Google Scholar 

  • Duncombe WG, Rising TJ (1973) Quantitative extraction and determination of nonesterified fatty acids in plasma. J Lipid Res 14:258–261

    CAS  PubMed  Google Scholar 

  • Ebdrup S, Sorensen LG, Olsen OH, Jacobsen P (2004) Synthesis and structure-activity relationship for a novel class of potent and selective carbamoyl-triazole based inhibitors for hormone sensitive lipase. J Med Chem 47:400–410

    CAS  PubMed  Google Scholar 

  • Edens NK, Leibel RL, Hirsch J (1990) Mechanism of free fatty acid re-esterification in human adipocytes in vitro. J Lipid Res 31:1423–1431

    CAS  PubMed  Google Scholar 

  • Egan J, Greenberg A, Chang M-K, Wek S, Moos J, Londos C (1992) Mechanism of hormone-stimulated lipolysis in adipocytes: translocation of hormone-sensitive lipase to the lipid storage droplet. Proc Natl Acad Sci U S A 89:8537–8541

    CAS  PubMed Central  PubMed  Google Scholar 

  • Folch J, Lees M, Sloane Stanley GH (1957) A simple method for the isolation and purification of total lipids from animal tissues. J Biol Chem 226:497–509

    CAS  PubMed  Google Scholar 

  • Franke WW, Hergt M, Grund C (1987) Rearrangement of the vimentin cytoskeleton during adipose conversion: formation of an intermediate filament cage around lipid globules. Cell 49:131–141

    CAS  PubMed  Google Scholar 

  • Frayn KN (2002) Adipose tissue as a buffer for daily lipid flux. Diabetologia 45:1201–1210

    CAS  PubMed  Google Scholar 

  • Fredrikson G et al (1981) Hormone-sensitive lipase of rat adipose tissue. Purification and some properties. J Biol Chem 256:6311–6320

    CAS  PubMed  Google Scholar 

  • Fujimoto Y, Iabe H, Sakai J, Makita M, Noda J, Mori M, Higashi Y, Kojima S, Takano T (2004) Identification of major proteins in the lipid droplet-enriched fraction isolated from the human hepatocyte cell line HuH7. Biochim Biophys Acta 1644:47–59

    CAS  PubMed  Google Scholar 

  • Gilbert CH, Kaye J, Galton DJ (1974) The effect of a glucose load on plasma fatty acids and lipolysis in adipose tissue of obese diabetic and non-diabetic patients. Diabetologia 10:135–138

    CAS  PubMed  Google Scholar 

  • Guilherme A, Soriano NA, Bose S, Holik J, Bose A, Pomerleau DP, Furcinitti P, Leszyk J, Corvera S, Czech MP (2004) CGI-58 interacts with perilipin and is localized to lipid droplets: possible involvement of GCI-58 mislocalization in Chanarin-Dorfman syndrome. J Biol Chem 279:30490–30497

    Google Scholar 

  • Haemmerle G, Lass A, Zimmermann R, Gorkiewicz G, Meyer C, Rozman J, Heldmaier G, Maier R, Theussl C, Eder S, Kratky D, Wagner EF, Klingenspor M, Hoefler G, Zechner R (2006) Defective lipolysis and altered energy metabolism in mice lacking adipose triglyceride lipase. Science 312:734–737

    Google Scholar 

  • Hämmerle G, Zimmermann R, Hayn M, Theussi C, Waeg G, Wagner E, Sattler W, Magin TM, Wagner EF, Zechner R (2002) Hormone-sensitive lipase deficiency in mice causes diglyceride accumulation in adipose tissue, muscle and testis. J Biol Chem 277:4806–4815

    Google Scholar 

  • Hammond VA, Johnston DG (1987) Substrate cycling between triglyceride and fatty acid in human adipocytes. Metabolism 36:308–313

    CAS  PubMed  Google Scholar 

  • Hendrickson HS (1994) Fluorescence-based assays of lipases, phospholipases, and other lipolytic enzymes. Anal Biochem 219:1–8

    CAS  PubMed  Google Scholar 

  • Hide WA, Chan L, Li WH (1992) Structure and evolution of the lipase superfamily. J Lipid Res 33:315–336

    Google Scholar 

  • Holm C (2003) Molecular mechanisms regulating hormone-sensitive lipase and lipolysis. Biochem Soc Trans 31:1120–1124

    CAS  PubMed  Google Scholar 

  • Holm C, Osterlund T (1999) Methods Mol Biol 109:109–121

    CAS  PubMed  Google Scholar 

  • Iverius P-H, Brunzell JD (1985) Human adipose tissue lipoprotein lipase: changes with feeding and relation to postheparin plasma enzyme. Am J Physiol 249:E107–E114

    CAS  PubMed  Google Scholar 

  • Jenkins CM, Mancuso DJ, Yan W, Sims HF, Gibson B, Gross RW (2004) Identification, cloning, expression, and purification of three novel human calcium-independent phospholipase A2 family members possessing triacylglycerol lipase and acylglycerol transacylase activities. J Biol Chem 279:48968–48975

    CAS  PubMed  Google Scholar 

  • Karpe F, Frayn KN (2004) The nicotinic acid receptor-a new mechanism for an old drug. Lancet 363:1892–1984

    CAS  PubMed  Google Scholar 

  • Kershaw EE, Hamm JK, Verhagen LAW, Peroni O, Katic M, Flier JS (2006) Adipose triglyceride lipase, function, regulation by insulin, and comparison with adiponutrin. Diabetes 55:148–157

    CAS  PubMed Central  PubMed  Google Scholar 

  • Kraemer FB, Shen WJ (2002) Hormone-sensitive lipase: control of intracellular tri-(di-)acylglycerol and cholesterylester hydrolysis. J Lipid Res 43:1585–1594

    CAS  PubMed  Google Scholar 

  • Lake AC, Sun Y, Li J-L, Kim JE, Johnson JW, Li D, Revett T, Shih HH, Liu W, Paulsen JE, Gimeno RE (2005) Expression, regulation, and triglyceride hydrolase activity of adiponutrin family members. J Lipid Res 46:2477–2487

    CAS  PubMed  Google Scholar 

  • Langin D (2006) Adipose tissue lipolysis as a metabolic pathway to define pharmacological strategies against obesity and the metabolic syndrome

    Google Scholar 

  • Langin D, Dicker A, Tavernier G, Hoffstedt J, Mairal A, Ryden M (2005) Adipocyte lipases and defect of lipolysis in human obesity. Diabetes 54:3190–3197

    CAS  PubMed  Google Scholar 

  • Laurell S, Tibbling G (1966) An enzymatic fluorometric micromethod for the determination of glycerol. Clin Chim Acta 13:317–322

    CAS  PubMed  Google Scholar 

  • Lehner R, Verger R (1997) Purification and characterization of a porcine liver microsomal triacylglycerol hydrolase. Biochemistry 36:1861–1868

    CAS  PubMed  Google Scholar 

  • Leibel RL, Forse RA, Hirsch J (1989) Effects of rapid glucose infusion on in vivo and in vitro free fatty acid reesterification by adipose tissue of fasted obese subjects. Int J Obes 13:661–671

    CAS  PubMed  Google Scholar 

  • Leibel RL, Hirsch J (1985) A radioisotopic technique for analysis of free fatty acid re-esterification in human adipose tissue. Am J Physiol 248:E140–E147

    CAS  PubMed  Google Scholar 

  • Leibel RL, Hirsch J, Berry EM, Gruen RK (1984) Radioisotopic method for the measurement of lipolysis in small samples of human adipose tissue. J Lipid Res 25:49–57

    CAS  PubMed  Google Scholar 

  • Lengsfeld H, Beaumier-Gallon G, Chahinian H, De Caro A, Verger R, Laugier R, Carriere F (2004) Physiology of gastrointestinal lipolysis and therapeutical use of lipases and digestive lipase inhibitors. In: Müller G, Petry S (eds) Lipases and phospholipases in drug development. Wiley-VCH, Weinheim, pp 195–229

    Google Scholar 

  • Lewin TM, Schwerbrock NM, Lee DP, Coleman RA (2004) Identification of a new glycerol-3-phosphate acyltransferase isoenzyme, mtGPAT2, in mitochondria. J Biol Chem 279:13488–13495

    CAS  PubMed  Google Scholar 

  • Lieber JG, Evans RM (1996) Disruption of the vimentin intermediate filament system durign adipose conversion of 3T3-L1 cells inhibits lipid droplet accumulation. J Cell Sci 109:3047–3058

    CAS  PubMed  Google Scholar 

  • Liu P, Rudick M, Anderson RG (2002) Multiple functions of caveolin-1. J Biol Chem 277:41295–41298

    CAS  PubMed  Google Scholar 

  • Liu P, Ying Y, Zhao Y, Mundy DI, Zhu M, Anderson RG (2004) Chinese hamster ovary K2 cell lipid droplets appear to be metabolic organelles involved in membrane traffic. J Biol Chem 279:3787–3792

    CAS  PubMed  Google Scholar 

  • Londos C, Sztalryd C, Tansey JT, Kimmel AR (2005) Role of PAT proteins in lipid metabolism. Biochimie 87:45–49

    CAS  PubMed  Google Scholar 

  • Marchesan D, Rutberg M, Andersson L, Asp L, Larsson T, Boren J, Johansson BR, Olofsson S-O (2003) A phospholipase D-dependent process forms lipid droplets containing caveolin, adipocyte differentiation-related protein, and vimentin in a cell-free system. J Biol Chem 278:27293–27300

    CAS  PubMed  Google Scholar 

  • Marcinkiewicz A, Gauthier D, Garcia A, Brasaemle DL (2006) The phosphorylation of serine 492 of perilipin a directs lipid droplet fragmentation and dispersion. J Biol Chem 281:11901–11909

    CAS  PubMed  Google Scholar 

  • Marial A, langin D, Arner P, Hoffstedt J (2006) Human adipose triglyceride lipase (PNPLA2) is not regulated by obesity and exhibits low in vitro triglyceride hydrolase activity. Diabetologia 49:1629–1636

    Google Scholar 

  • Martinez-Botas J, Andreson J, Tessler D, Lapillojnne A, Hung-Junn Chang B, Quast M, Gorenstein D, Chen K-H, Chan L (2000) Absence of perilipin results in leanness and reverses obesity in Lepr(db/db) mice. Nat Genet 26:474–479

    CAS  PubMed  Google Scholar 

  • Matsubara C, Nishikawa Y, Yoshida Y, Takamura K (1983) A spectrophotometric method for the determination of free fatty acid in serum using acyl-coenzyme A synthase and acyl-coenzyme A oxidase. Anal Biochem 130:128–133

    CAS  PubMed  Google Scholar 

  • Meshulam T, Herscovitz H, Casavant D, Bernardo J, Roman R, Haugland RP, Strohmeier GS, Diamond RD, Simons ER (1992) Flow cytometric kinetic measurements of neutrophil phospholipase A activation. J Biol Chem 267:21465–21470

    CAS  PubMed  Google Scholar 

  • Miles J, Glasscock R, Aikens J, Gerich J, Haymond M (1983) A microfluorometric method for the determination of free fatty acids in plasma. J Lipid Res 24:96–100

    CAS  PubMed  Google Scholar 

  • Miyoshi H, Souza SC, Zhang H-H, Strissel KJ, Christoffolete A, Kovsan J, Rudich A, Kraemer FB, Bianco AC, Obin MS, Greenberg AS (2006) Perilipin promotes HSL-mediated adipocyte lipolysis via phosphorylation-dependent and independent mechanisms. J Biol Chem 281:15837–15844

    CAS  PubMed  Google Scholar 

  • Morimoto C, Kameda K, Tsujita T, Okuda H (2001) Relationships between lipolysis induced by various lipolytic agents and hormone-sensitive lipase in rat fat cells. J Lipid Res 42:120–127

    CAS  PubMed  Google Scholar 

  • Morimoto C, Tsujita T, Sumida M, Okuda H (2000) Substratedependent lipolysis induced by isoproterenol. Biochem Biophys Res Commun 274:631–634

    CAS  PubMed  Google Scholar 

  • Moore HP, Silver RB, Mottillo EP, Bernlohr DA, Granneman JG (2005) Perilipin targets a novel pool of lipid droplets for lipolytic attack by hormone-sensitive lipase. J Biol Chem 280:43109–43120

    CAS  PubMed  Google Scholar 

  • Müller G, Petry S (2005) Triacylglycerol storage and mobilization, regulation of In: Meyers RA (ed) Encyclopedia of molecular cell biology and molecular medicine, vol 14, Wiley-VCH, Weinheim, p 621–704

    Google Scholar 

  • Müller G, Jordan H, Jung C, Kleine H, Petry S (2003) Analysis of lipolysis in adipocytes using a fluorescent fatty acid derivative. Biochimie 85:1245–1256

    PubMed  Google Scholar 

  • Müller G, Jordan H, Petry S, Wetekam E-M, Schindler P (1997b) Analysis of lipid metabolism in adipocytes using fluorescent fatty acids I. Insulin stimulation of lipogenesis. Biochim Biophys Acta 1347:23–39

    PubMed  Google Scholar 

  • Müller G, Wied S, Wetekam E-M, Crecelius A, Unkelbach A, Pünter J (1994a) Stimulation of glucose utilization in 3T3 adipocytes and rat diaphragm in vitro by the sulfonylureas, glimepiride and glibenclamide, is correlated with modulations of the cAMP regulatory cascade. Biochem Pharmacol 48:985–996

    PubMed  Google Scholar 

  • Näslund B, Bernström K, Lundin A, Arner P (1989) Free fatty acid determination by peroxidase-catalysed luminol chemiluminescence. J Biolumin Chemilumin 3:115–124

    PubMed  Google Scholar 

  • Näslund B, Bernström K, Lundin A, Arner P (1993) Release of small amounts of free fatty acids from human adipocytes as determined by chemiluminescence. J Lipid Res 34:633–641

    PubMed  Google Scholar 

  • Nisoli E, Carruba MO (2004) Emerging aspects of pharmacotherapy for obesity and metabolic syndrome. Pharmacol Res 50:453–469

    CAS  PubMed  Google Scholar 

  • Okabe H, Uji Y, Nagashima K, Noma A (1980) Enzymic determination of free fatty acids in serum. Clin Chem 26:1540–1543

    CAS  PubMed  Google Scholar 

  • Okuda H, Morimoto C, Tsujita T (1994) Effect of substrates on the cyclic AMP-dependent lipolytic reaction of hormonesensitive lipase. J Lipid Res 35:1267–1273

    CAS  PubMed  Google Scholar 

  • Osterlund T (2001) Structure-function relationships of hormone-sensitive lipase. Eur J Biochem 268:1899–1907

    CAS  PubMed  Google Scholar 

  • Osuga J, Ishibashi S, Oka T, Yagyu H, Tozawa R, Fujimoto A, Shionoiri F, Yahagi N, Kraemer FB, Tsutsumi O, Yamada N (2000) Targeted disruption of hormone-sensitive lipase results in male sterility and adipocyte hypertrophy, but not in obesity. Proc Natl Acad Sci U S A 97:787–792

    CAS  PubMed Central  PubMed  Google Scholar 

  • Pencreac’h G, Graille J, Pina M, Verger R (2002) An ultraviolet spectrophotometric assay for measuring lipase activity using long-chain triacylglycerols from Aleurites fordii seeds. Anal Biochem 303:17–24

    PubMed  Google Scholar 

  • Petry S, Ben Ali Y, Chahinian H, Jordan H, Kleine H, Müller G, Carriere F, Abousalham A (2005) Sensitive assay for hormone-sensitive lipase using NBD-labeled monoacylglycerol to detect low activities in rat adipocytes. J Lipid Res 46:603–614

    CAS  PubMed  Google Scholar 

  • Petry S, Baringhaus K-H, Schönafinger K, Jung C, Kleine H, Müller G (2004a) In: Müller G, Petry S (eds) Lipases and phospholipases in drug development. Wiley-VCH, Weinheim, pp 121–138

    Google Scholar 

  • Raben DM, Baldassare JJ (2005) A new lipase in regulating lipid mobilization: hormone-sensitive lipase is not alone. Trends Endocrinol Metab 16:35–36

    CAS  PubMed  Google Scholar 

  • Robenek H, Robenek MJ, Buers I, Lorkowski S, Hofnagel O, Troyer D, Severs NJ (2005a) Lipid droplets gain PAT family proteins by interaction with specialized plasma membrane proteins. J Biol Chem 280:26330–26338

    CAS  PubMed  Google Scholar 

  • Robenek H, Robenek MJ, Troyer D (2005b) PAT family proteins pervade lipid droplet cores. J Lipid Res 46:1331–1338

    CAS  PubMed  Google Scholar 

  • Robenek MJ, Severs NJ, Schlattmann K, Plenz G, Zimmer KP, Troyer D, Robenek H (2004) Lipids partition caveoln-1 from ER membranes into lipid droplets: updating the model of lipid droplet biogenesis. FASEB J 18:866–868

    CAS  PubMed  Google Scholar 

  • Schmid RD, Verger R (1998) Lipases: interfacial enzymes with attractive applications. Angew Chem Int Ed Engl 37:1608–1633

    Google Scholar 

  • Scholze H, Stutz H, Paltauf F, Hermetter A (1999a) Fluorescent inhibitors for the qualitative and quantitative analysis of lipolytic enzymes. Anal Biochem 276:72–80

    CAS  PubMed  Google Scholar 

  • Shimizu S, Tani Y, Yamada M, Tabata M, Murachi T (1980) Enzymatic determination of serum-free fatty acids: a colorimetric method. Anal Biochem 107:193–198

    CAS  PubMed  Google Scholar 

  • Slee DH, Bhat AS, Nguyen TN, Kish M, Lundeen K, Newman MJ (2003) Pyrrolopyrazinedione-based inhibitors of human hormone-sensitive lipase. J Med Chem 46:1120–1122

    CAS  PubMed  Google Scholar 

  • Soni KG, Lehner R, Metalnikov P, O'Donnell P, Semache M, Gao W, Ashman K, Pshezhetsky AV, Mitchell GA (2004) Carboxylesterase 3 (EC 3.1.1.1) is a major adipocyte lipase. J Biol Chem 279: 40683–40689

    Google Scholar 

  • Sooranna SR, Saggerson ED (1976b) Interactions of insulin and adrenaline with glycerol phosphate acylation processes in fat cells from rat. FEBS Lett 64:36–39

    CAS  PubMed  Google Scholar 

  • Subramanian V, Rothenberg A, Gomez C, Cohen AW, Garcia A, Bhattacharyya S, Shapiro L, Dolios G, Wang R, Lisanti M, Brasaemle DL (2004) Hydrophobic sequences target and anchor perilipin A to lipid droplets. J Biol Chem 279:42062–42071

    CAS  PubMed  Google Scholar 

  • Sztalryd C, Xu G, Dorward H, Tansey J, Contreras J, Kimmel A, Londos C (2003) Perilipin A is essential for the translocation of hormone-sensitive lipase during lipolytic activation. J Cell Biol 161:1093–1103

    CAS  PubMed Central  PubMed  Google Scholar 

  • Tansey JT, Huml AM, Vogt R, Davis KE, Jones JM, Fraser KA, Brasaemle DL, Kimmel AR, Londos C (2003) J Biol Chem 278:8401–8406

    CAS  PubMed  Google Scholar 

  • Tansey JT, Sztalryd C, Gruia-Gray J, Roush DL, Zee JV, Gavrilova O, Reitman ML, Deng CX, Li C, Kimmel AR, Londos C (2001) Proc Natl Acad Sci U S A 98:6494–6499

    CAS  PubMed Central  PubMed  Google Scholar 

  • Tiraby C, Langin D (2003) Conversion of white into brown adipocytes: a strategy for the control of fat mass? Trends Endocrinol Metab 14:439–441

    Google Scholar 

  • Tiss A, Miled N, Verger R, Gargouri Y, Abousalham A (2004) Digestive lipases inhibition: an in vitro study. In: Müller G, Petry S (eds) Lipases and phospholipases in drug development. Wiley-VCH, Weinheim, pp 155–193

    Google Scholar 

  • Tunaru S, Kero J, Schaub A, Wufka C, Blaukat A, Pfeffer K (2003) PUMA-G and HM74 are receptors for nicotinic acid and mediate its anti-lipolytic effect. Nat Med 9:352–355

    CAS  PubMed  Google Scholar 

  • Umlauf E, Csaszar E, Moertelmaier M, Schuetz GJ, Parton RG, Prohaska R (2004) Association of stomatin with lipid bodies. J Biol Chem 279:23699–23709

    CAS  PubMed  Google Scholar 

  • Vaughan M (1962) The production and release of glycerol by adipose tissue incubated in vitro. J Biol Chem 237:3354–3358

    CAS  PubMed  Google Scholar 

  • Verger R (1997) ‘Interfacial activation’ of lipases: facts and artefacts. Trends Biotechnol 15:32–38

    CAS  Google Scholar 

  • Vertesy L, Beck B, Brönstrup M, Ehrlich K, Kurz M, Müller G, Schummer D, Seibert G (2002) Cyclipostins, novel hormone-sensitive lipase inhibitors from Streptomyces sp. DSM 13381. II. Isolation, structure elucidation and biological properties. J Antibiot 55:480–494

    CAS  PubMed  Google Scholar 

  • Vila MDC, Milligan G, Standaert ML, Farese RV (1990a) Insulin activates glycerol-3-phosphate-acyltransferase (de novo phosphatidic acid synthesis) through a phospholipid derived mediator. Apparent involvement of Gi? and activation of a phospholipase C. Biochemistry 29:8735–8740

    CAS  PubMed  Google Scholar 

  • Villena JA, Roy S, Sarkadi-Nagy E, Kim KH, Sul HS (2004) Desnutrin, an adipocyte gene encoding a novel patatin domain-containing protein, is induced by fasting and glucocorticoids: ectopic expression of desnutrin increases triglyceride hydrolysis. J Biol Chem 279:47066–47075

    CAS  PubMed  Google Scholar 

  • Wang SP, Laurin N, Himms-Hagen J, Rudnicki MA, Levy E, Robert MF, Pan I, Oligny L, Mitchell GA (2001) The adipose tissue phenotype of hormone-sensitive lipase deficiency in mice. Obes Res 9:119–128

    CAS  PubMed  Google Scholar 

  • Wieland O (1974) Glycerin UV-methode. In: Bergmeyer HU (ed) Methoden der enzymatischen analyse. Verlag Chemie, Weinheim, pp 1448–1453

    Google Scholar 

  • Wise A, Foord SM, Fraser NJ, Barnes AA, Elshourbagy N, Eilert M (2003) Molecular identification of high and low affinity receptors for nicotinic acid. J Biol Chem 278:9869–9874

    CAS  PubMed  Google Scholar 

  • Wittenauer LA, Shirai K, Jackson RL, Johnson JD (1984) Hydrolysis of a fluorescent phospholipid substrate by phospholipase A2 and lipoprotein lipase. Biochem Biophys Res Commun 118:894–901

    CAS  PubMed  Google Scholar 

  • Wolins NE, Quaynor BK, Skinner JR, Schoenfish MJ, Tzekov A, Bickel PE (2005) S3–12, adipophilin, and TIP47 package lipid in adipocytes. J Biol Chem 280:19146–19155

    CAS  PubMed  Google Scholar 

  • Wu CC, Howell KE, Neville MC, Yates JR, McManaman JL (2000) Proteomics reveal a link between the endoplasmic reticulum and lipid secretory mechanisms in mammary epithelial cells. Electrophoresis 21:3470–3482

    CAS  PubMed  Google Scholar 

  • Yamaguchi T, Omatsu N, Matsushita S, Osumi T (2004) CGI-58 interacts with perilipin and is localized to lipid droplets. Possible involvement of CGI-58 mislocalization in Chanarin-Dorfman syndrome. J Biol Chem 279:30490–30497

    CAS  PubMed  Google Scholar 

  • Yeaman SJ (2004) Hormone-sensitive lipase: new roles for an old enzyme. Biochem J 379:11–22

    CAS  PubMed Central  PubMed  Google Scholar 

  • Zimmermann R, Strauss JG, Hämmerle G, Schoiswohl G, Birner-Grünberger R, Riederer M, Lass A, Neuberger G, Eisenhaber F, Hermetter A, Zechner R (2004a) Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science 306:1383–1386

    CAS  PubMed  Google Scholar 

Interaction of ATGL and CGI-58

  • Adam GC, Sorensen EJ, Cravatt BF (2002) Trifunctional chemical probes for the consolidated detection and identification of enzyme activities from complex proteomes. Mol Cell Proteomics 1:781–790

    CAS  PubMed  Google Scholar 

  • Arpigny JL, Jaeger KE (1999) Bacterial lipolytic enzymes: classification and properties. Biochem J 343:177–183

    CAS  PubMed Central  PubMed  Google Scholar 

  • Birner-Grünberger R, Susani-Etzerodt H, Waldhuber M, Riesenhuber G, Schmidinger H, Rechberger G, Kollroser M, Strauss JG, Lass A, Zimmermann R, Hämmerle G, Zechner R, Hermetter A (2005) The lipolytic proteome of mouse adipose tissue. Mol Cell Biol 4:1710–1717

    Google Scholar 

  • Carr S, Aebersold R, Baldwin M, Burlingame A, Clauser K, Nesvizhskii A (2004) The need for guidelines in publication of peptide and protein identification data: working group on publication guidelines for peptide and protein identification data. Mol Cell Proteomics 3:531–533

    CAS  PubMed  Google Scholar 

  • Chanarin I, Patel A, Slavin G, Wills EJ, Andrews TM, Stewart G (1975) Neutral-lipid storage disease: a new disorder of lipid metabolism. BMJ 1:553–555

    CAS  PubMed Central  PubMed  Google Scholar 

  • Dorfman ML, Hershko C, Eisenberg S, Sagher F (1974) Ichthyosiform dermatosis with systemic lipidosis. Arch Dermatol 110:261–266

    CAS  PubMed  Google Scholar 

  • Gorg A, Postel W, Gunther S, Weser J (1985) Improved horizontal two-dimensional electrophoresis with hybrid isoelectric-focusing in immobilized ph gradients in the 1st-dimension and laying-on transfer in the 2nd-dimension. Electrophoresis 6:599–604

    Google Scholar 

  • Gorg A, Postel W, Gunther S (1988) The current state of twodimensional electrophoresis with immobilized ph gradients. Electrophoresis 9:531–554

    CAS  PubMed  Google Scholar 

  • Greenbaum DC, Arnold WD, Lu F, Hayrapetian L, Baruch A, Krumrine J, Toba S, Chehade K, Bromee D, Kuntz ID, Bogyo M (2002) Small molecule affinity fingerprinting, a tool for enzyme family subclassification, target identification, and inhibitor design. Chem Biol 9:1085–1094

    CAS  PubMed  Google Scholar 

  • Lass A, Zimmermann R, Hämmerle G, Riederer M, Schoiswohl G, Schweiger M, Kienesberger P, Strauss JG, Gorkiewicz G, Zechner R (2006) Adipose triglyceride lipase-mediated lipolysis of cellular fat stores is activated by CGI-58 and defective in Chanarin-Dorfman syndrome. Cell Metab 3:309–319

    CAS  PubMed  Google Scholar 

  • Lefevre C, Jobard F, Caux F, Bouadjar B, Karaduman A, Heilig R, Lakhdar H, Wollenberg A, Verret A, Weissenbach J (2001) Mutations in CGI-58, the gene encoding a new protein of the esterase/lipase/thioesterase subfamily, in Chanarin-Dorfman syndrome. Am J Hum Genet 69:1002–1012

    CAS  PubMed Central  PubMed  Google Scholar 

  • Manesse MLM, Boots J-WP, Dijkman R, Slotboom AT, van der Hjiden HTWM, Egmond MR, Verhij HM, de Haas GH (1995) Phosphonate analogues of triacylglycerols are potent inhibitors of lipase. Biochim Biophys Acta 1259:56–64

    Google Scholar 

  • Martinelle M, Holmquist M, Hult K (1995) On the interfacial activation of Candida antarctica lipase A and B as compared with Humicola lanuginosa lipase. Biochim Biophys Acta 1258:272–276

    PubMed  Google Scholar 

  • Oskolkova OV, Saf R, Zenzmaier E, Hermetter A (2003) Fluorescent organophosphonates as inhibitors of microbial lipases. Chem Phys Lipids 125:103–114

    CAS  PubMed  Google Scholar 

  • Petry S, Baringhaus K-H, Schönafinger K, Jung C, Kleine H, Müller G (2004b) High-throughput screening of hormonesensitive lipase and subsequent computer-assisted compound optimization. In: Müller G, Petry S (eds) Lipases and phospholipases in drug development. Wiley-VCH, Weinheim, pp 121–136

    Google Scholar 

  • Pleiss J, Fischer M, Schmid RD (1998) Anatomy of lipase binding sites: the scissile fatty acid binding site. Chem Phys Lipids 93:67–80

    CAS  PubMed  Google Scholar 

  • Rotticci D, Norin T, Hult K, Martinelle M (2000) An activesite titration method for lipases. Biochim Biophys Acta 1483:132–140

    CAS  PubMed  Google Scholar 

  • Schmidinger H, Birner-Grünberger R, Riesenhuber G, Saf R, Susani-Etzerodt H, Hermetter A (2005) Novel fluorescent phosphonic acid esters for discrimination of lipases and esterases. Chembiochem 6:1–6

    Google Scholar 

  • Scholze H, Stutz H, Paltauf F, Hermetter A (1999b) Fluorescent inhibitors for the qualitative and quantitative analysis of lipolytic enzymes. Anal Biochem 276:72–80

    CAS  PubMed  Google Scholar 

  • Shevchenko A, Wilm M, Vorm O, Mann M (1996) Mass spectrometric sequencing of proteins from silver stained polyacrylamide gels. Anal Chem 68:850–858

    CAS  PubMed  Google Scholar 

  • Speers AE, Cravatt BF (2004a) Chemical strategies for activitybased proteomics. Chembiochem 5:41–47

    CAS  PubMed  Google Scholar 

  • Speers AE, Cravatt BF (2004b) Profiling enzyme activities in vivo using click chemistry methods. Chem Biol 11:535–546

    CAS  PubMed  Google Scholar 

  • Wang Q, Chan TR, Hilgraf R, Fokin VV, Sharpless KB, Finn MG (2003) Bioconjugation by copper(I)-catalyzed azide-alkyne [3+2] cycloaddition. J Am Chem Soc 125:3192–3193

    CAS  PubMed  Google Scholar 

  • Zimmermann R, Strauss JG, Hämmerle G, Schoiswohl G, Birner-Grünberger R, Riederer M, Lass A, Neuberger G, Eisenhaber F, Hermetter A, Zechner R (2004b) Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science 306:1383–1386

    CAS  PubMed  Google Scholar 

Protein Phosphatase (PP) Activity

  • Honnor RC, Dhillon GS, Londos C (1985a) cAMP-dependent protein kinase and lipolysis in rat adipocytes I. Cell preparation, manipulation, and predictability in behavior. J Biol Chem 260:15122–15129

    CAS  PubMed  Google Scholar 

  • Honnor RC, Dhillon GS, Londos C (1985b) cAMP-dependent protein kinase and lipolysis in rat adipocytes II. Definition of steady-state relationship with lipolytic and antilipolytic modulators. J Biol Chem 260:15130–15138

    CAS  PubMed  Google Scholar 

  • Kono T, Robinson FW, Sarver JA (1975) Insulin-sensitive phosphodiesterase. Its localization, hormonal stimulation, and oxidative stabilization. J Biol Chem 250:7826–7835

    CAS  PubMed  Google Scholar 

  • Londos C, Honnor RC, Dhillon GS (1985) cAMP-dependent protein kinase and lipolysis in rat adipocytes III. Multiple modes of insulin regulation of lipolysis and regulation of insulin responses by adenylate cyclase regulators. J Biol Chem 260:15139–15145

    CAS  PubMed  Google Scholar 

  • Müller G, Petry S (2004) Physiological and pharmacological regulation of triacylglycerol storage and mobilization. In: Müller G, Petry S (eds) Lipases and phospholipases in drug development. Wiley-VCH, Weinheim, pp 231–332

    Google Scholar 

  • Müller G, Wied S, Wetekam EM, Crecelius A, Punter J (1994b) Stimulation of glucose utilization in 3T3 adipocytes and rat diaphragm in vitro by the sulfonylureas glimepiride and glibenclamide, is correlated with modulations of the cAMP regulatory cycle. Biochem Pharmacol 48:985–996

    PubMed  Google Scholar 

  • Müller G, Grey S, Jung C, Bandlow W (2000) Insulin-like signaling in yeast: modulation of protein phosphatase 2A, protein kinase A, cAMP-specific phosphodiesterase, and glycosyl-phosphatidylinositol-specific phospholipase C activities. Biochemistry 39:1475–1488

    PubMed  Google Scholar 

  • Okuno S, Inaba M, Nishizawa Y, Inoue A, Morii H (1988) Effect of tolbutamide and glyburide on cAMP-dependent protein kinase activity in rat liver cytosol. Diabetes 37:857–861

    CAS  PubMed  Google Scholar 

  • Osegawa M, Makino H, Kanatsuka A, Kumagai A (1982) Effects of sulfonylureas on membrane-bound low Km cyclic AMP phosphodiesterase in rat fat cells. Biochim Biophys Acta 721:289–296

    CAS  PubMed  Google Scholar 

  • Roskoski R (1983) Assays of protein kinase. Methods Enzymol 99:3–6

    CAS  PubMed  Google Scholar 

  • Saltiel AR, Steigerwalt RW (1985) Purification of putative insulin-sensitive cAMP phosphodiesterase or its catalytic domain from adipose tissue. Diabetes 35:698–704

    Google Scholar 

  • Schölch C, Kuhlmann J, Gossel M, Müller G, Neumann-Hafelin C, Belz U, Kalisch J, Biemer-Daub G, Kramer W, Juretschke H-P, Herling A (2004) Characterization of adenosine-A1-receptor-mediated antilipolysis in rats by tissue-microdialysis, 1H-spectroscopy and glucose clamp studies. Diabetes 53:1920–1926

    Google Scholar 

  • Solomon SS, Deaton J, Shankar TP, Palazzolo M (1986) Cyclic AMP phosphodiesterase in diabetes. Effect of glyburide. Diabetes 35:1233–1236

    CAS  PubMed  Google Scholar 

  • Vila MDC, Milligan G, Standaert ML, Farese RV (1990b) Insulin activates glycerol-3-phosphate-acyltransferase (de novo phosphatidic acid synthesis) through a phospholipid-derived mediator. Apparent involvement of Giα and activation of a phospholipase C. Biochemistry 29:8735–8740

    CAS  PubMed  Google Scholar 

Measurement of Mitochondrial Membrane Potential

  • Bukowiecki L, Lindberg O (1974) Control of sn-glycerol 3-phosphate oxidation in brown adipose tissue mitochondria by calcium and acyl-CoA. Biochim Biophys Acta 348:115–125

    CAS  PubMed  Google Scholar 

  • Cannon B (1971) Control of fatty-acid oxidation in brown-adipose-tissue mitochondria. Eur J Biochem 23:125–135

    CAS  PubMed  Google Scholar 

  • Cannon B, Bernson VMS, Nedergaard J (1984) Metabolic consequences of limited substrate anion permeability in brown fat mitochondria from a hibernator, the golden hamster. Biochim Biophys Acta 766:483–491

    CAS  PubMed  Google Scholar 

  • Cannon B, Nedergaard J (2001) Cultures of adipose precursor cells from brown adipose tissue and of clonal brown-adipocyte-like cell lines. In: Ailhaud G (ed) Adipose tissue protocols. Humana Press, Totowa, pp 213–224

    Google Scholar 

  • Fain JN, Reed N, Saperstein R (1967) The isolation and metabolism of brown fat cells. J Cell Biol 242:1887–1894

    CAS  Google Scholar 

  • Lindberg O, DePierre J, Rylander E, Afzelius BA (1967) Studies of the mitochondrial energy transfer system of brown adipose tissue. J Cell Biol 34:293–310

    CAS  PubMed Central  PubMed  Google Scholar 

  • Marshall SE, McCormack JG, Denton RM (1984) Role of Ca2+ ions in the regulation of intramitochondrial metabolism in rat epididymal adipose tissue. Evidence against a role for Ca2+ in the activation of pyruvate dehydrogenase by insulin. Biochem J 218:249–260

    CAS  PubMed Central  PubMed  Google Scholar 

  • Matthias A, Jacobsson A, Cannon B, Nedergaard J (1999) The bioenergetics of brown fat mitochondria from UCP1-ablated mice. UCP1 is not involved in fatty acid-induced de-energization. J Biol Chem 274:28150–28160

    CAS  PubMed  Google Scholar 

  • Matthias A, Ohlson KEB, Fredriksson JM, Jacobsson A, Nedergaard J, Cannon B (2000) Thermogenic responses in brown-fat cells are fully UCP1-dependent: UCP2 or UCP3 do not substitute for UCP1 in adrenergically or fatty-acid induced thermogenesis. J Biol Chem 275:25073–25081

    CAS  PubMed  Google Scholar 

  • Monti M, Nilsson-Ehle P, Sörbis R, Wadsö I (1980) Microcalorimetric measurement of heat production in isolated human adipocytes. Scand J Clin Lab Invest 40:581–587

    CAS  PubMed  Google Scholar 

  • Nedergaard J (1982) Catecholamine sensitivity in brown fat cells from cold-acclimated hamsters and rats. Am J Physiol 242:C250–C257

    CAS  PubMed  Google Scholar 

  • Nedergaard J, Cannon B, Lindberg O (1977) Microcalorimetry of isolated mammalian cells. Nature 267:518–520

    CAS  PubMed  Google Scholar 

  • Nicholls DG (1974) Hamster brown-adipose-tissue mitochondria. The control of respiration of the proton electrochemical potential gradient by possible physiological effectors of the proton conductance of the inner membrane. Eur J Biochem 49:573–583

    CAS  PubMed  Google Scholar 

  • Nicholls DG, Grav HJ, Lindberg O (1972) Mitochondria from hamster brown-adipose tissue. Regulation of respiration in vitro by variation in volume of the matrix compartment. Eur J Biochem 37:526–533

    Google Scholar 

  • Nicholls DG, Lindberg O (1973) Brown-adipose-tissue mitochondria. The influence of albumin and nucleotides on ion permeabilities. Eur J Biochem 37:523–530

    CAS  PubMed  Google Scholar 

  • Olsson SA, Monti M, Sörbis R, Nilsson-Ehle P (1986) Adipocyte heat production before and after weight reduction by gastroplasty. Int J Obes 10:99–105

    CAS  PubMed  Google Scholar 

  • Prusiner SB, Cannon B, Lindberg O (1968) Oxidative metabolism in cells isolated from brown adipose tissue. I. Catecholamine and fatty acid stimulation of respiration. Eur J Biochem 6:15–22

    CAS  PubMed  Google Scholar 

  • Robinson PK (1994) The Clark oxygen electrode. In: Wilson K, Walker J (eds) Principles and techniques of practical biochemistry. Cambridge University Press, Cambridge, pp 555–562

    Google Scholar 

  • Rodbell M (1964) Metabolism of isolated fat cells. 1. Effects of hormones on glucose metabolism and lipolysis. J Biol Chem 239:375–380

    CAS  PubMed  Google Scholar 

  • Shabalina IG, Jacobsson A, Cannon B, Nedergaard J (2004) Native UCP1 displays simple competitive kinetics between the regulators purine nucleotides and fatty acids. J Biol Chem 279:38236–38248

    CAS  PubMed  Google Scholar 

  • Smith RE, Roberts JC, Hittelman KJ (1966) Nonphosphorylating respiration of mitochondria from brown adipose tissue of rats. Science 154:653–654

    CAS  PubMed  Google Scholar 

  • Svartengren J, Svoboda P, Cannon B (1982) Desensitization of ß-adrenergic responsiveness in vivo. Decreased coupling between receptors and adenylate cyclase in isolated brown fat cells. Eur J Biochem 128:481–488

    CAS  PubMed  Google Scholar 

  • Valdemarsson S, Fagher B, Hedner P, Monti M, Nilsson-Ehle P (1985) Platelet and adipocyte thermogenesis in hypothyroid patients: a microcalorimetric study. Acta Endocrinol 108:361–366

    CAS  PubMed  Google Scholar 

  • Zhao J, Cannon B, Nedergaard J (1998a) Thermogenesis is ß3- but not ß1-adrenergically mediated in brown fat cells, even after cold stimulation. Am J Physiol 275:R2002–R2011

    CAS  PubMed  Google Scholar 

  • Zhao J, Cannon B, Nedergaard J (1998b) Carteolol is a weak partial agonist on ß3-adrenergic receptor in brown adipocytes. Can J Physiol Pharmacol 76:428–433

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Günter Müller .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer-Verlag Berlin Heidelberg

About this entry

Cite this entry

Müller, G. (2015). Assays for Insulin and Insulin-Like Activity Based on Adipocytes. In: Hock, F. (eds) Drug Discovery and Evaluation: Pharmacological Assays. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-642-27728-3_154-1

Download citation

  • DOI: https://doi.org/10.1007/978-3-642-27728-3_154-1

  • Received:

  • Accepted:

  • Published:

  • Publisher Name: Springer, Berlin, Heidelberg

  • Online ISBN: 978-3-642-27728-3

  • eBook Packages: Springer Reference Biomedicine and Life SciencesReference Module Biomedical and Life Sciences

Publish with us

Policies and ethics