Next Article in Journal
Divergent Impact of Glucose Availability on Human Virus-Specific and Generically Activated CD8 T Cells
Next Article in Special Issue
Control of Adipose Cell Browning and Its Therapeutic Potential
Previous Article in Journal
1H-NMR-Based Analysis for Exploring Knee Synovial Fluid Metabolite Changes after Local Cryotherapy in Knee Arthritis Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Impact of Long-Term HFD Intake on the Peripheral and Central IGF System in Male and Female Mice

by
Santiago Guerra-Cantera
1,2,3,
Laura M. Frago
1,2,3,
María Jiménez-Hernaiz
1,3,
Purificación Ros
2,4,
Alejandra Freire-Regatillo
1,2,3,
Vicente Barrios
1,3,
Jesús Argente
1,2,3,5,* and
Julie A. Chowen
1,3,5,*
1
Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain
2
Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain
3
Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
4
Department of Pediatrics, Hospital Universitario Puerta de Hierro-Majadahonda, E-28222 Madrid, Spain
5
IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, E-28049 Madrid, Spain
*
Authors to whom correspondence should be addressed.
Metabolites 2020, 10(11), 462; https://doi.org/10.3390/metabo10110462
Submission received: 22 October 2020 / Revised: 10 November 2020 / Accepted: 11 November 2020 / Published: 13 November 2020
(This article belongs to the Special Issue Neuroendocrine Control of Energy Metabolism)

Abstract

:
The insulin-like growth factor (IGF) system is responsible for growth, but also affects metabolism and brain function throughout life. New IGF family members (i.e., pappalysins and stanniocalcins) control the availability/activity of IGFs and are implicated in growth. However, how diet and obesity modify this system has been poorly studied. We explored how intake of a high-fat diet (HFD) or commercial control diet (CCD) affects the IGF system in the circulation, visceral adipose tissue (VAT) and hypothalamus. Male and female C57/BL6J mice received HFD (60% fat, 5.1 kcal/g), CCD (10% fat, 3.7 kcal/g) or chow (3.1 % fat, 3.4 kcal/g) for 8 weeks. After 7 weeks of HFD intake, males had decreased glucose tolerance (p < 0.01) and at sacrifice increased plasma insulin (p < 0.05) and leptin (p < 0.01). Circulating free IGF1 (p < 0.001), total IGF1 (p < 0.001), IGF2 (p < 0.05) and IGFBP3 (p < 0.01) were higher after HFD in both sexes, with CCD increasing IGFBP2 in males (p < 0.001). In VAT, HFD reduced mRNA levels of IGF2 (p < 0.05), PAPP-A (p < 0.001) and stanniocalcin (STC)-1 (p < 0.001) in males. HFD increased hypothalamic IGF1 (p < 0.01), IGF2 (p < 0.05) and IGFBP5 (p < 0.01) mRNA levels, with these changes more apparent in females. Our results show that diet-induced changes in the IGF system are tissue-, sex- and diet-dependent.

Graphical Abstract

1. Introduction

The insulin-like growth factor (IGF) system has been widely studied in both pre- and postnatal growth and development, but this system is involved in a myriad of functions throughout life with much less known regarding many of these diverse roles. Members of this family include the ligands IGF1 and IGF2, which are crucial for longitudinal bone growth [1] and both also have additional important functions throughout life. Studies of IGF2 have focused mainly on its fundamental role during fetal life, as its expression decreases in multiple organs during aging in mice [2] as well as in the circulation in humans [3], but less is known about its postnatal actions. IGF1 and IGF2 bind to six different IGF binding proteins (IGFBPs) to increase their half-life and for transport to target tissues and importantly, to reduce their potential hypoglycemic effect. IGFBP3 is the most abundant binding protein in circulation, carrying from 70% to 90% of circulating IGF1 and 2 [4]. When IGF1 or IGF2 are bound to IGFBP3 or IGFBP5, it also binds to an acid-labile subunit (ALS) forming a trimolecular 150 KDa complex [5]. It is estimated that around 80% of the IGFs are bound in these trimolecular complexes, approximately 20% are bound to other IGFBPs and less than 1% are in the free form [6]. IGFBP2 is the second most abundant binding protein in serum, at least in some conditions [7,8], with a slight preference for IGF2 over IGF1 [9]. However, expression levels of the IGFBPs are tissue-specific, with, for example, IGFBP2 being the most abundant IGFBP in the postnatal central nervous system (CNS) [10], as well as in white preadipocytes during adipogenesis [11]; while IGFBP4 is the most abundant in cultured adult human adipose tissue [12].
When released from their binding proteins, the IGFs can activate the IGF1 receptor (IGF1R), which is structurally similar to the insulin receptor and when phosphorylated activates the PI3K/AKT pathway. Activation of this pathway underlies the anabolic effects of the IGFs [13]. In contrast, IGF2R is classified as a scavenger receptor for removing excess IGF2 from the circulation linked to lysosomal enzymes [14], with IGF2 having a 100-fold higher binding affinity for the IGF2R than IGF1 does [14].
New members of this system identified in recent years include pregnancy-associated plasma protein-A (PAPP-A) and PAPP-A2. These pappalysins modulate the biological activity of IGFs by cleaving the ligand from the binding protein and thus allowing them to bind to their receptors [15]. In addition, stanniocalcin (STC)-1 and 2 (STC-2) inhibit the proteolytic activity of pappalysins, reducing the release of IGF1 and 2 from the IGFBPs, hence their binding receptors [16,17].
The IGF system is important in anabolism, causing muscle hypertrophy [18], and is crucial in adipocyte maturation and differentiation [19]. Moreover, it can be altered by nutritional status [20]. In the brain it is important during development, but also throughout life. Indeed, in addition to peripheral IGF1 and IGF2 crossing the blood–brain barrier to reach the brain [21,22], IGFs are locally produced by brain cells, especially microglia and astrocytes [23,24]. These centrally produced IGFs can act as neuroprotective factors during events that result in gliosis and neuroinflammation. In the CNS, IGF1 is involved in postnatal synaptogenesis and neurogenesis [25], amyloid clearance [26], protection against neuroinflammation [24] and in neuroprotection [27], as well as with regulation of brain glucose metabolism [28]. The functions of IGF2 in the postnatal brain have been studied less, despite the fact that, for example, it is highly expressed in the hippocampus and has been associated with memory consolidation [29].
Gliosis, involving both astrocytes and microglia, is initially a protective response [30,31] but when prolonged can become damaging [32]. In response to high-fat diet (HFD) consumption, astrocytes are thought to participate in both the early protective response [33] and the detrimental effects of prolonged HFD intake and obesity [34]. The neuroprotective actions of astrocytes against oxidative stress [27], apoptosis and inflammation [35] are executed at least partially through the release of IGF1 [36,37] and consequently the downstream activation of the pro-survival pathway PI3K [38]. Thus, it is possible that the IGF system is involved in neuroprotection against the noxious effects of poor nutrition.
We have previously reported that short-term dietary changes modify the IGF system, with both a HFD and commercial control diet (CCD) inducing sex-specific changes peripherally and centrally compared to a normal rodent chow diet [39]. Here, our objective was to evaluate the long-term effects of HFD and CCD consumption on the central and peripheral IGF systems in male and female mice.

2. Results

2.1. Body Composition

Body weight changed with time throughout the study (F(8,53) = 325.6, p < 0.001; Figure 1A),and was also influenced by sex (F(1,53) = 235.6, p < 0.001) and diet (F(2,53) = 19.4, p < 0.001). At study onset, there were differences between the sexes (F(1,53) = 27.4, p < 0.001) in body weight (Figure 1A), with males weighing more than females and these sex differences were maintained throughout the study. HFD increased body weight in males from the first week (F(2,26) = 5.2, p < 0.05) until the last week of the study (F(2,26) = 62.2, p < 0.001). However, in females HFD intake induced a significant increase in weight only at the last week of the study (F(2,26) = 4.0, p < 0.05).
Weight gain (Figure 1B) was influenced by both sex (F(1,53) = 27.4, p < 0.001), with males gaining more weight than females, and diet (F(2,53) = 77.2, p < 0.001), with an interaction between these factors (F(2,53) = 6.4, p < 0.01). HFD induced weight gain in both males (F(2,26) = 116.2, p < 0.001) and females (F(2,26) = 13.5, p < 0.001).
Body weight at sacrifice (Table 1) was determined by sex (F(1,53) = 134.5, p < 0.001), with males weighing more than females, and by diet (F(2,53) = 55.4, p < 0.001), with an interaction between these two factors (F(2,53) = 8.5, p < 0.01). Body weight was higher after HFD consumption in both males (F(2,26) = 72.2, p < 0.001) and females (F(2,26) = 8.2, p < 0.01), with no effect of CCD intake on final body weight.
The amount of visceral adipose tissue (VAT; Table 1) was affected by both sex (F(1,53) = 24.8, p < 0.001), with males having higher levels than females, and diet (F(2,53) = 60.7, p < 0.001), with an interaction between these factors (F(2,53) = 9.1, p < 0.001). HFD intake increased the percentage of VAT in both males (F(2,26) = 94.9, p < 0.001) and females (F(2,26) = 8.6, p < 0.01). Subcutaneous adipose tissue content (Table 1) was influenced by diet (F(2,53) = 6.3, p < 0.001), with an interaction between sex and diet (F(2,53) = 4.9, p < 0.05). The percentage of subcutaneous adipose tissue was increased by HFD intake in both males (F(2,26) = 58.4, p < 0.001) and females (F(2,26) = 10.5, p < 0.01).
The number of kcal ingested per day was influenced by sex (F(1,17) = 62.5, p < 0.001; Table 1) and diet (F(2,17) = 267.9, p < 0.001). There was also an interaction between sex and diet (F(2,17) = 135.9, p < 0.001), with HFD only increasing energy intake per mouse in females (F(2,8) = 331.0, p < 0.001). When adjusted for body weight, females consumed more energy than males (F(1,17) = 232.4, p < 0.001), with this also being affected by diet (F(2,17) = 255.5, p < 0.001). There was an interaction between sex and diet (F(2,17) = 232.4, p < 0.001) as energy intake/g body weight was only increased by HFD in females (F(2,8) = 331.0, p < 0.001).
Energy efficiency, determined as the amount of weight gained per kilocalories consumed, was higher in males compared to females (F(1,17) = 156.4, p < 0.001; Table 1). Energy efficiency was also influenced by diet (F(2,17) = 37.3, p < 0.001), with an interaction between sex and diet (F(2,17) = 51.9, p < 0.001). This parameter was higher in males fed a HFD (F(2,8) = 63.2, p < 0.001) compared to those on chow.
Circulating leptin levels were affected by diet (F(2,35) = 19.8, p < 0.01; Table 1) with an interaction between sex and diet (F(2,35) = 4.3, p < 0.05). HFD increased plasma leptin levels in both males (F(2,18) = 16.3, p < 0.001) and females (F(2,16) = 5.5, p < 0.05).

2.2. Glucose Tolerance Test and Insulin Levels

Glycemia at sacrifice was affected by diet (F(2,53) = 7.6, p < 0.01), with an increase after HFD in both sexes (Table 1).
In the glucose tolerance test (Figure 2A), diet had an overall effect (F(2,34) = 7.9, p < 0.01) as did sex (F(1, 34) = 7.8, p < 0.01), with an interaction between these factors (F(2,34) = 3.3, p < 0.05) and a change over time (F(4,34) = 79.3, p < 0.001). Basal glycemia levels showed differences according to sex (F(1,34) = 18.9, p < 0.001) and diet (F(2,34) = 5.3, p < 0.05), with an interaction between these factors (F(2,34) = 3.2, p = 0.05). At baseline, there were sex differences when CCD was consumed (F(1,11) = 6.6, p < 0.05) as well as with HFD (F(1,11) = 30.1, p < 0.001), with males presenting a higher glycemia than females in both cases.
In males, HFD increased glycemia (F(2,17) = 7.0, p < 0.01). At 30 min (F(2,34) = 4.9, p < 0.05), 60 min (F(2,34) = 4.5, p < 0.05) and 90 min (F(2,34) = 6.6, p < 0.01) there was an effect of diet on the response to a glucose bolus with an increase in glycemia after HFD consumption. There was an effect of sex at 60 (F(1,34) = 4.5, p < 0.05) and 90 min (F(1,34) = 12.8, p < 0.01), with overall higher levels in males than females.
At 120 min, there was an effect of sex (F(1,34) = 9.5, p < 0.01) and diet (F(2,34) = 5.0, p < 0.05), and an interaction between these two factors (F(2,34) = 5.0, p < 0.05). There were differences between males and females on chow (F(1,10) = 5.3, p < 0.05) and HFD (F(1,11) = 12.1, p < 0.01), with males having increased glycemia in both cases. Males on a HFD had a higher glycemia compared to when consuming chow (F(2,17) = 6.3, p < 0.05).
The area under the curve (AUC; Figure 2B) was affected by sex (F(1,34) = 6.2, p < 0.05) and diet (F(2,34) = 7.6, p < 0.01), with the increase after HFD consumption being more apparent in males.
Plasma insulin levels (F(2,33) = 3.7, p < 0.05: Figure 2C) and the Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) (F(2,31) = 7.6, p < 0.01; Figure 2D) were affected by diet, with both HFD and CCD inducing an overall increase in these parameters, with the CCD effect on insulin being more apparent in females.

2.3. Circulating Levels of the IGF System

Free IGF1 levels were modified by diet (F(2,29) = 18.6, p < 0.001; Figure 3A), with an increase after HFD consumption in both sexes. Total IGF1 was affected by sex (F(1,34) = 19.0, p < 0.001; Figure 3B), with males having higher levels than females, and by diet (F(2,34) = 8.2, p < 0.01) with an increase after HFD intake in both sexes.
Circulating IGF2 levels were affected by sex (F(1,35) = 7.6, p < 0.05; Figure 3C), with females having overall higher levels than males, and diet (F(2,35) = 8.2, p < 0.01), with HFD inducing an overall increase in IGF2 levels. There was an effect of sex on plasma IGFBP2 levels (F(1,35) = 15.9, p < 0.001; Figure 3D), with males having higher levels of IGFBP2 than females. There was an interaction between sex and diet (F(2,35) = 5.1, p < 0.05) with sex differences in IGFBP2 levels after CCD consumption (F(1,11) = 19.6, p < 0.01). In males, there was an increase in IGFBP2 after CCD consumption (F(2,17) = 5.5, p = 0.05).
There was also an effect of sex (F(1,35) = 7.3, p < 0.05), with males having higher levels than females, and diet (F(2,35) = 9.5, p < 0.01) on plasma IGFBP3 levels (Figure 3E), with HFD increasing the levels of this binding protein in both sexes.

2.4. The IGF System in Visceral Adipose Tissue (VAT)

Relative IGF1 mRNA levels were modified by diet (F(2,33) = 4.5, p < 0.05; Figure 4A), with a decrease after HFD in males. There was an interaction between sex and diet regarding IGF2 mRNA levels (F(2,34) = 3.2, p = 0.05; Figure 4B). In males, relative IGF2 mRNA levels were decreased after HFD intake (F(2,16) = 4.9, p < 0.05), resulting in males having lower levels than females when on a HFD (F(1,10) = 11.9, p < 0.01). There was no effect of sex or diet on relative mRNA levels of IGF1R (Figure 4C), IGF2R (Figure 4D) or IGFBP2 (Figure 4E).
Relative PAPP-A mRNA levels were affected by both diet (F(2,34) = 5.3, p < 0.05; Figure 4F) and sex (F(1,34) = 12.1, p < 0.01), with an interaction between these factors (F(2,34) = 8.8, p < 0.01). In males, PAPP-A mRNA levels were reduced after HFD intake (F(2,16) = 7.7, p < 0.01), with no HFD effect in females resulting in males having lower levels when on a HFD (F(1,10) = 47.3, p < 0.001). Relative PAPP-A2 mRNA levels were only affected by sex (F(1,33) = 28.2, p < 0.001; Figure 4G), being higher in males than females.
Relative STC-1 mRNA levels were modified by diet (F(2,32) = 18.2, p < 0.001; Figure 4H) and sex (F(1,32) = 58.3, p < 0.001), with an interaction between these factors (F(2,32) = 33.8, p < 0.001). STC-1 mRNA levels were higher in males than females when on chow (F(1,10) = 52.8, p < 0.001) or a CCD (F(1,10) = 16.8, p < 0.01). HFD consumption led to decreased levels in males (F(2,15) = 25.8, p < 0.001) but increased in females (F(2,16) = 26.2, p < 0.001). In males, STC-1 mRNA levels were also decreased after CCD intake (F(2,15) = 25.8, p < 0.001). No significant differences in STC-2 mRNA levels were found (Figure 4I).

2.5. Hypothalamic Response to Dietary Change

2.5.1. The IGF System in the Hypothalamus

The hypothalamic levels of IGF1 mRNA were affected by diet (F(2,43) = 6.8, p < 0.01; Figure 5A), being increased after HFD consumption in both sexes. There was also an effect of diet on the hypothalamic mRNA levels of IGF2 (F(2,42) = 4.7, p < 0.05; Figure 5B), with an overall increase in response to HFD, with this being more apparent in females. Relative IGF1R mRNA levels were altered by diet (F(2,35) = 3.3, p = 0.05; Figure 5C) with an overall increase in response to the CCD. However, there were no effects of either sex or diet on relative IGF2R mRNA levels (Figure 5D).
Regarding IGFBP2 mRNA expression, despite the apparent increase after HFD consumption in females, there was no overall effect of sex or diet (Figure 5E). As previously reported [39], the relative mRNA levels of IGF2 and IGFBP2 showed a similar profile, with a significant positive correlation between these two factors (r = 0.843, p < 0.001; Figure 5F).
We observed no differences in the relative mRNA levels of IGFBP3 (Figure 5G) or IGFBP4 (Figure 5H). However, IGFBP5 mRNA levels were affected by diet (F(2,35) = 6.8, p < 0.01; Figure 5I) with an interaction between sex and diet (F(2,35) = 4.5, p < 0.05). When on a chow diet, females were found to have higher levels than males (F(1,11) = 13.1, p < 0.01). When separated by sex, CCD and HFD consumption were found to increase the mRNA levels of IGFBP5 in the hypothalamus in males (F(2,17) = 10.0, p < 0.01), with no significant effect of diet in females.
Relative hypothalamic mRNA levels of PAPP-A (Figure 5J), PAPP-A2 (Figure 5K), STC-1 (Figure 5L) and STC-2 (Figure 5M) were not affected by sex or diet.
IGFBP2 is proposed to be an antidiabetic factor, protecting against obesity onset [40,41]. In addition, taking into consideration that IGFBP2 has preference for IGF2 over IGF1 [9], we determined the statistical correlations between these factors in the circulation, VAT, and the hypothalamus with body weight and glycemia (Table 2). Plasma IGF2 was positively and IGFBP2 negatively correlated with body weight, but exclusively in males. No significant correlations were found with glycemia in either sex. In VAT, the only correlation observed was that relative IGF2 mRNA levels were negatively correlated with body weight in males. In the hypothalamus there was a positive correlation between IGF2 mRNA levels and both body weight and glycemia, but only in females. Hypothalamic IGFBP2 mRNA relative levels also correlated positively with both body weight and glycemia and again only in females.

2.5.2. Hypothalamic Neuropeptides

The relative mRNA levels of neuropeptide Y (NPY) (F(2,35) = 5.8, p < 0.01; Figure 6A) and Agouti-related protein (AgRP) (F(2,34) = 11.9, p < 0.001; Figure 6B) in the hypothalamus were affected by diet, with the mRNA levels of both orexigenic neuropeptides decreasing after HFD intake.
Proopiomelanocortin (POMC) mRNA levels (Figure 6C) were not altered by sex or diet, whereas relative cocaine and amphetamine regulated transcript (CART) expression was affected by diet (F(2,35) = 3.6, p < 0.05; Figure 6D), with HFD inducing an overall increase.

2.5.3. Gliosis and Hypothalamic Stress

We analyzed markers of gliosis and endoplasmic reticulum (ER) stress in the hypothalamus (Table 3). Gliosis (GFAP and Iba1) and ER stress markers (pJNK) were not altered in response to dietary intake (Table 3).

3. Discussion

As previously reported, long-term HFD consumption increased body weight, fat mass and circulating leptin levels in both sexes [42,43,44,45]. However, female mice needed more time before a significant weight gain was observed which is in concordance with previous observations in rodents [44,46], at least in young animals. Energy intake was also increased, particularly in females, with females on the HFD consuming more energy than males. Although both sexes gained significant weight, glucose tolerance was only affected in males, as previously shown after 16 weeks of HFD intake in C57/BL mice [45]. However, basal insulin levels and the HOMA index were increased in both sexes after HFD in agreement with previous studies [43].
Sex differences in the metabolic response to long-term dietary challenges have been previously reported [46,47,48]. Likewise, we and others have shown that the circulating IGF system also differs between male and female rodents, both at baseline [49,50] and in response to a short-term dietary change [39]. Male mice were found to have higher circulating levels of total IGF1, IGFBP2 and IGFBP3, whereas females have higher IGF2 levels, with no sex differences in free IGF1 levels, as previously reported in rats [50]. We previously reported higher levels of free and total IGF1, IGF2 and IGFBP3, but reduced IGFBP2 levels at baseline, in male compared to female rats [39]. The circulating IGF system has also been shown to differ between the sexes in humans [51,52]. Together, the sex differences in the growth hormone (GH)/IGF system in conjunction with sex steroids [53] underlie the differences in longitudinal growth between males and females.
In both male and female mice, the circulating IGF system was modified by HFD-induced obesity, similar to that observed in patients with obesity. In both sexes, circulating levels of free IGF1 [54], total IGF1, IGF2 [55,56,57] and IGFBP3 [58] are reported to be increased in humans and rodents with obesity, although some studies found no changes in total IGF1 in patients with obesity [54]. This general increase in the IGF system that is associated with obesity occurs despite GH hyposecretion, which contributes to adiposity [59,60]. Increased hepatic GH sensitivity most likely contributes to the maintenance of circulating IGF1 levels despite low GH secretion [61].
Rapid changes can be seen in the IGF system in response to a HFD even before weight gain is observed [39], suggesting that they may be caused by the diet per se. In contrast, the changes found here after long-term HFD consumption could be due to the modifications in overall nutritional status in addition to possible direct effects of the diet, whereas, in response to a short-term dietary change, the modifications observed in the IGF system were more abundant in response to CCD [39]—here, long-term effects were associated with HFD intake. This supports the hypothesis that changes associated with HFD intake are largely due to body weight gain.
Circulating IGFBP2, one of the most abundant IGFBPs in serum [62], is suggested to play a role in protection against HFD-induced obesity and diabetes onset [40,41]. A negative correlation between circulating IGFBP2 and body mass index (BMI) has been reported in obese patients [63] and, in children with obesity, IGFBP2 levels are reduced, whereas in anorexic adolescents they are increased [64]. We observed a negative correlation between IGFBP2 and body weight in male mice independently of the diet consumed, although the increase in IGFBP2 in male mice on a CCD was not associated with weight change. It is possible that changes in this binding protein are time-dependent and occur differently between males and females. Indeed, after one week of CCD IGFBP2 was decreased in female rats [39], while after a more long-term dietary change, this binding protein was not different from those on a chow diet. The augmented IGFBP2 in males after more long-term CCD could be an attempt to protect against the higher sucrose intake. The susceptibility to diet-induced obesity and its metabolic complications differs between the sexes [46,65] and this could be related to the differential changes in IGFBP2. However, more studies are clearly needed for a better comprehension of the role of IGFBP2 in metabolism.
IGF1 is important for adipocyte metabolism, maturation and differentiation [19,66,67]. In male C57 mice, IGF1 mRNA levels were reported to be decreased in adipocytes purified from perigonadal adipose tissue of obese compared to lean mice, despite the lack of change in IGF1 expression in lysates of this same tissue [68]. This could suggest that although IGF1 mRNA levels may be reduced in adipocytes, other cell types in adipose tissue, such as macrophages, could increase their expression of Igf1 such that global IGF1 mRNA levels in VAT do not change. Here, IGF1 mRNA levels were reduced in VAT after HFD in males, with the possibility that adipocytes in VAT decrease their IGF1 mRNA levels after HFD intake with the VAT macrophages or circulating IGF1 providing the source of IGF1 required to maintain adipocyte hypertrophy and hyperplasia [68]. This system is not altered in females, which are metabolically less affected by HFD consumption despite the increase in the VAT percentage observed in response to a HFD intake. If a longer exposition to this diet is required to alter it in females, or if this system is sex-dependent, deserves further study. As seen here, IGF2 mRNA levels in VAT were previously reported to be reduced in C57 mice in response to HFD-induced obesity; however in PWK mice, which are resistant to HFD-induced obesity, they are not [69]. These authors suggest that IGF2 has an anti-inflammatory effect on TNF-α-induced MCP-1 expression in adipocytes [69]. They also reported increased expression of IGF2R in VAT in HFD-exposed C57 mice [69], which was not found here possibly due to the shorter exposure to the HFD in our study.
During adipogenesis, IGFBP2 is the principal IGFBP produced by visceral white adipocytes, inhibiting both adipogenesis and lipogenesis [70], with levels reported to be reduced in obese ob/ob, db/db and high-fat-fed mice [71]. This reduction was accompanied by decreased IGFBP2 in circulation, suggesting that the decreased levels in VAT after HFD intake may lead to a decrease in circulating IGFBP2 levels [71]. Here we found no effect of HFD on IGFBP2 mRNA levels in VAT or circulating IGFBP2; again, it is possible that a longer exposition to a HFD may be needed.
In PAPP-A knockout mice, adipocyte size and lipid uptake are reduced in mesenteric fat, and visceral fat did not expand in PAPP-A KO mice exposed to a HFD, suggesting that the local cleavage of IGFBP4 is mandatory for adipocyte expansion [72]. Local free IGF1 levels are suggested to increase in adipose tissue in response to PAPP-A secretion in pregnant women [73], supporting the idea that IGF1 may contribute to the adipocyte hypertrophy associated with HFD intake through PAPP-A activity, and the reduction in these factors could be a negative feedback effect in attempt to block excess expansion. However, we found no effect of diet on PAPP-A2 expression, although males had significantly higher levels than females. Whether this protease is involved in sex differences in VAT function remains to be determined.
Both the CCD and HFD reduced STC-1 mRNA levels in male mice. In cultured adipocytes from male rats, STC-1 was shown to increase glucose uptake and the storage of triacylglycerol during the postprandial period [74]. Thus, this reduction in STC-1 mRNA levels may be a response conducted to control the lipid accumulation by VAT and this could possibly be induced by the higher amount of fat content in both the CCD and HFD compared to the chow diet. STC-2 mRNA levels were not altered in VAT, but the possible role of STCs regulating the IGFs availability in VAT, and potentially modifying the adipocyte size, deserves further research.
Hypothalamic IGF1 mRNA levels were increased after HFD consumption in both sexes, in agreement with our previous results in male mice after 7 weeks of HFD [75]. This increase could be a homeostatic mechanism to counteract the central effects of a HFD as increased central expression of IGF1 has been shown to improve glucose tolerance and insulin sensitivity [76]. Hypothalamic IGF2 mRNA levels were also increased after HFD, especially in females. Maternal HFD was shown to increase hypothalamic IGF2 mRNA levels in female Sprague Dawley rat offspring at postnatal day 10, with no changes in males [77], but little is known regarding central IGF2 in metabolism. In the adult brain, IGF2 promotes memory consolidation, neurogenesis and cognitive function [78] and it could be involved in structural changes induced by HFD and/or obesity, although this remains to be determined.
Hypothalamic levels of IGF2 and IGFBP2 were positively correlated, as previously reported in rats [39]. In females, both IGF2 and IGFBP2 were positively correlated with glycemia. A role of both IGFBP2 and IGF2 in glucose metabolism has been reported [41,79], but whether these factors participate in central glucose regulation remains unknown. The fact that females were metabolically less affected by long-term HFD consumption compared to males could suggest a possible metabolic effect of these factors at the level of the hypothalamus.
The role of IGFBP5 in metabolism has been poorly studied, particularly at the central level. Male IGFBP5-deficient mice are reported to have a greater body weight, impaired glucose tolerance at baseline and a larger increase in adipose tissue on a HFD compared to wild type [80]. This suggests that IGFBP5 is protective against obesity and glucose metabolism impairment, but the specific role of hypothalamic IGFPB5 in metabolic control is unknown. As this binding protein is increased after both HFD and CCD consumption in males one might speculate that it could be a protective response against these diets at the central level, and that this could be induced by the higher fat content in both of these diets compared to chow. No other members of the IGF system analyzed in the hypothalamus were affected by either diet or sex.
No evidence of gliosis or ER stress was found in the hypothalamus. Although various authors have reported both phenomena as well as central inflammation after 8 weeks of HFD consumption [81,82,83] or even earlier [81], other authors report no alterations in hypothalamic inflammation in male mice [84]. Thus, the causal relationship between hypothalamic inflammation and gliosis in weight gain is unclear [85]. In addition, the absence of gliosis observed in our study in response to HFD is accompanied by an increase in the gene expression of IGF1 and 2, which may play a neuroprotective role by reducing cell stress [27].
Diet composition is reported to be more important than caloric intake per se in determining NPY/AgRP neuron activity [86] and NPY and AgRP were reduced in the hypothalamus after HFD consumption in both sexes, as previously reported in male rats [75,87,88]. This is most likely related to the satiating properties of the HFD inducing a response that attempts to reduce the amount of energy consumed. However, in females this response is attenuated, at least regarding NPY, and this could underlie their higher energy intake compared to both HFD males and control females. Insulin decreases both NPY and AgRP expression in the hypothalamic neuronal line mHypoE-46 [89]. Considering the insulin-like actions of IGF1 and 2, it is possible that the rise in these factors with HFD intake contributes to the inhibition of orexigeneic neuropeptide expression. Administration of IGF icv has been shown to increase POMC mRNA levels in chicks [90], which was not observed here. This difference could be due to the difference in species. An overall effect of HFD to increase the anorexic neuropeptide CART was found, but no changes were observed in POMC expression. On the contrary, we previously found POMC mRNA levels to increase after HFD in male mice with no effect on CART relative expression [75], whereas other authors reported no significant changes in the hypothalamic POMC and CART mRNA expression in female mice on a long-term HFD [91], employing a different source of HFD (45% kcal from fat, 4.73 kcal/g). In our previous study [75], the exposition to HFD was for only 50 days instead of the 8 weeks here, which may explain some of these differences.
The CCD used here is what has been traditionally referred to as a control low-fat diet (LFD) as it was developed to be used as the control to the commercial HFD. However, it is now clear that these commercially developed control diets can also have metabolic effects as their nutrient composition differs from a normal chow diet. Here, the CCD did not modify body weight, but basal insulin levels and HOMA index were increased in both sexes. Although the caloric content is similar between the rodent chow and CCD, the amount of sucrose, 33% in CCD and 0.9% in chow, differs considerably. Dry sucrose or polycose consumption does not affect final body weight in rats [92,93], nor does consumption of a 33% sucrose solution, despite the metabolic alterations which are found [94,95]. However, these studies found fat mass and specific metabolic parameters to be modified. Thus, it is possible that the high sucrose composition in the CCD induces these observed metabolic alterations, again indicating that not only caloric intake, but dietary composition, is important.
The response of the IGF system to CCD intake is time- and sex-dependent, whereas, in female rats exposed to 1 week of CCD, IGFBP2 levels were reduced [39] and no changes were found in this sex after 8 weeks. In contrast, males showed no differences at one week of CCD intake [39] but had increased serum IGFBP2 levels after 8 weeks. This sex-specific response may be related to the protective role of IGFBP2 in glucose metabolism [40,41] as glucose metabolism is differently affected in males and females. Short- and a long-term HFD intake also differently affected IGF2 levels in males. Some authors suggest that an initial reduction in serum IGF2 levels may indicate a bad prognosis for weight gain [96], whereas longer HFD exposition leads to an increase in the circulating IGF2 as in obesity [56]. What is clear is that each diet has differential metabolic implications that are time- and sex-dependent.
In conclusion, long-term HFD intake modulates the IGF system in a tissue-dependent manner, which may respond to different tissue requirements, and suggests an active role of this system in the metabolic response to this dietary challenge. Moreover, most of the observed changes are sex-specific and this could be involved in the differential metabolic responses between males and females.

4. Materials and Methods

4.1. Ethical Statement

This study was designed following the European Communities Council Directive (2010/63/UE) and the Spanish Royal Decree 53/2013 concerning the protection of experimental animals. In addition, the study was approved by the Ethical Committee of Animal Experimentation of the Hospital Puerta de Hierro de Madrid and the Animal Welfare Organ of the Comunidad Autónoma de Madrid (07/346225.9/15, 9 April 2015). All care was taken to use the minimum number of animals.

4.2. Animals and Diets

In this study, 54 seven-week-old C57BL/6J mice (27 males and 27 females) were purchased from Charles River Laboratories. Upon arrival, animals were weighed and randomly distributed into cages according to sex (3 mice per cage) and allowed to acclimate for a week with free access to a normal chow diet and tap water. The mice were weighed again and randomly distributed into the different experimental groups. They were fed ad libitum with a high-fat diet (HFD; 62% kcal from fat, 18% kcal from proteins, 20% kcal from carbohydrates, 5.1 kcal/g, LabDiet), a commercial control diet (CCD; 10% kcal from fat, 18% kcal from proteins, 72% kcal from carbohydrates, 3.76 kcal/g, LabDiet) or standard rodent chow (6% kcal from fat, 17% kcal from proteins, 77% from carbohydrates, 3.41 kcal/g, Panlab) for 8 weeks, with free access to tap water throughout the study. This resulted in a total of 6 groups with nine animals per diet and sex (n = 9). Body weight and food intake were monitored each week. Animals were maintained at 22 ± 2 °C throughout the study.

4.3. Glucose Tolerance Test (GTT)

A week before sacrifice, six mice per group (n = 6) were fasted for 6 h prior to testing. They were weighed and then intraperitoneally injected with 2 mg of D-glucose per gram of body weight. Glycemia was determined in a drop of blood from the tail by using a Freestyle Optimum Neo glucometer (Abbott, Witney, UK). Glycemia was measured just prior to the injection (basal, 0 min) and at 30, 60, 90 and 120 min after the D-glucose injection.

4.4. Tissue Collection

Mice were weighed and fasted 12 h before sacrifice by decapitation, which took place between 9:00 and 11:00 am. A few days before sacrifice, sawdust from male cages was mixed in the females’ cage to equalize estrous cycle in females, which was determined by vaginal cytology at sacrifices. In total, 18.5% of the females were on proestrus phase, 77.8% were in estrus, 0% in metaestrus and 3.7% in diestrus. Trunk blood was collected in tubes containing a 0.5 M ethylenediaminetetraacetic acid (EDTA) solution. Samples were then centrifuged at 3000 rpm for 15 min at 4 °C and plasma was collected and aliquoted (to avoid freeze–thaw cycles) and stored at −80 °C until used. Perigonadal visceral adipose tissue (VAT) was dissected, weighted and frozen at −80 °C. The hypothalamus (rostrally limited by the optic chiasm and caudally by the mammillary bodies) was also dissected and kept at −80 °C until processed.

4.5. ELISA Assays

Plasma levels of free IGF1 (AnshLabs, Webster, TX, USA), total IGF1 (Mediagnost, Reutlingen, Germany), IGF2 (R&D Systems, Minneapolis, MN, USA), IGFBP2 (Millipore, Burlington, MA, USA), IGFBP3 (Mediagnost), insulin (Millipore) and leptin (Millipore) were measured following the manufacturers’ instructions. Absorbance was read by spectrophotometry (Tecan Infinite M200, Grödig, Austria).
Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) was calculated by using the following equation: HOMA-IR = glycemia (mmol/L) × insulin (mU/L)/22.5

4.6. RNA and Protein Extraction

An RNeasy Lipid Tissue Mini Kit (Qiagen, Hilden, Germany) was used for visceral adipose tissue RNA extraction according to manufacturer’s instructions, whereas an RNeasy Plus Mini Kit (Qiagen) was used following the manufacturer’s instructions for hypothalamic RNA isolation. Protein was extracted from the eluate after tissue lysis. The eluate was mixed with 4 volumes of cold acetone and stored overnight at −20 °C. The samples were centrifuged 10 min at 3000 rpm and the pellets containing the protein were resuspended in a CHAPS hydrate (Sigma-Aldrich, Saint Louis, MO, USA) solution, containing 7 M urea, 2 M thiourea, 4% CHAPS, 0.5% 1M Tris pH 8.8 in distilled water and stored at −80°C. For protein quantification, Protein Assay Dye Reagent Concentrate (Bio-Rad Laboratories, Hercules, CA, USA) was used to perform the Bradford assay.

4.7. Western Blotting

Depending on the expected signal and molecular weight of each target, 10 to 40 µg of protein were resolved on 8, 10 or 12% sodium dodecyl sulphate-denaturing polyacrylamide gels. After electrophoresis, proteins were transferred to a previously activated polyvinylidine difluoride (PVDF) membrane and then blocked with a Tris-buffered saline buffer containing 0.1% Tween 20 and 5% non-fat dried milk or bovine serum albumin (BSA) when phosphorylated proteins were assayed. Primary antibodies (Table 4) were diluted in the same buffer and incubated O/N with agitation at 4 °C. The next day, the corresponding horseradish peroxidase-conjugated secondary antibody was diluted in the same buffer and the membranes incubated for 1.5 h. Clarity Western ECL Substrate (Bio-Rad Laboratories, Hercules, CA, USA) was employed to visualize the peroxidase activity and the chemiluminescent signal was detected by using ImageQuant Las 4000 Software (GE Healthcare Life Sciences, Barcelona, Spain). To normalize for protein loading, GAPDH was used, as indicated.

4.8. Real-Time qPCR

For RT-qPCR, RNA (0.5–1 µg) was retro-transcribed to copy DNA (cDNA) by using an NZY First-Strand cDNA Synthesis Kit (NZYTech, Lisbon, Portugal). TaqMan probes of target genes (Table 5) were used for qPCR in a QuantStudio 3 Real-Time PCR System (Applied Biosystems, Carlsbad, CA, USA). Mouse glyceraldehyde 3-phosphate dehydrogenase (GAPDH) endogenous control (Applied Biosystems) was employed as the housekeeping gene, except on visceral adipose tissue, for which Ppia was chosen (Table 5). For the mathematical analysis, the ΔΔCT method was performed. Relative levels of expression are expressed as percentage of the chow male group.

4.9. Statistical Analysis

Statistics analyses were performed using SPSS 15.0 (SPSS Inc., Chicago, IL, USA) software. A two-way ANOVA was employed using the diet and sex factors. Weight gain and food intake over time were calculated by a repeated measures ANOVA, as well as the weekly data for these variables. For the glucose tolerance test, the area under the curve (AUC) was calculated by using GraphPad Prism 5 software (San Diego, CA, USA). Data are presented as the mean ± standard error of the mean (SEM). Graphs were made by GraphPad Prism 5 software. For the linear correlation between variables, a Pearson correlation coefficient was calculated. p < 0.05 was considered significant.

Author Contributions

Conceptualization: J.A.C., L.M.F. and J.A.; methodology: J.A.C., L.M.F. and J.A.; validation: S.G.-C. and L.M.F.; formal analysis: S.G.-C. and J.A.C.; investigation: S.G.-C., M.J.-H., P.R., A.F.-R. and V.B.; resources: J.A.C., L.F.M. and J.A.; data curation: S.G.-C. and J.A.C.; writing—original draft preparation: S.G.-C.; writing—review and editing: J.A.C., L.M.F. and J.A.; visualization: S.G.-C. and J.A.C.; supervision: L.M.F. and J.A.C.; project administration: J.A.C.; funding acquisition: J.A.C., L.M.F. and J.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by grants from the Spanish Ministry of Science and Innovation (BFU2017-82565-C21-R2 to J.A.C. and L.M.F.), Spanish Ministry of Education, Culture and Sports (university training grant PU13/00909 to A.F.-R.), Fondo de Investigación Sanitaria (PI1900166 to J.A.) and Fondos FEDER. Centro de Investigación Biomédica en Red Fisiopatología de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (J.A.).

Acknowledgments

We would like to thank Francisca Díaz and Sandra Canelles for their excellent technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yakar, S.; Rosen, C.J.; Beamer, W.G.; Ackert-Bicknell, C.L.; Wu, Y.; Liu, J.L.; Ooi, G.T.; Setser, J.; Frystyk, J.; Boisclair, Y.R.; et al. Circulating levels of IGF-1 directly regulate bone growth and density. J. Clin. Investig. 2002, 110, 771–781. [Google Scholar] [CrossRef] [PubMed]
  2. Lui, J.C.; Finkielstain, G.P.; Barnes, K.M.; Baron, J. An imprinted gene network that controls mammalian somatic growth is down-regulated during postnatal growth deceleration in multiple organs. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008, 295, R189–R196. [Google Scholar] [CrossRef] [PubMed]
  3. Bann, D.; Holly, J.M.; Lashen, H.; Hardy, R.; Adams, J.; Kuh, D.; Ong, K.K.; Ben-Shlomo, Y. Changes in insulin-like growth factor-I and -II associated with fat but not lean mass in early old age. Obesity 2015, 23, 692–698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Blum, W.F.; Ranke, M.B. Insulin-like growth factor binding proteins (IGFBPs) with special reference to IGFBP-3. Acta Paediatr. Scand. Suppl. 1990, 367, 55–62. [Google Scholar] [CrossRef] [PubMed]
  5. Baxter, R.C. Insulin-like growth factor (IGF)-binding proteins: Interactions with IGFs and intrinsic bioactivities. Am. J. Physiol. Endocrinol. Metab. 2000, 278, E967–E976. [Google Scholar] [CrossRef] [PubMed]
  6. Ranke, M.B. Insulin-like growth factor binding-protein-3 (IGFBP-3). Best Pract. Res. Clin. Endocrinol. Metab. 2015, 29, 701–711. [Google Scholar] [CrossRef]
  7. Probst-Hensch, N.M.; Steiner, J.H.; Schraml, P.; Varga, Z.; Zürrer-Härdi, U.; Storz, M.; Korol, D.; Fehr, M.K.; Fink, D.; Pestalozzi, B.C.; et al. IGFBP2 and IGFBP3 protein expressions in human breast cancer: Association with hormonal factors and obesity. Clin. Cancer Res. 2010, 16, 1025–1032. [Google Scholar] [CrossRef] [Green Version]
  8. Russo, V.C.; Azar, W.J.; Yau, S.W.; Sabin, M.A.; Werther, G.A. IGFBP-2: The dark horse in metabolism and cancer. Cytokine Growth Factor Rev. 2015, 26, 329–346. [Google Scholar] [CrossRef]
  9. Bach, L.A. IGF-binding proteins. J. Mol. Endocrinol. 2018, 61, T11–T28. [Google Scholar] [CrossRef] [Green Version]
  10. Lee, W.H.; Michels, K.M.; Bondy, C.A. Localization of insulin-like growth factor binding protein-2 messenger RNA during postnatal brain development: Correlation with insulin-like growth factors I and II. Neuroscience 1993, 53, 251–265. [Google Scholar] [CrossRef]
  11. Boney, C.M.; Moats-Staats, B.M.; Stiles, A.D.; D’Ercole, A.J. Expression of insulin-like growth factor-I (IGF-I) and IGF-binding proteins during adipogenesis. Endocrinology 1994, 135, 1863–1868. [Google Scholar] [CrossRef] [PubMed]
  12. Gude, M.F.; Frystyk, J.; Flyvbjerg, A.; Bruun, J.M.; Richelsen, B.; Pedersen, S.B. The production and regulation of IGF and IGFBPs in human adipose tissue cultures. Growth Horm. IGF Res. 2012, 22, 200–205. [Google Scholar] [CrossRef] [PubMed]
  13. Velloso, C.P. Regulation of muscle mass by growth hormone and IGF-I. Br. J. Pharmacol. 2008, 154, 557–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kiess, W.; Yang, Y.; Kessler, U.; Hoeflich, A. Insulin-like growth factor II (IGF-II) and the IGF-II/mannose-6-phosphate receptor: The myth continues. Horm. Res. 1994, 41, 66–73. [Google Scholar] [CrossRef]
  15. Overgaard, M.T.; Boldt, H.B.; Laursen, L.S.; Sottrup-Jensen, L.; Conover, C.A.; Oxvig, C. Pregnancy-associated plasma protein-A2 (PAPP-A2), a novel insulin-like growth factor-binding protein-5 proteinase. J. Biol. Chem. 2001, 276, 21849–21853. [Google Scholar] [CrossRef] [Green Version]
  16. Kløverpris, S.; Mikkelsen, J.H.; Pedersen, J.H.; Jepsen, M.R.; Laursen, L.S.; Petersen, S.V.; Oxvig, C. Stanniocalcin-1 Potently Inhibits the Proteolytic Activity of the Metalloproteinase Pregnancy-associated Plasma Protein-A. J. Biol. Chem. 2015, 290, 21915–21924. [Google Scholar] [CrossRef] [Green Version]
  17. Jepsen, M.R.; Kløverpris, S.; Mikkelsen, J.H.; Pedersen, J.H.; Füchtbauer, E.M.; Laursen, L.S.; Oxvig, C. Stanniocalcin-2 inhibits mammalian growth by proteolytic inhibition of the insulin-like growth factor axis. J. Biol. Chem. 2015, 290, 3430–3439. [Google Scholar] [CrossRef] [Green Version]
  18. Hennebry, A.; Oldham, J.; Shavlakadze, T.; Grounds, M.D.; Sheard, P.; Fiorotto, M.L.; Falconer, S.; Smith, H.K.; Berry, C.; Jeanplong, F.; et al. IGF1 stimulates greater muscle hypertrophy in the absence of myostatin in male mice. J. Endocrinol. 2017, 234, 187–200. [Google Scholar] [CrossRef] [Green Version]
  19. Scavo, L.M.; Karas, M.; Murray, M.; Leroith, D. Insulin-like growth factor-I stimulates both cell growth and lipogenesis during differentiation of human mesenchymal stem cells into adipocytes. J. Clin. Endocrinol. Metab. 2004, 89, 3543–3553. [Google Scholar] [CrossRef]
  20. Argente, J.; Caballo, N.; Barrios, V.; Muñoz, M.T.; Pozo, J.; Chowen, J.A.; Hernández, M. Disturbances in the growth hormone-insulin-like growth factor axis in children and adolescents with different eating disorders. Horm. Res. 1997, 48, 16–18. [Google Scholar] [CrossRef]
  21. Reinhardt, R.R.; Bondy, C.A. Insulin-like growth factors cross the blood-brain barrier. Endocrinology 1994, 135, 1753–1761. [Google Scholar] [CrossRef] [PubMed]
  22. Pan, W.; Kastin, A.J. Interactions of IGF-1 with the blood-brain barrier in vivo and in situ. Neuroendocrinology 2000, 72, 171–178. [Google Scholar] [CrossRef] [PubMed]
  23. Quesada, A.; Romeo, H.E.; Micevych, P. Distribution and localization Patterns of Estrogen Receptor-β and Insulin-Like Growth Factor-1 Receptors in Neurons and Glial Cells of the Female Rat Substantia Nigra. J. Comp. Neurol. 2007, 503, 198–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Labandeira-Garcia, J.L.; Costa-Besada, M.A.; Labandeira, C.M.; Villar-Cheda, B.; Rodríguez-Perez, A.I. Insulin-Like Growth Factor-1 and Neuroinflammation. Front. Aging Neurosci. 2017, 9, 365. [Google Scholar] [CrossRef] [PubMed]
  25. O’Kusky, J.R.; Ye, P.; D’Ercole, A.J. Insulin-like growth factor-I promotes neurogenesis and synaptogenesis in the hippocampal dentate gyrus during postnatal development. J. Neurosci. 2000, 20, 8435–8442. [Google Scholar] [CrossRef] [Green Version]
  26. Carro, E.; Torres-Aleman, I. The role of insulin and insulin-like growth factor I in the molecular and cellular mechanisms underlying the pathology of Alzheimer’s disease. Eur. J. Pharmacol. 2004, 490, 127–133. [Google Scholar] [CrossRef] [Green Version]
  27. Genis, L.; Dávila, D.; Fernandez, S.; Pozo-Rodrigálvarez, A.; Martínez-Murillo, R.; Torres-Aleman, I. Astrocytes require insulin-like growth factor I to protect neurons against oxidative injury. F1000 Res. 2014, 3, 28. [Google Scholar] [CrossRef]
  28. Hernandez-Garzón, E.; Fernandez, A.M.; Perez-Alvarez, A.; Genis, L.; Bascuñana, P.; Fernandez de la Rosa, R.; Delgado, M.; Angel Pozo, M.; Moreno, E.; McCormick, P.J.; et al. The insulin-like growth factor I receptor regulates glucose transport by astrocytes. Glia 2016, 64, 1962–1971. [Google Scholar] [CrossRef]
  29. Chen, D.Y.; Stern, S.A.; Garcia-Osta, A.; Saunier-Rebori, B.; Pollonini, G.; Bambah-Mukku, D.; Blitzer, R.D.; Alberini, C.M. A critical role for IGF-II in memory consolidation and enhancement. Nature 2011, 469, 491–497. [Google Scholar] [CrossRef]
  30. Myer, D.J.; Gurkoff, G.G.; Lee, S.M.; Hovda, D.A.; Sofroniew, M.V. Essential protective roles of reactive astrocytes in traumatic brain injury. Brain 2006, 129, 2761–2772. [Google Scholar] [CrossRef]
  31. Faulkner, J.R.; Herrmann, J.E.; Woo, M.J.; Tansey, K.E.; Doan, N.B.; Sofroniew, M.V. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J. Neurosci. 2004, 24, 2143–2155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Liddelow, S.; Guttenplan, K.; Clarke, L.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [PubMed]
  33. Buckman, L.B.; Thompson, M.M.; Lippert, R.N.; Blackwell, T.S.; Yull, F.E.; Ellacott, K.L. Evidence for a novel functional role of astrocytes in the acute homeostatic response to high-fat diet intake in mice. Mol. Metab. 2014, 4, 58–63. [Google Scholar] [CrossRef] [PubMed]
  34. Douglass, J.D.; Dorfman, M.D.; Fasnacht, R.; Shaffer, L.D.; Thaler, J.P. Astrocyte IKKβ/NF-κB signaling is required for diet-induced obesity and hypothalamic inflammation. Mol. Metab. 2017, 6, 366–373. [Google Scholar] [CrossRef] [PubMed]
  35. Iglesias, J.; Morales, L.; Barreto, G.E. Metabolic and Inflammatory Adaptation of Reactive Astrocytes: Role of PPARs. Mol. Neurobiol. 2017, 54, 2518–2538. [Google Scholar] [CrossRef] [PubMed]
  36. Komoly, S.; Hudson, L.D.; Webster, H.D.; Bondy, C.A. Insulin-like growth factor I gene expression is induced in astrocytes during experimental demyelination. Proc. Natl. Acad. Sci. USA 1992, 89, 1894–1898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Fernandez, A.M.; Garcia-Estrada, J.; Garcia-Segura, L.M.; Torres-Aleman, I. Insulin-like growth factor I modulates c-fos induction and astrocytosis in response to neurotoxic insult. Neuroscience 1996, 76, 117–122. [Google Scholar] [CrossRef] [Green Version]
  38. Ryu, B.R.; Ko, H.W.; Jou, I.; Noh, J.S.; Gwag, B.J. Phosphatidylinositol 3-kinase-mediated regulation of neuronal apoptosis and necrosis by insulin and IGF-I. J. Neurobiol. 1999, 39, 536–546. [Google Scholar] [CrossRef]
  39. Guerra-Cantera, S.; Frago, L.M.; Díaz, F.; Ros, P.; Jiménez-Hernaiz, M.; Freire-Regatillo, A.; Barrios, V.; Argente, J.; Chowen, J.A. Short-Term Diet Induced Changes in the Central and Circulating IGF Systems Are Sex Specific. Front. Endocrinol. 2020, 11, 513. [Google Scholar] [CrossRef]
  40. Hedbacker, K.; Birsoy, K.; Wysocki, R.W.; Asilmaz, E.; Ahima, R.S.; Farooqi, I.S.; Friedman, J.M. Antidiabetic effects of IGFBP2, a leptin-regulated gene. Cell Metab. 2010, 11, 11–22. [Google Scholar] [CrossRef] [Green Version]
  41. Wheatcroft, S.B.; Kearney, M.T.; Shah, A.M.; Ezzat, V.A.; Miell, J.R.; Modo, M.; Williams, S.C.; Cawthorn, W.P.; Medina-Gomez, G.; Vidal-Puig, A.; et al. IGF-binding protein-2 protects against the development of obesity and insulin resistance. Diabetes 2007, 56, 285–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Paruthiyil, S.; Hagiwara, S.I.; Kundassery, K.; Bhargava, A. Sexually dimorphic metabolic responses mediated by CRF2 receptor during nutritional stress in mice. Biol. Sex Differ. 2018, 9, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. de Wilde, J.; Smit, E.; Mohren, R.; Boekschoten, M.V.; de Groot, P.; van den Berg, S.A.; Bijland, S.; Voshol, P.J.; van Dijk, K.W.; de Wit, N.W.; et al. An 8-week high-fat diet induces obesity and insulin resistance with small changes in the muscle transcriptome of C57BL/6J mice. J. Nutrigenet. Nutr. 2009, 2, 280–291. [Google Scholar] [CrossRef]
  44. Ros, P.; Díaz, F.; Freire-Regatillo, A.; Argente-Arizón, P.; Barrios, V.; Argente, J.; Chowen, J.A. Sex Differences in Long-term Metabolic Effects of Maternal Resveratrol Intake in Adult Rat Offspring. Endocrinology 2020, 161, bqaa090. [Google Scholar] [CrossRef]
  45. Samuel, P.; Khan, M.A.; Nag, S.; Inagami, T.; Hussain, T. Angiotensin AT(2) receptor contributes towards gender bias in weight gain. PLoS ONE 2013, 8, e48425. [Google Scholar] [CrossRef] [PubMed]
  46. Pettersson, U.S.; Waldén, T.B.; Carlsson, P.O.; Jansson, L.; Phillipson, M. Female mice are protected against high-fat diet induced metabolic syndrome and increase the regulatory T cell population in adipose tissue. PLoS ONE 2012, 7, e46057. [Google Scholar] [CrossRef] [PubMed]
  47. Pinos, H.; Carrillo, B.; Díaz, F.; Chowen, J.A.; Collado, P. Differential vulnerability to adverse nutritional conditions in male and female rats: Modulatory role of estradiol during development. Front. Neuroendocrinol. 2018, 48, 13–22. [Google Scholar] [CrossRef]
  48. Chowen, J.A.; Freire-Regatillo, A.; Argente, J. Neurobiological characteristics underlying metabolic differences between males and females. Prog. Neurobiol. 2019, 176, 18–32. [Google Scholar] [CrossRef]
  49. Peshti, V.; Obolensky, A.; Nahum, L.; Kanfi, Y.; Rathaus, M.; Avraham, M.; Tinman, S.; Alt, F.W.; Banin, E.; Cohen, H.Y. Characterization of physiological defects in adult SIRT6-/- mice. PLoS ONE 2017, 12, e0176371. [Google Scholar] [CrossRef] [Green Version]
  50. Frystyk, J.; Grønbaek, H.; Skjaerbaek, C.; Flyvbjerg, A.; Orskov, H.; Baxter, R.C. Developmental changes in serum levels of free and total insulin-like growth factor I (IGF-I), IGF-binding protein-1 and -3, and the acid-labile subunit in rats. Endocrinology 1998, 139, 4286–4292. [Google Scholar] [CrossRef]
  51. Argente, J.; Barrios, V.; Pozo, J.; Muñoz, M.T.; Hervás, F.; Stene, M.; Hernández, M. Normative data for insulin-like growth factors (IGFs), IGF-binding proteins, and growth hormone-binding protein in a healthy Spanish pediatric population: Age- and sex-related changes. J. Clin. Endocrinol. Metab. 1993, 77, 1522–1528. [Google Scholar] [CrossRef] [PubMed]
  52. Waters, D.L.; Yau, C.L.; Montoya, G.D.; Baumgartner, R.N. Serum Sex Hormones, IGF-1, and IGFBP3 Exert a Sexually Dimorphic Effect on Lean Body Mass in Aging. J. Gerontol. A Biol. Sci. Med. Sci. 2003, 58, 648–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Borski, R.J.; Tsai, W.; DeMott-Friberg, R.; Barkan, A.L. Regulation of somatic growth and the somatotropic axis by gonadal steroids: Primary effect on insulin-like growth factor I gene expression and secretion. Endocrinology 1996, 137, 3253–3259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Nam, S.Y.; Lee, E.J.; Kim, K.R.; Cha, B.S.; Song, Y.D.; Lim, S.K.; Lee, H.C.; Huh, K.B. Effect of obesity on total and free insulin-like growth factor (IGF)-1, and their relationship to IGF-binding protein (BP)-1, IGFBP-2, IGFBP-3, insulin, and growth hormone. Int. J. Obes. Relat. Metab. Disord. 1997, 21, 355–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Doyle, S.L.; Donohoe, C.L.; Finn, S.P.; Howard, J.M.; Lithander, F.E.; Reynolds, J.V.; Pidgeon, G.P.; Lysaght, J. IGF-1 and its receptor in esophageal cancer: Association with adenocarcinoma and visceral obesity. Am. J. Gastroenterol. 2012, 107, 196–204. [Google Scholar] [CrossRef] [PubMed]
  56. Xuan, L.; Ma, J.; Yu, M.; Yang, Z.; Huang, Y.; Guo, C.; Lu, Y.; Yan, L.; Shi, S. Insulin-like growth factor 2 promotes adipocyte proliferation, differentiation and lipid deposition in obese type 2 diabetes. J. Transl. Sci. 2019, 6, 1–7. [Google Scholar] [CrossRef]
  57. Buchanan, C.M.; Phillips, A.R.; Cooper, G.J. Preptin derived from proinsulin-like growth factor II (proIGF-II) is secreted from pancreatic islet beta cells and enhances insulin secretion. Biochem. J. 2001, 360, 431–439. [Google Scholar] [CrossRef]
  58. Argente, J.; Caballo, N.; Barrios, V.; Pozo, J.; Muñoz, M.T.; Chowen, J.A.; Hernández, M. Multiple endocrine abnormalities of the growth hormone and insulin-like growth factor axis in prepubertal children with exogenous obesity: Effect of short- and long-term weight reduction. J. Clin. Endocrinol. Metab. 1997, 82, 2076–2083. [Google Scholar] [CrossRef] [Green Version]
  59. Vahl, N.; Klausen, I.; Christiansen, J.S.; Jørgensen, J.O. Growth hormone (GH) status is an independent determinant of serum levels of cholesterol and triglycerides in healthy adults. Clin. Endocrinol. 1999, 51, 309–316. [Google Scholar] [CrossRef]
  60. Berryman, D.E.; Glad, C.A.; List, E.O.; Johannsson, G. The GH/IGF-1 axis in obesity: Pathophysiology and therapeutic considerations. Nat. Rev. Endocrinol. 2013, 9, 346–356. [Google Scholar] [CrossRef]
  61. Lewitt, M.S.; Dent, M.S.; Hall, K. The Insulin-Like Growth Factor System in Obesity, Insulin Resistance and Type 2 Diabetes Mellitus. J. Clin. Med. 2014, 3, 1561–1574. [Google Scholar] [CrossRef] [PubMed]
  62. Clemmons, D.R.; Snyder, D.K.; Busby, W.H., Jr. Variables controlling the secretion of insulin-like growth factor binding protein-2 in normal human subjects. J. Clin. Endocrinol. Metab. 1991, 73, 727–733. [Google Scholar] [CrossRef] [PubMed]
  63. Yau, S.W.; Harcourt, B.E.; Kao, K.T.; Alexander, E.J.; Russo, V.C.; Werther, G.A.; Sabin, M.A. Serum IGFBP-2 levels are associated with reduced insulin sensitivity in obese children. Clin. Obes. 2018, 8, 184–190. [Google Scholar] [CrossRef] [PubMed]
  64. Barrios, V.; Buño, M.; Pozo, J.; Muñoz, M.T.; Argente, J. Insulin-like growth factor-binding protein-2 levels in pediatric patients with growth hormone deficiency, eating disorders and acute lymphoblastic leukemia. Horm. Res. 2000, 53, 221–227. [Google Scholar] [CrossRef] [PubMed]
  65. Hwang, L.L.; Wang, C.H.; Li, T.L.; Chang, S.D.; Lin, L.C.; Chen, C.P.; Chen, C.T.; Liang, K.C.; Ho, I.K.; Yang, W.S.; et al. Sex differences in high-fat diet-induced obesity, metabolic alterations and learning, and synaptic plasticity deficits in mice. Obesity 2010, 18, 463–469. [Google Scholar] [CrossRef]
  66. Wabitsch, M.; Hauner, H.; Heinze, E.; Teller, W.M. The role of growth hormone/insulin-like growth factors in adipocyte differentiation. Metabolism 1995, 44, 45–49. [Google Scholar] [CrossRef]
  67. Smith, P.J.; Wise, L.S.; Berkowitz, R.; Wan, C.; Rubin, C.S. Insulin-like growth factor-I is an essential regulator of the differentiation of 3T3-L1 adipocytes. J. Biol. Chem. 1988, 263, 9402–9408. [Google Scholar]
  68. Chang, H.R.; Kim, H.J.; Xu, X.; Ferrante, A.W., Jr. Macrophage and adipocyte IGF1 maintain adipose tissue homeostasis during metabolic stresses. Obesity 2016, 24, 172–183. [Google Scholar] [CrossRef] [Green Version]
  69. Morita, S.; Horii, T.; Kimura, M.; Arai, Y.; Kamei, Y.; Ogawa, Y.; Hatada, I. Paternal allele influences high fat diet-induced obesity. PLoS ONE 2014, 9, e85477. [Google Scholar] [CrossRef] [Green Version]
  70. Yau, S.W.; Russo, V.C.; Clarke, I.J.; Dunshea, F.R.; Werther, G.A.; Sabin, M.A. IGFBP-2 inhibits adipogenesis and lipogenesis in human visceral, but not subcutaneous, adipocytes. Int. J. Obes. 2015, 39, 770–781. [Google Scholar] [CrossRef]
  71. Li, Z.; Picard, F. Modulation of IGFBP2 mRNA expression in white adipose tissue upon aging and obesity. Horm. Metab. Res. 2010, 42, 787–791. [Google Scholar] [CrossRef] [PubMed]
  72. Conover, C.A.; Harstad, S.L.; Tchkonia, T.; Kirkland, J.L. Preferential impact of pregnancy-associated plasma protein-A deficiency on visceral fat in mice on high-fat diet. Am. J. Physiol. Endocrinol. Metab. 2013, 305, E1145–E1153. [Google Scholar] [CrossRef] [PubMed]
  73. Rojas-Rodriguez, R.; Lifshitz, L.M.; Bellve, K.D.; Min, S.Y.; Pires, J.; Leung, K.; Boeras, C.; Sert, A.; Draper, J.T.; Corvera, S.; et al. Human adipose tissue expansion in pregnancy is impaired in gestational diabetes mellitus. Diabetologia 2015, 58, 2106–2114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Sarapio, E.; De Souza, S.K.; Model, J.F.A.; Trapp, M.; Da Silva, R.S.M. Stanniocalcin-1 and -2 effects on glucose and lipid metabolism in white adipose tissue from fed and fasted rats. Can. J. Physiol. Pharmacol. 2019, 97, 916–923. [Google Scholar] [CrossRef]
  75. Baquedano, E.; Ruiz-Lopez, A.M.; Sustarsic, E.G.; Herpy, J.; List, E.O.; Chowen, J.A.; Frago, L.M.; Kopchick, J.J.; Argente, J. The absence of GH signaling affects the susceptibility to high-fat diet-induced hypothalamic inflammation in male mice. Endocrinology 2014, 155, 4856–4867. [Google Scholar] [CrossRef] [Green Version]
  76. Hong, H.; Cui, Z.Z.; Zhu, L.; Fu, S.P.; Rossi, M.; Cui, Y.H.; Zhu, B.M. Central IGF1 improves glucose tolerance and insulin sensitivity in mice. Nutr. Diabetes 2017, 7, 2. [Google Scholar] [CrossRef] [Green Version]
  77. Barrand, S.; Crowley, T.M.; Wood-Bradley, R.J.; De Jong, K.A.; Armitage, J.A. Impact of maternal high fat diet on hypothalamic transcriptome in neonatal Sprague Dawley rats. PLoS ONE 2017, 12, e0189492. [Google Scholar] [CrossRef] [Green Version]
  78. Iwamoto, T.; Ouchi, Y. Emerging evidence of insulin-like growth factor 2 as a memory enhancer: A unique animal model of cognitive dysfunction with impaired adult neurogenesis. Rev. Neurosci. 2014, 25, 559–574. [Google Scholar] [CrossRef]
  79. Uchimura, T.; Hollander, J.M.; Nakamura, D.S.; Liu, Z.; Rosen, C.J.; Georgakoudi, I.; Zeng, L. An essential role for IGF2 in cartilage development and glucose metabolism during postnatal long bone growth. Development 2017, 144, 3533–3546. [Google Scholar] [CrossRef] [Green Version]
  80. Gleason, C.E.; Ning, Y.; Cominski, T.P.; Gupta, R.; Kaestner, K.H.; Pintar, J.E.; Birnbaum, M.J. Role of insulin-like growth factor-binding protein 5 (IGFBP5) in organismal and pancreatic beta-cell growth. Mol. Endocrinol. 2010, 24, 178–192. [Google Scholar] [CrossRef] [Green Version]
  81. Thaler, J.P.; Yi, C.X.; Schur, E.A.; Guyenet, S.J.; Hwang, B.H.; Dietrich, M.O.; Zhao, X.; Sarruf, D.A.; Izgur, V.; Maravilla, K.R.; et al. Obesity is associated with hypothalamic injury in rodents and humans. J. Clin. Investig. 2012, 122, 153–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Valdearcos, M.; Robblee, M.M.; Benjamin, D.I.; Nomura, D.K.; Xu, A.W.; Koliwad, S.K. Microglia dictate the impact of saturated fat consumption on hypothalamic inflammation and neuronal function. Cell Rep. 2014, 9, 2124–2138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Lemus, M.B.; Bayliss, J.A.; Lockie, S.H.; Santos, V.V.; Reichenbach, A.; Stark, R.; Andrews, Z.B. A stereological analysis of NPY, POMC, Orexin, GFAP astrocyte, and Iba1 microglia cell number and volume in diet-induced obese male mice. Endocrinology 2015, 156, 1701–1713. [Google Scholar] [CrossRef] [PubMed]
  84. Baufeld, C.; Osterloh, A.; Prokop, S.; Miller, K.R.; Heppner, F.L. High-fat diet-induced brain region-specific phenotypic spectrum of CNS resident microglia. Acta Neuropathol. 2016, 132, 361–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Dorfman, M.D.; Thaler, J.P. Hypothalamic inflammation and gliosis in obesity. Curr. Opin. Endocrinol. Diabetes Obes. 2015, 22, 325–330. [Google Scholar] [CrossRef] [Green Version]
  86. Wei, W.; Pham, K.; Gammons, J.W.; Sutherland, D.; Liu, Y.; Smith, A.; Kaczorowski, C.C.; O’Connell, K.M. Diet composition, not calorie intake, rapidly alters intrinsic excitability of hypothalamic AgRP/NPY neurons in mice. Sci. Rep. 2015, 5, 16810. [Google Scholar] [CrossRef] [Green Version]
  87. Hassan, A.M.; Mancano, G.; Kashofer, K.; Fröhlich, E.E.; Matak, A.; Mayerhofer, R.; Reichmann, F.; Olivares, M.; Neyrinck, A.M.; Delzenne, N.M.; et al. High-fat diet induces depression-like behaviour in mice associated with changes in microbiome, neuropeptide Y, and brain metabolome. Nutr. Neurosci. 2019, 22, 877–893. [Google Scholar] [CrossRef] [Green Version]
  88. de Araujo, T.M.; Razolli, D.S.; Correa-da-Silva, F.; de Lima-Junior, J.C.; Gaspar, R.S.; Sidarta-Oliveira, D.; Victorio, S.C.; Donato, J., Jr.; Kim, Y.B.; Velloso, L.A. The partial inhibition of hypothalamic IRX3 exacerbates obesity. EBioMedicine 2019, 39, 448–460. [Google Scholar] [CrossRef] [Green Version]
  89. Mayer, C.M.; Belsham, D.D. Insulin directly regulates NPY and AgRP gene expression via the MAPK MEK/ERK signal transduction pathway in mHypoE-46 hypothalamic neurons. Mol. Cell Endocrinol. 2009, 307, 99–108. [Google Scholar] [CrossRef]
  90. Fujita, S.; Honda, K.; Yamaguchi, M.; Fukuzo, S.; Saneyasu, T.; Kamisoyama, H. Role of Insulin-like Growth Factor-1 in the Central Regulation of Feeding Behavior in Chicks. J. Poult. Sci. 2019, 56, 270–276. [Google Scholar] [CrossRef] [Green Version]
  91. Roepke, T.A.; Yasrebi, A.; Villalobos, A.; Krumm, E.A.; Yang, J.A.; Mamounis, K.J. Loss of ERα partially reverses the effects of maternal high-fat diet on energy homeostasis in female mice. Sci. Rep. 2017, 7, 6381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Sclafani, A. Carbohydrate-induced hyperphagia and obesity in the rat: Effects of saccharide type, form, and taste. Neurosci. Biobehav. Rev. 1987, 11, 155–162. [Google Scholar] [CrossRef]
  93. Sclafani, A.; Xenakis, S. Influence of diet form on the hyperphagia-promoting effect of polysaccharide in rats. Life Sci. 1984, 34, 1253–1259. [Google Scholar] [CrossRef]
  94. Fuente-Martín, E.; García-Cáceres, C.; Granado, M.; Sánchez-Garrido, M.A.; Tena-Sempere, M.; Frago, L.M.; Argente, J.; Chowen, J.A. Early postnatal overnutrition increases adipose tissue accrual in response to a sucrose-enriched diet. Am. J. Physiol. Endocrinol. Metab. 2012, 302, E1586–E1598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Fuente-Martín, E.; Granado, M.; García-Cáceres, C.; Sanchez-Garrido, M.A.; Frago, L.M.; Tena-Sempere, M.; Argente, J.; Chowen, J.A. Early nutritional changes induce sexually dimorphic long-term effects on body weight gain and the response to sucrose intake in adult rats. Metabolism 2012, 61, 812–822. [Google Scholar] [CrossRef]
  96. Sandhu, M.S.; Gibson, J.M.; Heald, A.H.; Dunger, D.B.; Wareham, N.J. Low circulating IGF-II concentrations predict weight gain and obesity in humans. Diabetes 2003, 52, 1403–1408. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Body weight progression (A) and accumulated weight gain in percentage (B) in male and female mice exposed to a high-fat diet (HFD), commercial control diet (CCD) or a chow diet for 8 weeks. *** p < 0.001; #: different from chow in the same sex, @: different between sexes on the same diet. n = 6.
Figure 1. Body weight progression (A) and accumulated weight gain in percentage (B) in male and female mice exposed to a high-fat diet (HFD), commercial control diet (CCD) or a chow diet for 8 weeks. *** p < 0.001; #: different from chow in the same sex, @: different between sexes on the same diet. n = 6.
Metabolites 10 00462 g001
Figure 2. Glucose tolerance test and insulin levels. Glucose levels over time (A), area under curve (B), plasma insulin levels (C) and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR; D) in male and female mice on a high-fat diet (HFD), commercial control diet (CCD) or a chow diet for 8 weeks. a: overall effect of sex, b: overall effect of diet, #: different from chow on the same sex, @: differences between sexes on the same diet, n = 6.
Figure 2. Glucose tolerance test and insulin levels. Glucose levels over time (A), area under curve (B), plasma insulin levels (C) and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR; D) in male and female mice on a high-fat diet (HFD), commercial control diet (CCD) or a chow diet for 8 weeks. a: overall effect of sex, b: overall effect of diet, #: different from chow on the same sex, @: differences between sexes on the same diet, n = 6.
Metabolites 10 00462 g002
Figure 3. Circulating levels of free IGF-1 (A), total IGF-1 (B), IGF2 (C), IGFBP2 (D) and IGFBP3 (E) in mice on a high-fat diet (HFD), commercial control diet (CCD) or chow diet for 8 weeks. *** p < 0.001, a: effect of sex, b: effect of diet, n = 6.
Figure 3. Circulating levels of free IGF-1 (A), total IGF-1 (B), IGF2 (C), IGFBP2 (D) and IGFBP3 (E) in mice on a high-fat diet (HFD), commercial control diet (CCD) or chow diet for 8 weeks. *** p < 0.001, a: effect of sex, b: effect of diet, n = 6.
Metabolites 10 00462 g003
Figure 4. Relative gene expression of the insulin-like growth factor (IGF) system in visceral adipose tissue: IGF1 (A), IGF2 (B), IGF1R (C), IGF2R (D), IGFBP2 (E), PAPP-A (F), PAPP-A2 (G), stanniocalcin (STC)-1 (H) and STC-2 (I) in mice on a high-fat diet (HFD), commercial control diet (CCD) or a chow diet for 8 weeks. * p < 0.05, *** p < 0.001, a: effect of sex, b: effect of diet, ns = non-significant, n = 6.
Figure 4. Relative gene expression of the insulin-like growth factor (IGF) system in visceral adipose tissue: IGF1 (A), IGF2 (B), IGF1R (C), IGF2R (D), IGFBP2 (E), PAPP-A (F), PAPP-A2 (G), stanniocalcin (STC)-1 (H) and STC-2 (I) in mice on a high-fat diet (HFD), commercial control diet (CCD) or a chow diet for 8 weeks. * p < 0.05, *** p < 0.001, a: effect of sex, b: effect of diet, ns = non-significant, n = 6.
Metabolites 10 00462 g004
Figure 5. Relative hypothalamic mRNA levels of the IGF system: IGF1 (A), IGF2 (B), IGF1R (C), IGF2R (D), IGFBP2 (E), IGFBP3 (G), IGFBP4 (H), IGFBP5 (I), PAPP-A (J), PAPP-A2 (K), STC-1 (L) and STC-2 (M). Correlation of relative hypothalamic mRNA levels of IGF2 and IGFBP2 (F). ** p < 0.01, b: effect of diet, ns = non-significant, HFD = high-fat diet, CCD = commercial control diet, n = 6.
Figure 5. Relative hypothalamic mRNA levels of the IGF system: IGF1 (A), IGF2 (B), IGF1R (C), IGF2R (D), IGFBP2 (E), IGFBP3 (G), IGFBP4 (H), IGFBP5 (I), PAPP-A (J), PAPP-A2 (K), STC-1 (L) and STC-2 (M). Correlation of relative hypothalamic mRNA levels of IGF2 and IGFBP2 (F). ** p < 0.01, b: effect of diet, ns = non-significant, HFD = high-fat diet, CCD = commercial control diet, n = 6.
Metabolites 10 00462 g005aMetabolites 10 00462 g005b
Figure 6. Relative mRNA levels of orexigenic and anorexigenic neuropeptides in the hypothalamus: Neuropeptide Y (NPY; A), Agouti-related protein (AgRP; B), proopiomelanocortin (POMC; C) and cocaine and amphetamine regulated transcript (CART; D). b: effect of diet, ns = non-significant. HFD = high-fat diet; CCD = commercial control diet, n = 6.
Figure 6. Relative mRNA levels of orexigenic and anorexigenic neuropeptides in the hypothalamus: Neuropeptide Y (NPY; A), Agouti-related protein (AgRP; B), proopiomelanocortin (POMC; C) and cocaine and amphetamine regulated transcript (CART; D). b: effect of diet, ns = non-significant. HFD = high-fat diet; CCD = commercial control diet, n = 6.
Metabolites 10 00462 g006
Table 1. Body weight, fat mass, glycemia, energy intake and circulating leptin levels in male and female mice exposed for 8 weeks to a high-fat diet (HFD), commercial control diet (CCD) or a chow diet. a: effect of sex, b: effect of diet, c: interaction between sex and diet, #: different from chow on the same sex, @: differences between sexes on the same diet, n = 6, except energy intake was n = 3 (number of cages/group).
Table 1. Body weight, fat mass, glycemia, energy intake and circulating leptin levels in male and female mice exposed for 8 weeks to a high-fat diet (HFD), commercial control diet (CCD) or a chow diet. a: effect of sex, b: effect of diet, c: interaction between sex and diet, #: different from chow on the same sex, @: differences between sexes on the same diet, n = 6, except energy intake was n = 3 (number of cages/group).
Chow MalesCCD MalesHFD
Males
Chow FemalesCCD FemalesHFD FemalesSignificance
Final body weight (g)24.6
± 0.5
26.4
± 0.3
35.2
± 1.0 #
19.6
± 0.3 @
20.3
± 0.3 @
24.2
± 1.5 #,@
a, p < 0.001
b, p < 0.001
c, p < 0.01
Visceral adipose tissue (%)1.16
± 0.07
1.22
± 0.14
4.31
± 0.28 #
0.62
± 0.07 @
0.99
± 0.12
2.19
± 0.46 #,@
a, p < 0.001
b, p < 0.001
c, p < 0.001
Subcutaneous adipose tissue (%)0.52
± 0.04
0.57
± 0.05
1.86
± 0.16 #
0.67
± 0.04 @
0.96
± 0.09 @
1.52
± 0.21 #,@
b, p < 0.001
c, p < 0.001
Glycemia (mg/dl)67.8
± 2.0
71.8
± 3.7
80.6
± 3.2
63.6
± 3.9
68.7
± 4.3
79.6
± 4.9
b, p < 0.01
Kcal/mouse/
day
11.7
± 0.1
10.9
± 0.8
13.8
± 0.5
10.0
± 0.2 @
9.9
± 0.2
24.9
± 0.4 #,@
a, p < 0.001
b, p < 0.001
c, p < 0.001
Kcal/mouse/
day/100 g
46.2
± 0.7
42.3
± 2.0
45.8
± 0.7
48.7
± 1.2
47.6
± 0.9
114.5
± 3.3 #,@
a, p < 0.001
b, p < 0.001
c, p < 0.001
Energy efficiency (%)0.68
± 0.02
0.96
± 0.11
1.84
± 0.06 #
0.56
± 0.02 @
0.50
± 0.05 @
0.45
± 0.07 @
a, p < 0.001
b, p < 0.001
c, p < 0.001
Leptin (ng/mL)0.77
± 0.28
0.65
± 0.21
10.05
± 2.10 #
1.06
± 0.34
1.22
± 0.49
4.52
± 1.43 #
b, p < 0.01
c, p < 0.05
Table 2. Linear correlation between circulating levels of IGF2 and IGFBP2, relative mRNA levels of IGF2 and IGFBP2 in visceral adipose tissue (VAT) and in the hypothalamus with body weight and glycemia in both sexes. * p < 0.05, ** p < 0.01, n = 6.
Table 2. Linear correlation between circulating levels of IGF2 and IGFBP2, relative mRNA levels of IGF2 and IGFBP2 in visceral adipose tissue (VAT) and in the hypothalamus with body weight and glycemia in both sexes. * p < 0.05, ** p < 0.01, n = 6.
Plasma IGF2Plasma IGFBP2VAT
IGF2 mRNA
VAT IGFBP2 mRNAHPT
IGF2 mRNA
HPT IGFBP2 mRNA
Body weight (males)0.661 **−0.470 *−0.523 *−0.3060.0880.090
Body weight (females)0.286−0.225−0.107−0.1320.579 **0.598 **
Glycemia (males)0.159−0.102−0.2180.075−0.0920.138
Glycemia (females)0.268−0.016−0.157−0.2930.570 **0.623 **
Table 3. Effects of 8 weeks on a high-fat diet (HFD), commercial control diet (CCD) or chow diet on glial structural protein (GFAPs: Glial fibrillary acidic protein, Iba1: Ionized calcium binding adaptor molecule 1) and endoplasmic reticulum (ER)-stress markers (pJNK: phosphorylated c-Jun N-terminal kinases). ns = non-significant, n = 6.
Table 3. Effects of 8 weeks on a high-fat diet (HFD), commercial control diet (CCD) or chow diet on glial structural protein (GFAPs: Glial fibrillary acidic protein, Iba1: Ionized calcium binding adaptor molecule 1) and endoplasmic reticulum (ER)-stress markers (pJNK: phosphorylated c-Jun N-terminal kinases). ns = non-significant, n = 6.
Chow MalesCCD MalesHFD MalesChow FemalesCCD FemalesHFD FemalesSignificance
GFAP100.0
± 2.1
115.6
± 6.8
107.4
± 9.5
103.9
± 7.6
97.9
± 3.0
103.3
± 4.5
ns
Iba1100.0
± 3.2
117.1
± 3.6
110.4
± 12.5
114.8
± 7.4
101.5
± 4.5
97.7
± 6.9
ns
pJNK100.0
± 10.8
84.0
± 11.5
71.6
± 5.2
107.5
± 18.3
86.7
± 13.9
95.3
± 14.1
ns
Table 4. Antibodies used for Western blotting.
Table 4. Antibodies used for Western blotting.
AntibodyClassDilutionHostCommercial SourceReference
pJNKPolyclonal1:3000RabbitPromegaV7932
GAPDHPolyclonal1:10,000RabbitSigma-AldrichG9545
GFAPPolyclonal1:5000Guinea pigSynaptic Systems173004
Iba1Polyclonal1:1000RabbitSynaptic Systems234003
α-guinea pig HRP-conjugatedPolyclonal1:2000GoatAbD SerotecAHP861P
α-rabbit HRP-conjugatedPolyclonal1:20,000GoatInvitrogen31460
Table 5. List of TaqMan probes used for qPCR.
Table 5. List of TaqMan probes used for qPCR.
NameGeneReferenceCommercial Source
Agouti-related proteinAgrpMm00475829_g1Applied Biosystems
Cyclophilin A (Peptidylprolyl isomerase A)PpiaMm02342430_g1Applied Biosystems
Cocaine and amphetamine regulated transcript prepropeptideCartptMm04210469_m1Applied Biosystems
Insulin-like growth factor 1Igf1Mm00439560_m1Applied Biosystems
Insulin-like growth factor 1 receptorIgf1rMm00802831_m1Applied Biosystems
Insulin-like growth factor 2Igf2Mm00439564_m1Applied Biosystems
Insulin-like growth factor 2 receptorIgf2rMm00439576_m1Applied Biosystems
Insulin-like growth factor-binding protein 2Igfbp2Mm00492632_m1Applied Biosystems
Insulin-like growth factor-binding protein 3Igfbp3Mm01187817_m1Applied Biosystems
Insulin-like growth factor-binding protein 4Igfbp4Mm00494922_m1Applied Biosystems
Insulin-like growth factor-binding protein 5Igfbp5Mm00516037_m1Applied Biosystems
Neuropeptide YNpyMm03048253_m1Applied Biosystems
Pregnancy-associated plasma protein APappaMm01259244_m1Applied Biosystems
Pregnancy-associated plasma protein A-2Pappa2Mm01284029_m1Applied Biosystems
Pro-opiomelanocortinPomcMm00435874_m1Applied Biosystems
Stanniocalcin-1Stc1Mm01322191_m1Applied Biosystems
Stanniocalcin-2Stc2Mm00441560_m1Applied Biosystems
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Guerra-Cantera, S.; Frago, L.M.; Jiménez-Hernaiz, M.; Ros, P.; Freire-Regatillo, A.; Barrios, V.; Argente, J.; Chowen, J.A. Impact of Long-Term HFD Intake on the Peripheral and Central IGF System in Male and Female Mice. Metabolites 2020, 10, 462. https://doi.org/10.3390/metabo10110462

AMA Style

Guerra-Cantera S, Frago LM, Jiménez-Hernaiz M, Ros P, Freire-Regatillo A, Barrios V, Argente J, Chowen JA. Impact of Long-Term HFD Intake on the Peripheral and Central IGF System in Male and Female Mice. Metabolites. 2020; 10(11):462. https://doi.org/10.3390/metabo10110462

Chicago/Turabian Style

Guerra-Cantera, Santiago, Laura M. Frago, María Jiménez-Hernaiz, Purificación Ros, Alejandra Freire-Regatillo, Vicente Barrios, Jesús Argente, and Julie A. Chowen. 2020. "Impact of Long-Term HFD Intake on the Peripheral and Central IGF System in Male and Female Mice" Metabolites 10, no. 11: 462. https://doi.org/10.3390/metabo10110462

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop