Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Oct 23.
Published in final edited form as: Exp Gerontol. 2009 Apr 14;44(0):372–374. doi: 10.1016/j.exger.2009.04.001

The somatotropic axis and aging: Mechanisms and persistent questions about practical implications

Andrzej Bartke 1
PMCID: PMC4207209  NIHMSID: NIHMS634644  PMID: 19371777

Abstract

Reduced somatotropic (GH/IGF-1) signaling delays aging and extends longevity in laboratory mice. However, it is unclear whether the physiological decrease of GH and IGF-1 levels with age represents a symptom of declining neuroendocrine function, a cause of age-related alterations in body composition and functionality or a protective mechanism against age-associated disease. Although available clinical evidence does not support the use of recombinant GH as an anti-aging therapy, many studies suggest the potential utility of GH and GH secretagogues in the treatment of sarcopenia and frailty.

Introduction; long-lived mutant mice

In laboratory populations of house mice (Mus musculus), mutations that reduce growth hormone (GH) and/or insulin-like growth factor 1 (IGF-1) levels or actions produce significant, often major increases in longevity (Brown-Borg et al., 1996; Flurkey et al., 2001; Coschigano et al., 2003; Holzenberger et al., 2003; Conover and Bale, 2007). In some of these long-lived mutants, there is evidence for extended maintenance of “youthful” characteristics, delayed onset of age-related disease and other indications that the biological process of aging has been postponed and/or slowed down (Flurkey et al., 2001; Silberberg, 1972; Ikeno et al., 2003; Kinney et al., 2001a, b; Vergara et al., 2004). These findings inescapably lead to a somewhat counterintuitive conclusion that the amounts and physiological actions of GH and IGF-1 normally present in these animals are not optimal for long-term survival. It could be argued that these negative effects of GH and/or IGF-1 on longevity may be unique to laboratory stocks of mice in which hundreds of generations of domestication and purposeful or inadvertant selection for growth and fecundity produced high levels of somatotropic signaling that lead to rapid growth, early maturation and high fertility at the expense of life expectancy. Endocrine and reproductive characteristics of mice derived from animals recently caught in the wild support this possibility (Miller et al., 2002). However, the impact of GH and IGF-1 on lifespan is not limited to laboratory populations of house mice, and there is strong evidence that IGF-1 and homologous signaling is involved in the control of aging in organisms ranging from worms to mammals (details below).

GH vs. IGF-1 effects

In mice, the effects of mutations influencing GH secretion or actions on longevity are greater and more consistent than the effects of mutations that directly impact IGF-1 signaling. For example, longevity is significantly increased in both sexes of GH resistant Ghr -/- mice maintained in different laboratories on different genetic backgrounds and different diets (Coschigano et al., 2003; Bartke et al., 2004; Bonkowski et al., 2006) while extension of longevity in partially IGF-1-resistant Igf1r+/- and IGF-1 hypomorphic Midi-mice is limited to females (Holzenberger et al., 2003; Sell and Lorenzini, 2007) and, in Igf1r+/- mice on a long-lived genetic background, male longevity is reduced rather than increased (Richardson, personal communication). Moreover, while complete disruption of GH signaling produces a long-lived mouse (Brown-Borg et al., 1996; Flurkey et al., 2001; Coschigano et al., 2003), complete disruption of IGF-1 signaling is lethal or detrimental (Liu et al., 1993). It must be concluded that at least some of the mechanisms by which GH influences longevity are unrelated to reduction of peripheral IGF-1 levels. Effects of GH on insulin signaling and on production of adiponectin and other secretory products by fat cells are among the most likely candidate mechanisms linking GH and aging independently of IGF-1. Further studies in animals with altered local availability of IGF-1 (Conover and Bale, 2007) or disruption of somatotropic signaling in specific organs or cell types (as in LeRoith et al., 2008; Taguchi et al., 2007; Kappeler et al., 2008) should clarify the mechanisms by which GH and IGF-1 influence aging.

How mutant mouse data fit into the “big picture”

Extended longevity of mice with reduced activity of the somatotropic axis complements the results of elegant studies in invertebrates showing that reduced IGF-1 /insulin-like signaling in these animals increases lifespan, often very dramatically (Tatar et al., 2003; Kenyon, 2005; Piper et al., 2008). In fact demonstration of homology of the key “longevity gene” in Caenorhabditis elegans, daf-2 with mammalian IGF-1 and insulin receptor genes (Kimura et al., 1997) was followed by elucidation of conserved genetic mechanisms of aging that extend from yeast to mammals. Diminutive phenotype of most of the long-lived mouse GH/IGF-1 related mutants fits very well with the negative correlation of longevity with adult body size in genetically normal mice (Rollo, 2002; Miller et al., 2002) as well as in rats, dogs, horses and, in many comparisons, also in humans (Rollo, 2002; Patronek et al., 1997; Greer et al., 2007; Brosnahan and Paradis, 2003; Samaras, 2007).

However, these findings are difficult to reconcile with the thoroughly documented age-related decline in GH and IGF-1 levels in all mammals studied to date (Sonntag et al., 1980; Crew et al., 1987; Müller et al., 1993; Veldhuis et al., 1997) and with numerous reports of reversal of age related changes in body composition and various functional parameters by therapy with GH, GH secretagogues or IGF-1 (Rudman et al., 1990; Welle et al., 1996; Khan et al., 2001; Sonntag et al., 2005; Roy et al., 2007). Although species differences may contribute to or perhaps even account for some of these discrepancies, more likely explanations are related to the divergent physiological roles of GH during different stages of life history and its relationship to longevity within the framework of the concept of antagonistic pleiotropy. Long-term survival presumably confers little or no reproductive and thus evolutionary advantage. Therefore, robust somatotropic signaling early in life may have been selected for as a promoter of sexual maturation and reproductive fitness, regardless of the “costs” in terms of longevity and age-related disease.

Will growth hormone find legitimate use in anti-aging medicine?

Similarly to other alterations of endocrine function during aging, it is difficult to decide whether reduced activity of the somatotropic axis should be viewed primarily as a symptom or a mechanism of aging. It could also be argued that the age-related decline in GH secretion constitutes a natural protection from insulin resistance and cancer or perhaps activation of survival mechanisms (Bartke, 2003; Shechter et al., 2008) rather than simply reflecting a progressive failure of the hypothalamus-somatotrope axis.

Congenital GH deficiency and resistance lead to obesity and have been traditionally considered as serious risk factors for cardiovascular disease, but recent studies uncovered unexpected protection from atherosclerosis (Schumacher et al., 2008; Oliveira et al., 2007) and the impact of these endocrine syndromes on longevity remains controversial (Krzisnik et al., 1999; Besson et al., 2003; Laron, 2005). Studies of anti-aging interventions further fuel these controversies. While suppression of somatotropic signaling and circulating IGF-1 levels have long been considered among likely mechanisms by which calorie restriction (CR) delays aging and extends life, recent studies indicate that both short- and long-term CR in humans improves numerous aging-related risk factors without reducing either GH or IGF-1 levels (Redman et al., 2008; Fontana et al., 2008).

When viewed against this background, it is not surprising that opinions about the potential use of GH or GH secretagogues as anti-aging agents differ widely and are highly controversial. Data from well designed clinical studies reported to date indicate unfavorable risk:benefit ratio for treatment of endocrinologically normal elderly subjects with GH (Blackman et al., 2002; Liu et al., 2007). Prescribing GH to individuals who are not GH deficient in an attempt to slow aging is not supported by the available evidence, is not included among its approved uses and, in the US, is specifically disallowed. Increasing IGF-1 levels is believed to be risky, and inhibitors of IGF-1 signaling are presently undergoing clinical trials as therapy for different types of cancer.

Nevertheless, the use of GH as an anti-aging medication continues to be commercially promoted, and it is believed to be widespread in many countries. Numerous clinical and animal studies support the ability of GH to increase muscle mass (although strength is not affected), reduce adiposity and improve various aspects of cardiac function and brain blood supply (Rudman et al., 1990; Welle et al., 1996; Khan et al., 2001; Sonntag et al., 2005; Papadakis et al., 1996; Le Corvoisier et al., 2007; Giovannini et al., 2008), but more data are needed to develop a well-informed consensus on the potential utility of GH in geriatrics. Some of the available information supports a very intriguing possibility that judicious use of low doses of GH, ghrelin analogs or other GH-releasing agents may find its place in legitimate medical practice as a treatment of sarcopenia or for other age-associated indications (Giovannini et al., 2008; Mekala and Tritos, 2009; White et al., in press; Nass et al., in press OR Thorner et al., in press).

Acknowledgments

Research in our laboratory and preparation of this article were supported by NIA (NIH) via grants AG19899 and U19 AG023122, by the Ellison Medical Foundation, the Glenn Foundation for Medical Research and by the SIU Geriatrics Research Initiative. The author apologizes to those whose work pertinent to this topic was not mentioned due to format limitations of this article.

References

  1. Bartke A. Is growth hormone deficiency a beneficial adaptation to aging? Evidence from experimental animals. Trends Endocrinol Metabol. 2003;14:340–344. doi: 10.1016/s1043-2760(03)00115-2. [DOI] [PubMed] [Google Scholar]
  2. Bartke A, Peluso MR, Moretz N, Wright C, Bonkowski M, Winters TA, Shanahan MF, Kopchick JJ, Banz WJ. Effects of soy-derived diets on plasma and liver lipids, glucose tolerance, and longevity in normal, long-lived and short-lived mice. Horm Metab Res. 2004;36:550–558. doi: 10.1055/s-2004-825796. [DOI] [PubMed] [Google Scholar]
  3. Besson A, Salemi S, Gallati S, Jenal A, Horn R, Mullis PS, Mullis PE. Reduced longevity in untreated patients with isolated growth hormone deficiency. J Clin Endocrinol Metab. 2003;88:3664–3667. doi: 10.1210/jc.2002-021938. [DOI] [PubMed] [Google Scholar]
  4. Blackman MR, Sorkin JD, Munzer T, Bellantoni MF, Busby-Whitehead J, Stevens TE, Jayme J, O’Connor KG, O’Connor KG, Christmas C, Tobin JD, Stewart KJ, Cottrell E, St Clair C, Pabst KM, Harman SM. Growth hormone and sex steroid administration in healthy aged women and men: A randomized controlled trial. JAMA. 2002;288:2282–2292. doi: 10.1001/jama.288.18.2282. [DOI] [PubMed] [Google Scholar]
  5. Bonkowski MS, Rocha JS, Masternak MM, Al-Regaiey KA, Bartke A. Targeted disruption of growth hormone receptor interferes with the beneficial actions of calorie restriction. Proc Natl Acad Sci U S A. 2006;103:7901–7905. doi: 10.1073/pnas.0600161103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Brosnahan MM, Paradis MR. Demographic and clinical characteristics of geriatric horses: 467 cases (1989-1999) J Am Vet Med Assoc. 2003;223:93–98. doi: 10.2460/javma.2003.223.93. [DOI] [PubMed] [Google Scholar]
  7. Brown-Borg HM, Borg KE, Meliska CJ, Bartke A. Dwarf mice and the ageing process. Nature. 1996;384:33. doi: 10.1038/384033a0. [DOI] [PubMed] [Google Scholar]
  8. Conover CA, Bale LK. Loss of pregnancy-associated plasma protein A extends lifespan in mice. Aging Cell. 2007;6:727–729. doi: 10.1111/j.1474-9726.2007.00328.x. [DOI] [PubMed] [Google Scholar]
  9. 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 insulin-like growth factor I levels and increased life span. Endocrinology. 2003;144:3799–3810. doi: 10.1210/en.2003-0374. [DOI] [PubMed] [Google Scholar]
  10. Crew MD, Spindler SR, Walford RL, Koizumi A. Age-related decrease of growth hormone and prolactin gene expression in the mouse pituitary. Endocrinology. 1987;121:1251–1255. doi: 10.1210/endo-121-4-1251. [DOI] [PubMed] [Google Scholar]
  11. Flurkey K, Papaconstantinou J, Miller RA, Harrison DE. 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;98:6736–6741. doi: 10.1073/pnas.111158898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Fontana L, Weiss EP, Villareal DT, Klein S, Holloszy JO. Long-term effects of calorie or protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell. 2008;7:681–687. doi: 10.1111/j.1474-9726.2008.00417.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Giovannini S, Marzetti E, Borst SE, Leuwenburgh C. Modulation of GH/IGF-1 axis: Potential strategies to counteract sarcopenia in older adults. Mech Ageing Dev. 2008;129:593–601. doi: 10.1016/j.mad.2008.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Greer KA, Canterberry SC, Murphy KE. Statistical analysis regarding the effects of height and weight on life span of the domestic dog. Res Vet Sci. 2007;82:208–214. doi: 10.1016/j.rvsc.2006.06.005. [DOI] [PubMed] [Google Scholar]
  15. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Evens P, Cervera P, LeBouc Y. IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature. 2003;421:182–187. doi: 10.1038/nature01298. [DOI] [PubMed] [Google Scholar]
  16. Ikeno Y, Bronson RT, Hubbard GB, Lee S, Bartke A. Delayed occurrence of fatal neoplastic diseases in Ames dwarf mice: correlation to extended longevity. J Gerontol A Biol Sci Med Sci. 2003;58A:291–296. doi: 10.1093/gerona/58.4.b291. [DOI] [PubMed] [Google Scholar]
  17. Kappeler L, De Magalhaes Filho CM, Dupont J, Leneuve P, Cervera P, Perin L, Loudes C, Blaise A, Klein R, Epelbaum J, Le Bouc Y, Holzenberger M. Brain IGF-1 receptors control mammalian growth and lifespan through a neuroendocrine mechanism. PLoS Biol. 2008;6:e254. doi: 10.1371/journal.pbio.0060254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Kenyon C. The plasticity of aging: insights from long-lived mutants. Cell. 2005;120:449–460. doi: 10.1016/j.cell.2005.02.002. [DOI] [PubMed] [Google Scholar]
  19. Khan AS, Lynch CD, Sane DC, Willingham MC, Sonntag WE. Growth hormone increases regional coronary blood flow and capillary density in aged rats. J Gerontol A Biol Sci Med Sci. 2001;56:B364–371. doi: 10.1093/gerona/56.8.b364. [DOI] [PubMed] [Google Scholar]
  20. Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G. daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans. Science. 1997;277:942–946. doi: 10.1126/science.277.5328.942. [DOI] [PubMed] [Google Scholar]
  21. Kinney BA, Coschigano KT, Kopchick JJ, Bartke A. Evidence that age-induced decline in memory retention is delayed in growth hormone resistant GH-R-KO (Laron) mice. Physiol Behav. 2001;72:653–660. doi: 10.1016/s0031-9384(01)00423-1. [DOI] [PubMed] [Google Scholar]
  22. Kinney BA, Meliska CJ, Steger RW, Bartke A. Evidence that Ames dwarf mice age differently from their normal siblings in behavioral and learning and memory parameters. Horm Behav. 2001;39:277–284. doi: 10.1006/hbeh.2001.1654. [DOI] [PubMed] [Google Scholar]
  23. Krzisnik C, Kolacio Z, Battelino T, Brown M, Parks JS, Laron Z. The “Little People” of the island of Krk - revisited. Etiology of hypopituitarism revealed. J Endocr Genet. 1999;1:9–19. [Google Scholar]
  24. Laron Z. Do deficiencies in growth hormone and insulin-like growth factor-1 (IGF-1) shorten or prolong longevity? Mech Ageing Dev. 2005;126:305–307. doi: 10.1016/j.mad.2004.08.022. [DOI] [PubMed] [Google Scholar]
  25. Le Corvoisier P, Hittinger L, Chanson P, Montagne O, Macquin-Mavier I, Maison P. Cardiac effects of growth hormone treatment in chronic heart failure: A meta-analysis. J Clin Endocrinol Metab. 2007;92:180–185. doi: 10.1210/jc.2006-1313. [DOI] [PubMed] [Google Scholar]
  26. LeRoith D. Clinical relevance of systemic and local IGF-I: lessons from animal models. Pediatr Endocrinol Rev. 2008;5(Suppl 2):739–743. [PubMed] [Google Scholar]
  27. Liu H, Bravata DM, Olkin I, Nayak S, Roberts B, Garber AM, Hoffman AR. Systematic review: the safety and efficacy of growth hormone in the healthy elderly. Ann Intern Med. 2007;146:104–115. doi: 10.7326/0003-4819-146-2-200701160-00005. [DOI] [PubMed] [Google Scholar]
  28. Liu J-P, Baker J, Perkins AS, Robertson EJ, Efstratiadis A. Mice carrying null mutations of the genes encoding insulin-like growth factor 1 (Igf1) and type 1 IGF receptor (Igf2r) Cell. 1993;75:59–72. [PubMed] [Google Scholar]
  29. Mekala KC, Tritos NA. Effects of recombinant human growth hormone therapy in obesity in adults: a metaanalysis. J Clin Endocrinol Metab. 2009;94:130–7. doi: 10.1210/jc.2008-1357. [DOI] [PubMed] [Google Scholar]
  30. Miller RA, Harper JM, Dysko RC, Durkee SJ, Austad SN. Longer life spans and delayed maturation in wild-derived mice. Exp Biol Med (Maywood) 2002;227:500–508. doi: 10.1177/153537020222700715. [DOI] [PubMed] [Google Scholar]
  31. Miller RA, Harper JM, Galecki A, Burke DT. Big mice die young: early life body weight predicts longevity in genetically heterogeneous mice. Aging Cell. 2002;1:22–29. doi: 10.1046/j.1474-9728.2002.00006.x. [DOI] [PubMed] [Google Scholar]
  32. Müller EE, Cella SG, De Gennaro Colonna V, Parenti M, Cocchi D, Locatelli V. Aspects of the neuroendocrine control of growth hormone secretion in ageing mammals. In: Falvo R, Bartke A, Giacobini E, Thorne A, editors. Neurobiology and Neuroendocrinology of Ageing. J Reprod Fert Suppl. Vol. 46. 1993. pp. 99–114. [PubMed] [Google Scholar]
  33. Nass R, Johannsson G, Christiansen JS, Kopchick JJ, Thorner MO. The aging population - Is there a role for endocrine interventions? Growth Horm IGF Res. 2008 doi: 10.1016/j.ghir.2008.09.002. [Epub ahead of print] [Or Thorner M.O.et al Statement by the Growth Hormone Research Society on GH/IGF-1 axis in extending healthspan [epub ahead of print…] [DOI] [PubMed] [Google Scholar]
  34. Oliveira JL, Aguiar-Oliveira MH, D’Oliveira A, Jr, Pereira RM, Oliveira CR, Farias CT, Barreto-Filho JA, Anjos-Andrade FD, Marques-Santos C, Nascimento-Junior AC, Alves EO, Oliveira FT, Campos VC, Ximenes R, Blackford A, Parmigiani G, Salvatori R. Congenital growth hormone (GH) deficiency and atherosclerosis: effects of GH replacement in GH-naive adults. J Clin Endocrinol Metab. 2007;92:4664–4670. doi: 10.1210/jc.2007-1636. [DOI] [PubMed] [Google Scholar]
  35. Papadakis MA, Grady D, Black D, Tierney MJ, Gooding GA, Schambelan M, Grunfeld C. Growth hormone replacement in healthy older men improves body composition but not functional ability. Ann Intern Med. 1996;124:708–716. doi: 10.7326/0003-4819-124-8-199604150-00002. [DOI] [PubMed] [Google Scholar]
  36. Patronek GJ, Waters DJ, Glickman LT. Comparative longevity of pet dogs and humans: implications for gerontology research. J Gerontol. A Biol Sci Med Sci. 1997;52A:B171–B178. doi: 10.1093/gerona/52a.3.b171. [DOI] [PubMed] [Google Scholar]
  37. Piper MDW, Selman C, McElwee JJ, Partridge L. Separating cause from effect: how does insulin/IGF signalling control lifespan in worms, flies and mice? J Intern Med. 2008;263:179–191. doi: 10.1111/j.1365-2796.2007.01906.x. [DOI] [PubMed] [Google Scholar]
  38. Redman LM, Martin CK, Williamson DA, Ravussin E. Effect of caloric restriction in non-obese humans on physiological, psychological and behavioral outcomes. Physiol Behav. 2008;94:643–648. doi: 10.1016/j.physbeh.2008.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Richardson A. Personal comment [Google Scholar]
  40. Rollo CD. Growth negatively impacts the life span of mammals. Evol Dev. 2002;4:55–61. doi: 10.1046/j.1525-142x.2002.01053.x. [DOI] [PubMed] [Google Scholar]
  41. Rudman D, Feller AG, Nagraj HS, Gergans GA, Lalitha PY, Goldberg AF, Schlenker RA, Cohn L, Rudman IW, Mattson DE. Effects of human growth hormone in men over 60 years old. N Engl J Med. 1990;323:1–6. doi: 10.1056/NEJM199007053230101. [DOI] [PubMed] [Google Scholar]
  42. Samaras TT. Human body size and the laws of scaling: physiological, performance, growth, longevity and ecological ramifications. Nova Science Publishers, Inc; New York: 2007. [Google Scholar]
  43. Shechter M, Ginsberg S, Scheinowitz M, Feinberg MS, Laron Z. Obese adults with primary growth hormone resistance (Laron Syndrome) have normal endothelial function. Growth Horm IGF Res. 2007;17(2):165–170. doi: 10.1016/j.ghir.2007.01.009. [DOI] [PubMed] [Google Scholar]
  44. Sell C, Lorenzini S. Aging in IGF-1 hypomorphic mice. The American Aging Association 36th Annual Meeting; San Antonio, TX. American Aging Association; 2007. [Google Scholar]
  45. Silberberg R. Articular aging and osteoarthrosis in dwarf mice. Pathol Microbiol (Basel) 1972;38:417–430. doi: 10.1159/000162458. [DOI] [PubMed] [Google Scholar]
  46. Smith RG, Sun Y, Jiang H, Albarran-Zeckler R, Timchenko N. Ghrelin receptor (GHS-R1A) agonists show potential as interventive agents during aging. Ann N Y Acad Sci. 2007;1119:147–164. doi: 10.1196/annals.1404.023. [DOI] [PubMed] [Google Scholar]
  47. Sonntag EW, Steger RW, Forman JL, Meites J. Decreased pulsatile release of growth hormone in old male rats. Endocrinology. 1980;107:1875–1879. doi: 10.1210/endo-107-6-1875. [DOI] [PubMed] [Google Scholar]
  48. Sonntag WE, Ramsey M, Carter CS. Growth hormone and insulin-like growth factor-1 (IGF-1) and their influence on cognitive aging. Ageing Res Rev. 2005;4:195–212. doi: 10.1016/j.arr.2005.02.001. [DOI] [PubMed] [Google Scholar]
  49. Taguchi A, Wartschow LM, White MF. Brain IRS2 signaling coordinates life span and nutrient homeostasis. Science. 2007;317:369–372. doi: 10.1126/science.1142179. [DOI] [PubMed] [Google Scholar]
  50. Tatar M, Bartke A, Antebi A. The endocrine regulation of aging by insulin-like signals. Science. 2003;299:1346–1351. doi: 10.1126/science.1081447. [DOI] [PubMed] [Google Scholar]
  51. Veldhuis J, Iranmanesh A, Weltman A. Elements in the pathophysiology of diminished growth hormone (GH) secretion in aging humans. Endocrine. 1997;7:41–48. doi: 10.1007/BF02778061. [DOI] [PubMed] [Google Scholar]
  52. Vergara M, Smith-Wheelock M, Harper JM, Sigler R, Miller RA. Hormone-treated Snell dwarf mice regain fertility but remain long lived and disease resistant. J Gerontol A Biol Sci Med Sci. 2004;59:1244–1250. doi: 10.1093/gerona/59.12.1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Welle S, Thornton C, Statt M, McHenry B. Growth hormone increases muscle mass and strength but does not rejuvenate myofibrillar protein synthesis in healthy subjects over 60 years old. J Clin Endocrinol Metab. 1996;81:3239–3243. doi: 10.1210/jcem.81.9.8784075. [DOI] [PubMed] [Google Scholar]
  54. White HK, Petrie CD, Landschulz W, Maclean D, Taylor A, Lyles K, Wei JY, Hoffman AR, Salvatori R, Ettinger MP, Morey MC, Blackman MR, Merriam GR. Effects of an oral growth hormone secretagogue in older adults. J Clin Endocrinol Metab. 2009 doi: 10.1210/jc.2008-0632. in press. [DOI] [PubMed] [Google Scholar]

RESOURCES