当前位置: 首页 > 期刊 > 《内分泌学杂志》 > 2005年第9期 > 正文
编号:11295337
Minireview: Role of the Growth Hormone/Insulin-Like Growth Factor System in Mammalian Aging
http://www.100md.com 《内分泌学杂志》
     Departments of Physiology and Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois 62794

    Abstract

    The important role of IGF and insulin-related signaling pathways in the control of longevity of worms and insects is very well documented. In the mouse, several spontaneous or experimentally induced mutations that interfere with GH biosynthesis, GH actions, or sensitivity to IGF-I lead to extended longevity. Increases in the average life span in these mutants range from approximately 20–70% depending on the nature of the endocrine defect, gender, diet, and/or genetic background. Extended longevity of hypopituitary and GH-resistant mice appears to be due to multiple mechanisms including reduced insulin levels, enhanced insulin sensitivity, alterations in carbohydrate and lipid metabolism, reduced generation of reactive oxygen species, enhanced resistance to stress, reduced oxidative damage, and delayed onset of age-related disease. There is considerable evidence to suggest that the genetic and endocrine mechanisms that influence aging and longevity in mice may play a similar role in other mammalian species, including the human.

    Introduction

    THE SOMATOTROPIC AXIS, consisting of pituitary-derived GH and IGF-I, the main mediator of GH actions, is the key determinant of somatic growth and adult body size. Moreover, the GH/IGF-I system is involved in the regulation of puberty and gonadal function and influences body composition as well as structural and functional maintenance of adult tissues. Loss of skeletal muscle mass, increased adiposity, and other unwelcome accompaniments of aging have been linked to age-related decline in pituitary GH secretion (1) and some of these changes can be ameliorated by GH treatment of elderly individuals (1, 2). On this basis, administration of GH is often advocated as an "anti-aging" therapy (3). In sharp contrast to these findings, GH deficiency, GH resistance, and reduced IGF-I signaling in mice are associated with symptoms of delayed aging and markedly extended longevity (4, 5, 6, 7). Although most normal mice die at approximately 2 yr of age, hypopituitary and GH-resistant mutants often survive beyond the age of 3 yr and occasionally past the age of 4 yr, i.e. outside the range encountered in various laboratory strains of this species. In this brief review, we will list the key characteristics of long-lived GH-deficient, GH-resistant, and IGF-I-resistant mice, discuss mechanisms that are believed to link reduced somatotropic signaling with prolonged longevity, and speculate on the possible relevance of findings obtained in genetically altered mice to the physiological control of aging in normal mice and in other mammalian species including the human.

    Long-Lived Mutant Mice

    Primary defects in somatotropic signaling and some of the secondary endocrine changes in different types of long-lived mutant mice are listed in Table 1. In three of these mutants [Ames dwarf, Snell dwarf, and GH receptor knockout (GHRKO)], robust increases in longevity were reproducibly observed in both genders (4, 5, 6, 9, 13, 14, 15). In addition, increased longevity of GHRKO mice was observed in different laboratories (5, 13, 14) and in stocks with different genetic backgrounds (13). In "little" (GHRHRlit) mice, significant extension of longevity was detected only when the animals were fed a low-fat diet to prevent obesity (6). In IGFIR+/– mice, extension of average life span was significant only in females (7). However, it should also be mentioned that IGFIR+/– mice are unique among these long-lived mutants by having near-normal growth and adult body size and no detectable alterations in reproductive development and function (7). Preliminary findings from ongoing studies in mice expressing a hypomorphic IGF-I mutation support the conclusion that reduction of IGF-I signaling in mice leads to increased longevity (C. Sell, personal communication). Surprisingly, life span is not altered in transgenic mice expressing an antagonistic analog of GH (13). We suspect that this may be due to a very modest reduction in circulating IGF-I levels in these animals (13). Moreover, in contrast to GH-deficient and GH-resistant mutants, GH antagonist transgenic mice exhibit no reduction in insulin and only very minor, age-dependent suppression of glucose levels (13), with some of these characteristics likely being related to conspicuous obesity of these animals.

    With the exception of IGFIR+/– animals, mice with mutations affecting somatotropic signaling exhibit a series of phenotypic characteristics consistent with GH deficiency or resistance including reduced postnatal (and particularly, postweaning) growth, diminutive adult body size, delayed puberty, and reduced fertility. In addition, Ames and Snell dwarf as well as GHRKO mice have reduced plasma insulin and glucose levels (16, 17, 18, 19). Snell and Ames dwarf mice have primary endocrine defects unrelated to the somatotropic axis. Both of these mutants lack thyroid stimulating hormone and, therefore, are hypothyroid and are also prolactin (PRL)-deficient. Reduction of body temperature in dwarf mice (20) is presumably related to hypothyroidism, although lack of GH and reduced levels of insulin are likely to contribute to this characteristic. PRL deficiency leads to female sterility, because in the mouse, PRL is absolutely required for luteal function, implantation, and maintenance of pregnancy (21).

    In hypopituitary Ames and Snell dwarf mice as well as in GH-resistant GHRKO mice, a significant increase in life span is associated with various symptoms of delayed aging (Table 2). These findings suggest that the biological process of aging is altered in these animals and their increased life expectancy is not due to removing causes of early mortality.

    Suspected Mechanisms of Prolonged Longevity

    Association of reduced somatotropic signaling with extended longevity in laboratory stocks of house mice (Mus musculus) is robust, reproducible, and consistent across several mutants, genetic backgrounds, and diets (4, 5, 6, 7, 13, 14, 15, 22). However, at this point, we can only speculate which of the seemingly endless list of direct and indirect consequences of GH deficiency or resistance are causally related to prolonged longevity. In Snell and Ames dwarf mice, the situation is further complicated by concomitant PRL and thyroid stimulating hormone deficiency, the likely importance of hypothyroidism, and possible interactions between the consequences of different hormonal deficits. Despite these limitations, studies conducted to date and comparisons with data obtained in genetically normal animals in which life span was extended by reducing the amount of food they are allowed to consume (the so called "caloric restriction") suggest several potential mechanisms that may link reduced GH/IGF-I signaling with delayed aging.

    Metabolic rate and oxidative damage

    Reduced levels of GH, IGF-I, insulin, and thyroid hormone would be expected to lead to reduced oxidative metabolism and reduced generation of reactive oxygen species (ROS), two very plausible mechanisms of delayed aging. Reduced oxygen consumption (28), body core temperature (20, 29) and mitochondrial ROS production (30), combined with increased expression and/or activity of enzymes involved in antioxidant defenses (31, 32, 33, 34) in hypopituitary and/or GH-resistant mice are consistent with the suspected importance of these mechanisms. In further support of the role of reduced ROS production and enhanced antioxidant defenses as potential mechanisms of delayed aging, oxidative damage of proteins, lipids, and mitochondrial DNA are reduced in Ames dwarf mice (30, 35).

    Stress resistance

    It was recently shown that skin fibroblasts isolated from either Snell dwarf, Ames dwarf, or GHRKO mice survive significantly longer than fibroblasts isolated from normal mice when exposed to various forms of cytotoxic stress in vitro (36, 37). Association of increased stress resistance with delayed aging and extended longevity was described previously in long-lived invertebrate mutants (38) and in mammals subjected to caloric restriction (39).

    Insulin signaling

    Hypoinsulinemia combined with enhanced sensitivity to insulin and reduced or "low normal" glucose levels represent another likely mechanism of extended longevity of GH-deficient and GH-resistant mice (Fig. 1). Reduced mass of pancreatic islets (17, 40), absence of "anti-insulinemic" actions of GH, enhanced secretion of adiponectin (41), and altered expression of genes related to insulin sensitivity including peroxisome proliferators activated receptor and its coactivator PGC1 (41, 42, 43, 78) are among likely mechanisms of reduced insulin release and enhanced insulin sensitivity in these animals. It is well documented that reduced function of homologous (IGF/insulin-like) signaling pathways leads to major extension of life span in worms and flies (reviewed in Refs. 44 and 45). In the human, centenarians were reported to be exceptionally insulin sensitive (46) and to have reduced incidence of diabetes (47). These characteristics of exceptionally long-lived people are remarkable, because human aging is normally associated with a progressive increase in insulin resistance and a significant age-related reduction in insulin sensitivity was evident in normal individuals from the same population (46). Moreover, insulin resistance and "metabolic syndrome" (a condition roughly opposite to the characteristics of insulin signaling in dwarf and GHRKO mice) is a major risk factor for age-associated diseases (48).

    Recent findings raise an interesting possibility that in the absence of GH signaling, the actions of lactogenic hormones during early development may set the stage for enhanced insulin sensitivity in adult life. Fleenor et al. (49) reported that mice with combined GH deficiency and PRL resistance (created by crossing little GHRHRlit and PRL receptor knockout animals) develop insulin resistance and characteristics of metabolic syndrome. This contrasts sharply with the situation described previously in Ames dwarf mice in which combined GH and PRL deficiency is associated with enhanced rather than reduced sensitivity to insulin (18). It is tempting to ascribe this difference to the inability of PRL receptor knockout mice to respond to placental lactogens and maternal PRL (including PRL present in the milk) during prenatal and early postnatal development. However, in adult mice, lactogenic (PRL receptor-mediated) signaling presumably plays little role in regulating sensitivity to insulin since enhanced responses to insulin are present in both Ames dwarfs that are PRL deficient (18, 21) and GHRKO mice (17) that are mildly hyperprolactinemic (50).

    Body size: data from normal mice and other species

    Diminutive body size is a striking characteristic of most long-lived mouse mutants. Association of small stature with prolonged longevity applies also to genetically normal mice and is supported by meta-analysis of multiple studies (51) and by the recent demonstration that small body size is a significant predictor of longer lifespan in individual animals from a genetically heterogeneous population of normal mice (52). Negative correlation of body size and longevity is well documented in domestic dogs (53, 54) and in laboratory stocks of rats (51) and was reported also in other species including the human (55). Presumably, small stature is a phenotypic marker of some developmental and/or metabolic characteristics that predispose to increased life expectancy. However, reduced body size per se is unlikely to lead to delayed aging. Vergara et al. (15) recently reported that 7 wk of treatment of young Snell dwarf mice with GH or GH plus thyroxine failed to shorten their lifespan even though it significantly increased their adult size.

    Gonadal function

    Important interactions between the somatotropic and the hypothalamic-pituitary-gonadal axes and numerous examples of trade-offs between longevity and fecundity (56) suggest that delayed reproductive development and reduced fertility of long-lived mutant mice could be contributing to their extended survival. Existence of such a relationship would be consistent with a concept of antagonistic pleiotropy. Thus, high levels of GH and IGF-I would be favored by natural selection because of their positive impact on sexual maturation and reproductive competence, but would exert negative impacts (e.g. promoting tumorigenesis or accelerating the rate of aging) later in life when the force of natural selection is diminished. However, the potential impact of reduced reproductive competence on aging in hypopituitary and GH-resistant mice appears to be very minor. Comparable life extension and similar maximal life spans were recorded among Snell dwarf mice from a stock in which dwarfs are sterile (6), Snell dwarfs rendered fertile by hormone therapy (15), Ames dwarf mice from a stock in which many females cycle and most males are fertile (4), and in GHRKO mice, in which both sexes can reproduce (12).

    Alterations at the Level of Gene Expression

    Microarray analysis of wide profiles of gene expression (43, 57, 58) and real-time PCR studies of genes related to insulin and IGF-I signaling in various organs (41, 42, 79) strengthen the evidence for reduced IGF-I/insulin signaling in long-lived mutant mice, and demonstrate alternations in gene expression that are consistent with enhanced insulin sensitivity and oxidative stress resistance in these animals. Results of these studies also suggest a possible role of enhanced gluconeogenesis and -oxidation of fatty acids and reduced glycolysis and lipogenesis in the liver, as well as increased expression of Foxo family forkhead transcription factors, peroxisome proliferators-activated receptor , and PGC1 in extending longevity of GH-deficient or GH-resistant mice. Reduced lipogenesis in long-lived mutant mice is strongly supported by elegant studies of various steps of cholesterol biosynthesis in the liver of Snell dwarf mice (59). Interestingly, adiposity as measured by percent of body fat is increased rather than decreased in GHRKO and in young adult Ames dwarf mice (60, 61). We suspect that coexistence of adiposity and enhanced insulin sensitivity in long-lived mouse mutants is due to altered secretion of adiponectin, TNF, and IL-6 by fat cells of these animals (41, 80).

    Brain IGF-I and cognitive function

    Demonstration of enhanced IGF-I expression in the hippocampus of Ames dwarf mice (62, 63) raised an intriguing possibility that preservation and/or compensatory increases in the local production and paracrine/autocrine actions of IGF-I may account for some unexpected phenotypic characteristics of long-lived GH-deficient mutants. Ames dwarf and GHRKO mice do not differ from normal animals in performance of tasks designed to measure learning and memory (25, 26). Moreover these animals maintain youthful levels of cognitive function into a very advanced age (25, 26, 64) despite deficiency of GH signaling and reduction of circulating IGF-I below the level of detectability. Both GH and IGF-I are well documented to exert important neurostimulatory and neuroprotective effects (65, 66). We suspect that the paradox of apparently normal cognitive function and its preservation during aging in animals with deficient somatotropic signaling may be due to normal or enhanced IGF-I biosynthesis and actions in brain regions providing neural substrate for learning and memory.

    Somatotropic Axis and Longevity of Rats

    Although results obtained in rats are in general agreement with the data concerning the effects of GH and IGF-I on aging, some important differences also exist. Heterozygous transgenic rats expressing antisense GH live longer than normal rats, but animals homozygous for the expression of the same transgene have a reduced rather than extended life span (67). Longevity is not affected in dwarf rats which are GH deficient and have plasma IGF-I levels reduced to approximately 50% of normal values (68). Intriguingly, adult onset GH deficiency produced in dwarf rats by GH replacement therapy between 4 and 14 wk of age increased longevity of males significantly above the values measured in either mutated dwarf rats or their normal siblings (68). In the Lou C/Jall rats which are long-lived, lean, and were described as a model of healthy aging, pulsatile GH secretion is maintained during aging, but IGF-I levels are reduced and exhibit a pronounced age-related decrease, implying partial GH resistance (69). Results of meta-analysis of data from over 400 groups of rats revealed significant negative correlation of maximal body size with maximal longevity in this species (51) resembling data obtained in mice.

    Relevance to the Human

    Can the findings in mice summarized above be extrapolated to the human This question is not easy to answer. Because mutations affecting IGF-I/insulin or homologous signaling pathways exert major effects on longevity in organisms ranging from unicellular yeast, through worms and insects, to rodents, it seems entirely reasonable to expect that the involvement of these signaling pathways in the control of aging is universal and includes humans. In the human, some individuals with hypopituitarism due to Prop1 mutations (homologous to Ames dwarfism in mice), or with Laron dwarfism (GH resistance, equivalent to the phenotype of GHRKO mice) can survive into very advanced age (70, 71), but dwarfs with isolated GH deficiency were reported to have reduced life span (72). Pathologically elevated levels of GH are associated with reduced life expectancy in both acromegalic humans (73, 74) and transgenic mice (reviewed in Ref. 75). However, increased mortality rate of acromegalics is due mostly to greater incidence of cardiovascular disease, diabetes, and cancer and thus it could be debated whether the biological process of aging per se is altered in these individuals. As was mentioned earlier in this Minireview, there is also evidence that a negative correlation of body size and longevity, which is very robust in mice and dogs, may apply to the human (55). Association of greater height with shorter lifespan was very striking in some of the examined cohorts, for example in professional baseball players (55). Height can be assumed to represent a biomarker of the activity of the somatotropic axis, including circulating IGF-I levels and responsiveness of target tissues to IGF-I.

    Studies of polymorphic variants of genes related to GH biosynthesis, IGF-I signaling and insulin action (47, 76, 77), provided evidence for altered frequency of several of these variants in exceptionally long-lived people. For example, Kojimra et al. (47) recently reported that one insulin receptor haplotype comprised of two SNPs in linkage disequilibrium was significantly more frequent in semisuper centenarians (individuals who were 105 yr old or older) than in healthy younger controls. A study by van Heemst et al. (76) examined frequency of variants of five genes related to GH and insulin signaling in relation to height and longevity in humans. Based on the expected effects of these genetic variants on hormonal signaling, these authors concluded that a composite score reflecting low activity of this pathway was significantly associated with lower height and improved old age survival in women. Carriers of a SNP variant of the GH1 gene were shorter by 2 cm and had 0.80-fold reduced mortality compared with carriers of a wild-type allele (76). Increased insulin sensitivity in centenarians (46) and detrimental effects of insulin resistance (48) mentioned earlier in this review provide additional indications that at least some of the suspected mechanisms of delayed aging in mice with reduced somatotropic signaling may apply broadly, including humans.

    Acknowledgments

    Apologies are extended to all those whose findings or opinions pertinent to this topic were not referenced or discussed due to limitations of space or inadvertent omissions.

    Footnotes

    This work was supported by the National Institutes of Health (RO1 AG19899 and 1U19 AG023122-01A) and by the Ellison Medical Foundation.

    Abbreviations: GHRKO, GH receptor knockout; PRL, prolactin; ROS, reactive oxygen species.

    References

    Rudman D, Feller AG, Nagraj HS, Gergans GA, Lalitha PY, Goldberg AF, Schlenker RA, Cohn L, Rudman IW, Mattson DE 1990 Effects of human growth hormone in men over 60 years old. New Engl J Med 323:1–6

    Blackman MR, Sorkin JD, Munzer T, Bellantoni M, Busby-Whitehead J, Stevens TE, Jayme J, O’Connor K, Christmas C, Tobin JD, Stewart KJ, Cottrell E, St. Clair C, Pabst KM, Harman SM 2002 Growth hormone and sex steroid administration in healthy aged women and men: a randomized controlled trial. J Am Med Assoc 288:2282–2292

    Klatz R 1997 Grow young with HGH: the amazing medically proven plan to reverse aging. New York: Harper Collins

    Brown-Borg HM, Borg KE, Meliska CJ, Bartke A 1996 Dwarf mice and the ageing process. Nature 384:33–34

    Coschigano KT, Clemmons D, Bellush LL, Kopchick JJ 2000 Assessment of growth parameters and life span of GHR/BP gene-disrupted mice. Endocrinology 141:2608–2613

    Flurkey K, Papaconstantinou J, Miller RA, Harrison DE 2001 Lifespan extension and delayed immune and collagen aging in mutant mice with defects in growth hormone production. Proc Nat Acad Sci USA 98:6736–6741

    Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Evens P, Cervera P, LeBouc Y 2003 IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421:182–187

    Schaible R, Gowen JW 1961 A new dwarf mouse. Genetics 46:896

    Bartke A, Peluso MR, Moretz N, Wright C, Bonkowski M, Winters TA, Shanahan MF, Kopchick JJ, Banz WJ 2004 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 36:550–558

    Snell GD 1929 Dwarf, a new mendelian recessive character of the house mouse. Proc Natl Acad Sci USA 15:733–734

    Eicher EM, Beamer WG 1976 Inherited ateliotic dwarfism in mice. Characteristics of the mutation, little, on chromosome 6. J Hered 67:87–91

    Zhou Y, Xu BC, Maheshwari HG, He L, Reed M, Lozykowski M, Okada S, Wagner TE, Cataldo LA, Coschigano K, Baumann G, Kopchick JJ 1997 A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone receptor/binding protein gene (the Laron mouse). Proc Nat Acad Sci USA 94:13215–13220

    Coschigano KT, Holland AN, Riders ME, List EO, Flyvbjerg A, Kopchick JJ 2003 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 144:3799–3810

    Bartke A, Chandrashekar V, Bailey B, Zaczek D, Turyn D 2002 Consequences of growth hormone (GH) overexpression and GH resistance. Neuropeptides 36:201–208

    Vergara M, Smith-Wheelock M, Harper JM, Sigler R, Miller RA 2004 Hormone-treated Snell dwarf mice regain fertility but remain long lived and disease resistant. J Gerontol A Biol Sci Med Sci 59:1244–1250

    Borg KE, Brown-Borg HM, Bartke A 1995 Assessment of the primary adrenal cortical and pancreatic hormone basal levels in relation to plasma glucose and age in the unstressed Ames dwarf mouse. Proc Soc Exp Biol Med 210:126–133

    Liu J-L, Coschigano KT, Robertson K, Lipsett M, Guo Y, Kopchick JJ, Kumar U, Liu YL 2004 Disruption of growth hormone receptor gene causes diminished pancreatic islet size and increased insulin sensitivity in mice. Am J Physiol Endocrinol Metab 287:E405–E413

    Dominici FP, Hauck S, Argentino DP, Bartke A, Turyn D 2002 Increased insulin sensitivity and upregulation of insulin receptor, insulin receptor substrate (IRS)-I and IRS-2 in liver of Ames dwarf mice. J Endocrinol 173:81–94

    Hsieh C-C, DeFord JH, Flurkey K, Harrison DE, Papaconstantinou J 2002 Implications of the insulin signaling pathway in Snell dwarf mouse longevity: a similarity with the C. elegans longevity paradigm. Mech Ageing Dev 123:1229–1243

    Hunter WS, Croson WB, Bartke A, Gentry MV, Meliska CJ 1999 Low body temperature in long-lived Ames dwarf mice at rest and during stress. Physiol Behav 67:433–437

    Bartke A 1965 Influence of luteotrophin on fertility of dwarf mice. J Reprod Fertil 10:93–103

    Bartke A, Wright JC, Mattison J, Ingram DK, Miller RA, Roth GS 2001 Extending the lifespan of long-lived mice. Nature 414:412

    de Magalhaes JP, Cabral J, Magalhaes D 2005 The influence of genes on the aging process of mice: a statistical assessment of the genetics of aging. Genetics 169:265–274

    Ikeno Y, Bronson RT, Hubbard GB, Lee S, Bartke A 2003 Delayed occurrence of fatal neoplastic diseases in Ames dwarf mice: correlation to extended longevity. J Gerontol A Biol Sci Med Sci 58A:291–296

    Kinney BA, Meliska CJ, Steger RW, Bartke A 2001 Evidence that Ames dwarf mice age differently from their normal siblings in behavioral and learning and memory parameters. Horm Behav 39:277–284

    Kinney BA, Coschigano KT, Kopchick JJ, Bartke A 2001 Evidence that age-induced decline in memory retention is delayed in growth hormone resistant GH-R-KO (Laron) mice. Physiol Behav 72:653–660

    Silberberg R 1972 Articular aging and osteoarthrosis in dwarf mice. Pathol Microbiol 38:417–430

    Benedict FG, Lee RC 1936 La production de chaleur de la souris. Etude de plusieurs races de souris. Ann Physiol Physicochim Biol 12:983–1064

    Hauck SJ, Hunter WS, Danilovich N, Kopchick JJ, Bartke A 2001 Reduced levels of thyroid hormones, insulin, and glucose, and lower body core temperature in the growth hormone receptor/binding protein knockout mouse. Exp Biol Med 226:552–558

    Brown-Borg H, Johnson W, Rakoczy S, Romanick M 2001 Mitochondrial oxidant generation and oxidative damage in Ames dwarf and GH transgenic mice. Amer Aging Assoc 24:85–96

    Brown-Borg HM, Bode AM, Bartke A 1999 Antioxidative mechanisms and plasma growth hormone levels. Endocrine 11:41–48

    Brown-Borg H, Rakoczy S 2000 Catalase expression in delayed and premature aging mouse models. Exp Gerontol 35:199–212

    Hauck S, Bartke A 2000 Effects of growth hormone on hypothalamic catalase and Cu/Zn superoxide dismutase. Free Rad Biol Med 28:970–978

    Brown-Borg HM, Rakoczy SG 2005 Glutathione metabolism in long-living Ames dwarf mice. Exp Gerontol 40:115–120

    Sanz A, Bartke A, Barja G 2002 Long-lived Ames dwarf mice: oxidative damage to mitochondrial DNA in heart and brain. J Am Aging Assoc 25:119–122

    Murakami S, Salmon A, Miller R 2003 Multiplex stress resistance in cells from long-lived dwarf mice. FASEB J 17:1565–1566

    Salmon AB, Murakami S, Bartke A, Kopchick J, Yasumura K, Miller RA 2005 Fibroblast cell lines from young adult mice of long-lived mutant strains are resistant to multiple forms of stress. Am J Physiol Endocrinol Metab 289:E23–E29

    Johnson TE, Lithgow GJ, Murakami S 1996 Hypothesis: interventions that increase the response to stress offer the potential for effective life prolongation and increased health. J Gerontol A Biol Sci Med Sci 51:B392–B395

    Masoro EJ 2001 Dietary restriction: an experimental approach to the study of the biology of aging. In: Masoro EJ, Austad SN, eds. Handbook of the biology of aging. 5th ed. San Diego: Academic Press; 396–420

    Parsons JA, Bartke A, Sorenson RL 1995 Number and size of islets of langerhans in pregnant, human growth hormone-expressing transgenic, and pituitary dwarf mice: effect of lactogenic hormones. Endocrinology 136:2013–2021

    Al-Regaiey KA, Masternak MM, Bonkowski M, Sun L, Bartke A 2005 Long-lived growth hormone receptor knockout mice: interaction of reduced insulin-like growth factor 1/insulin signaling and caloric restriction. Endocrinology 146:851–860

    Masternak M, Al-Regaiey K, Bonkowski M, Panici J, Sun L, Wang J, Przybylski GK, Bartke A 2004 Divergent effects of caloric restriction on gene expression in normal and long-lived mice. J Gerontol A Biol Sci Med Sci 59:784–788

    Tsuchiya T, Dhahbi JM, Cui X, Mote PL, Bartke A, Spindler SR 2004 Additive regulation of hepatic gene expression by dwarfism and caloric restriction. Physiol Genomics 17:307–315

    Guarente L, Kenyon C 2000 Genetic pathways that regulate ageing in model organisms. Nature 408:255–262

    Tatar M, Bartke A, Antebi A 2003 The endocrine regulation of aging by insulin-like signals. Science 299:1346–1351

    Paolisso G, Gambardella A, Ammendola S, D’Amore A, Balbi V, Varricchio M, D’Onofrio F 1996 Glucose tolerance and insulin action in healthy centenarians. Am J Physiol 270:E890–E894

    Kojima T, Kamei H, Aizu T, Arai Y, Takayama M, Nakazawa S, Ebihara Y, Inagaki H, Masui Y, Gondo Y 2004 Association analysis between longevity in the Japanese population and polymorphic variants of genes involved in insulin and insulin-like growth factor 1 signaling pathways. Exp Gerontol 39:1595–1598

    Facchini FS, Hua N, Abbasi F, Reaven GM 2001 Insulin resistance as a predictor of age-related diseases. J Clin Endocrinol Metab 86:3574–3578

    Fleenor D, Oden J, Kelly PA, Mohan S, Alliouachene S, Pende M, Wentz S, Kerr J, Freemark M 2005 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 146:103–112

    Chandrashekar V, Bartke A, Coschigano KT, Kopchick JJ 1999 Pituitary and testicular function in growth hormone receptor gene knockout mice. Endocrinology 140:1082–1088

    Rollo CD 2002 Growth negatively impacts the life span of mammals. Evol Dev 4:55–61

    Miller RA, Harper JM, Galecki A, Burke DT 2002 Big mice die young: early life body weight predicts longevity in genetically heterogeneous mice. Aging Cell 1:22–29

    Eigenmann JE, Amador A, Patterson DF 1988 Insulin-like growth factor I levels in proportionate dogs, chondrodystrophic dogs and in giant dogs. Acta Endocrinol 118:105–108

    Patronek GJ, Waters DJ, Glickman LT 1997 Comparative longevity of pet dogs and humans: implications for gerontology research. J Gerontol 52A:B171–B178

    Samaras T, Elrick H, Storms L 2003 Is height related to longevity Life Sci 72:1781–1802

    Partridge L, Gems D, Withers DJ 2005 Sex and death: What is the connection Cell 120:461–472

    Dozmorov I, Bartke A, Miller RA 2001 Array-based expression analysis of mouse liver genes: Effect of age and of the longevity mutant Prop 1df. J Gerontol A Biol Sci Med Sci 56A:B72–B80

    Miller RA, Chang Y, Galecki AT, Al-Regaiey K, Kopchick JJ, Bartke A 2002 Gene expression patterns in calorically restricted mice: partial overlap with long-lived mutant mice. Mol Endocrinol 16:2657–2666

    Boylston WH, Gerstner A, DeFord JH, Madsen M, Flurkey K, Harrison DE, Papaconstantinou J 2004 Altered cholesterologenic and lipogenic transcriptional profile in livers of aging Snell dwarf (Pit1dw/dwJ) mice. Aging Cell 3:283–296

    Heiman M, Tinsley F, Mattison J, Hauck S, Bartke A 2003 Body composition of prolactin-, growth hormone-, and thyrotropin-deficient Ames dwarf mice. Endocrine 20:149–154

    Berryman DE, List EO, Coschigano KT, Behar K, Kim JK, Kopchick JJ 2004 Comparing adiposity profiles in three mouse models with altered GH signaling. Growth Horm IGF Res 14:309–318

    Sun LY, Al-Regaiey K, Masternak MM, Wang J, Bartke A 2005 Local expression of GH and IGF-1 in the hippocampus of GH-deficient long-lived mice. Neurobiol Aging 26:929–937

    Sun LY, Evans MS, Hsieh J, Panici J, Bartke A 2005 Increased neurogenesis in dentate gyrus of long-lived Ames dwarf mice. Endocrinology 146:1138–1144

    Kinney-Forshee B, Kinney N, Steger R, Bartke A 2004 Could a deficiency in growth hormone signaling be beneficial to the aging brain Physiol Behav 80:589–594

    Markowska AL, Mooney M, Sonntag WE 1998 Insulin-like growth factor-I (IGF-I) ameliorates age-related behavioral deficits. Neuroscience 87:559–569

    Veiga S, Melcangi RC, DonCarlos LL, Garcia-Segura LM, Azcoitia I 2004 Sex hormones and brain aging. Exp Gerontol 39:1623–1631

    Shimokawa I, Higami Y, Utsuyama M, Tuchiya T, Komatsu T, Chiba T, Yamaza H 2002 Lifespan extension by reduction of the growth hormone-insulin-like growth factor-1 axis: relation to caloric restriction. Am J Pathol 160:2259–2265

    Sonntag WE, Carter CS, Ikeno Y, Ekenstedt K, Carlson CS, Loeser RF, Chakrabarty S, Lee S, Bennett C, Ingram R, Moore T, Ramsey M 24 March 2005 Adult-onset growth hormone and IGF-1 deficiency reduces neoplastic disease, modifies age-related pathology and increases lifespan. Endocrinology 10.1210/en.2005-0058

    Kappeler L, Zizzari P, Grouselle D, Epelbaum J, Bluet-Pajot MT 2004 Plasma and hypothalamic peptide-hormone levels regulating somatotroph function and energy balance in fed and fasted states: a comparative study in four strains of rats. J Neuroendocrinol 16:980–988

    Krzisnik C, Kolacio Z, Battelino T, Brown M, Parks JS, Laron Z 1999 The "Little People" of the island of Krk—revisited. Etiology of hypopituitarism revealed. Journal Endocr Genet 1:9–19

    Laron Z 2005 Do deficiencies in growth hormone and insulin-like growth factor-1 (IGF-1) shorten or prolong longevity Mech Ageing Dev 126:305–307

    Besson A, Salemi S, Gallati S, Jenal A, Horn R, Mullis PS, Mullis, PE 2003 Reduced longevity in untreated patients with isolated growth hormone deficiency. J Clin Endocrinol Metab 88:3664–3667

    Jadresic A, Banks LM, Child DF, Diamant L, Doyle FH, Fraser TR, Joplin F 1982 The acromegaly syndrome. Q J Med 202:189–204

    Orme SM, McNally RJQ, Cartwright RA, Belchetz PE 1998 Mortality and cancer incidence in acromegaly: a retrospective cohort study. J Clin Endocrinol Metab 83:2730–2734

    Bartke A 2003 Can growth hormone (GH) accelerate aging Evidence from GH-transgenic mice. Neuroendocrinology 78:210–216

    van Heemst D, Beekman M, Mooijaart SP, Heijmans BT, Brandt BW, Zwaan BJ, Slagboom PE, Westendorp GJ Reduced insulin/IGF-1 signalling and human longevity. Aging Cell 4:79–85

    Bonafe M, Barbieri M, Marchegiani F, Olivieri F, Ragno E, Giampieri C, Mugianesi E, Centurelli M, Franceschi C, Paolisso Gl 2003 Polymorphic variants of insulin-like growth factor I (IGF-I) receptor and phosphoinositide 3-kinase genes affect IGF-I plasma levels and human longevity: cues for an evolutionarily conserved mechanism of life span control. J Clin Endocrinol Metab 88:3299–3304

    Masternak MM, Al-Regaiey KA, Del Rosario Lim MM, Jimenez-Ortega V, Panici JA, Bonkowski MS, Kopchick JJ, Bartke A, Effects of caloric restriction and GH resistance on the expression level of peroxisome proliferator-activated receptors (PPARs) superfamily in the liver of normal and long-lived GHR-KO mice. J Gerontol A Biol Sci Med Sci, in press

    Masternak MM, Al-Regaiey KA, Del Rosario Lim MM, Jimenez-Ortega V, Panici JA, Bonkowski MS, Bartke A, Effects of caloric restriction on insulin pathway gene expression in the skeletal muscle and liver of normal and long-lived GHR-KO mice. Exp Gerontol, in press

    Wang Z, Al-Regaiey KA, Bartke A, The changes of adipocytokines and fat metabolism in Ames dwarf mice and growth hormone transgenic mice. Program of the 87th Annual Meeting of The Endocrine Society, San Diego, CA, 2005, p 360 (Abstract P2-16)(Andrzej Bartke)