Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Oct 1.
Published in final edited form as: Front Neuroendocrinol. 2018 Jun 8;51:125–131. doi: 10.1016/j.yfrne.2018.06.002

Homeostatic sensing of dietary protein restriction: A case for FGF21

Cristal M Hill 1, Hans-Rudolf Berthoud 1, Heike Münzberg 1, Christopher D Morrison 1,*
PMCID: PMC6175661  NIHMSID: NIHMS978204  PMID: 29890191

Abstract

Restriction of dietary protein intake increases food intake and energy expenditure, reduces growth, and alters amino acid, lipid, and glucose metabolism. While these responses suggest that animals ‘sense’ insufficient consumption of amino acids, the basic physiological mechanism mediating the adaptive response to protein restriction has been largely undescribed. In this review we make the case that the liver-derived metabolic hormone FGF21 is the key signal which communicates and coordinates the homeostatic response to dietary protein restriction. Support for this model centers on the evidence that FGF21 is induced by settings of insufficient dietary protein or amino acid intake and is required for adaptive changes in metabolism and behavior. FGF21 occupies a unique endocrine niche, being induced when energy intake is adequate but protein and carbohydrate are imbalanced. Collectively, the evidence thus suggests that FGF21 is the first known endocrine signal of dietary protein restriction.

Keywords: macronutrient, FGF21, dietary protein, nutrition

1. Introduction

The ability to sense and respond to nutrient restriction is one of the most essential physiological functions in biology. Survival is dependent on the organism procuring sufficient nutrients to meet metabolic needs and doing so in a fashion that responds to changes in the external environment or internal physiology. However, free-feeding animals often face a complex nutritional landscape where individual food sources vary in macronutrient content, energy density, palatability, and availability. Effectively navigating this nutritional landscape requires choices that maximize nutrient intake while minimizing procurement cost. Although considerable progress has been made in understanding the neural regulation of feeding behavior, much of this work has focused on food intake in terms of total food (grams) or energy (calories). Yet it seems intuitive that we eat for more than just calories/energy. Roughly 6 years ago we summarized the evidence for the homeostatic regulation of dietary protein intake while also noting that the neuroendocrine mechanisms governing this behavior were largely undefined (Morrison et al., 2012). Here we provide an update by making the case that the metabolic hormone FGF21 is a key component in the homeostatic mechanism mediating responses to protein restriction.

2. Impact of Dietary Protein Restriction on Behavior and Metabolism

Consumption of essential amino acids is required for life, and regulatory systems exist to detect insufficient protein intake and coordinate adaptive changes in feeding behavior and metabolism. For instance, a large amount of evidence indicates that reduced dietary protein content increases total food intake across a range of species, while also increasing energy expenditure, reducing growth, and altering the expression of a variety of genes associated with amino acid, glucose and lipid metabolism within the liver and elsewhere (Berthoud et al., 2012; Morrison et al., 2012; Anthony et al., 2013; Davidenko et al., 2013; Ghosh et al., 2014; Gosby et al., 2014; Martens & Westerterp-Plantenga, 2014; Morrison & Laeger, 2015). Additional evidence suggests that variations in amino acid composition can also influence feeding behavior, such that animals increase their consumption of diets that are moderately restricted in one or a few amino acids, avoid diets that are severely imbalanced in AA composition, and selectively find and consume the missing amino acid when faced with multiple food options (Hrupka et al., 1997; Torii & Niijima, 2001; Anthony & Gietzen, 2013; Cummings et al., 2017; Wanders et al., 2017). Finally, the specific response is often dependent on the magnitude of the dietary restriction, with methionine restriction producing adaptive changes in food intake and metabolism within a specific ‘window’ of dietary restriction (Forney et al., 2017a; Forney et al., 2017b). Methionine intakes higher than this threshold producing no overt phenotype and methionine intakes below producing negative physiological outcomes that are more accurately described as deprivation.

In addition to changes in feeding behavior, early work by Rothwell and Stock demonstrated that increases in food intake during protein restriction were not accompanied by the predicted increase in body weight (Rothwell et al., 1982, 1983). More recent work provides clear evidence that protein restriction increases whole-body energy expenditure, both at room temperature and thermoneutrality, and that these effects are associated with activation of brown adipose tissue, increases in UCP1, and the browning of white fat (Rothwell & Stock, 1987; Laeger et al., 2016; Hill et al., 2017). Although the physiological reason for this increase in energy expenditure is unclear, it could be argued that the increase in energy expenditure allows the animal to burn off excess calories in an effort to procure the missing amino acids (Felicetti et al., 2003; Sorensen et al., 2008). However, we recently demonstrated that preventing hyperphagia in protein-restricted mice increased weight loss and did not block the increase in energy expenditure, while deletion of UCP1 blocked both the increase in both food intake and the increase in energy expenditure during protein restriction (Hill et al., 2017). Taken together, these data not only suggest that increased energy expenditure is independent of hyperphagia, but that increased energy expenditure may in fact drive food intake.

As expected, dietary protein restriction also influences metabolism within multiple tissues, including muscle, fat and liver. The liver in particular responds to reduced amino acid intake by inhibiting protein synthesis and amino acid catabolism and increasing amino acid biosynthesis (Anthony et al., 2004; Kilberg et al., 2005), effects which buffer against dramatic or persistent falls in circulating amino acids during protein restriction (Anderson et al., 1990; Kalhan et al., 2011; Laeger et al., 2014b). Protein restriction promotes other changes within the liver, altering the expression of genes associated with amino acid and lipid metabolism, enhancing insulin sensitivity, and increasing autophagy (Mortimore & Schworer, 1977; Guo & Cavener, 2007; Hasek et al., 2013; Stone et al., 2014; Fontana et al., 2016; Henagan et al., 2016; Maida et al., 2016; Cummings et al., 2017). The liver is also uniquely positioned to sense and respond to alterations in dietary protein content due to its direct sensing of absorbed amino acids via the portal circulation. The observations led our lab to focus on the possibility that the liver produced a signal that communicated protein status, and this effort eventually led to the link between FGF21 and dietary protein restriction (Laeger et al., 2014a).

3. Nutritional Regulation of Liver FGF21 Expression

FGF21 is a member of a large group of fibroblast growth factors that influence an array of physiological and cellular functions (Nishimura et al., 2000). While most FGFs act in a paracrine fashion, FGF21, FGF15/19 and FGF23 form a subgroup of ‘endocrine FGFs’ which circulate in appreciable amounts within the bloodstream and thereby act as true endocrine hormones (Angelin et al., 2012; Itoh et al., 2015). Initial interest in FGF21 stemmed from the discovery that FGF21 promoted glucose uptake in adipocytes (Kharitonenkov et al., 2005). Soon it was demonstrated that FGF21 treatment reduced body weight, glucose and lipid concentrations in models of obesity (Kharitonenkov et al., 2005; Kharitonenkov et al., 2007; Coskun et al., 2008), leading to substantial interest within both the basic research and pharmaceutical communities regarding FGF21’s potential as treatment for obesity/diabetes. Multiple prior reviews thoroughly cover the discovery of FGF21 and the early description of its metabolic effects (Potthoff et al., 2012; Kharitonenkov & Adams, 2014; Fisher & Maratos-Flier, 2016; Kharitonenkov & DiMarchi, 2017; Potthoff, 2017).

In addition to defining the pharmacological potential of FGF21, significant effort also focused on defining the physiological significance of FGF21. Early work in rodent models shaped the FGF21 narrative by demonstrating that circulating FGF21 is induced by fasting and ketogenic diets and principally produced by the liver, that PPARα was the key molecular regulator of FGF21 expression, and that FGF21 contributed to the adaptive responses to fasting (Badman et al., 2007; Inagaki et al., 2007; Badman et al., 2009; Potthoff et al., 2009; Markan et al., 2014). However, the effect of fasting and ketogenic diets to increase FGF21 is not nearly as robust in humans as initially observed in mice (Galman et al., 2008; Christodoulides et al., 2009; Dushay et al., 2010). FGF21 is also increased in settings of obesity (Zhang et al., 2008; Chavez et al., 2009; Dushay et al., 2010), which appears contradictory for a fasting hormone which lowers body weight and improves glucose homeostasis. The elevated FGF21 levels observed during obesity has led some to suggest that obesity is an FGF21 resistant state, and it has been demonstrated that the FGF21 co-receptor beta-Klotho (Klb) is significantly downregulated and FGF21 signaling attenuated in liver and fat of obese models (Fisher et al., 2010; Markan et al., 2017). However, subsequent studies indicate that FGF21’s effects on metabolic endpoints are not impaired in models of obesity (Hale et al., 2012; Laeger et al., 2017), and it has been speculated that loss of FGF21 signaling in white adipose tissue could represent an adaptive effort to shunt lipids to brown adipose during obesity (Schlein et al., 2016; Markan et al., 2017). Finally, several recent studies suggest that alcohol intake also increases FGF21 and that FGF21 inhibits alcohol intake (Schumann et al., 2016; Talukdar et al., 2016; Desai et al., 2017; Soberg et al., 2018; Song et al., 2018). The connection between liver FGF21 secretion and obesity, steatosis, alcohol consumption and metabolic stress has led to a broader view of FGF21 as a signal of metabolic or cellular stress (Maratos-Flier, 2017). Indeed, various manipulations which trigger cellular stress increase FGF21 production, even in tissues such as muscle which normally do not produce significant quantities of FGF21 (Kim et al., 2012; Brahma et al., 2014; Keipert et al., 2014; Guridi et al., 2015; Harris et al., 2015; Touvier et al., 2015; Vandanmagsar et al., 2016; Pereira et al., 2017b). These data collectively suggest that diverse signals are capable of regulating FGF21 production in liver and other tissues, but considerable debate remains regarding the relevance of these various inputs in a physiological context. In the next section we suggest that one key physiological effect of FGF21 is to signal macronutrient imbalance, particularly insufficient protein in the context of high energy/carbohydrate.

3.1. Increased FG21 in response to high carbohydrate.

The first connection between carbohydrate intake and FGF21 derived from a study demonstrating that refeeding with high carbohydrate diets induced a robust increase in liver and circulating FGF21 (Sanchez et al., 2009). This effect appeared to be specific for liver, as treatment of primary rat hepatocytes with glucose also increased FGF21 mRNA expression (Iizuka et al., 2009). However, the role of carbohydrate as a regulator of FGF21 gained significant momentum when it was demonstrated that fructose ingestion acutely increased circulating FGF21 in humans (Dushay et al., 2015). Multiple studies have since demonstrated that both acute ingestion of carbohydrate (generally sucrose, fructose or glucose), as well as longer-term exposure to high carbohydrate diets, leads to increases in both hepatic FGF21 mRNA expression and circulating FGF21 protein (Solon-Biet et al., 2016; von Holstein-Rathlou et al., 2016; Fisher et al., 2017; Iroz et al., 2017; Lundsgaard et al., 2017; Maekawa et al., 2017; Pereira et al., 2017a). The mechanism of this carbohydrate-induced increase in FGF21 in the liver appears to be primarily driven by carbohydrate response element binding protein (ChREBP). The FGF21 promoter contains ChREBP elements, carbohydrate intake induces ChREBP binding to the FGF21 promoter, and deletion of ChREBP blocks the carbohydrate-induced increase in FGF21 (Iizuka et al., 2009; von Holstein-Rathlou et al., 2016; Fisher et al., 2017; Iroz et al., 2017). Interestingly, the transcription factor PPARα is also required for glucose-induced increases in FGF21 expression, and available evidence highlights an important interaction between PPARα and ChREPB signaling in the context of FGF21 regulation (Iroz et al., 2017). Taken together, these data provide compelling evidence that dietary carbohydrates engage a specific transcriptional pathway to control FGF21 production.

3.2. Regulation of FGF21 by dietary protein restriction.

The identification of FGF21 as a signal of dietary protein restriction was initiated by De Sousa-Coelho and colleagues (De Sousa-Coelho et al., 2012). Their work demonstrated that the FGF21 promoter contained amino acid response elements (AARE) and that depletion of the amino acid leucine promoted binding of the transcription factor ATF4 to these AAREs, leading to robust increases in FGF21 expression in vitro and in vivo. Subsequent work demonstrated that FGF21 was increased by ER stress via the same ATF4-dependent pathway (Schaap et al., 2013; Jiang et al., 2014), thereby supporting ATF4 as an alternative mechanism for FGF21 regulation in the liver. ATF4 is a key molecular mediator of the classic integrated stress response (ISR), which coordinates the cellular response to various stressors, including amino acid depletion (Wek et al., 2006; Kilberg et al., 2009). This connection between FGF21, amino acid restriction and the ISR led our lab to hypothesize that perhaps the restriction of dietary protein contributed to the increases in FGF21 observed during fasting. Our work demonstrated that liver FGF21 expression and circulating FGF21 protein levels are increased by protein restriction in mice, rats and humans, and that this effect is independent of any change in energy intake (Laeger et al., 2014a). In fact, restricting energy intake without restricting protein decreased FGF21 expression. Subsequent work by multiple groups has confirmed that FGF21 is robustly increased by dietary protein restriction (Fournier et al., 2014; Ozaki et al., 2015; Chalvon-Demersay et al., 2016; Gosby et al., 2016; Maida et al., 2016; Pezeshki et al., 2016; Solon-Biet et al., 2016; Larson et al., 2017; Pereira et al., 2017a). This effect to increase FGF21 also extends to other situations of amino acid restriction, including the restriction of leucine, methionine/cysteine, the depletion asparagine, and the restriction of multiple non-essential amino acids (De Sousa-Coelho et al., 2012; Wanders et al., 2015; Wilson et al., 2015; Maida et al., 2016; Wanders et al., 2017). Contrastingly, available evidence suggests that the restriction of branched-chain amino acids may not increase FGF21, at least not persistently (Fontana et al., 2016; Cummings et al., 2017). Importantly, multiple groups have independently extended this observation to humans, suggesting that increases in FGF21 during dietary protein restriction is conserved across mammalian species (Laeger et al., 2014a; Fontana et al., 2016; Gosby et al., 2016; Maida et al., 2016).

Currently the mechanisms driving increases in liver FGF21 in response to protein restriction are not fully clear. Initial work implicated the classic integrated stress response pathway, particularly activation of the amino acid sensor GCN2, increased elF2a phosphorylation, the binding of ATF4 to the FGF21 promoter, and increases in FGF21 expression (De Sousa-Coelho et al., 2012). Indeed, deletion of GCN2 markedly attenuates the increase in FGF21 during protein restriction as well as in a model of asparagine depletion (Wilson et al., 2015; Laeger et al., 2016). However, GCN2-deficient mice appear to compensate over time, such that FGF21 expression eventually increases even in the absence of GCN2 (Laeger et al., 2016). Consistent with changes in FGF21 levels, the metabolic response to protein restriction is also impaired in GCN2-deficient mice for only the first few weeks of dietary protein restriction. The identity of this compensatory, GCN2-independent mechanism is currently unclear, but it seems possible that other components of the integrated stress response could contribute. For instance, PERK is known to promote FGF21 expression in response to classic ER stress signals and could potentially compensate for loss of GCN2 (Schaap et al., 2013), while other integrated stress response components such as Nupr1 and IRE1α-XBP1 have been implicated on FGF21 regulation (Jiang et al., 2014; Maida et al., 2016). GCN2 seems to be wholly unnecessary for increases in FGF21 in the context of methionine restriction, and while initial work suggested that PERK was activated, PERK is also dispensable for changes in FGF21 expression and metabolism during methionine restriction (Wanders et al., 2016; Pettit et al., 2017). Contrastingly, PPARα-deficient mice fail to increase FGF21 during protein restriction (Laeger et al., 2014a), and therefore PPARα is required for the effects of protein restriction, fasting and carbohydrate excess to increase FGF21 (Badman et al., 2007; Inagaki et al., 2007; Iroz et al., 2017). Although PPARα is required, there is no evidence that PPARα signaling is activated by protein or amino acid restriction (Ghosh et al., 2014; Laeger et al., 2014a), an outcome which suggests that PPARα likely plays a structural or constitutive role in FGF21 transcriptional activity during protein restriction, just as it appears to do in response to carbohydrate excess (Iroz et al., 2017). In summary, multiple lines of evidence suggest that protein restriction potently and persistently increases liver FGF21 expression and circulating FGF21 concentrations.

3.3. FGF21 as a signal of protein:carbohydrate imbalance

The above discussion suggests that FGF21 is not simply a fasting hormone, but is regulated by multiple nutritional inputs. One complication of many nutritional studies, including our own, is that protein and carbohydrate are often concomitantly altered to maintain a diet that is isocaloric with the control. As such, low protein diets are also marginally higher in carbohydrate, and it could be argued that this carbohydrate contributes to the increase in FGF21 induced by low protein diets. However, FGF21 is also increased by the restriction of individual amino acids acids (De Sousa-Coelho et al., 2012; Wanders et al., 2015; Wilson et al., 2015; Maida et al., 2016; Wanders et al., 2017; Cummings et al., 2018), and in this situation there is no change in carbohydrate content. High carbohydrate cannot explain the effect of ketogenic diets to increase FGF21, as ketogenic diets are virtually devoid of carbohydrate. Interestingly, several studies demonstrate that ketogenic diets increase FGF21 only when protein is reduced (Laeger et al., 2014a; Stemmer et al., 2015), and therefore dietary protein or amino acid restriction is sufficient to increase FGF21 regardless of the carbohydrate content of the diet. A similar argument could be made of pure sucrose/glucose/fructose/alcohol solutions, as these treatments are devoid of protein and therefore dilute protein intake. However, a recent study demonstrated that high carbohydrate diets increase FGF21 even when protein intake is controlled (Lundsgaard et al., 2017), and the specific, critical role played ChREBP in mediating the effects of glucose provides a specific mechanism linking carbohydrate intake to FGF21 expression.

Collectively these data suggest that FGF21 is independently regulated by multiple macronutrient inputs, and this conclusion is supported by a study using the geometric framework to define the macronutrient regulation of FGF21 in 858 mice eating 25 diets which varied in protein, carbohydrate, fat, and energy density (Solon-Biet et al., 2016). This study provides strong evidence that low protein and high carbohydrate independently increase FGF21, but energy intake itself does not influence FGF21. Taken together these observations suggest that FGF21 is primarily regulated by an imbalance between protein and carbohydrate intake, being particularly increased when protein intake is restricted and carbohydrate intake is excessive. FGF21 therefore responds to a nutritional state that is different from leptin and other energy balance signals. Two alternative nutritional scenarios can be envisioned which highlight these unique roles. The first scenario is one in which food is generally scarce, resulting in the restriction of energy intake. In this scenario, classic energy balance signals (leptin, insulin, ghrelin, etc.) govern metabolic and behavioral adaptions to dietary (energy) restriction. The second scenario is one in which low protein, high carbohydrate foods are readily available, but protein rich foods are scare. In this case energy balance signals are not engaged because energy intake is sufficient, but FGF21 is specifically increased because protein intake is low but carbohydrate intake is high. It is thus attractive to hypothesize that the induction of FGF21 in this state provides an endocrine mechanism to coordinate adaptive responses to imbalanced, protein-poor diets (Felicetti et al., 2003; Sorensen et al., 2008; Morrison et al., 2012). However, for this scenario to be relevant FGF21 must be physiologically required for adaptive responses to protein restriction.

4. FGF21 is essential for metabolic responses to protein restriction

It is generally accepted that physiological systems sense ‘nutrient restriction’ and engage adaptive mechanisms which both mitigate the consequences of restriction and promote a restoration of physiological function once food becomes available. Powerful examples exist for responses to the restriction of energy, sodium and water. While available evidence suggests that animals sense and respond to protein restriction, the primary mechanisms mediating the adaptive response to protein restriction is largely unknown (Morrison et al., 2012; Morrison & Laeger, 2015). However, work from our lab and others increasingly suggest that FGF21 is robustly increased by dietary protein restriction and required for adaptive responses to low protein diets.

The initial observations demonstrating FGF21’s required role stems from straightforward studies assessing the impact of low protein diets in FGF21-deficient mice. Protein restriction in mice produces an array of metabolic responses, including reduced body weight and adiposity and increased energy expenditure and food intake. These effects are lost in FGF21-deficient mice, whose growth rate is not reduced by the low protein diet (Laeger et al., 2014a). These data therefore indicate that FGF21 is the missing endocrine signal which coordinates adaptive behavioral and metabolic responses to dietary protein restriction. Subsequent work from multiple groups has replicated these core observations. Metabolic responses to protein restriction persist beyond 6 months of exposure, and FGF21 is fully required for this persistent response (Laeger et al., 2016). FGF21 also contributes to the effects of protein restriction on other metabolic endpoints, most notably improving insulin sensitivity in both diet and genetic models of obesity (Maida et al., 2016). The contribution of FGF21 to dietary protein restriction has also been extended to methionine restricting diets, where it was shown that FGF21 was required of the changes in energy expenditure and glucose homeostasis during MR, but not effects on lipid metabolism (Forney et al., 2017a; Wanders et al., 2017).

We have also indirectly validated the contribution of FGF21 via a separate set of studies focusing on the mechanism for FGF21 induction during protein restriction (Laeger et al., 2016). As described above, the amino acid sensor GCN2 is required for acute increases in FGF21 during protein restriction, such that GCN2-KO mice phenocopy FGF21-KO in their failure to respond to protein restriction, at least initially. However, GCN2-KO mice exhibit a delayed response to protein restriction, and this delayed metabolic response is explained by a delayed increase in FGF21 (Laeger et al., 2016). Because the response to protein restriction in GCN2-KO mice is tied to the increase in FGF21, this observation provides independent evidence that FGF21 is required for metabolic responses to protein restriction. Therefore, an increase in circulating FGF21 levels during protein restriction is required for the animal to ‘sense’ protein restriction and adapt metabolically and behaviorally, and this novel mechanism may provide a window into the biology underlying this robust but poorly understood response.

5. Biology of FGF21 action in the context of a low protein diet.

FGF21 is required for metabolic responses to protein restriction, but the mechanisms through which FGF21 produces these metabolic responses are less clear. More generally, understanding where and how FGF21 acts to regulate metabolic endpoints, both pharmacologically and physiologically, remains an important scientific question. While a full review of the FGF21 signal transduction cascade and the gamut of effects and sites of action implicated in the response to pharmacological FGF21 treatment is beyond the scope of this review, the growing consensus from this work suggests FGF21 largely acts through a heterodimer of FGFR1 and the co-receptor beta-Klotho (Klb) (Ding et al., 2012; Lee et al., 2018). Klb appears to be essential for the biological effects of FGF21, with adipose tissue and brain being key targets (Sarruf et al., 2010; Yang et al., 2012; Owen et al., 2013; Owen et al., 2014; BonDurant et al., 2017). Indeed, the brain has proven to be an essential mediator of FGF21 action in the context of pharmacological treatment (Sarruf et al., 2010; Bookout et al., 2013; Owen et al., 2014; von Holstein-Rathlou et al., 2016), although effects on adipose tissue or an interacting effect between multiple tissues remains a possibility (BonDurant et al., 2017). To date none of these questions have been rigorously tested within the context of protein restriction. Currently the mechanisms through which the brain senses and regulates macronutrient specific intake is poorly understood (Berthoud et al., 2012), but recent work has implicated FGF21 in this context by demonstrating that FGF21 acts to specifically reduce sweet taste (Talukdar et al., 2016; von Holstein-Rathlou et al., 2016) and alcohol consumption (Schumann et al., 2016; Talukdar et al., 2016). Similarly, genetic studies have linked FGF21 to macronutrient intake (Chu et al., 2013; Tanaka et al., 2013; Heianza et al., 2016; Schumann et al., 2016; Soberg et al., 2017). Finally, we contend that dietary protein restriction represents a valuable physiological context in which to test the mechanism of FGF21 action, both because it provides an alternative to studies using pharmacological doses/administration and because signaling of protein restriction represents an important physiological role for FGF21.

6. Conclusions and Future Directions

The above discussion highlights three main conclusions: First, dietary protein restriction triggers an array of adaptive responses that include changes in feeding behavior, energy expenditure, growth rate, and metabolism. Second, protein restriction produces a large increase in circulating levels of FGF21 within a range of mammalian species. Third, the metabolic effects of protein restriction appear, in large part, to require increased circulating FGF21. Collectively, these data provide strong evidence that FGF21 is the first known hormone that coordinates adaptive behavioral and metabolic responses to protein restriction. However, many unanswered questions remain. First, the specific mechanisms connecting dietary protein intake to FGF21 production remain unclear. For instance, are certain amino acids preferentially sensed or are all amino acids (essential and non-essential) treated equally, and what are the intracellular signaling events that drive increased FGF21 expression in the liver during protein restriction. It will also be important to define how these mechanisms transcriptionally interact with other nutritional signals that also engage FGF21. As noted earlier in this review, FGF21 is independently increased by high carbohydrate intake and low protein intake, and could therefore be more broadly labeled as a hormone signaling macronutrient imbalance. Second, if FGF21 indeed functions to coordinate adaptive responses to protein restriction/macronutrient imbalance, then how does FGF21 signaling interact with classic signals of energy balance such as leptin? To what extent do these ‘protein’ and ‘energy’ signals interact to control feeding behavior or metabolism? A third set of questions surround the mechanism through which FGF21 produces its metabolic effects in the context of dietary protein restriction. In which tissues (brain, fat, liver) does FGF21 primarily act; how does FGF21 signaling translate into changes in energy expenditure, feeding or glucose homeostasis; which of these effects are primary and which are secondary. A fourth set of questions surround the extent to which signals beyond FGF21 contribute to the overall adaptive response to protein restriction, as it seems unlikely that a single hormone mediates the broad and extensive effects that protein restriction induces. Finally, although evidence demonstrates that protein restriction also increases FGF21 in humans, the extent to which the specific metabolic effects produced by protein restriction in rodents translate to humans is an important question. Within the fields of obesity and metabolism it is almost axiomatic that adequate or even high protein diets are ‘healthy’, as they tend to reduce food intake and promote fat loss while also supporting the maintenance of lean mass. However, recent studies in flies and rodents suggest that high protein diets may have negative effects on insulin sensitivity and longevity while low protein diets exert metabolic benefits and extend lifespan (Grandison et al., 2009; Piper et al., 2011; Levine et al., 2014; Solon-Biet et al., 2014; Solon-Biet et al., 2015; Fontana et al., 2016; Cummings et al., 2017; Piper et al., 2017). These latter studies support the longstanding consensus that dietary restriction promotes healthspan and lifespan and suggest that the restriction of protein may be a contributing mechanism. Future studies are therefore needed to specifically define whether FGF21 contributes to these effects, and how these novel pathways can be leveraged to improve health in humans.

Highlights.

  1. Dietary protein restriction triggers adaptive changes in metabolism and behavior

  2. FGF21 is increased by excess carbohydrate and/or insufficient protein intake

  3. Adaptive responses to protein restriction require intact FGF21

Acknowledgments

Funding

This work was supported by the National Institutes of Health R01DK105032 to CDM and F32DK115137 to CMH.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Anderson SA, Tews JK & Harper AE, 1990. Dietary branched-chain amino acids and protein selection by rats. J Nutr. 120, 52–63. [DOI] [PubMed] [Google Scholar]
  2. Angelin B, Larsson TE & Rudling M, 2012. Circulating fibroblast growth factors as metabolic regulators-a critical appraisal. Cell Metab. 16, 693–705. [DOI] [PubMed] [Google Scholar]
  3. Anthony TG & Gietzen DW, 2013. Detection of amino acid deprivation in the central nervous system. Curr Opin Clin Nutr Metab Care. 16, 96–101. [DOI] [PubMed] [Google Scholar]
  4. Anthony TG, McDaniel BJ, Byerley RL, McGrath BC, Cavener DR, McNurlan MA & Wek RC, 2004. Preservation of liver protein synthesis during dietary leucine deprivation occurs at the expense of skeletal muscle mass in mice deleted for eIF2 kinase GCN2. J Biol Chem. 279, 36553–36561. [DOI] [PubMed] [Google Scholar]
  5. Anthony TG, Morrison CD & Gettys TW, 2013. Remodeling of lipid metabolism by dietary restriction of essential amino acids. Diabetes. 62, 2635–2644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Badman MK, Koester A, Flier JS, Kharitonenkov A & Maratos-Flier E, 2009. Fibroblast growth factor 21-deficient mice demonstrate impaired adaptation to ketosis. Endocrinology. 150, 4931–4940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Badman MK, Pissios P, Kennedy AR, Koukos G, Flier JS & Maratos-Flier E, 2007. Hepatic fibroblast growth factor 21 is regulated by PPARalpha and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab. 5, 426–437. [DOI] [PubMed] [Google Scholar]
  8. Berthoud HR, Munzberg H, Richards BK & Morrison CD, 2012. Neural and metabolic regulation of macronutrient intake and selection. The Proceedings of the Nutrition Society. 71, 390–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. BonDurant LD, Ameka M, Naber MC, Markan KR, Idiga SO, Acevedo MR, Walsh SA, Ornitz DM & Potthoff MJ, 2017. FGF21 Regulates Metabolism Through Adipose-Dependent and - Independent Mechanisms. Cell Metab. 25, 935–944 e934 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bookout AL, de Groot MH, Owen BM, Lee S, Gautron L, Lawrence HL, Ding X, Elmquist JK, Takahashi JS, Mangelsdorf DJ & Kliewer SA, 2013. FGF21 regulates metabolism and circadian behavior by acting on the nervous system. Nat Med. 19, 1147–1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Brahma MK, Adam RC, Pollak NM, Jaeger D, Zierler KA, Pocher N, Schreiber R, Romauch M, Moustafa T, Eder S, Ruelicke T, Preiss-Landl K, Lass A, Zechner R & Haemmerle G, 2014. Fibroblast growth factor 21 is induced upon cardiac stress and alters cardiac lipid homeostasis. Journal of lipid research. 55, 2229–2241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chalvon-Demersay T, Even PC, Tome D, Chaumontet C, Piedcoq J, Gaudichon C & Azzout-Marniche D, 2016. Low-protein diet induces, whereas high-protein diet reduces hepatic FGF21 production in mice, but glucose and not amino acids up-regulate FGF21 in cultured hepatocytes. J Nutr Biochem. 36, 60–67. [DOI] [PubMed] [Google Scholar]
  13. Chavez AO, Molina-Carrion M, Abdul-Ghani MA, Folli F, Defronzo RA & Tripathy D, 2009. 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. 32, 1542–1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Christodoulides C, Dyson P, Sprecher D, Tsintzas K & Karpe F, 2009. Circulating fibroblast growth factor 21 is induced by peroxisome proliferator-activated receptor agonists but not ketosis in man. J Clin Endocrinol Metab. 94, 3594–3601. [DOI] [PubMed] [Google Scholar]
  15. Chu AY, Workalemahu T, Paynter NP, Rose LM, Giulianini F, Tanaka T, Ngwa JS, Qi Q, Curhan GC, Rimm EB, Hunter DJ, Pasquale LR, Ridker PM, Hu FB, Chasman DI & Qi L, 2013. Novel locus including FGF21 is associated with dietary macronutrient intake. Hum Mol Genet. 22, 1895–1902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Coskun T, Bina HA, Schneider MA, Dunbar JD, Hu CC, Chen Y, Moller DE & Kharitonenkov A, 2008. Fibroblast growth factor 21 corrects obesity in mice. Endocrinology. 149, 6018–6027. [DOI] [PubMed] [Google Scholar]
  17. Cummings NE, Williams EM, Kasza I, Konon EN, Schaid MD, Schmidt BA, Poudel C, Sherman DS, Yu D, Arriola Apelo SI, Cottrell SE, Geiger G, Barnes ME, Wisinski JA, Fenske RJ, Matkowskyj KA, Kimple ME, Alexander CM, Merrins MJ & Lamming DW, 2017. Restoration of metabolic health by decreased consumption of branched-chain amino acids. J Physiol. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cummings NE, Williams EM, Kasza I, Konon EN, Schaid MD, Schmidt BA, Poudel C, Sherman DS, Yu D, Arriola Apelo SI, Cottrell SE, Geiger G, Barnes ME, Wisinski JA, Fenske RJ, Matkowskyj KA, Kimple ME, Alexander CM, Merrins MJ & Lamming DW, 2018. Restoration of metabolic health by decreased consumption of branched-chain amino acids. J Physiol. 596, 623–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Davidenko O, Darcel N, Fromentin G & Tome D, 2013. Control of protein and energy intake - brain mechanisms. Eur J Clin Nutr. 67, 455–461. [DOI] [PubMed] [Google Scholar]
  20. De Sousa-Coelho AL, Marrero PF & Haro D, 2012. Activating transcription factor 4-dependent induction of FGF21 during amino acid deprivation. Biochem J. 443, 165–171. [DOI] [PubMed] [Google Scholar]
  21. Desai BN, Singhal G, Watanabe M, Stevanovic D, Lundasen T, Fisher FM, Mather ML, Vardeh HG, Douris N, Adams AC, Nasser IA, FitzGerald GA, Flier JS, Skarke C & Maratos-Flier E, 2017. Fibroblast growth factor 21 (FGF21) is robustly induced by ethanol and has a protective role in ethanol associated liver injury. Molecular metabolism. 6, 1395–1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Ding X, Boney-Montoya J, Owen BM, Bookout AL, Coate KC, Mangelsdorf DJ & Kliewer SA, 2012. betaKlotho is required for fibroblast growth factor 21 effects on growth and metabolism. Cell Metab. 16, 387–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Dushay J, Chui PC, Gopalakrishnan GS, Varela-Rey M, Crawley M, Fisher FM, Badman MK, Martinez-Chantar ML & Maratos-Flier E, 2010. Increased fibroblast growth factor 21 in obesity and nonalcoholic fatty liver disease. Gastroenterology. 139, 456–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Dushay JR, Toschi E, Mitten EK, Fisher FM, Herman MA & Maratos-Flier E, 2015. Fructose ingestion acutely stimulates circulating FGF21 levels in humans. Molecular metabolism. 4, 51–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Felicetti LA, Robbins CT & Shipley LA, 2003. Dietary protein content alters energy expenditure and composition of the mass gain in grizzly bears (Ursus arctos horribilis). Physiol Biochem Zool. 76, 256–261. [DOI] [PubMed] [Google Scholar]
  26. Fisher FM, Chui PC, Antonellis PJ, Bina HA, Kharitonenkov A, Flier JS & Maratos-Flier E, 2010. Obesity is a fibroblast growth factor 21 (FGF21)-resistant state. Diabetes. 59, 2781–2789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Fisher FM, Kim M, Doridot L, Cunniff JC, Parker TS, Levine DM, Hellerstein MK, Hudgins LC, Maratos-Flier E & Herman MA, 2017. A critical role for ChREBP-mediated FGF21 secretion in hepatic fructose metabolism. Molecular metabolism. 6, 14–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Fisher FM & Maratos-Flier E, 2016. Understanding the Physiology of FGF21. Annu Rev Physiol. 78, 223–241. [DOI] [PubMed] [Google Scholar]
  29. Fontana L, Cummings NE, Arriola Apelo SI, Neuman JC, Kasza I, Schmidt BA, Cava E, Spelta F, Tosti V, Syed FA, Baar EL, Veronese N, Cottrell SE, Fenske RJ, Bertozzi B, Brar HK, Pietka T, Bullock AD, Figenshau RS, Andriole GL, Merrins MJ, Alexander CM, Kimple ME & Lamming DW, 2016. Decreased Consumption of Branched-Chain Amino Acids Improves Metabolic Health. Cell reports. 16, 520–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Forney LA, Stone KP, Wanders D & Gettys TW, 2017a. Sensing and signaling mechanisms linking dietary methionine restriction to the behavioral and physiological components of the response. Front Neuroendocrinol. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Forney LA, Wanders D, Stone KP, Pierse A & Gettys TW, 2017b. Concentration-dependent linkage of dietary methionine restriction to the components of its metabolic phenotype. Obesity (Silver Spring). 25, 730–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Fournier C, Rizzoli R & Ammann P, 2014. Low calcium-phosphate intakes modulate the low-protein diet-related effect on peak bone mass acquisition: a hormonal and bone strength determinants study in female growing rats. Endocrinology. 155, 4305–4315. [DOI] [PubMed] [Google Scholar]
  33. Galman C, Lundasen T, Kharitonenkov A, Bina HA, Eriksson M, Hafstrom I, Dahlin M, Amark P, Angelin B & Rudling M, 2008. The circulating metabolic regulator FGF21 is induced by prolonged fasting and PPARalpha activation in man. Cell Metab. 8, 169–174. [DOI] [PubMed] [Google Scholar]
  34. Ghosh S, Wanders D, Stone KP, Van NT, Cortez CC & Gettys TW, 2014. A systems biology analysis of the unique and overlapping transcriptional responses to caloric restriction and dietary methionine restriction in rats. FaSeB J. 28, 2577–2590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Gosby AK, Conigrave AD, Raubenheimer D & Simpson SJ, 2014. Protein leverage and energy intake. Obes Rev. 15, 183–191. [DOI] [PubMed] [Google Scholar]
  36. Gosby AK, Lau NS, Tam CS, Iglesias MA, Morrison CD, Caterson ID, Brand-Miller J, Conigrave AD, Raubenheimer D & Simpson SJ, 2016. Raised FGF-21 and Triglycerides Accompany Increased Energy Intake Driven by Protein Leverage in Lean, Healthy Individuals: A Randomised Trial. PLoS One. 11, e0161003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Grandison RC, Piper MD & Partridge L, 2009. Amino-acid imbalance explains extension of lifespan by dietary restriction in Drosophila. Nature. 462, 1061–1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Guo F & Cavener DR, 2007. The GCN2 eIF2alpha kinase regulates fatty-acid homeostasis in the liver during deprivation of an essential amino acid. Cell Metab. 5, 103–114. [DOI] [PubMed] [Google Scholar]
  39. Guridi M, Tintignac LA, Lin S, Kupr B, Castets P & Ruegg MA, 2015. Activation of mTORC1 in skeletal muscle regulates whole-body metabolism through FGF21. Sci Signal. 8, ra113. [DOI] [PubMed] [Google Scholar]
  40. Hale C, Chen MM, Stanislaus S, Chinookoswong N, Hager T, Wang M, Veniant MM & Xu J, 2012. Lack of overt FGF21 resistance in two mouse models of obesity and insulin resistance. Endocrinology. 153, 69–80. [DOI] [PubMed] [Google Scholar]
  41. Harris LA, Skinner JR, Shew TM, Pietka TA, Abumrad NA & Wolins NE, 2015. Perilipin 5-Driven Lipid Droplet Accumulation in Skeletal Muscle Stimulates the Expression of Fibroblast Growth Factor 21. Diabetes. 64, 2757–2768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Hasek BE, Boudreau A, Shin J, Feng D, Hulver M, Van NT, Laque A, Stewart LK, Stone KP, Wanders D, Ghosh S, Pessin JE & Gettys TW, 2013. Remodeling the integration of lipid metabolism between liver and adipose tissue by dietary methionine restriction in rats. Diabetes. 62, 3362–3372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Heianza Y, Ma W, Huang T, Wang T, Zheng Y, Smith SR, Bray GA, Sacks FM & Qi L, 2016. Macronutrient Intake-Associated FGF21 Genotype Modifies Effects of Weight-Loss Diets on 2-Year Changes of Central Adiposity and Body Composition: The POUNDS Lost Trial. Diabetes Care. 39, 1909–1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Henagan TM, Laeger T, Navard AM, Albarado D, Noland RC, Stadler K, Elks CM, Burk D & Morrison CD, 2016. Hepatic autophagy contributes to the metabolic response to dietary protein restriction. Metabolism. 65, 805–815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Hill CM, Laeger T, Albarado DC, McDougal DH, Berthoud H-R, Münzberg H & Morrison CD, 2017. Low protein-induced increases in FGF21 drive UCP1-dependent metabolic but not thermoregulatory endpoints. Scientific reports. 7, 8209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Hrupka BJ, Lin YM, Gietzen DW & Rogers QR, 1997. Small changes in essential amino acid concentrations alter diet selection in amino acid-deficient rats. J Nutr. 127, 777–784. [DOI] [PubMed] [Google Scholar]
  47. Iizuka K, Takeda J & Horikawa Y, 2009. Glucose induces FGF21 mRNA expression through ChREBP activation in rat hepatocytes. FEBS Lett. 583, 2882–2886. [DOI] [PubMed] [Google Scholar]
  48. Inagaki T, Dutchak P, Zhao G, Ding X, Gautron L, Parameswara V, Li Y, Goetz R, Mohammadi M, Esser V, Elmquist JK, Gerard RD, Burgess SC, Hammer RE, Mangelsdorf DJ & Kliewer SA, 2007. Endocrine regulation of the fasting response by PPARalpha-mediated induction of fibroblast growth factor 21. Cell Metab. 5, 415–425. [DOI] [PubMed] [Google Scholar]
  49. Iroz A, Montagner A, Benhamed F, Levavasseur F, Polizzi A, Anthony E, Regnier M, Fouche E, Lukowicz C, Cauzac M, Tournier E, Do-Cruzeiro M, Daujat-Chavanieu M, Gerbal-Chalouin S, Fauveau V, Marmier S, Burnol AF, Guilmeau S, Lippi Y, Girard J, Wahli W, Dentin R, Guillou H & Postic C, 2017. A Specific ChREBP and PPARalpha Cross-Talk Is Required for the Glucose-Mediated FGF21 Response. Cell reports. 21, 403–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Itoh N, Ohta H & Konishi M, 2015. Endocrine FGFs: Evolution, Physiology, Pathophysiology, and Pharmacotherapy. Front Endocrinol (Lausanne). 6, 154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Jiang S, Yan C, Fang QC, Shao ML, Zhang YL, Liu Y, Deng YP, Shan B, Liu JQ, Li HT, Yang L, Zhou J, Dai Z, Liu Y & Jia WP, 2014. Fibroblast growth factor 21 is regulated by the IRE1alpha-XBP1 branch of the unfolded protein response and counteracts endoplasmic reticulum stress-induced hepatic steatosis. J Biol Chem. 289, 29751–29765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Kalhan SC, Uppal SO, Moorman JL, Bennett C, Gruca LL, Parimi PS, Dasarathy S, Serre D & Hanson RW, 2011. Metabolic and genomic response to dietary isocaloric protein restriction in the rat. J Biol Chem. 286, 5266–5277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Keipert S, Ost M, Johann K, Imber F, Jastroch M, van Schothorst EM, Keijer J & Klaus S, 2014. Skeletal muscle mitochondrial uncoupling drives endocrine cross-talk through the induction of FGF21 as a myokine. Am J Physiol Endocrinol Metab. 306, E469–482. [DOI] [PubMed] [Google Scholar]
  54. Kharitonenkov A & Adams AC, 2014. Inventing new medicines: The FGF21 story. Molecular metabolism. 3, 221–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Kharitonenkov A & DiMarchi R, 2017. Fibroblast growth factor 21 night watch: advances and uncertainties in the field. J Intern Med. 281, 233–246. [DOI] [PubMed] [Google Scholar]
  56. Kharitonenkov A, Shiyanova TL, Koester A, Ford AM, Micanovic R, Galbreath EJ, Sandusky GE, Hammond LJ, Moyers JS, Owens RA, Gromada J, Brozinick JT, Hawkins ED, Wroblewski VJ, Li DS, Mehrbod F, Jaskunas SR & Shanafelt AB, 2005. FGF-21 as a novel metabolic regulator. J Clin Invest. 115, 1627–1635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Kharitonenkov A, Wroblewski VJ, Koester A, Chen YF, Clutinger CK, Tigno XT, Hansen BC, Shanafelt AB & Etgen GJ, 2007. The metabolic state of diabetic monkeys is regulated by fibroblast growth factor-21. Endocrinology. 148, 774–781. [DOI] [PubMed] [Google Scholar]
  58. Kilberg MS, Pan YX, Chen H & Leung-Pineda V, 2005. Nutritional control of gene expression: how mammalian cells respond to amino acid limitation. Annu Rev Nutr. 25, 59–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Kilberg MS, Shan J & Su N, 2009. ATF4-dependent transcription mediates signaling of amino acid limitation. Trends Endocrinol Metab. 20, 436–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kim KH, Jeong YT, Oh H, Kim SH, Cho JM, Kim YN, Kim SS, Kim DH, Hur KY, Kim HK, Ko T, Han J, Kim HL, Kim J, Back SH, Komatsu M, Chen H, Chan DC, Konishi M, Itoh N, Choi CS & Lee MS, 2012. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat Med. [DOI] [PubMed] [Google Scholar]
  61. Laeger T, Albarado DC, Burke SJ, Trosclair L, Hedgepeth JW, Berthoud HR, Gettys TW, Collier JJ, Munzberg H & Morrison CD, 2016. Metabolic Responses to Dietary Protein Restriction Require an Increase in FGF21 that Is Delayed by the Absence of GCN2. Cell reports. 16, 707–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Laeger T, Baumeier C, Wilhelmi I, Wurfel J, Kamitz A & Schurmann A, 2017. FGF21 improves glucose homeostasis in an obese diabetes-prone mouse model independent of body fat changes. Diabetologia. 60, 2274–2284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Laeger T, Henagan TM, Albarado DC, Redman LM, Bray GA, Noland RC, Munzberg H, Hutson SM, Gettys TW, Schwartz MW & Morrison CD, 2014a. FGF21 is an endocrine signal of protein restriction. J Clin Invest. 124, 3913–3922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Laeger T, Reed SD, Henagan TM, Fernandez DH, Taghavi M, Addington A, Munzberg H, Martin RJ, Hutson SM & Morrison CD, 2014b. Leucine acts in the brain to suppress food intake but does not function as a physiological signal of low dietary protein. Am J Physiol Regul Integr Comp Physiol. 307, R310–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Larson KR, Russo KA, Fang Y, Mohajerani N, Goodson ML & Ryan KK, 2017. Sex Differences in the Hormonal and Metabolic Response to Dietary Protein Dilution. Endocrinology. 158, 3477–3487. [DOI] [PubMed] [Google Scholar]
  66. Lee S, Choi J, Mohanty J, Sousa LP, Tome F, Pardon E, Steyaert J, Lemmon MA, Lax I & Schlessinger J, 2018. Structures of beta-klotho reveal a ‘zip code’-like mechanism for endocrine FGF signalling. Nature. 553, 501–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Levine ME, Suarez JA, Brandhorst S, Balasubramanian P, Cheng CW, Madia F, Fontana L, Mirisola MG, Guevara-Aguirre J, Wan J, Passarino G, Kennedy BK, Wei M, Cohen P, Crimmins EM & Longo VD, 2014. 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. 19, 407–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Lundsgaard AM, Fritzen AM, Sjoberg KA, Myrmel LS, Madsen L, Wojtaszewski JF, Richter EA & Kiens B, 2017. Circulating FGF21 in humans is potently induced by short term overfeeding of carbohydrates. Molecular metabolism. 6, 22–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Maekawa R, Seino Y, Ogata H, Murase M, Iida A, Hosokawa K, Joo E, Harada N, Tsunekawa S, Hamada Y, Oiso Y, Inagaki N, Hayashi Y & Arima H, 2017. Chronic high-sucrose diet increases fibroblast growth factor 21 production and energy expenditure in mice. J Nutr Biochem. 49, 71–79. [DOI] [PubMed] [Google Scholar]
  70. Maida A, Zota A, Sjoberg KA, Schumacher J, Sijmonsma TP, Pfenninger A, Christensen MM, Gantert T, Fuhrmeister J, Rothermel U, Schmoll D, Heikenwalder M, Iovanna JL, Stemmer K, Kiens B, Herzig S & Rose AJ, 2016. A liver stress-endocrine nexus promotes metabolic integrity during dietary protein dilution. J Clin Invest. 126, 3263–3278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Maratos-Flier E, 2017. Fatty liver and FGF21 physiology. Exp Cell Res. 360, 2–5. [DOI] [PubMed] [Google Scholar]
  72. Markan KR, Naber MC, Ameka MK, Anderegg MD, Mangelsdorf DJ, Kliewer SA, Mohammadi M & Potthoff MJ, 2014. Circulating FGF21 is Liver Derived and Enhances Glucose Uptake During Refeeding and Overfeeding. Diabetes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Markan KR, Naber MC, Small SM, Peltekian L, Kessler RL & Potthoff MJ, 2017. FGF21 resistance is not mediated by downregulation of beta-klotho expression in white adipose tissue. Molecular metabolism. 6, 602–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Martens EA & Westerterp-Plantenga MS, 2014. Protein diets, body weight loss and weight maintenance. Curr Opin Clin Nutr Metab Care. 17, 75–79. [DOI] [PubMed] [Google Scholar]
  75. Morrison CD & Laeger T, 2015. Protein-dependent regulation of feeding and metabolism. Trends Endocrinol Metab. 26, 256–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Morrison CD, Reed SD & Henagan TM, 2012. Homeostatic regulation of protein intake: in search of a mechanism. Am J Physiol Regul Integr Comp Physiol. 302, R917–928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Mortimore GE & Schworer CM, 1977. Induction of autophagy by amino-acid deprivation in perfused rat liver. Nature. 270, 174–176. [DOI] [PubMed] [Google Scholar]
  78. Nishimura T, Nakatake Y, Konishi M & Itoh N, 2000. Identification of a novel FGF, FGF-21, preferentially expressed in the liver. Biochim Biophys Acta. 1492, 203–206. [DOI] [PubMed] [Google Scholar]
  79. Owen BM, Bookout AL, Ding X, Lin VY, Atkin SD, Gautron L, Kliewer SA & Mangelsdorf DJ, 2013. FGF21 contributes to neuroendocrine control of female reproduction. Nat Med. 19, 1153–1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Owen BM, Ding X, Morgan DA, Coate KC, Bookout AL, Rahmouni K, Kliewer SA & Mangelsdorf DJ, 2014. FGF21 Acts Centrally to Induce Sympathetic Nerve Activity, Energy Expenditure, and Weight Loss. Cell Metab. 20, 670–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Ozaki Y, Saito K, Nakazawa K, Konishi M, Itoh N, Hakuno F, Takahashi S, Kato H & Takenaka A, 2015. Rapid increase in fibroblast growth factor 21 in protein malnutrition and its impact on growth and lipid metabolism. Br J Nutr. 114, 1410–1418. [DOI] [PubMed] [Google Scholar]
  82. Pereira MP, Ferreira LAA, da Silva FHS, Christoffolete MA, Metsios GS, Chaves VE, de Franca SA, Damazo AS, Flouris AD & Kawashita NH, 2017a. A low-protein, highcarbohydrate diet increases browning in perirenal adipose tissue but not in inguinal adipose tissue. Nutrition. 42, 37–45. [DOI] [PubMed] [Google Scholar]
  83. Pereira RO, Tadinada SM, Zasadny FM, Oliveira KJ, Pires KMP, Olvera A, Jeffers J, Souvenir R, McGlauflin R, Seei A, Funari T, Sesaki H, Potthoff MJ, Adams CM, Anderson EJ & Abel ED, 2017b. OPA1 deficiency promotes secretion of FGF21 from muscle that prevents obesity and insulin resistance. EMBO J. 36, 2126–2145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Pettit AP, Jonsson WO, Bargoud AR, Mirek ET, Peelor FF 3rd, Wang Y, Gettys TW, Kimball SR, Miller BF, Hamilton KL, Wek RC & Anthony TG, 2017. Dietary Methionine Restriction Regulates Liver Protein Synthesis and Gene Expression Independently of Eukaryotic Initiation Factor 2 Phosphorylation in Mice. J Nutr. 147, 1031–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Pezeshki A, Zapata RC, Singh A, Yee NJ & Chelikani PK, 2016. Low protein diets produce divergent effects on energy balance. Scientific reports. 6, 25145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Piper MD, Partridge L, Raubenheimer D & Simpson SJ, 2011. Dietary restriction and aging: a unifying perspective. Cell Metab. 14, 154–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Piper MD, Soultoukis GA, Blanc E, Mesaros A, Herbert SL, Juricic P, He X, Atanassov I, Salmonowicz H, Yang M, Simpson SJ, Ribeiro C & Partridge L, 2017. Matching Dietary Amino Acid Balance to the In Silico-Translated Exome Optimizes Growth and Reproduction without Cost to Lifespan. Cell Metab. 25, 610–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Potthoff MJ, 2017. FGF21 and metabolic disease in 2016: A new frontier in FGF21 biology. Nature reviews Endocrinology. 13, 74–76. [DOI] [PubMed] [Google Scholar]
  89. Potthoff MJ, Inagaki T, Satapati S, Ding X, He T, Goetz R, Mohammadi M, Finck BN, Mangelsdorf DJ, Kliewer SA & Burgess SC, 2009. FGF21 induces PGC-1alpha and regulates carbohydrate and fatty acid metabolism during the adaptive starvation response. Proc Natl Acad Sci U S A. 106, 10853–10858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Potthoff MJ, Kliewer SA & Mangelsdorf DJ, 2012. Endocrine fibroblast growth factors 15/19 and 21: from feast to famine. Genes Dev. 26, 312–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Rothwell NJ & Stock MJ, 1987. Influence of carbohydrate and fat intake on diet-induced thermogenesis and brown fat activity in rats fed low protein diets. J Nutr. 117, 1721–1726. [DOI] [PubMed] [Google Scholar]
  92. Rothwell NJ, Stock MJ & Tyzbir RS, 1982. Energy balance and mitochondrial function in liver and brown fat of rats fed “cafeteria” diets of varying protein content. J Nutr. 112, 1663–1672. [DOI] [PubMed] [Google Scholar]
  93. Rothwell NJ, Stock MJ & Tyzbir RS, 1983. Mechanisms of thermogenesis induced by low protein diets. Metabolism. 32, 257–261. [DOI] [PubMed] [Google Scholar]
  94. Sanchez J, Palou A & Pico C, 2009. Response to carbohydrate and fat refeeding in the expression of genes involved in nutrient partitioning and metabolism: striking effects on fibroblast growth factor-21 induction. Endocrinology. 150, 5341–5350. [DOI] [PubMed] [Google Scholar]
  95. Sarruf DA, Thaler JP, Morton GJ, German J, Fischer JD, Ogimoto K & Schwartz MW, 2010. Fibroblast growth factor 21 action in the brain increases energy expenditure and insulin sensitivity in obese rats. Diabetes. 59, 1817–1824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Schaap FG, Kremer AE, Lamers WH, Jansen PL & Gaemers IC, 2013. Fibroblast growth factor 21 is induced by endoplasmic reticulum stress. Biochimie. 95, 692–699. [DOI] [PubMed] [Google Scholar]
  97. Schlein C, Talukdar S, Heine M, Fischer AW, Krott LM, Nilsson SK, Brenner MB, Heeren J & Scheja L, 2016. FGF21 Lowers Plasma Triglycerides by Accelerating Lipoprotein Catabolism in White and Brown Adipose Tissues. Cell Metab. 23, 441–453. [DOI] [PubMed] [Google Scholar]
  98. Schumann G, Liu C, O’Reilly P, Gao H, Song P, Xu B, Ruggeri B, Amin N, Jia T, Preis S, Segura Lepe M, Akira S, Barbieri C, Baumeister S, Cauchi S, Clarke TK, Enroth S, Fischer K, Hallfors J, Harris SE, Hieber S, Hofer E, Hottenga JJ, Johansson A, Joshi PK, Kaartinen N, Laitinen J, Lemaitre R, Loukola A, Luan J, Lyytikainen LP, Mangino M, Manichaikul A, Mbarek H, Milaneschi Y, Moayyeri A, Mukamal K, Nelson C, Nettleton J, Partinen E, Rawal R, Robino A, Rose L, Sala C, Satoh T, Schmidt R, Schraut K, Scott R, Smith AV, Starr JM, Teumer A, Trompet S, Uitterlinden AG, Venturini C, Vergnaud AC, Verweij N, Vitart V, Vuckovic D, Wedenoja J, Yengo L, Yu B, Zhang W, Zhao JH, Boomsma DI, Chambers J, Chasman DI, Daniela T, de Geus E, Deary I, Eriksson JG, Esko T, Eulenburg V, Franco OH, Froguel P, Gieger C, Grabe HJ, Gudnason V, Gyllensten U, Harris TB, Hartikainen AL, Heath AC, Hocking L, Hofman A, Huth C, Jarvelin MR, Jukema JW, Kaprio J, Kooner JS, Kutalik Z, Lahti J, Langenberg C, Lehtimaki T, Liu Y, Madden PA, Martin N, Morrison A, Penninx B, Pirastu N, Psaty B, Raitakari O, Ridker P, Rose R, Rotter JI, Samani NJ, Schmidt H, Spector TD, Stott D, Strachan D, Tzoulaki I, van der Harst P, van Duijn CM, Marques-Vidal P, Vollenweider P, Wareham NJ, Whitfield JB, Wilson J, Wolffenbuttel B, Bakalkin G, Evangelou E, Liu Y, Rice KM, Desrivieres S, Kliewer SA, Mangelsdorf DJ, Muller CP, Levy D & Elliott P, 2016. KLB is associated with alcohol drinking, and its gene product beta-Klotho is necessary for FGF21 regulation of alcohol preference. Proc Natl Acad Sci U S A. 113, 14372–14377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Soberg S, Andersen ES, Dalgaard NB, Jarlhelt I, Hansen NL, Hoffmann N, Vilsboll T, Chenchar A, Jensen M, Grevengoed TJ, Trammell SAJ, Knop FK & Gillum MP, 2018. FGF21, a liver hormone that inhibits alcohol intake in mice, increases in human circulation after acute alcohol ingestion and sustained binge drinking at Oktoberfest. Molecular metabolism. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Soberg S, Sandholt CH, Jespersen NZ, Toft U, Madsen AL, von Holstein-Rathlou S, Grevengoed TJ, Christensen KB, Bredie WLP, Potthoff MJ, Solomon TPJ, Scheele C, Linneberg A, Jorgensen T, Pedersen O, Hansen T, Gillum MP & Grarup N, 2017. FGF21 Is a Sugar-Induced Hormone Associated with Sweet Intake and Preference in Humans. Cell Metab. 25, 1045–1053 e1046. [DOI] [PubMed] [Google Scholar]
  101. Solon-Biet SM, Cogger VC, Pulpitel T, Heblinski M, Wahl D, McMahon AC, Warren A, Durrant-Whyte J, Walters KA, Krycer JR, Ponton F, Gokarn R, Wali JA, Ruohonen K, Conigrave AD, James DE, Raubenheimer D, Morrison CD, Le Couteur DG & Simpson SJ, 2016Defining the Nutritional and Metabolic Context of FGF21 Using the Geometric Framework. Cell Metab. 24, 555–565. [DOI] [PubMed] [Google Scholar]
  102. Solon-Biet SM, McMahon AC, Ballard JW, Ruohonen K, Wu LE, Cogger VC, Warren A, Huang X, Pichaud N, Melvin RG, Gokarn R, Khalil M, Turner N, Cooney GJ, Sinclair DA, Raubenheimer D, Le Couteur DG & Simpson SJ, 2014. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 19, 418–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Solon-Biet SM, Walters KA, Simanainen UK, McMahon AC, Ruohonen K, Ballard JW, Raubenheimer D, Handelsman DJ, Le Couteur DG & Simpson SJ, 2015. Macronutrient balance, reproductive function, and lifespan in aging mice. Proc Natl Acad Sci U S A. 112, 3481–3486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Song P, Zechner C, Hernandez G, Canovas J, Xie Y, Sondhi V, Wagner M, Stadlbauer V, Horvath A, Leber B, Hu MC, Moe OW, Mangelsdorf DJ & Kliewer SA, 2018. The Hormone FGF21 Stimulates Water Drinking in Response to Ketogenic Diet and Alcohol. Cell Metab. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Sorensen A, Mayntz D, Raubenheimer D & Simpson SJ, 2008. Protein-leverage in mice: the geometry of macronutrient balancing and consequences for fat deposition. Obesity (Silver Spring). 16, 566–571. [DOI] [PubMed] [Google Scholar]
  106. Stemmer K, Zani F, Habegger KM, Neff C, Kotzbeck P, Bauer M, Yalamanchilli S, Azad A, Lehti M, Martins PJ, Muller TD, Pfluger PT & Seeley RJ, 2015. FGF21 is not required for glucose homeostasis, ketosis or tumour suppression associated with ketogenic diets in mice. Diabetologia. 58, 2414–2423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Stone KP, Wanders D, Orgeron M, Cortez CC & Gettys TW, 2014. Mechanisms of increased in vivo insulin sensitivity by dietary methionine restriction in mice. Diabetes. 63, 3721–3733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Talukdar S, Owen BM, Song P, Hernandez G, Zhang Y, Zhou Y, Scott WT, Paratala B, Turner T, Smith A, Bernardo B, Muller CP, Tang H, Mangelsdorf DJ, Goodwin B & Kliewer SA, 2016. FGF21 Regulates Sweet and Alcohol Preference. Cell Metab. 23, 344–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Tanaka T, Ngwa JS, van Rooij FJ, Zillikens MC, Wojczynski MK, Frazier-Wood AC, Houston DK, Kanoni S, Lemaitre RN, Luan J, Mikkila V, Renstrom F, Sonestedt E, Zhao JH, Chu AY, Qi L, Chasman DI, de Oliveira Otto MC, Dhurandhar EJ, Feitosa MF, Johansson I, Khaw KT, Lohman KK, Manichaikul A, McKeown NM, Mozaffarian D, Singleton A, Stirrups K, Viikari J, Ye Z, Bandinelli S, Barroso I, Deloukas P, Forouhi NG, Hofman A, Liu Y, Lyytikainen LP, North KE, Dimitriou M, Hallmans G, Kahonen M, Langenberg C, Ordovas JM, Uitterlinden AG, Hu FB, Kalafati IP, Raitakari O, Franco OH, Johnson A, Emilsson V, Schrack JA, Semba RD, Siscovick DS, Arnett DK, Borecki IB, Franks PW, Kritchevsky SB, Lehtimaki T, Loos RJ, Orho-Melander M, Rotter JI, Wareham NJ, Witteman JC, Ferrucci L, Dedoussis G, Cupples LA & Nettleton JA, 2013. Genome-wide meta-analysis of observational studies shows common genetic variants associated with macronutrient intake. Am J Clin Nutr. 97, 1395–1402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Torii K & Niijima A, 2001. Effect of lysine on afferent activity of the hepatic branch of the vagus nerve in normal and L-lysine-deficient rats. Physiol Behav. 72, 685–690. [DOI] [PubMed] [Google Scholar]
  111. Touvier T, De Palma C, Rigamonti E, Scagliola A, Incerti E, Mazelin L, Thomas JL, D’Antonio M, Politi L, Schaeffer L, Clementi E & Brunelli S, 2015. Muscle-specific Drp1 overexpression impairs skeletal muscle growth via translational attenuation. Cell Death Dis. 6, e1663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Vandanmagsar B, Warfel JD, Wicks SE, Ghosh S, Salbaum JM, Burk D, Dubuisson OS, Mendoza TM, Zhang J, Noland RC & Mynatt RL, 2016. Impaired Mitochondrial Fat Oxidation Induces FGF21 in Muscle. Cell reports. 15, 1686–1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. von Holstein-Rathlou S, BonDurant LD, Peltekian L, Naber MC, Yin TC, Claflin KE, Urizar AI, Madsen AN, Ratner C, Holst B, Karstoft K, Vandenbeuch A, Anderson CB, Cassell MD, Thompson AP, Solomon TP, Rahmouni K, Kinnamon SC, Pieper AA, Gillum MP & Potthoff MJ, 2016. FGF21 Mediates Endocrine Control of Simple Sugar Intake and Sweet Taste Preference by the Liver. Cell Metab. 23, 335–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Wanders D, Forney LA, Stone KP, Burk DH, Pierse A & Gettys TW, 2017. FGF21 Mediates the Thermogenic and Insulin-Sensitizing Effects of Dietary Methionine Restriction but not its Effects on Hepatic Lipid Metabolism. Diabetes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Wanders D, Stone KP, Dille K, Simon J, Pierse A & Gettys TW, 2015. Metabolic responses to dietary leucine restriction involve remodeling of adipose tissue and enhanced hepatic insulin signaling. Biofactors. 41, 391–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Wanders D, Stone KP, Forney LA, Cortez CC, Dille KN, Simon J, Xu M, Hotard EC, Nikonorova IA, Pettit AP, Anthony TG & Gettys TW, 2016. Role of GCN2-Independent Signaling Through a Noncanonical PERK/NRF2 Pathway in the Physiological Responses to Dietary Methionine Restriction. Diabetes. 65, 1499–1510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Wek RC, Jiang HY & Anthony TG, 2006. Coping with stress: eIF2 kinases and translational control. Biochem Soc Trans. 34, 7–11. [DOI] [PubMed] [Google Scholar]
  118. Wilson GJ, Lennox BA, She P, Mirek ET, Al Baghdadi RJ, Fusakio ME, Dixon JL, Henderson GC, Wek RC & Anthony TG, 2015. GCN2 is required to increase fibroblast growth factor 21 and maintain hepatic triglyceride homeostasis during asparaginase treatment. Am J Physiol Endocrinol Metab. 308, E283–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Yang C, Jin C, Li X, Wang F, McKeehan WL & Luo Y, 2012. Differential specificity of endocrine FGF19 and FGF21 to FGFR1 and FGFR4 in complex with KLB. PLoS ONE. 7, e33870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Zhang X, Yeung DC, Karpisek M, Stejskal D, Zhou ZG, Liu F, Wong RL, Chow WS, Tso AW, Lam KS & Xu A, 2008. Serum FGF21 levels are increased in obesity and are independently associated with the metabolic syndrome in humans. Diabetes. 57, 1246–1253. [DOI] [PubMed] [Google Scholar]

RESOURCES