Skip to main content

Adipose Tissue and Ceramide Biosynthesis in the Pathogenesis of Obesity

  • Chapter
Sphingolipids and Metabolic Disease

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 721))

Abstract

Although obesity is a complex metabolic disorder often associated with insulin resistance, hyperinsulinemia and Type 2 diabetes, as well as with accelerated atherosclerosis, the molecular changes in obesity that promote these disorders are not completely understood. Several mechanisms have been proposed to explain how increased adipose tissue mass affects whole body insulin resistance and cardiovascular risk. One theory is that increased adipose derived inflammatory cytokines induces a chronic inflammatory state that not only increases cardiovascular risk, but also antagonizes insulin signaling and mitochondrial function and thereby impair glucose hemostasis. Another suggests that lipid accumulation in nonadipose tissues not suited for fat storage leads to the buildup of bioactive lipids that inhibit insulin signaling and metabolism. Recent evidence demonstrates that sphingolipid metabolism is dysregulated in obesity and specific sphingolipids may provide a common pathway that link excess nutrients and inflammation to increased metabolic and cardiovascular risk. This chapter will focus primarily on the expression and regulation of adipose and plasma ceramide biosynthesis in obesity and, its potential contribution to the pathogenesis of obesity and the metabolic syndrome.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Ogden CL, Carroll MD, Curtin LR et al. Prevalence of overweight and obesity in the United States, 1999–2004. JAMA 2006; 295(13):1549–1555.

    PubMed  CAS  Google Scholar 

  2. Hotamisligil GS. Inflammation and metabolic disorders. Nature 2006; 444(7121):860–867.

    PubMed  CAS  Google Scholar 

  3. Hotamisligil GS. Molecular mechanisms of insulin resistance and the role of the adipocyte. Int J Obes Relat Metab Disord 2000; 24(Suppl 4):S23–S27.

    PubMed  CAS  Google Scholar 

  4. Mohamed-Ali V, Goodrick S, Rawesh A et al. Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-alpha, in vivo. J Clin Endocrinol Metab 1997; 82:4196–4200.

    PubMed  CAS  Google Scholar 

  5. Christiansen T, Richelsen B, Bruun JM. Monocyte chemoattractant protein-1 is produced in isolated adipocytes, associated with adiposity and reduced after weight loss in morbid obese subjects. Int J Obes (Lond) 2005; 29(1): 146–150.

    CAS  Google Scholar 

  6. Bruun JM, Pedersen SB, Richelsen B. Regulation of interleukin 8 production and gene expression in human adipose tissue in vitro. J Clin Endocrinol Metab 2001; 86(3):1267–1273.

    PubMed  CAS  Google Scholar 

  7. Wellen KE, Hotamisligil GS. Inflammation, stress and diabetes. J Clin Invest 2005; 115(5):1111–1119.

    PubMed  CAS  Google Scholar 

  8. Fantuzzi G. Adipose tissue, adipokines and inflammation. J Allergy Clin Immunol 2005; 115(5):911–919.

    PubMed  CAS  Google Scholar 

  9. Dandona P, Aljada A, Bandyopadhyay A. Inflammation: the link between insulin resistance, obesity and diabetes. Trends Immunol 2004; 25(1):4–7.

    PubMed  CAS  Google Scholar 

  10. Hotamisligil GS. Inflammatory pathways and insulin action. Int J Obes Relat Metab Disord 2003; 27(Suppl 3):S53–S55.

    PubMed  CAS  Google Scholar 

  11. Neels JG, Badeanlou L, Hester KD et al. Keratinocyte-derived chemokine in obesity: Expression and role in adipose macrophage infiltration and glucose homeostasis. J Biol Chem 2009.

    Google Scholar 

  12. Dellas C, Loskutoff DJ. Historical analysis of PAI-1 from its discovery to its potential role in cell motility and disease. Thromb Haemost 2005; 93(4):631–640.

    PubMed  CAS  Google Scholar 

  13. Samad F, Loskutoff DJ. Tissue distribution and regulation of plasminogen activator inhibitor-1 in obese mice. Mol Med 1996; 2:568–582.

    PubMed  CAS  Google Scholar 

  14. Juhan-Vague I, Alessi MC, Mavri A et al. Plasminogen activator inhibitor-1, inflammation, obesity, insulin resistance and vascular risk. J Thromb Haemost 2003; 1:1575–1579.

    PubMed  CAS  Google Scholar 

  15. Loskutoff DJ, Samad F. The adipocyte and hemostatic balance in obesity: Studies of PAI-1. Arterioscler Thromb Vasc Biol 1998; 18:1–6.

    PubMed  CAS  Google Scholar 

  16. Eriksson P, Reynisdottir S, Lönnqvist F et al. Adipose tissue secretion of plasminogen activator inhibitor-1 in non-obese and obese individuals. Diabetologia 1998; 41:65–71.

    PubMed  CAS  Google Scholar 

  17. Loskutoff DJ, Ihara H, Yamamoto K, et al. The fat mouse: A powerful genetic model to study alterations in hemostatic gene expression in obesity. In: Suzuki K, Ikeda Y, Maruyama I, eds. New frontier in Vascular Biology; Thrombosis and Hemostasis. Osaka: Eibun Press, Inc., 2000:151–160.

    Google Scholar 

  18. De TB, Smith LH, Vaughan DE. Plasminogen activator inhibitor-1: a common denominator in obesity, diabetes and cardiovascular disease. Curr Opin Pharmacol 2005; 5(2): 149–154.

    Google Scholar 

  19. Schäfer K, Fujisawa K, Konstantinides S et al. Disruption of the plasminogen activator inhibitor-1 gene reduces the adiposity and improves the metabolic profile of geneticallly obese and diabetic ob/ob mice. FASEB J 2001; (June 27, 2001) 10.1096/fj.00-0750fje.

    Google Scholar 

  20. Ma L-J, Mao S-L, Taylor KL et al. Prevention of obesity and insulin resistance in mice lacking plasminogen activator inhibitor 1. Diabetes 2004; 53:336–346.

    PubMed  CAS  Google Scholar 

  21. Kraegen EW, Cooney GJ, Ye JM et al. The role of lipids in the pathogenesis of muscle insulin resistance and beta cell failure in type II diabetes and obesity. Exp Clin Endocrinol Diabetes 2001;109(Suppl2):S189–S201.

    PubMed  CAS  Google Scholar 

  22. Lelliott C, Vidal-Puig AJ. Lipotoxicity, an imbalance between lipogenesis de novo and fatty acid oxidation. Int J Obes Relat Metab Disord 2004; 28(Suppl 4):S22–S28.

    PubMed  CAS  Google Scholar 

  23. Unger RH. Minireview: weapons of lean body mass destruction: the role of ectopic lipids in the metabolic syndrome. Endocrinol 2003; 144(12):5159–5165.

    CAS  Google Scholar 

  24. McGarry JD. Banting lecture 2001: dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes 2002; 51(1):7–18.

    PubMed  CAS  Google Scholar 

  25. Summers SA. Ceramides in insulin resistance and lipotoxicity. Prog Lipid Res 2006; 45(1):42–72.

    PubMed  CAS  Google Scholar 

  26. Hanada K. Serine palmitoyltransferase, a key enzyme of sphingolipid metabolism. Biochim Biophys Acta 2003; 1632(1–3):16–30.

    PubMed  CAS  Google Scholar 

  27. Futerman AH, Hannun YA. The complex life of simple sphingolipids. EMBO Rep 2004; 5(8):777–782.

    PubMed  CAS  Google Scholar 

  28. Hannun YA, Obeid LM. The ceramide-centric universe of lipid-mediated cell regulation: Stress encounters of the lipid kind. J Biol Chem 2002; 277:25847–25850.

    PubMed  CAS  Google Scholar 

  29. Pewzner-jung Y, Ben-Dor S, Futerman AH. When do Lasses (longevity assurance genes) become CerS (ceramide synthases)?: Insights into the regulation of ceramide synthesis. J Biol Chem 2006; 281(35):25001–25005.

    PubMed  CAS  Google Scholar 

  30. Sugiura M, Kono K, Liu H et al. Ceramide kinase, a novel lipid kinase. Molecular cloning and functional characterization. J Biol Chem 2002; 277:23294–23300.

    PubMed  CAS  Google Scholar 

  31. Hirabayashi Y, Osuka S, Nagatsuka Y. [Biology of glucosylceramide synthase: importance of lipid glycosylation]. Tanpakushitsu Kakusan Koso 2003; 48(8 Suppl):958–962.

    PubMed  CAS  Google Scholar 

  32. Samad F, Hester KD, Yang G et al. Altered adipose and plasma sphingolipid metabolism in obesity: a potential mechanism for cardiovascular and metabolic risk. Diabetes 2006; 55(9):2579–2587.

    PubMed  CAS  Google Scholar 

  33. Holland WL, Brozinick JT, Wang LP et al. Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-and obesity-induced insulin resistance. Cell Metab 2007; 5(3): 167–179.

    PubMed  CAS  Google Scholar 

  34. Shah C, Yang G, Lee I, et al. Protection from high fat diet-induced increase in ceramide in mice lacking plasminogen activator inhibitor 1. J Biol Chem 2008; 283(20):13538–13548.

    PubMed  CAS  Google Scholar 

  35. Haus JM, Kashyap SR, Kasumov T et al. Plasma ceramides are elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance. Diabetes 2009; 58(2):337–343.

    PubMed  CAS  Google Scholar 

  36. Gorska M, Dobrzyn A, Baranowski M. Concentrations of sphingosine and sphinganine in plasma of patients with type 2 diabetes. Med Sci Monit 2005; 11(1):CR35–CR38.

    PubMed  CAS  Google Scholar 

  37. Kolak M, Westerbacka J, Velagapudi VR et al. Adipose tissue inflammation and increased ceramide content characterize subjects with high liver fat content independent of obesity. Diabetes 2007; 56(8):1960–1968.

    PubMed  CAS  Google Scholar 

  38. Wu D, Ren Z, Pae M et al. Aging up-regulates expression of inflammatory mediators in mouse adipose tissue. J Immunol 2007; 179(7):4829–4839.

    PubMed  CAS  Google Scholar 

  39. Hotamisligil GS, Spiegelman BM. Tumor necrosis factor α: A key component of the obesity-diabetes link. Diabetes 1994; 43:1271–1278.

    PubMed  CAS  Google Scholar 

  40. Chavez JA, Summers SA. Characterizing the effects of saturated fatty acids on insulin signaling and ceramide and diacylglycerol accumulation in 3T3-L1 adipocytes and C2C12 myotubes. Arch Biochem Biophys 2003; 419(2):101–109.

    PubMed  CAS  Google Scholar 

  41. Michailidou Z, Jensen MD, Dumesic DA et al. Omental 11beta-hydroxysteroid dehydrogenase 1 correlates with fat cell size independently of obesity. Obesity (Silver Spring) 2007; 15(5):1155–1163.

    CAS  Google Scholar 

  42. Kotelevtsev Y, Holmes MC, Burchell A et al 11beta-hydroxysteroid dehydrogenase type 1 knockout mice show attenuated glucocorticoid-inducible responses and resist hyperglycemia on obesity or stress. Proc Natl Acad Sci USA 1997; 94(26): 14924–14929.

    PubMed  CAS  Google Scholar 

  43. Masuzaki H, Paterson J, Shinyama H et al. A transgenic model of visceral obesity and the metabolic syndrome. Science 2001; 294(5549):2166–2170.

    PubMed  CAS  Google Scholar 

  44. Masuzaki H, Yamamoto H, Kenyon CJ et al. Transgenic amplification of glucocorticoid action in adipose tissue causes high blood pressure in mice. J Clin Invest 2003; 112(1):83–90.

    PubMed  CAS  Google Scholar 

  45. Arai N, Masuzaki H, Tanaka T et al. Ceramide and adenosine 5′-monophosphate-activated protein kinase are two novel regulators of 11beta-hydroxysteroid dehydrogenase type 1 expression and activity in cultured preadipocytes. Endocrinol 2007; 148(11):5268–5277.

    CAS  Google Scholar 

  46. Murray DK, Ruhmann-Wennhold A, Nelson DH. Dexamethasone effect on the phospholipid content of isolated fat cell ghosts from adrenalectomized rats. Endocrinol 1979; 105(3):774–777.

    CAS  Google Scholar 

  47. Johnston D, Matthews ER, Melnykovych G. Glucocorticoid effects on lipid metabolism in HeLa cells: inhibition of cholesterol synthesis and increased sphingomyelin synthesis. Endocrinol 1980; 107(5):1482–1488.

    CAS  Google Scholar 

  48. Murray DK, Ruhmann-Wennhold A, Nelson DH. Adrenalectomy decreases the sphingomyelin and cholesterol content of fat cell ghosts. Endocrinol 1982; 111(2):452–455.

    CAS  Google Scholar 

  49. Evans JL, Goldfine ID, Maddux BA et al. Oxidative stress and stress-activated signaling pathways: a unifying hypothesis of type 2 diabetes. Endocr Rev 2002; 23(5):599–622.

    PubMed  CAS  Google Scholar 

  50. Furukawa S, Fujita T, Shimabukuro M et al. Increased oxidative stress in obesity and its impact onmetabolic syndrome. J Clin Invest 2004; 114(12):1752–1761.

    PubMed  CAS  Google Scholar 

  51. Gregor MF, Hotamisligil GS. Thematic review series: Adipocyte Biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res 2007; 48(9): 1905–1914.

    PubMed  CAS  Google Scholar 

  52. Martinez JA. Mitochondrial oxidative stress and inflammation: an slalom to obesity and insulin resistance. J Physiol Biochem 2006; 62(4):303–306.

    PubMed  CAS  Google Scholar 

  53. Andrieu-Abadie N, Gouaze V, Salvayre R et al. Ceramide in apoptosis signaling: relationship with oxidative stress. Free Radic Biol Med 2001; 31(6):717–728.

    PubMed  CAS  Google Scholar 

  54. Birbes H, el BS, Obeid LM et al. Mitochondria and ceramide: intertwined roles in regulation of apoptosis. Adv Enzyme Regul 2002; 42:113–129.

    PubMed  CAS  Google Scholar 

  55. Novgorodov SA, Szulc ZM, Luberto C et al. Positively charged ceramide is apotent inducer of mitochondrial permeabilization. J Biol Chem 2005; 280(16): 16096–16105.

    PubMed  CAS  Google Scholar 

  56. Kashkar H, Wiegmann K, Yazdanpanah B et al. Acid sphingomyelinase is indispensable for UV light-induced Bax conformational change at the mitochondrial membrane. J Biol Chem 2005; 280(21):20804–20813.

    PubMed  CAS  Google Scholar 

  57. Birbes H, Luberto C, Hsu YT et al. A mitochondrial pool of sphingomyelin is involved in TNFalpha-induced Bax translocation to mitochondria. Biochem J 2005; 386(Pt 3):445–451.

    PubMed  CAS  Google Scholar 

  58. Sharpe JC, Arnoult D, Youle RJ. Control of mitochondrial permeability by Bcl-2 family members. Biochim Biophys Acta 2004; 1644(2–3):107–113.

    PubMed  CAS  Google Scholar 

  59. Zhang Y, Proenca R, Maffei M et al. Positional cloning of the mouse obese gene and its human homologue. Nature 1994; 372:425–432.

    PubMed  CAS  Google Scholar 

  60. Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature 1998; 395:763–770.

    PubMed  CAS  Google Scholar 

  61. Friedman JM, Leibel RL. Tackling a weighty problem. Cell 1992; 69:217–220.

    PubMed  CAS  Google Scholar 

  62. Hamann A, Matthaei S. Regulation of energy balance by leptin. Exper and Clin Endocrin and Diabetes 1996; 104:293–300.

    CAS  Google Scholar 

  63. Van Heek M, Compton DS, France CF et al. Diet-induced obese mice develop peripheral, but not central, resistance to leptin. J Clin Invest 1997; 99:385–390.

    Google Scholar 

  64. Heymsfield SB, Greenberg AS, Fujioka K et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA 1999; 282:1568–1575.

    PubMed  CAS  Google Scholar 

  65. Prpic V, Watson PM, Frampton IC et al. Differential mechanisms and development of leptin resistance in A/J versus c57BL/6J mice during diet-induced obesity. Endocrinol 2003; 144(4): 1155–1163.

    CAS  Google Scholar 

  66. Wang MY, Orci L, Ravazzola M et al. Fat storage in adipocytes requiRes inactivation of leptin’s paracrine activity: implications for treatment of human obesity. Proc Natl Acad Sci USA 2005; 102(50): 18011–18016.

    PubMed  CAS  Google Scholar 

  67. Hojjati MR, Li Z, Zhou H et al. Effect of myriocin on plasma sphingolipid metabolism and atherosclerosis in apoE-deficient mice. J Biol Chem 2005; 280(11):10284–10289.

    PubMed  CAS  Google Scholar 

  68. Park TS, Panek RL, Mueller SB et al. Inhibition of sphingomyelin synthesis reduces atherogenesis in apolipoprotein E-knockout mice. Circulation 2004; 110(22):3465–3471.

    PubMed  CAS  Google Scholar 

  69. Yang GD, Badeanlou L, Bielawski JD et al. Central role of ceramide biosynthesis in body weight regulation, energy metabolism and the metabolic syndrome. Am J Physiol Endocrinol Metab 2009.

    Google Scholar 

  70. Kanatani Y, Usui I, Ishizuka K et al. Effects of pioglitazone on suppressor of cytokine signaling 3 expression: potential mechanisms for its effects on insulin sensitivity and adiponectin expression. Diabetes 2007; 56(3):795–803.

    PubMed  CAS  Google Scholar 

  71. Howard JK, Flier JS. Attenuation of leptin and insulin signaling by SOCS proteins. Trends Endocrinol Metab 2006; 17(9):365–371.

    PubMed  CAS  Google Scholar 

  72. Steinberg GR, McAinch AJ, Chen MB et al. The suppressor of cytokine signaling 3 inhibits leptin activation of AMP-kinase in cultured skeletal muscle of obese humans. J Clin Endocrinol Metab 2006; 91(9):3592–3597.

    PubMed  CAS  Google Scholar 

  73. Cernkovich ER, Deng J, Bond MC et al. Adipose-specific disruption of signal transducer and activator of transcription 3 increases body weight and adiposity. Endocrinol 2008; 149(4): 1581–1590.

    CAS  Google Scholar 

  74. Wang ZW, Pan WT, Lee Y et al. The role of leptin resistance in the lipid abnormalities of aging. FASEB J 2001; 15(1):108–114.

    PubMed  CAS  Google Scholar 

  75. Orci L, Cook WS, Ravazzola M et al. Rapid transformation of white adipocytes into fat-oxidizing machines. Proc Natl Acad Sci USA 2004; 101(7):2058–2063.

    PubMed  CAS  Google Scholar 

  76. Park BH, Wang MY, Lee Y et al. Combined leptin actions on adipose tissue and hypothalamus are required to deplete adipocyte fat in lean rats: implications for obesity treatment. J Biol Chem 2006; 281(52):40283–40291.

    PubMed  CAS  Google Scholar 

  77. Unger RH, Zhou YT, Orci L. Regulation of fatty acid homeostasis in cells: novel role of leptin. Proc Natl Acad Sci USA 1999; 96(5):2327–2332.

    PubMed  CAS  Google Scholar 

  78. Martin TL, Alquier T, Asakura K et al. Diet-induced obesity alters AMP kinase activity in hypothalamus and skeletal muscle. J Biol Chem 2006; 281(28): 18933–18941.

    PubMed  CAS  Google Scholar 

  79. Nicholls DG, Locke RM. Thermogenic mechanisms in brown fat. Physiol Rev 1984; 64(l):1–64.

    PubMed  CAS  Google Scholar 

  80. Shimeno H, Soeda S, Sakamoto M et al. Partial purification and characterization of sphingosine N-acyltransferase (ceramide synthase) from bovine liver mitochondrion-rich fraction. Lipids 1998; 33(6):601–605.

    PubMed  CAS  Google Scholar 

  81. Santana P, Pena LA, Haimovitz-Friedman A et al. Acid sphingomyelinase-deficient human lymphoblasts and mice are defective in radiation-induced apoptosis. Cell 1996; 86(2):189–199.

    PubMed  CAS  Google Scholar 

  82. Bionda C, Portoukalian J, Schmitt D et al. Subcellular compartmentalization of ceramide metabolism: MaM (mitochondria-associated membrane) and/or mitochondria? Biochem J 2004; 382(Pt 2):527–533.

    PubMed  CAS  Google Scholar 

  83. Garcia-Ruiz C, Colell A, Mari M et al. Direct effect of ceramide on the mitochondrial electron transport chain leads to generation of reactive oxygen species. Role of mitochondrial glutathione. J Biol Chem 1997; 272(17):11369–11377.

    PubMed  CAS  Google Scholar 

  84. Soeda S, Honda O, Shimeno H et al. Sphingomyelinase and cell-permeable ceramide analogs increase the release of plasminogen activator inhibitor-1 from cultured endothelial cells. Thromb Res 1995; 80(6):509–518.

    PubMed  CAS  Google Scholar 

  85. Kimura M, Soeda S, Oda M et al. Release of plasminogen activator inhibitor-1 from human astrocytes is regulated by intracellular ceramide. J Neurosci Res 2000; 62:781–788.

    PubMed  CAS  Google Scholar 

  86. Holland WL, Summers SA. Sphingolipids, insulin resistance and metabolic disease: new insights from in vivo manipulation of sphingolipid metabolism. Endocr Rev 2008; 29(4):381–402.

    PubMed  CAS  Google Scholar 

  87. Summers SA, Garza LA, Zhou H et al. Regulation of insulin-stimulated glucose transporter GLUT4 translocation and Akt kinase activity by ceramide. Mol cell Biol 1998; 18:5457–5464.

    PubMed  CAS  Google Scholar 

  88. Nelson DH, Murray DK. Sphingolipids inhibit insulin and phorbol ester stimulated uptake of 2-deoxyglucose. Biochem Biophys Res Commun 1986; 138(1):463–467.

    PubMed  CAS  Google Scholar 

  89. Hajduch E, Balendran A, Batty IH et al. Ceramide impairs the insulin-dependent membrane recruitment of protein kinase B leading to a loss in downstream signalling in L6 skeletal muscle cells. Diabetologia 2001;44(2):173–183.

    PubMed  CAS  Google Scholar 

  90. Summers SA, Yin VP, Whiteman EL et al. Signaling pathways mediating insulin-stimulated glucose transport. Ann NY Acad Sci 1999; 892:169–186.

    PubMed  CAS  Google Scholar 

  91. Liu P, Leffler BJ, Weeks LK et al. Sphingomyelinase activates GLUT4 translocation via a cholesterol-dependent mechanism. Am J Physiol Cell Physiol 2004; 286(2):C317–C329.

    PubMed  CAS  Google Scholar 

  92. Summers SA, Kao AW, Kohn AD et al. The role of glycogen synthase kinase 3beta in insulin-stimulated glucose metabolism. J Biol Chem 1999; 274(25):17934–17940.

    PubMed  CAS  Google Scholar 

  93. Kralik SF, Liu P, Leffler BJ et al. Ceramide and glucosamine antagonism of alternate signaling pathways regulating insulin-and osmotic shock-induced glucose transporter 4 translocation. Endocrinol 2002; 143(1):37–46.

    CAS  Google Scholar 

  94. Paz K, Hemi R, LeRoith D et al.A molecular basis for insulin resistance. Elevated serine/threonine phosphorylation of IRS-1 and IRS-2 inhibits their binding to the juxtamembrane region of the insulin receptor and impairs their ability to undergo insulin-induced tyrosine phosphorylation. J Biol Chem 1997; 272(47):29911–29918.

    PubMed  CAS  Google Scholar 

  95. Kanety H, Hemi R, Papa MZ et al. Sphingomyelinase and ceramide suppress insulin-induced tyrosine phosphorylation of the insulin receptor substrate-1. J Biol Chem 1996; 271(17):9895–9897.

    PubMed  CAS  Google Scholar 

  96. Chavez JA, Knotts TA, Wang LP et al. A role for ceramide, but not diacylglycerol, in the antagonism of insulin signal transduction by saturated fatty acids. J Biol Chem 2003; 278(12):10297–10303.

    PubMed  CAS  Google Scholar 

  97. Bourbon NA, Sandirasegarane L, Kester M. Ceramide-induced inhibition of Akt is mediated through protein kinase Czeta: implications for growth arrest. J Biol Chem 2002; 277(5):3286–3292.

    PubMed  CAS  Google Scholar 

  98. Salinas M, Lopez-Valdaliso R, Martin D et al. Inhibition of PKB/Akt1 by C2-ceramide involves activation of ceramide-activated protein phosphatase in PC12 cells. Mol Cell Neurosci 2000; 15(2): 156–169.

    PubMed  CAS  Google Scholar 

  99. Schubertk M, Scheid MP, Duronio V. Ceramide inhibits protein kinaseB/Akt by promoting dephosphorylation of serine 473. J Biol Chem 2000; 275(18): 13330–13335.

    Google Scholar 

  100. Stratford S, De Wald DB, Summers SA. Ceramide dissociates 3′-phosphoinositide production from pleckstrin homology domain translocation. Biochem J 2001; 354(Pt 2):359–368.

    PubMed  CAS  Google Scholar 

  101. Stratford S, Hoehn KL, Liu F et al. Regulation of insulin action by ceramide: dual mechanisms linking ceramide accumulation to the inhibition of Akt/protein kinase B. J Biol Chem 2004; 279(35):36608–36615.

    PubMed  CAS  Google Scholar 

  102. Teruel T, Hernandez R, Lorenzo M. Ceramide mediates insulin resistance by tumor necrosis factor-alpha in brown adipocytes by maintaining Akt in an inactive dephosphorylated state. Diabetes 2001; 50:2563–2571.

    PubMed  CAS  Google Scholar 

  103. Zhou H, Summers SA, Birnbaum MJ et al. Inhibition of Akt kinase by cell-permeable ceramide and its implications for ceramide-induced apoptosis. J Biol Chem 1998; 273(26):16568–16575.

    PubMed  CAS  Google Scholar 

  104. Dobrowsky RT, Kamibayashi C, Mumby MC et al. Ceramide activates heterotrimeric protein phosphatase 2A. J Biol Chem 1993; 268(21):15523–15530.

    PubMed  CAS  Google Scholar 

  105. Resjo S, Goransson O, Harndahl L et al. Protein phosphatase 2A is the main phosphatase involved in the regulation of protein kinase B in rat adipocytes. Cell Signal 2002; 14(3):231–238.

    PubMed  CAS  Google Scholar 

  106. Powell DJ, Hajduch E, Kular G et al. Ceramide disables 3-phosphoinositide binding to the pleckstrin homology domain of protein kinase B (PKB)/Akt by a PKCzeta-dependent mechanism. Mol Cell Biol 2003;23(21):7794–7808.

    PubMed  CAS  Google Scholar 

  107. Powell DJ, Turban S, Gray A et al. Intracellular ceramide synthesis and protein kinase Czeta activation play an essential role in palmitate-induced insulin resistance in rat L6 skeletal muscle cells. Biochem J 2004; 382(Pt2):619–629.

    PubMed  CAS  Google Scholar 

  108. Yuan M, Konstantopoulos N, Lee J et al. Reversal of obesity-and diet-induced insulin resistance with salicylates or targeted disruption of Ikkbeta. Science 2001; 293(5535):1673–1677.

    PubMed  CAS  Google Scholar 

  109. Hirosumi J, Tuncman G, Chang L et al. A central role for JNK in obesity and insulin resistance. Nature 2002;420(6913):333–336.

    PubMed  CAS  Google Scholar 

  110. Ruvolo PP. Intracellular signal transduction pathways activated by ceramide and its metabolites. Pharmacol Res 2003; 47(5):383–392.

    PubMed  CAS  Google Scholar 

  111. Tagami S, Inokuchi J, Kabayama K et al. Ganglioside GM3 participates in the pathological conditions of insulin resistance. J Biol Chem 2002; 277:3085–3092.

    PubMed  CAS  Google Scholar 

  112. Kabayama K, Sato T, Kitamura F et al. TNFalpha-induced insulin resistance in adipocytes as a membrane microdomain disorder: involvement of ganglioside GM3. Glycobiology 2005; 15(1):21–29.

    PubMed  CAS  Google Scholar 

  113. Aerts JM, Ottenhoff R, Powlson AS et al. Pharmacological inhibition of glucosylceramide synthesis enhances insulin sensitivity. Diabetes 2007.

    Google Scholar 

  114. Yamashita T, Hashiramoto A, Haluzik M et al. Enhanced insulin sensitivity in mice lacking ganglioside GM3. Proc Natl Acad Sci USA 2003; 100(6):3445–3449.

    PubMed  CAS  Google Scholar 

  115. Donnelly KL, Smith CI, Schwarzenberg SJ et al. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest 2005; 115(5): 1343–1351.

    PubMed  CAS  Google Scholar 

  116. Greco D, Kotronen A, Westerbacka J et al. Gene expression in humanNAFLD. Am J Physiol Gastrointest Liver Physiol 2008; 294(5):G1281–G1287.

    PubMed  CAS  Google Scholar 

  117. Yetukuri L, Katajamaa M, Medina-Gomez G et al. Bioinformatics strategies for lipidomics analysis: characterization of obesity related hepatic steatosis. BMC Syst Biol 2007; 1:12.

    PubMed  Google Scholar 

  118. Kotronen A, Seppanen-Laakso T, Westerbacka J et al. Hepatic stearoyl-CoA desaturase (ScD)-1 activity and diacylglycerol but not ceramide concentrations are increased in the nonalcoholic human fatty liver. Diabetes 2009; 58(1):203–208.

    PubMed  CAS  Google Scholar 

  119. Deevska GM, Rozenova KA, Giltiay NV et al. Acid Sphingomyelinase Deficiency Prevents Diet-induced hepatic triacylglycerol accumulation and hyperglycemia in Mice. J Biol Chem 2009; 284(13):8359–8368.

    PubMed  CAS  Google Scholar 

  120. Ueki K, Kondo T, Tseng YH et al. Central role of suppressors of cytokine signaling proteins in hepatic steatosis, insulin resistance and the metabolic syndrome in the mouse. Proc Natl Acad Sci USA 2004; 101(28):10422–10427.

    PubMed  CAS  Google Scholar 

  121. Krauss RM, Winston M, Fletcher RN et al. Obesity: impact of cardiovascular disease. Circulation 1998; 98(14): 1472–1476.

    CAS  Google Scholar 

  122. Lavie CJ, Milani RV, Ventura HO. Obesity and cardiovascular disease: risk factor, paradox and impact of weight loss. J Am Coll Cardiol 2009; 53(21): 1925–1932.

    PubMed  Google Scholar 

  123. Vaughan DE. Plasminogen activator inhibitor-1: A common denominator in cardiovascular disease. J Invest Med 1998; 46:370–376.

    CAS  Google Scholar 

  124. Chatterjee S. Sphingolipids in atherosclerosis and vascular biology. Arterioscler Thromb Vasc Biol 1998; 18(10):1523–1533.

    PubMed  CAS  Google Scholar 

  125. Auge N, Negre-Salvayre A, Salvayre R et al. Sphingomyelin metabolites in vascular cell signaling and atherogenesis. Prog Lipid Res 2000; 39(3):207–229.

    PubMed  CAS  Google Scholar 

  126. Li H, Junk P, Huwiler A et al. Dual effect of ceramide on human endothelial cells: induction of oxidative stress and transcriptional upregulation of endothelial nitric oxide synthase. Circulation 2002; 106(17):2250–2256.

    PubMed  CAS  Google Scholar 

  127. Auge N, Maupas-Schwalm F, Elbaz M et al. Role for matrix metalloproteinase-2 in oxidized low-density lipoprotein-induced activation of the sphingomyelin/ceramide pathway and smoothmuscle cell proliferation. Circulation 2004; 110(5):571–578.

    PubMed  CAS  Google Scholar 

  128. Nixon GF, Mathieson FA, Hunter I. The potential roles of sphingolipids in vascular smooth-muscle function. Biochem Soc Trans 2007; 35(Pt 5):908–909.

    PubMed  CAS  Google Scholar 

  129. Li Z, Basterr MJ, Hailemariam TK et al. The effect of dietary sphingolipids on plasma sphingomyelin metabolism and atherosclerosis. Biochim Biophys Acta 2005; 1735(2):130–134.

    PubMed  CAS  Google Scholar 

  130. Jiang XC, Paultre F, Pearson TA et al. Plasma sphingomyelin level as a risk factor for coronary artery disease. Arterioscler Thromb Vasc Biol 2000; 20:2614–2618.

    PubMed  CAS  Google Scholar 

  131. Paumen MB, Ishida Y, Muramatsu M et al. Inhibition of carnitine palmitoyltransferase I augments sphingolipid synthesis and palmitate-induced apoptosis. J Biol Chem 1997; 272(6):3324–3329.

    PubMed  CAS  Google Scholar 

  132. De Vries JE, Vork MM, Roemen TH et al. Saturated but not mono-unsaturated fatty acids induce apoptotic cell death in neonatal rat ventricular myocytes. J Lipid Res 1997; 38(7): 1384–1394.

    PubMed  Google Scholar 

  133. Dyntar D, Eppenberger-Eberhardt M, Maedler K et al. Glucose and palmitic acid induce degeneration of myofibrils and modulate apoptosis in rat adult cardiomyocytes. Diabetes 2001; 50(9):2105–2113.

    PubMed  CAS  Google Scholar 

  134. Ren J, Relling DP. Leptin-induced suppression of cardiomyocyte contraction is amplified by ceramide. Peptides 2006; 27(6): 1415–1419.

    PubMed  CAS  Google Scholar 

  135. Marathe S, Choi Y, Leventhal AR et al. Sphingomyelinase converts lipoproteins from apolipoprotein E knockout mice into potent iducers of macrophage foam cell formation. Arterioscler Thromb Vasc Biol 2000; 20:2607–2613.

    PubMed  CAS  Google Scholar 

  136. Schissel SL, Tweedie-Hardman J, Rapp JH et al. Tabas I. rabbit aorta and human atherosclerotic lesions hydrolyze the sphingomyelin of retained low-density lipoprotein. Proposed role for arterial-wall sphingomyelinase in subendothelial retention and aggregation of atherogenic lipoproteins. J Clin Invest 1996; 98(6): 1455–1464.

    PubMed  CAS  Google Scholar 

  137. Mitchinson MJ, Hardwick SJ, Bennett MR. Cell death in atherosclerotic plaques. Curr Opin Lipidol 1996; 7(5):324–329.

    PubMed  CAS  Google Scholar 

  138. Mallat Z, Tedgui A. Apoptosis in the vasculature: mechanisms and functional importance. Br J Pharmacol 2000; 130(5):947–962.

    PubMed  CAS  Google Scholar 

  139. Spiegel S, Milstien S. Sphingosine-1-phosphate: an enigmatic signalling lipid. Nat rev Mol cell Biol 2003; 4(5):397–407.

    PubMed  CAS  Google Scholar 

  140. Alewijnse AE, Peters SL, Michel MC. Cardiovascular effects of sphingosine-1-phosphate and other sphingomyelin metabolites. Br J Pharmacol 2004; 143(6):666–684.

    PubMed  CAS  Google Scholar 

  141. Yatomi Y, Ohmori T, Rile G et al. Sphingosine 1-phosphate as a major bioactive lysophospholipid that is released from platelets and interacts with endothelial cells. Blood 2000; 96(10):3431–3438.

    PubMed  CAS  Google Scholar 

  142. Murata N, Sato K, Kon J et al. Interaction of sphingosine 1-phosphate with plasma components, including lipoproteins, regulates the lipid receptor-mediated actions. Biochem J 2000; 352 Pt 3:809–815.

    PubMed  CAS  Google Scholar 

  143. Nofer JR, van der GM, Tolle M et al. HDL induces NO-dependent vasorelaxation via the lysophospholipid receptor S1P3. J Clin Invest 2004; 113(4):569–581.

    PubMed  CAS  Google Scholar 

  144. Karliner JS. Toward solving the riddle: the enigma becomes less mysterious. Circ Res 2006; 99(5):465–467.

    PubMed  CAS  Google Scholar 

  145. Auge N, Nikolova-Karakashian M, Carpentier S et al. Role of sphingosine 1-phosphate in the mitogenesis induced by oxidized low density lipoprotein in smooth muscle cells via activation of sphingomyelinase, ceramidase and sphingosine kinase. J Biol Chem 1999; 274(31):21533–21538.

    PubMed  CAS  Google Scholar 

  146. Ipatova OM, Torkhovskaya TI, Zakharova TS et al. Sphingolipids and cell signaling: involvement in apoptosis and atherogenesis. Biochemistry (Mosc) 2006; 71(7):713–722.

    CAS  Google Scholar 

  147. Xia P, Gamble JR, Rye KA et al. Tumor necrosis factor-alpha induces adhesion molecule expression through the sphingosine kinase pathway. Proc Natl Acad Sci USA 1998; 95(24):14196–14201.

    PubMed  CAS  Google Scholar 

  148. Bot M, Nofer JR, van Berkel TJC et al. Lysophospholipids: two-faced mediators in atherosclerosis. Future Lipidol 2007; 2(3): 341–356.

    CAS  Google Scholar 

  149. Karliner JS, Honbo N, Summers K et al. The lysophospholipids sphingosine-1-phosphate and lysophosphatidic acid enhance survival during hypoxia in neonatal rat cardiac myocytes. J Mol cell Cardiol 2001;33(9):1713–1717.

    PubMed  CAS  Google Scholar 

  150. Jin ZQ, Zhou HZ, Zhu P et al. Cardioprotection mediated by sphingosine-1-phosphate and ganglioside GM-1 in wild-type and PKC epsilon knockout mouse hearts. Am J Physiol Heart Circ Physiol 2002; 282(6):H1970–H1977.

    PubMed  CAS  Google Scholar 

  151. Lecour S, Smith RM, Woodward B et al. Identification of a novel role for sphingolipid signaling in TNF alpha and ischemic preconditioning mediated cardioprotection. J Mol Cell Cardiol 2002; 34(5):509–518.

    PubMed  CAS  Google Scholar 

  152. Nofer JR, Assmann G. Atheroprotective effects of high-density lipoprotein-associated lysosphingolipids. Trends Cardiovasc Med 2005; 15(7):265–271.

    PubMed  CAS  Google Scholar 

  153. Kimura T, Sato K, Kuwabara A et al. Sphingosine 1-phosphate may be a major component of plasma lipoproteins responsible for the cytoprotective actions in human umbilical vein endothelial cells. J Biol Chem 2001; 276(34):31780–31785.

    PubMed  CAS  Google Scholar 

  154. Kimura T, Sato K, Malchinkhuu E et al. High-density lipoprotein stimulates endothelial cell migration and survival through sphingosine 1-phosphate and its receptors. Arterioscler Thromb Vasc Biol 2003;23(7):1283–1288.

    PubMed  CAS  Google Scholar 

  155. Nofer JR, Bot M, Brodde M et al. FTY720, a synthetic sphingosine 1 phosphate analogue, inhibits development of atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation 2007; 115(4):501–508.

    PubMed  CAS  Google Scholar 

  156. Keul P, Tolle M, Lucke S et al. The sphingosine-1-phosphate analogue FTY720 reduces atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2007; 27(3):607–613.

    PubMed  CAS  Google Scholar 

  157. Kahan BD. Update on pharmacokinetic/pharmacodynamic studies with FTY720 and sirolimus. Ther Drug Monit 2002; 24(1):47–52.

    PubMed  CAS  Google Scholar 

  158. Kovarik JM, Schmouder RL, Slade AJ. Overview of FTY720 clinical pharmacokinetics and pharmacology. Ther Drug Monit 2004; 26(6):585–587.

    PubMed  CAS  Google Scholar 

  159. Pettus BJ, Bielawski J, Porcelli AM et al. The sphingosine kinase 1/sphingosine-1-phospate pathway mediates COX-2 induction and PGE2 production in response to TNf-α. FASEB J 2003; 17:1411–1421.

    PubMed  CAS  Google Scholar 

  160. Pettus BJ, Kitatani K, Chalfant CE et al. The coordination of prostaglandin E2 production by sphingosine-1-phosphate and ceramide-1-phosphate. Mol Pharmacol 2005; 68(2):330–335.

    PubMed  CAS  Google Scholar 

  161. Cipollone F, Fazia ML. COX-2 and atherosclerosis. J Cardiovasc Pharmacol 2006; 47(Suppl 1):S26–S36.

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2011 Landes Bioscience and Springer Science+Business Media, LLC

About this chapter

Cite this chapter

Samad, F., Badeanlou, L., Shah, C., Yang, G. (2011). Adipose Tissue and Ceramide Biosynthesis in the Pathogenesis of Obesity. In: Cowart, L.A. (eds) Sphingolipids and Metabolic Disease. Advances in Experimental Medicine and Biology, vol 721. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-0650-1_5

Download citation

Publish with us

Policies and ethics