Abstract
Dietary restriction (DR) refers to a broad set of interventions that limit the intake of specific nutrients or overall food consumption, either in quantity or timing, without causing malnutrition. DR has long been considered the most robust intervention for increasing healthspan and lifespan. This includes, not exhaustively, caloric restriction (CR), protein restriction (PR), amino acid restriction (AAR), intermittent fasting (IF), and time-restricted fasting (TRF), each with overlapping but distinct metabolic and physiological effects. This brief review examines the current scientific understanding of how some of the most commonly employed DR regimens may impact metabolism, lifespan, and healthspan. Particular attention is given to the underlying biological mechanisms and supporting evidence derived from both human clinical studies and fundamental biological research conducted with model organisms ranging from yeast to non-human primates.
Keywords: Dietary restriction, Caloric restriction, Protein restriction, Intermittent fasting, Time-restricted fasting, Longevity and aging, Metabolic reprogramming, Nutrient-sensing, Multiple model organisms, Anti-aging interventions
Introduction to dietary restriction
Dietary restriction (DR) refers to a wide range of dietary regimens that limit the intake of certain nutrients or overall food consumption, either in quantity or timing, while ensuring adequate nutrition is maintained. This includes, not exhaustively, caloric restriction (CR), protein restriction (PR), amino acid restriction (AAR), intermittent fasting (IF), and time-restricted fasting (TRF). Among all potential anti-aging interventions, DR has been regarded as the most robust and conserved intervention for increasing healthspan and lifespan across taxa [1]. Initial studies of each dietary restriction regimen in different model organisms are summarized in Table 1.
Table 1.
List of initial studies of different DR regimens in various model organisms. Here, we list the initial studies of CR, PR, AAR, IF, or TRF in different model organisms that we were able to find. The treatment column indicates the exact dietary regimens used in each study
| Type of dietary restriction | Model organism | Treatment | Reference | 
|---|---|---|---|
| Caloric restriction (CR) | Yeast | Glucose reduced from 2% to 0.5% | Lin et al., (RLS); Smith et al., (CLS) [8, 9] | 
| Nematode | Bacterial dilutions from 1x10^10 to 10^8 CFU | Klass et al., [11] | |
| Fruit fly | 35% less yeast and sugar | Chapman et al., [15] | |
| Rat | 25–65% CR | McCay et al., [3] | |
| Mouse | 55–65% CR | Weindruch et al., [18] | |
| Grey mouse lemur | 30% CR | Pifferi et al., [31] | |
| Rhesus monkey | 30% CR | Colman et al., [28] | |
| Protein/amino acid restriction (PR/AAR) | Yeast | Remove all amino acids, except 4 essential a.a. | Jiang et al., [41] | 
| Yeast | 50-90% less methionine | Koc et al., [46] | |
| Fruit fly | 57% less protein (with 28% CR) | Mair et al., [52] | |
| Fruit fly | 67%–89% less methionine | Troen et al., [48] | |
| Fruit fly | 85% less BCAAs | Juricic et al., [49] | |
| Rat | 40% less protein | Yu et al., [55] | |
| Rat | 80% less methionine | Orentreich et al., [56] | |
| Rat | Tryptophan deficiency | Segall et al., [66] | |
| Mouse | 83% less protein | Stoltzner et al., [54] | |
| Mouse | 67% less BCAAs | Richardson et al., [62] | |
| Mouse | 77% less methionine | Miller et al., [57] | |
| Mouse | 60% less tryptophan | De Marte et al., [64] | |
| Intermittent/time-restricted fasting (IF/TRF) | Nematode | ADF | Honjoh et al., [72] | 
| Fruit fly | 2-day fed:5-day fasted IF | Catterson et al., [73] | |
| Rat | ADF | Goodrick et al., [78] | |
| Mouse | ADF | Goodrick et al., [75] | |
| Mouse | TRF for 8 hr per day | Hatori et al., [84] | 
Calorie restriction
Caloric restriction (CR) is a reduction in energy intake below the level of calorie consumption under ad libitum conditions without causing malnutrition. The concept of caloric restriction has been recognized for centuries, with early philosophical and medical texts acknowledging the potential benefits of reduced food intake [2]. Scientific exploration of CR began in the early twentieth century, notably with studies on rats that demonstrated a significant extension of both mean and maximal lifespan through calorie reduction [3]. McCay’s groundbreaking research in the 1930s provided some of the first experimental evidence linking CR to longevity.
Subsequent studies expanded across various model organisms, consistently reaffirming the longevity benefits of CR across multiple species, including yeast, worms, flies, rodents, and primates [4–7]. In yeast, CR typically involves lowering glucose concentrations in the growth medium from 2% to 0.5% or less. This reduction significantly extends both the replicative lifespan (RLS; the number of daughter cells that a yeast mother cell can produce) [8] and the chronological lifespan (CLS; how long non-dividing cells remain viable) [9]. In nematode Caenorhabditis elegans, CR extends lifespan by up to 50%, depending on the method used. Approaches include bacterial dilution (reducing food availability) [10, 11] and genetic manipulation (such as eat-2 mutants with reduced feeding rate) [12]. CR also improves proteostasis and reduces the accumulation of age-related protein aggregates, contributing to improved cellular function and organismal health in worms [13]. In fruit flies, Drosophila, studies have observed CR-induced lifespan extension by 30–50%, particularly when CR is initiated early in life [14, 15]. CR has also been shown to preserve physical activity and delay the onset of age-related decline in reproductive functions in flies [16, 17].
In rodents, the impact of CR is typically investigated by reducing calorie intake to around 60–70% of the ad libitum consumption. Reductions of calorie intake at this level extend both mean and maximum lifespan across various rodent species, including mice and rats [3, 18–20]. Rodent studies have also been instrumental in understanding the long-term effects of CR on disease onset. In studies where rodents are subjected to CR, the onset of age-related diseases, such as cancer, cardiovascular disease, and neurodegeneration, is delayed or attenuated [18, 21, 22]. For example, CR has been shown to reduce the incidence and multiplicity of spontaneous and chemically induced tumors in mice and rats, particularly in tissues such as the liver, lung, and mammary glands [23]. Moreover, CR markedly lowers the development of atherosclerosis [24], one major contributor to cardiovascular morbidity in aging populations. In terms of neurodegeneration, CR has been linked to enhanced synaptic plasticity and decreased accumulation of amyloid-beta, a hallmark of Alzheimer’s disease pathology, in mouse models [25–27].
Research on non-human primates, such as rhesus monkeys (Macaca mulatta) and grey mouse lemurs (Microcebus murinus), has significantly contributed to the understanding of the potential effects of CR on aging and healthspan in humans, as these primates share many physiological and genetic similarities with humans. Results from these studies have demonstrated that CR improved overall health and longevity while delaying the onset of age-related diseases in these primates [28–31]. For example, CR has been linked to a reduced incidence of cancer, cardiovascular diseases, and metabolic disorders [28, 29]. Additionally, it helps preserve brain volume and enhances cognitive function in aged monkeys [28, 32], suggesting its neuroprotective effects.
Research on CR in humans faces challenges due to difficulties in conducting long-term controlled studies. However, evidence gathered from clinical trials, epidemiological research, and observational studies strongly indicates that reducing calorie intake provides significant health advantages [33, 34]. One of the most well-known human studies is the CALERIE (Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy), a 2-year, randomized controlled trial for non-obese individuals [35]. The results showed that moderate caloric restriction (around 25% reduction in daily caloric intake) led to significant improvements in several key biomarkers of aging, such as insulin sensitivity, blood pressure, inflammatory markers, and lipid profiles [36, 37]. Participants in the CALERIE trial also showed improvements in muscle function and immune system performance [38, 39], indicating that CR can have broad, positive effects on tissue maintenance during aging. (All the human clinical studies mentioned here and in the later sections are summarized in Table 2.)
Table 2.
List of all human clinical studies discussed in the main text
| Study | Subject characteristics | Study protocol | Duration | Primary endpoints | Key observations | References | 
|---|---|---|---|---|---|---|
| CALERIE trial | Non-obese healthy adults (aged 21-50) | Randomized controlled trial with 25% reduction in daily caloric intake | 2 years | Biomarkers of aging, cardiometabolic health | Significant improvements in insulin sensitivity, blood pressure, inflammatory markers, lipid profiles, muscle function, and immune system performance | Meydani et al.,Most et al., Kraus et al., Das et al., [36–39] | 
| NHANES III analysis | Adults aged 50-65 vs 65+ | Epidemiological analysis of protein intake levels | Long-term follow-up | Mortality | High protein intake (≥20% of calories) associated with 75% increase in overall and cancer mortality in 50-65 age group; this negative association is not observed in >65 age group | Levine et al., [68] | 
| Plant vs animal protein study | Health care professionals | Observational analysis of protein source | Long-term follow-up | Mortality | Plant-based proteins associated with reduced mortality risk; animal-based protein linked to increased risk | Song et al., [69] | 
| Protein restriction (7%–9% protein diet) | Middle-aged overweight and mildly obese men | Randomized clinical trials of reduced protein intake | 43 ± 11days | Glucose metabolism | Reducing protein intake improved glucose homeostasis | Fontana et al., [70] | 
| Low-protein diet study | Lean, healthy men | Low-protein, high-carbohydrate (LPHC; 8 E% protein) diet vs a habitual higher protein (16 E% protein) diet | 5 weeks | Insulin sensitivity, metabolic parameters | Improved whole-body insulin sensitivity when proteins replaced by carbohydrates; tendency to consume more calories from fats/carbohydrates to maintain body weight | Nicolaisen et al., [71] | 
| Early time-restricted feeding | Prediabetic men | eTRF (6-hour eating window) vs control schedule (12-hr) | 5 weeks | Insulin sensitivity, metabolic markers | Improved insulin sensitivity even without weight loss | Sutton et al., [92] | 
| 10-hour time-restricted feeding | Individuals with metabolic syndrome | 10-hour time-restricted feeding | 2-week baseline and 12-week intervention periods | Metabolic and cardiovascular outcomes | Reduced fasting glucose, lowered HbA1c, improved blood pressure and atherogenic lipid levels | Wilkinson et al., [93] | 
| IF and brain function | Overweight adults aged 55-70 | 5:2 intermittent fasting (480 Kcal/day for 2 days; USDA diet for 5 days ) | 8 weeks | Cognitive function, brain aging | Enhanced executive function and memory; attenuated rate of brain aging | Kapogiannis et al., [94] | 
Protein/amino acid restriction
Protein restriction (PR) or amino acid restriction (AAR) refers to dietary interventions that lower total protein intake or limit specific amino acids, respectively, while maintaining overall nutritional adequacy. Common PR strategies include lowering daily protein intake, which also leads to reduced caloric intake, or substituting protein content in the diet with carbohydrates without altering the total caloric intake. The longevity effects of PR have been well-documented in several model organisms, including yeast, fruit flies, and rodents [40]. On the other hand, targeting specific amino acids such as methionine, tryptophan, or branched-chain amino acids (BCAAs) like leucine, isoleucine, and valine has also been demonstrated to increase longevity.
In yeast, limiting the availability of all nonessential amino acids [41] or specific amino acids such as methionine, asparagine, or glutamate extends RLS or CLS independently of calorie intake [42–46]. Due to the lack of a precisely defined food source for cultivating C. elegans in the laboratory, direct implementation of protein or amino acid restriction has not been feasible. However, indirect evidence from worms cultured on metformin-treated bacteria suggests that methionine restriction may be responsible for the observed longevity phenotype in metformin-treated worms [47]. In Drosophila, both protein and amino acid restriction have been shown to extend lifespan, particularly when methionine or BCAA is restricted [48–52]. Grandison et al. demonstrated that reducing dietary protein, while supplementing only methionine, reversed the lifespan-extending effect, highlighting methionine’s central role in aging regulation [53]. In addition to methionine restriction, recent studies have suggested that BCAA restriction may also increase survival in flies [49].
In rodents, reducing dietary protein by 40–83% typically leads to a 10–20% increase in lifespan [54, 55], whereas methionine restriction alone has been shown to extend lifespan by 30–40%, independent of total caloric intake [56–58]. Besides lifespan extension, methionine restriction has been shown to reduce oxidative stress and promote leanness in rodents [59, 60]. Similarly, emerging evidence suggests that restricting BCAAs may promote longevity and improve metabolic profiles in mice [61, 62]. Notably, both protein restriction and BCAA restriction exhibit sex-dependent effects in mice [62, 63]. Finally, dietary restrictions of tryptophan have been reported to extend the lifespan of mice and rats and delay the onset of age-associated diseases in various studies dating back to the 1970s and 1980s [64–66]. Interestingly, a recent study suggests that tryptophan restriction may be an evolutionarily conserved intervention for promoting longevity. The administration of ibuprofen, an inhibitor of tryptophan uptake, extends the lifespan of yeast, worms, and flies [67]. However, the underlying mechanism of how tryptophan restriction may exhibit its longevity benefits remains largely unknown.
Direct evidence linking PR to lifespan extension remains limited in humans. However, accumulating data suggest that reducing total protein intake, particularly from animal sources, and specific amino acids may yield metabolic benefits and promote healthy aging. Epidemiological studies, such as analyses of data from the National Health and Nutrition Examination Survey (NHANES), have shown that adults aged 50–65 consuming high levels of protein (≥ 20% of daily calories) had a 75% increase in overall mortality and a fourfold increase in cancer mortality, compared to those with moderate protein intake [68]. Interestingly, these associations were not observed in individuals over 65, highlighting the importance of age-specific recommendations. Importantly, the source of protein appears to matter; plant-based proteins were associated with reduced mortality risk, while animal-based protein was linked to increased risk [69]. Short-term intervention trials in humans further support the health benefits of protein restriction. For instance, randomized controlled studies have shown that reducing protein intake improves glucose homeostasis [70]. A recent study by Nicolaisen et al. demonstrates that when lean and healthy men followed a low-protein diet (meeting minimum requirements) for 5 weeks, they tended to consume more calories, either from fats or carbohydrates, to maintain their body weight. Intriguingly, the protein-restricted diet improved whole-body insulin sensitivity when proteins were replaced by carbohydrates, which may involve FGF21 (fibroblast growth factor 21) [71].
Intermittent fasting
Intermittent fasting (IF) has gained considerable attention in recent years, not only as a weight management strategy but also for its potential effects on longevity and healthspan. Intermittent fasting refers to eating patterns that cycle between periods of eating and fasting. Common approaches include alternate-day fasting (ADF) and periodic fasting (PF). The emerging time-restricted fasting (TRF) may also be considered as an alternative form of IF. TRF limits daily food consumption to a specific time window (e.g., 8 h) and fasting for the rest of the day. ADF alternates between regular eating days and fasting days with little or no caloric intake. PF involves restricting calories or complete fasting for longer periods (e.g., 2–5 days) at regular intervals.
Compelling evidence for the longevity-promoting effects of IF comes from research in various model organisms. In organisms such as worms [72] and flies [73, 74], intermittent fasting has been shown to extend lifespan by 20–40%. A variety of IF regimens, including ADF, PF, and TRF, have been tested in mice and rats, yielding robust evidence that IF can enhance longevity and delay the onset of age-related pathologies [75–78]. One of the earliest studies by Goodrick et al. demonstrated that ADF significantly extended both median and maximum lifespan in several strains of mice. These benefits were observed even when fasting was initiated in mid-life, suggesting that IF can be beneficial even if not practiced lifelong. Subsequent studies confirmed that rodents subjected to IF exhibit increased lifespan comparable to those under caloric restriction (CR), although the degree of extension varies with strain, sex, and fasting protocol [79, 80]. IF in rodents consistently results in improved insulin sensitivity, lower fasting glucose and insulin levels, and protection against high-fat diet-induced obesity [81–84]. Rodent studies have also shown that IF reduces the incidence and progression of age-related cancers and neurodegenerative diseases [27, 85–87]. Notably, several studies also indicate that the beneficial effects of IF are not attributable to reduced calorie intake [81, 84, 88]. It is also worth noting that the CR regimens implemented in early rodent studies were often associated with TRF, as the animals were fed once daily and consumed most of their meal within a short timeframe.
Although conclusive evidence that intermittent fasting (IF) extends human lifespan remains absent, numerous surrogate endpoints and health-related outcomes suggest that IF may provide longevity benefits [89–91]. IF elicits significant metabolic improvements. A controlled trial conducted by Sutton et al. in prediabetic men found that limiting food intake to a 6-h window improved insulin sensitivity, even in the absence of weight loss [92]. Complementary findings from Wilkinson et al. showed that 10-h TRF reduced fasting glucose levels and lowered HbA1c (Hemoglobin A1c) in individuals with metabolic syndrome [93]. In the same study, TRF also improved cardiovascular outcomes, such as blood pressure and atherogenic lipid levels, in patients. Recent findings by Kapogiannis et al. indicate that IF enhances executive function and memory while also attenuating the rate of brain aging in older adults [94].
The underlying molecular and cellular mechanisms of dietary restriction
Metabolic reprogramming
At the cellular level, DR promotes a transition from glucose-dependent metabolism toward alternative fuel utilization pathways. Different DR regimens, including CR, PR, and IF, trigger overlapping yet distinct metabolic adaptations. Previous studies in C. elegans and Drosophila demonstrate that CR leads to a metabolic shift toward fatty acid metabolism [95, 96], enhancing the organism’s ability to utilize stored lipids for energy production. Similarly, elevated lipogenesis and lipolysis are observed in mice subjected to CR or IF [97, 98], indicating that dynamic remodeling of lipid metabolism is needed to maintain metabolic homeostasis during periods of nutrient scarcity. It is worth noting that PR also influences lipid metabolism. In flies fed a high-fat diet, PR has been shown to alter lipid metabolic pathways [99]. Dietary methionine restriction persistently increases total energy expenditure and enhances metabolic flexibility while increasing uncoupled respiration in both fed and fasted states [100]. Enhanced fatty acid oxidation has long been linked to various longevity pathways [101, 102]. Research in C. elegans and Drosophila has also demonstrated that genetic upregulation of lipolytic pathways and fatty acid β-oxidation contributes to lifespan extension [103, 104], suggesting that a metabolic shift toward more efficient energy utilization may be critical for the longevity benefits.
IF, which involves cycling between periods of feeding and fasting, also leads to elevated ketogenesis, which is not found in CR or PR. Growing evidence supports the health benefits of ketone bodies in animal models [105, 106]. However, Tomita et al. found that ketone body supplementation may produce opposing effects, depending on the timing of administration. Ketone body supplementation prolongs lifespan when given to aged mice but increases mortality when administered early in life [107]. These findings suggest that the broader metabolic reprogramming caused by IF may have a greater impact on promoting longevity than the elevation of ketone bodies alone.
Nutrient-sensing signaling networks
Insulin/IGF-1 signaling (IIS)
The insulin/IGF-1 signaling (IIS) is a highly conserved signaling pathway that regulates both metabolism and lifespan (Fig. 1). Reduced IIS activity and subsequent activation of pro-longevity FOXO (forkhead box O) transcription factor have been consistently demonstrated to extend lifespan in model organisms such as worms, flies, and mice [108, 109]. Accumulating evidence from vertebrate models or humans has suggested that CR leads to systemic decreases in glucose and insulin, thereby improving insulin sensitivity [37, 110]. Remarkably, PR triggers a similar metabolic benefit even in the absence of calorie reduction. In healthy individuals, PR has been shown to reduce blood glucose and insulin levels [111]. Furthermore, methionine restriction can mimic many of the effects of CR, including reduced blood glucose and insulin [112]. Similar effects in systemic glucose homeostasis could also be found in IF animals [81]. Therefore, the DR-induced longevity benefits may be mediated by inactivating the IIS pathway in vertebrates. Consistent with this model, studies in rodents have shown that the FOXOs become activated under DR conditions and are required for DR-mediated lifespan extension [113, 114].
Fig. 1.
A schematic illustration summarizes the signaling pathways discussed in the text that might mediate the longevity effects of different DR regimens. Key components of each signaling pathway and their downstream effectors are included here. We list only the most recognized names of their mammalian homologs. The arrows indicate whether the activities or the levels of these key components are up- or down-regulated in response to different DR regimens
mTORC1 signaling
The mammalian target of rapamycin (mTOR) pathway serves as a central hub for integrating signals from multiple nutrients and growth factors [115]. mTOR exists in two distinct complexes: mTORC1 and mTORC2, with mTORC1 being the primary nutrient sensor. When activated, mTORC1 promotes anabolic processes essential for cell growth and proliferation, including protein synthesis, lipid synthesis, nucleotide biosynthesis, and glycolysis. Conversely, mTORC1 activation suppresses catabolic processes such as autophagy and lysosomal biogenesis. Accumulating studies have established mTORC1 inhibition as an evolutionarily conserved mechanism for lifespan extension [116]. Pharmacological inhibition of mTORC1 with rapamycin also promotes health and longevity in diverse model organisms [117], making it an attractive target for anti-aging therapy.
Given the role of mTORC1 as a cellular nutrient sensor, DR is anticipated to suppress mTORC1 activity by reducing nutrient availability. Indeed, this has been demonstrated across multiple experimental paradigms, with CR, PR, and IF showing significant decreases in mTORC1 signaling activity in various tissues of mouse model [118–122]. mTORC1 regulates longevity mainly through modulating both protein synthesis and autophagy [116], two critical processes for protein homeostasis. When nutrients are abundant, mTORC1 drives cellular growth by activating translation regulators such as S6K1 and 4E-BP1, stimulating protein synthesis. In response to DR, reduced mTORC1 activity leads to a decrease in protein synthesis. The significance of this regulation is further highlighted by recent studies showing that genetic reduction of protein synthesis alone can extend lifespan [123]. mTORC1 also suppresses autophagy by suppressing the autophagy initiators, such as ULK1 and ATG13 (Autophagy related 13). Under DR conditions, reduced mTORC1 activity alleviates this inhibition, boosting autophagy for clearing damaged cellular components. Additionally, mTORC1 inhibition activates TFEB, a transcription factor that boosts lysosomal biogenesis and autophagy-related gene expression during nutrient scarcity. This elevating autophagy further promotes longevity, as impaired autophagy has been shown to block the health benefits of DR [124, 125].
AMPK signaling
AMP-activated protein kinase (AMPK) is a highly conserved serine/threonine kinase that functions as a central regulator of cellular energy homeostasis, activated in response to energy stress to restore ATP levels by modulating metabolic pathways [126]. Under DR conditions, reduced nutrient availability leads to decreased ATP levels and increased AMP/ATP ratios, which directly activate AMPK. Research in C. elegans demonstrates that AMPK activity is required for DR-induced longevity [127, 128]. Furthermore, genetic overexpression of AMPK extends lifespan in worms and flies [129, 130]. In rodents, DR activates AMPK in various tissues, including the heart and skeletal muscle [131–133]. Upon activation, AMPK triggers several longevity-promoting cellular mechanisms, such as autophagy. AMPK promotes autophagy by phosphorylating ULK1 (Unc-51-like kinase 1) and inhibiting mTORC1 [134]. The negative regulation of mTORC1 signaling by AMPK also results in decreased protein translation. Moreover, AMPK activates eukaryotic elongation factor 2 kinase (eEF2K), which suppresses translation elongation, thereby further reducing global protein synthesis [135]. Furthermore, the AMPK-eEF2K axis has also been implicated in the regulation of stress granule formation, which also contributes to DR-induced longevity in worms [128]. AMPK regulates lipid metabolism by phosphorylating acetyl-CoA carboxylase 1/2 (ACC1/2), thereby inhibiting the production of malonyl-CoA, a critical substrate for fatty acid synthase and a precursor for de novo palmitate synthesis. Since malonyl-CoA potently inhibits mitochondrial carnitine palmitoyl transferase 1 (CPT1) and thus limits fatty acid β-oxidation, AMPK-mediated ACC phosphorylation simultaneously suppresses lipogenesis while promoting fatty acid oxidation [126], a metabolic pattern typically observed in animals under DR.
Sirtuin signaling
Sirtuins are evolutionarily conserved enzymes that catalyze the removal of acetyl groups from lysine residues on target proteins. This reaction produces nicotinamide, O-acetyl-ADP-ribose, and a deacetylated protein, linking sirtuin activity to cellular NAD⁺ levels and metabolic state. The mammalian sirtuin family comprises seven members (SIRT1-7) with distinct subcellular localizations and substrate specificities [136]. SIRT1 emerges as the most extensively studied sirtuin in the context of DR and longevity regulation. In yeast, overexpression of Sir2, the homolog of mammalian SIRT1, extends lifespan by approximately 30% [137]. The role of Sir2/SIRT1 in mediating DR-induced longevity has been well-demonstrated in worms and flies, and mice as well [138–140], underscoring the evolutionary significance of sirtuins in lifespan regulation. Moreover, upregulation of SIRT1 expression has been observed in mice subjected to CR and fasting [140, 141], further supporting its involvement in nutrient-responsive pathways. While whole-body SIRT1 overexpression protects against metabolic disorders without extending lifespan, brain-specific overexpression of SIRT1 significantly increases lifespan and delays aging-associated decline [142, 143], suggesting that SIRT1 activity in the central nervous system might play a critical role in regulating aging. DR-induced activation of SIRT1 leads to the deacetylation of various transcription factors and metabolic regulators such as PGC-1α, FOXO proteins, and p53 [144]. This activation promotes a transcriptome-wide shift, contributing to improved cellular function and stress resistance. Supporting this, Levin et al. demonstrated that the effects of TRF-CR on hepatic gene expression in mice are largely mediated by an increase in the daytime peak of hepatic NADH levels, which inhibit SIRT1 activity to modulate transcriptional and metabolic responses [145]. Besides SIRT1, SIRT6 shares several pro-longevity attributes with SIRT1, including involvement in genome stability, epigenetic regulation, and attenuation of inflammation [146–148].
As sirtuin activity requires NAD+, the contributions of sirtuin and NAD+ metabolism for the beneficial effects of DR are difficult to separate. Thus, it is not surprising to find that the DR-induced lifespan extension requires both Sir2 and NAD+ synthesis pathway in yeast [149]. Altering key enzymes involved in the NAD+ metabolism, such as PNC1 or NAMPT, has been shown to affect lifespan in yeast, worms, and mice [150–153]. Consistent with this idea, nutritional supplementation with NAD+ or its precursors has been proposed to promote healthy aging, potentially via sirtuin activation [154].
The GCN-ATF4-FGF21 axis
GCN2 (general control nonderepressible 2) is a conserved serine/threonine kinase that also acts as an amino acid sensor to regulate various cellular and physiological responses, including immune system homeostasis and integrated stress responses [155–157]. Activation of GCN2 results in the phosphorylation of eIF2 (eukaryotic translation initiation factor 2), which inhibits the translation of most mRNA, while selectively upregulating the translation of certain proteins, including ATF4 (activating transcription factor 4) [158–160]. ATF4 is required for the elevated expression of several PR or AAR-responsive genes, including the hormone FGF21 (fibroblast growth factor 21) [161]. The roles of GCN2 in DR have been reported in both worms and rodents. In worms, gcn-2 is required for the lifespan extension by the genetic model of CR (i.e., eat-2 mutants) and TOR inactivation [162]. In rodents, Gcn2-knockout mice subjected to PR demonstrate a delayed metabolic response adaptation in response to reduced protein intake [163].
The FGF21 is a liver-derived hormone that has been implicated in many metabolic effects of PR, such as increased insulin sensitivity [164, 165]. It has also been reported that both the hepatic expression and circulating level of FGF21 are elevated in response to chronic PR in both rodents and humans [70, 164]. Moreover, mice lacking fgf21 are less responsive to PR [164, 166]. The most compelling evidence came from the mouse studies, in which Zhang et al. have shown that FGF21 overexpression could extend mouse lifespan [167] and Hill et al. reported that FGF21 is required for PR to extend lifespan in male mice [168].
Translational perspectives
The translation of DR research to human anti-aging interventions faces fundamental obstacles rooted in the complexity of human biology and lifestyle factors. While controlled studies in model organisms demonstrate clear benefits, human studies present unique challenges. Long-term adherence to restrictive eating patterns, individual genetic variability, and the influence of social and environmental factors all complicate the direct translation of laboratory findings. Recent clinical trials have shown promising results for IF and CR protocols in humans [37, 169], with observed improvements in insulin sensitivity, cardiovascular markers, and inflammatory profiles. However, the optimal timing (i.e., the age of the participant), duration, and intensity of dietary restriction interventions remain poorly defined.
Understanding the molecular mechanisms underlying DR benefits has opened new therapeutic avenues beyond traditional DR limitations. The identification of key signaling pathways, including mTOR, AMPK, and sirtuins, has led to the development of DR mimetics. These compounds aim to activate the same beneficial cellular responses as DR without requiring actual food limitation, potentially offering more practical alternatives for clinical application.
Rapamycin has emerged as one of the most promising DR mimetics through its specific inhibition of mTORC1. Rapamycin recapitulates many of the longevity benefits observed in DR animals, including enhanced autophagy, improved protein quality control, and increased stress resistance [170]. Clinical studies with rapamycin have demonstrated remarkable results, including improved immune function in elderly populations and enhanced vaccine responses [171]. However, chronic rapamycin use presents challenges, including increased risk of infections, delayed wound healing, and metabolic complications [172]. Recent research has focused on developing rapamycin analogs and intermittent dosing regimens that might maximize longevity benefits while minimizing adverse effects [173, 174].
Metformin, the world's most widely prescribed diabetes medication, has gained even more attention as a potential anti-aging intervention due to its ability to activate AMPK [175]. Large-scale epidemiological studies have shown that diabetic patients taking metformin often exhibit lower rates of age-related diseases, including cardiovascular disease [176], cancer [177, 178], and neurodegenerative disorders [179, 180], compared to those on other diabetes medications. Those observations have led to the TAME (Targeting Aging with Metformin) study, which aims to demonstrate metformin's potential as a geroprotective agent in non-diabetic populations [181]. Metformin's excellent safety profile, established through decades of clinical use, makes it a promising candidate for long-term use in non-diabetic or healthy populations.
The complementary mechanisms of rapamycin and metformin have sparked interest in combination approaches that target multiple pathways simultaneously. Preliminary studies in model systems suggest that combination therapy might provide additive or synergistic benefits [182–184], potentially allowing for lower doses of each drug while maintaining efficacy. The new strategies represent a compelling intersection of basic science discovery and clinical application.
It is worth noting that while DR has been regarded as the most robust intervention to promote health and increase lifespan, it does come with some trade-offs. The most commonly known trade-off is reduced fecundity [185–187]. However, some studies suggested that longevity and reproduction may be uncoupled under certain DR conditions [187, 188]. Furthermore, it has been reported that men and populations with elevated BMI who undergo DR may have mental health consequences, such as increased depressive symptoms [189]. Therefore, the use of DR mimetics as an anti-aging intervention in humans should still be cautious before more clinical studies are completed.
Conclusion
The overlapping yet distinct mechanisms of different DR regimens, including CR, PR, AAR, IF, and TRF, offer us numerous potential targets for future translational research. As our understanding of the underlying mechanisms of these DR regimens continues to grow, the development of safe and effective interventions that mimic the benefits of DR holds significant promise for promoting healthy aging and preventing age-related diseases in human populations.
Acknowledgements
We thank the National Science and Technology Council, Taiwan, and Academia Sinica for the funding support to T.T. Ching (NSTC 112-2320-B-A49-014-MY3) and A.L. Hsu (NSTC 113-2320-B-A49-016-MY3). We also thank Academia Sinica for the funding support (AS-ASSA-113-03).
Author contributions
T.T. Ching and A.L. Hsu drafted and edited the manuscript together.
Funding
The following funding sources supported the completion of this manuscript. National Science and Technology Council, Taiwan (NSTC 112–2320-B-A49-014-MY3 to T.T.C. and NSTC 113–2320-B-A49-016-MY3 to A.L.H.); Academia Sinica (AS-ASSA-113–03 to A.L.H.).
Data availability
No datasets were generated or analysed during the current study.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Contributor Information
Tsui-Ting Ching, Email: [email protected].
Ao-Lin Hsu, Email: [email protected].
References
- 1.Fontana L, Partridge L, Longo VD. Extending healthy life span–from yeast to humans. Science. 2010;328:321–6. 10.1126/science.1172539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Longo VD, Anderson RM. Nutrition, longevity and disease: from molecular mechanisms to interventions. Cell. 2022;185:1455–70. 10.1016/j.cell.2022.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.McCay CM, Crowell MF, Maynard LA. The effect of retarded growth upon the length of life span and upon the ultimate body size. J Nutr. 1935;10:63–79. [PubMed] [Google Scholar]
- 4.Guarente L, Picard F. Calorie restriction–the SIR2 connection. Cell. 2005;120:473–82. 10.1016/j.cell.2005.01.029. [DOI] [PubMed] [Google Scholar]
- 5.Heilbronn LK, Ravussin E. Calorie restriction and aging: review of the literature and implications for studies in humans. Am J Clin Nutr. 2003;78:361–9. 10.1093/ajcn/78.3.361. [DOI] [PubMed] [Google Scholar]
- 6.Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: an update. Ageing Res Rev. 2017;39:36–45. 10.1016/j.arr.2016.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Partridge L, Piper MD, Mair W. Dietary restriction in Drosophila. Mech Ageing Dev. 2005;126:938–50. 10.1016/j.mad.2005.03.023. [DOI] [PubMed] [Google Scholar]
- 8.Lin SJ, et al. Calorie restriction extends Saccharomyces cerevisiae lifespan by increasing respiration. Nature. 2002;418:344–8. 10.1038/nature00829. [DOI] [PubMed] [Google Scholar]
- 9.Smith DL Jr, McClure JM, Matecic M, Smith JS. Calorie restriction extends the chronological lifespan of Saccharomyces cerevisiae independently of the Sirtuins. Aging Cell. 2007;6:649–62. 10.1111/j.1474-9726.2007.00326.x. [DOI] [PubMed] [Google Scholar]
- 10.Panowski SH, Wolff S, Aguilaniu H, Durieux J, Dillin A. PHA-4/Foxa mediates diet-restriction-induced longevity of C. elegans. Nature. 2007;447:550–5. [DOI] [PubMed] [Google Scholar]
- 11.Klass MR. Aging in the nematode Caenorhabditis elegans: major biological and environmental factors influencing life span. Mech Ageing Dev. 1977;6:413–29. 10.1016/0047-6374(77)90043-4. [DOI] [PubMed] [Google Scholar]
- 12.Lakowski B, Hekimi S. The genetics of caloric restriction in Caenorhabditis elegans. Proc Natl Acad Sci U S A. 1998;95:13091–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Matai L, et al. Dietary restriction improves proteostasis and increases life span through endoplasmic reticulum hormesis. Proc Natl Acad Sci U S A. 2019;116:17383–92. 10.1073/pnas.1900055116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mair W, Goymer P, Pletcher SD, Partridge L. Demography of dietary restriction and death in Drosophila. Science. 2003;301:1731–3. 10.1126/science.1086016. [DOI] [PubMed] [Google Scholar]
- 15.Chapman T, Partridge L. Female fitness in Drosophila melanogaster: an interaction between the effect of nutrition and of encounter rate with males. Proc Biol Sci. 1996;263:755–9. 10.1098/rspb.1996.0113. [DOI] [PubMed] [Google Scholar]
- 16.Bross TG, Rogina B, Helfand SL. Behavioral, physical, and demographic changes in Drosophila populations through dietary restriction. Aging Cell. 2005;4:309–17. 10.1111/j.1474-9726.2005.00181.x. [DOI] [PubMed] [Google Scholar]
- 17.Tu MP, Tatar M. Juvenile diet restriction and the aging and reproduction of adult Drosophila melanogaster. Aging Cell. 2003;2:327–33. [DOI] [PubMed] [Google Scholar]
- 18.Weindruch R, Walford RL, Fligiel S, Guthrie D. The retardation of aging in mice by dietary restriction: longevity, cancer, immunity and lifetime energy intake. J Nutr. 1986;116:641–54. 10.1093/jn/116.4.641. [DOI] [PubMed] [Google Scholar]
- 19.Stuchlikova E, Juricova-Horakova M, Deyl Z. New aspects of the dietary effect of life prolongation in rodents. What is the role of obesity in aging? Exp Gerontol. 1975;10:141–4. 10.1016/0531-5565(75)90043-1. [DOI] [PubMed] [Google Scholar]
- 20.Turturro A, et al. Growth curves and survival characteristics of the animals used in the biomarkers of aging program. J Gerontol A Biol Sci Med Sci. 1999;54:B492-501. 10.1093/gerona/54.11.b492. [DOI] [PubMed] [Google Scholar]
- 21.Weiss EP, Fontana L. Caloric restriction: powerful protection for the aging heart and vasculature. Am J Physiol Heart Circ Physiol. 2011;301:H1205-1219. 10.1152/ajpheart.00685.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wei L, Tao Q, Yao M, Zhao Z, Ge S. Caloric restriction can ameliorate postoperative cognitive dysfunction by upregulating the expression of Sirt1, MeCP2 and BDNF in the hippocampal CA1 region of aged C57BL/6 mice. Brain Sci. 2023. 10.3390/brainsci13030462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hursting SD, Lavigne JA, Berrigan D, Perkins SN, Barrett JC. Calorie restriction, aging, and cancer prevention: mechanisms of action and applicability to humans. Annu Rev Med. 2003;54:131–52. 10.1146/annurev.med.54.101601.152156. [DOI] [PubMed] [Google Scholar]
- 24.Guo Z, et al. Dietary restriction reduces atherosclerosis and oxidative stress in the aorta of apolipoprotein E-deficient mice. Mech Ageing Dev. 2002;123:1121–31. 10.1016/s0047-6374(02)00008-8. [DOI] [PubMed] [Google Scholar]
- 25.Patel NV, et al. Caloric restriction attenuates Abeta-deposition in Alzheimer transgenic models. Neurobiol Aging. 2005;26:995–1000. 10.1016/j.neurobiolaging.2004.09.014. [DOI] [PubMed] [Google Scholar]
- 26.Wang J, et al. Caloric restriction attenuates beta-amyloid neuropathology in a mouse model of Alzheimer’s disease. FASEB J. 2005;19:659–61. 10.1096/fj.04-3182fje. [DOI] [PubMed] [Google Scholar]
- 27.Halagappa VK, et al. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer’s disease. Neurobiol Dis. 2007;26:212–20. 10.1016/j.nbd.2006.12.019. [DOI] [PubMed] [Google Scholar]
- 28.Colman RJ, et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science. 2009;325:201–4. 10.1126/science.1173635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mattison JA, et al. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature. 2012;489:318–21. 10.1038/nature11432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mattison JA, et al. Caloric restriction improves health and survival of rhesus monkeys. Nat Commun. 2017;8:14063. 10.1038/ncomms14063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Pifferi F, et al. Caloric restriction increases lifespan but affects brain integrity in grey mouse lemur primates. Commun Biol. 2018;1:30. 10.1038/s42003-018-0024-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Willette AA, et al. Calorie restriction reduces the influence of glucoregulatory dysfunction on regional brain volume in aged rhesus monkeys. Diabetes. 2012;61:1036–42. 10.2337/db11-1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Omodei D, Fontana L. Calorie restriction and prevention of age-associated chronic disease. FEBS Lett. 2011;585:1537–42. 10.1016/j.febslet.2011.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Flanagan EW, Most J, Mey JT, Redman LM. Calorie restriction and aging in humans. Annu Rev Nutr. 2020;40:105–33. 10.1146/annurev-nutr-122319-034601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Fleischer JG, Das SK, Bhapkar M, Manoogian ENC, Panda S. Associations between the timing of eating and weight-loss in calorically restricted healthy adults: findings from the CALERIE study. Exp Gerontol. 2022;165:111837. 10.1016/j.exger.2022.111837. [DOI] [PubMed] [Google Scholar]
- 36.Most J, et al. Significant improvement in cardiometabolic health in healthy nonobese individuals during caloric restriction-induced weight loss and weight loss maintenance. Am J Physiol Endocrinol Metab. 2018;314:E396–405. 10.1152/ajpendo.00261.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kraus WE, et al. 2 years of calorie restriction and cardiometabolic risk (CALERIE): exploratory outcomes of a multicentre, phase 2, randomised controlled trial. Lancet Diabetes Endocrinol. 2019;7:673–83. 10.1016/S2213-8587(19)30151-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Das JK, et al. Calorie restriction modulates the transcription of genes related to stress response and longevity in human muscle: the CALERIE study. Aging Cell. 2023;22:e13963. 10.1111/acel.13963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Meydani SN, et al. Long-term moderate calorie restriction inhibits inflammation without impairing cell-mediated immunity: a randomized controlled trial in non-obese humans. Aging. 2016;8:1416–31. 10.18632/aging.100994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Mirzaei H, Raynes R, Longo VD. The conserved role of protein restriction in aging and disease. Curr Opin Clin Nutr Metab Care. 2016;19:74–9. 10.1097/MCO.0000000000000239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Jiang JC, Jaruga E, Repnevskaya MV, Jazwinski SM. An intervention resembling caloric restriction prolongs life span and retards aging in yeast. FASEB J. 2000;14:2135–7. 10.1096/fj.00-0242fje. [DOI] [PubMed] [Google Scholar]
- 42.Ruckenstuhl C, et al. Lifespan extension by methionine restriction requires autophagy-dependent vacuolar acidification. PLoS Genet. 2014;10:e1004347. 10.1371/journal.pgen.1004347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wu Z, Liu SQ, Huang D. Dietary restriction depends on nutrient composition to extend chronological lifespan in budding yeast Saccharomyces cerevisiae. PLoS ONE. 2013;8:e64448. 10.1371/journal.pone.0064448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Johnson JE, Johnson FB. Methionine restriction activates the retrograde response and confers both stress tolerance and lifespan extension to yeast, mouse and human cells. PLoS ONE. 2014;9:e97729. 10.1371/journal.pone.0097729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Powers RW 3rd, Kaeberlein M, Caldwell SD, Kennedy BK, Fields S. Extension of chronological life span in yeast by decreased TOR pathway signaling. Genes Dev. 2006;20:174–84. 10.1101/gad.1381406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Koc A, Gasch AP, Rutherford JC, Kim HY, Gladyshev VN. Methionine sulfoxide reductase regulation of yeast lifespan reveals reactive oxygen species-dependent and -independent components of aging. Proc Natl Acad Sci U S A. 2004;101:7999–8004. 10.1073/pnas.0307929101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Cabreiro F, et al. Metformin retards aging in C. elegans by altering microbial folate and methionine metabolism. Cell. 2013;153:228–39. 10.1016/j.cell.2013.02.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Troen AM, et al. Lifespan modification by glucose and methionine in Drosophila melanogaster fed a chemically defined diet. Age. 2007;29:29–39. 10.1007/s11357-006-9018-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Juricic P, Gronke S, Partridge L. Branched-chain amino acids have equivalent effects to other essential amino acids on lifespan and aging-related traits in Drosophila. J Gerontol A Biol Sci Med Sci. 2020;75:24–31. 10.1093/gerona/glz080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kosakamoto H, et al. Early-adult methionine restriction reduces methionine sulfoxide and extends lifespan in Drosophila. Nat Commun. 2023;14:7832. 10.1038/s41467-023-43550-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lee BC, et al. Methionine restriction extends lifespan of Drosophila melanogaster under conditions of low amino-acid status. Nat Commun. 2014;5:3592. 10.1038/ncomms4592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mair W, Piper MD, Partridge L. Calories do not explain extension of life span by dietary restriction in Drosophila. PLoS Biol. 2005;3:e223. 10.1371/journal.pbio.0030223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Grandison RC, Piper MD, Partridge L. Amino-acid imbalance explains extension of lifespan by dietary restriction in Drosophila. Nature. 2009;462:1061–4. 10.1038/nature08619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Stoltzner G. Effects of life-long dietary protein restriction on mortality, growth, organ weights, blood counts, liver aldolase and kidney catalase in Balb/C mice. Growth. 1977;41:337–48. [PubMed] [Google Scholar]
- 55.Yu BP, Masoro EJ, McMahan CA. Nutritional influences on aging of Fischer 344 rats: I. Physical, metabolic, and longevity characteristics. J Gerontol. 1985;40:657–70. 10.1093/geronj/40.6.657. [DOI] [PubMed] [Google Scholar]
- 56.Orentreich N, Matias JR, DeFelice A, Zimmerman JA. Low methionine ingestion by rats extends life span. J Nutr. 1993;123:269–74. [DOI] [PubMed] [Google Scholar]
- 57.Miller RA, et al. Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell. 2005;4:119–25. 10.1111/j.1474-9726.2005.00152.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zimmerman JA, Malloy V, Krajcik R, Orentreich N. Nutritional control of aging. Exp Gerontol. 2003;38:47–52. 10.1016/s0531-5565(02)00149-3. [DOI] [PubMed] [Google Scholar]
- 59.Fang H, Stone KP, Wanders D, Forney LA, Gettys TW. The origins, evolution, and future of dietary methionine restriction. Annu Rev Nutr. 2022;42:201–26. 10.1146/annurev-nutr-062320-111849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ayala V, et al. Dietary protein restriction decreases oxidative protein damage, peroxidizability index, and mitochondrial complex I content in rat liver. J Gerontol A Biol Sci Med Sci. 2007;62:352–60. 10.1093/gerona/62.4.352. [DOI] [PubMed] [Google Scholar]
- 61.Trautman ME, Richardson NE, Lamming DW. Protein restriction and branched-chain amino acid restriction promote geroprotective shifts in metabolism. Aging Cell. 2022;21:e13626. 10.1111/acel.13626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Richardson NE, et al. Lifelong restriction of dietary branched-chain amino acids has sex-specific benefits for frailty and lifespan in mice. Nat Aging. 2021;1:73–86. 10.1038/s43587-020-00006-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Forney LA, et al. Sexually dimorphic effects of dietary methionine restriction are dependent on age when the diet is introduced. Obesity. 2020;28:581–9. 10.1002/oby.22721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.De Marte ML, Enesco HE. Influence of low tryptophan diet on survival and organ growth in mice. Mech Ageing Dev. 1986;36:161–71. 10.1016/0047-6374(86)90017-5. [DOI] [PubMed] [Google Scholar]
- 65.Segall PE, Timiras PS. Patho-physiologic findings after chronic tryptophan deficiency in rats: a model for delayed growth and aging. Mech Ageing Dev. 1976;5:109–24. 10.1016/0047-6374(76)90012-9. [DOI] [PubMed] [Google Scholar]
- 66.Ooka H, Segall PE, Timiras PS. Histology and survival in age-delayed low-tryptophan-fed rats. Mech Ageing Dev. 1988;43:79–98. 10.1016/0047-6374(88)90099-1. [DOI] [PubMed] [Google Scholar]
- 67.He C, et al. Enhanced longevity by ibuprofen, conserved in multiple species, occurs in yeast through inhibition of tryptophan import. PLoS Genet. 2014;10:e1004860. 10.1371/journal.pgen.1004860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Levine ME, et al. Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 2014;19:407–17. 10.1016/j.cmet.2014.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Song M, et al. Association of animal and plant protein intake with all-cause and cause-specific mortality. JAMA Intern Med. 2016;176:1453–63. 10.1001/jamainternmed.2016.4182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Fontana L, et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 2016;16:520–30. 10.1016/j.celrep.2016.05.092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Nicolaisen TS, et al. Dietary protein restriction elevates FGF21 levels and energy requirements to maintain body weight in lean men. Nat Metab. 2025;7:602–16. 10.1038/s42255-025-01236-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Honjoh S, Yamamoto T, Uno M, Nishida E. Signalling through RHEB-1 mediates intermittent fasting-induced longevity in C. elegans. Nature. 2009;457:726–30. 10.1038/nature07583. [DOI] [PubMed] [Google Scholar]
- 73.Catterson JH, et al. Short-term, intermittent fasting induces long-lasting gut health and TOR-independent lifespan extension. Curr Biol. 2018. 10.1016/j.cub.2018.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ulgherait M, et al. Circadian autophagy drives iTRF-mediated longevity. Nature. 2021;598:353–8. 10.1038/s41586-021-03934-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Goodrick CL, Ingram DK, Reynolds MA, Freeman JR, Cider N. Effects of intermittent feeding upon body weight and lifespan in inbred mice: interaction of genotype and age. Mech Ageing Dev. 1990;55:69–87. 10.1016/0047-6374(90)90107-q. [DOI] [PubMed] [Google Scholar]
- 76.Mitchell SJ, et al. Daily fasting improves health and survival in male mice independent of diet composition and calories. Cell Metab. 2019. 10.1016/j.cmet.2018.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Brandhorst S, et al. A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metab. 2015;22:86–99. 10.1016/j.cmet.2015.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Goodrick CL, Ingram DK, Reynolds MA, Freeman JR, Cider NL. Effects of intermittent feeding upon growth and life span in rats. Gerontology. 1982;28:233–41. 10.1159/000212538. [DOI] [PubMed] [Google Scholar]
- 79.Mattson MP, Longo VD, Harvie M. Impact of intermittent fasting on health and disease processes. Ageing Res Rev. 2017;39:46–58. 10.1016/j.arr.2016.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Longo VD, Di Tano M, Mattson MP, Guidi N. Intermittent and periodic fasting, longevity and disease. Nat Aging. 2021;1:47–59. 10.1038/s43587-020-00013-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Anson RM, et al. Intermittent fasting dissociates beneficial effects of dietary restriction on glucose metabolism and neuronal resistance to injury from calorie intake. Proc Natl Acad Sci USA. 2003;100:6216–20. 10.1073/pnas.1035720100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Liu B, et al. Intermittent fasting improves glucose tolerance and promotes adipose tissue remodeling in male mice fed a high-fat diet. Endocrinology. 2019;160:169–80. 10.1210/en.2018-00701. [DOI] [PubMed] [Google Scholar]
- 83.Liang BJ, et al. Intermittent fasting therapy promotes insulin sensitivity by inhibiting NLRP3 inflammasome in rat model. Ann Palliat Med. 2021;10:5299–309. 10.21037/apm-20-2410. [DOI] [PubMed] [Google Scholar]
- 84.Hatori M, et al. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab. 2012;15:848–60. 10.1016/j.cmet.2012.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Varady KA, Hellerstein MK. Alternate-day fasting and chronic disease prevention: a review of human and animal trials. Am J Clin Nutr. 2007;86:7–13. 10.1093/ajcn/86.1.7. [DOI] [PubMed] [Google Scholar]
- 86.Whittaker DS, et al. Circadian modulation by time-restricted feeding rescues brain pathology and improves memory in mouse models of Alzheimer’s disease. Cell Metab. 2023. 10.1016/j.cmet.2023.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Dogan S, Rogozina OP, Lokshin AE, Grande JP, Cleary MP. Effects of chronic vs. intermittent calorie restriction on mammary tumor incidence and serum adiponectin and leptin levels in MMTV-TGF-α mice at different ages. Oncol Lett. 2010;1:167–76. 10.3892/ol_00000031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kim YH, et al. Thermogenesis-independent metabolic benefits conferred by isocaloric intermittent fasting in ob/ob mice. Sci Rep. 2019;9:2479. 10.1038/s41598-019-39380-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.de Cabo R, Mattson MP. Effects of intermittent fasting on health, aging, and disease. N Engl J Med. 2019;381:2541–51. 10.1056/NEJMra1905136. [DOI] [PubMed] [Google Scholar]
- 90.Sun ML, et al. Intermittent fasting and health outcomes: an umbrella review of systematic reviews and meta-analyses of randomised controlled trials. E Clin Med. 2024;70:102519. 10.1016/j.eclinm.2024.102519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Patterson RE, et al. Intermittent fasting and human metabolic health. J Acad Nutr Diet. 2015;115:1203–12. 10.1016/j.jand.2015.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Sutton EF, et al. Early time-restricted feeding improves insulin sensitivity, blood pressure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab. 2018. 10.1016/j.cmet.2018.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wilkinson MJ, et al. Ten-hour time-restricted eating reduces weight, blood pressure, and atherogenic lipids in patients with metabolic syndrome. Cell Metab. 2020. 10.1016/j.cmet.2019.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kapogiannis D, et al. Brain responses to intermittent fasting and the healthy living diet in older adults. Cell Metab. 2024;36:1900–4. 10.1016/j.cmet.2024.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Yuan Y, et al. Enhanced energy metabolism contributes to the extended life span of calorie-restricted Caenorhabditis elegans. J Biol Chem. 2012;287:31414–26. 10.1074/jbc.M112.377275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Katewa SD, et al. Intramyocellular fatty-acid metabolism plays a critical role in mediating responses to dietary restriction in Drosophila melanogaster. Cell Metab. 2012;16:97–103. 10.1016/j.cmet.2012.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Bruss MD, Khambatta CF, Ruby MA, Aggarwal I, Hellerstein MK. Calorie restriction increases fatty acid synthesis and whole body fat oxidation rates. Am J Physiol Endocrinol Metab. 2010;298:E108-116. 10.1152/ajpendo.00524.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Haran A, et al. Differential effects of bariatric surgery and caloric restriction on hepatic one-carbon and fatty acid metabolism. iScience. 2023;26:107046. 10.1016/j.isci.2023.107046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wang R, et al. Periodic protein-restricted diets extend the lifespan of high-fat diet-induced Drosophila melanogaster males. Aging Cell. 2024;23:e14327. 10.1111/acel.14327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hasek BE, et al. Dietary methionine restriction enhances metabolic flexibility and increases uncoupled respiration in both fed and fasted states. Am J Physiol Regul Integr Comp Physiol. 2010;299:R728-739. 10.1152/ajpregu.00837.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Mutlu AS, Duffy J, Wang MC. Lipid metabolism and lipid signals in aging and longevity. Dev Cell. 2021;56:1394–407. 10.1016/j.devcel.2021.03.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Watanabe K, et al. Lifespan-extending interventions induce consistent patterns of fatty acid oxidation in mouse livers. Commun Biol. 2023;6:768. 10.1038/s42003-023-05128-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ramachandran PV, et al. Lysosomal signaling promotes longevity by adjusting mitochondrial activity. Dev Cell. 2019. 10.1016/j.devcel.2018.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Mourikis P, Hurlbut GD, Artavanis-Tsakonas S. Enigma, a mitochondrial protein affecting lifespan and oxidative stress response in Drosophila. Proc Natl Acad Sci U S A. 2006;103:1307–12. 10.1073/pnas.0510564103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017;25:262–84. 10.1016/j.cmet.2016.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Tsuruta H, Yamahara K, Yasuda-Yamahara M, Kume S. Emerging pathophysiological roles of ketone bodies. Physiology. 2024. 10.1152/physiol.00031.2023. [DOI] [PubMed] [Google Scholar]
- 107.Tomita I, et al. Ketone bodies: a double-edged sword for mammalian life span. Aging Cell. 2023;22:e13833. 10.1111/acel.13833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Barbieri M, Bonafe M, Franceschi C, Paolisso G. Insulin/IGF-I-signaling pathway: an evolutionarily conserved mechanism of longevity from yeast to humans. Am J Physiol Endocrinol Metab. 2003;285:E1064-1071. 10.1152/ajpendo.00296.2003. [DOI] [PubMed] [Google Scholar]
- 109.Martins R, Lithgow GJ, Link W. Long live FOXO: unraveling the role of FOXO proteins in aging and longevity. Aging Cell. 2016;15:196–207. 10.1111/acel.12427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Dos Santos C, et al. Calorie restriction increases insulin sensitivity to promote beta cell homeostasis and longevity in mice. Nat Commun. 2024;15:9063. 10.1038/s41467-024-53127-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lariviere F, Chiasson JL, Schiffrin A, Taveroff A, Hoffer LJ. Effects of dietary protein restriction on glucose and insulin metabolism in normal and diabetic humans. Metabolism. 1994;43:462–7. 10.1016/0026-0495(94)90077-9. [DOI] [PubMed] [Google Scholar]
- 112.Malloy VL, et al. Methionine restriction decreases visceral fat mass and preserves insulin action in aging male Fischer 344 rats independent of energy restriction. Aging Cell. 2006;5:305–14. 10.1111/j.1474-9726.2006.00220.x. [DOI] [PubMed] [Google Scholar]
- 113.Shimokawa I, et al. The life-extending effect of dietary restriction requires Foxo3 in mice. Aging Cell. 2015;14:707–9. 10.1111/acel.12340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Kim DH, Kim JY, Yu BP, Chung HY. The activation of NF-kappaB through Akt-induced FOXO1 phosphorylation during aging and its modulation by calorie restriction. Biogerontology. 2008;9:33–47. 10.1007/s10522-007-9114-6. [DOI] [PubMed] [Google Scholar]
- 115.Liu GY, Sabatini DM. Mtor at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21:183–203. 10.1038/s41580-019-0199-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Johnson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and age-related disease. Nature. 2013;493:338–45. 10.1038/nature11861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Mannick JB, Lamming DW. Targeting the biology of aging with mTOR inhibitors. Nat Aging. 2023;3:642–60. 10.1038/s43587-023-00416-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Wu Y, et al. Very-low-protein diets lead to reduced food intake and weight loss, linked to inhibition of hypothalamic mTOR signaling, in mice. Cell Metab. 2021;33:1264–6. 10.1016/j.cmet.2021.04.016. [DOI] [PubMed] [Google Scholar]
- 119.Lamming DW, et al. Restriction of dietary protein decreases mTORC1 in tumors and somatic tissues of a tumor-bearing mouse xenograft model. Oncotarget. 2015;6:31233–40. 10.18632/oncotarget.5180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Alirezaei M, et al. Short-term fasting induces profound neuronal autophagy. Autophagy. 2010;6:702–10. 10.4161/auto.6.6.12376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Sengupta S, Peterson TR, Laplante M, Oh S, Sabatini DM. mTORC1 controls fasting-induced ketogenesis and its modulation by ageing. Nature. 2010;468:1100–4. 10.1038/nature09584. [DOI] [PubMed] [Google Scholar]
- 122.Igarashi M, Guarente L. mTORC1 and SIRT1 cooperate to foster expansion of gut adult stem cells during calorie restriction. Cell. 2016;166:436–50. 10.1016/j.cell.2016.05.044. [DOI] [PubMed] [Google Scholar]
- 123.Kim HS, Pickering AM. Protein translation paradox: implications in translational regulation of aging. Front Cell Dev Biol. 2023;11:1129281. 10.3389/fcell.2023.1129281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Jia K, Levine B. Autophagy is required for dietary restriction-mediated life span extension in C. elegans. Autophagy. 2007;3:597–9. 10.4161/auto.4989. [DOI] [PubMed] [Google Scholar]
- 125.Hansen M, et al. A role for autophagy in the extension of lifespan by dietary restriction in C. elegans. PLoS Genet. 2008;4:e24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Herzig S, Shaw RJ. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 2018;19:121–35. 10.1038/nrm.2017.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Greer EL, et al. An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Curr Biol. 2007;17:1646–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kuo CT, et al. AMPK-mediated formation of stress granules is required for dietary restriction-induced longevity in Caenorhabditis elegans. Aging Cell. 2020;19:e13157. 10.1111/acel.13157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Stenesen D, et al. Adenosine nucleotide biosynthesis and AMPK regulate adult life span and mediate the longevity benefit of caloric restriction in flies. Cell Metab. 2013;17:101–12. 10.1016/j.cmet.2012.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Apfeld J, O’Connor G, McDonagh T, DiStefano PS, Curtis R. The AMP-activated protein kinase AAK-2 links energy levels and insulin-like signals to lifespan in C. elegans. Genes Dev. 2004;18:3004–9. 10.1101/gad.1255404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Shinmura K, et al. Cardioprotective effects of short-term caloric restriction are mediated by adiponectin via activation of AMP-activated protein kinase. Circulation. 2007;116:2809–17. 10.1161/CIRCULATIONAHA.107.725697. [DOI] [PubMed] [Google Scholar]
- 132.Zheng A, et al. Greater phosphorylation of AMPK and multiple AMPK substrates in the skeletal muscle of 24-month-old calorie restricted compared to ad-libitum fed male rats. J Gerontol A Biol Sci Med Sci. 2023;78:177–85. 10.1093/gerona/glac218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.de Lange P, et al. Sequential changes in the signal transduction responses of skeletal muscle following food deprivation. FASEB J. 2006;20:2579–81. 10.1096/fj.06-6025fje. [DOI] [PubMed] [Google Scholar]
- 134.Li Y, Chen Y. AMPK and autophagy. Adv Exp Med Biol. 2019;1206:85–108. 10.1007/978-981-15-0602-4_4. [DOI] [PubMed] [Google Scholar]
- 135.Johanns M, et al. Direct and indirect activation of eukaryotic elongation factor 2 kinase by AMP-activated protein kinase. Cell Signal. 2017;36:212–21. 10.1016/j.cellsig.2017.05.010. [DOI] [PubMed] [Google Scholar]
- 136.Wu QJ, et al. The sirtuin family in health and disease. Signal Transduct Target Ther. 2022;7:402. 10.1038/s41392-022-01257-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kaeberlein M, McVey M, Guarente L. The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes Dev. 1999;13:2570–80. 10.1101/gad.13.19.2570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Rogina B, Helfand SL. Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proc Natl Acad Sci U S A. 2004;101:15998–6003. 10.1073/pnas.0404184101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Wang Y, Tissenbaum HA. Overlapping and distinct functions for a Caenorhabditis elegans SIR2 and DAF-16/FOXO. Mech Ageing Dev. 2006;127:48–56. 10.1016/j.mad.2005.09.005. [DOI] [PubMed] [Google Scholar]
- 140.Mercken EM, et al. SIRT1 but not its increased expression is essential for lifespan extension in caloric-restricted mice. Aging Cell. 2014;13:193–6. 10.1111/acel.12151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Liu Y, et al. A fasting inducible switch modulates gluconeogenesis via activator/coactivator exchange. Nature. 2008;456:269–73. 10.1038/nature07349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Herranz D, et al. Sirt1 improves healthy ageing and protects from metabolic syndrome-associated cancer. Nat Commun. 2010;1:3. 10.1038/ncomms1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Satoh A, et al. Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 2013;18:416–30. 10.1016/j.cmet.2013.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Chang HC, Guarente L. SIRT1 and other sirtuins in metabolism. Trends Endocrinol Metab. 2014;25:138–45. 10.1016/j.tem.2013.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Levine DC, et al. NADH inhibition of SIRT1 links energy state to transcription during time-restricted feeding. Nat Metab. 2021;3:1621–32. 10.1038/s42255-021-00498-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.You Y, Liang W. SIRT1 and SIRT6: the role in aging-related diseases. Biochim Biophys Acta Mol Basis Dis. 2023;1869:166815. 10.1016/j.bbadis.2023.166815. [DOI] [PubMed] [Google Scholar]
- 147.Li Y, Jin J, Wang Y. SIRT6 widely regulates aging, immunity, and cancer. Front Oncol. 2022;12:861334. 10.3389/fonc.2022.861334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Korotkov A, Seluanov A, Gorbunova V. Sirtuin 6: linking longevity with genome and epigenome stability. Trends Cell Biol. 2021;31:994–1006. 10.1016/j.tcb.2021.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Lin SJ, Defossez PA, Guarente L. Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. Science. 2000;289:2126–8. [DOI] [PubMed] [Google Scholar]
- 150.Moroz N, et al. Dietary restriction involves NAD(+) -dependent mechanisms and a shift toward oxidative metabolism. Aging Cell. 2014;13:1075–85. 10.1111/acel.12273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Medvedik O, Lamming DW, Kim KD, Sinclair DA. MSN2 and MSN4 link calorie restriction and TOR to sirtuin-mediated lifespan extension in Saccharomyces cerevisiae. PLoS Biol. 2007;5:e261. 10.1371/journal.pbio.0050261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Anderson RM, Bitterman KJ, Wood JG, Medvedik O, Sinclair DA. Nicotinamide and PNC1 govern lifespan extension by calorie restriction in Saccharomyces cerevisiae. Nature. 2003;423:181–5. 10.1038/nature01578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Yoshida M, et al. Extracellular vesicle-contained eNAMPT delays aging and extends lifespan in mice. Cell Metab. 2019. 10.1016/j.cmet.2019.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Covarrubias AJ, Perrone R, Grozio A, Verdin E. NAD(+) metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cell Biol. 2021;22:119–41. 10.1038/s41580-020-00313-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Ravindran R, et al. The amino acid sensor GCN2 controls gut inflammation by inhibiting inflammasome activation. Nature. 2016;531:523–7. 10.1038/nature17186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Ye J, et al. The GCN2-ATF4 pathway is critical for tumour cell survival and proliferation in response to nutrient deprivation. EMBO J. 2010;29:2082–96. 10.1038/emboj.2010.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Bunpo P, et al. The eIF2 kinase GCN2 is essential for the murine immune system to adapt to amino acid deprivation by asparaginase. J Nutr. 2010;140:2020–7. 10.3945/jn.110.129197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Vattem KM, Wek RC. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc Natl Acad Sci U S A. 2004;101:11269–74. 10.1073/pnas.0400541101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Harding HP, et al. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell. 2000;6:1099–108. 10.1016/s1097-2765(00)00108-8. [DOI] [PubMed] [Google Scholar]
- 160.Dever TE, et al. Phosphorylation of initiation factor 2 alpha by protein kinase GCN2 mediates gene-specific translational control of GCN4 in yeast. Cell. 1992;68:585–96. 10.1016/0092-8674(92)90193-g. [DOI] [PubMed] [Google Scholar]
- 161.De Sousa-Coelho AL, Marrero PF, Haro D. Activating transcription factor 4-dependent induction of FGF21 during amino acid deprivation. Biochem J. 2012;443:165–71. 10.1042/BJ20111748. [DOI] [PubMed] [Google Scholar]
- 162.Rousakis A, et al. The general control nonderepressible-2 kinase mediates stress response and longevity induced by target of rapamycin inactivation in Caenorhabditis elegans. Aging Cell. 2013;12:742–51. 10.1111/acel.12101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Laeger T, et al. Metabolic responses to dietary protein restriction require an increase in FGF21 that is delayed by the absence of GCN2. Cell Rep. 2016;16:707–16. 10.1016/j.celrep.2016.06.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Laeger T, et al. FGF21 is an endocrine signal of protein restriction. J Clin Invest. 2014;124:3913–22. 10.1172/JCI74915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Nishimura T, Nakatake Y, Konishi M, Itoh N. Identification of a novel FGF, FGF-21, preferentially expressed in the liver. Biochim Biophys Acta. 2000;1492:203–6. 10.1016/s0167-4781(00)00067-1. [DOI] [PubMed] [Google Scholar]
- 166.Hill CM, et al. FGF21 signals protein status to the brain and adaptively regulates food choice and metabolism. Cell Rep. 2019. 10.1016/j.celrep.2019.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Zhang Y, et al. The starvation hormone, fibroblast growth factor-21, extends lifespan in mice. Elife. 2012;1:e00065. 10.7554/eLife.00065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Hill CM, et al. FGF21 is required for protein restriction to extend lifespan and improve metabolic health in male mice. Nat Commun. 2022;13:1897. 10.1038/s41467-022-29499-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Moro T, et al. Effects of eight weeks of time-restricted feeding (16/8) on basal metabolism, maximal strength, body composition, inflammation, and cardiovascular risk factors in resistance-trained males. J Transl Med. 2016;14:290. 10.1186/s12967-016-1044-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Rubinsztein DC, Marino G, Kroemer G. Autophagy and aging. Cell. 2011;146:682–95. 10.1016/j.cell.2011.07.030. [DOI] [PubMed] [Google Scholar]
- 171.Mannick JB, et al. Mtor inhibition improves immune function in the elderly. Sci Transl Med. 2014;6:268ra179. 10.1126/scitranslmed.3009892. [DOI] [PubMed] [Google Scholar]
- 172.Lamming DW, et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science. 2012;335:1638–43. 10.1126/science.1215135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Baghdadi M, et al. Intermittent rapamycin feeding recapitulates some effects of continuous treatment while maintaining lifespan extension. Mol Metab. 2024;81:101902. 10.1016/j.molmet.2024.101902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Lee DJW, Hodzic Kuerec A, Maier AB. Targeting ageing with rapamycin and its derivatives in humans: a systematic review. Lancet Healthy Longev. 2024;5:e152–62. 10.1016/S2666-7568(23)00258-1. [DOI] [PubMed] [Google Scholar]
- 175.Soukas AA, Hao H, Wu L. Metformin as anti-aging therapy: is it for everyone? Trends Endocrinol Metab. 2019;30:745–55. 10.1016/j.tem.2019.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Han Y, et al. Effect of metformin on all-cause and cardiovascular mortality in patients with coronary artery diseases: a systematic review and an updated meta-analysis. Cardiovasc Diabetol. 2019;18:96. 10.1186/s12933-019-0900-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Cao X, Wu Y, Wang J, Liu K, Wang X. The effect of metformin on mortality among diabetic cancer patients: a systematic review and meta-analysis. JNCI Cancer Spectr. 2017;1:pkx007. 10.1093/jncics/pkx007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Gandini S, et al. Metformin and cancer risk and mortality: a systematic review and meta-analysis taking into account biases and confounders. Cancer Prev Res. 2014;7:867–85. 10.1158/1940-6207.CAPR-13-0424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Zhang Y, et al. Metformin and the risk of neurodegenerative diseases in patients with diabetes: a meta-analysis of population-based cohort studies. Diabet Med. 2022;39:e14821. 10.1111/dme.14821. [DOI] [PubMed] [Google Scholar]
- 180.Ji S, et al. Metformin and the risk of dementia based on an analysis of 396,332 participants. Ther Adv Chronic Dis. 2022;13:20406223221109454. 10.1177/20406223221109454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA. Metformin as a tool to target aging. Cell Metab. 2016;23:1060–5. 10.1016/j.cmet.2016.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Reifsnyder PC, et al. Rapamycin/metformin co-treatment normalizes insulin sensitivity and reduces complications of metabolic syndrome in type 2 diabetic mice. Aging Cell. 2022;21:e13666. 10.1111/acel.13666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Huang X, et al. Reducing signs of aging and increasing lifespan by drug synergy. Aging Cell. 2013;12:652–60. 10.1111/acel.12090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Gkioni L, et al. The geroprotectors trametinib and rapamycin combine additively to extend mouse healthspan and lifespan. Nat Aging. 2025. 10.1038/s43587-025-00876-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Crawford D, Libina N, Kenyon C. Caenorhabditis elegans integrates food and reproductive signals in lifespan determination. Aging Cell. 2007;6:715–21. 10.1111/j.1474-9726.2007.00327.x. [DOI] [PubMed] [Google Scholar]
- 186.Chippindale AK, Leroi AM, Kim SB, Rose MR. Phenotypic plasticity and selection in Drosophila life-history evolution. 1. nutrition and the cost of reproduction. J Evolution Biol. 1993;6:171–93. 10.1046/j.1420-9101.1993.6020171.x. [Google Scholar]
- 187.Moatt JP, Nakagawa S, Lagisz M, Walling CA. The effect of dietary restriction on reproduction: a meta-analytic perspective. BMC Evol Biol. 2016;16:199. 10.1186/s12862-016-0768-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Mair W, Sgro CM, Johnson AP, Chapman T, Partridge L. Lifespan extension by dietary restriction in female Drosophila melanogaster is not caused by a reduction in vitellogenesis or ovarian activity. Exp Gerontol. 2004;39:1011–9. 10.1016/j.exger.2004.03.018. [DOI] [PubMed] [Google Scholar]
- 189.Menniti G, et al. Mental health consequences of dietary restriction: increased depressive symptoms in biological men and populations with elevated BMI. BMJ Nutr Prev Health. 2025;8:e001167. 10.1136/bmjnph-2025-001167. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.

