• No results found

In this thesis, the effects of GH and PPARα on lipid metabolism have been investigated, especially with respect to hepatic genes involved in lipogenesis and VLDL assembly.

• The importance of increased insulin levels for the effects of GH on lipoprotein metabolism was investigated in Hx rats. GH increased the triglyceride secretion rate and content in the liver (Figure 8), which could be explained by increased gene expression of enzymes in lipogenesis. Insulin suppressed the effect of GH on the hepatic triglyceride secretion rate and content, but this was not due to decreased expression of SCD-1, FAS or MTP. Insulin also suppressed the effect of GH on SREBP-1c mRNA. The GH-antagonistic effect of insulin may therefore be explained by changed expression of other SREBP-1c regulated genes or decreased availability of FFA. In conclusion, insulin does not mediate the effects of GH but inhibits the stimulatory effect of GH on hepatic SREBP-1c expression and hepatic triglyceride secretion rate and content.

Figure 8. The effects of continuous GH (GHc) administration in this thesis. *TG (triglyceride) synthesis was measured as total TG biosynthesis in the whole cell (results from Sjöberg et al, 1996 [136]).

apoB MTP

TG ER lumen

TG Pre-VLDL

TG

TG

Cytosol

TG TG

FAS, GPAT mRNA

+ GHc

+

+ +

+ +

TG synthesis*

+ +

TG synthesis*

• The regulation of hepatic lipogenic genes and MTP by the sex-differentiated GH secretory pattern was studied in Hx rats administered GH in a mode that mimics either the female or male plasma pattern of GH. FAS and GPAT mRNA levels were increased in females compared to males and specifically upregulated by female-like GH administration, probably due to decreased insulin sensitivity. SCD-1 was not sex-differentiated and increased by GH irrespective of administration mode. The MTP expression was higher in female rats due to the female GH secretory pattern. In conclusion, increased expression of FAS, GPAT and MTP could help to explain the previously described stimulatory effects of female sex and the female GH secretory pattern on VLDL assembly and secretion (Figure 8).

• Treatment of mice and rats with a PPARα agonist (WY) increased MTP expression and activity in mouse and rat liver. Incubation of primary cultures of mouse and rat hepatocytes with WY also increased MTP expression. By using primary hepatocyte cultures from PPARα KO mice, this effect of WY was shown to be PPARα specific. In rat hepatocytes incubated with WY, MTP protein expression and apoB-100 secretion increased between 24 and 72 h of incubation. In conclusion, PPARα activation increases MTP expression and activity that could explain the increased apoB-100 secretion from hepatocytes following PPARα activation (Figure 9).

Figure 9. The effects of PPARα treatment in this thesis. *Results from Lindén et al, 2002 [92]. TG (triglyceride) synthesis was measured as total TG biosynthesis in the whole cell.

--ACKNOWLEDGEMENTS

I am grateful to all the persons who in one way or another have contributed to this thesis or who have made my life as a PhD student much more enjoyable. I would especially like to thank:

Jan Oscarsson, my supervisor, for all your support and excellent guidance throughout the years. You have shared both your great knowledge and enthusiasm for science with me, which I have very much appreciated.

Past and present members of the Endocrine division, for all the good times and for creating such a nice atmosphere to work in. It was sad leaving you…

…but I am very happy for the warm welcome at Wallenberg laboratory! My time here has been a pleasure thanks to all nice people and the cheerful atmosphere!

All co-authors, for help with lab work and valuable comments on the manuscripts.

My present fellow PhD students, Anna Ljungberg, for being a good friend and for sharing my passion for apples and aversion for cold, and Ulrika Edvardsson, for nice company in the lab and for being my personal Maniatis.

My former fellow PhD students, Linda Carlsson, for good company in the “booth”

and for the way you kept the Endocrine division in order, Daniel Lindén, for nice collaboration that is still ongoing, and Fredrik Frick, for your sense of humour that makes everyone feel good at work.

Mikael Alsterholm, for being such a nice person and for always drinking tea with me.

I miss you at the lab (are you coming back soon?)!

Heimir Snorrason, the enchanting wizard of computers, for invaluable help with everything concerning computers. I have really appreciated all your kind help!

All my friends outside the lab - no one mentioned, no one forgotten. I’m sorry that I have been asocial the last couple of months… Now I will definitively try to spend more time with you, my excellent friends!

Jonas Boström, for all the fun we had throughout the years and for being my best friend. Especially many thanks for your excellent Thai food and the warm bath that saved me from the radioactivity disaster.

My parents, Ulla and Torkel, for all your love and for always always being there.

Kajsa, the best sister you can think of! Thanks for being such a cool and kind sister (except when you don’t get food in time) and, of course, for regularly getting rid of

“the ball”.

Sandra, for endless love and support, thank you for everything.

And remember, “Acknowledgements” is NOT the important part of a thesis! Now go on read from the beginning…

REFERENCES

1. Bengtsson, B.A., et al., Epidemiology and long-term survival in acromegaly. A study of 166 cases diagnosed between 1955 and 1984. Acta Med Scand, 1988.

223(4): p. 327-35.

2. Rosen, T., et al., Cardiovascular risk factors in adult patients with growth hormone deficiency. Acta Endocrinol (Copenh), 1993. 129(3): p. 195-200.

3. Franek, E., et al., Abnormal pulsatile secretion of growth hormone in non-insulin-dependent diabetes mellitus. Clin Endocrinol (Oxf), 1997. 47(4): p. 471-8.

4. Packard, C.J., Triacylglycerol-rich lipoproteins and the generation of small, dense low-density lipoprotein. Biochem Soc Trans, 2003. 31(Pt 5): p. 1066-9.

5. Grundy, S.M., Small LDL, atherogenic dyslipidemia, and the metabolic syndrome. Circulation, 1997. 95(1): p. 1-4.

6. Austin, M.A., et al., Atherogenic lipoprotein phenotype. A proposed genetic marker for coronary heart disease risk. Circulation, 1990. 82(2): p. 495-506.

7. Guerin, M., et al., Fenofibrate reduces plasma cholesteryl ester transfer from HDL to VLDL and normalizes the atherogenic, dense LDL profile in combined hyperlipidemia. Arterioscler Thromb Vasc Biol, 1996. 16(6): p. 763-72.

8. Tulenko, T.N. and A.E. Sumner, The physiology of lipoproteins. J Nucl Cardiol, 2002. 9(6): p. 638-49.

9. Taskinen, M.R., Diabetic dyslipidaemia: from basic research to clinical practice.

Diabetologia, 2003. 46(6): p. 733-49.

10. Chen, S.H., et al., Apolipoprotein B-48 is the product of a messenger RNA with an organ- specific in-frame stop codon. Science, 1987. 238(4825): p. 363-6.

11. Powell, L.M., et al., A novel form of tissue-specific RNA processing produces apolipoprotein- B48 in intestine. Cell, 1987. 50(6): p. 831-40.

12. Davidson, N.O., et al., Proposed nomenclature for the catalytic subunit of the mammalian apolipoprotein B mRNA editing enzyme: APOBEC-1. Rna, 1995.

1(1): p. 3.

13. Teng, B., C.F. Burant, and N.O. Davidson, Molecular cloning of an apolipoprotein B messenger RNA editing protein. Science, 1993. 260(5115): p.

1816-9.

14. Davidson, N.O. and G.S. Shelness, APOLIPOPROTEIN B: mRNA editing, lipoprotein assembly, and presecretory degradation. Annu Rev Nutr, 2000. 20: p.

169-93.

15. Innerarity, T.L., et al., Biosynthesis of apolipoprotein B48-containing lipoproteins. Regulation by novel post-transcriptional mechanisms. J Biol Chem, 1996. 271(5): p. 2353-6.

16. Olofsson, S.O., L. Asp, and J. Boren, The assembly and secretion of apolipoprotein B-containing lipoproteins. Curr Opin Lipidol, 1999. 10(4): p. 341-6.

17. Alexander, C.A., R.L. Hamilton, and R.J. Havel, Subcellular localization of B apoprotein of plasma lipoproteins in rat liver. J Cell Biol, 1976. 69(2): p. 241-63.

18. Boren, J., S. Rustaeus, and S.O. Olofsson, Studies on the assembly of apolipoprotein B-100- and B-48-containing very low density lipoproteins in McA-RH7777 cells. J Biol Chem, 1994. 269(41): p. 25879-88.

19. Boren, J., et al., The assembly and secretion of ApoB 100-containing lipoproteins in Hep G2 cells. ApoB 100 is cotranslationally integrated into lipoproteins. J Biol Chem, 1992. 267(14): p. 9858-67.

20. Rustaeus, S., et al., Brefeldin A reversibly inhibits the assembly of apoB containing lipoproteins in McA-RH7777 cells. J Biol Chem, 1995. 270(48): p.

28879-86.

21. Fisher, E.A., et al., The degradation of apolipoprotein B100 is mediated by the ubiquitin-proteasome pathway and involves heat shock protein 70. J Biol Chem, 1997. 272(33): p. 20427-34.

22. Yeung, S.J., S.H. Chen, and L. Chan, Ubiquitin-proteasome pathway mediates intracellular degradation of apolipoprotein B. Biochemistry, 1996. 35(43): p.

13843-8.

23. Adeli, K., et al., Apolipoprotein B is intracellularly associated with an ER-60 protease homologue in HepG2 cells. J Biol Chem, 1997. 272(36): p. 22489-94.

24. Wetterau, J.R., M.C. Lin, and H. Jamil, Microsomal triglyceride transfer protein.

Biochim Biophys Acta, 1997. 1345(2): p. 136-50.

25. Bulleid, N.J. and R.B. Freedman, Defective co-translational formation of disulphide bonds in protein disulphide-isomerase-deficient microsomes. Nature, 1988. 335(6191): p. 649-51.

26. Freedman, R.B., T.R. Hirst, and M.F. Tuite, Protein disulphide isomerase:

building bridges in protein folding. Trends Biochem Sci, 1994. 19(8): p. 331-6.

27. Jamil, H., et al., Microsomal triglyceride transfer protein. Specificity of lipid binding and transport. J Biol Chem, 1995. 270(12): p. 6549-54.

28. Patel, S.B. and S.M. Grundy, Interactions between microsomal triglyceride transfer protein and apolipoprotein B within the endoplasmic reticulum in a heterologous expression system. J Biol Chem, 1996. 271(31): p. 18686-94.

29. Wu, X., et al., Demonstration of a physical interaction between microsomal triglyceride transfer protein and apolipoprotein B during the assembly of ApoB-containing lipoproteins. J Biol Chem, 1996. 271(17): p. 10277-81.

30. Mann, C.J., et al., The structure of vitellogenin provides a molecular model for the assembly and secretion of atherogenic lipoproteins. J Mol Biol, 1999. 285(1): p.

391-408.

31. Benoist, F., E. Nicodeme, and T. Grand-Perret, Microsomal triacylglycerol transfer protein prevents presecretory degradation of apolipoprotein B-100. A dithiothreitol-sensitive protease is involved. Eur J Biochem, 1996. 240(3): p. 713-20.

32. Liao, W., K. Kobayashi, and L. Chan, Adenovirus-mediated overexpression of microsomal triglyceride transfer protein (MTP): mechanistic studies on the role of MTP in apolipoprotein B-100 biogenesis, by. Biochemistry, 1999. 38(31): p.

10215.

33. Benoist, F. and T. Grand-Perret, Co-translational degradation of apolipoprotein B100 by the proteasome is prevented by microsomal triglyceride transfer protein.

Synchronized translation studies on HepG2 cells treated with an inhibitor of microsomal triglyceride transfer protein. J Biol Chem, 1997. 272(33): p. 20435-42.

34. Gordon, D.A. and H. Jamil, Progress towards understanding the role of microsomal triglyceride transfer protein in apolipoprotein-B lipoprotein assembly. Biochim Biophys Acta, 2000. 1486(1): p. 72-83.

35. Zhou, M., E.A. Fisher, and H.N. Ginsberg, Regulated Co-translational ubiquitination of apolipoprotein B100. A new paradigm for proteasomal degradation of a secretory protein. J Biol Chem, 1998. 273(38): p. 24649-53.

36. Gretch, D.G., et al., The amino terminus of apolipoprotein B is necessary but not sufficient for microsomal triglyceride transfer protein responsiveness. J Biol Chem, 1996. 271(15): p. 8682-91.

37. Vuori, K., et al., Site-directed mutagenesis of human protein disulphide isomerase: effect on the assembly, activity and endoplasmic reticulum retention of human prolyl 4-hydroxylase in Spodoptera frugiperda insect cells. Embo J, 1992.

11(11): p. 4213-7.

38. Wetterau, J.R., et al., Protein disulfide isomerase appears necessary to maintain the catalytically active structure of the microsomal triglyceride transfer protein.

Biochemistry, 1991. 30(40): p. 9728-35.

39. Tietge, U.J., et al., Hepatic overexpression of microsomal triglyceride transfer protein (MTP) results in increased in vivo secretion of VLDL triglycerides and apolipoprotein B. J Lipid Res, 1999. 40(11): p. 2134-9.

40. Raabe, M., et al., Knockout of the abetalipoproteinemia gene in mice: reduced lipoprotein secretion in heterozygotes and embryonic lethality in homozygotes.

Proc Natl Acad Sci U S A, 1998. 95(15): p. 8686-91.

41. Leung, G.K., et al., A deficiency of microsomal triglyceride transfer protein reduces apolipoprotein B secretion. J Biol Chem, 2000. 275(11): p. 7515-20.

42. Berriot-Varoqueaux, N., et al., The role of the microsomal triglygeride transfer protein in abetalipoproteinemia. Annu Rev Nutr, 2000. 20: p. 663-97.

43. Raabe, M., et al., Analysis of the role of microsomal triglyceride transfer protein in the liver of tissue-specific knockout mice. J Clin Invest, 1999. 103(9): p. 1287-98.

44. Gordon, D.A., et al., Inhibition of the microsomal triglyceride transfer protein blocks the first step of apolipoprotein B lipoprotein assembly but not the addition of bulk core lipids in the second step. J Biol Chem, 1996. 271(51): p. 33047-53.

45. Lin, M.C., D. Gordon, and J.R. Wetterau, Microsomal triglyceride transfer protein (MTP) regulation in HepG2 cells: insulin negatively regulates MTP gene expression. J Lipid Res, 1995. 36(5): p. 1073-81.

46. Hagan, D.L., et al., Transcriptional regulation of human and hamster microsomal triglyceride transfer protein genes. Cell type-specific expression and response to metabolic regulators. J Biol Chem, 1994. 269(46): p. 28737-44.

47. Carpentier, A., et al., Ameliorated hepatic insulin resistance is associated with normalization of microsomal triglyceride transfer protein expression and reduction in very low density lipoprotein assembly and secretion in the fructose-fed hamster. J Biol Chem, 2002. 277(32): p. 28795-802.

48. Taghibiglou, C., et al., Mechanisms of hepatic very low density lipoprotein overproduction in insulin resistance. Evidence for enhanced lipoprotein assembly, reduced intracellular ApoB degradation, and increased microsomal triglyceride

transfer protein in a fructose-fed hamster model. J Biol Chem, 2000. 275(12): p.

8416-25.

49. Bartels, E.D., M. Lauritsen, and L.B. Nielsen, Hepatic expression of microsomal triglyceride transfer protein and in vivo secretion of triglyceride-rich lipoproteins are increased in obese diabetic mice. Diabetes, 2002. 51(4): p. 1233-9.

50. Bennett, A.J., et al., Regulation of hamster hepatic microsomal triglyceride transfer protein mRNA levels by dietary fats. Biochem Biophys Res Commun, 1995. 212(2): p. 473-8.

51. Lin, M.C., et al., Cloning and regulation of hamster microsomal triglyceride transfer protein. The regulation is independent from that of other hepatic and intestinal proteins which participate in the transport of fatty acids and triglycerides. J Biol Chem, 1994. 269(46): p. 29138-45.

52. Taguchi, H., et al., Dietary diacylglycerol suppresses high fat diet-induced hepatic fat accumulation and microsomal triacylglycerol transfer protein activity in rats. J Nutr Biochem, 2002. 13(11): p. 678-683.

53. Bennett, A.J., et al., Hepatic microsomal triglyceride transfer protein messenger RNA concentrations are increased by dietary cholesterol in hamsters. FEBS Lett, 1996. 394(3): p. 247-50.

54. Sato, R., et al., Sterol regulatory element-binding protein negatively regulates microsomal triglyceride transfer protein gene transcription. J Biol Chem, 1999.

274(35): p. 24714-20.

55. Horton, J.D., et al., Activation of cholesterol synthesis in preference to fatty acid synthesis in liver and adipose tissue of transgenic mice overproducing sterol regulatory element-binding protein-2. J Clin Invest, 1998. 101(11): p. 2331-9.

56. Wang, S.L., et al., Coordinate regulation of lipogenesis, the assembly and secretion of apolipoprotein B-containing lipoproteins by sterol response element binding protein 1. J Biol Chem, 1997. 272(31): p. 19351-8.

57. Brown, M.S. and J.L. Goldstein, The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell, 1997.

89(3): p. 331-40.

58. Hua, X., et al., SREBP-2, a second basic-helix-loop-helix-leucine zipper protein that stimulates transcription by binding to a sterol regulatory element. Proc Natl Acad Sci U S A, 1993. 90(24): p. 11603-7.

59. Shimano, H., et al., Isoform 1c of sterol regulatory element binding protein is less active than isoform 1a in livers of transgenic mice and in cultured cells. J Clin Invest, 1997. 99(5): p. 846-54.

60. Shimomura, I., et al., Differential expression of exons 1a and 1c in mRNAs for sterol regulatory element binding protein-1 in human and mouse organs and cultured cells. J Clin Invest, 1997. 99(5): p. 838-45.

61. Shimano, H., et al., Overproduction of cholesterol and fatty acids causes massive liver enlargement in transgenic mice expressing truncated SREBP-1a. J Clin Invest, 1996. 98(7): p. 1575-84.

62. Shimano, H., et al., Sterol regulatory element-binding protein-1 as a key transcription factor for nutritional induction of lipogenic enzyme genes. J Biol Chem, 1999. 274(50): p. 35832-9.

63. Shimomura, I., et al., Nuclear sterol regulatory element-binding proteins activate genes responsible for the entire program of unsaturated fatty acid biosynthesis in transgenic mouse liver. J Biol Chem, 1998. 273(52): p. 35299-306.

64. Pai, J.T., et al., Differential stimulation of cholesterol and unsaturated fatty acid biosynthesis in cells expressing individual nuclear sterol regulatory element-binding proteins. J Biol Chem, 1998. 273(40): p. 26138-48.

65. Sakai, J., et al., Sterol-regulated release of SREBP-2 from cell membranes requires two sequential cleavages, one within a transmembrane segment. Cell, 1996. 85(7): p. 1037-46.

66. Wang, X., et al., SREBP-1, a membrane-bound transcription factor released by sterol-regulated proteolysis. Cell, 1994. 77(1): p. 53-62.

67. Kim, J.B., et al., Dual DNA binding specificity of ADD1/SREBP1 controlled by a single amino acid in the basic helix-loop-helix domain. Mol Cell Biol, 1995.

15(5): p. 2582-8.

68. Hasty, A.H., et al., Sterol regulatory element-binding protein-1 is regulated by glucose at the transcriptional level. J Biol Chem, 2000. 275(40): p. 31069-77.

69. Foretz, M., et al., ADD1/SREBP-1c is required in the activation of hepatic lipogenic gene expression by glucose. Mol Cell Biol, 1999. 19(5): p. 3760-8.

70. Shimomura, I., et al., Insulin selectively increases SREBP-1c mRNA in the livers of rats with streptozotocin-induced diabetes. Proc Natl Acad Sci U S A, 1999.

96(24): p. 13656-61.

71. Yahagi, N., et al., A crucial role of sterol regulatory element-binding protein-1 in the regulation of lipogenic gene expression by polyunsaturated fatty acids. J Biol Chem, 1999. 274(50): p. 35840-4.

72. Shimano, H., Sterol regulatory element-binding protein-1 as a dominant transcription factor for gene regulation of lipogenic enzymes in the liver. Trends Cardiovasc Med, 2000. 10(7): p. 275-8.

73. Azzout-Marniche, D., et al., Insulin effects on sterol regulatory-element-binding protein-1c (SREBP-1c) transcriptional activity in rat hepatocytes. Biochem J, 2000. 350 Pt 2: p. 389-93.

74. Foretz, M., et al., Sterol regulatory element binding protein-1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis- related genes. Proc Natl Acad Sci U S A, 1999. 96(22): p. 12737-42.

75. Lopez, J.M., et al., Sterol regulation of acetyl coenzyme A carboxylase: a mechanism for coordinate control of cellular lipid. Proc Natl Acad Sci U S A, 1996. 93(3): p. 1049-53.

76. Kim, J.B. and B.M. Spiegelman, ADD1/SREBP1 promotes adipocyte differentiation and gene expression linked to fatty acid metabolism. Genes Dev, 1996. 10(9): p. 1096-107.

77. Ericsson, J., et al., Identification of glycerol-3-phosphate acyltransferase as an adipocyte determination and differentiation factor 1- and sterol regulatory element-binding protein-responsive gene. J Biol Chem, 1997. 272(11): p. 7298-305.

78. Munday, M.R. and C.J. Hemingway, The regulation of acetyl-CoA carboxylase--a potential target for the action of hypolipidemic agents. Adv Enzyme Regul, 1999.

39: p. 205-34.

79. Moustaid, N., et al., Regulation of fatty acid synthase gene transcription.

Sequences that confer a positive insulin effect and differentiation-dependent expression in 3T3-L1 preadipocytes are present in the 332 bp promoter. Biochem J, 1993. 292 ( Pt 3): p. 767-72.

80. Semenkovich, C.F., T. Coleman, and R. Goforth, Physiologic concentrations of glucose regulate fatty acid synthase activity in HepG2 cells by mediating fatty acid synthase mRNA stability. J Biol Chem, 1993. 268(10): p. 6961-70.

81. Ntambi, J.M., The regulation of stearoyl-CoA desaturase (SCD). Prog Lipid Res, 1995. 34(2): p. 139-50.

82. Miyazaki, M., et al., The biosynthesis of hepatic cholesterol esters and triglycerides is impaired in mice with a disruption of the gene for stearoyl-CoA desaturase 1. J Biol Chem, 2000. 275(39): p. 30132-8.

83. Miyazaki, M., W.C. Man, and J.M. Ntambi, Targeted disruption of stearoyl-CoA desaturase1 gene in mice causes atrophy of sebaceous and meibomian glands and depletion of wax esters in the eyelid. J Nutr, 2001. 131(9): p. 2260-8.

84. Sul, H.S. and D. Wang, Nutritional and hormonal regulation of enzymes in fat synthesis: studies of fatty acid synthase and mitochondrial glycerol-3-phosphate acyltransferase gene transcription. Annu Rev Nutr, 1998. 18: p. 331-51.

85. Hammond, L.E., et al., Mitochondrial glycerol-3-phosphate acyltransferase-deficient mice have reduced weight and liver triacylglycerol content and altered glycerolipid fatty acid composition. Mol Cell Biol, 2002. 22(23): p. 8204-14.

86. Peters, J.M., et al., Alterations in lipoprotein metabolism in peroxisome proliferator- activated receptor alpha-deficient mice. J Biol Chem, 1997. 272(43):

p. 27307-12.

87. Schoonjans, K., B. Staels, and J. Auwerx, The peroxisome proliferator activated receptors (PPARS) and their effects on lipid metabolism and adipocyte differentiation. Biochim Biophys Acta, 1996. 1302(2): p. 93-109.

88. Torra, I.P., et al., Peroxisome proliferator-activated receptors: from transcriptional control to clinical practice. Curr Opin Lipidol, 2001. 12(3): p.

245-54.

89. Kersten, S., et al., Peroxisome proliferator-activated receptor alpha mediates the adaptive response to fasting. J Clin Invest, 1999. 103(11): p. 1489-98.

90. Vu-Dac, N., et al., The nuclear receptors peroxisome proliferator-activated receptor alpha and Rev-erbalpha mediate the species-specific regulation of apolipoprotein A-I expression by fibrates. J Biol Chem, 1998. 273(40): p. 25713-20.

91. Staels, B., et al., Mechanism of action of fibrates on lipid and lipoprotein metabolism. Circulation, 1998. 98(19): p. 2088-93.

92. Linden, D., et al., Influence of peroxisome proliferator activated receptoralpha (PPARalpha) agonists on the intracellular turnover and secretion of apoB-100 and apoB-48. J Biol Chem, 2002. 29: p. 29.

93. Seeburg, P.H., et al., Nucleotide sequence and amplification in bacteria of structural gene for rat growth hormone. Nature, 1977. 270(5637): p. 486-94.

94. Giustina, A. and J.D. Veldhuis, Pathophysiology of the neuroregulation of growth hormone secretion in experimental animals and the human. Endocr Rev, 1998.

19(6): p. 717-97.

95. Eden, S., Age- and sex-related differences in episodic growth hormone secretion in the rat. Endocrinology, 1979. 105(2): p. 555-60.

96. Jaffe, C.A., et al., Regulatory mechanisms of growth hormone secretion are sexually dimorphic. J Clin Invest, 1998. 102(1): p. 153-64.

97. Tannenbaum, G.S. and J.B. Martin, Evidence for an endogenous ultradian rhythm governing growth hormone secretion in the rat. Endocrinology, 1976. 98(3): p.

562-70.

98. Jansson, J.O., et al., Influence of gonadal steroids on age- and sex-related secretory patterns of growth hormone in the rat. Endocrinology, 1984. 114(4): p.

1287-94.

99. van den Berg, G., et al., An amplitude-specific divergence in the pulsatile mode of growth hormone (GH) secretion underlies the gender difference in mean GH concentrations in men and premenopausal women. J Clin Endocrinol Metab, 1996. 81(7): p. 2460-7.

100. Ho, K.Y. and A.J. Weissberger, Secretory patterns of growth hormone according to sex and age. Horm Res, 1990. 33 Suppl 4: p. 7-11.

101. Mode, A., et al., Association between plasma level of growth hormone and sex differentiation of hepatic steroid metabolism in the rat. Endocrinology, 1982.

111(5): p. 1692-7.

102. Eden, S., et al., The influence of gonadal steroids and the pituitary on the levels and composition of plasma phospholipids in the rat. Metabolism, 1987. 36(6): p.

527-32.

103. Wehrenberg, W.B. and A. Giustina, Basic counterpoint: mechanisms and pathways of gonadal steroid modulation of growth hormone secretion. Endocr Rev, 1992. 13(2): p. 299-308.

104. Aguila, M.C. and S.M. McCann, Stimulation of somatostatin release in vitro by synthetic human growth hormone-releasing factor by a nondopaminergic mechanism. Endocrinology, 1985. 117(2): p. 762-5.

105. Clark, R.G., et al., Intravenous growth hormone: growth responses to patterned infusions in hypophysectomized rats. J Endocrinol, 1985. 104(1): p. 53-61.

105. Clark, R.G., et al., Intravenous growth hormone: growth responses to patterned infusions in hypophysectomized rats. J Endocrinol, 1985. 104(1): p. 53-61.

Related documents