Comprehensive Physiology Wiley Online Library

Impact of Growth Hormone on Regulation of Adipose Tissue

Full Article on Wiley Online Library



ABSTRACT

Increasing prevalence of obesity and obesity‐related conditions worldwide has necessitated a more thorough understanding of adipose tissue (AT) and expanded the scope of research in this field. AT is now understood to be far more complex and dynamic than previously thought, which has also fueled research to reevaluate how hormones, such as growth hormone (GH), alter the tissue. In this review, we will introduce properties of AT important for understanding how GH alters the tissue, such as anatomical location of depots and adipokine output. We will provide an overview of GH structure and function and define several human conditions and cognate mouse lines with extremes in GH action that have helped shape our understanding of GH and AT. A detailed discussion of the GH/AT relationship will be included that addresses adipokine production, immune cell populations, lipid metabolism, senescence, differentiation, and fibrosis, as well as brown AT and beiging of white AT. A brief overview of how GH levels are altered in an obese state, and the efficacy of GH as a therapeutic option to manage obesity will be given. As we will reveal, the effects of GH on AT are numerous, dynamic and depot‐dependent. © 2017 American Physiological Society. Compr Physiol 7:819‐840, 2017.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. AT depots in mice. Locations and classifications of four major WAT depots in a male mouse are shown on the left image. Mesenteric fat (top left) runs along the intestines and is, by strictest definition, a true visceral depot. The inguinal fat pad (bottom left) is classified as subQ and resides just beneath the skin. The epididymal depot (bottom right) is located next to the testes and has been moved outside of the abdominal space for better visualization. While epididymal fat is given an intra‐abdominal (IA) designation and sometimes referred to as visceral, many do not consider it a true visceral depot due to lack of drainage into the portal vein. The retroperitoneal depot (top right) is also visible, located behind the kidney (K). The location of the interscapular BAT depot is shown on the right. This is the only dissectible BAT fat pad in mice. Figure on the left was adapted from Sackmann‐Sala et al. (185) and reused from Berryman et al. (26) with permission.
Figure 2. Figure 2. AT depots in humans. Several prominent human AT depots are illustrated. Subcutaneous depots include gluteal, femoral, and two distinct abdominal fat pads, one superficial and one deep. Intraabdominal depots in humans include intraperitoneal, also known as visceral, and retroperitoneal AT. Perinephric fat surrounds the kidney, and retroperitoneal fat is located in the retroperitoneal space behind the kidneys. Visceral depots in humans line organs of the digestive system and include mesenteric and omental AT. Reused with permission from Lee et al. (112).
Figure 3. Figure 3. Changes accompanying pathological WAT expansion in obesity. Expansion of healthy WAT (left) mainly occurs through adipocyte hyperplasia. At this stage, the tissue exists in an anti‐inflammatory state, characterized by higher levels of M2 macrophage populations as well as T regulatory cells and T helper cells. The ECM remains loose, allowing for unhindered tissue expansion. Vascularization is also adequate to support the tissue. Conversely, pathological expansion, as seen in obesity (right), favors hypertrophy of adipocytes and exhibits a pro‐inflammatory profile, hallmarked by increased infiltration of M1 macrophages, cytotoxic T cells, and natural killer cells, with decreased T regulatory cells and T helper cells. Expression of pro‐inflammatory cytokines TNF‐α, IL‐6, and MCP‐1 are also increased in this state. The ECM becomes rigid and deposition increases, leading to a state of fibrosis. Vascularization is insufficient to support the growing tissue, which results in hypoxia and ultimately adipocyte death. Adapted with permission from (21).
Figure 4. Figure 4. Regulation of pituitary GH. The hypothalamus secretes GHRH and somatostatin (SST), which stimulate and inhibit GH secretion, respectively. GH induced intracellular signaling via the GH receptor results in IGF‐1 production by target tissues, which acts, along with GH, to decrease GH secretion through feedback inhibition. Other factors such as estrogen, leptin, and free fatty acids (FFAs) also modulate GH secretion. Adapted with permission from (21,95).
Figure 5. Figure 5. The intracellular signaling cascade of GHR in response to GH binding. The canonical pathway consists of GHR‐JAK2‐STAT5. Phosphorylation of STAT5 in response to GH activates STAT5, which regulates target gene transcription. GHR also signals through IRS to activate mTOR and through Src to activate ERK and JNK.
Figure 6. Figure 6. Mice with altered GH action in a C57BL/6J genetic background. From top to bottom: a wild‐type (WT) mouse, a bGH transgenic mouse with elevated levels of GH action, a GHA transgenic mouse with decreased levels of GH action and a GHR gene disrupted mouse with no GH action via the GHR. Reused with permission from (26).
Figure 7. Figure 7. Comparison of body fat percentage over time in mice with altered GH action. Male and female bGH mice have greater body fat percent than WT controls earlier in life, a trend which begins to reverse at 4 and 6 months of age, respectively. Percent fat mass is greater in male and female GHA mice compared to controls at all‐time points measured and continues to increase throughout life. Male GHR−/− mice have markedly increased body fat percent compared to controls and appear to rapidly accumulate fat during the first 4 months of life. Increases in percent fat are also observed in female GHR−/− mice respective to controls, though the differences are not as dramatic. Select data presented from previously published works with permission from (25,27,160).
Figure 8. Figure 8. Comparison of collagen staining in AT. SubQ AT from 6‐month‐old bGH, WT controls and GHR−/− mice stained with picrosirius red, a commonly used histological technique to visualize collagen in paraffin‐embedded tissue sections.


Figure 1. AT depots in mice. Locations and classifications of four major WAT depots in a male mouse are shown on the left image. Mesenteric fat (top left) runs along the intestines and is, by strictest definition, a true visceral depot. The inguinal fat pad (bottom left) is classified as subQ and resides just beneath the skin. The epididymal depot (bottom right) is located next to the testes and has been moved outside of the abdominal space for better visualization. While epididymal fat is given an intra‐abdominal (IA) designation and sometimes referred to as visceral, many do not consider it a true visceral depot due to lack of drainage into the portal vein. The retroperitoneal depot (top right) is also visible, located behind the kidney (K). The location of the interscapular BAT depot is shown on the right. This is the only dissectible BAT fat pad in mice. Figure on the left was adapted from Sackmann‐Sala et al. (185) and reused from Berryman et al. (26) with permission.


Figure 2. AT depots in humans. Several prominent human AT depots are illustrated. Subcutaneous depots include gluteal, femoral, and two distinct abdominal fat pads, one superficial and one deep. Intraabdominal depots in humans include intraperitoneal, also known as visceral, and retroperitoneal AT. Perinephric fat surrounds the kidney, and retroperitoneal fat is located in the retroperitoneal space behind the kidneys. Visceral depots in humans line organs of the digestive system and include mesenteric and omental AT. Reused with permission from Lee et al. (112).


Figure 3. Changes accompanying pathological WAT expansion in obesity. Expansion of healthy WAT (left) mainly occurs through adipocyte hyperplasia. At this stage, the tissue exists in an anti‐inflammatory state, characterized by higher levels of M2 macrophage populations as well as T regulatory cells and T helper cells. The ECM remains loose, allowing for unhindered tissue expansion. Vascularization is also adequate to support the tissue. Conversely, pathological expansion, as seen in obesity (right), favors hypertrophy of adipocytes and exhibits a pro‐inflammatory profile, hallmarked by increased infiltration of M1 macrophages, cytotoxic T cells, and natural killer cells, with decreased T regulatory cells and T helper cells. Expression of pro‐inflammatory cytokines TNF‐α, IL‐6, and MCP‐1 are also increased in this state. The ECM becomes rigid and deposition increases, leading to a state of fibrosis. Vascularization is insufficient to support the growing tissue, which results in hypoxia and ultimately adipocyte death. Adapted with permission from (21).


Figure 4. Regulation of pituitary GH. The hypothalamus secretes GHRH and somatostatin (SST), which stimulate and inhibit GH secretion, respectively. GH induced intracellular signaling via the GH receptor results in IGF‐1 production by target tissues, which acts, along with GH, to decrease GH secretion through feedback inhibition. Other factors such as estrogen, leptin, and free fatty acids (FFAs) also modulate GH secretion. Adapted with permission from (21,95).


Figure 5. The intracellular signaling cascade of GHR in response to GH binding. The canonical pathway consists of GHR‐JAK2‐STAT5. Phosphorylation of STAT5 in response to GH activates STAT5, which regulates target gene transcription. GHR also signals through IRS to activate mTOR and through Src to activate ERK and JNK.


Figure 6. Mice with altered GH action in a C57BL/6J genetic background. From top to bottom: a wild‐type (WT) mouse, a bGH transgenic mouse with elevated levels of GH action, a GHA transgenic mouse with decreased levels of GH action and a GHR gene disrupted mouse with no GH action via the GHR. Reused with permission from (26).


Figure 7. Comparison of body fat percentage over time in mice with altered GH action. Male and female bGH mice have greater body fat percent than WT controls earlier in life, a trend which begins to reverse at 4 and 6 months of age, respectively. Percent fat mass is greater in male and female GHA mice compared to controls at all‐time points measured and continues to increase throughout life. Male GHR−/− mice have markedly increased body fat percent compared to controls and appear to rapidly accumulate fat during the first 4 months of life. Increases in percent fat are also observed in female GHR−/− mice respective to controls, though the differences are not as dramatic. Select data presented from previously published works with permission from (25,27,160).


Figure 8. Comparison of collagen staining in AT. SubQ AT from 6‐month‐old bGH, WT controls and GHR−/− mice stained with picrosirius red, a commonly used histological technique to visualize collagen in paraffin‐embedded tissue sections.
References
 1.Abreu A, Tovar AP, Castellanos R, Valenzuela A, Giraldo CM, Pinedo AC, Guerrero DP, Barrera CA, Franco HI, Ribeiro‐Oliveira A, Jr., Vilar L, Jallad RS, Duarte FG, Gadelha M, Boguszewski CL, Abucham J, Naves LA, Musolino NR, de Faria ME, Rossato C, Bronstein MD. Challenges in the diagnosis and management of acromegaly: A focus on comorbidities. Pituitary 19: 448‐457, 2016.
 2.Agladioglu SY, Cetinkaya S, Savas Erdeve S, Onder A, Kendirci HN, Bas VN, Aycan Z. Diabetes mellitus with Laron syndrome: Case report. J Pediatr Endocrinol Metab 26: 955‐958, 2013.
 3.Al‐Regaiey KA, Masternak MM, Bonkowski M, Sun L, Bartke A. Long‐lived growth hormone receptor knockout mice: Interaction of reduced insulin‐like growth factor i/insulin signaling and caloric restriction. Endocrinology 146: 851‐860, 2005.
 4.Alatzoglou KS, Webb EA, Le Tissier P, Dattani MT. Isolated growth hormone deficiency (GHD) in childhood and adolescence: Recent advances. Endocr Rev 35: 376‐432, 2014.
 5.Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, Hotta K, Shimomura I, Nakamura T, Miyaoka K, Kuriyama H, Nishida M, Yamashita S, Okubo K, Matsubara K, Muraguchi M, Ohmoto Y, Funahashi T, Matsuzawa Y. Paradoxical decrease of an adipose‐specific protein, adiponectin, in obesity. Biochem Biophys Res Commun 257: 79‐83, 1999.
 6.Arner P, Andersson DP, Thorne A, Wiren M, Hoffstedt J, Naslund E, Thorell A, Ryden M. Variations in the size of the major omentum are primarily determined by fat cell number. J Clin Endocrinol Metab 98: E897‐901, 2013.
 7.Asada N, Takahashi Y, Honjo M. Effects of 22K or 20K human growth hormone on lipolysis, leptin production in adipocytes in the presence and absence of human growth hormone binding protein. Horm Res 54: 203‐207, 2000.
 8.Asayama K, Amemiya S, Kusano S, Kato K. Growth‐hormone‐induced changes in postheparin plasma lipoprotein lipase and hepatic triglyceride lipase activities. Metabolism 33: 129‐131, 1984.
 9.Bado A, Levasseur S, Attoub S, Kermorgant S, Laigneau JP, Bortoluzzi MN, Moizo L, Lehy T, Guerre‐Millo M, Le Marchand‐Brustel Y, Lewin MJM. The stomach is a source of leptin. Nature 394: 790‐793, 1998.
 10.Baldelli R, Durante C, D'Amico E, Diacono F, Tamburrano G, Casanueva FF. Serum leptin levels in acromegalic patients before and during somatostatin analogs therapy. J Endocrinol Invest 26: 1219‐1224, 2003.
 11.Bamshad M, Song CK, Bartness TJ. CNS origins of the sympathetic nervous system outflow to brown adipose tissue. Am J Physiol 276: R1569‐1578, 1999.
 12.Barnea M, Shamay A, Stark AH, Madar Z. A high‐fat diet has a tissue‐specific effect on adiponectin and related enzyme expression. Obesity 14: 2145‐2153, 2006.
 13.Bartke A. Can growth hormone (GH) accelerate aging? Evidence from GH‐transgenic mice. Neuroendocrinology 78: 210‐216, 2003.
 14.Bartke A, Brown‐Borg H. Life extension in the dwarf mouse. Curr Top Dev Biol 63: 189‐225, 2004.
 15.Bartke A, Westbrook R. Metabolic characteristics of long‐lived mice. Front Genet 3: 288, 2012.
 16.Baum HB, Biller BM, Finkelstein JS, Cannistraro KB, Oppenhein DS, Schoenfeld DA, Michel TH, Wittink H, Klibanski A. Effects of physiologic growth hormone therapy on bone density and body composition in patients with adult‐onset growth hormone deficiency. A randomized, placebo‐controlled trial. Ann Intern Med 125: 883‐890, 1996.
 17.Beauregard C, Utz A, Schaub AE, Nachtigall L, Biller BM, Miller KK, Klibanski A. Growth hormone decreases visceral fat and improves cardiovascular risk markers in women with hypopituitarism: A randomized, placebo‐controlled study. J Clin Endocrinol Metab 93: 2063‐2071, 2008.
 18.Benencia F, Harshman S, Duran‐Ortiz S, Lubbers ER, List EO, Householder L, Al‐Naeeli M, Liang X, Welch L, Kopchick JJ, Berryman DE. Male bovine GH transgenic mice have decreased adiposity with an adipose depot‐specific increase in immune cell populations. Endocrinology 156: 1794‐1803, 2015.
 19.Bengtsson BA, Brummer RJ, Eden S, Bosaeus I, Lindstedt G. Body composition in acromegaly: The effect of treatment. Clin Endocrinol (Oxf) 31: 481‐490, 1989.
 20.Bengtsson BA, Eden S, Lonn L, Kvist H, Stokland A, Lindstedt G, Bosaeus I, Tolli J, Sjostrom L, Isaksson OG. Treatment of adults with growth hormone (GH) deficiency with recombinant human GH. J Clin Endocrinol Metab 76: 309‐317, 1993.
 21.Berryman D, Householder L, Lesende V, List E, Kopchick JJ. Living large: What mouse models reveal about growth hormone. In: Berger NA, editor. Murine Models, Energy, Balance, and Cancer. New York: Springer, 2015, p. 65‐95.
 22.Berryman DE, Glad CA, List EO, Johannsson G. The GH/IGF‐1 axis in obesity: Pathophysiology and therapeutic considerations. Nat Rev Endocrinol 9: 346‐356, 2013.
 23.Berryman DE, List EO, Coschigano KT, Behar K, Kim JK, Kopchick JJ. Comparing adiposity profiles in three mouse models with altered GH signaling. Growth Horm IGF Res 14: 309‐318, 2004.
 24.Berryman DE, List EO, Kohn DT, Coschigano KT, Seeley RJ, Kopchick JJ. Effect of growth hormone on susceptibility to diet‐induced obesity. Endocrinology 147: 2801‐2808, 2006.
 25.Berryman DE, List EO, Palmer AJ, Chung MY, Wright‐Piekarski J, Lubbers E, O'Connor P, Okada S, Kopchick JJ. Two‐year body composition analyses of long‐lived GHR null mice. J Gerontol A Biol Sci Med Sci 65: 31‐40, 2010.
 26.Berryman DE, List EO, Sackmann‐Sala L, Lubbers E, Munn R, Kopchick JJ. Growth hormone and adipose tissue: beyond the adipocyte. Growth Horm IGF Res 21: 113‐123, 2011.
 27.Berryman DE, Lubbers ER, Magon V, List EO, Kopchick JJ. A dwarf mouse model with decreased GH/IGF‐1 activity that does not experience life‐span extension: potential impact of increased adiposity, leptin, and insulin with advancing age. J Gerontol A Biol Sci Med Sci 69: 131‐141, 2014.
 28.Boot AM, Engels MA, Boerma GJ, Krenning EP, De Muinck Keizer‐Schrama SM. Changes in bone mineral density, body composition, and lipid metabolism during growth hormone (GH) treatment in children with GH deficiency. J Clin Endocrinol Metab 82: 2423‐2428, 1997.
 29.Borland CA, Barber MC, Travers MT, Vernon RG. Growth hormone inhibition of lipogenesis in sheep adipose tissue: Requirement for gene transcription and polyamines. J Endocrinol 142: 235‐243, 1994.
 30.Brooks AJ, Waters MJ. The growth hormone receptor: Mechanism of activation and clinical implications. Nat Rev Endocrinol 6: 515‐525, 2010.
 31.Brown RJ, Adams JJ, Pelekanos RA, Wan Y, McKinstry WJ, Palethorpe K, Seeber RM, Monks TA, Eidne KA, Parker MW, Waters MJ. Model for growth hormone receptor activation based on subunit rotation within a receptor dimer. Nat Struct Mol Biol 12: 814‐821, 2005.
 32.Carro E, Senaris R, Considine RV, Casanueva FF, Dieguez C. Regulation of in vivo growth hormone secretion by leptin. Endocrinology 138: 2203‐2206, 1997.
 33.Chang YH, Chang DM, Lin KC, Shin SJ, Lee YJ. Visfatin in overweight/obesity, type 2 diabetes mellitus, insulin resistance, metabolic syndrome and cardiovascular diseases: a meta‐analysis and systemic review. Diabetes Metab Res Rev 27: 515‐527, 2011.
 34.Chapman IM, Bach MA, Van Cauter E, Farmer M, Krupa D, Taylor AM, Schilling LM, Cole KY, Skiles EH, Pezzoli SS, Hartman ML, Veldhuis JD, Gormley GJ, Thorner MO. Stimulation of the growth hormone (GH)‐insulin‐like growth factor I axis by daily oral administration of a GH secretogogue (MK‐677) in healthy elderly subjects. J Clin Endocrinol Metab 81: 4249‐4257, 1996.
 35.Chen, Brychta RJ, Linderman JD, Smith S, Courville A, Dieckmann W, Herscovitch P, Millo CM, Remaley A, Lee P, Celi FS. Brown fat activation mediates cold‐induced thermogenesis in adult humans in response to a mild decrease in ambient temperature. J Clin Endocrinol Metab 98: E1218‐1223, 2013.
 36.Chen H, Charlat O, Tartaglia LA, Woolf EA, Weng X, Ellis SJ, Lakey ND, Culpepper J, Moore KJ, Breitbart RE, Duyk GM, Tepper RI, Morgenstern JP. Evidence that the diabetes gene encodes the leptin receptor: Identification of a mutation in the leptin receptor gene in db/db mice. Cell 84: 491‐495, 1996.
 37.Chen WY, White ME, Wagner TE, Kopchick JJ. Functional antagonism between endogenous mouse growth hormone (GH) and a GH analog results in dwarf transgenic mice. Endocrinology 129: 1402‐1408, 1991.
 38.Chen WY, Wight DC, Chen NY, Coleman TA, Wagner TE, Kopchick JJ. Mutations in the third alpha‐helix of bovine growth hormone dramatically affect its intracellular distribution in vitro and growth enhancement in transgenic mice. J Biol Chem 266: 2252‐2258, 1991.
 39.Chen WY, Wight DC, Mehta BV, Wagner TE, Kopchick JJ. Glycine 119 of bovine growth hormone is critical for growth‐promoting activity. Mol Endocrinol 5: 1845‐1852, 1991.
 40.Cinti S. The adipose organ. Prostaglandins Leukot Essent Fatty Acids 73: 9‐15, 2005.
 41.Ciresi A, Amato MC, Pizzolanti G, Giordano C. Serum visfatin levels in acromegaly: Correlation with disease activity and metabolic alterations. Growth Horm IGF Res 25: 240‐246, 2015.
 42.Ciresi A, Pizzolanti G, Leotta M, Guarnotta V, Teresi G, Giordano C. Resistin, visfatin, leptin and omentin are differently related to hormonal and metabolic parameters in growth hormone‐deficient children. J Endocrinol Invest, 2016.
 43.Ciresi A, Radellini S, Guarnotta V, Giordano C. The visceral adiposity index is associated with insulin sensitivity and IGF‐I levels in adults with growth hormone deficiency. Endocrine, 2016.
 44.Clement K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, Gourmelen M, Dina C, Chambaz J, Lacorte JM, Basdevant A, Bougneres P, Lebouc Y, Froguel P, Guy‐Grand B. A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392: 398‐401, 1998.
 45.Comisford R, Lubbers ER, Householder LA, Suer O, Tchkonia T, Kirkland JL, List EO, Kopchick JJ, Berryman DE. 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 62: 163‐172, 2015.
 46.Coschigano KT, Holland AN, Riders ME, List EO, Flyvbjerg A, Kopchick JJ. Deletion, but not antagonism, of the mouse growth hormone receptor results in severely decreased body weights, insulin and IGF‐1 levels and increased lifespan. Endocrinology 144: 3799‐3810, 2003.
 47.Curat CA, Wegner V, Sengenes C, Miranville A, Tonus C, Busse R, Bouloumie A. Macrophages in human visceral adipose tissue: Increased accumulation in obesity and a source of resistin and visfatin. Diabetologia 49: 744‐747, 2006.
 48.Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, Kuo FC, Palmer EL, Tseng YH, Doria A, Kolodny GM, Kahn CR. Identification and importance of brown adipose tissue in adult humans. N Engl J Med 360: 1509‐1517, 2009.
 49.Cypess AM, White AP, Vernochet C, Schulz TJ, Xue R, Sass CA, Huang TL, Roberts‐Toler C, Weiner LS, Sze C, Chacko AT, Deschamps LN, Herder LM, Truchan N, Glasgow AL, Holman AR, Gavrila A, Hasselgren PO, Mori MA, Molla M, Tseng YH. Anatomical localization, gene expression profiling and functional characterization of adult human neck brown fat. Nat Med 19: 635‐639, 2013.
 50.De Boer H, Blok GJ, Voerman HJ, De Vries PM, van der Veen EA. Body composition in adult growth hormone‐deficient men, assessed by anthropometry and bioimpedance analysis. J Clin Endocrinol Metab 75: 833‐837, 1992.
 51.de Jesus LA, Carvalho SD, Ribeiro MO, Schneider M, Kim SW, Harney JW, Larsen PR, Bianco AC. The type 2 iodothyronine deiodinase is essential for adaptive thermogenesis in brown adipose tissue. J Clin Invest 108: 1379‐1385, 2001.
 52.de Souza Batista CM, Yang RZ, Lee MJ, Glynn NM, Yu DZ, Pray J, Ndubuizu K, Patil S, Schwartz A, Kligman M, Fried SK, Gong DW, Shuldiner AR, Pollin TI, McLenithan JC. Omentin plasma levels and gene expression are decreased in obesity. Diabetes 56: 1655‐1661, 2007.
 53.Di Somma C, Ciresi A, Amato MC, Savastano S, Savanelli MC, Scarano E, Colao A, Giordano C. Alteration of the growth hormone axis, visceral fat dysfunction, and early cardiometabolic risk in adults: The role of the visceral adiposity index. Endocrine 49: 492‐502, 2015.
 54.Doessing S, Holm L, Heinemeier KM, Feldt‐Rasmussen U, Schjerling P, Qvortrup K, Larsen JO, Nielsen RH, Flyvbjerg A, Kjaer M. GH and IGF1 levels are positively associated with musculotendinous collagen expression: Experiments in acromegalic and GH deficiency patients. Eur J Endocrinol 163: 853‐862, 2010.
 55.Donahue LR, Beamer WG. Growth hormone deficiency in ‘little’ mice results in aberrant body composition, reduced insulin‐like growth factor‐I and insulin‐like growth factor‐binding protein‐3 (IGFBP‐3), but does not affect IGFBP‐2, ‐1 or ‐4. J Endocrinol 136: 91‐104, 1993.
 56.dos Santos MC, Nascimento GC, Nascimento AG, Carvalho VC, Lopes MH, Montenegro R, Montenegro R, Jr., Vilar L, Albano MF, Alves AR, Parente CV, dos Santos Faria M. Thyroid cancer in patients with acromegaly: A case‐control study. Pituitary 16: 109‐114, 2013.
 57.Drake WM, Coyte D, Camacho‐Hubner C, Jivanji NM, Kaltsas G, Wood DF, Trainer PJ, Grossman AB, Besser GM, Monson JP. Optimizing growth hormone replacement therapy by dose titration in hypopituitary adults. J Clin Endocrinol Metab 83: 3913‐3919, 1998.
 58.Enerback S, Jacobsson A, Simpson EM, Guerra C, Yamashita H, Harper ME, Kozak LP. Mice lacking mitochondrial uncoupling protein are cold‐sensitive but not obese. Nature 387: 90‐94, 1997.
 59.Fantuzzi G. Adiponectin and inflammation: Consensus and controversy. J Allergy Clin Immunol 121: 326‐330, 2008.
 60.Farooqi IS, Keogh JM, Kamath S, Jones S, Gibson WT, Trussell R, Jebb SA, Lip GY, O'Rahilly S. Partial leptin deficiency and human adiposity. Nature 414: 34‐35, 2001.
 61.Fei H, Okano HJ, Li C, Lee GH, Zhao C, Darnell R, Friedman JM. Anatomic localization of alternatively spliced leptin receptors (Ob‐R) in mouse brain and other tissues. Proc Natl Acad Sci U S A 94: 7001‐7005, 1997.
 62.Fernandez‐Perez L, Guerra B, Diaz‐Chico JC, Flores‐Morales A. Estrogens regulate the hepatic effects of growth hormone, a hormonal interplay with multiple fates. Front Endocrinol (Lausanne) 4: 66, 2013.
 63.Flint DJ, Binart N, Boumard S, Kopchick JJ, Kelly P. 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 191: 101‐111, 2006.
 64.Florini JR, Ewton DZ, Coolican SA. Growth hormone and the insulin‐like growth factor system in myogenesis. Endocr Rev 17: 481‐517, 1996.
 65.Franco C, Brandberg J, Lonn L, Andersson B, Bengtsson BA, Johannsson G. Growth hormone treatment reduces abdominal visceral fat in postmenopausal women with abdominal obesity: a 12‐month placebo‐controlled trial. J Clin Endocrinol Metab 90: 1466‐1474, 2005.
 66.Freda PU, Shen W, Heymsfield SB, Reyes‐Vidal CM, Geer EB, Bruce JN, Gallagher D. Lower visceral and subcutaneous but higher intermuscular adipose tissue depots in patients with growth hormone and insulin‐like growth factor I excess due to acromegaly. J Clin Endocrinol Metab 93: 2334‐2343, 2008.
 67.Gibney J, Wolthers T, Burt MG, Leung KC, Umpleby AM, Ho KK. Protein metabolism in acromegaly: Differential effects of short‐ and long‐term treatment. J Clin Endocrinol Metab 92: 1479‐1484, 2007.
 68.Gilchrist FJ, Murray RD, Shalet SM. The effect of long‐term untreated growth hormone deficiency (GHD) and 9 years of GH replacement on the quality of life (QoL) of GH‐deficient adults. Clin Endocrinol (Oxf) 57: 363‐370, 2002.
 69.Ginsberg S, Laron Z, Bed MA, Vaisman N. The obesity of patients with Laron Syndrome is not associated with excessive nutritional intake. Obes Res Clin Pract 3: 1‐52, 2009.
 70.Green ED, Maffei M, Braden VV, Proenca R, DeSilva U, Zhang Y, Chua SC, Jr., Leibel RL, Weissenbach J, Friedman JM. The human obese (OB) gene: RNA expression pattern and mapping on the physical, cytogenetic, and genetic maps of chromosome 7. Genome Res 5: 5‐12, 1995.
 71.Green H, Morikawa M, Nixon T. A dual effector theory of growth‐hormone action. Differentiation 29: 195‐198, 1985.
 72.Guevara‐Aguirre J, Balasubramanian P, Guevara‐Aguirre M, Wei M, Madia F, Cheng CW, Hwang D, Martin‐Montalvo A, Saavedra J, Ingles S, de Cabo R, Cohen P, Longo VD. Growth hormone receptor deficiency is associated with a major reduction in pro‐aging signaling, cancer, and diabetes in humans. Sci Transl Med 3: 70ra13, 2011.
 73.Hada Y, Yamauchi T, Waki H, Tsuchida A, Hara K, Yago H, Miyazaki O, Ebinuma H, Kadowaki T. Selective purification and characterization of adiponectin multimer species from human plasma. Biochem Biophys Res Commun 356: 487‐493, 2007.
 74.Hara K, Horikoshi M, Yamauchi T, Yago H, Miyazaki O, Ebinuma H, Imai Y, Nagai R, Kadowaki T. Measurement of the high‐molecular weight form of adiponectin in plasma is useful for the prediction of insulin resistance and metabolic syndrome. Diabetes Care 29: 1357‐1362, 2006.
 75.Heiman ML, Tinsley FC, Mattison JA, Hauck S, Bartke A. Body composition of prolactin‐, growth hormone, and thyrotropin‐deficient Ames dwarf mice. Endocrine 20: 149‐154, 2003.
 76.Hioki C, Yoshida T, Kogure A, Takakura Y, Umekawa T, Yoshioka K, Shimatsu A, Yoshikawa T. Effects of growth hormone (GH) on mRNA levels of uncoupling proteins 1, 2, and 3 in brown and white adipose tissues and skeletal muscle in obese mice. Horm Metab Res 36: 607‐613, 2004.
 77.Hirose H, Yamamoto Y, Seino‐Yoshihara Y, Kawabe H, Saito I. Serum high‐molecular‐weight adiponectin as a marker for the evaluation and care of subjects with metabolic syndrome and related disorders. J Atheroscler Thromb 17: 1201‐1211.
 78.Hochberg I, Tran QT, Barkan AL, Saltiel AR, Chandler WF, Bridges D. Gene expression signature in adipose tissue of acromegaly patients. PLoS One 10: e0129359, 2015.
 79.Holdaway IM, Bolland MJ, Gamble GD. A meta‐analysis of the effect of lowering serum levels of GH and IGF‐I on mortality in acromegaly. Eur J Endocrinol 159: 89‐95, 2008.
 80.Houssay B. The hypophysis and metabolism. N Engl J Med 214: 961‐985, 1936.
 81.Ibrahim MM. Subcutaneous and visceral adipose tissue: structural and functional differences. Obes Rev 11: 11‐18, 2010.
 82.Ikeno Y, Hubbard GB, Lee S, Cortez LA, Lew CM, Webb CR, Berryman DE, List EO, Kopchick JJ, Bartke A. Reduced incidence and delayed occurrence of fatal neoplastic diseases in growth hormone receptor/binding protein knockout mice. J Gerontol A Biol Sci Med Sci 64: 522‐529, 2009.
 83.Iranmanesh A, Lizarralde G, Veldhuis JD. Age and relative adiposity are specific negative determinants of the frequency and amplitude of growth hormone (GH) secretory bursts and the half‐life of endogenous GH in healthy men. J Clin Endocrinol Metab 73: 1081‐1088, 1991.
 84.Isgaard J, Carlsson L, Isaksson OG, Jansson JO. Pulsatile intravenous growth hormone (GH) infusion to hypophysectomized rats increases insulin‐like growth factor I messenger ribonucleic acid in skeletal tissues more effectively than continuous GH infusion. Endocrinology 123: 2605‐2610, 1988.
 85.Jamaluddin MS, Weakley SM, Yao Q, Chen C. Resistin: functional roles and therapeutic considerations for cardiovascular disease. Br J Pharmacol 165: 622‐632, 2012.
 86.Jeffery E, Wing A, Holtrup B, Sebo Z, Kaplan JL, Saavedra‐Pena R, Church CD, Colman L, Berry R, Rodeheffer MS. The adipose tissue microenvironment regulates depot‐specific adipogenesis in obesity. Cell Metab 24: 142‐150, 2016.
 87.Jenkins PJ. Cancers associated with acromegaly. Neuroendocrinology 83: 218‐223, 2006.
 88.Kanety H, Hemi R, Ginsberg S, Pariente C, Yissachar E, Barhod E, Funahashi T, Laron Z. Total and high molecular weight adiponectin are elevated in patients with Laron syndrome despite marked obesity. Eur J Endocrinol 161: 837‐844, 2009.
 89.Karastergiou K, Bredella MA, Lee MJ, Smith SR, Fried SK, Miller KK. Growth hormone receptor expression in human gluteal versus abdominal subcutaneous adipose tissue: Association with body shape. Obesity 24: 1090‐1096, 2016.
 90.Kelder B, Berryman DE, Clark R, Li A, List EO, Kopchick JJ. CIDE‐A gene expression is decreased in white adipose tissue of growth hormone receptor/binding protein gene disrupted mice and with high‐fat feeding of normal mice. Growth Horm IGF Res 17: 346‐351, 2007.
 91.Khadilkar V, Ekbote V, Kajale N, Khadilkar A, Chiplonkar S, Kinare A. Effect of one‐year growth hormone therapy on body composition and cardio‐metabolic risk in Indian children with growth hormone deficiency. Endocr Res 39: 73‐78, 2014.
 92.Khalfallah Y, Sassolas G, Borson‐Chazot F, Vega N, Vidal H. Expression of insulin target genes in skeletal muscle and adipose tissue in adult patients with growth hormone deficiency: Effect of one year recombinant human growth hormone therapy. J Endocrinol 171: 285‐292, 2001.
 93.Khan T, Muise ES, Iyengar P, Wang ZV, Chandalia M, Abate N, Zhang BB, Bonaldo P, Chua S, Scherer PE. Metabolic dysregulation and adipose tissue fibrosis: Role of collagen VI. Mol Cell Biol 29: 1575‐1591, 2009.
 94.Kim JY, van de Wall E, Laplante M, Azzara A, Trujillo ME, Hofmann SM, Schraw T, Durand JL, Li H, Li G, Jelicks LA, Mehler MF, Hui DY, Deshaies Y, Shulman GI, Schwartz GJ, Scherer PE. Obesity‐associated improvements in metabolic profile through expansion of adipose tissue. J Clin Invest 117: 2621‐2637, 2007.
 95.Kopchick JJ, Andry JM. Growth hormone (GH), GH receptor, and signal transduction. Mol Genet Metab 71: 293‐314, 2000.
 96.Kopchick JJ, Bellush LL, Coschigano KT. Transgenic models of growth hormone action. Annu Rev Nutr 19: 437‐461, 1999.
 97.Kopchick JJ, Laron Z. Laron Syndrome—From Man to Mouse. Berlin: Springer, 2011.
 98.Kopchick JJ, List EO, Kelder B, Gosney ES, Berryman DE. Evaluation of growth hormone (GH) action in mice: Discovery of GH receptor antagonists and clinical indications. Mol Cell Endocrinol 386: 34‐45, 2014.
 99.Kreitschmann‐Andermahr I, Suarez P, Jennings R, Evers N, Brabant G. GH/IGF‐I regulation in obesity–‐mechanisms and practical consequences in children and adults. Horm Res Paediatr 73: 153‐160, 2010.
 100.Kusminski CM, Bickel PE, Scherer PE. Targeting adipose tissue in the treatment of obesity‐associated diabetes. Nat Rev Drug Discov 15: 639‐660, 2016.
 101.Lam KS, Xu A, Tan KC, Wong LC, Tiu SC, Tam S. Serum adiponectin is reduced in acromegaly and normalized after correction of growth hormone excess. J Clin Endocrinol Metab 89: 5448‐5453, 2004.
 102.Laron Z. Emerging treatment options for patients with Laron syndrome. Expert Opin Orphan Drugs 2: 681‐694, 2014.
 103.Laron Z. Lessons from 50 Years of Study of Laron Syndrome. Endocr Pract 21: 1395‐1402, 2015.
 104.Laron Z, Blum W, Chatelain P, Ranke M, Rosenfeld R, Savage M, Underwood L. Classification of growth hormone insensitivity syndrome [editorial]. J Pediatr 122: 241, 1993.
 105.Laron Z, Ginsberg S, Lilos P, Arbiv M, Vaisman N. Body composition in untreated adult patients with Laron syndrome (primary GH insensitivity). Clin Endocrinol (Oxf) 65: 114‐117, 2006.
 106.Laron Z, Ginsberg S, Lilos P, Arbiv M, Vaisman N. Long‐term IGF‐I treatment of children with Laron syndrome increases adiposity. Growth Horm IGF Res 16: 61‐64, 2006.
 107.Laron Z, Kauli R. Fifty seven years of follow‐up of the Israeli cohort of Laron Syndrome patients‐From discovery to treatment. Growth Horm IGF Res 28: 53‐56, 2016.
 108.Laron Z, Klinger B. IGF‐I treatment of adult patients with Laron syndrome: Preliminary results. Clin Endocrinol (Oxf) 41: 631‐638, 1994.
 109.Laron Z, Kopchick J. Laron Syndrome ‐ From Man to Mouse Lessons from Clinical and Experimental Experience. Berlin; New York: Springer, 2011, p. 1 online resource (xiv, 531 p.).
 110.Laron Z, Pertzelan A, Karp M. Pituitary dwarfism with high serum levels of growth hormone. Isr J Med Sci 4: 883‐894, 1968.
 111.Laron Z, Silbergeld A, Lilos P, Blum FW. Serum leptin in obese patients with Laron syndrome before and during IGF‐I treatment. J Pediatr Endocrinol Metab 11: 653‐656, 1998.
 112.Lee MJ, Wu Y, Fried SK. Adipose tissue heterogeneity: Implication of depot differences in adipose tissue for obesity complications. Mol Aspects Med 34: 1‐11, 2013.
 113.Lewitt MS, Hilding A, Brismar K, Efendic S, Ostenson CG, Hall K. IGF‐binding protein 1 and abdominal obesity in the development of type 2 diabetes in women. Eur J Endocrinol 163: 233‐242, 2010.
 114.Li S, Shin HJ, Ding EL, van Dam RM. Adiponectin levels and risk of type 2 diabetes: A systematic review and meta‐analysis. JAMA 302: 179‐188, 2009.
 115.Li Y, Knapp JR, Kopchick JJ. Enlargement of interscapular brown adipose tissue in growth hormone antagonist transgenic and in growth hormone receptor gene‐disrupted dwarf mice. Exp Biol Med (Maywood) 228: 207‐215, 2003.
 116.Licinio J, Caglayan S, Ozata M, Yildiz BO, de Miranda PB, O'Kirwan F, Whitby R, Liang L, Cohen P, Bhasin S, Krauss RM, Veldhuis JD, Wagner AJ, DePaoli AM, McCann SM, Wong ML. Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin‐deficient adults. Proc Natl Acad Sci U S A 101: 4531‐4536, 2004.
 117.Lidell ME, Betz MJ, Dahlqvist Leinhard O, Heglind M, Elander L, Slawik M, Mussack T, Nilsson D, Romu T, Nuutila P, Virtanen KA, Beuschlein F, Persson A, Borga M, Enerback S. Evidence for two types of brown adipose tissue in humans. Nat Med 19: 631‐634, 2013.
 118.Lidell ME, Betz MJ, Enerback S. Brown adipose tissue and its therapeutic potential. J Intern Med 276: 364‐377, 2014.
 119.Lim S, Honek J, Xue Y, Seki T, Cao Z, Andersson P, Yang X, Hosaka K, Cao Y. Cold‐induced activation of brown adipose tissue and adipose angiogenesis in mice. Nat Protoc 7: 606‐615, 2012.
 120.List EO, Berryman DE, Funk K, Gosney ES, Jara A, Kelder B, Wang X, Kutz L, Troike K, Lozier N, Mikula V, Lubbers ER, Zhang H, Vesel C, Junnila RK, Frank SJ, Masternak MM, Bartke A, Kopchick JJ. The role of GH in adipose tissue: Lessons from adipose‐specific GH receptor gene‐disrupted mice. Mol Endocrinol 27: 524‐535, 2013.
 121.List EO, Berryman DE, Funk K, Jara A, Kelder B, Wang F, Stout MB, Zhi X, Sun L, White TA, LeBrasseur NK, Pirtskhalava T, Tchkonia T, Jensen EA, Zhang W, Masternak MM, Kirkland JL, Miller RA, Bartke A, Kopchick JJ. 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 155: 1793‐1805, 2014.
 122.List EO, Palmer AJ, Berryman DE, Bower B, Kelder B, Kopchick JJ. Growth hormone improves body composition, fasting blood glucose, glucose tolerance and liver triacylglycerol in a mouse model of diet‐induced obesity and type 2 diabetes. Diabetologia 52: 1647‐1655, 2009.
 123.Longobardi S, Keay N, Ehrnborg C, Cittadini A, Rosen T, Dall R, Boroujerdi MA, Bassett EE, Healy ML, Pentecost C, Wallace JD, Powrie J, Jorgensen JO, Sacca L. 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 85: 1505‐1512, 2000.
 124.Lu C, Kumar PA, Sun J, Aggarwal A, Fan Y, Sperling MA, Lumeng CN, Menon RK. 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 288: 15725‐15735, 2013.
 125.Lubbers ER, List EO, Jara A, Sackmann‐Sala L, Cordoba‐Chacon J, Gahete M, Kineman RD, Boparai R, Bartke A, Kopchick J, Berryman DE. Adiponectin in mice with altered growth hormone action: Links to insulin sensitivity and longevity? J Endocrinol 216: 363‐374, 2012.
 126.Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 117: 175‐184, 2007.
 127.Lupu F, Terwilliger JD, Lee K, Segre GV, Efstratiadis A. Roles of growth hormone and insulin‐like growth factor 1 in mouse postnatal growth. Dev Biol 229: 141‐162, 2001.
 128.Makimura H, Stanley T, Mun D, You SM, Grinspoon S. The effects of central adiposity on growth hormone (GH) response to GH‐releasing hormone‐arginine stimulation testing in men. J Clin Endocrinol Metab 93: 4254‐4260, 2008.
 129.Martos‐Moreno GA, Sackmann‐Sala L, Berryman DE, Blome DW, Argente J, Kopchick JJ. Anatomical heterogeneity in the proteome of human subcutaneous adipose tissue. An Pediatr (Barc) 78: 140‐148, 2013.
 130.Masternak MM, Bartke A, Wang F, Spong A, Gesing A, Fang Y, Salmon AB, Hughes LF, Liberati T, Boparai R, Kopchick JJ, Westbrook R. Metabolic effects of intra‐abdominal fat in GHRKO mice. Aging Cell 11: 73‐81, 2012.
 131.Masuzaki H, Ogawa Y, Isse N, Satoh N, Okazaki T, Shigemoto M, Mori K, Tamura N, Hosoda K, Yoshimasa Y, et al. Human obese gene expression. Adipocyte‐specific expression and regional differences in the adipose tissue. Diabetes 44: 855‐858, 1995.
 132.Matsubara M, Maruoka S, Katayose S. Inverse relationship between plasma adiponectin and leptin concentrations in normal‐weight and obese women. Eur J Endocrinol 147: 173‐180, 2002.
 133.Mauras N, Haymond MW. Are the metabolic effects of GH and IGF‐I separable? Growth Horm IGF Res 15: 19‐27, 2005.
 134.Meazza C, Elsedfy HH, Pagani S, Bozzola E, El Kholy M, Bozzola M. Metabolic parameters and adipokine profile in growth hormone deficient (GHD) children before and after 12‐month GH treatment. Horm Metab Res 46: 219‐223, 2014.
 135.Melmed S. Acromegaly pathogenesis and treatment. J Clin Invest 119: 3189‐3202, 2009.
 136.Meyer LK, Ciaraldi TP, Henry RR, Wittgrove AC, Phillips SA. Adipose tissue depot and cell size dependency of adiponectin synthesis and secretion in human obesity. Adipocyte 2: 217‐226, 2013.
 137.Miller KK, Biller BM, Lipman JG, Bradwin G, Rifai N, Klibanski A. Truncal adiposity, relative growth hormone deficiency, and cardiovascular risk. J Clin Endocrinol Metab 90: 768‐774, 2005.
 138.Miquet JG, Freund T, Martinez CS, Gonzalez L, Diaz ME, Micucci GP, Zotta E, Boparai RK, Bartke A, Turyn D, Sotelo AI. Hepatocellular alterations and dysregulation of oncogenic pathways in the liver of transgenic mice overexpressing growth hormone. Cell Cycle 12: 1042‐1057, 2013.
 139.Moller N, Jorgensen JO. Effects of growth hormone on glucose, lipid, protein metabolism in human subjects. Endocr Rev 30: 152‐177, 2009.
 140.Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH, Earley AR, Barnett AH, Prins JB, O'Rahilly S. Congenital leptin deficiency is associated with severe early‐onset obesity in humans. Nature 387: 903‐908, 1997.
 141.Montague CT, Prins JB, Sanders L, Digby JE, O'Rahilly S. Depot‐ and sex‐specific differences in human leptin mRNA expression: implications for the control of regional fat distribution. Diabetes 46: 342‐347, 1997.
 142.Muller AF, Lamberts SW, Janssen JA, Hofland LJ, Koetsveld PV, Bidlingmaier M, Strasburger CJ, Ghigo E, Van der Lely AJ. Ghrelin drives GH secretion during fasting in man. Eur J Endocrinol 146: 203‐207, 2002.
 143.Muller EE. Clinical implications of growth hormone feedback mechanisms. Horm Res 33(Suppl 4): 90‐96, 1990.
 144.Muller EE, Locatelli V, Cocchi D. Neuroendocrine control of growth hormone secretion. Physiol Rev 79: 511‐607, 1999.
 145.Mullis PE. Genetics of growth hormone deficiency. Endocrinol Metab Clin North Am 36: 17‐36, 2007.
 146.Murase T, Yamada N, Ohsawa N, Kosaka K, Morita S, Yoshida S. Decline of postheparin plasma lipoprotein lipase in acromegalic patients. Metabolism 29: 666‐672, 1980.
 147.Nam SY, Lobie PE. The mechanism of effect of growth hormone on preadipocyte and adipocyte function. Obes Rev 1: 73‐86, 2000.
 148.Nawrocki AR, Rajala MW, Tomas E, Pajvani UB, Saha AK, Trumbauer ME, Pang Z, Chen AS, Ruderman NB, Chen H, Rossetti L, Scherer PE. Mice lacking adiponectin show decreased hepatic insulin sensitivity and reduced responsiveness to peroxisome proliferator‐activated receptor gamma agonists. J Biol Chem 281: 2654‐2660, 2006.
 149.Nedergaard J, Bengtsson T, Cannon B. Three years with adult human brown adipose tissue. Ann N Y Acad Sci 1212: E20‐36, 2010.
 150.Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in adult humans. Am J Physiol Endocrinol Metab 293: E444‐452, 2007.
 151.Nilsson A, Isgaard J, Lindahl A, Dahlstrom A, Skottner A, Isaksson OG. Regulation by growth hormone of number of chondrocytes containing IGF‐I in rat growth plate. Science 233: 571‐574, 1986.
 152.Nilsson L, Binart N, Bohlooly YM, Bramnert M, Egecioglu E, Kindblom J, Kelly PA, Kopchick JJ, Ormandy CJ, Ling C, Billig H. Prolactin and growth hormone regulate adiponectin secretion and receptor expression in adipose tissue. Biochem Biophys Res Commun 331: 1120‐1126, 2005.
 153.Okada S, Chen WY, Wiehl P, Kelder B, Goodman HM, Guller S, Sonenberg M, Kopchick JJ. A growth hormone (GH) analog can antagonize the ability of native GH to promote differentiation of 3T3‐F442A preadipocytes and stimulate insulin‐like and lipolytic activities in primary rat adipocytes. Endocrinology 130: 2284‐2290, 1992.
 154.Olarescu NC, Berryman DE, Householder LA, Lubbers ER, List EO, Benencia F, Kopchick JJ, Bollerslev J. GH action influences adipogenesis of mouse adipose tissue‐derived mesenchymal stem cells. J Endocrinol 226: 13‐23, 2015.
 155.Olarescu NC, Heck A, Godang K, Ueland T, Bollerslev J. The metabolic risk in newly diagnosed patients with acromegaly is related to fat distribution and circulating adipokines and improves after treatment. Neuroendocrinology, 2015.
 156.Olarescu NC, Ueland T, Godang K, Lindberg‐Larsen R, Jorgensen JO, Bollerslev J. Inflammatory adipokines contribute to insulin resistance in active acromegaly and respond differently to different treatment modalities. Eur J Endocrinol 170: 39‐48, 2014.
 157.Olsson B, Bohlooly YM, Fitzgerald SM, Frick F, Ljungberg A, Ahren B, Tornell J, Bergstrom G, Oscarsson J. Bovine growth hormone transgenic mice are resistant to diet‐induced obesity but develop hyperphagia, dyslipidemia, and diabetes on a high‐fat diet. Endocrinology 146: 920‐930, 2005.
 158.Ouchi N, Kihara S, Funahashi T, Matsuzawa Y, Walsh K. Obesity, adiponectin and vascular inflammatory disease. Curr Opin Lipidol 14: 561‐566, 2003.
 159.Pajvani UB, Du X, Combs TP, Berg AH, Rajala MW, Schulthess T, Engel J, Brownlee M, Scherer PE. Structure‐function studies of the adipocyte‐secreted hormone Acrp30/adiponectin. Implications fpr metabolic regulation and bioactivity. J Biol Chem 278: 9073‐9085, 2003.
 160.Palmer AJ, Chung MY, List EO, Walker J, Okada S, Kopchick JJ, Berryman DE. Age‐related changes in body composition of bovine growth hormone transgenic mice. Endocrinology 150: 1353‐1360, 2009.
 161.Palmer BF, Clegg DJ. The sexual dimorphism of obesity. Mol Cell Endocrinol 402: 113‐119, 2015.
 162.Parkinson C, Kassem M, Heickendorff L, Flyvbjerg A, Trainer PJ. Pegvisomant‐induced serum insulin‐like growth factor‐I normalization in patients with acromegaly returns elevated markers of bone turnover to normal. J Clin Endocrinol Metab 88: 5650‐5655, 2003.
 163.Pasarica M, Gowronska‐Kozak B, Burk D, Remedios I, Hymel D, Gimble J, Ravussin E, Bray GA, Smith SR. Adipose tissue collagen VI in obesity. J Clin Endocrinol Metab 94: 5155‐5162, 2009.
 164.Pelleymounter MA, Cullen MJ, Baker MB, Hecht R, Winters D, Boone T, Collins F. Effects of the obese gene product on body weight regulation in ob/ob mice. Science 269: 540‐543, 1995.
 165.Pombo M, Pombo CM, Astorga R, Cordido F, Popovic V, Garcia‐Mayor RV, Dieguez C, Casanueva FF. Regulation of growth hormone secretion by signals produced by the adipose tissue. J Endocrinol Invest 22: 22‐26, 1999.
 166.Pond CM. Physiological specialisation of adipose tissue. Prog Lipid Res 38: 225‐248, 1999.
 167.Pond CM, Mattacks CA. In vivo evidence for the involvement of the adipose tissue surrounding lymph nodes in immune responses. Immunol Lett 63: 159‐167, 1998.
 168.Pond CM, Mattacks CA. Interactions between adipose tissue around lymph nodes and lymphoid cells in vitro. J Lipid Res 36: 2219‐2231, 1995.
 169.Pope M, Budge H, Symonds ME. The developmental transition of ovine adipose tissue through early life. Acta Physiol 210: 20‐30, 2014.
 170.Psilopanagioti A, Papadaki H, Kranioti EF, Alexandrides TK, Varakis JN. Expression of adiponectin and adiponectin receptors in human pituitary gland and brain. Neuroendocrinology 89: 38‐47, 2009.
 171.Rabe K, Lehrke M, Parhofer KG, Broedl UC. Adipokines and insulin resistance. Mol Med 14: 741‐751, 2008.
 172.Raguso CA, Kyle U, Kossovsky MP, Roynette C, Paoloni‐Giacobino A, Hans D, Genton L, Pichard C. A 3‐year longitudinal study on body composition changes in the elderly: role of physical exercise. Clin Nutr 25: 573‐580, 2006.
 173.Rasmussen MH, Hvidberg A, Juul A, Main KM, Gotfredsen A, Skakkebaek NE, Hilsted J, Skakkebae NE. Massive weight loss restores 24‐hour growth hormone release profiles and serum insulin‐like growth factor‐I levels in obese subjects. J Clin Endocrinol Metab 80: 1407‐1415, 1995.
 174.Reddy NL, Tan BK, Barber TM, Randeva HS. Brown adipose tissue: Endocrine determinants of function and therapeutic manipulation as a novel treatment strategy for obesity. BMC Obes 1: 13, 2014.
 175.Reed ML, Merriam GR, Kargi AY. Adult growth hormone deficiency—benefits, side effects, and risks of growth hormone replacement. Front Endocrinol (Lausanne) 4: 64, 2013.
 176.Reyes‐Vidal C, Fernandez JC, Bruce JN, Crisman C, Conwell IM, Kostadinov J, Geer EB, Post KD, Freda PU. Prospective study of surgical treatment of acromegaly: Effects on ghrelin, weight, adiposity, and markers of CV risk. J Clin Endocrinol Metab 99: 4124‐4132, 2014.
 177.Richelsen B, Pedersen SB, Kristensen K, Borglum JD, Norrelund H, Christiansen JS, Jorgensen JO. 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 49: 906‐911, 2000.
 178.Rodriguez‐Arnao J, Jabbar A, Fulcher K, Besser GM, Ross RJ. Effects of growth hormone replacement on physical performance and body composition in GH deficient adults. Clin Endocrinol (Oxf) 51: 53‐60, 1999.
 179.Rogozinski A, Furioso A, Glikman P, Junco M, Laudi R, Reyes A, Lowenstein A. Thyroid nodules in acromegaly. Arq Bras Endocrinol Metabol 56: 300‐304, 2012.
 180.Rokkas T, Pistiolas D, Sechopoulos P, Margantinis G, Koukoulis G. Risk of colorectal neoplasm in patients with acromegaly: a meta‐analysis. World J Gastroenterol 14: 3484‐3489, 2008.
 181.Romero CJ, Ng Y, Luque RM, Kineman RD, Koch L, Bruning JC, Radovick S. Targeted deletion of somatotroph insulin‐like growth factor‐I signaling in a cell‐specific knockout mouse model. Mol Endocrinol 24: 1077‐1089, 2010.
 182.Romero CJ, Wolfe A, Law YY, Costelloe CZ, Miller R, Wondisford F, Radovick S. Altered somatotroph feedback regulation improves metabolic efficiency and limits adipose deposition in male mice. Metabolism 65: 557‐568, 2016.
 183.Rosilio M, Blum WF, Edwards DJ, Shavrikova EP, Valle D, Lamberts SW, Erfurth EM, Webb SM, Ross RJ, Chihara K, Henrich G, Herschbach P, Attanasio AF. Long‐term improvement of quality of life during growth hormone (GH) replacement therapy in adults with GH deficiency, as measured by questions on life satisfaction‐hypopituitarism (QLS‐H). J Clin Endocrinol Metab 89: 1684‐1693, 2004.
 184.Rowlinson SW, Yoshizato H, Barclay JL, Brooks AJ, Behncken SN, Kerr LM, Millard K, Palethorpe K, Nielsen K, Clyde‐Smith J, Hancock JF, Waters MJ. An agonist‐induced conformational change in the growth hormone receptor determines the choice of signalling pathway. Nat Cell Biol 10: 740‐747, 2008.
 185.Sackmann‐Sala L, Berryman DE, Munn RD, Lubbers ER, Kopchick JJ. Heterogeneity among white adipose tissue depots in male C57BL/6J mice. Obesity (Silver Spring) 20: 101‐111, 2012.
 186.Saito M, Okamatsu‐Ogura Y, Matsushita M, Watanabe K, Yoneshiro T, Nio‐Kobayashi J, Iwanaga T, Miyagawa M, Kameya T, Nakada K, Kawai Y, Tsujisaki M. High incidence of metabolically active brown adipose tissue in healthy adult humans: effects of cold exposure and adiposity. Diabetes 58: 1526‐1531, 2009.
 187.Savage DB, Sewter CP, Klenk ES, Segal DG, Vidal‐Puig A, Considine RV, O'Rahilly S. Resistin/Fizz3 expression in relation to obesity and peroxisome proliferator‐activated receptor‐gamma action in humans. Diabetes 50: 2199‐2202, 2001.
 188.Savastano S, Barbato A, Di Somma C, Guida B, Pizza G, Barrea L, Avallone S, Schiano di Cola M, Strazzullo P, Colao A. Beyond waist circumference in an adult male population of Southern Italy: Is there any role for subscapular skinfold thickness in the relationship between insulin‐like growth factor‐I system and metabolic parameters? J Endocrinol Invest 35: 925‐929, 2012.
 189.Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem 270: 26746‐26749, 1995.
 190.Sharp LZ, Shinoda K, Ohno H, Scheel DW, Tomoda E, Ruiz L, Hu H, Wang L, Pavlova Z, Gilsanz V, Kajimura S. Human BAT possesses molecular signatures that resemble beige/brite cells. PLoS One 7: e49452, 2012.
 191.Shevah O, Laron Z. Patients with congenital deficiency of IGF‐I seem protected from the development of malignancies: A preliminary report. Growth Horm IGF Res 17: 54‐57, 2007.
 192.Silha JV, Krsek M, Hana V, Marek J, Jezkova J, Weiss V, Murphy LJ. Perturbations in adiponectin, leptin and resistin levels in acromegaly: Lack of correlation with insulin resistance. Clin Endocrinol (Oxf) 58: 736‐742, 2003.
 193.Simsolo RB, Ezzat S, Ong JM, Saghizadeh M, Kern PA. Effects of acromegaly treatment and growth hormone on adipose tissue lipoprotein lipase. J Clin Endocrinol Metab 80: 3233‐3238, 1995.
 194.Smuel K, Kauli R, Lilos P, Laron Z. Growth, development, puberty and adult height before and during treatment in children with congenital isolated growth hormone deficiency. Growth Horm IGF Res 25: 182‐188, 2015.
 195.Sotiropoulos A, Ohanna M, Kedzia C, Menon RK, Kopchick JJ, Kelly PA, Pende M. Growth hormone promotes skeletal muscle cell fusion independent of insulin‐like growth factor 1 up‐regulation. Proc Natl Acad Sci U S A 103: 7315‐7320, 2006.
 196.Spadaro O, Goldberg EL, Camell CD, Youm YH, Kopchick JJ, Nguyen KY, Bartke A, Sun LY, Dixit VD. Growth hormone receptor deficiency protects against age‐related NLRP3 inflammasome activation and immune senescence. Cell Rep 14: 1571‐1580, 2016.
 197.Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA. The hormone resistin links obesity to diabetes. Nature 409: 307‐312, 2001.
 198.Steuerman R, Shevah O, Laron Z. Congenital IGF1 deficiency tends to confer protection against post‐natal development of malignancies. Eur J Endocrinol 164: 485‐489, 2011.
 199.Stout MB, Swindell WR, Zhi X, Rohde K, List EO, Berryman DE, Kopchick JJ, Gesing A, Fang Y, Masternak MM. Transcriptome profiling reveals divergent expression shifts in brown and white adipose tissue from long‐lived GHRKO mice. Oncotarget 6: 26702‐26715, 2015.
 200.Stout MB, Tchkonia T, Pirtskhalava T, Palmer AK, List EO, Berryman DE, Lubbers ER, Escande C, Spong A, Masternak MM, Oberg AL, LeBrasseur NK, Miller RA, Kopchick JJ, Bartke A, Kirkland JL. Growth hormone action predicts age‐related white adipose tissue dysfunction and senescent cell burden in mice. Aging (Albany NY) 6: 575‐586, 2014.
 201.Stubbins RE, Holcomb VB, Hong J, Nunez NP. Estrogen modulates abdominal adiposity and protects female mice from obesity and impaired glucose tolerance. Eur J Nutr 51: 861‐870, 2012.
 202.Sucunza N, Barahona MJ, Resmini E, Fernandez‐Real JM, Ricart W, Farrerons J, Rodriguez Espinosa J, Marin AM, Puig T, Webb SM. A link between bone mineral density and serum adiponectin and visfatin levels in acromegaly. J Clin Endocrinol Metab 94: 3889‐3896, 2009.
 203.Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J Clin Invest 121: 2094‐2101, 2011.
 204.Sun K, Tordjman J, Clement K, Scherer PE. Fibrosis and adipose tissue dysfunction. Cell Metab 18: 470‐477, 2013.
 205.Tchkonia T, Morbeck DE, Von Zglinicki T, Van Deursen J, Lustgarten J, Scrable H, Khosla S, Jensen MD, Kirkland JL. Fat tissue, aging, and cellular senescence. Aging Cell 9: 667‐684, 2010.
 206.Tchkonia T, Thomou T, Zhu Y, Karagiannides I, Pothoulakis C, Jensen MD, Kirkland JL. Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab 17: 644‐656, 2013.
 207.Ueland T, Fougner SL, Godang K, Lekva T, Schurgers LJ, Scholz H, Halvorsen B, Schreiner T, Aukrust P, Bollerslev J. Associations between body composition, circulating interleukin‐1 receptor antagonist, osteocalcin, and insulin metabolism in active acromegaly. J Clin Endocrinol Metab 95: 361‐368, 2010.
 208.Vahl N, Jorgensen JO, Jurik AG, Christiansen JS. Abdominal adiposity and physical fitness are major determinants of the age associated decline in stimulated GH secretion in healthy adults. J Clin Endocrinol Metab 81: 2209‐2215, 1996.
 209.Vahl N, Jorgensen JO, Skjaerbaek C, Veldhuis JD, Orskov H, Christiansen JS. Abdominal adiposity rather than age and sex predicts mass and regularity of GH secretion in healthy adults. Am J Physiol 272: E1108‐E1116, 1997.
 210.Vickers MH, Gilmour S, Gertler A, Breier BH, Tunny K, Waters MJ, Gluckman PD. 20‐kDa placental hGH‐V has diminished diabetogenic and lactogenic activities compared with 22‐kDa hGH‐N while retaining antilipogenic activity. Am J Physiol Endocrinol Metab 297: E629‐E637, 2009.
 211.Vikman K, Carlsson B, Billig H, Eden S. Expression and regulation of growth hormone (GH) receptor messenger ribonucleic acid (mRNA) in rat adipose tissue, adipocytes, and adipocyte precursor cells: GH regulation of GH receptor mRNA. Endocrinology 129: 1155‐1161, 1991.
 212.Vitali A, Murano I, Zingaretti MC, Frontini A, Ricquier D, Cinti S. The adipose organ of obesity‐prone C57BL/6J mice is composed of mixed white and brown adipocytes. J Lipid Res 53: 619‐629, 2012.
 213.Vizner B, Reiner Z, Sekso M. Effect of l‐dopa on growth hormone, glucose, insulin, and cortisol response in obese subjects. Exp Clin Endocrinol 81: 41‐48, 1983.
 214.Wang GX, Zhao XY, Meng ZX, Kern M, Dietrich A, Chen Z, Cozacov Z, Zhou D, Okunade AL, Su X, Li S, Bluher M, Lin JD. The brown fat‐enriched secreted factor Nrg4 preserves metabolic homeostasis through attenuation of hepatic lipogenesis. Nat Med 20: 1436‐1443, 2014.
 215.Wang QA, Tao C, Gupta RK, Scherer PE. Tracking adipogenesis during white adipose tissue development, expansion and regeneration. Nat Med 19: 1338‐1344, 2013.
 216.Wang Z, Al‐Regaiey KA, Masternak MM, Bartke A. Adipocytokines and lipid levels in Ames dwarf and calorie‐restricted mice. J Gerontol A Biol Sci Med Sci 61: 323‐331, 2006.
 217.Wells JA. Binding in the growth hormone receptor complex. Proc Natl Acad Sci U S A 93: 1‐6, 1996.
 218.Williams T, Berelowitz M, Joffe SN, Thorner MO, Rivier J, Vale W, Frohman LA. Impaired growth hormone responses to growth hormone‐releasing factor in obesity. A pituitary defect reversed with weight reduction. N Engl J Med 311: 1403‐1407, 1984.
 219.Xue Y, Gao Y, Wang S, Wang P. An examination of the effects of different doses of recombinant human growth hormone on children with growth hormone deficiency. Exp Ther Med 11: 1647‐1652, 2016.
 220.Yang T, Householder LA, Lubbers ER, List EO, Troike K, Vesel C, Duran‐Ortiz S, Kopchick JJ, Berryman DE. Growth hormone receptor antagonist transgenic mice are protected from hyperinsulinemia and glucose intolerance despite obesity when placed on a HF diet. Endocrinology 156: 555‐564, 2015.
 221.Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature 372: 425‐432, 1994.
 222.Zhao JT, Cowley MJ, Lee P, Birzniece V, Kaplan W, Ho KK. Identification of novel GH‐regulated pathway of lipid metabolism in adipose tissue: A gene expression study in hypopituitary men. J Clin Endocrinol Metab 96: E1188‐E1196, 2011.
 223.Zhou Y, Xu BC, Maheshwari HG, He L, Reed M, Lozykowski M, Okada S, Cataldo L, Coschigamo K, Wagner TE, Baumann G, Kopchick JJ. A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone receptor/binding protein gene (the Laron mouse). Proc Natl Acad Sci U S A 94: 13215‐13220, 1997.
 224.Zou L, Menon RK, Sperling MA. Induction of mRNAs for the growth hormone receptor gene during mouse 3T3‐L1 preadipocyte differentiation. Metabolism 46: 114‐118, 1997.

Further Reading

Berryman DE, Henry B, Hjortebjerg R, List EO. Developments in our understanding of the effects of growth hormone on white adipose tissue: implications on the clinic. Expert Reviews in Endocrinology and Metabolism. 2016; 11(2): 197-207.

Ho KKY and Works GDC. Consensus guidelines for the diagnosis and treatment of adults with GH deficiency II: a statement of the GH research society in association with the European Society for Pediatric Endocrinology, Lawson Wilkins Society, European Society of Endocrinology, Japan Endocrine Society, and Endocrine Society of Australia. European Journal of Endocrinology 157: 695-700, 2007.

Israel E, Attie KM, Bengtsson BA, Blethen SL, Blum W, Cameron F, Carel JC, Carlsson L, Chipman JJ, Christiansen JS, Clayton P, Clemmons DR, Cohen P, Drop S, Fujieda K, Ghigo E, Hintz RL, Ho K, Ilondo MM, Jasper H, Jesussek B, Kappelgaard AM, Laron Z, Lippe BM, Malozowski S, Mullis PE, de Munick-Keizer-Schrama S, Nishi Y, Parks JS, Phelps C, Ranke M, Robinson I, Rosenfeld RG, Rose S, Saenger P, Saggese G, Savage M, Shalet S, Sizonenko PC, Strasburger C, Tachibana K, Tanaka T, Thorner MO, Wikland KA, Zadik Z, and Soc GR. Consensus guidelines for the diagnosis and treatment of growth hormone (GH) deficiency in childhood and adolescence: Summary statement of the GH Research Society. J Clin Endocrinol Metab 85: 3990-3993, 2000.

 

 

Teaching Material

 

 

K. M. Troike, B. E. Henry, E. A. Jensen, J. A. Young, E. O. List, J. J. Kopchick, D. E. Berryman. Impact of Growth Hormone on Regulation of Adipose Tissue. Compr Physiol 7 2017, 819-840.

 

Didactic Synopsis




 

 

 

 

 

Major Teaching Points:

     

  1. Adipose tissue (AT) is a complex, dynamic endocrine organ that secretes and responds to various hormones and cytokines.
  2.  

     

  3. Growth hormone (GH) has a profound impact on AT.
  4.  

       

    1. In AT, GH acts to inhibit insulin action, decrease glucose uptake, stimulate lipolysis, and inhibit lipogenesis.
    2.  

       

    3. GH regulates other AT properties, such as endocrine output, immune cell function, senescence, differentiation, beiging, and extracellular matrix deposition.
    4.  

     

  5. Perturbations in GH-induced signaling are responsible for a number of clinical conditions (e.g., acromegaly, Laron Syndrome, and GH deficiency). Many genetically altered mouse lines have been developed to study these diseases.
  6.  

     

  7. The GH-IGF-1 axis plays a role in obesity.
  8.  

 

  1. GH secretory response is blunted in obesity.
  2.  

     

  3. Visceral WAT accumulation is negatively correlated with GH secretion.
  4.  

     

  5. Treatment of obesity using rhGH is not currently advised with additional studies needed to demonstrate its safety and efficacy.
  6.  

 

 

 

Didactic Legends

 




 

 

 

 

 

The figures—in a freely downloadable PowerPoint format—can be found on the Images tab along with the formal legends published in the article. The following legends to the same figures are written to be useful for teaching.


 

 

 

Figure 1. Teaching points: AT is distributed throughout the body in various discrete locations, known as depots. These areas of fat deposition are important to consider when studying AT because the composition and secretory profile can vary drastically depending on AT location. For example, while visceral fat pads are often considered "unhealthy" because of their positive correlation with metabolic disease and inflammation, subQ fat does not appear to impart disease risk and is generally regarded as a "healthy" fat pad.

Figure 2. Teaching points: Understanding the differences in AT distribution between humans and mice is important when attempting to translate findings from mouse studies to those performed using human subjects. Human AT distribution shares some common features with that of mice, although there are notable differences. For example, the omental depot in human WAT is much less prominent in mice. Additionally, humans lack the perigonadal fat pad that is commonly used in studies of mouse WAT.

Figure 3. Teaching points: WAT can undergo extensive remodeling in states of obesity. Expansion of WAT occurs in two ways: namely, an increase in cell number (hyperplasia) or cell size (hypertrophy). "Healthy", or normal, fat expansion usually involves an increase in cell number, while "unhealthy", or pathological, expansion often occurs during obesity when there is a shift to increasing cell size. Pathological expansion induces a number of changes within WAT that lead to increased pro-inflammatory immune cell and adipokine response, tissue fibrosis, and hypoxia.

Figure 4. Teaching points: Understanding the regulation of GH secretion is essential for understanding the clinical conditions associated with excess or reduced GH action. Hormones released by the hypothalamus control the secretion of GH, which binds receptors on the liver and stimulates the production of IGF-1. GH exerts its effects both directly and indirectly, via IGF-1, on a number of tissues including fat, muscle, and bone.

Figure 5. Teaching points: The action of GH is mediated through its binding to the GHR. GH-induced intracellular signaling can occur through several pathways.

Figure 6. Teaching points: Genetic alteration of GH action in mice results in many phenotypic changes. Specifically, the effect of GH on body composition is readily observable when comparing these different mouse lines. bGH mice, which have excess GH action, are giant and lean with reduced fat mass. Conversely, GHA and GHR–/– mice have reduced and absent GH action, respectively, resulting in a dwarf phenotype and increased fat mass.

Figure 7. Teaching points: Comparisons of body fat percentage in mice with altered GH action further showcase the negative regulation of fat mass by GH. Initially, bGH mice have greater body fat than WT controls, but have significantly reduced fat mass later in life. In contrast, GHA and GHR-/- mice have greater percentage body fat than WT controls at all-time points.

Figure 8. Teaching points: Picrosirius red staining reveals the effect of GH on collagen deposition in WAT. bGH have increased collagen deposition compared to those with normal or absent GH action.

 

 

 

 

 

 

 

 

 

 

 

 


Related Articles:

Effect of dietary and endocrine factors on adipose tissue growth1
The Metabolic Actions of Growth Hormone
Feedback Regulation of Growth Hormone Secretion
Hormone‐sensitive lipolytic activity of adipose tissue1
Adipose Organ Development and Remodeling

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Katie M. Troike, Brooke E. Henry, Elizabeth A. Jensen, Jonathan A. Young, Edward O. List, John J. Kopchick, Darlene E. Berryman. Impact of Growth Hormone on Regulation of Adipose Tissue. Compr Physiol 2017, 7: 819-840. doi: 10.1002/cphy.c160027