Summary
Mutations resulting in reduced signaling of the growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis are associated with increased life and health-span across model organisms. Similar findings have been noted in human cohorts with functional mutations in the somatotropic axis, suggesting that this pathway may also be relevant to human aging and protection from age-related diseases. While epidemiological data indicates that low circulating IGF-1 level may protect aging populations from cancer, results remain inconclusive regarding most other diseases. We propose that studies in humans and animals need to consider differences in sex, pathway function, organs, and time-specific effects of GH/IGF-1 signaling in order to better define the role of the somatotropic axis in aging. Agents that modulate signaling of the GH/IGF-1 pathway are available for human use, but before they can be implemented in clinical studies that target aging and age-related diseases, researchers need to address the challenges discussed in this review.
Introduction
A growing body of evidence points to the importance of the growth hormone/insulin-like growth factor-1 (GH/IGF-1) signaling pathway in the regulation of aging and disease. Although the beneficial effects of attenuated GH/IGF-1 signaling on lifespan and some aspects of healthspan in invertebrates (Kenyon et al., 1993) and rodents (Brown-Borg et al., 1996; Ikeno et al., 2003) have long been recognized, only recently have similar benefits been observed in humans (Guevara-Aguirre et al., 2011; Milman et al., 2014; Suh et al., 2008; van der Spoel et al., 2015). GH and IGF-1 levels change throughout the human lifespan. Circulating GH/IGF-1 levels peak in the 2nd decade of life, a time of considerable cell proliferation and linear growth, but then rapidly decline until the 6th decade, at which time levels plateau (Yamamoto et al., 1991). The effect of this age-related decline in the somatotropic hormones on health in humans is complex and remains unresolved, though there is consensus among scientists that GH replacement is not a viable treatment option in the older adult population.
Data is accumulating from dwarf humans (Guevara-Aguirre et al., 2011) and individuals with exceptional longevity (Milman et al., 2014; Suh et al., 2008; van der Spoel et al., 2015) about the potential beneficial effects of attenuated somatotropic signaling on human lifespan and healthspan, yet results of epidemiological studies remain inconclusive. This ambiguity stems from several shortcomings of epidemiological studies, such as failure to account for sex, age, organ-specific effects and function, rather than levels of GH/IGF-1. Although it is possible to target the GH and/or IGF-1 signaling pathway in humans with pharmaceutical agents, many important questions remain unanswered. At what point in the lifespan and for what duration and magnitude would attenuating the GH and/or IGF-1 pathway impact aging? Does an early decline in somatotropic hormones reflect premature aging or enhanced adaptation to and protection from the consequences of aging? Are there organ-systems in older adults that might suffer detrimental effects from reduced IGF-1 signaling? How are these effects expected to differ between males and females? Is it a reduction in GH, IGF-1 or both that is necessary to achieve these effects? What is the contribution of environmental factors, such as nutrition, to the function of the somatotropic axis? We focus our review on the current state of knowledge about the role of the somatotropic axis in human aging and present some supporting data from animal studies in an effort to establish a framework to guide future research in addressing the knowledge gaps in this area of science.
Regulation of the GH/IGF-1 pathway in humans
GH is secreted from the anterior pituitary gland in pulses, in response to stimulation by growth hormone releasing hormone (GHRH), ghrelin, and dietary stimuli such as protein. GH secretion is inhibited by IGF-1 in a feedback loop, as well as by somatostatin, and other neuroendocrine signals, including insulin (Ceda et al., 1985), which act by binding to their respective receptors. While GH is a major regulator of hepatic IGF-1 expression, IGF-1 is also regulated and secreted by other organs in an autocrine/paracrine manner(Stewart and Rotwein, 1996). Insulin has also been shown to potentiate hepatic IGF-1 secretion in response to GH via up-regulation of GH receptors (Leung et al., 2000). IGF-1 initiates complex intracellular signal cascades through binding to the high-affinity IGF-1R on the cell surface(Holly and Perks, 2012) and activating the phosphorylation of the insulin receptor substrate (IRS) molecules, the (phosphoinositide 3-kinase) PI3K-Akt pathway, and the mitogen activated protein kinase (MAPK) signaling cascade, controlling multiple functions including the mechanistic target of rapamycin (mTOR) activity and FoxO translocation (Taniguchi et al., 2006). IGF-1 is bound to a family of six structurally related binding proteins, termed IGF-binding proteins (IGFBP-1 to 6) that sequester IGF-1 in high-affinity complexes. Hepatic production of IGFBP-1 is under the negative control of insulin and it is inversely correlated with IGF-1 levels, while IGFBP-3 production is under positive regulation of GH (Holly and Perks, 2012). IGFBP-1 circulates in small concentrations, whereas IGFBP-3 is the most abundant of these binding proteins. In circulation, most IGF-1 is bound to IGFBP-3 as part of a 150 kD ternary complex involving a third protein, the acid-labile subunit (ALS)(Holly and Perks, 2012). This highly regulated process protects IGF-1 and IGFBPs from rapid degradation or elimination from the serum and keeps the circulating concentrations of biologically active, un-complexed ‘free’ IGF-1 at relatively low levels (Baxter, 2000; Rajaram et al., 1997). Of note, some of the IGFBPs exhibit actions that are independent of IGF-1 and impact functions such as glucose metabolism (Kim et al., 2007; Muzumdar et al., 2006) and cell-cycle (Cobb et al., 2009). With aging, the amplitude and frequency of GH pulses decline, resulting in decreased circulating IGF-1 levels (Rudman et al., 1981). However, the impact of this decline on autocrine production of IGF-1 by brain and peripheral tissues in humans is unknown.
Human “models” of GH/IGF-1 attenuation and aging
Humans with genetic variations or mutations within the GH/IGF-1 pathway serve as models for understanding the relationship between the somatotropic axis, health, and longevity. Individuals with non-functional GH receptor (GHR), resulting from a mutation in the GHR gene (Laron dwarfs), manifest a significantly reduced risk of developing cancer, stroke, and type 2 diabetes mellitus (T2DM) (Guevara-Aguirre et al., 2011; Steuerman et al., 2011). Nonagenarian female carriers of genetic polymorphisms that attenuate the action of the GH/IGF-1 pathway exhibit longer survival (van Heemst et al., 2005). Likewise, individuals with exceptional longevity are enriched with functional mutations in the IGF1R gene, which confer partial IGF-1 resistance (Suh et al., 2008; Tazearslan et al., 2011). Variants in AKT1 and FOXO3A genes, which are elements of the GH/IGF-1 signaling pathway, have been identified in long-lived individuals from several different cohorts of diverse ethnic backgrounds, although their functional role has not yet been defined (He et al., 2014; Pawlikowska et al., 2009). Furthermore, centenarians exhibit a higher than age-predicted level of a specific microRNA, hsa-miR-363*, which regulates AKT1 and IGFBP-5 (Gombar et al., 2012). This indicates that posttranslational regulation of the GH/IGF-1 axis may also play a role in longevity. However, since carriers of these variants have been exposed to reduced activity in various signaling or downstream effectors of this pathway throughout the lifecourse, the question of whether these beneficial effects require developmental exposure or could be alternatively achieved by pharmacologic interventions at later ages remains unresolved.
Humans with exceptional longevity serve as models of successful aging. Aging is the major underlying risk factor for numerous diseases, including cardiovascular disease (CVD), cancer, diabetes mellitus type 2 (T2DM), and dementia. Yet, centenarians demonstrate a significant delay in the onset of most age-associated diseases and frequently escape from disease altogether (Andersen et al., 2012). Circulating IGF-1 levels have been inversely correlated with survival in those with lifespan extending into the tenth decade and beyond. Our group demonstrated that lower IGF-1 levels can predict extended survival in Ashkenazi Jewish female nonagenarians and nonagenarians of both sexes with a history of cancer (Milman et al., 2014). In Dutch nonagenarians of both sexes, the hazard of death was 27% lower among those with IGF-1/IGFBP-3 ratios in the first quartile compared to those in the fourth quartile (van der Spoel et al., 2015). Furthermore, those with the lowest IGF-1/IGFBP-3 demonstrated higher scores on performance of activities of daily living (ADL) (van der Spoel et al., 2015). Such evidence suggests that diminished function of the GH/IGF-1 axis may be beneficial to human aging and promotes longevity by conferring protection against age-related diseases and dysfunction.
GH/IGF-1 axis and age-related diseases
A number of human studies now support the notion that attenuation of GH/IGF-1 signaling may protect from age-related diseases and functional decline. This evidence is more robust for several site-specific cancers, but remains equivocal when it comes to cardiovascular disease, T2DM, and neurodegenerative conditions. On the other hand, age-related osteoporotic bone loss is offset by higher circulating IGF-1 levels, but this effect may be sex and age specific. Below, we review the available evidence from human and animal studies that associate the GH/IGF-1 axis with age-related diseases and mortality.
Neoplastic diseases
Epidemiological evidence reveals that higher levels of circulating IGF-1 are associated with future increased risk of multiple cancers in human populations (Cao et al., 2015; Key et al., 2010; Renehan et al., 2004; Rinaldi et al., 2010). Mechanistically, IGF-1 is believed to promote malignancy by activating downstream signaling via the IRS/Akt/MAPK pathway, which promotes cell growth and proliferation (Novosyadlyy and Leroith, 2012). The evidence linking IGF-1 and cancer in humans is strongest for breast (Key et al., 2010; Renehan et al., 2004) and prostate cancer (Cao et al., 2015; Renehan et al., 2004; Rowlands et al., 2009), while moderate evidence exists for a relationship with colorectal cancer (Rinaldi et al., 2010). However, the association of IGF-1 and lung cancer is equivocal (Cao et al., 2012; Renehan et al., 2004). Additionally, IGFBP levels have been related to cancer risk. Higher IGFBP-3 level has been linked to breast cancer (Renehan et al., 2004), whereas lower IGFBP-1 was associated with reduced risk for prostate cancer (Cao et al., 2015).
The role of GH per se on cancer risk in humans is less clear. Patients with acromegaly, characterized by excess GH secretion, have been noted to have an increased risk of multiple cancers, including colon, thyroid, and prostate (Boguszewski and Ayuk, 2016), while Laron dwarfs appear to be protected from malignancy (Guevara-Aguirre et al., 2011; Steuerman et al., 2011).. However, alterations in GH signaling are also accompanied by concurrent changes in IGF-1 and insulin levels. This relationship has made it challenging to characterize the independent contribution of GH to aging and diseases of aging, though recent generation of animal models with extrahepatic targeted deletion of GH receptors (GHR) represent a significant step forward in overcoming these limitations to better clarify the role of GH (Jara et al., 2016; List et al., 2015).
Numerous studies in rodents have demonstrated the importance of GH, IGF-1 and insulin to tumorigenesis (Ikeno et al., 2003). Indeed, caloric restriction (CR), which reduces GH/IGF-1 and insulin levels in rodents (Breese et al., 1991), potently retards tumor transformation and progression (Huffman et al., 2007; Reddy et al., 1987). Moreover, replacing GH or IGF-1 completely abrogated the protective effects of CR on carcinogenesis (Hursting et al., 1993). Furthermore, pre-clinical studies employing IGF-1R antibodies have proven effective in retarding cancer progression (Fahrenholtz et al., 2013; Galet et al., 2013), though its use as a monotherapy in clinical trials for advanced stage cancers were not as successful (Fuchs et al., 2015; Robertson et al., 2013). GH may impact tumor growth, independent of circulating IGF-1, since deletion of hepatic Igf1 did not reduce prostate tumor progression in mice (Anzo et al., 2008), although the LID model used to generate this deficiency is also markedly insulin resistant (Haluzik et al., 2003). Human cancer cell lines, and in particular metastatic melanoma cell lines, express GHR, with some lines displaying increased proliferation in response to GH (Sustarsic et al., 2013). Whether these effects on cancer progression are directly mediated by GH or via GH-stimulated local production of IGF-1 remains to be determined. It should also be noted that whole body, rather than tissue-specific modulation of IGF-1 or its receptor, impact on GH secretion and insulin sensitivity, may explain some differences in findings between systemic and tissue specific models.
Several analyses of cancer-associated mortality and circulating IGF-1 levels in humans have revealed a U-shaped relationship, with both lowest and highest levels of IGF-1 associated with greater hazard of cancer-related mortality (Burgers et al., 2011; Svensson et al., 2012). Yet, not all studies have confirmed this association, with many finding no link between IGF-1 and cancer-specific mortality (Friedrich et al., 2009; Saydah et al., 2007; van Bunderen et al., 2010). However, it is important to note that several of these studies have analyzed data from individuals over a wide age range, beginning as young as 20 years old (Friedrich et al., 2009; Saydah et al., 2007), without regard for the possibility that the relationship between IGF-1 and mortality may differ significantly according to age group. One study identified a positive correlation between self-reported protein intake and circulating IGF-1 level in people age 50–65 from the National Health and Nutrition Examination Survey III (NHANES III), with consumers of a high protein diet having a 4-fold increased risk in cancer mortality over 18-years of follow-up compared to those who consumed a low-protein diet (Levine et al., 2014). However, protein consumption did not correlate with IGF-1 levels in individuals age 66 and older in that study (Levine et al., 2014).
Cardiovascular disease
The relationship between cardiovascular disease, cardiovascular-associated mortality and IGF-1 levels in humans is conflicting. Higher levels of circulating IGF-1 have been positively correlated with risk for congestive heart failure (Andreassen et al., 2009), but not consistently across studies (Vasan et al., 2003). No significant association has been found between IGF-1 and future risk for cardiovascular events, such as myocardial infarction or stroke (Andreassen et al., 2009; Kaplan et al., 2007; Ricketts et al., 2011). On the other hand, several population studies have found about 1.5–2 times increased risk for cardiovascular mortality among individuals with lower circulating IGF-1 levels (Friedrich et al., 2009; Juul et al., 2002; Laughlin et al., 2004; Svensson et al., 2012). Analysis of data from NHANES III did not find a significant association between IGF-1 levels and cardiovascular mortality (Saydah et al., 2007), whereas a population study of older individuals in the Netherlands and a meta-analysis revealed a U-shaped relationship between IGF-1 and cardiovascular mortality (Burgers et al., 2011; van Bunderen et al., 2010). Further, while data is far more limited, one study reported high IGFBP-3 was associated with greater risk of death from ischemic heart disease (Juul et al., 2002).
Animal studies examining the role of GH and IGF-1 signaling on the vasculature and myocardium have proven to be equally complex. GH transgenic mice have cardiac hypertrophy and increased fibrosis (Bollano et al., 2000), similar to humans with acromegaly who have excess circulating GH and IGF-1 (Lopez-Velasco et al., 1997), while GHRKO mice have generally normal cardiac function and less fibrosis with aging (Egecioglu et al., 2007). On the other hand, Lewis dwarf rats, which have GH and IGF-1 deficiency, have increased incidence of stroke and arterial stress (Csiszar et al., 2008; Ungvari and Csiszar, 2012). However, because these models cannot distinguish from concomitant changes in circulating IGF-1, a model was generated with targeted disruption of GHR in cardiomyocytes (Jara et al., 2016); yet, no alteration in cardiac performance was observed in this model. In contrast, cardiac specific deletion of IGF-1R in adult mice was shown to impair diastolic function, while adult onset IGF-1 deficiency mice demonstrated multiple impairments in cardiovascular function (Bailey-Downs et al., 2012; Li et al., 2008). Collectively, these data suggest that IGF-1R signaling is most critical to cardiac structure and function (Moellendorf et al., 2012).
Animal models suggest that IGF-1 may protect from endothelial dysfunction and reduce pro-inflammatory signaling, thereby contributing to protection form cardiovascular disease (Higashi et al., 2012). In contrast, others have observed that whole-body haploinsufficiency or conditional deletion of the IGF-1R gene in endothelial cells resulted in beneficial effects on endothelial nitric oxide synthase (eNOS) phosphorylation (Abbas et al., 2011) and vascular wall repair (Yuldasheva et al., 2014). Interestingly, Ames hypopituitary mice, which are deficient in circulating GH/IGF-1, manifest delayed aging, yet they exhibit increased vascular oxidative stress (Csiszar et al., 2008). While these studies, particularly the recent inclusion of conditional models, have shed greater insights into the role of GH versus IGF-1 signaling on the cardiovascular system, it should be noted that the vast majority of studies have been performed in young, male rodents. Moving forward, age and sex differences, as well as timing of deficiencies, should be carefully considered in order to more clearly define the role of GH and IGF-1 signaling on the developing and aging myocardium and vasculature.
Diabetes Mellitus Type 2
The somatotropic axis is intricately linked to the insulin pathway and thereby glucose homeostasis. The insulin and IGF-1 signaling pathways are evolutionary conserved among species. Reduced signaling through these pathways results in lifespan extensions in both invertebrate and mammalian models. Unlike mammals, invertebrates have a single orthologue of the IGF-1 and insulin receptor that binds insulin-like peptides (Russell and Kahn, 2007). In mammals on the other hand, signaling through the IGF-1R and insulin receptor (InsR) converge at the PI3K-Akt and MAPK pathways (Novosyadlyy and Leroith, 2012; Russell and Kahn, 2007; Taniguchi et al., 2006). IGF-1 and insulin can each bind to the IGF-1R and InsR, although each ligand binds to its own receptor with 100 to 1,000 fold greater affinity (Novosyadlyy and Leroith, 2012). Furthermore, IGF-1R and InsR can combine to form heterodimers known as IGF-1R/InsR hybrids, and the expression of these hybrid receptors can account for a significant proportion of total receptor stoichiometry in some tissues, including heart, muscle and brain (Bailyes et al., 1997). Physiologically, IGF-1 has been shown to have similar effects as insulin both in the periphery (Hawkins et al., 1996) and in the brain (Muzumdar et al., 2006) to regulate glucose homeostasis, though centrally-acting IGF-1 proved more potent than insulin in regulating peripheral glucose fluxes in aging (Huffman et al., 2016).
Despite the integration of the two systems, prospective studies in humans that look at the risk of developing diabetes mellitus type 2 (T2DM) in association with the GH/IGF-1 axis are sparse. A prospective study of middle-aged men and women found that IGF-1 levels above the median were associated with 50% lower odds of developing impaired glucose tolerance or T2DM (Sandhu et al., 2002). Another study that looked at the risk of developing T2DM in middle-aged women did not find an association between T2DM and serum total IGF-1 level, but did note a negative association between IGFBP-1 level and T2DM (Rajpathak et al., 2012). This study also noted an increased risk for T2DM in women in the highest quintile of IGFBP-3 compared to those in the lowest quintile (Rajpathak et al., 2012). The results of these cohort studies differ substantially from the findings in the Laron dwarfs (Guevara-Aguirre et al., 2011), in whom lifelong inactivity of GH and deficiency of IGF-1 is protective from T2DM, although the absence of GH action may be largely responsible for this protection. Indeed, while haploinsufficiency for the IGF-1R results in impaired glucose tolerance (Holzenberger et al., 2003), mice deficient in GH and IGF-1 (GHRKO) are more insulin sensitive (Al-Regaiey et al., 2005).
Neurodegenerative diseases
The association between circulating IGF-1 levels and cognitive function in humans remains an issue of current debate. One cross-sectional analysis showed that higher circulating IGF-1 level was positively correlated with cognitive function in older men, but not women (Al-Delaimy et al., 2009). Similarly, a separate analysis revealed an association between mild cognitive impairment and IGF-1 among individuals in the lowest quartile of IGF-1 levels (Doi et al., 2015). The results of prospective studies have been less inconsistent, with some suggesting poorer cognitive performance and increased risk of Alzheimer’s disease in individuals with lower IGF-1 levels (Okereke et al., 2007; Westwood et al., 2014). Meanwhile, another prospective study conducted in men revealed no association between cognitive function and circulating IGF-1 level (Green et al., 2014). On the contrary, a recent analysis of men older than 40 revealed that individuals with circulating IGF-1 levels in the top quintile at baseline demonstrated poorer processing capacity and global cognitive scores after an average of 8 years of follow-up (Tumati et al., 2016). Interestingly, a year-long supplementation trial with GH in postmenopausal women had no affect on memory (Friedlander et al., 2001). Despite the conflicting results related to circulating GH and IGF-1, the paracrine effect of locally-produced GH and IGF-1 in the CNS remains an important caveat for which little is currently known, and therefore should also be considered.
Studies in rodents focused on the IGF-1 axis have been most well studied, and have revealed that IGF-1 levels in circulation, CSF and brain tissue decline with advancing age in some strains, while expression of the IGF-1R in various regions of the CNS either increases, decreases, or remains unchanged (Ashpole et al., 2015; Huffman et al., 2016). Evidence regarding the role of local GH and IGF-1 in cognition is limited, but expression of GH and IGF-1 in the hippocampus of dwarf mice was found to be paradoxically increased, suggesting that maintenance of adequate IGF-1 levels in the brain may be critical to normal cognitive function in these mice (Sun et al., 2005). In addition to local production, the brain receives peripheral input of IGF-1 via a transport mechanism in the choroid plexus involving the low-density lipoprotein receptor-related protein 2 (LRP2) and the IGF-1R (Fernandez and Torres-Aleman, 2012), but some evidence suggests that IGF-1 input from CSF into the brain is impaired with aging (Muller et al., 2012). However, IGF-1 replacement targeting the CNS in aged rodents, can improve learning and memory. The exact mechanisms involved in this effect are not clear, but appear to involve the cerebrovascular system, neurovascular coupling, regulation of N-methyl-D-aspartate (NMDA) receptors, synaptic function, and synaptic plasticity (Ashpole et al., 2015; Sonntag et al., 2005; Toth et al., 2015). IGF-1 may also influence cognition in aging via its effect on neurogenesis (Lichtenwalner et al., 2001). In contrast, some evidence in nematodes suggests that insulin/IGF-1 signaling may actually inhibit regeneration in aging neurons (Byrne et al., 2014). Similarly, knocking out the IGF-1R in the rodent brain has been linked to protection against various neurological conditions and functions (Chaker et al., 2015; Gontier et al., 2015). Thus, the role of IGF-1 in the brain is highly complex and to fully understand it will likely require a better appreciation of not only the site(s) of action, but also its specific role in the context of the aging central nervous system.
Osteoporosis
Decreased bone mineral density (BMD) that results in bone fragility, termed osteoporosis, leads to increased risk of fracture and often presents as a manifestation of aging. Both GH and IGF-1 regulate bone metabolism through endocrine and paracrine/autocrine signaling. GH induces IGF-1 production by osteoblasts. IGF-1 affects osteoblast differentiation and bone mineralization. Parathyroid hormone also contributes to IGF-1 gene transcription in bone. Although the role of GH/IGF-1 axis on bone mineral density maintenance has long been accepted, the relative contribution of GH and IGF-1 to skeletal acquisition remains difficult to interpret, particularly as it may differ between the sexes (Liu et al., 2016). Several cross-sectional studies conducted in older populations have found a positive correlation between IGF-1 levels and BMD specific to women or men only (Janssen et al., 1998; Langlois et al., 1998). Prospective analysis confirmed a similar association in women (van Varsseveld et al., 2015). Low IGF-1 level has been linked to increased risk of future fracture in men and women, although not all studies have shown consistent sex-specific effects (Ohlsson et al., 2011; van Varsseveld et al., 2015; Yamaguchi et al., 2006).
Interestingly, recent evidence in a mouse model of inducible IGF-1 deficiency suggests that low IGF-1 levels early, but not later in life, led to reductions in cortical bone structure and strength in both males and females, though shifts in trabecular bone were only observed in males (Ashpole et al., 2016). Similarly, Gong et al observed that IGF-1 deficiency, despite an excess of GH, led to compromised skeletal integrity and accelerated bone loss (Gong et al., 2014). In contrast, restoration of endocrine IGF-1 was insufficient to restore skeletal integrity in the absence of GHR, suggesting that both GH and IGF-1 are required for normal skeletal development and integrity (Wu et al., 2013). Therefore, in designing age-delaying strategies targeting the GH/IGF-1 axis, it will be critical to consider the importance of both hormones, as well as ramifications of age, sex and other factors on the skeletal system.
All-cause Mortality
Defects in the somatotrophic axis have been linked to longevity in invertebrate and rodent models (Bokov et al., 2011; Brown-Borg et al., 1996; Kenyon et al., 1993; Xu et al., 2014). While several models of combined GH and IGF-1 deficiency have been linked to improved longevity in males and females, the effect of isolated IGF-1 or IGF-1R deficiency has often been sex-specific, with females, rather than males, demonstrating improved survival (Holzenberger et al., 2003; Xu et al., 2014). Despite lower IGF-1 being predictive of extended survival in individuals with exceptional longevity (Milman et al., 2014; van der Spoel et al., 2015), its association with all-cause mortality in the general population remains inconsistent. Several assessments in older adult cohorts have found increased hazard of death among people with lower IGF-1 levels (Roubenoff et al., 2003; van Bunderen et al., 2010), although a study in men did not find consistent results (Maggio et al., 2007). A population-wide study found no association between IGF-1 levels and all-cause mortality (Saydah et al., 2007). There was also no association between the rate of IGF-1 decline over time and mortality in a cohort aged 77 and older (Kaplan et al., 2012). A number of analyses conducted in middle aged and older populations demonstrated a positive association between IGF-1 levels and mortality, with several revealing a U-shaped relationship (Andreassen et al., 2009; Burgers et al., 2011; Svensson et al., 2012).
Unraveling the Inconsistencies
As exemplified above, much of the knowledge about the affects of the GH/IGF-1 axis on age-associated diseases and aging in humans remains inconsistent. Several aspects of this controversy could potentially be unraveled by employing innovative study approaches in human populations. In addition, novel animal models can help to expand our understanding of GH/IGF-1 related mechanisms that impact human aging and disease. In the following sections we highlight several important issues that should be addressed through epidemiological, clinical and basic research. We also propose methodologies that can be employed to clarify some of the uncertainties. Only after these issues are better addressed can clinical trials that target the somatotropic axis be considered as a strategy to delay aging and its diseases.
Consideration of temporal factors that mediate the effect of GH/IGF-1 on disease and lifespan
Aging researchers recognize that the effect of a hormone may differ dramatically in a youthful organism compared to an older one. The concept of antagonistic pleiotropy, which posits that some factors may have been evolutionarily selected due to beneficial effects for growth and reproduction, yet could have harmful effects later, may be highly relevant to the somatotropic axis. Indeed, whereas sufficient levels of GH/IGF-1 are beneficial for the development and growth in the young, they may be detrimental to an aging organism by attenuating putative stress resistant pathways and accelerating malignancy. Whether a “switch” from beneficial to harmful exists for GH and IGF-1, and at what point in the lifespan it occurs remains unknown. IGF-1 is required for normal brain development (Liu et al., 2009) and appears to play an important role in cardiovascular health and bone acquisition, but persistently high circulating IGF-1 levels throughout mid and late life may come at the expense of greater risk for malignancy (Renehan et al., 2004). Therefore, when analyzing data on IGF-1 it is important to stratify analysis by age rather than pool all ages. Statistically adjusting for age does not satisfy this need because it assumes a linear relationship between age and IGF-1, whereas the true relationship may actually be positive during youth and inverse thereafter.
Other important questions center on the duration of exposure to increased or diminished GH/IGF-1 levels that is required for the onset of the beneficial age-associated effect and in what organ-systems these pathways should be targeted. Furthermore, lower IGF-1 levels may serve as either a protective mechanism during aging or a sign of accelerated aging. Low circulating IGF-1 level can be induced by illness, malnutrition, and debilitation; thus in cross-sectional analyses poor health status would be linked to lower IGF-1. Thus prospective studies in humans and animals are necessary to answer many of these questions.
Sexual dimorphism in GH/IGF-1 action
It is now becoming increasingly recognized that the effects of GH, and particularly IGF-1, on physiology, disease and survival are sex-specific (Bokov et al., 2011; Holzenberger et al., 2003; Milman et al., 2014; Xu et al., 2014). This has been observed to varying degrees in lifespan studies utilizing IGF-1R+/− animals (Bokov et al., 2011; Holzenberger et al., 2003; Xu et al., 2014), as well as disease-specific analyses in rodent models and humans (Liu et al., 2016; Milman et al., 2014; Wang et al., 2006). Going forward, sex stratification should be employed in all studies looking at the effects of the somatotropic axis in aging and disease. Furthermore, uncovering the mechanism(s) involved in these sexually-dimorphic effects on aging phenotypes should be a priority for future investigations in humans and animal models in order to enable tailored approaches to modulate these pathways in males and females. This can be further enhanced by studies addressing the potential role of sex steroids on the response to GH, IGF-1 and insulin signaling, with specific consideration of both developmental and later life exposure to androgens and estrogen.
Addressing the apparent paradoxical effect of GH/IGF-1 in different organs and diseases
IGF-1 functions both as a circulating endocrine hormone and as a paracrine/autocrine signaling molecule. Detectable circulating IGF-1 levels may not be reflective of the concentration of IGF-1 at the tissue level, where it may be locally produced and exert paracrine and autocrine function. In fact, Ames dwarf mice, which are deficient in circulating GH and IGF-1, among other hormones, have elevated levels of hippocampal GH and IGF-1 (Sun et al., 2005). Similarly, some cohorts of Laron dwarfs who exhibit GH insensitivity appear to have normal cognitive function, presumptively resulting from local production of IGF-1 in the brain (Kranzler et al., 1998). Based on this evidence we propose that declining circulating levels of IGF-1 may lead to greater local tissue production of IGF-1 as a result of loosened negative feedback that may be exerted by circulating IGF-1. This may explain how different organ systems that have varying IGF-1 requirements may maintain optimal function despite systemic decline in circulating IGF-1.
Alternatively, the beneficial health effects of higher circulating IGF-1 may be age dependent. As mentioned above, GH and IGF-1 regulated bone accrual was observed only in younger animals, but not in the older rodents (Ashpole et al., 2016). Thus, the age-associated decline in GH/IGF-1 levels may results in protection from malignancies without compromise of the musculoskeletal system in the aged individuals. Furthermore, the effect of IGF-1 on disease incidence or progression may differ depending on the underlying health of the organ in question. Gaining a better understanding of the production and regulation of tissue specific IGF-1 in humans is important for clarifying the physiology. Although some experiments may be prohibitive in humans, studies conducted in other model organisms may inform our understanding.
Genetic and functional approaches to resolve epidemiologic uncertainty
While lower IGF-1 levels are strongly associated with protection from various types of cancer and increased risk of osteoporosis, the effect of attenuated IGF-1 function on CVD, T2DM, dementia, and mortality remains inconclusive. This uncertainty, in part, results from observational studies that pool diverse groups of people based solely on the circulating IGF-1 phenotype (e.g. level of circulating IGF-1), not accounting for the individual genotypes that might impact on the response to this ligand. This approach would have resulted in the cohort of centenarians harboring functional IGF-1R mutation(s), which causes elevation in the IGF-1 level, to be grouped together with all other individuals with elevated IGF-1 levels, despite evidence that carriers of this mutation demonstrate IGF-1 resistance and decreased IGF-1 function (Suh et al., 2008; Tazearslan et al., 2011). These functional differences are not reflected in simple measurements of IGF-1 levels, but can be deduced from the genome coupled with functional assays and models. Transgenic animal models that harbor mutations or genomic variants identified in humans would help characterize the function of these genetic elements and assist with proper characterization of subjects, as was demonstrated by our group (Tazearslan et al., 2011). Thus future epidemiologic or experimental studies should consider phenotyping other components related to the IGF-1 response in addition to IGF-1, which should result in more accurate subject characterization and study conclusions. This novel approach will foster a more physiologically relevant assessment of the role of IGF-1 in age-associated diseases and longevity.
Unique populations serving as human “models”
Understanding the role of GH/IGF-1 on aging by utilizing human “models,” as described above, is unparalleled. Through studying the genetic make-up of centenarians, who are prototypes of healthy aging, one can uncover the effects of genomic variations that modulate the GH/IGF-1 system on healthspan and lifespan without following an individual for a lifetime. Furthermore, by studying the offspring of centenarians, who inherit, at least partially, the centenarian genome, permits one to validate prospectively the effect of these candidate genes against an age-matched control group.
Contribution of environmental factors to the regulation of the somatotropic axis
Certain environmental factors have also been shown to influence IGF-1 levels. Animal protein intake has been linked to higher circulating IGF-1 levels in individuals age 50–65 years (Levine et al., 2014). On the other hand, prolonged fasting was associated with reduction in IGF-1 levels, although moderate caloric restriction of 9% in humans has not been shown to reduce circulating IGF-1 (Fontana et al., 2016). Hormone replacement therapy in women may also modulate IGF-1 and IGFBP levels (Posaci et al., 2001). These factors should be considered in future studies.
Future Directions
Therapeutic modalities that attenuate GH/IGF-1 signaling
A number of available therapeutics that have been designed for human use can block the signaling of the GH/IGF-1 pathway. These include somatostatin analogs, growth hormone receptor antagonists and anti-IGF-1R antibodies. The effect of these agents on health and lifespan is currently being investigated in several different experimental models. Future studies conducted in humans and animal models should focus on elucidating the highlighted uncertainties that currently surround the association between GH/IGF-1 axis and aging. Only after these questions have been answered can we consider therapeutic trials in aging humans.
Consideration of other health-related outcomes
The impact of modulating GH/IGF-1 signaling on human health should not only consider effects on diseases per se, but also on health-span, robustness against common stressors encountered with aging and maintenance of independent function. Indeed, data from rodent models of GH and/or IGF-1 deficiency suggest improved stress resistance (Wang and Miller, 2012). Furthermore, evidence from long-lived humans indicates that lower IGF-1/IGFBP-3 ratio is related to improved performance of activities of daily living (ADLs) (van der Spoel et al., 2015). On the other hand, recent data from long-lived c. elegans daf-2 mutants suggests that a significant proportion of their lifespan is spent in suboptimal functional status (Bansal et al., 2015). Thus, these important outcomes should also be considered.
Here, we have laid out a framework for research that is needed to address the knowledge gaps surrounding the somatotropic axis and its impact on aging. Animal models can specifically be utilized to help clarify questions arising from human studies, where human studies would not be feasible. However, the utility of rodents as a model of IGF-1 and age-related disease should consider important limitations, including the fact that mortality is typically attributable to malignancy and renal disease in rats and mice, where IGF-1 may have harmful effects with age. In contrast, aging humans commonly suffer from cardiovascular disease, dementia, T2DM and osteoporosis, ailments where the role of IGF-1 is less defined. Finally, little is known about the role of other IGF axis components on aging biology, including IGF-2, despite the fact that circulating levels of this related ligand are several fold greater than IGF-1 in humans. Beyond its importance to development, evidence suggests a potentially important role in malignancy and cardiovascular disease, but its relevance to other diseases is not known. Certainly, the fact that adult rodents lack circulating IGF-2 has impaired progress in our understanding of this hormone (Bergman et al., 2013).
Conclusion
Genomic studies in humans and animals reveal a beneficial impact of attenuated IGF-1 action on several age-related diseases and lifespan; thus, bringing into question the assertion that the decline in circulating IGF-1 levels during aging are inherently detrimental. Despite the aforementioned evidence from dwarf humans and individuals with exceptional longevity, the results from epidemiological studies have not provided the same degree of clarity on these relationships. The inconsistencies among human study results are disease-specific, with reduced incidence of site-specific cancers clearly associated with low IGF-1 levels, while relationships with CVD, T2DM and cognitive decline are less consistent. These discrepancies partially stem from research methodological factors, including the lack of sex and age stratification in the analysis, functional and organ-specific characterization of the GH/IGF-1, and limited information regarding the degree of modulation observed in these pathways. An interdisciplinary approach will be required in order to better characterize the role of GH/IGF-1 in humans. Such an approach will require an increased emphasis on controlling for factors highlighted above in human investigations, while utilizing studies in model organisms to better recapitulate the pleiotropic actions of these hormones across cells and systems. Such efforts will be necessary to successfully translate approaches to modulate the GH/IGF-1 axis to promote healthy aging in humans.
Acknowledgments
This work was funded by 1K23AG051148-01 (S.M.), American Federation for Aging Research (S.M., D.M.H.), R00AG037574 (D.M.H.), Grants from the National Institutes of Health (P01AG021654) (N.B.), The Nathan Shock Center of Excellence for the Biology of Aging (P30AG038072) (N.B.), the Glenn Center for the Biology of Human Aging (Paul Glenn Foundation for Medical Research) (N.B.), NIH R37 AG18381 (Barzilai Merit Award), and NIH/NIA 1 R01AG044829 (PIs-Veghese/Barzilai).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Abbas A, Imrie H, Viswambharan H, Sukumar P, Rajwani A, Cubbon RM, Gage M, Smith J, Galloway S, Yuldeshava N, et al. The insulin-like growth factor-1 receptor is a negative regulator of nitric oxide bioavailability and insulin sensitivity in the endothelium. Diabetes. 2011;60:2169–2178. doi: 10.2337/db11-0197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al-Delaimy WK, von Muhlen D, Barrett-Connor E. Insulinlike growth factor-1, insulinlike growth factor binding protein-1, and cognitive function in older men and women. J Am Geriatr Soc. 2009;57:1441–1446. doi: 10.1111/j.1532-5415.2009.02343.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. 2005;146:851–860. doi: 10.1210/en.2004-1120. [DOI] [PubMed] [Google Scholar]
- Andersen SL, Sebastiani P, Dworkis DA, Feldman L, Perls TT. Health span approximates life span among many supercentenarians: compression of morbidity at the approximate limit of life span. J Gerontol A Biol Sci Med Sci. 2012;67:395–405. doi: 10.1093/gerona/glr223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andreassen M, Raymond I, Kistorp C, Hildebrandt P, Faber J, Kristensen LO. IGF1 as predictor of all cause mortality and cardiovascular disease in an elderly population. Eur J Endocrinol. 2009;160:25–31. doi: 10.1530/EJE-08-0452. [DOI] [PubMed] [Google Scholar]
- Anzo M, Cobb LJ, Hwang DL, Mehta H, Said JW, Yakar S, LeRoith D, Cohen P. Targeted deletion of hepatic Igf1 in TRAMP mice leads to dramatic alterations in the circulating insulin-like growth factor axis but does not reduce tumor progression. Cancer research. 2008;68:3342–3349. doi: 10.1158/0008-5472.CAN-07-3165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ashpole NM, Herron JC, Estep PN, Logan S, Hodges EL, Yabluchanskiy A, Humphrey MB, Sonntag WE. Differential effects of IGF-1 deficiency during the life span on structural and biomechanical properties in the tibia of aged mice. Age (Dordr) 2016;38:38. doi: 10.1007/s11357-016-9902-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ashpole NM, Sanders JE, Hodges EL, Yan H, Sonntag WE. Growth hormone, insulin-like growth factor-1 and the aging brain. Exp Gerontol. 2015;68:76–81. doi: 10.1016/j.exger.2014.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bailey-Downs LC, Mitschelen M, Sosnowska D, Toth P, Pinto JT, Ballabh P, Valcarcel-Ares MN, Farley J, Koller A, Henthorn JC, et al. Liver-specific knockdown of IGF-1 decreases vascular oxidative stress resistance by impairing the Nrf2-dependent antioxidant response: a novel model of vascular aging. J Gerontol A Biol Sci Med Sci. 2012;67:313–329. doi: 10.1093/gerona/glr164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bailyes EM, Nave BT, Soos MA, Orr SR, Hayward AC, Siddle K. Insulin receptor/IGF-I receptor hybrids are widely distributed in mammalian tissues: quantification of individual receptor species by selective immunoprecipitation and immunoblotting. The Biochemical journal. 1997;327(Pt 1):209–215. doi: 10.1042/bj3270209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bansal A, Zhu LJ, Yen K, Tissenbaum HA. Uncoupling lifespan and healthspan in Caenorhabditis elegans longevity mutants. Proc Natl Acad Sci U S A. 2015;112:E277–286. doi: 10.1073/pnas.1412192112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baxter RC. Insulin-like growth factor (IGF)-binding proteins: interactions with IGFs and intrinsic bioactivities. Am J Physiol Endocrinol Metab. 2000;278:E967–976. doi: 10.1152/ajpendo.2000.278.6.E967. [DOI] [PubMed] [Google Scholar]
- Bergman D, Halje M, Nordin M, Engstrom W. Insulin-like growth factor 2 in development and disease: a mini-review. Gerontology. 2013;59:240–249. doi: 10.1159/000343995. [DOI] [PubMed] [Google Scholar]
- Boguszewski CL, Ayuk J. Management of Endocrine Disease: Acromegaly and Cancer: An Old Debate Revisited. Eur J Endocrinol. 2016 doi: 10.1530/EJE-16-0178. [DOI] [PubMed] [Google Scholar]
- Bokov AF, Garg N, Ikeno Y, Thakur S, Musi N, DeFronzo RA, Zhang N, Erickson RC, Gelfond J, Hubbard GB, et al. Does reduced IGF-1R signaling in Igf1r+/− mice alter aging? PLoS One. 2011;6:e26891. doi: 10.1371/journal.pone.0026891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bollano E, Omerovic E, Bohlooly-y M, Kujacic V, Madhu B, Tornell J, Isaksson O, Soussi B, Schulze W, Fu ML, et al. Impairment of cardiac function and bioenergetics in adult transgenic mice overexpressing the bovine growth hormone gene. Endocrinology. 2000;141:2229–2235. doi: 10.1210/endo.141.6.7486. [DOI] [PubMed] [Google Scholar]
- Breese CR, Ingram RL, Sonntag WE. Influence of age and long-term dietary restriction on plasma insulin-like growth factor-1 (IGF-1), IGF-1 gene expression, and IGF-1 binding proteins. J Gerontol. 1991;46:B180–187. doi: 10.1093/geronj/46.5.b180. [DOI] [PubMed] [Google Scholar]
- 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]
- Burgers AM, Biermasz NR, Schoones JW, Pereira AM, Renehan AG, Zwahlen M, Egger M, Dekkers OM. Meta-analysis and dose-response metaregression: circulating insulin-like growth factor I (IGF-I) and mortality. J Clin Endocrinol Metab. 2011;96:2912–2920. doi: 10.1210/jc.2011-1377. [DOI] [PubMed] [Google Scholar]
- Byrne AB, Walradt T, Gardner KE, Hubbert A, Reinke V, Hammarlund M. Insulin/IGF1 signaling inhibits age-dependent axon regeneration. Neuron. 2014;81:561–573. doi: 10.1016/j.neuron.2013.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao H, Wang G, Meng L, Shen H, Feng Z, Liu Q, Du J. Association between circulating levels of IGF-1 and IGFBP-3 and lung cancer risk: a meta-analysis. PLoS One. 2012;7:e49884. doi: 10.1371/journal.pone.0049884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao Y, Nimptsch K, Shui IM, Platz EA, Wu K, Pollak MN, Kenfield SA, Stampfer MJ, Giovannucci EL. Prediagnostic plasma IGFBP-1, IGF-1 and risk of prostate cancer. Int J Cancer. 2015;136:2418–2426. doi: 10.1002/ijc.29295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ceda GP, Hoffman AR, Silverberg GD, Wilson DM, Rosenfeld RG. Regulation of growth hormone release from cultured human pituitary adenomas by somatomedins and insulin. J Clin Endocrinol Metab. 1985;60:1204–1209. doi: 10.1210/jcem-60-6-1204. [DOI] [PubMed] [Google Scholar]
- Chaker Z, Aid S, Berry H, Holzenberger M. Suppression of IGF-I signals in neural stem cells enhances neurogenesis and olfactory function during aging. Aging Cell. 2015;14:847–856. doi: 10.1111/acel.12365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cobb LJ, Mehta H, Cohen P. Enhancing the apoptotic potential of insulin-like growth factor-binding protein-3 in prostate cancer by modulation of CK2 phosphorylation. Molecular endocrinology. 2009;23:1624–1633. doi: 10.1210/me.2008-0365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Csiszar A, Labinskyy N, Perez V, Recchia FA, Podlutsky A, Mukhopadhyay P, Losonczy G, Pacher P, Austad SN, Bartke A, et al. Endothelial function and vascular oxidative stress in long-lived GH/IGF-deficient Ames dwarf mice. American journal of physiology Heart and circulatory physiology. 2008;295:H1882–1894. doi: 10.1152/ajpheart.412.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doi T, Shimada H, Makizako H, Tsutsumimoto K, Hotta R, Nakakubo S, Suzuki T. Association of insulin-like growth factor-1 with mild cognitive impairment and slow gait speed. Neurobiology of aging. 2015;36:942–947. doi: 10.1016/j.neurobiolaging.2014.10.035. [DOI] [PubMed] [Google Scholar]
- Egecioglu E, Andersson IJ, Bollano E, Palsdottir V, Gabrielsson BG, Kopchick JJ, Skott O, Bie P, Isgaard J, Bohlooly YM, et al. Growth hormone receptor deficiency in mice results in reduced systolic blood pressure and plasma renin, increased aortic eNOS expression, and altered cardiovascular structure and function. Am J Physiol Endocrinol Metab. 2007;292:E1418–1425. doi: 10.1152/ajpendo.00335.2006. [DOI] [PubMed] [Google Scholar]
- Fahrenholtz CD, Beltran PJ, Burnstein KL. Targeting IGF-IR with ganitumab inhibits tumorigenesis and increases durability of response to androgen-deprivation therapy in VCaP prostate cancer xenografts. Molecular cancer therapeutics. 2013;12:394–404. doi: 10.1158/1535-7163.MCT-12-0648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernandez AM, Torres-Aleman I. The many faces of insulin-like peptide signalling in the brain. Nature reviews Neuroscience. 2012;13:225–239. doi: 10.1038/nrn3209. [DOI] [PubMed] [Google Scholar]
- Fontana L, Villareal DT, Das SK, Smith SR, Meydani SN, Pittas AG, Klein S, Bhapkar M, Rochon J, Ravussin E, et al. Effects of 2-year calorie restriction on circulating levels of IGF-1, IGF-binding proteins and cortisol in nonobese men and women: a randomized clinical trial. Aging Cell. 2016;15:22–27. doi: 10.1111/acel.12400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friedlander AL, Butterfield GE, Moynihan S, Grillo J, Pollack M, Holloway L, Friedman L, Yesavage J, Matthias D, Lee S, et al. One year of insulin-like growth factor I treatment does not affect bone density, body composition, or psychological measures in postmenopausal women. J Clin Endocrinol Metab. 2001;86:1496–1503. doi: 10.1210/jcem.86.4.7377. [DOI] [PubMed] [Google Scholar]
- Friedrich N, Haring R, Nauck M, Ludemann J, Rosskopf D, Spilcke-Liss E, Felix SB, Dorr M, Brabant G, Volzke H, et al. Mortality and serum insulin-like growth factor (IGF)-I and IGF binding protein 3 concentrations. J Clin Endocrinol Metab. 2009;94:1732–1739. doi: 10.1210/jc.2008-2138. [DOI] [PubMed] [Google Scholar]
- Fuchs CS, Azevedo S, Okusaka T, Van Laethem JL, Lipton LR, Riess H, Szczylik C, Moore MJ, Peeters M, Bodoky G, et al. A phase 3 randomized, double-blind, placebo-controlled trial of ganitumab or placebo in combination with gemcitabine as first-line therapy for metastatic adenocarcinoma of the pancreas: the GAMMA trial. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO. 2015;26:921–927. doi: 10.1093/annonc/mdv027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galet C, Gray A, Said JW, Castor B, Wan J, Beltran PJ, Calzone FJ, Elashoff D, Cohen P, Aronson WJ. Effects of calorie restriction and IGF-1 receptor blockade on the progression of 22Rv1 prostate cancer xenografts. International journal of molecular sciences. 2013;14:13782–13795. doi: 10.3390/ijms140713782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gombar S, Jung HJ, Dong F, Calder B, Atzmon G, Barzilai N, Tian XL, Pothof J, Hoeijmakers JH, Campisi J, et al. Comprehensive microRNA profiling in B-cells of human centenarians by massively parallel sequencing. BMC Genomics. 2012;13:353. doi: 10.1186/1471-2164-13-353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong Z, Kennedy O, Sun H, Wu Y, Williams GA, Klein L, Cardoso L, Matheny RW, Jr, Hubbard GB, Ikeno Y, et al. Reductions in serum IGF-1 during aging impair health span. Aging Cell. 2014;13:408–418. doi: 10.1111/acel.12188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gontier G, George C, Chaker Z, Holzenberger M, Aid S. Blocking IGF Signaling in Adult Neurons Alleviates Alzheimer’s Disease Pathology through Amyloid-beta Clearance. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2015;35:11500–11513. doi: 10.1523/JNEUROSCI.0343-15.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green CJ, Holly JM, Bayer A, Fish M, Ebrahim S, Gallacher J, Ben-Shlomo Y. The role of IGF-I, IGF-II, and IGFBP-3 in male cognitive aging and dementia risk: the Caerphilly Prospective Study. Journal of Alzheimer’s disease : JAD. 2014;41:867–875. doi: 10.3233/JAD-132183. [DOI] [PubMed] [Google Scholar]
- Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, Wei M, Madia F, Cheng CW, Hwang D, Martin-Montalvo A, Saavedra J, Ingles S, et al. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med. 2011;3:70ra13. doi: 10.1126/scitranslmed.3001845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haluzik M, Yakar S, Gavrilova O, Setser J, Boisclair Y, LeRoith D. Insulin resistance in the liver-specific IGF-1 gene-deleted mouse is abrogated by deletion of the acid-labile subunit of the IGF-binding protein-3 complex: relative roles of growth hormone and IGF-1 in insulin resistance. Diabetes. 2003;52:2483–2489. doi: 10.2337/diabetes.52.10.2483. [DOI] [PubMed] [Google Scholar]
- Hawkins M, Barzilai N, Chen W, Angelov I, Hu M, Cohen P, Rossetti L. Increased hexosamine availability similarly impairs the action of insulin and IGF-1 on glucose disposal. Diabetes. 1996;45:1734–1743. doi: 10.2337/diab.45.12.1734. [DOI] [PubMed] [Google Scholar]
- He YH, Lu X, Yang LQ, Xu LY, Kong QP. Association of the insulin-like growth factor binding protein 3 (IGFBP-3) polymorphism with longevity in Chinese nonagenarians and centenarians. Aging (Albany NY) 2014;6:944–956. doi: 10.18632/aging.100703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Higashi Y, Sukhanov S, Anwar A, Shai SY, Delafontaine P. Aging, atherosclerosis, and IGF-1. J Gerontol A Biol Sci Med Sci. 2012;67:626–639. doi: 10.1093/gerona/gls102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holly JM, Perks CM. Insulin-like growth factor physiology: what we have learned from human studies. Endocrinol Metab Clin North Am. 2012;41:249–263. v. doi: 10.1016/j.ecl.2012.04.009. [DOI] [PubMed] [Google Scholar]
- Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC, Cervera P, Le Bouc 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]
- Huffman DM, Farias Quipildor G, Mao K, Zhang X, Wan J, Apontes P, Cohen P, Barzilai N. Central insulin-like growth factor-1 (IGF-1) restores whole-body insulin action in a model of age-related insulin resistance and IGF-1 decline. Aging Cell. 2016;15:181–186. doi: 10.1111/acel.12415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huffman DM, Johnson MS, Watts A, Elgavish A, Eltoum IA, Nagy TR. Cancer progression in the transgenic adenocarcinoma of mouse prostate mouse is related to energy balance, body mass, and body composition, but not food intake. Cancer research. 2007;67:417–424. doi: 10.1158/0008-5472.CAN-06-1244. [DOI] [PubMed] [Google Scholar]
- Hursting SD, Switzer BR, French JE, Kari FW. The growth hormone: insulin-like growth factor 1 axis is a mediator of diet restriction-induced inhibition of mononuclear cell leukemia in Fischer rats. Cancer research. 1993;53:2750–2757. [PubMed] [Google Scholar]
- 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;58:291–296. doi: 10.1093/gerona/58.4.b291. [DOI] [PubMed] [Google Scholar]
- Janssen JA, Burger H, Stolk RP, Grobbee DE, de Jong FH, Lamberts SW, Pols HA. Gender-specific relationship between serum free and total IGF-I and bone mineral density in elderly men and women. Eur J Endocrinol. 1998;138:627–632. doi: 10.1530/eje.0.1380627. [DOI] [PubMed] [Google Scholar]
- Jara A, Liu X, Sim D, Benner CM, Duran-Ortiz S, Qian Y, List EO, Berryman DE, Kim JK, Kopchick JJ. Cardiac-Specific Disruption of GH Receptor Alters Glucose Homeostasis While Maintaining Normal Cardiac Performance in Adult Male Mice. Endocrinology. 2016;157:1929–1941. doi: 10.1210/en.2015-1686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Juul A, Scheike T, Davidsen M, Gyllenborg J, Jorgensen T. Low serum insulin-like growth factor I is associated with increased risk of ischemic heart disease: a population-based case-control study. Circulation. 2002;106:939–944. doi: 10.1161/01.cir.0000027563.44593.cc. [DOI] [PubMed] [Google Scholar]
- Kaplan RC, Buzkova P, Cappola AR, Strickler HD, McGinn AP, Mercer LD, Arnold AM, Pollak MN, Newman AB. Decline in circulating insulin-like growth factors and mortality in older adults: cardiovascular health study all-stars study. J Clin Endocrinol Metab. 2012;97:1970–1976. doi: 10.1210/jc.2011-2967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaplan RC, McGinn AP, Pollak MN, Kuller LH, Strickler HD, Rohan TE, Cappola AR, Xue X, Psaty BM. Association of total insulin-like growth factor-I, insulin-like growth factor binding protein-1 (IGFBP-1), and IGFBP-3 levels with incident coronary events and ischemic stroke. J Clin Endocrinol Metab. 2007;92:1319–1325. doi: 10.1210/jc.2006-1631. [DOI] [PubMed] [Google Scholar]
- Kenyon C, Chang J, Gensch E, Rudner A, Tabtiang R. A C. elegans mutant that lives twice as long as wild type. Nature. 1993;366:461–464. doi: 10.1038/366461a0. [DOI] [PubMed] [Google Scholar]
- Key TJ, Appleby PN, Reeves GK, Roddam AW. Insulin-like growth factor 1 (IGF1), IGF binding protein 3 (IGFBP3), and breast cancer risk: pooled individual data analysis of 17 prospective studies. Lancet Oncol. 2010;11:530–542. doi: 10.1016/S1470-2045(10)70095-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim HS, Ali O, Shim M, Lee KW, Vuguin P, Muzumdar R, Barzilai N, Cohen P. Insulin-like growth factor binding protein-3 induces insulin resistance in adipocytes in vitro and in rats in vivo. Pediatric research. 2007;61:159–164. doi: 10.1203/pdr.0b013e31802d8a30. [DOI] [PubMed] [Google Scholar]
- Kranzler JH, Rosenbloom AL, Martinez V, Guevara-Aguirre J. Normal intelligence with severe insulin-like growth factor I deficiency due to growth hormone receptor deficiency: a controlled study in a genetically homogeneous population. J Clin Endocrinol Metab. 1998;83:1953–1958. doi: 10.1210/jcem.83.6.4863. [DOI] [PubMed] [Google Scholar]
- Langlois JA, Rosen CJ, Visser M, Hannan MT, Harris T, Wilson PW, Kiel DP. Association between insulin-like growth factor I and bone mineral density in older women and men: the Framingham Heart Study. J Clin Endocrinol Metab. 1998;83:4257–4262. doi: 10.1210/jcem.83.12.5308. [DOI] [PubMed] [Google Scholar]
- Laughlin GA, Barrett-Connor E, Criqui MH, Kritz-Silverstein D. The prospective association of serum insulin-like growth factor I (IGF-I) and IGF-binding protein-1 levels with all cause and cardiovascular disease mortality in older adults: the Rancho Bernardo Study. J Clin Endocrinol Metab. 2004;89:114–120. doi: 10.1210/jc.2003-030967. [DOI] [PubMed] [Google Scholar]
- Leung KC, Doyle N, Ballesteros M, Waters MJ, Ho KK. Insulin regulation of human hepatic growth hormone receptors: divergent effects on biosynthesis and surface translocation. J Clin Endocrinol Metab. 2000;85:4712–4720. doi: 10.1210/jcem.85.12.7017. [DOI] [PubMed] [Google Scholar]
- Levine ME, Suarez JA, Brandhorst S, Balasubramanian P, Cheng CW, Madia F, Fontana L, Mirisola MG, Guevara-Aguirre J, Wan J, et al. Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 2014;19:407–417. doi: 10.1016/j.cmet.2014.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Q, Ceylan-Isik AF, Li J, Ren J. Deficiency of insulin-like growth factor 1 reduces sensitivity to aging-associated cardiomyocyte dysfunction. Rejuvenation research. 2008;11:725–733. doi: 10.1089/rej.2008.0717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lichtenwalner RJ, Forbes ME, Bennett SA, Lynch CD, Sonntag WE, Riddle DR. Intracerebroventricular infusion of insulin-like growth factor-I ameliorates the age-related decline in hippocampal neurogenesis. Neuroscience. 2001;107:603–613. doi: 10.1016/s0306-4522(01)00378-5. [DOI] [PubMed] [Google Scholar]
- List EO, Berryman DE, Ikeno Y, Hubbard GB, Funk K, Comisford R, Young JA, Stout MB, Tchkonia T, Masternak MM, et al. Removal of growth hormone receptor (GHR) in muscle of male mice replicates some of the health benefits seen in global GHR−/− mice. Aging (Albany NY) 2015;7:500–512. doi: 10.18632/aging.100766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu W, Ye P, O’Kusky JR, D’Ercole AJ. Type 1 insulin-like growth factor receptor signaling is essential for the development of the hippocampal formation and dentate gyrus. Journal of neuroscience research. 2009;87:2821–2832. doi: 10.1002/jnr.22129. [DOI] [PubMed] [Google Scholar]
- Liu Z, Mohan S, Yakar S. Does the GH/IGF-1 axis contribute to skeletal sexual dimorphism? Evidence from mouse studies. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 2016;27:7–17. doi: 10.1016/j.ghir.2015.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lopez-Velasco R, Escobar-Morreale HF, Vega B, Villa E, Sancho JM, Moya-Mur JL, Garcia-Robles R. Cardiac involvement in acromegaly: specific myocardiopathy or consequence of systemic hypertension? J Clin Endocrinol Metab. 1997;82:1047–1053. doi: 10.1210/jcem.82.4.3876. [DOI] [PubMed] [Google Scholar]
- Maggio M, Lauretani F, Ceda GP, Bandinelli S, Ling SM, Metter EJ, Artoni A, Carassale L, Cazzato A, Ceresini G, et al. Relationship between low levels of anabolic hormones and 6-year mortality in older men: the aging in the Chianti Area (InCHIANTI) study. Arch Intern Med. 2007;167:2249–2254. doi: 10.1001/archinte.167.20.2249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milman S, Atzmon G, Huffman DM, Wan J, Crandall JP, Cohen P, Barzilai N. Low insulin-like growth factor-1 level predicts survival in humans with exceptional longevity. Aging Cell. 2014 doi: 10.1111/acel.12213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moellendorf S, Kessels C, Peiseler L, Raupach A, Jacoby C, Vogt N, Lindecke A, Koch L, Bruning J, Heger J, et al. IGF-IR signaling attenuates the age-related decline of diastolic cardiac function. Am J Physiol Endocrinol Metab. 2012;303:E213–222. doi: 10.1152/ajpendo.00538.2011. [DOI] [PubMed] [Google Scholar]
- Muller AP, Fernandez AM, Haas C, Zimmer E, Portela LV, Torres-Aleman I. Reduced brain insulin-like growth factor I function during aging. Molecular and cellular neurosciences. 2012;49:9–12. doi: 10.1016/j.mcn.2011.07.008. [DOI] [PubMed] [Google Scholar]
- Muzumdar RH, Ma X, Fishman S, Yang X, Atzmon G, Vuguin P, Einstein FH, Hwang D, Cohen P, Barzilai N. Central and opposing effects of IGF-I and IGF-binding protein-3 on systemic insulin action. Diabetes. 2006;55:2788–2796. doi: 10.2337/db06-0318. [DOI] [PubMed] [Google Scholar]
- Novosyadlyy R, Leroith D. Insulin-like growth factors and insulin: at the crossroad between tumor development and longevity. J Gerontol A Biol Sci Med Sci. 2012;67:640–651. doi: 10.1093/gerona/gls065. [DOI] [PubMed] [Google Scholar]
- Ohlsson C, Mellstrom D, Carlzon D, Orwoll E, Ljunggren O, Karlsson MK, Vandenput L. Older men with low serum IGF-1 have an increased risk of incident fractures: the MrOS Sweden study. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2011;26:865–872. doi: 10.1002/jbmr.281. [DOI] [PubMed] [Google Scholar]
- Okereke O, Kang JH, Ma J, Hankinson SE, Pollak MN, Grodstein F. Plasma IGF-I levels and cognitive performance in older women. Neurobiology of aging. 2007;28:135–142. doi: 10.1016/j.neurobiolaging.2005.10.012. [DOI] [PubMed] [Google Scholar]
- Pawlikowska L, Hu D, Huntsman S, Sung A, Chu C, Chen J, Joyner AH, Schork NJ, Hsueh WC, Reiner AP, et al. Association of common genetic variation in the insulin/IGF1 signaling pathway with human longevity. Aging Cell. 2009;8:460–472. doi: 10.1111/j.1474-9726.2009.00493.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Posaci C, Altunyurt S, Islekel H, Onvural A. Effects of HRT on serum levels of IGF-I in postmenopausal women. Maturitas. 2001;40:69–74. doi: 10.1016/s0378-5122(01)00230-4. [DOI] [PubMed] [Google Scholar]
- Rajaram S, Baylink DJ, Mohan S. Insulin-like growth factor-binding proteins in serum and other biological fluids: regulation and functions. Endocr Rev. 1997;18:801–831. doi: 10.1210/edrv.18.6.0321. [DOI] [PubMed] [Google Scholar]
- Rajpathak SN, He M, Sun Q, Kaplan RC, Muzumdar R, Rohan TE, Gunter MJ, Pollak M, Kim M, Pessin JE, et al. Insulin-like growth factor axis and risk of type 2 diabetes in women. Diabetes. 2012;61:2248–2254. doi: 10.2337/db11-1488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reddy BS, Wang CX, Maruyama H. Effect of restricted caloric intake on azoxymethane-induced colon tumor incidence in male F344 rats. Cancer research. 1987;47:1226–1228. [PubMed] [Google Scholar]
- Renehan AG, Zwahlen M, Minder C, O’Dwyer ST, Shalet SM, Egger M. Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: systematic review and meta-regression analysis. Lancet. 2004;363:1346–1353. doi: 10.1016/S0140-6736(04)16044-3. [DOI] [PubMed] [Google Scholar]
- Ricketts SL, Rensing KL, Holly JM, Chen L, Young EH, Luben R, Ashford S, Song K, Yuan X, Dehghan A, et al. Prospective study of insulin-like growth factor-I, insulin-like growth factor-binding protein 3, genetic variants in the IGF1 and IGFBP3 genes and risk of coronary artery disease. International journal of molecular epidemiology and genetics. 2011;2:261–285. [PMC free article] [PubMed] [Google Scholar]
- Rinaldi S, Cleveland R, Norat T, Biessy C, Rohrmann S, Linseisen J, Boeing H, Pischon T, Panico S, Agnoli C, et al. Serum levels of IGF-I, IGFBP-3 and colorectal cancer risk: results from the EPIC cohort, plus a meta-analysis of prospective studies. Int J Cancer. 2010;126:1702–1715. doi: 10.1002/ijc.24927. [DOI] [PubMed] [Google Scholar]
- Robertson JF, Ferrero JM, Bourgeois H, Kennecke H, de Boer RH, Jacot W, McGreivy J, Suzuki S, Zhu M, McCaffery I, et al. Ganitumab with either exemestane or fulvestrant for postmenopausal women with advanced, hormone-receptor-positive breast cancer: a randomised, controlled, double-blind, phase 2 trial. Lancet Oncol. 2013;14:228–235. doi: 10.1016/S1470-2045(13)70026-3. [DOI] [PubMed] [Google Scholar]
- Roubenoff R, Parise H, Payette HA, Abad LW, D’Agostino R, Jacques PF, Wilson PW, Dinarello CA, Harris TB. Cytokines, insulin-like growth factor 1, sarcopenia, and mortality in very old community-dwelling men and women: the Framingham Heart Study. Am J Med. 2003;115:429–435. doi: 10.1016/j.amjmed.2003.05.001. [DOI] [PubMed] [Google Scholar]
- Rowlands MA, Gunnell D, Harris R, Vatten LJ, Holly JM, Martin RM. Circulating insulin-like growth factor peptides and prostate cancer risk: a systematic review and meta-analysis. Int J Cancer. 2009;124:2416–2429. doi: 10.1002/ijc.24202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rudman D, Kutner MH, Rogers CM, Lubin MF, Fleming GA, Bain RP. Impaired growth hormone secretion in the adult population: relation to age and adiposity. J Clin Invest. 1981;67:1361–1369. doi: 10.1172/JCI110164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Russell SJ, Kahn CR. Endocrine regulation of ageing. Nature reviews Molecular cell biology. 2007;8:681–691. doi: 10.1038/nrm2234. [DOI] [PubMed] [Google Scholar]
- Sandhu MS, Heald AH, Gibson JM, Cruickshank JK, Dunger DB, Wareham NJ. Circulating concentrations of insulin-like growth factor-I and development of glucose intolerance: a prospective observational study. Lancet. 2002;359:1740–1745. doi: 10.1016/S0140-6736(02)08655-5. [DOI] [PubMed] [Google Scholar]
- Saydah S, Graubard B, Ballard-Barbash R, Berrigan D. Insulin-like growth factors and subsequent risk of mortality in the United States. Am J Epidemiol. 2007;166:518–526. doi: 10.1093/aje/kwm124. [DOI] [PubMed] [Google Scholar]
- 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]
- Steuerman R, Shevah O, Laron Z. Congenital IGF1 deficiency tends to confer protection against post-natal development of malignancies. Eur J Endocrinol. 2011;164:485–489. doi: 10.1530/EJE-10-0859. [DOI] [PubMed] [Google Scholar]
- Stewart CE, Rotwein P. Growth, differentiation, and survival: multiple physiological functions for insulin-like growth factors. Physiol Rev. 1996;76:1005–1026. doi: 10.1152/physrev.1996.76.4.1005. [DOI] [PubMed] [Google Scholar]
- Suh Y, Atzmon G, Cho MO, Hwang D, Liu B, Leahy DJ, Barzilai N, Cohen P. Functionally significant insulin-like growth factor I receptor mutations in centenarians. Proc Natl Acad Sci U S A. 2008;105:3438–3442. doi: 10.1073/pnas.0705467105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun LY, Al-Regaiey K, Masternak MM, Wang J, Bartke A. Local expression of GH and IGF-1 in the hippocampus of GH-deficient long-lived mice. Neurobiology of aging. 2005;26:929–937. doi: 10.1016/j.neurobiolaging.2004.07.010. [DOI] [PubMed] [Google Scholar]
- Sustarsic EG, Junnila RK, Kopchick JJ. Human metastatic melanoma cell lines express high levels of growth hormone receptor and respond to GH treatment. Biochemical and biophysical research communications. 2013;441:144–150. doi: 10.1016/j.bbrc.2013.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Svensson J, Carlzon D, Petzold M, Karlsson MK, Ljunggren O, Tivesten A, Mellstrom D, Ohlsson C. Both low and high serum IGF-I levels associate with cancer mortality in older men. J Clin Endocrinol Metab. 2012;97:4623–4630. doi: 10.1210/jc.2012-2329. [DOI] [PubMed] [Google Scholar]
- Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pathways: insights into insulin action. Nature reviews Molecular cell biology. 2006;7:85–96. doi: 10.1038/nrm1837. [DOI] [PubMed] [Google Scholar]
- Tazearslan C, Huang J, Barzilai N, Suh Y. Impaired IGF1R signaling in cells expressing longevity-associated human IGF1R alleles. Aging Cell. 2011;10:551–554. doi: 10.1111/j.1474-9726.2011.00697.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toth P, Tarantini S, Ashpole NM, Tucsek Z, Milne GL, Valcarcel-Ares NM, Menyhart A, Farkas E, Sonntag WE, Csiszar A, et al. IGF-1 deficiency impairs neurovascular coupling in mice: implications for cerebromicrovascular aging. Aging Cell. 2015;14:1034–1044. doi: 10.1111/acel.12372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tumati S, Burger H, Martens S, van der Schouw YT, Aleman A. Association between Cognition and Serum Insulin-Like Growth Factor-1 in Middle-Aged & Older Men: An 8 Year Follow-Up Study. PLoS One. 2016;11:e0154450. doi: 10.1371/journal.pone.0154450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ungvari Z, Csiszar A. The emerging role of IGF-1 deficiency in cardiovascular aging: recent advances. J Gerontol A Biol Sci Med Sci. 2012;67:599–610. doi: 10.1093/gerona/gls072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Bunderen CC, van Nieuwpoort IC, van Schoor NM, Deeg DJ, Lips P, Drent ML. The association of serum insulin-like growth factor-I with mortality, cardiovascular disease, and cancer in the elderly: a population-based study. J Clin Endocrinol Metab. 2010;95:4616–4624. doi: 10.1210/jc.2010-0940. [DOI] [PubMed] [Google Scholar]
- van der Spoel E, Rozing MP, Houwing-Duistermaat JJ, Slagboom PE, Beekman M, de Craen AJ, Westendorp RG, van Heemst D. Association analysis of insulin-like growth factor-1 axis parameters with survival and functional status in nonagenarians of the Leiden Longevity Study. Aging (Albany NY) 2015;7:956–963. doi: 10.18632/aging.100841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Heemst D, Beekman M, Mooijaart SP, Heijmans BT, Brandt BW, Zwaan BJ, Slagboom PE, Westendorp RG. Reduced insulin/IGF-1 signalling and human longevity. Aging Cell. 2005;4:79–85. doi: 10.1111/j.1474-9728.2005.00148.x. [DOI] [PubMed] [Google Scholar]
- van Varsseveld NC, Sohl E, Drent ML, Lips P. Gender-Specific Associations of Serum Insulin-Like Growth Factor-1 With Bone Health and Fractures in Older Persons. J Clin Endocrinol Metab. 2015;100:4272–4281. doi: 10.1210/jc.2015-2549. [DOI] [PubMed] [Google Scholar]
- Vasan RS, Sullivan LM, D’Agostino RB, Roubenoff R, Harris T, Sawyer DB, Levy D, Wilson PW. Serum insulin-like growth factor I and risk for heart failure in elderly individuals without a previous myocardial infarction: the Framingham Heart Study. Ann Intern Med. 2003;139:642–648. doi: 10.7326/0003-4819-139-8-200310210-00007. [DOI] [PubMed] [Google Scholar]
- Wang M, Miller RA. Fibroblasts from long-lived mutant mice exhibit increased autophagy and lower TOR activity after nutrient deprivation or oxidative stress. Aging Cell. 2012;11:668–674. doi: 10.1111/j.1474-9726.2012.00833.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y, Sakata T, Elalieh HZ, Munson SJ, Burghardt A, Majumdar S, Halloran BP, Bikle DD. Gender differences in the response of CD-1 mouse bone to parathyroid hormone: potential role of IGF-I. The Journal of endocrinology. 2006;189:279–287. doi: 10.1677/joe.1.06351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Westwood AJ, Beiser A, Decarli C, Harris TB, Chen TC, He XM, Roubenoff R, Pikula A, Au R, Braverman LE, et al. Insulin-like growth factor-1 and risk of Alzheimer dementia and brain atrophy. Neurology. 2014;82:1613–1619. doi: 10.1212/WNL.0000000000000382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woods KA, Camacho-Hubner C, Savage MO, Clark AJ. Intrauterine growth retardation and postnatal growth failure associated with deletion of the insulin-like growth factor I gene. N Engl J Med. 1996;335:1363–1367. doi: 10.1056/NEJM199610313351805. [DOI] [PubMed] [Google Scholar]
- Wu Y, Sun H, Basta-Pljakic J, Cardoso L, Kennedy OD, Jasper H, Domene H, Karabatas L, Guida C, Schaffler MB, et al. Serum IGF-1 is insufficient to restore skeletal size in the total absence of the growth hormone receptor. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2013;28:1575–1586. doi: 10.1002/jbmr.1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu J, Gontier G, Chaker Z, Lacube P, Dupont J, Holzenberger M. Longevity effect of IGF-1R(+/−) mutation depends on genetic background-specific receptor activation. Aging Cell. 2014;13:19–28. doi: 10.1111/acel.12145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamaguchi T, Kanatani M, Yamauchi M, Kaji H, Sugishita T, Baylink DJ, Mohan S, Chihara K, Sugimoto T. Serum levels of insulin-like growth factor (IGF); IGF-binding proteins-3, -4, and -5; and their relationships to bone mineral density and the risk of vertebral fractures in postmenopausal women. Calcif Tissue Int. 2006;78:18–24. doi: 10.1007/s00223-005-0163-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamamoto H, Sohmiya M, Oka N, Kato Y. Effects of aging and sex on plasma insulin-like growth factor I (IGF-I) levels in normal adults. Acta Endocrinol (Copenh) 1991;124:497–500. doi: 10.1530/acta.0.1240497. [DOI] [PubMed] [Google Scholar]
- Yuldasheva NY, Rashid ST, Haywood NJ, Cordell P, Mughal R, Viswambharan H, Imrie H, Sukumar P, Cubbon RM, Aziz A, et al. Haploinsufficiency of the insulinlike growth factor-1 receptor enhances endothelial repair and favorably modifies angiogenic progenitor cell phenotype. Arterioscler Thromb Vasc Biol. 2014;34:2051–2058. doi: 10.1161/ATVBAHA.114.304121. [DOI] [PubMed] [Google Scholar]