J Lipid Atheroscler.  2020 Jan;9(1):50-65. 10.12997/jla.2020.9.1.50.

Ceramides: Nutrient Signals that Drive Hepatosteatosis

Affiliations
  • 1Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA. scott.a.summers@health.utah.edu

Abstract

Ceramides are minor components of the hepatic lipidome that have major effects on liver function. These products of lipid and protein metabolism accumulate when the energy needs of the hepatocyte have been met and its storage capacity is full, such that free fatty acids start to couple to the sphingoid backbone rather than the glycerol moiety that is the scaffold for glycerolipids (e.g., triglycerides) or the carnitine moiety that shunts them into mitochondria. As ceramides accrue, they initiate actions that protect cells from acute increases in detergent-like fatty acids; for example, they alter cellular substrate preference from glucose to lipids and they enhance triglyceride storage. When prolonged, these ceramide actions cause insulin resistance and hepatic steatosis, 2 of the underlying drivers of cardiometabolic diseases. Herein the author discusses the mechanisms linking ceramides to the development of insulin resistance, hepatosteatosis and resultant cardiometabolic disorders.

Keyword

Ceramides; Steatohepatitis; Insulin resistance; Non-alcoholic fatty liver disease; Diabetes

MeSH Terms

Carnitine
Ceramides*
Fatty Acids
Fatty Acids, Nonesterified
Fatty Liver
Glucose
Glycerol
Hepatocytes
Insulin Resistance
Liver
Metabolism
Mitochondria
Non-alcoholic Fatty Liver Disease
Triglycerides
Carnitine
Ceramides
Fatty Acids
Fatty Acids, Nonesterified
Glucose
Glycerol

Figure

  • Fig. 1 Schematic depicting the key reactions in the ceramide biosynthesis pathway. CoA, coenzyme A; SPT, serine palmitoyltransferase; KDHR, 3-ketodihydrosphingosine reductase; CERS, (dihydro)ceramide synthases (isoforms in parentheses); DES, dihydroceramide desaturase (isoforms in parentheses); GCS, glucosyceramide synthase; SMS, sphingomyelin synthase (isoforms in parentheses); CERK, ceramide kinase.

  • Fig. 2 Schematic depicting the pathways that incorporate fatty acids entering the cell. (A) When detergent-like fatty acid levels get too high, they emulsify cellular membranes. The lysis occurs over 3 stages: fatty acid penetration into the bilayer; their equilibration into both faces of the bilayer; and the transition of the bilayer into a micelle. (B) To prevent lysis, fatty acids entering the cell are quickly coupled to CoA and then incorporated into backbones to produce acylcarnitines, glycerolipids, and sphingolipids. Acylcarnitines are formed as fatty acids translocate into mitochondria. Glycerolipids that are produced make up the bulk of membrane bilayers and create the triglyceride stores that form the majority of the lipid droplet. Sphingolipids are signals of lipid excess that signal to upregulate the other 2 pathways under conditions of lipid excess. CoA, coenzyme A.

  • Fig. 3 Schematic depicting the means by which ceramides influence cellular metabolism. Mechanisms described in the text and depicted in greater detail in Fig. 4. SM, sphingomyelin; GLUT4, the insulin-responsive glucose transporter; Akt, the serine threonine kinase Akt/PKB; HSL, hormone-sensitive lipase; SREBP, sterol response element binding protein; MFF, mitochondrial fission factor; BAX, the pro-apoptotic BCL2 family member BAX; FFA, free fatty acid. Green denotes a process that is increased and red denotes decreased.

  • Fig. 4 Schematic depicting the mechanisms of ceramide action. MFF, mitochondrial fission factor; PKCζ, protein kinase C-zeta; CREB3L1, cAMP responsive element binding protein 3 like 1; PP2A, protein phosphatase 2A; BAX, the pro-apoptotic BCL2 family member BAX; Akt, the serine threonine kinase Akt/PKB; SREBP, sterol response element binding protein; HSL, hormone sensitive lipase.

  • Fig. 5 Schematic depicting the interventions that have been shown to ameliorate cardiometabolic disorders. Pharmaceutical interventions or genetic knockout/knockdown of ceramide-synthesizing genes improves insulin sensitivity, resolves hepatic steatosis, and prevents the development of resultant metabolic disorders including diabetes, cardiomyopathy, coronary artery disease, hypertension, and heart failure. Sptlc2, Serine palmitoyltransferase subunit 2; Cers, ceramide synthase genes; Degs1, dihydroceramide desaturase-1 gene; CoA, coenzyme A.


Reference

1. Cahill GF Jr. Starvation in man. Clin Endocrinol Metab. 1976; 5:397–415.
Article
2. Chaurasia B, Tippetts TS, Mayoral Monibas R, Liu J, Li Y, Wang L, et al. Targeting a ceramide double bond improves insulin resistance and hepatic steatosis. Science. 2019; 365:386–392.
Article
3. Merrill AH Jr. De novo sphingolipid biosynthesis: a necessary, but dangerous, pathway. J Biol Chem. 2002; 277:25843–25846.
4. Lone MA, Santos T, Alecu I, Silva LC, Hornemann T. 1-Deoxysphingolipids. Biochim Biophys Acta Mol Cell Biol Lipids. 2019; 1864:512–521.
Article
5. Davis D, Kannan M, Wattenberg B. Orm/ORMDL proteins: gate guardians and master regulators. Adv Biol Regul. 2018; 70:3–18.
Article
6. Cingolani F, Futerman AH, Casas J. Ceramide synthases in biomedical research. Chem Phys Lipids. 2016; 197:25–32.
Article
7. Park JW, Park WJ, Futerman AH. Ceramide synthases as potential targets for therapeutic intervention in human diseases. Biochim Biophys Acta. 2014; 1841:671–681.
Article
8. Raichur S, Wang ST, Chan PW, Li Y, Ching J, Chaurasia B, et al. Cers2 haploinsufficiency inhibits β-oxidation and confers susceptibility to diet-induced steatohepatitis and insulin resistance. Cell Metab. 2014; 20:687–695.
Article
9. Raichur S, Brunner B, Bielohuby M, Hansen G, Pfenninger A, Wang B, et al. The role of C16:0 ceramide in the development of obesity and type 2 diabetes: Cers6 inhibition as a novel therapeutic approach. Mol Metab. 2019; 21:36–50.
Article
10. Turpin SM, Nicholls HT, Willmes DM, Mourier A, Brodesser S, Wunderlich CM, et al. Obesity-induced Cers6-dependent C16:0 ceramide production promotes weight gain and glucose intolerance. Cell Metab. 2014; 20:678–686.
Article
11. Russo SB, Baicu CF, Van Laer A, Geng T, Kasiganesan H, Zile MR, et al. Ceramide synthase 5 mediates lipid-induced autophagy and hypertrophy in cardiomyocytes. J Clin Invest. 2012; 122:3919–3930.
Article
12. Hammerschmidt P, Ostkotte D, Nolte H, Gerl MJ, Jais A, Brunner HL, et al. Cers6-derived sphingolipids interact with MFF and promote mitochondrial fragmentation in obesity. Cell. 2019; 177:1536–1552.e23.
Article
13. Turpin-Nolan SM, Hammerschmidt P, Chen W, Jais A, Timper K, Awazawa M, et al. Cers1-derived C18:0 ceramide in skeletal muscle promotes obesity-induced insulin resistance. Cell Rep. 2019; 26:1–10.e7.
14. Sociale M, Wulf AL, Breiden B, Klee K, Thielisch M, Eckardt F, et al. Ceramide synthase schlank is a transcriptional regulator adapting gene expression to energy requirements. Cell Reports. 2018; 22:967–978.
Article
15. Pillai BK, Jasuja R, Simard JR, Hamilton JA. Fast diffusion of very long chain saturated fatty acids across a bilayer membrane and their rapid extraction by cyclodextrins: implications for adrenoleukodystrophy. J Biol Chem. 2009; 284:33296–33304.
Article
16. Khan MA, Bishop RE. Molecular mechanism for lateral lipid diffusion between the outer membrane external leaflet and a beta-barrel hydrocarbon ruler. Biochemistry. 2009; 48:9745–9756.
Article
17. Brunaldi K, Huang N, Hamilton JA. Fatty acids are rapidly delivered to and extracted from membranes by methyl-beta-cyclodextrin. J Lipid Res. 2010; 51:120–131.
Article
18. Guo W, Huang N, Cai J, Xie W, Hamilton JA. Fatty acid transport and metabolism in HepG2 cells. Am J Physiol Gastrointest Liver Physiol. 2006; 290:G528–G534.
Article
19. Hamilton JA, Johnson RA, Corkey B, Kamp F. Fatty acid transport: the diffusion mechanism in model and biological membranes. J Mol Neurosci. 2001; 16:99–108.
Article
20. Glatz JF. Lipids and lipid binding proteins: a perfect match. Prostaglandins Leukot Essent Fatty Acids. 2015; 93:45–49.
Article
21. Jay AG, Hamilton JA. The enigmatic membrane fatty acid transporter CD36: new insights into fatty acid binding and their effects on uptake of oxidized LDL. Prostaglandins Leukot Essent Fatty Acids. 2018; 138:64–70.
Article
22. Xu S, Jay A, Brunaldi K, Huang N, Hamilton JA. CD36 enhances fatty acid uptake by increasing the rate of intracellular esterification but not transport across the plasma membrane. Biochemistry. 2013; 52:7254–7261.
Article
23. Xia JY, Holland WL, Kusminski CM, Sun K, Sharma AX, Pearson MJ, et al. Targeted induction of ceramide degradation leads to improved systemic metabolism and reduced hepatic steatosis. Cell Metab. 2015; 22:266–278.
Article
24. Ehehalt R, Sparla R, Kulaksiz H, Herrmann T, Füllekrug J, Stremmel W. Uptake of long chain fatty acids is regulated by dynamic interaction of FAT/CD36 with cholesterol/sphingolipid enriched microdomains (lipid rafts). BMC Cell Biol. 2008; 9:45.
Article
25. Pohl J, Ring A, Ehehalt R, Schulze-Bergkamen H, Schad A, Verkade P, et al. Long-chain fatty acid uptake into adipocytes depends on lipid raft function. Biochemistry. 2004; 43:4179–4187.
Article
26. Covey SD, Brunet RH, Gandhi SG, McFarlane N, Boreham DR, Gerber GE, et al. Cholesterol depletion inhibits fatty acid uptake without affecting CD36 or caveolin-1 distribution in adipocytes. Biochem Biophys Res Commun. 2007; 355:67–71.
Article
27. Jiang C, Xie C, Li F, Zhang L, Nichols RG, Krausz KW, et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J Clin Invest. 2015; 125:386–402.
Article
28. Worgall TS, Juliano RA, Seo T, Deckelbaum RJ. Ceramide synthesis correlates with the posttranscriptional regulation of the sterol-regulatory element-binding protein. Arterioscler Thromb Vasc Biol. 2004; 24:943–948.
Article
29. Taniguchi CM, Kondo T, Sajan M, Luo J, Bronson R, Asano T, et al. Divergent regulation of hepatic glucose and lipid metabolism by phosphoinositide 3-kinase via Akt and PKClambda/zeta. Cell Metab. 2006; 3:343–353.
Article
30. Chen TC, Lee RA, Tsai SL, Kanamaluru D, Gray NE, Yiv N, et al. An ANGPTL4-ceramide-protein kinase Cζ axis mediates chronic glucocorticoid exposure-induced hepatic steatosis and hypertriglyceridemia in mice. J Biol Chem. 2019; 294:9213–9224.
Article
31. Summers SA, Garza LA, Zhou H, Birnbaum MJ. Regulation of insulin-stimulated glucose transporter GLUT4 translocation and Akt kinase activity by ceramide. Mol Cell Biol. 1998; 18:5457–5464.
Article
32. Wang CN, O'Brien L, Brindley DN. Effects of cell-permeable ceramides and tumor necrosis factor-alpha on insulin signaling and glucose uptake in 3T3-L1 adipocytes. Diabetes. 1998; 47:24–31.
Article
33. Hyde R, Hajduch E, Powell DJ, Taylor PM, Hundal HS. Ceramide down-regulates System A amino acid transport and protein synthesis in rat skeletal muscle cells. FASEB J. 2005; 19:461–463.
Article
34. Finicle BT, Ramirez MU, Liu G, Selwan EM, McCracken AN, Yu J, et al. Sphingolipids inhibit endosomal recycling of nutrient transporters by inactivating ARF6. J Cell Sci. 2018; 131:jcs213314.
Article
35. Guenther GG, Peralta ER, Rosales KR, Wong SY, Siskind LJ, Edinger AL. Ceramide starves cells to death by downregulating nutrient transporter proteins. Proc Natl Acad Sci U S A. 2008; 105:17402–17407.
Article
36. Edinger AL. Starvation in the midst of plenty: making sense of ceramide-induced autophagy by analysing nutrient transporter expression. Biochem Soc Trans. 2009; 37:253–258.
Article
37. Cowart LA, Obeid LM. Yeast sphingolipids: recent developments in understanding biosynthesis, regulation, and function. Biochim Biophys Acta. 2007; 1771:421–431.
Article
38. Chung N, Mao C, Heitman J, Hannun YA, Obeid LM. Phytosphingosine as a specific inhibitor of growth and nutrient import in Saccharomyces cerevisiae . J Biol Chem. 2001; 276:35614–35621.
Article
39. Hajduch E, Balendran A, Batty IH, Litherland GJ, Blair AS, Downes CP, 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; 44:173–183.
Article
40. Stratford S, DeWald DB, Summers SA. Ceramide dissociates 3′-phosphoinositide production from pleckstrin homology domain translocation. Biochem J. 2001; 354:359–368.
Article
41. Stratford S, Hoehn KL, Liu F, Summers SA. Regulation of insulin action by ceramide: dual mechanisms linking ceramide accumulation to the inhibition of Akt/protein kinase B. J Biol Chem. 2004; 279:36608–36615.
42. Powell DJ, Hajduch E, Kular G, Hundal HS. Ceramide disables 3-phosphoinositide binding to the pleckstrin homology domain of protein kinase B (PKB)/Akt by a PKCζ-dependent mechanism. Mol Cell Biol. 2003; 23:7794–7808.
Article
43. Chavez JA, Knotts TA, Wang LP, Li G, Dobrowsky RT, Florant GL, et al. A role for ceramide, but not diacylglycerol, in the antagonism of insulin signal transduction by saturated fatty acids. J Biol Chem. 2003; 278:10297–10303.
Article
44. Salinas M, López-Valdaliso R, Martín D, Alvarez A, Cuadrado A. Inhibition of PKB/Akt1 by C2-ceramide involves activation of ceramide-activated protein phosphatase in PC12 cells. Mol Cell Neurosci. 2000; 15:156–169.
Article
45. 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.
Article
46. Zinda MJ, Vlahos CJ, Lai MT. Ceramide induces the dephosphorylation and inhibition of constitutively activated Akt in PTEN negative U87mg cells. Biochem Biophys Res Commun. 2001; 280:1107–1115.
Article
47. Bourbon NA, Sandirasegarane L, Kester M. Ceramide-induced inhibition of Akt is mediated through protein kinase Cζ: implications for growth arrest. J Biol Chem. 2002; 277:3286–3292.
48. Fox TE, Houck KL, O'Neill SM, Nagarajan M, Stover TC, Pomianowski PT, et al. Ceramide recruits and activates protein kinase C ζ (PKCζ) within structured membrane microdomains. J Biol Chem. 2007; 282:12450–12457.
Article
49. Hajduch E, Turban S, Le Liepvre X, Le Lay S, Lipina C, Dimopoulos N, et al. Targeting of PKCζ and PKB to caveolin-enriched microdomains represents a crucial step underpinning the disruption in PKB-directed signalling by ceramide. Biochem J. 2008; 410:369–379.
Article
50. Dey D, Basu D, Roy SS, Bandyopadhyay A, Bhattacharya S. Involvement of novel PKC isoforms in FFA induced defects in insulin signaling. Mol Cell Endocrinol. 2006; 246:60–64.
Article
51. Blouin CM, Prado C, Takane KK, Lasnier F, Garcia-Ocana A, Ferré P, et al. Plasma membrane subdomain compartmentalization contributes to distinct mechanisms of ceramide action on insulin signaling. Diabetes. 2010; 59:600–610.
Article
52. Gudz TI, Tserng KY, Hoppel CL. Direct inhibition of mitochondrial respiratory chain complex III by cell-permeable ceramide. J Biol Chem. 1997; 272:24154–24158.
Article
53. Di Paola M, Cocco T, Lorusso M. Ceramide interaction with the respiratory chain of heart mitochondria. Biochemistry. 2000; 39:6660–6668.
Article
54. Zigdon H, Kogot-Levin A, Park JW, Goldschmidt R, Kelly S, Merrill AH Jr, et al. Ablation of ceramide synthase 2 causes chronic oxidative stress due to disruption of the mitochondrial respiratory chain. J Biol Chem. 2013; 288:4947–4956.
Article
55. Park M, Kaddai V, Ching J, Fridianto KT, Sieli RJ, Sugii S, et al. A role for ceramides, but not sphingomyelins, as antagonists of insulin signaling and mitochondrial metabolism in C2C12 myotubes. J Biol Chem. 2016; 291:23978–23988.
Article
56. Wang X, Rao RP, Kosakowska-Cholody T, Masood MA, Southon E, Zhang H, et al. Mitochondrial degeneration and not apoptosis is the primary cause of embryonic lethality in ceramide transfer protein mutant mice. J Cell Biol. 2009; 184:143–158.
Article
57. Turner N, Lim XY, Toop HD, Osborne B, Brandon AE, Taylor EN, et al. A selective inhibitor of ceramide synthase 1 reveals a novel role in fat metabolism. Nat Commun. 2018; 9:3165.
Article
58. Smith ME, Tippetts TS, Brassfield ES, Tucker BJ, Ockey A, Swensen AC, et al. Mitochondrial fission mediates ceramide-induced metabolic disruption in skeletal muscle. Biochem J. 2013; 456:427–439.
Article
59. Obeid LM, Linardic CM, Karolak LA, Hannun YA. Programmed cell death induced by ceramide. Science. 1993; 259:1769–1771.
Article
60. Ganesan V, Perera MN, Colombini D, Datskovskiy D, Chadha K, Colombini M. Ceramide and activated Bax act synergistically to permeabilize the mitochondrial outer membrane. Apoptosis. 2010; 15:553–562.
Article
61. Dadsena S, Bockelmann S, Mina JG, Hassan DG, Korneev S, Razzera G, et al. Ceramides bind VDAC2 to trigger mitochondrial apoptosis. Nat Commun. 2019; 10:1832.
Article
62. Siskind LJ, Kolesnick RN, Colombini M. Ceramide forms channels in mitochondrial outer membranes at physiologically relevant concentrations. Mitochondrion. 2006; 6:118–125.
Article
63. Siskind LJ, Davoody A, Lewin N, Marshall S, Colombini M. Enlargement and contracture of C2-ceramide channels. Biophys J. 2003; 85:1560–1575.
Article
64. Siskind LJ, Kolesnick RN, Colombini M. Ceramide channels increase the permeability of the mitochondrial outer membrane to small proteins. J Biol Chem. 2002; 277:26796–26803.
Article
65. Siskind LJ, Colombini M. The lipids C2- and C16-ceramide form large stable channels. Implications for apoptosis. J Biol Chem. 2000; 275:38640–38644.
66. Perera MN, Lin SH, Peterson YK, Bielawska A, Szulc ZM, Bittman R, et al. Bax and Bcl-xL exert their regulation on different sites of the ceramide channel. Biochem J. 2012; 445:81–91.
Article
67. Ganesan V, Colombini M. Regulation of ceramide channels by Bcl-2 family proteins. FEBS Lett. 2010; 584:2128–2134.
Article
68. Siskind LJ, Feinstein L, Yu T, Davis JS, Jones D, Choi J, et al. Anti-apoptotic Bcl-2 family proteins disassemble ceramide channels. J Biol Chem. 2008; 283:6622–6630.
Article
69. Chen Q, Denard B, Lee CE, Han S, Ye JS, Ye J. Inverting the topology of a transmembrane protein by regulating the translocation of the first transmembrane helix. Mol Cell. 2016; 63:567–578.
Article
70. Chen Q, Lee CE, Denard B, Ye J. Sustained induction of collagen synthesis by TGF-β requires regulated intramembrane proteolysis of CREB3L1. PLoS One. 2014; 9:e108528.
Article
71. Denard B, Lee C, Ye J. Doxorubicin blocks proliferation of cancer cells through proteolytic activation of CREB3L1. eLife. 2012; 1:e00090.
Article
72. Brown MS, Goldstein JL. Selective versus total insulin resistance: a pathogenic paradox. Cell Metab. 2008; 7:95–96.
Article
73. Park TS, Rosebury W, Kindt EK, Kowala MC, Panek RL. Serine palmitoyltransferase inhibitor myriocin induces the regression of atherosclerotic plaques in hyperlipidemic ApoE-deficient mice. Pharmacol Res. 2008; 58:45–51.
Article
74. Glaros EN, Kim WS, Quinn CM, Jessup W, Rye KA, Garner B. Myriocin slows the progression of established atherosclerotic lesions in apolipoprotein E gene knockout mice. J Lipid Res. 2008; 49:324–331.
Article
75. Glaros EN, Kim WS, Wu BJ, Suarna C, Quinn CM, Rye KA, et al. Inhibition of atherosclerosis by the serine palmitoyl transferase inhibitor myriocin is associated with reduced plasma glycosphingolipid concentration. Biochem Pharmacol. 2007; 73:1340–1346.
Article
76. Park TS, Panek RL, Rekhter MD, Mueller SB, Rosebury WS, Robertson A, et al. Modulation of lipoprotein metabolism by inhibition of sphingomyelin synthesis in ApoE knockout mice. Atherosclerosis. 2006; 189:264–272.
Article
77. Hojjati MR, Li Z, Zhou H, Tang S, Huan C, Ooi E, et al. Effect of myriocin on plasma sphingolipid metabolism and atherosclerosis in apoE-deficient mice. J Biol Chem. 2005; 280:10284–10289.
Article
78. Park TS, Panek RL, Mueller SB, Hanselman JC, Rosebury WS, Robertson AW, et al. Inhibition of sphingomyelin synthesis reduces atherogenesis in apolipoprotein E-knockout mice. Circulation. 2004; 110:3465–3471.
Article
79. Holland WL, Brozinick JT, Wang LP, Hawkins ED, Sargent KM, Liu Y, et al. Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance. Cell Metab. 2007; 5:167–179.
Article
80. Dekker MJ, Baker C, Naples M, Samsoondar J, Zhang R, Qiu W, et al. Inhibition of sphingolipid synthesis improves dyslipidemia in the diet-induced hamster model of insulin resistance: evidence for the role of sphingosine and sphinganine in hepatic VLDL-apoB100 overproduction. Atherosclerosis. 2013; 228:98–109.
Article
81. Ussher JR, Koves TR, Cadete VJ, Zhang L, Jaswal JS, Swyrd SJ, et al. Inhibition of de novo ceramide synthesis reverses diet-induced insulin resistance and enhances whole-body oxygen consumption. Diabetes. 2010; 59:2453–2464.
Article
82. Yang G, Badeanlou L, Bielawski J, Roberts AJ, Hannun YA, Samad F. Central role of ceramide biosynthesis in body weight regulation, energy metabolism, and the metabolic syndrome. Am J Physiol Endocrinol Metab. 2009; 297:E211–E224.
Article
83. Blachnio-Zabielska AU, Hady HR, Markowski AR, Kurianiuk A, Karwowska A, Górski J, et al. Inhibition of ceramide de novo synthesis affects adipocytokine secretion and improves systemic and adipose tissue insulin sensitivity. Int J Mol Sci. 2018; 19:E3995.
84. Kurek K, Piotrowska DM, Wiesiołek-Kurek P, Łukaszuk B, Chabowski A, Górski J, et al. Inhibition of ceramide de novo synthesis reduces liver lipid accumulation in rats with nonalcoholic fatty liver disease. Liver Int. 2014; 34:1074–1083.
Article
85. Correnti JM, Juskeviciute E, Swarup A, Hoek JB. Pharmacological ceramide reduction alleviates alcohol-induced steatosis and hepatomegaly in adiponectin knockout mice. Am J Physiol Gastrointest Liver Physiol. 2014; 306:G959–G973.
Article
86. Kasumov T, Li L, Li M, Gulshan K, Kirwan JP, Liu X, et al. Ceramide as a mediator of non-alcoholic fatty liver disease and associated atherosclerosis. PLoS One. 2015; 10:e0126910.
Article
87. Chaurasia B, Kaddai VA, Lancaster GI, Henstridge DC, Sriram S, Galam DL, et al. Adipocyte ceramides regulate subcutaneous adipose browning, inflammation, and metabolism. Cell Metab. 2016; 24:820–834.
Article
88. Bikman BT, Guan Y, Shui G, Siddique MM, Holland WL, Kim JY, et al. Fenretinide prevents lipid-induced insulin resistance by blocking ceramide biosynthesis. J Biol Chem. 2012; 287:17426–17437.
Article
89. Shimabukuro M, Higa M, Zhou YT, Wang MY, Newgard CB, Unger RH. Lipoapoptosis in beta-cells of obese prediabetic fa/fa rats. Role of serine palmitoyltransferase overexpression. J Biol Chem. 1998; 273:32487–32490.
90. Ji R, Akashi H, Drosatos K, Liao X, Jiang H, Kennel PJ, et al. Increased de novo ceramide synthesis and accumulation in failing myocardium. JCI Insight. 2017; 2:96203.
Article
91. Park TS, Hu Y, Noh HL, Drosatos K, Okajima K, Buchanan J, et al. Ceramide is a cardiotoxin in lipotoxic cardiomyopathy. J Lipid Res. 2008; 49:2101–2112.
Article
Full Text Links
  • JLA
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr