• No results found

University of Groningen Manipulating age-related metabolic flexibility Dommerholt, Marleen

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Manipulating age-related metabolic flexibility Dommerholt, Marleen"

Copied!
19
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Manipulating age-related metabolic flexibility

Dommerholt, Marleen

DOI:

10.33612/diss.172053834

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2021

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Dommerholt, M. (2021). Manipulating age-related metabolic flexibility: using pharmacological and dietary interventions. University of Groningen. https://doi.org/10.33612/diss.172053834

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter

General discussion

(3)

While the worldwide average lifespan is increasing, the incidence of frailty and the development of related chronic diseases is elevated. The accumulation of age-related damage in cells affects metabolic flexibility leading to changes in whole-body energy metabolism. Hyperinsulinemia, increased adiposity, and muscle loss are some of the age-related changes contributing to a reduction in the quality of life. Poor metabolic health, caused by bad lifestyle habits such as an unhealthy diet and physical inactivity, also contributes to the progression of chronic metabolic diseases, meaning that the extended lifespan is rarely disease-free. In this thesis, we investigated the potential to increase metabolic health via modulation of age-related changes in metabolic flexibility using genetic-, pharmacological- and dietary interventions.

Metabolic flexibility as a mechanism to adapt to dietary changes at

an advanced age

In Chapter 1, we describe the metabolic alterations associated with ageing, such as white adipose tissue (WAT) redistribution, ectopic lipid accumulation, hyperinsulinemia, and muscle deterioration, that affect metabolic flexibility. While the exact mechanisms contributing to these metabolic alterations remain incompletely understood, this thesis aimed to characterized metabolic flexibility in aged mice. We investigated whether dietary interventions, such as caloric restriction (CR), protein restriction (PR), or high-fat diet (HFD), has a different effect on metabolic health in aged mice as compared to young animals.

First, it has been long known that insulin sensitivity is decreased during ageing, leading to compensatory hyperinsulinemia to maintain normal glycemia [1,2]. In Chapters 4&5, we confirmed the age-related hyperinsulinemia, but surprisingly discovered an improved glucose tolerance compared to young animals. Similarly, an HFD aggravates age-related hyperinsulinemia, as aged animals had to increase insulin levels up to 3.5 times higher during glucose administration compared to young mice, but it did not yet deteriorate glucose homeostasis (Chapter 4). Next, we investigated whether age-related hyperinsulinemia could be affected by PR, but in contrast to our hypothesis, we found no effects (Chapter 5). Glucose homeostasis and subsequent insulin secretion were not improved by a 3-month intervention of either low- or high protein intake. Interestingly, mice with a genetically reduced insulin dosage, that are protected from diet-induced hyperinsulinemia [3], still developed an age-related increase in insulin secretion (Chapter 6). While we describe an improvement of glucose metabolism by caloric restriction at all ages tested, these results appear to be independent of a reduction in fasting insulin levels. Together these results suggest that age-related hyperinsulinemia develops independent of insulin dosage, cannot be regulated by dietary interventions, but can deteriorate fast by an unhealthy diet.

Second, age-associated muscle decline is an important factor to reduce the quality of life in the elderly. It has been suggested that aged muscles require high dietary protein intake to maintain protein synthesis. While basal protein synthesis is maintained during ageing, sensitivity to detect amino acids declines and muscle protein synthesis after protein ingestion is reduced in aged individuals due to anabolic resistance involving insulin and mTOR signalling [4,5]. We determined an age-related decline in muscle

(4)

7

strength but this was not affected by dietary protein intake (Chapter 5). At 18 months

of age, a low protein diet was not detrimental to muscle strength and mitochondrial function, and a high protein diet did not prevent age-related decline. Previously, it has been shown that the decline of mitochondrial respiratory capacity can be prevented until 18 months of age by voluntary running, independent of the administered low- or high-fat diet [6]. Furthermore, muscle mitochondrial function of 18 months animals was not yet affected compared to young animals but decreased rapidly in response to 3 months HFD. Aged animals were incapable of increasing oxidative capacity in the muscle in response to nutrient excess, leading to lipid accumulation (Chapter 4). Muscle strength after high-fat feeding, measured by hanging wire or grip strength also deteriorated, but this could also be a secondary effect of increased weight gain. The insulin sensitivity, to which skeletal muscle is the greatest contributor, was already strongly reduced in aged animals compared to young and was further decreased upon a high-fat diet. Together, these results indicate that more research is necessary to find mechanisms to improve muscle function at an advanced age.

Third, age-related adipose tissue redistribution towards visceral WAT (vWAT), ectopic lipid accumulation and increased accumulation of senescent cells affect metabolic health at an advanced age [7]. Changes in sex hormones and subcutaneous WAT (scWAT) dysfunction have been shown to contribute to the relocation of lipids to vWAT, liver and muscle [8,9]. Ageing is associated with alterations in the immune cell environment of vWAT, subsequently causing adipose tissue dysfunction, insulin resistance, and ectopic lipid storage [10,11]. In Chapter 4, we observed that the aged animals have limited abilities to compensate when challenged with an HFD, leading to ectopic lipid accumulation in the muscle. We found that the expansion of scWAT was similar between young and aged animals, but the vWAT expanded only by 75% in aged animals while it was 3-fold in young animals. Based on these observations, we hypothesize that limited adipose tissue expansion due to age-related vWAT dysfunction resulted in ectopic lipid accumulation in the muscle after 3 months HFD. Furthermore, we indicate that the aged adipose tissue is still sensitive to changes in dietary protein intake and subsequent fibroblast growth factor 21 (FGF21) signalling, indicating metabolic flexibility. In Chapter 5, a low protein diet increased energy expenditure and stimulated the rejuvenation of scWAT by increasing the beige adipocyte population. Together, this thesis shows that a healthy dietary lifestyle can induce rejuvenation of adipose tissue, indicating the presence of metabolic flexibility at an advanced age.

Altogether, we conclude that healthy dietary interventions can improve metabolic health at an advanced age by stimulating metabolic flexibility, but that an unhealthy lifestyle is highly detrimental due to the loss of compensatory mechanisms. Together these results suggest that dietary composition is important during ageing. Whether these dietary interventions at advanced age also affect longevity remains to be investigated. The study of Hahn et al. demonstrated that a nutritional memory of aged adipose tissue prevented 21-months old mice from CR-induced longevity and metabolic remodelling of adipose tissue [12]. While differences in the experimental design, such as age at the start of the experiment, duration of the diet, or gender could explain these results, it remains unknown whether the improvements in longevity and

(5)

metabolic health are connected. The results in this thesis further suggest that diet composition is an important factor for the effectiveness of late-life dietary intervention strategies and age-specific recommendations might be necessary.

The complexity of IGF1 and FGF21 signalling in ageing and longevity

As discussed in Chapter 1, reduced growth hormone (GH) / insulin-like growth factor 1 (IGF1) / insulin signalling is an essential factor in longevity and healthy ageing. It has been suggested that dietary interventions promote metabolic health by affecting these metabolic targets, however, the role of these hormones in physiological ageing is paradoxical. While reduced IGF1-insulin signalling is an important aspect of increased longevity by caloric restriction, models of premature ageing also exhibited reduced IGF1 [13]. Analysis of significantly over-represented biological processes in both delayed and premature aging revealed highly similar profiles. Reduced expression of the GH-IGF1 pathway is found in natural ageing, accelerated ageing, and long-lived mouse models [13]. In an attempt to extend lifespan, the IGF1-insulin axis was reduced in order to limit cell growth [14]. However, in aged mice, this results in rapid and progressive decline in organ function, as dysfunctional or dying cells cannot be replaced [15]. Also in humans, secretion of IGF1 declines continuously after reaching adulthood, resulting in very low levels in aged individuals [16,17]. The total serum concentration of IGF1 in humans reaches a peak in young adults and then progressively declines, reaching a relatively steady level at the age of 75 years [18,19]. In humans, lower IGF1 concentrations have been associated with the development of age-related cognitive impairment and higher mortality [20], including all-cause mortality, cancer, cardiovascular diseases, chronic heart failure, and ischemic heart diseases [21–24]. Low protein intake has been associated with reduced IGF1 levels and improved disease incidence in the population below 65 years of age, but not in the older population [25]. However, the IGF1 axis is also associated with increased survival and better functional status in nonagenarians [17]. Low IGF1 predicts life expectancy in exceptionally long-lived individuals and even increased survival in elderly (95+) with a history of cancer [16]. These contradicting results suggest a complex role of IGF1 signalling during ageing which in healthy individuals can improve life, while others will be detrimentally affected by reduced IGF1 levels. It should therefore be further investigated how reduced IGF1 signalling can have divergent effects in elderly, and whether this will affect the efficacy of the response of dietary interventions.

Similarly, FGF21 has a paradoxical role in metabolic health and disease. FGF21 is produced by the liver and adipose tissues in response to prolonged fasting, thereby suppressing lipogenesis and stimulating hepatic fatty acid oxidation and ketogenesis [26,27]. FGF21 administration improves insulin sensitivity, hepatic triglyceride accumulation, and energy expenditure [28,29]. However, elevated FGF21 levels have also been ascribed during obesity, metabolic diseases and ageing, see Table 1 [30–42]. Plasma FGF21 levels have been reported to be significantly associated with body mass index, plasma triglyceride levels and insulin resistance [34,36]. In addition, the expression of βKlotho is downregulated in several pathophysiological conditions that display high FGF21 levels, such as in adipose tissue after an HFD,

(6)

7

liver during non-alcoholic fatty liver disease, and pancreatic islets of diabetic ob/ob

mice [43]. Due to elevated FGF21 production by both liver and WAT, obesity has been classified as an FGF21-resistant state, but it remains unclear whether elevated FGF21 levels have functionality in obese individuals [44]. Classification of FGF21 resistance should therefore include the phosphorylation and expression of downstream signalling proteins (ERK, AMPK, Akt, Egr1, or cFos), as well as the physiological response to exogenous FGF21 both in vivo and in vitro [45]. However, studies investigating the state of ‘’FGF21 resistance’’ during obesity have found inconclusive results. While exogenous FGF21 administration in obese mice resulted in a strong reduction in the sensitivity of WAT and liver, measured by ERK phosphorylation and target gene mRNA expression (Egr1/cFos), the physiological responses to exogenous FGF21 like glucose-lowering and the reduction of body weight remain preserved in obese mice [46,47]. To the same extent, specific age-related changes in FGF21 signalling need to be better understood. Increased levels of FGF21 have been determined linearly across the age groups (from 20 to 80 years) in a healthy population [42]. However, centenarians display an interesting low level of FGF21 levels compared to elderly controls [33], and adipose explants from elderly humans respond similarly to FGF21 administration [32]. These results are in line with our findings in Chapter 5, in which aged animals were responsive to PR-induced elevation of plasma FGF21 and aged adipocytes could be stimulated to increase thermogenesis, indicating that ageing itself does not induce FGF21 resistance. However, the physiological relevance of elevated FGF21 levels remains elusive and it needs to be further studied whether chronically high FGF21 levels can interfere with the FGF21-induced effects of dietary interventions.

Table 1: FGF21 levels are associated with obesity, ageing, and metabolic diseases in humans. OBESITY Increased levels of FGF21 [30,31,34,35]

T2D Inconclusively increased by glucose intolerance [30,31,35–39] Inconclusively increased in T2D patients

Drug treatment of T2D patient does not affect FGF21 levels

T1D Decreased levels of FGF21 [39]

NAFLD/NASH Increased levels of FGF21, [41,48] most pronounced in NAFLD, not NASH

HYPERTENSION Increased levels of FGF21 [30]

AGEING Increasing levels of FGF21 over time [32,33,42] Reduced levels of FGF21 in centenarians

T2D; Type 2 Diabetes, T1D; Type 1 Diabetes, NAFLD; nonalcoholic fatty liver disease, NASH; nonalcoholic steatohepatitis.

The effect of weight loss on metabolic health

Changes in body weight and body composition are important aspects to take into account when using dietary interventions as a tool to study metabolic health. In this thesis, we investigated the effects of dietary interventions in aged mice, in which body weight loss, and especially lean mass deterioration, is not necessarily beneficial. It is therefore important to establish whether improved health by a dietary intervention depends on body weight. CR primarily interferes with the state of energy imbalance

(7)

by reducing energy intake and subsequently reducing body weight to juvenile levels. Normally, C57Bl/6 mice rapidly gain body weight during development by the expansion of lean mass, and body weight continues to increase after adolescence as a result of general overnutrition and low energy expenditure, resulting in overweight adult and aged mice [49]. However, since the effectiveness of CR varies significantly between different strains [50], it suggests that the effectiveness of CR correlates with the induced weight loss. For example, DBA/2 mice, which have a reduced weight gain over time due to a high rate of energy expenditure and therefore exhibit limited weight loss by CR, also appear to be unresponsive to the effects of CR on longevity [51–54]. This hypothesis is also in line with our findings in Chapter 6, in which we investigated the role of age in the effectiveness of CR. CR reduced body weight to a juvenile level, independent of age and its associated body weight. Our results indicate that there was a strong correlation between the reduction in body weight and the improvements in metabolic health at different ages. In addition, both short-term and a lifelong CR diet affected both body mass and metabolic health regardless of insulin gene dosage [55], suggesting that the duration of the diet and fasting insulin levels have limited effects on the metabolic improvements induced by CR. These effects have also been described in non-obese humans, in which long-term CR does not have a greater effect on metabolic health compared to short-term studies. In those studies, cardiovascular risk factors, adipokine secretion, and bone mineral density are not further improved after 12 months CR [56–59]. The weight loss (more specifically fat mass) declines between 12 and 24 months of CR, possibly due to a decline in the adherence to a CR diet. Together, these results suggest that changes in body weight and composition are the main drivers of metabolic changes by caloric restriction.

Body composition is another important factor when determining the efficiency of dietary interventions in the elderly. Especially at an advanced age, when immobility is complicated by medical or environmental factors, lean mass deterioration increases the risk of metabolic decline. In contrast, available research on protein restriction in young mice reported increased longevity and improved metabolic health, such as energy expenditure and glucose tolerance, together with a reduction in body weight and lean mass. A low protein diet of 5% or 7% induced a significant reduction in body weight and lean mass after 2 weeks [60,61]. Whether the metabolic effects of PR depend on changes in body weight or lean mass has not been investigated. However, the use of Fgf21 KO mice showed that PR-induced effects on energy expenditure and thermogenesis are FGF21 dependent. Furthermore, these mice also exhibit a different response in body weight and composition, in which the effects on body weight and lean mass are less severe compared to wild type mice [60,62–64]. Together, these studies suggest a synergy between the effects on body composition and energy expenditure. In contrast to these observations, we found no effect of protein restriction in aged mice on body weight or lean mass (Chapter 5). Since we observed an increase in energy expenditure and browning, this suggested that these effects are independent of body weight. As we didn’t observe an improvement in glucose homeostasis, we hypothesize that body composition is important for PR-induced glucose tolerance and insulin sensitivity. Together, these studies indicate that PR can improve metabolic

(8)

7

health independent of weight loss and suggest distinct mechanisms between insulin

resistance and energy expenditure. While further studies to determine this distinction are necessary, these observations indicate important differences between CR and PR in the treatment of obesity and its comorbidities.

Gender differences in WAT flexibility and effects on metabolic health

Metabolic research has traditionally been focused on using males. A smaller variability, due to the lack of hormonal fluctuations during the female reproductive cycle, has frequently been used as justification [65]. However, many gender-related differences can complicate the extrapolation of male-derived results of dietary restriction. Most appreciated of the gender differences are inherent differences in fat distribution and function. While women have a higher percentage of fat mass, which is mainly located subcutaneously (specifically around the hips and thighs), men have higher fat storage in visceral adipose tissue (belly) [65]. This distinction is important as the site of adipose tissue expansion in obesity is a major determinant of cardiometabolic disease risk. The high visceral adiposity in men is associated with an increased incidence of diabetes and atherosclerosis, while high subcutaneous adiposity, is associated with a reduced incidence of metabolic disease [66]. Subsequently, insulin resistance occurs more frequently in males than females in both mice and humans [65,67]. As a result of gender-specific distribution and function of adipose tissue, the progression of obesity occurs differently in males and females. Female mice gain weight at a slower pace, store lipids in scWAT rather than vWAT, and expand adipose tissue through hyperplasia rather than hypertrophy [68–71]. Altogether, gender differences contribute substantially to diet-induced obesity. In Chapter 4, we describe that the development of obesity in aged male mice is associated with impaired insulin sensitivity, ectopic lipid accumulation in the muscle, and the inability to induce beta-oxidation. However, as the adipose tissue expansion is different between males and females, it remains unclear whether aged female mice respond similarly. Distinct scWAT function could contribute to differences in insulin resistance and ectopic lipid accumulation in the muscle. Additional research is needed to determine whether a high-fat diet impacts muscle oxidation and lipid accumulation in aged female mice, independent of adipose tissue function.

Furthermore, the more obesogenic scWAT of aged females could be less sensitive to cues that promote browning of scWAT by protein restriction (Chapter 5). Unfortunately, only a few studies have investigated the effects induced by PR or methionine restriction (MR) in females, and report inconclusive findings between males and females on lean mass deterioration, adiposity, glucose tolerance, insulin sensitivity, and energy expenditure [72–75]. Gender-specific mechanisms to increase energy expenditure have been suggested as plasma FGF21 and uncoupling protein 1 (Ucp1) in scWAT are not induced by MR in females and the PR-induced magnitude of this response was diminished compared to males [72,73]. These results suggest that energy expenditure can be induced independently of the FGF21-UCP1 axis in females [73]. Furthermore, MR produced sexually dimorphic changes in body composition in young growing animals, while the effects on energy balance were comparable between sexes when the diet was initiated after reaching physical maturity [75]. Together these results

(9)

indicate the importance of the timing of dietary interventions to determine its effects in males and females. These findings are in line with our observations in Chapter 5, which highlight the importance of the timing of initiating a dietary intervention. We hypothesized that early intervention with a low protein diet affects the growth and body composition of mice, while these effects are absent when starting the diet after maturation. Together, these results indicate the importance of the life stage when applying the dietary intervention, and suggest important gender-specific differences.

Gender differences in IGF1-insulin signalling and effects on longevity

A gender-specific response to reduced IGF1-insulin signalling could also greatly contribute to differences induced by caloric restriction. It has been observed that extended longevity as a result of reduced nutrient-sensing is gender-biased [76,77]. While a range of pharmacological treatments to extend longevity is favoured in males, extended longevity by genetic models with IGF1-insulin deficiency is more pronounced in females, see Table 2 [78–100]. Only in

female and male Ames Dwarf and Snell Dwarf mice lifespan is affected equally. In line with these results, the mouse model used in Chapter 6 is also potentially affected by gender differences. Female Ins2+/- mice (on an Ins1 null background) were protected against high-fat diet-induced obesity, and extended longevity both on chow and HFD [83,101], while male Ins2+/- mice only showed highly variable results when challenged with an HFD [102]. In contrast, male Ins1+/- mice (on an Ins2 null background) were protected against obesity as a result of increased energy expenditure by immediate upregulation of mitochondrial oxidation [3,103], but no changes in metabolic health were observed in either chow-fed, high-fat diet-fed, or caloric restricted female Ins2-/- mice [3,55]. The

underlying mechanisms concerning these difference could be due to study design, timing or housing conditions, however, the impact of both insulin alleles in the regulation of insulin secretion between genders have not been systemically addressed.

Furthermore, differences in average lifespan exist between males and females, with women having a greater life expectancy at birth than men [76]. However, gender differences in animal models regarding

Table 2: Some models with reduced

GH-IGF1-insulin signalling show major gender-specific effects on longevity.

Males Females Ref. Snell dwarf ↑↑ ↑↑ [78] Ames dwarf ↑↑ ↑↑↑ [79] lit/lit ↑↑ ↑↑ [78] Ghr-/- ↑↑ ↑↑ [90] Ghrh-/- ↑↑ [94] Liver Igf1-/- [95] Liver Igf1-/- [96] Igf1r+/- ↑↑ [97] Igf1r+/- [98,99] brain Igf1r+/- [100] PAPP-A-/- ↑↑ [80] IR+/- [81] FIRKO ↑ ↑ [82] Ins1-/-;Ins2+/- ? [83] IRS1+/- [84] IRS1-/- ↑↑ [84,85] IRS2+/- [84] IRS2+/- [86] IRS2-/- ↓↓ [84,87] brain IRS2+/- [86] p110a+/- [88] mtor+/-;mist8+/- [89] mtorΔ/Δ [91] TSC TG − ↑ [92] S6K-/- [93]

− = no change, ↑ = small increase (<10%), ↑↑ = large increase (20-50%), ↑↑↑ = extra-large increase (>50%), ↓ = small reduction (<10%), ↓↓ = large reduction (20-50%)

(10)

7

longevity are small and inconsistent. Depending on the species and strain, it has been

reported that males are the longer-lived sex, that there is no sex difference, or that females live longer [76]. However, as the average lifespan differs between males and females, the generic effects of caloric restriction cannot be assumed. Caloric restriction, defined as a 40% reduction in caloric intake, effectively extends lifespan and improves metabolic health, but the most optimal level of restriction may be strain- and gender-specific. A meta-analysis illustrated that the lifespan-extending effects of 40% CR in mice ranged between 4–27% [104]. In C57Bl/6, various levels of caloric restriction suggest that longevity in females is only extended by 20% CR, not 40%, while the beneficial effects on glucose homeostasis are not different between levels of CR and gender [105]. In contrast, we observed the expected rapid decrease in body weight and blood glucose levels after CR treatment, but found that it was more pronounced in females than in males (Chapter 6). Female mice showed a reduction in their islets insulin content due to CR, which was not found in males, suggesting that female Ins2−/− mice may be more susceptible to the effects of CR. Furthermore, the effects of CR on adipose tissue size seem stronger in females compared to males. Upregulation of genes involved in lipogenesis and thermogenesis in scWAT were more pronounced in females and brown adipose tissue (BAT) activity was still active in the female CR

Ins2−/− mice while it was diminished in males. Together, these results highlight the complexity of CR in the context of gender and suggest the uncoupling of healthspan and lifespan [105]. The molecular mechanisms underlying the sex differences in the metabolic responses to CR clearly require further study.

Translation of pharmacological and dietary intervention to humans

While this discussion describes several factors that confound the interpretation of data of dietary interventions in mouse models, species differences also need to be taken into account to predict the human outcome of these lifestyle changes or therapeutical interventions. This thesis described an important role of brown and white adipose tissue activation by protein restriction or FGF21 administration on metabolic health. However, it remains elusive whether FGF21 acts similarly in mice and men. In Chapter 2, we describe the clinical trials that are currently conducted with FGF21-analogues and highlight some differences in results compared to preclinical research. While very effective in reducing plasma TG and hepatic lipid accumulation, the glucose-lowering effects are not observed in NAFLD- and T2D patients [106]. These differences between humans and rodents are potentially explained by different mechanisms.

First, different body composition and adipose tissue function between mice and humans can lead to distinct effects. Many adipose tissue depots in mice are barely present in humans and vice versa, such as the human omentum or the murine epididymal fat [107]. Also, mice have relatively large amounts of BAT as they heavily rely on heat generation to maintain body temperature, while humans lose most of their BAT within a few years after birth [108]. Recently, the existence of genuine brown fat in adult humans has been proven by the detection of bona fide UCP1+ adipose tissue

in biopsies [109–111]. These UCP1+ cells in humans display molecular markers of

(11)

Pax7+Myf5+ precursor cells that give rise to classic BAT [107]. However, while the exact origin of these cells remains unknown, a specific CD81+ precursor population has

recently been discovered in the human subcutaneous adipose depot that correlates negatively with metabolic health [112,115,116]. Further studies need to determine how human brown/beige adipocytes are induced by dietary and pharmacological interventions. However, these relatively little amounts of BAT compared to rodents may account for the attenuated glucose-lowering response by FGF21 treatment observed in human clinical trials.

Second, FGF21-induced effects in mice and humans can be affected by species differences within FGFR and βKlotho expression. Previous results suggest that FGF21-induced effects in mice are dependent on βKlotho expression in the central nervous system to stimulate BAT and WAT [117]. However, while FGF-receptor 4 and βKlotho were detected in several regions of the murine brain, including the hypothalamus, mRNA encoding these receptors were not detected in cynomolgus monkey or human brain samples [118]. These species differences are not only important for FGF21 signalling, but possibly also affect the response of other FGF-based therapeutics. In this thesis, we have suggested species differences by fibroblast growth factor 1 (FGF1) signalling, independent of βKlotho (Chapter 3). As the therapeutic application of FGF1 is limited by its high mitogenic properties, several FGF1 analogues have been developed to reduce these effects [119–121]. While these compounds retain their metabolic activity in vivo, determined by glucose-lowering and antisteatotic effects in several mouse models, it remains to be determined whether species differences will affect the outcome. We determined the metabolic and mitogenic potential of these FGF1 analogues in a rat hepatocyte cell line to assess the potential as a NAFLD target, but observed an unexpected mitogenic potential of these FGF1 analogues that can potentially be explained by species differences. However, more research is needed to exclude tissue-specific or cell-specific effects.

Conclusion and further perspectives

Altogether, the research in this thesis highlights the perspective of dietary or pharmacological interventions in the context of ageing. While we know that ageing is associated with several processes that reduce metabolic flexibility, limited studies are available that study metabolic health in aged mice after dietary intervention. We found that the macronutrient composition of the diets is of vital importance to maintain insulin sensitivity, muscle strength, and adipose tissue function during ageing. Based on these findings, we conclude that healthy dietary interventions can improve metabolic health at an advanced age by stimulating metabolic flexibility, but that an unhealthy lifestyle is highly detrimental due to the loss of compensatory mechanisms. However, further studies are needed to investigate the metabolic impact of dietary interventions at different ages, with age-induced obesity taken into account. Ageing is a generic stochastic process, causing significant variation in body weight but also in other metabolic parameters such as hormone levels and incidence of metabolic diseases. Further research should therefore focus on various subcategories within the elderly to determine whether body weight and its associated metabolic status is a confounding

(12)

7

factor in the efficiency of dietary interventions. This will potentially contribute to the

improvement of personalized dietary- or pharmacological interventions in the elderly, and further highlight age-related metabolic changes.

References

1. Fink RI, Kolterman OG, Griffin J, Olefsky JM. Mechanisms of insulin resistance in aging. J Clin Invest. 1983;71(6):1523–35.

2. DeFronzo RA. Glucose intolerance and aging. Vol. 4, Diabetes Care. 1981. p. 493–501. 3. Mehran AE, Templeman NM, Brigidi GS, Lim GE, Chu K-YY, Hu XK, et al. Hyperinsulinemia drives

diet-induced obesity independently of brain insulin production. Cell Metab. 2012;16(6):723–37. 4. Breen L, Phillips SM. Skeletal muscle protein metabolism in the elderly: Interventions to counteract

the “anabolic resistance” of ageing. Nutr Metab. 2011;8:1–11.

5. Robinson SM, Reginster JY, Rizzoli R, Shaw SC, Kanis JA, Bautmans I, et al. Does nutrition play a role in the prevention and management of sarcopenia? Clin Nutr. 2018;37(4):1121–32. 6. Stolle S, Ciapaite J, Reijne AC, Talarovicova A, Wolters JC, Aguirre-Gamboa R, et al.

Running-wheel activity delays mitochondrial respiratory flux decline in aging mouse muscle via a post-transcriptional mechanism. Aging Cell. 2017;(September):e12700.

7. Tchkonia T, Morbeck DE, Von Zglinicki T, Van Deursen J, Lustgarten J, Scrable H, et al. Fat tissue, aging, and cellular senescence. Aging Cell. 2010;9(5):667–84.

8. Mancuso P, Bouchard B. The impact of aging on adipose function and adipokine synthesis. Front Endocrinol (Lausanne). 2019;10(MAR):1–12.

9. Rogers NH, Landa A, Park S, Smith RG. Aging leads to a programmed loss of brown adipocytes in murine subcutaneous white adipose tissue. Aging Cell. 2012;11(6):1074–83.

10. van der Heijden RA, Sheedfar F, Morrison MC, Hommelberg PPH, Kor D, Kloosterhuis NJ, et al. High-fat diet induced obesity primes inflammation in adipose tissue prior to liver in C57BL/6j mice. Aging (Albany NY). 2015;7(4):256–68.

11. Khan S, Chan YT, Revelo XS, Winer DA. The Immune Landscape of Visceral Adipose Tissue During Obesity and Aging. Front Endocrinol (Lausanne). 2020;11(May):1–18.

12. Hahn O, Drews LF, Nguyen A, Tatsuta T, Gkioni L, Hendrich O, et al. A nutritional memory effect counteracts the benefits of dietary restriction in old mice. Nat Metab. 2019;1(11):1059–73. 13. Schumacher B, Van Der Pluijm I, Moorhouse MJ, Kosteas T, Robinson AR, Suh Y, et al. Delayed

and accelerated aging share common longevity assurance mechanisms. PLoS Genet. 2008;4(8). 14. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell.

2013;153(6).

15. Anversa P. Aging and longevity: The IGF-1 enigma. Circ Res. 2005;97(5):411–4.

16. Milman S, Atzmon G, Huffman DM, Wan J, Crandall JP, Cohen P, et al. Low insulin-like growth factor-1 level predicts survival in humans with exceptional longevity. Aging Cell. 2014 Aug 1;13(4):769–71.

17. van der Spoel E, Rozing MP, Houwing-Duistermaat JJ, Slagboom PE, Beekman M, de Craen AJM, et al. 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 Nov;7(11):956–63.

18. Strasburger CJ, Bidlingmaier M, Wu Z, Morrison KM. Normal values of insulin-like growth factor I and their clinical utility in adults. Horm Res. 2001;55(SUPPL. 2):100–5.

19. 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(5):497–500. 20. Yeap BB, Hui J, Knuiman MW, Paul Chubb SA, Ho KKY, Flicker L, et al. Associations of plasma

IGF1, IGFBP3 and estradiol with leucocyte telomere length, a marker of biological age, in men. Eur J Endocrinol. 2019 Jan 1;182(1):23–33.

21. 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(1):114–20.

(13)

22. Andreassen M, Raymond I, Kistorp C, Hildebrandt P, Eaber J, Kristensen LØ. IGF1 as predictor of all cause mortality and cardiovascular disease in an elderly population. Eur J Endocrinol. 2009;160(1):25–31.

23. Friedrich N, Haring R, Nauck M, Lüdemann J, Rosskopf D, Spilcke-Liss E, et al. Mortality and serum insulin-like growth factor (IGF)-I and IGF binding protein 3 concentrations. J Clin Endocrinol Metab. 2009;94(5):1732–9.

24. Brugts MP, Van Den Beld AW, Hofland LJ, Van Der Wansem K, Van Koetsveld PM, Frystyk J, et al. Low circulating insulin-like growth factor I bioactivity in elderly men is associated with increased mortality. J Clin Endocrinol Metab. 2008;93(7):2515–22.

25. Levine ME, Suarez JA, Brandhorst S, Balasubramanian P, Cheng CW, Madia F, 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(3):407–17.

26. Markan KR, Naber MC, Ameka MK, Anderegg MD, Mangelsdorf DJ, Kliewer SA, et al. Circulating FGF21 is liver derived and enhances glucose uptake during refeeding and overfeeding. Diabetes. 2014;63(12):4057–63.

27. Owen BMBM, Mangelsdorf DJDJDJ, Kliewer SAS a. Tissue-specific actions of the metabolic hormones FGF15/19 and FGF21. Trends Endocrinol Metab. 2015;26(1):22–9.

28. Kharitonenkov A, Shiyanova TL, Koester A, Ford AM, Micanovic R, Galbreath EJ, et al. FGF-21 as a novel metabolic regulator. J Clin Invest. 2005 Jun 1;115(6):1627–35.

29. Xu J, Lloyd DJ, Hale C, Stanislaus S, Chen M, Sivits G, et al. FGF21 Reverses Hepatic Steatosis, Increases Energy Expenditure and Improves Insulin Sensitivity in Diet-induced Obese Mice. Diabetes. 2008;58(January):250–9.

30. Zhang X, Yeung DCY, Karpisek M, Stejskal D, Zhou Z, Liu F, et al. Serum FGF21 Levels Are Increased in Obesity and Are Independently associated with the metabolic syndrome in Humans. Diabetes. 2008;57(May):1246–53.

31. Mraz M, Bartlova M, Lacinova Z, Michalsky D, Kasalicky M, Haluzikova D, et al. Serum concentrations and tissue expression of a novel endocrine regulator fibroblast growth factor-21 in patients with type 2 diabetes and obesity. Clin Endocrinol (Oxf). 2009;71(3):369–75.

32. Villarroya J, Gallego-Escuredo JM, Delgado-Anglés A, Cairó M, Moure R, Gracia Mateo M, et al. Aging is associated with increased FGF21 levels but unaltered FGF21 responsiveness in adipose tissue. Aging Cell. 2018;17(5):17–21.

33. Sanchis-Gomar F, Pareja-Galeano H, Santos-Lozano A, Garatachea N, Fiuza-Luces C, Venturini L, et al. A preliminary candidate approach identifies the combination of chemerin, fetuin-A, and fibroblast growth factors 19 and 21 as a potential biomarker panel of successful aging. Age (Omaha). 2015;37(3).

34. Gallego-Escuredo JM, Gómez-Ambrosi J, Catalan V, Domingo P, Giralt M, Frühbeck G, et al. Opposite alterations in FGF21 and FGF19 levels and disturbed expression of the receptor machinery for endocrine FGFs in obese patients. Int J Obes. 2015 Jan 12;39(1):121–9. 35. Chavez AO, Molina-Carrion M, Abdul-Ghani MA, Folli F, DeFronzo RA, Tripathy D. Circulating

fibroblast growth factor-21 is elevated in impaired glucose tolerance and type 2 diabetes and correlates with muscle and hepatic insulin resistance. Diabetes Care. 2009;32(8):1542–6. 36. Hong ES, Lim C, Choi HY, Lee YK, Ku EJ, Moon JH, et al. Plasma fibroblast growth factor 21

levels increase with ectopic fat accumulation and its receptor levels are decreased in the visceral fat of patients with type 2 diabetes. BMJ Open Diabetes Res Care. 2019;7(1):1–7.

37. Chen C, Cheung BMY, Tso AWK, Wang Y, Law LSC, Ong KL, et al. High plasma level of fibroblast growth factor 21 is an independent predictor of type 2 diabetes: A 5.4-year population-based prospective study in Chinese subjects. Diabetes Care. 2011;34(9):2113–5.

38. Bobbert T, Schwarz F, Fischer-Rosinsky A, Pfeiffer AFHH, Möhlig M, Mai K, et al. Fibroblast growth factor 21 predicts the metabolic syndrome and type 2 diabetes in Caucasians. Diabetes Care. 2013;36(1):145–9.

39. Xiao Y, Xu A, Law LSC, Chen C, Li H, Li X, et al. Distinct changes in serum fibroblast growth factor 21 levels in different subtypes of diabetes. J Clin Endocrinol Metab. 2012;97(1):54–8.

40. Li H, Wu G, Fang Q, Zhang M, Hui X, Sheng B, et al. Fibroblast growth factor 21 increases insulin sensitivity through specific expansion of subcutaneous fat. Nat Commun. 2018;9(1).

(14)

7

41. Dushay J, Chui PC, Gopalakrishnan GS, Varela-Rey M, Crawley M, Fisher FM, et al. Increased fibroblast growth factor 21 in obesity and nonalcoholic fatty liver disease. Gastroenterology. 2010;139(2):456–63.

42. Hanks LJ, Gutiérrez OM, Bamman MM, Ashraf A, McCormick KL, Casazza K. Circulating levels of fibroblast growth factor-21 increase with age independently of body composition indices among healthy individuals. J Clin Transl Endocrinol. 2015;2(2):72–82.

43. Salminen A, Kaarniranta K, Kauppinen A. Regulation of longevity by FGF21: Interaction between energy metabolism and stress responses. Ageing Res Rev. 2017;37:79–93.

44. Markan KR. Defining “FGF21 Resistance” during obesity: Controversy, criteria and unresolved questions. F1000Research. 2018;7:289.

45. Tanajak P. Letter to the editor: Parameters, characteristics, and criteria for defining the term “fGF21 resistance.” Endocrinology. 2017;158(5):1523–4.

46. Fisher FM, Chui PC, Antonellis PJ, Bina HA, Kharitonenkov A, Flier JS, et al. Obesity is a fibroblast growth factor 21 (FGF21)-resistant state. Diabetes. 2010;59(11):2781–9.

47. Hale C, Chen MM, Stanislaus S, Chinookoswong N, Hager T, Wang M, et al. Lack of overt FGF21 resistance in two mouse models of obesity and insulin resistance. Endocrinology. 2012;153(1):69–80.

48. Li H, Fang Q, Gao F, Fan J, Zhou J, Wang X, et al. Fibroblast growth factor 21 levels are increased in nonalcoholic fatty liver disease patients and are correlated with hepatic triglyceride. J Hepatol. 2010;53(5):934–40.

49. Sohal RS, Ferguson M, Sohal BH, Forster MJ. Life Span Extension in Mice by Food Restriction Depends on an Energy Imbalance. J Nutr. 2009;139(3):533–9.

50. Liao C-YY, Rikke BA, Johnson TE, Diaz V, Nelson JF. Genetic variation in the murine lifespan response to dietary restriction: From life extension to life shortening. Aging Cell. 2010;9(1):92–5. 51. Ferguson M, Rebrin I, Forster MJ, Sohal RS. Comparison of metabolic rate and oxidative stress between two different strains of mice with varying response to caloric restriction. Exp Gerontol. 2008;43(8):757–63.

52. Ferguson M, Sohal BH, Forster MJ, Sohal RS. Effect of long-term caloric restriction on oxygen consumption and body temperature in two different strains of mice. Mech Ageing Dev. 2007;128(10):539–45.

53. Forster MJ, Morris P, Sohal RS. Genotype and age influence the effect of caloric intake on mortality in mice. FASEB J. 2003;17(6):690–2.

54. Sohal RS, Forster MJ. Caloric restriction and the aging process: A critique. Free Radic Biol Med. 2014;73:366–82.

55. Dionne DA, Skovsø S, Templeman NM, Clee SM, Johnson JD. Caloric restriction paradoxically increases adiposity in mice with genetically reduced insulin. Endocrinology. 2016;157(7):2724–34. 56. Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: An update. Ageing Res

Rev. 2017;39:36–45.

57. Ravussin E, Redman LM, Rochon J, Das SK, Fontana L, Kraus WE, et al. A 2-year randomized controlled trial of human caloric restriction: Feasibility and effects on predictors of health span and longevity. Journals Gerontol - Ser A Biol Sci Med Sci. 2015;70(9):1097–104.

58. Villareal DT, Fontana L, Das SK, Redman L, Smith SR, Saltzman E, et al. Effect of Two-Year Caloric Restriction on Bone Metabolism and Bone Mineral Density in Non-Obese Younger Adults: A Randomized Clinical Trial. J Bone Miner Res. 2016 Jan 1;31(1):40–51.

59. Fontana L, Villareal DT, Das SK, Smith SR, Meydani SN, Pittas AG, 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. 2015;(August):n/a-n/a.

60. Laeger T, Münzberg H, Henagan TM, Hutson SM, Albarado DC, Gettys TW. FGF21 is an endocrine signal of protein restriction. J Clin Invest. 2014;124(9):1–3.

61. Fontana L, Cummings NE, Arriola Apelo SI, Neuman JC, Kasza I, Schmidt BA, et al. Decreased consumption of branched chain amino acids improves metabolic health. Cell Rep. 2016;16(2):520–30.

62. Hill CM, Laeger T, Albarado DiC, McDougal DH, Berthoud HR, Münzberg H, et al. Low protein-induced increases in FGF21 drive UCP1-dependent metabolic but not thermoregulatory endpoints.

(15)

Sci Rep. 2017;7(1):1–11.

63. Laeger T, Albarado DC, Burke SJ, Trosclair L, Hedgepeth JW, Berthoud HR, et al. Metabolic Responses to Dietary Protein Restriction Require an Increase in FGF21 that Is Delayed by the Absence of GCN2. Cell Rep. 2016;16(3):707–16.

64. Pérez-Martí A, Garcia-Guasch M, Tresserra-Rimbau A, Carrilho-Do-Rosário A, Estruch R, Salas-Salvadó J, et al. A low-protein diet induces body weight loss and browning of subcutaneous white adipose tissue through enhanced expression of hepatic fibroblast growth factor 21 (FGF21). Mol Nutr Food Res. 2017;61(8):1–10.

65. Link JC, Reue K. Genetic Basis for Sex Differences in Obesity and Lipid Metabolism. Annu Rev Nutr. 2017;37:225–45.

66. Sebo ZL, Church C, Berry R, Rodeheffer MS, Rodeheffer M. Adipose tissue developmental growth constraints uncouple fat distribution from glucose metabolism in two mouse models of obesity. 2020;1–28.

67. Parks BW, Sallam T, Mehrabian M, Psychogios N, Hui ST, Norheim F, et al. Genetic architecture of insulin resistance in the mouse. Cell Metab. 2015 Feb 3;21(2):334–47.

68. Jeffery E, Church CD, Holtrup B, Colman L, Rodeheffer MS. Rapid depot-specific activation of adipocyte precursor cells at the onset of obesity. Nat Cell Biol. 2015;17(4):376–85.

69. Wang QA, Tao C, Gupta RK, Scherer PE. Tracking adipogenesis during white adipose tissue development, expansion and regeneration. Nat Med. 2013 Oct 1;19(10):1338–44.

70. Arner P, Andersson DP, Thörne A, Wirén M, Hoffstedt J, Näslund E, et al. Variations in the Size of the Major Omentum Are Primarily Determined by Fat Cell Number. J Clin Endocrinol Metab. 2013 May 1;98(5):E897–901.

71. Jeffery E, Wing A, Holtrup B, Sebo Z, Kaplan JL, Saavedra-Peña R, et al. The Adipose Tissue Microenvironment Regulates Depot-Specific Adipogenesis in Obesity. Cell Metab. 2016;24(1):142–50.

72. Larson KR, Russo KA, Fang Y, Mohajerani N, Goodson ML, Ryan KK. Sex Differences in the Hormonal and Metabolic Response to Dietary Protein Dilution. 2017;158(October):3477–87. 73. Yu D, Yang SE, Miller BR, Wisinski JA, Sherman DS, Brinkman JA, et al. Short-Term methionine

deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms. FASEB J. 2018;32(6):3471–82.

74. Chaumontet C, Azzout-Marniche D, Blais A, Piedcoq J, Tomé D, Gaudichon C, et al. Low-protein and methionine, high-starch diets increase energy intake and expenditure, increase FGF21, decrease IGF-1, and have little effect on adiposity in mice. Am J Physiol Regul Integr Comp Physiol. 2019;316(5):R486–501.

75. Forney LA, Stone KP, Gibson AN, Vick AM, Sims LC, Fang H, et al. Sexually Dimorphic Effects of Dietary Methionine Restriction are Dependent on Age when the Diet is Introduced. Obesity. 2020;28(3):581–9.

76. Austad SN, Bartke A. Sex Differences in Longevity and in Responses to Anti-Aging Interventions: A Mini-Review. Gerontology. 2015;62(1):40–6.

77. Junnila RK, List EO, Berryman DE, Murrey JW, Kopchick JJ. The GH/IGF-1 axis in ageing and longevity. Nat Rev Endocrinol. 2013;9(6):366–76.

78. Flurkey K, Papaconstantinou J, Miller RA, Harrison DE. Lifespan extension and delayed immune and collagen aging in mutant mice with defects in growth hormone production. Proc Natl Acad Sci U S A. 2001 Jun 5;98(12):6736–41.

79. Brown-Borg HM, Borg KE, Meliska CJ, Bartke A. Dwarf mice and the ageing process. Nature. 1996;384(6604):33.

80. Conover CA, Bale LK, Mader JR, Mason MA, Keenan KP, Marler RJ. Longevity and age-related pathology of mice deficient in pregnancy-associated plasma protein-a. Journals Gerontol - Ser A Biol Sci Med Sci. 2010 Jun;65 A(6):590–9.

81. Nelson JF, Strong R, Bokov A, Diaz V, Ward W. Probing the relationship between insulin sensitivity and longevity using genetically modified mice. Journals Gerontol - Ser A Biol Sci Med Sci. 2012;67(12):1332–8.

82. Blüher M, Kahn BBM, Kahn CR, Bluher M, Kahn BBM, Kahn CR, et al. Extended Longevity in Mice Lacking the Insulin Receptor in Adipose Tissue. Science (80- ). 2003 Jan 24;299(January):2000–2.

(16)

7

83. Templeman NM, Flibotte S, Chik JHL, Sinha S, Lim GE, Foster LJ, et al. Reduced Circulating Insulin Enhances Insulin Sensitivity in Old Mice and Extends Lifespan. Cell Rep. 2017;20(2):451–63. 84. Selman C, Lingard S, Choudhury AI, Batterham RL, Claret M, Clements M, et al. Evidence for

lifespan extension and delayed age-related biomarkers in insulin receptor substrate 1 null mice. FASEB J. 2007 Mar;22(3):807–18.

85. Selman C, Partridge L, Withers DJ. Replication of extended lifespan phenotype in mice with deletion of insulin receptor substrate 1. PLoS One. 2011;6(1):3–5.

86. Taguchi A, Wartschow LM, White MF. Brain IRS2 signaling coordinates life span and nutrient homeostasis. Science (80- ). 2007 Jul 20;317(5836):369–72.

87. Selman C, Lingard S, Gems D, Partridge L, Withers DJ. Comment on “Brain IRS2 signaling coordinates life span and nutrient homeostasis.” Vol. 320, Science. American Association for the Advancement of Science; 2008. p. 1012–1012.

88. Foukas LC, Bilanges B, Bettedi L, Pearce W, Ali K, Sancho S, et al. Long-term p110α PI3K inactivation exerts a beneficial effect on metabolism. EMBO Mol Med. 2013;5(4):563–71. 89. Lamming DW, Ye A, Katajisto P, Goncalves MD, Saitoh M, Stevens DM, et al.

Rapamycin-Induced Insulin Resistance Is Mediated by mTORC2 Loss and Uncoupled from Longevity. 2012;335(March).

90. Coschigano KT, Holland AN, Riders ME, List EO, Flyvbjerg A, Kopchick JJ. Deletion, but not antagonism, of the mouse growth hormone receptor results in severely decreased body weights, insulin, and insulin-like growth factor I levels and increased life span. Endocrinology. 2003;144(9):3799–810.

91. Wu JJ, Liu J, Chen EB, Wang JJ, Cao L, Narayan N, et al. Increased mammalian lifespan and a segmental and tissue-specific slowing of aging after genetic reduction of mTOR expression. Cell Rep. 2013;4(5):913–20.

92. Zhang HM, Diaz V, Walsh ME, Zhang Y. Moderate lifelong overexpression of tuberous sclerosis complex 1 (TSC1) improves health and survival in mice. Sci Rep. 2017;7(1):1–14.

93. Selman C, Tullet JMA, Wieser D, Irvine E, Lingard SJ, Choudhury AI, et al. Ribosomal protein S6 kinase 1 signaling regulates mammalia life span. Science (80- ). 2009;263(October):140–4. 94. Sun LY, Spong A, Swindell WR, Fang Y, Hill C, Huber JA, et al. Growth hormone-releasing

hormone disruption extends lifespan and regulates response to caloric restriction in mice. Elife. 2013;2013(2):1–24.

95. Gong Z, Kennedy O, Sun H, Wu Y, Williams GA, Klein L, et al. Reductions in serum IGF-1 during aging impair health span. Aging Cell. 2014;13(3):408–18.

96. Svensson J, Sjögren K, Fäldt J, Andersson N, Isaksson O, Jansson J-O, et al. Liver-derived IGF-I regulates mean life span in mice. PLoS One. 2011;6(7):e22640.

97. Holzenberger M, Dupont J, Ducos B, Leneuve P, Géloën A, Even PC, et al. IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature. 2003;421(6919):182–7. 98. Bokov AF, Garg N, Ikeno Y, Thakur S, Musi N, DeFronzo RA, et al. Does Reduced IGF-1R

Signaling in Igf1r+/− Mice Alter Aging? Lo ACY, editor. PLoS One. 2011 Nov 23;6(11):e26891. 99. 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(1):19–28. 100. Kappeler L, De Magalhaes Filho C, Dupont J, Leneuve P, Cervera P, Périn L, et al. Brain IGF-1 receptors control mammalian growth and lifespan through a neuroendocrine mechanism. PLoS Biol. 2008;6(10):2144–53.

101. Templeman NM, Clee SM, Johnson JD. Suppression of hyperinsulinaemia in growing female mice provides long-term protection against obesity. Diabetologia. 2015;58(10):2392–402. 102. Templeman NM, Mehran AE, Johnson JD. Hyper-Variability in Circulating Insulin, High Fat

Feeding Outcomes, and Effects of Reducing Ins2 Dosage in Male Ins1-Null Mice in a Specific Pathogen-Free Facility. PLoS One. 2016;11(4):e0153280.

103. Botezelli JD, Overby P, Lindo L, Wang S, Haïda O, Lim GE, et al. Adipose depot-specific upregulation of Ucp1 or mitochondrial oxidative complex 3 proteins are early consequences of genetic insulin reduction in mice. Am J Physiol Endocrinol Metab. 2020;21(1):1–9.

104. Swindell WR. Dietary restriction in rats and mice: A meta-analysis and review of the evidence for genotype-dependent effects on lifespan. Ageing Res Rev. 2012;11(2):254–70.

(17)

105. Mitchell SJ, Madrigal-Matute J, Scheibye-Knudsen M, Fang E, Aon M, González-Reyes JA, et al. Effects of Sex, Strain, and Energy Intake on Hallmarks of Aging in Mice. Cell Metab. 2016;23(6):1093–112.

106. Struik D, Dommerholt MB, Jonker JW. Fibroblast growth factors in control of lipid metabolism: from biological function to clinical application. Curr Opin Lipidol. 2019 Jun 1;30(3):235–43. 107. Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell. 2014;156(1–

2):20–44.

108. Zoico E, Rubele S, De Caro A, Nori N, Mazzali G, Fantin F, et al. Brown and beige adipose tissue and aging. Front Endocrinol (Lausanne). 2019;10(JUN):1–10.

109. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, et al. Identification and Importance of Brown Adipose Tissue in Adult Humans. N Engl J Med. 2009 Apr 9;360(15):1509–17.

110. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JMAFL, Kemerink GJ, Bouvy ND, et al. Cold-Activated Brown Adipose Tissue in Healthy Men. N Engl J Med. 2009;360(15):1500–8.

111. Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren R, Niemi T, et al. Functional Brown Adipose Tissue in Healthy Adults. N Engl J Med. 2009 Apr 9;360(15):1518–25.

112. Wu J, Bostrom P, Sparks LM, Ye L, Choi JH, Giang A-H, et al. Beige Adipocytes are a Distinct Type of Thermogenic Fat Cell in Mouse and Human. Cell Rep. 2012;150(6):366–76.

113. Schulz T, Tseng Y-H. Brown adipose tissue: development, metabolism and beyond. Biochem J. 2014;453(2):1–24.

114. Cypess AM, White AP, Vernochet C, Schulz TJ, Xue R, Sass CA, et al. Anatomical localization, gene expression profiling and functional characterization of adult human neck brown fat. Nat Med. 2013;19(5):635–9.

115. Oguri Y, Shinoda K, Kim H, Alba DL, Bolus WR, Wang Q, et al. CD81 Controls Beige Fat Progenitor Cell Growth and Energy Balance via FAK Signaling. Cell. 2020;182(3):563-577.e20.

116. Villanueva CJ. Finding a Needle in a Haystack: Identification of a Beige Fat Progenitor. Cell. 2020;182(3):537–9.

117. Owen BM, Ding X, Morgan DA, Coate KC, Bookout AL, Rahmouni K, et al. FGF21 acts centrally to induce sympathetic nerve activity, energy expenditure, and weight loss. Cell Metab. 2014;20(4):670–7.

118. Hultman K, Scarlett JM, Baquero AF, Cornea A, Zhang Y, Salinas CBG, et al. The central fibroblast growth factor receptor/beta klotho system: Comprehensive mapping in Mus musculus and comparisons to nonhuman primate and human samples using an automated in situ hybridization platform. J Comp Neurol. 2019;527(12):2069–85.

119. Suh JM, Jonker JW, Ahmadian M, Goetz R, Lackey D, Osborn O, et al. Endocrinization of FGF1 produces a neomorphic and potent insulin sensitizer. Nature. 2014;513(7518):436–9.

120. Klingenberg O, Wiçdłocha A, Rapak A, Muñoz R, Falnes P, Olsnes S. Inability of the acidic fibroblast growth factor mutant K132E to stimulate DNA synthesis after translocation into cells. J Biol Chem. 1998 May 1;273(18):11164–72.

121. Mori S, Tran V, Nishikawa K, Kaneda T, Hamada Y, Kawaguchi N, et al. A Dominant-Negative FGF1 Mutant (the R50E Mutant) Suppresses Tumorigenesis and Angiogenesis. PLoS One. 2013;8(2).

(18)
(19)

Referenties

GERELATEERDE DOCUMENTEN

We determined several parameters of metabolic health and revealed that short-term dietary protein restriction in aged mice ameliorated the age-related decline of some aspects

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.. Downloaded

The research described in this thesis was carried out at the Laboratory of Pediatrics (Head: prof. Kuipers), Section Molecular Metabolism and Nutrition, University Medical

Apart from interfering with glucose metabolism in the heart, muscle, and liver, elevated plasma fatty acid levels also drive hepatic lipid accumulation.. Previous studies have

Reduced levels of FGF19 are generally observed in obesity and related disorders, including T2D, gestational diabetes, non- alcoholic fatty liver disease (NAFLD) and (NASH)

Pegbelfermin (BMS-986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: a randomised, double- blind, placebo-controlled,

In addition to its potent anti-steatotic action, rFGF1 also suppressed hepatic inflammation in ob/ob mice as indicated by reduced mRNA expression of a range of hepatic

Een verhoogde sympathische activiteit lijkt ook een rol te spelen bij de metabole effecten van FGF19, FGF21 en na toediening van FGF1 in het brein.. Gezamenlijk geven