93
Views
8
CrossRef citations to date
0
Altmetric
Reviews

Developments in our understanding of the effects of growth hormone on white adipose tissue from mice: implications to the clinic

, , , &
Pages 197-207 | Received 17 Nov 2015, Accepted 26 Jan 2016, Published online: 24 Feb 2016

References

  • Prunet-Marcassus B, Cousin B, Caton D, et al. From heterogeneity to plasticity in adipose tissues: site-specific differences. Exp Cell Res. 2006;312(6):727–736.
  • Huh JY, Park YJ, Ham M, et al. Crosstalk between adipocytes and immune cells in adipose tissue inflammation and metabolic dysregulation in obesity. Mol Cells. 2014;37(5):365–371.
  • Chazenbalk G, Bertolotto C, Heneidi S, et al. Novel pathway of adipogenesis through cross-talk between adipose tissue macrophages, adipose stem cells and adipocytes: evidence of cell plasticity. PLoS One. 2011;6(3):e17834.
  • Grant RW, Dixit VD. Adipose tissue as an immunological organ. Obesity (Silver Spring). 2015;23(3):512–518.
  • Deiuliis J, Shah Z, Shah N, et al. Visceral adipose inflammation in obesity is associated with critical alterations in tregulatory cell numbers. PLoS One. 2011;6(1):e16376.
  • Hausman GJ, Dodson MV. Stromal vascular cells and adipogenesis: cells within adipose depots regulate adipogenesis. J Genomics. 2013;1:56–66.
  • Bertola A, Ciucci T, Rousseau D, et al. Identification of adipose tissue dendritic cells correlated with obesity-associated insulin-resistance and inducing Th17 responses in mice and patients. Diabetes. 2012;61(9):2238–2247.
  • Winer DA, Winer S, Shen L, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med. 2011;17(5):610–617.
  • Sepe A, Tchkonia T, Thomou T, et al. Aging and regional differences in fat cell progenitors - a mini-review. Gerontology. 2011;57(1):66–75.
  • Billon N, Dani C. Developmental origins of the adipocyte lineage: new insights from genetics and genomics studies. Stem Cell Rev. 2012;8(1):55–66.
  • Von Der Weid P-Y, Rainey KJ. Review article: lymphatic system and associated adipose tissue in the development of inflammatory bowel disease. Aliment Pharmacol Ther. 2010;32(6):697–711.
  • Gehrke S, Brueckner B, Schepky A, et al. Epigenetic regulation of depot-specific gene expression in adipose tissue. PLoS One. 2013;8(12):e82516.
  • Betz MJ, Enerbäck S. Human brown adipose tissue: what we have learned so far. Diabetes. 2015;64(7):2352–2360.
  • Cypess AM, Lehman S, Williams G, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med. 2009;360(15):1509–1517.
  • Divoux A, Tordjman J, Lacasa D, et al. Fibrosis in human adipose tissue: composition, distribution, and link with lipid metabolism and fat mass loss. Diabetes. 2010;59(11):2817–2825.
  • Khan T, Muise ES, Iyengar P, et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol Cell Biol. 2009;29(6):1575–1591.
  • Sun K, Tordjman J, Clément K, et al. Fibrosis and adipose tissue dysfunction. Cell Metab. 2013;18(4):470–477.
  • Londos C, Brasaemle DL, Schultz CJ, et al. Perilipins, ADRP, and other proteins that associate with intracellular neutral lipid droplets in animal cells. Semin Cell Dev Biol. 1999;10(1):51–58.
  • Childs GV, Akhter N, Haney A, et al. The somatotrope as a metabolic sensor: deletion of leptin receptors causes obesity. Endocrinology. 2011;152(1):69–81.
  • Watts SW. Trash talk by fat: chemerin as a reactive oxygen species provocateur in the vasculature. Hypertension. 2015;66(3):466–468.
  • Neves KB, Nguyen Dinh Cat A, Lopes RAM, et al. Chemerin regulates crosstalk between adipocytes and vascular cells through Nox. Hypertension. 2015;66(3):657–666.
  • Rao RR, Long JZ, White JP, et al. Meteorin-like is a hormone that regulates immune-adipose interactions to increase beige fat thermogenesis. Cell. 2014;157(6):1279–1291.
  • Li Z-Y, Zheng S-L, Wang P, et al. Subfatin is a novel adipokine and unlike Meteorin in adipose and brain expression. CNS Neurosci Ther. 2014;20(4):344–354.
  • Tchkonia T, Zhu Y, Van Deursen J, et al. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 2013;123(3):966–972.
  • Møller N, Jørgensen JO. Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocr Rev. 2009;30(2):152–177.
  • Brooks AJ, Dai W, O’Mara ML, et al. Mechanism of activation of protein kinase JAK2 by the growth hormone receptor. Science. 2014;344(6185):1249783.
  • Laron Z, Blum W, Chatelain P, et al. Classification of growth hormone insensitivity syndrome [editorial]. J Pediatr. 1993;122(2):241.
  • Steuerman R, Shevah O, Laron Z. Congenital IGF1 deficiency tends to confer protection against post-natal development of malignancies. Eur J Endocrinology/European Fed Endocr Societies. 2011;164(4):485–489.
  • Laron Z, Ginsberg S, Lilos P, et al. Body composition in untreated adult patients with Laron syndrome (primary GH insensitivity). Clin Endocrinol (Oxf). 2006;65(1):114–117.
  • Guevara-Aguirre J, Rosenbloom AL, Balasubramanian P, et al. GH receptor deficiency in ecuadorian adults is associated with obesity and enhanced insulin sensitivity. J Clin Endocrinol Metab. 2015;100(7):2589–2596.
  • Kanety H, Hemi R, Ginsberg S, et al. Total and high molecular weight adiponectin are elevated in patients with Laron syndrome despite marked obesity. Eur J Endocrinol. 2009;161(6):837–844.
  • Laron Z, Silbergeld A, Lilos P, et al. Serum leptin in obese patients with Laron syndrome before and during IGF-I treatment. J Pediatr Endocrinol Metab. 1998;11(5):653–656.
  • Gomes-Santos E, Salvatori R, Ferrão TO, et al. Increased visceral adiposity and cortisol to cortisone ratio in adults with congenital lifetime isolated GH deficiency. J Clin Endocrinol Metabolism. 2014;99(9):3285–3289.
  • Pereira RM, Aguiar-Oliveira MH, Sagazio A, et al. Heterozygosity for a mutation in the growth hormone-releasing hormone receptor gene does not influence adult stature, but affects body composition. J Clin Endocrinol Metab. 2007;92(6):2353–2357.
  • Aguiar-Oliveira MH, Oliveira FT, Pereira RMC, et al. Longevity in untreated congenital growth hormone deficiency due to a homozygous mutation in the GHRH receptor gene. J Clin Endocrinol Metab. 2010;95(2):714–721.
  • Vicente TAR, Rocha IES, Salvatori R, et al. Lifetime congenital isolated GH deficiency does not protect from the development of diabetes. Endocr Connect. 2013;2(2):112–117.
  • Chaves VE, Júnior FM, Bertolini GL. The metabolic effects of growth hormone in adipose tissue. Endocrine. 2013;44(2):293–302.
  • Ukropec J, Penesová A, Skopková M, et al. Adipokine protein expression pattern in growth hormone deficiency predisposes to the increased fat cell size and the whole body metabolic derangements. J Clin Endocrinol Metab. 2008;93(6):2255–2262.
  • Molitch ME, Clemmons DR, Malozowski S, et al. Evaluation and treatment of adult growth hormone deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96(6):1587–1609.
  • Melmed S. Acromegaly pathogenesis and treatment. J Clin Invest. 2009;119(11):3189–3202.
  • Reyes-Vidal CM, Mojahed H, Shen W, et al. Adipose tissue redistribution and ectopic lipid deposition in active acromegaly and effects of surgical treatment. J Clin Endocrinol Metab. 2015;100(8):2946–2955.
  • Ciresi A, Amato MC, Pizzolanti G, et al. Serum visfatin levels in acromegaly: correlation with disease activity and metabolic alterations. Growth Hormone IGF Research. 2015;25(5):240–246.
  • Olarescu NC, Ueland T, Lekva T, et al. Adipocytes as a source of increased circulating levels of nicotinamide phosphoribosyltransferase/visfatin in active acromegaly. J Clin Endocrinol Metab. 2012;97(4):1355–1362.
  • Luque RM, Lin Q, Córdoba-Chacón J, et al. Metabolic impact of adult-onset, isolated, growth hormone deficiency (AOiGHD) due to destruction of pituitary somatotropes. PLoS One. 2011;6(1):e15767.
  • List EO, Berryman DE, Funk K, et al. Liver-specific GH receptor gene-disrupted (LiGHRKO) mice have decreased endocrine IGF-I, increased local IGF-I, and altered body size, body composition, and adipokine profiles. Endocrinology. 2014;155(5):1793–1805.
  • List EO, Berryman DE, Funk K, et al. The role of GH in adipose tissue: lessons from adipose-specific GH receptor gene-disrupted mice. Mol Endocrinol. 2013;27(3):524–535.
  • Lu C, Kumar PA, Sun J, et al. Targeted deletion of growth hormone (GH) receptor in macrophage reveals novel osteopontin-mediated effects of GH on glucose homeostasis and insulin sensitivity in diet-induced obesity. J Biol Chem. 2013;288(22):15725–15735.
  • Berryman DE, List EO, Kohn DT, et al. Effect of growth hormone on susceptibility to diet-induced obesity. Endocrinology. 2006;147(6):2801–2808.
  • Berryman DE, List EO, Coschigano KT, et al. Comparing adiposity profiles in three mouse models with altered GH signaling. Growth Horm IGF Res. 2004;14(4):309–318.
  • Wiesenborn DS, Menon V, Zhi X, et al. The effect of calorie restriction on insulin signaling in skeletal muscle and adipose tissue of Ames dwarf mice. Aging. 2014;6(10):900–912.
  • Brown-Borg HM, Bartke A. GH and IGF1: roles in energy metabolism of long-living GH mutant mice. J Gerontol A Biol Sci Med Sci. 2012;67A(6):652–660.
  • Bartke A, Brown-Borg H. Life extension in the dwarf mouse. Curr Top Dev Biol. 2004;63:189–225.
  • Flurkey K, Papaconstantinou J, Miller RA, et al. Lifespan extension and delayed immune and collagen aging in mutant mice with defects in growth hormone production. Proc Natl Acad Sci U S A. 2001;98(12):6736–6741.
  • Fleenor D, Oden J, Kelly PA, et al. Roles of the lactogens and somatogens in perinatal and postnatal metabolism and growth: studies of a novel mouse model combining lactogen resistance and growth hormone deficiency. Endocrinology. 2005;146(1):103–112.
  • Berryman DE, List EO, Sackmann-Sala L, et al. Growth hormone and adipose tissue: beyond the adipocyte. Growth Horm IGF Res. 2011;21(3):113–123.
  • Menon V, Zhi X, Hossain T, et al. The contribution of visceral fat to improved insulin signaling in Ames dwarf mice. Aging Cell. 2014;13(3):497–506.
  • Richelsen B, Pedersen SB, Kristensen K, et al. Regulation of lipoprotein lipase and hormone-sensitive lipase activity and gene expression in adipose and muscle tissue by growth hormone treatment during weight loss in obese patients. Metabolism. 2000;49(7):906–911.
  • Kraemer FB, Shen W-J. Hormone-sensitive lipase: control of intracellular tri-(di-)acylglycerol and cholesteryl ester hydrolysis. J Lipid Res. 2002;43(10):1585–1594.
  • Yang HL, Sun C, Sun C, et al. Effect of suppressor of cytokine signaling 2 (SOCS2) on fat metabolism induced by growth hormone (GH) in porcine primary adipocyte. Mol Biol Rep. 2012;39(9):9113–9122.
  • Fain JN, Cheema P, Tichansky DS, et al. Stimulation of human omental adipose tissue lipolysis by growth hormone plus dexamethasone. Mol Cell Endocrinol. 2008;295(1–2):101–105.
  • Nam SY, Lobie PE. The mechanism of effect of growth hormone on preadipocyte and adipocyte function. Obes Rev. 2000;1(2):73–86.
  • Flint DJ, Binart N, Boumard S, et al. Developmental aspects of adipose tissue in GH receptor and prolactin receptor gene disrupted mice: site-specific effects upon proliferation, differentiation and hormone sensitivity. J Endocrinol. 2006;191(1):101–111.
  • Olarescu NC, Berryman DE, Householder LA, et al. GH action influences adipogenesis of mouse adipose tissue-derived mesenchymal stem cells. J Endocrinol. 2015;226(1):13–23.
  • Masternak MM, Bartke A, Wang F, et al. Metabolic effects of intra-abdominal fat in GHRKO mice. Aging Cell. 2012;11(1):73–81.
  • Lubbers ER, List EO, Jara A, et al. Adiponectin in mice with altered growth hormone action: links to insulin sensitivity and longevity? J Endocrinol. 2012;216(3):363–374.
  • Wölfing B, Neumeier M, Buechler C, et al. Interfering effects of insulin, growth hormone and glucose on adipokine secretion. Exp Clin Endocrinol Diabetes. 2008;116(1):47–52.
  • Tchkonia T, Thomou T, Zhu Y, et al. Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab. 2013;17(5):644–656.
  • Tchkonia T, Morbeck DE, Von Zglinicki T, et al. Fat tissue, aging, and cellular senescence. Aging Cell. 2010;9(5):667–684.
  • Stout MB, Tchkonia T, Pirtskhalava T, et al. Growth hormone action predicts age-related white adipose tissue dysfunction and senescent cell burden in mice. Aging (Albany NY). 2014;6(7):575–586.
  • Comisford R, Lubbers ER, Householder LA, et al. Growth hormone receptor antagonist transgenic mice have increased subcutaneous adipose tissue mass, altered glucose homeostasis and no change in white adipose tissue cellular senescence. Gerontology. 2015;62(2). [Epub ahead of print]. Available form: http://www.ncbi.nlm.nih.gov/pubmed/26372907
  • Villaret A, Galitzky J, Decaunes P, et al. Adipose tissue endothelial cells from obese human subjects: differences among depots in angiogenic, metabolic, and inflammatory gene expression and cellular senescence. Diabetes. 2010;59(11):2755–2763.
  • Hattori N. Expression, regulation and biological actions of growth hormone (GH) and ghrelin in the immune system. Growth Horm IGF Res. 2009;19(3):187–197.
  • Lu C, Kumar PA, Fan Y, et al. A novel effect of growth hormone on macrophage modulates macrophage-dependent adipocyte differentiation. Endocrinology. 2010;151(5):2189–2199.
  • Benencia F, Harshman S, Duran-Ortiz S, et al. Male bovine GH transgenic mice have decreased adiposity with an adipose depot-specific increase in immune cell populations. Endocrinology. 2014;156(5):1794–1803. en20141794.
  • Stout MB, Swindell WR, Zhi X, et al. Transcriptome profiling reveals divergent expression shifts in brown and white adipose tissue from long-lived GHRKO mice. Oncotarget. 2015;6(29):26702–26715.
  • Doessing S, Holm L, Heinemeier KM, et al. GH and IGF1 levels are positively associated with musculotendinous collagen expression: experiments in acromegalic and GH deficiency patients. Eur J Endocrinol. 2010;163(6):853–862.
  • Longobardi S, Keay N, Ehrnborg C, et al. Growth hormone (GH) effects on bone and collagen turnover in healthy adults and its potential as a marker of GH abuse in sports: a double blind, placebo-controlled study. The GH-2000 Study Group. J Clin Endocrinol Metab. 2000;85(4):1505–1512.
  • Amato G, Mazziotti G, Di Somma C, et al. Recombinant growth hormone (GH) therapy in GH-deficient adults: a long-term controlled study on daily versus thrice weekly injections. J Clin Endocrinol Metab. 2000;85(10):3720–3725.
  • Hazem A, Elamin MB, Bancos I, et al. Body composition and quality of life in adults treated with GH therapy: a systematic review and meta-analysis. Eur J Endocrinology/European Fed Endocr Societies. 2012;166(1):13–20.
  • Meazza C, Elsedfy HH, Pagani S, et al. Metabolic parameters and adipokine profile in growth hormone deficient (GHD) children before and after 12-month GH treatment. Horm Metab Res. 2014;46(3):219–223.
  • Rosenbloom AL. A half-century of studies of growth hormone insensitivity/Laron syndrome: a historical perspective. Growth Horm IGF Res. 2015. doi:10.1016/j.ghir.2015.08.001. [Epub ahead of print]. Available from: http://www.sciencedirect.com/science/article/pii/S1096637415300216
  • Laron Z, Ginsberg S, Lilos P, et al. Long-term IGF-I treatment of children with Laron syndrome increases adiposity. Growth Horm IGF Res. 2006;16(1):61–64.
  • Guevara-Aguirre J, Rosenbloom AL, Guevara-Aguirre M, et al. Recommended IGF-I dosage causes greater fat accumulation and osseous maturation than lower dosage and may compromise long-term growth effects. J Clin Endocrinol Metabolism. 2013;98(2):839–845.
  • Ogden CL, Carroll MD, Kit BK, et al. Prevalence of childhood and adult obesity in the United States, 2011-2012. Jama. 2014;311(8):806–814.
  • Berryman DE, Glad CAM, List EO, et al. The GH/IGF-1 axis in obesity: pathophysiology and therapeutic considerations. Nat Rev Endocrinol. 2013;9(6):346–356.
  • Lewitt MS, Dent MS, Hall K. The insulin-like growth factor system in obesity, insulin resistance and type 2 diabetes mellitus. J Clin Med. 2014;3(4):1561–1574.
  • Franco C, Brandberg J, Lönn L, et al. Growth hormone treatment reduces abdominal visceral fat in postmenopausal women with abdominal obesity: a 12-month placebo-controlled trial. J Clin Endocrinol Metab. 2005;90(3):1466–1474.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.