A Comprehensive Report on Methionine Restriction, Glycine Mimicry, and Associated Metabolic Modulators
I. Executive Summary
Methionine Restriction (MR), the deliberate reduction of the essential amino acid methionine in the diet, stands as one of the most robust and consistently replicated interventions for extending healthspan and lifespan in a diverse array of model organisms, from yeast to rodents. Its effects are profound, often rivaling or exceeding those of caloric restriction, yet it can be achieved without a net reduction in caloric intake. The benefits of MR are not isolated to longevity alone; they encompass a broad spectrum of health improvements, including enhanced metabolic flexibility, reduced adiposity, increased insulin sensitivity, potent anti-cancer effects, and neuroprotection. These outcomes are not coincidental but are the result of a fundamental reprogramming of cellular metabolism and signaling. The core mechanism hinges on the reduction of intracellular S-adenosylmethionine (SAM), the body's principal methyl donor, which in turn modulates key nutrient-sensing and stress-response pathways. Specifically, MR attenuates the activity of the mechanistic target of rapamycin complex 1 (mTORC1) and lowers circulating levels of insulin-like growth factor-1 (IGF-1), two of the most critical hubs in the regulation of aging. Concurrently, MR potently induces the production of the beneficial metabolic hormone fibroblast growth factor 21 (FGF21) and enhances cellular housekeeping processes like autophagy, while mitigating oxidative stress.
While direct dietary MR in humans is challenging due to compliance issues and the risk of malnutrition, a compelling and mechanistically sound strategy has emerged to capture its benefits: high-dose glycine supplementation. This report delineates the extensive evidence suggesting that glycine can function as a powerful MR mimetic. The primary biochemical link is the enzyme Glycine N-Methyltransferase (GNMT), which is abundant in the liver. Glycine serves as the key substrate for GNMT, which catalyzes its methylation into sarcosine. This reaction consumes SAM. By providing a surplus of glycine, this pathway is driven forward, creating a metabolic "sink" that actively depletes the hepatic SAM pool. This depletion of SAM effectively replicates the central biochemical event of dietary MR, thereby triggering the same cascade of downstream pro-longevity signaling events—namely, the inhibition of mTORC1, the reduction of IGF-1, and the induction of FGF21.
The success of this glycine-based strategy, however, is critically dependent on a nuanced understanding of the surrounding metabolic network. The efficacy of the intervention is profoundly influenced by two key modulators: essential cofactors and a direct metabolic antagonist.
First, the increased metabolic flux through the methionine cycle, instigated by high-dose glycine, places a significant demand on the enzymes responsible for processing metabolic intermediates. This necessitates an adequate supply of B-vitamin cofactors—specifically folate (B9), cobalamin (B12), and pyridoxine (B6)—to ensure the cycle runs efficiently and to prevent the accumulation of potentially deleterious byproducts like homocysteine. Additional support from methyl donors such as betaine (trimethylglycine) may provide further synergistic benefits. These cofactors are not merely supportive but are integral to the safety and efficacy of the protocol.
Second, and of paramount importance, is the direct metabolic antagonism posed by creatine supplementation. Endogenous creatine synthesis is one of the single largest consumers of SAM in the body. Supplementing with exogenous creatine powerfully suppresses this endogenous production via feedback inhibition, thereby "sparing" the SAM pool. This action is in direct biochemical opposition to the goal of the glycine strategy, which is to deplete SAM. Co-supplementation of creatine and glycine for the purpose of mimicking MR is therefore mechanistically contradictory and self-defeating.
In conclusion, this report synthesizes the vast body of preclinical and clinical evidence to build a comprehensive case for glycine as a practical MR mimetic. A carefully designed protocol involving high-dose glycine supplementation, supported by essential B-vitamin cofactors and coupled with the strict avoidance of supplemental creatine, represents a highly plausible, evidence-based, and actionable strategy for capturing a significant portion of the profound health and longevity benefits of dietary methionine restriction in humans.
II. The Foundational Science and Health Implications of Methionine Restriction
A. The Methionine Cycle: A Master Regulator of Metabolism and Aging
The profound effects of methionine restriction can only be understood through a detailed examination of the biochemical pathways it governs. Methionine is not merely a building block for proteins; it sits at the nexus of several fundamental metabolic processes that are intimately linked to cellular health, epigenetic regulation, and the aging process itself. These interconnected pathways—the Methionine Cycle, the Folate Cycle, and the Transsulfuration Pathway—collectively form the engine of one-carbon metabolism, a system responsible for the transfer of single-carbon methyl groups that are vital for life.1
One-Carbon Metabolism: The Folate and Methionine Cycles
One-carbon metabolism comprises a set of biochemical reactions that manage the transfer of one-carbon units, primarily in the form of methyl groups (CH3). This system is driven by two interlocking cycles: the Folate Cycle and the Methionine Cycle.1 The Folate Cycle, utilizing various forms of vitamin B9 (folate), processes one-carbon units derived from amino acids like serine. The Methionine Cycle, in turn, accepts these methyl groups to regenerate methionine, which then becomes "activated" to fuel methylation reactions throughout the body. The seamless integration of these two cycles is essential for a vast array of cellular functions, including the synthesis of nucleotides (the building blocks of DNA), the regulation of gene expression via epigenetic marks, and the production of key neurotransmitters.1
S-Adenosylmethionine (SAM): The Universal Methyl Donor
At the heart of the Methionine Cycle lies S-adenosylmethionine, commonly known as SAM or AdoMet. Synthesized from methionine and adenosine triphosphate (ATP), SAM is the principal methyl donor for virtually all methylation reactions in the body.4 It is often referred to as the "activated" form of methionine. When SAM donates its methyl group to a substrate—be it DNA, a histone protein, or a precursor to a neurotransmitter—it is converted into S-adenosylhomocysteine (SAH).7 The ratio of SAM to SAH is a critical barometer of the cell's "methylation potential" or "methylation capacity." A high SAM/SAH ratio indicates a robust capacity to carry out essential methylation reactions, while a low ratio signifies impaired methylation capacity. The regulation of this ratio is a key element in cellular homeostasis, and as will be discussed, its modulation is central to the effects of methionine restriction.4
The Transsulfuration Pathway: Clearance and Antioxidant Synthesis
Once SAH is formed, it is hydrolyzed to homocysteine. Homocysteine stands at a critical metabolic crossroads with two primary fates. The first is remethylation back to methionine, a salvage pathway that conserves this essential amino acid. The second fate is irreversible entry into the transsulfuration pathway.9 This pathway catabolizes homocysteine, converting it first to cystathionine and then to the amino acid cysteine. This serves two vital purposes: it provides a mechanism to clear excess methionine from the body, preventing its accumulation to toxic levels, and it supplies the cysteine necessary for the synthesis of glutathione (GSH).5 Glutathione is the body's most abundant endogenous antioxidant, playing a master role in neutralizing reactive oxygen species (ROS) and protecting cells from oxidative damage.10
Homocysteine Crossroads and B-Vitamin Cofactors
The fate of homocysteine—whether it is remethylated to methionine or shunted into the transsulfuration pathway—is governed by a series of enzymes that depend on specific B-vitamin cofactors.1
Remethylation to Methionine: This reaction is primarily catalyzed by the enzyme methionine synthase (MS), which requires vitamin B12 (cobalamin) as a direct cofactor and receives its methyl group from 5-methyltetrahydrofolate (5-MTHF), the active form of vitamin B9 (folate). The production of 5-MTHF is dependent on the enzyme methylenetetrahydrofolate reductase (MTHFR), which uses vitamin B2 (riboflavin) as a cofactor.1
Transsulfuration to Cysteine: The first and rate-limiting step of this pathway is catalyzed by the enzyme cystathionine β-synthase (CBS), which requires vitamin B6 (pyridoxine) as an essential cofactor.12
The availability of these B-vitamins is therefore critical for maintaining the proper flow of metabolites through the entire one-carbon network. Deficiencies can lead to bottlenecks, most notably the accumulation of homocysteine, a state linked to various pathologies.12 The intricate interplay of these pathways is illustrated in the figure below.
!(https://i.imgur.com/8Qj8m1L.png)
Figure 1: A simplified schematic of one-carbon metabolism, illustrating the interconnected Methionine and Folate cycles, the Transsulfuration pathway, and the critical roles of key amino acids and B-vitamin cofactors. Methionine is converted to SAM, the universal methyl donor. After donating a methyl group, SAM becomes SAH, which is hydrolyzed to homocysteine (Hcy). Hcy can be remethylated back to methionine in reactions requiring Folate (B9) and B12, or it can be irreversibly converted to cysteine (a precursor for glutathione) via the transsulfuration pathway, which requires Vitamin B6. Glycine plays a key role as a substrate for the GNMT enzyme, which consumes SAM, and is also involved in the folate cycle via SHMT.
B. The Pro-Longevity Effects of MR: A Review of the Evidence
Methionine restriction is one of the most powerful dietary interventions known to extend lifespan and improve healthspan in laboratory animals. The effects are not only large in magnitude but are also remarkably consistent across different species and genetic backgrounds, pointing to the manipulation of a deeply conserved, fundamental biological process.15
Magnitude of Lifespan Extension in Rodent Models
The most striking evidence for MR comes from studies in rats and mice, where the intervention has been shown to extend lifespan by as much as 45%.15 One of the foundational studies in this field, conducted by Orentreich and colleagues in 1993, demonstrated that feeding male Fischer 344 rats a diet with 80% less methionine (0.17% methionine by weight compared to a 0.86% control diet) resulted in a 30% to 40% increase in both median and maximum lifespan.18 This dramatic extension of life was achieved despite the fact that the MR animals were slightly hyperphagic, meaning they consumed more food per gram of body weight than their control-fed counterparts.4 This crucial observation, replicated in subsequent studies, definitively separated the effects of MR from those of simple caloric restriction (CR), establishing MR as a distinct and potent longevity intervention. Subsequent research confirmed these findings across various rodent strains, including Sprague-Dawley, Wistar Hannover, and genetically heterogeneous mice, demonstrating that the response is not specific to a single genetic background.4
Cross-Species Conservation of Longevity Benefits
The pro-longevity effects of MR are not a quirk of rodent physiology but are conserved across a vast evolutionary distance. Lifespan extension has been documented in simpler organisms such as budding yeast (Saccharomyces cerevisiae), fruit flies (Drosophila melanogaster), and nematodes (Caenorhabditis elegans).15 Furthermore, MR has been shown to extend the replicative lifespan of cultured human diploid fibroblasts, delaying the onset of cellular senescence.15 This remarkable conservation strongly suggests that MR targets ancient and fundamental mechanisms of aging that are shared among a wide range of eukaryotic organisms, including humans.
Robust Improvements in Healthspan
Beyond simply extending the number of days an organism lives, MR profoundly improves the quality of those days by extending healthspan—the period of life spent free from chronic disease and disability. Animals on MR diets exhibit a delayed onset and reduced incidence of a wide range of age-related pathologies.4 These benefits include:
Improved Metabolic Health: MR leads to lower body weight, reduced adiposity (particularly visceral fat), improved glucose tolerance, and dramatically increased insulin sensitivity.4
Reduced Cancer Incidence: As many tumors are methionine-dependent, MR has been shown to reduce the incidence of spontaneous tumors in aging rodents.19
Preserved Organ Function: MR has been shown to protect against age-related decline in multiple organs, including the liver, heart, and kidneys.24
Critically, these benefits are not limited to lifelong interventions. A recent study demonstrated that initiating MR late in life, in mice already 18 months old (equivalent to a human in their late 50s or early 60s), still conferred significant healthspan benefits. These late-life MR mice showed improved neuromuscular function, better lung function, and a significant reduction in a composite frailty score compared to age-matched controls.26 This finding is particularly significant as it suggests that MR-based strategies could have therapeutic potential for reversing or mitigating existing age-related decline, not just preventing it.
The following table summarizes key findings from several landmark preclinical studies, highlighting the consistent and robust effects of MR on lifespan and healthspan.
Table 1: Summary of Lifespan and Healthspan Effects of Methionine Restriction in Preclinical Models
Species/Strain
MR Level (% Restriction)
Key Lifespan Outcome
Key Healthspan Outcomes
Key Mechanistic Changes
Reference(s)
Fischer 344 Rats
80% (0.17% diet)
30-40% increase in median & max lifespan
Reduced adiposity, lower incidence of lethal diseases
↓ IGF-1
18
(BALB/cJ × C57BL/6J) F1 Mice
83% (0.12% diet)
~30% increase in median lifespan
Lower body weight, reduced visceral fat, preserved insulin sensitivity
↓ Plasma IGF-1, insulin, glucose, T4
4
Multiple Rat Strains (BN, SD, WH)
80%
~45% increase in lifespan
Reduced age-related pathologies
-
15
Drosophila melanogaster
Varies
Lifespan extension (context-dependent)
Mimics dietary restriction effects
↓ Reproduction, involves TOR signaling
16
C57BL/6J Mice (Late-life)
83% (started at 18 mo)
N/A
Improved neuromuscular function, lung function, metabolic health; reduced frailty
-
26
Human Diploid Fibroblasts
Genetic MR
Extended replicative lifespan
Delayed cellular senescence, increased stress resistance
↑ NF-κB signaling
15
C. Core Mechanisms of Action: How MR Rewires the Aging Process
The remarkable health and longevity benefits of methionine restriction stem from its ability to trigger a coordinated, multi-systemic response that fundamentally alters cellular signaling and metabolism. This response is not a simple reaction to nutrient scarcity but a highly specific program initiated by the cell's sensing of low methionine availability. This program converges on three of the most well-established pillars of aging research: nutrient-sensing pathways, endocrine signaling, and cellular maintenance systems.
1. Nutrient Sensing and Growth Pathways (mTOR and IGF-1)
At the highest level, MR acts by modulating the primary pathways that link nutrient availability to cell growth, proliferation, and survival.
mTORC1 Inhibition: The mechanistic target of rapamycin (mTOR) is a protein kinase that forms two distinct complexes, mTORC1 and mTORC2. mTORC1, in particular, is a master regulator of cellular growth that integrates signals from nutrients, growth factors, and cellular energy status.28 High mTORC1 activity promotes anabolic processes like protein and lipid synthesis while inhibiting catabolic processes like autophagy. While beneficial for growth and development, chronic hyperactivation of mTORC1 is strongly implicated in accelerating the aging process.28 Methionine restriction directly inhibits mTORC1 signaling. The key mechanism is the reduction in intracellular S-adenosylmethionine (SAM). SAM has been identified as a critical allosteric activator of mTORC1.5 When dietary methionine is scarce, SAM levels fall, leading to reduced mTORC1 activity.5 This effect is analogous to that of the drug rapamycin, a direct mTORC1 inhibitor and a well-validated geroprotector that extends lifespan in numerous species.28
IGF-1 Reduction: A second, and equally critical, effect of MR is a significant and consistent reduction in circulating levels of Insulin-Like Growth Factor-1 (IGF-1).4 The GH/IGF-1 axis is a major endocrine pathway controlling growth and metabolism, and its downregulation is one of the most conserved mechanisms for lifespan extension across phylogeny.22 The reduction in IGF-1 signaling is not merely a correlation but appears to be a primary driver of MR's benefits. This is powerfully illustrated by a landmark study in which long-lived dwarf mice, which are genetically deficient in growth hormone (GH) signaling and thus already have very low IGF-1 levels, were placed on a methionine-restricted diet. The MR diet failed to provide any additional lifespan extension to these mice.22 This lack of an additive effect strongly suggests that MR exerts its longevity benefits primarily, and perhaps entirely, through the same pathway that is already suppressed in these animals: the reduction of IGF-1 signaling.
2. The Endocrine Response (FGF21)
Beyond the canonical nutrient-sensing pathways, MR engages a potent endocrine response mediated by the hormone Fibroblast Growth Factor 21 (FGF21). FGF21 is primarily produced by the liver in response to various metabolic stresses, including starvation and, most potently, the restriction of specific amino acids.4
FGF21 is an essential mediator for many of MR's most beneficial metabolic effects. Studies using mice genetically engineered to lack FGF21 (Fgf21-/-) have been instrumental in dissecting its role. When these knockout mice are placed on an MR diet, many of the hallmark benefits are lost. They fail to exhibit the characteristic increase in energy expenditure, the browning of white adipose tissue (activation of UCP-1), and the profound improvements in insulin sensitivity seen in normal mice on the same diet.4 This demonstrates that FGF21 is not just associated with MR but is causally required for these specific metabolic improvements.
However, the MR response is not solely dependent on FGF21. Even in Fgf21-/- mice, MR can still reduce body weight and adiposity (albeit through different mechanisms, such as reduced food intake) and can still exert anti-inflammatory effects in the liver and adipose tissue.23 This highlights the multifaceted nature of the MR phenotype, where FGF21 is a critical but not exclusive effector.
Furthermore, FGF21 signaling is crucial for the neuroprotective effects of MR. In mouse models of both age-related cognitive decline and diabetes-associated cognitive impairment, MR was found to improve memory and synaptic plasticity. These benefits were linked to MR-induced increases in circulating FGF21, which then acted on the brain to improve glucose metabolism, reduce neuroinflammation, and mitigate oxidative damage.33
3. Cellular Homeostasis (Autophagy and Oxidative Stress)
At the cellular level, MR promotes longevity by enhancing the machinery responsible for maintaining cellular quality control and resilience.
Autophagy Induction: By inhibiting mTORC1, MR robustly induces autophagy, the cell's intrinsic recycling program.5 Autophagy involves the engulfment of old, damaged, or unnecessary cellular components—such as misfolded proteins and dysfunctional organelles—within double-membraned vesicles called autophagosomes, which then fuse with lysosomes for degradation and recycling of the constituent parts. This process is essential for cellular health, and its decline with age is considered a hallmark of the aging process. Studies in yeast have shown that the lifespan-extending benefits of MR are completely dependent on the presence of core autophagy genes like
Atg5 and Atg7.15 Further investigation has revealed that MR may be particularly effective at inducing mitophagy, the specialized form of autophagy that selectively targets and removes damaged mitochondria.15 By clearing out dysfunctional mitochondria, mitophagy prevents the accumulation of ROS and maintains a healthy, functional mitochondrial pool.Oxidative Stress Reduction: MR combats age-related oxidative stress through a multi-pronged approach. First, by promoting mitophagy and improving the efficiency of the mitochondrial electron transport chain, it directly reduces the production of mitochondrial reactive oxygen species (ROS), particularly from Complex I and Complex III.5 Studies in rats have shown that even a moderate 40% MR can significantly decrease mitochondrial ROS generation and subsequent oxidative damage to mitochondrial DNA.19 Second, MR upregulates the body's endogenous antioxidant and cytoprotective systems. It has been shown to increase the production of hydrogen sulfide (H2S), a gaseous signaling molecule with potent antioxidant and anti-inflammatory properties, via the transsulfuration pathway.5 It also increases levels of spermidine, a polyamine known to induce autophagy and extend lifespan.5 The net result is a cellular environment with lower levels of damaging ROS and a greater capacity to handle oxidative insults, contributing to extended healthspan.
D. Clinical Manifestations of MR: Disease-Specific Benefits
While the bulk of the evidence for MR comes from preclinical models, a growing body of research, including limited human trials, indicates that these benefits are translatable and have significant implications for the prevention and management of major age-related diseases.
Metabolic Syndrome
Metabolic syndrome is a cluster of conditions—including central obesity, high blood pressure, high blood sugar, and abnormal cholesterol or triglyceride levels—that together increase the risk of heart disease, stroke, and type 2 diabetes. The metabolic reprogramming induced by MR directly targets the core components of this syndrome. A pivotal 16-week human clinical trial investigated the effects of an ~80% MR diet in obese subjects with metabolic syndrome.38 While both the MR group and a weight-loss-matched control group saw improvements in insulin sensitivity, the MR group experienced unique benefits. Specifically, dietary MR produced a significant 12.1% increase in the rate of fat oxidation, while the control group saw an 8.1% decrease. Furthermore, the MR diet led to a significant reduction in intrahepatic lipid content (fatty liver), a key driver of metabolic dysfunction.38 These benefits occurred independently of weight loss, demonstrating that MR confers metabolic advantages beyond simple caloric balance. These findings directly mirror the metabolic improvements observed consistently in rodent models.4
Cancer Biology
The link between methionine metabolism and cancer is one of the most compelling areas of MR research. Decades of in vitro work have established the phenomenon of "methionine dependency," where a wide variety of human cancer cell lines—including those from colon, breast, prostate, ovary, and melanoma—are unable to proliferate or survive in a culture medium lacking methionine, a condition under which normal, non-cancerous cells thrive.29 This metabolic vulnerability, termed the "Hoffman effect," arises because many tumors lose the ability to efficiently recycle homocysteine back into methionine and thus become absolutely dependent on an external supply.40
This dependency makes MR a highly attractive therapeutic strategy. In animal models, MR has been shown to inhibit tumor growth, induce apoptosis (programmed cell death) in cancer cells, and, critically, enhance the efficacy of conventional treatments like chemotherapy and radiation therapy.11 For example, combining an MR diet with radiation slowed tumor growth in a mouse model of aggressive sarcoma by 50% more than radiation alone.43 These promising preclinical results have led to a number of early-phase clinical trials investigating MR diets, often in combination with standard-of-care treatments, for various human malignancies.39
Neuroprotection and Cognitive Function
Emerging evidence suggests that the benefits of MR extend to the brain, offering a potential strategy to combat age-related cognitive decline and neurodegenerative diseases. In mouse models, MR has been shown to ameliorate cognitive impairments associated with both normal aging and type 2 diabetes.33 In aged mice, a 3-month MR intervention improved working, short-term, and spatial memory. This was accompanied by preserved synaptic structure, increased mitochondrial biogenesis, and reduced markers of oxidative damage (e.g., malondialdehyde, MDA) and neuroinflammation in the hippocampus.34 The mechanisms appear to be mediated, at least in part, by the induction of FGF21. MR increases FGF21 expression in the liver, serum, and brain, which in turn activates protective signaling pathways (e.g., FGFR1/AMPK/GLUT4) that enhance brain glucose metabolism and antioxidant defenses.33 These findings provide a strong rationale for investigating MR-based strategies to promote cognitive healthspan in humans.
III. Glycine Supplementation as a Viable Methionine Restriction Mimetic
While the health and longevity benefits of dietary methionine restriction are well-established in preclinical models, its practical application in humans is fraught with challenges. Sustaining a diet that restricts a specific essential amino acid by ~80% is difficult, unpalatable for many, and carries a risk of other nutritional deficiencies if not carefully managed.27 This has spurred the search for "MR mimetics"—interventions that can replicate the biochemical and physiological effects of MR without requiring such a stringent diet. Among the most promising candidates is high-dose supplementation with the non-essential amino acid glycine. A compelling body of evidence suggests that glycine can function as a potent and practical MR mimetic by targeting the central hub of the methionine cycle.
A. The Biochemical Bridge: Glycine N-Methyltransferase (GNMT)
The core hypothesis linking glycine supplementation to the effects of MR centers on a single, highly abundant liver enzyme: Glycine N-Methyltransferase (GNMT).
GNMT: The Methionine Sink
GNMT's primary function is to regulate the concentration of S-adenosylmethionine (SAM) in the liver, which is the main site of methionine metabolism.6 It accomplishes this by catalyzing the methylation of glycine to form N-methylglycine, more commonly known as sarcosine. This reaction is a major consumer of methyl groups, using one molecule of SAM as the methyl donor for every molecule of glycine it processes.51 Under normal conditions, GNMT acts as a buffer, preventing the accumulation of excess SAM when methionine intake is high.
The Mimicry Mechanism
The MR-mimetic strategy leverages this buffering capacity. By providing a large, supplemental dose of glycine, the availability of one of GNMT's key substrates is massively increased. According to the principles of enzyme kinetics, this surplus of substrate drives the GNMT-catalyzed reaction forward at a higher rate. This accelerated reaction creates a powerful metabolic "sink" that actively consumes and depletes the hepatic SAM pool.51 The resulting state of low SAM availability is the key biochemical event that mimics the effects of directly restricting dietary methionine intake. By lowering the SAM/SAH ratio, glycine supplementation is hypothesized to trigger the same downstream signaling cascades—mTORC1 inhibition, IGF-1 reduction, and FGF21 induction—that are responsible for the benefits of MR.
Causal Evidence from Genetic Models
The pivotal role of the GNMT-methionine-SAM axis is powerfully supported by genetic studies in mice.
GNMT Knockout Models: Mice that are genetically engineered to lack the GNMT enzyme (GNMT-/-) cannot effectively clear excess SAM. As a result, they accumulate extraordinarily high levels of SAM in the liver—up to 75-fold higher than normal—and spontaneously develop severe non-alcoholic fatty liver disease (NAFLD) and liver steatosis.8 This demonstrates that dysregulation of GNMT and the subsequent rise in SAM are sufficient to cause significant metabolic pathology. Critically, when these
GNMT-/- mice are placed on a methionine-restricted diet, their liver SAM levels normalize, and the liver pathology is reversed.8 This provides unequivocal genetic proof that GNMT is the central link between dietary methionine intake, SAM homeostasis, and hepatic health.GNMT Overexpression Models: Conversely, studies in Drosophila have shown that genetically overexpressing the GNMT enzyme is sufficient on its own to extend lifespan and reduce systemic methionine levels.51 This finding solidifies GNMT's status as a bona fide pro-longevity gene, whose activation is a desirable therapeutic target.
Together, these genetic models provide strong causal evidence for the central hypothesis: activating GNMT (via glycine supplementation) to lower SAM is a viable strategy for achieving the benefits associated with MR.
B. Evidence for Glycine as a Geroprotector
Beyond the mechanistic plausibility of the GNMT hypothesis, direct evidence from animal and human studies supports the role of glycine as a geroprotective agent, capable of extending lifespan and improving healthspan.
Lifespan Extension in Animal Models
Glycine supplementation has been shown to extend lifespan in multiple model organisms, validating its potential as an anti-aging intervention.
Mice: A large, rigorous study conducted by the prestigious Interventions Testing Program (ITP)—a multi-institutional consortium funded by the National Institute on Aging to test potential longevity drugs—evaluated the effect of an 8% glycine diet in genetically heterogeneous mice. The results showed a statistically significant, albeit modest, increase in lifespan of 4-6% in both males (p=0.002) and females (p<0.001).53 While the effect size is smaller than the dramatic extensions seen with severe MR in inbred rodent strains, it is a highly significant finding in a genetically diverse population, which is more relevant to humans.
Rats: Earlier, smaller studies in Fischer 344 rats reported more dramatic results, with glycine supplementation leading to lifespan extensions of approximately 20-40%.60
Other Organisms: Lifespan extension from glycine has also been observed in simpler organisms like C. elegans.61
Healthspan Benefits in Humans
While long-term human lifespan trials are not feasible, numerous short-term clinical trials have demonstrated that glycine supplementation confers tangible healthspan benefits that align with the known effects of MR. A 2023 systematic review analyzed 50 human studies of glycine administration and found consistent positive effects across multiple physiological systems.61 Key findings include:
Nervous System and Sleep: Oral glycine supplementation, typically at a dose of 3 grams taken before bedtime, has been repeatedly shown to improve subjective sleep quality, reduce the time it takes to fall asleep, lessen daytime sleepiness, and improve cognitive performance and alertness upon waking.61
Metabolic Health: In patients with metabolic syndrome, a daily dose of 15 grams of glycine (divided into three 5g doses) for three months was shown to reduce oxidative stress and lower HbA1c, a marker of long-term blood sugar control.62 In healthy individuals, glycine enhances insulin secretion in response to a glucose challenge, suggesting it improves metabolic flexibility.66
Inflammation: Glycine exhibits systemic anti-inflammatory properties. In patients with Type 2 Diabetes, 15g of glycine per day decreased levels of pro-inflammatory cytokines such as TNF-α and IL-6.61
Other Synergistic Mechanisms
In addition to its primary role as an MR mimetic via GNMT, glycine possesses other intrinsic properties that contribute to its geroprotective profile.
Glutathione Precursor: Glycine is one of the three amino acids (along with cysteine and glutamate) required for the synthesis of glutathione (GSH), the body's master intracellular antioxidant.51 Glutathione levels are known to decline with age, leading to increased oxidative stress. Supplementing with glycine (often alongside N-acetylcysteine, or NAC, a cysteine precursor) can help replenish GSH stores, thereby bolstering the body's antioxidant defenses.65
Anti-inflammatory and Cytoprotective Effects: Glycine has direct cytoprotective (cell-protecting) and anti-inflammatory effects, independent of its roles in the methionine cycle or GSH synthesis. It can activate glycine receptors on immune cells like macrophages, which dampens their inflammatory response.64
C. Projecting Human Lifespan and Healthspan Gains: A Data-Driven Extrapolation
Projecting the precise impact of a nutritional intervention like glycine supplementation on human lifespan is inherently speculative and complex. No validated life expectancy calculator exists for such a strategy.72 However, by synthesizing the evidence from preclinical longevity studies, human clinical trials on intermediate biomarkers, and large-scale epidemiological data, it is possible to construct a robust, data-driven model of the likely effects on healthspan and disease risk. The overall picture that emerges is one of a multi-pronged strategy that mitigates risk across several of the most common age-related diseases, thereby plausibly extending the period of healthy life.
Modeling Approach: From Biomarkers to Risk Reduction
The projection model employed here does not attempt to calculate a specific number of "extra years." Instead, it translates the observed effect sizes of glycine and MR on key physiological and biochemical markers into quantifiable reductions in the risk of developing major age-related diseases. This approach is grounded in the established links between these biomarkers and long-term health outcomes.
Metabolic Disease Risk Reduction
The evidence is strong that an MR-mimetic strategy directly combats the drivers of metabolic syndrome and type 2 diabetes.
Improved Insulin Sensitivity and Hepatic Fat: The 16-week human MR trial demonstrated a significant reduction in intrahepatic lipid content and a 12.1% increase in fat oxidation, effects that were independent of weight loss.38 Glycine supplementation in humans has been shown to improve insulin secretion and lower HbA1c.64 Given that insulin resistance and non-alcoholic fatty liver disease are primary drivers of type 2 diabetes, improvements in these markers translate to a substantial reduction in disease risk.
FGF21 Induction: The induction of FGF21 by MR is a key mechanism for these metabolic benefits.23 FGF21 analogs are currently in clinical development as treatments for metabolic dysfunction, highlighting the therapeutic relevance of this pathway.75 A glycine-based strategy that increases endogenous FGF21 would thus be expected to confer similar protective effects.
Cardiovascular Disease (CVD) Risk Reduction
The intervention is projected to lower CVD risk through several mechanisms.
Reduced Inflammation: Chronic, low-grade inflammation is a key driver of atherosclerosis. Both MR and glycine have demonstrated potent anti-inflammatory effects in preclinical and clinical settings, including the reduction of circulating pro-inflammatory cytokines like IL-6 and TNF-α.19
Improved Metabolic Parameters: By reducing adiposity, improving insulin sensitivity, and lowering triglycerides, the strategy addresses several of the core risk factors for CVD that are part of the metabolic syndrome. Some studies also suggest glycine may help lower blood pressure.62
Homocysteine Regulation: While high homocysteine is a risk marker for CVD, large-scale trials have shown that lowering it with B-vitamins alone has an inconsistent and often null effect on cardiovascular events.76 The focus of this report's strategy is not solely on lowering homocysteine, but on maintaining the health of the entire one-carbon metabolism cycle. The included B-vitamin cofactors are intended to prevent pathway bottlenecks, which is a more holistic approach than targeting a single biomarker.
Cancer Risk Reduction
The potential for cancer risk reduction is one of the most compelling aspects of an MR-mimetic strategy.
Targeting Methionine Dependency: The well-documented phenomenon of methionine dependency in many common cancers (colon, breast, prostate) suggests that systemically reducing methionine availability could create a less permissive environment for tumor initiation and growth.40
Lowering IGF-1: A core effect of MR is the reduction of circulating IGF-1.22 High levels of IGF-1 are a known risk factor for several cancers, particularly prostate, premenopausal breast, and colorectal cancer, as IGF-1 is a potent promoter of cell growth and proliferation.28 By lowering IGF-1, a glycine-based strategy directly targets a major hormonal driver of carcinogenesis.
Neurocognitive Healthspan
The strategy is projected to support brain health and preserve cognitive function into old age.
Preclinical Evidence: Animal models clearly show that MR can protect against age-related and diabetes-related cognitive decline, with benefits mediated by FGF21 signaling, reduced neuroinflammation, and lower oxidative stress in the brain.33
Human Evidence: Clinical trials in humans have shown that glycine supplementation directly improves sleep quality, which is fundamental for memory consolidation and brain health, and can improve daytime alertness and cognitive performance.61
Overall Longevity Projection (Caveated)
Synthesizing these points, a glycine-based MR mimetic strategy is projected to significantly extend healthspan by simultaneously reducing the risk of the primary diseases of aging: metabolic disease, cardiovascular disease, cancer, and neurodegeneration. While a precise quantitative prediction for human lifespan extension is not possible, the available evidence allows for a reasoned, qualitative projection. The modest but statistically significant 4-6% lifespan extension observed in the ITP mouse study of glycine supplementation provides the most direct and relevant animal evidence for a positive longevity effect.58 It is plausible that a similar modest extension of maximum lifespan, coupled with a more significant compression of morbidity (i.e., a longer period of healthy life), could be achievable in humans who adhere to a well-designed protocol.
IV. Modulating the Methionine-Glycine Axis: Cofactors and Antagonists
The implementation of a glycine-based methionine restriction mimetic strategy is not as simple as taking a single supplement. The methionine cycle is a complex and highly regulated metabolic network. Intentionally increasing the flux through one part of this network—the consumption of SAM by GNMT—has cascading effects on the entire system. A successful protocol must therefore be designed to support the increased metabolic demand and, critically, to avoid interventions that directly counteract the intended biochemical goal. This requires the inclusion of essential cofactors and the strict avoidance of a key metabolic antagonist, creatine.
A. Essential Cofactors for Metabolic Efficacy and Safety
Driving the GNMT pathway with high-dose glycine accelerates the entire methionine cycle. Each molecule of SAM consumed produces one molecule of SAH, which is then converted to homocysteine. This increased production of homocysteine places a higher demand on the two pathways responsible for its clearance: remethylation and transsulfuration. Without adequate support, these pathways can become overwhelmed, leading to a bottleneck and the potential accumulation of homocysteine. Therefore, ensuring an adequate supply of the B-vitamin cofactors required by these pathways is not an optional add-on but a fundamental component of a safe and effective protocol.1
The B-Vitamin Trio (B6, B9, B12): These three vitamins are the indispensable cofactors for homocysteine metabolism.1
Folate (Vitamin B9) and Vitamin B12: These are absolutely required for the primary remethylation pathway catalyzed by methionine synthase (MS), which recycles homocysteine back to methionine. B12 acts as a direct cofactor for MS, while folate provides the methyl group in its active form, 5-MTHF.1
Vitamin B6: This vitamin, in its active form pyridoxal-5'-phosphate (P5P), is the essential cofactor for cystathionine β-synthase (CBS), the rate-limiting enzyme of the transsulfuration pathway that irreversibly clears homocysteine by converting it to cysteine.12 Vitamin B6 is also a cofactor for serine hydroxymethyltransferase (SHMT), which interconverts serine and glycine, linking the folate and glycine pathways.1
Betaine (Trimethylglycine - TMG): Betaine offers an additional, parallel pathway for homocysteine clearance, particularly within the liver. The enzyme betaine-homocysteine S-methyltransferase (BHMT) uses a methyl group from betaine to convert homocysteine to methionine.1 This pathway is independent of folate and B12, providing a valuable secondary route for managing homocysteine load. Furthermore, betaine supplementation has been shown in its own right to increase circulating levels of the beneficial hormone FGF21, suggesting it may have synergistic effects with the MR-mimetic strategy.83
B. The Creatine Conundrum: A Direct Metabolic Conflict
While the need for B-vitamin co-supplementation is a logical consequence of increasing metabolic flux, the interaction with creatine represents a more profound and direct biochemical conflict. Understanding this conflict is arguably the most critical insight for the successful implementation of a glycine-based MR mimetic strategy.
Endogenous Creatine Synthesis: A Major Consumer of SAM
Creatine is a critical molecule for energy buffering in high-energy tissues like muscle and brain. The body synthesizes creatine endogenously in a two-step process that consumes three amino acids: arginine, glycine, and methionine.84 The final, irreversible step of this synthesis is the methylation of a precursor molecule, guanidinoacetate (GAA), to form creatine. This reaction is catalyzed by the enzyme guanidinoacetate N-methyltransferase (GAMT) and, crucially, it consumes one molecule of SAM as the methyl donor.85 The demand for this pathway is enormous; endogenous creatine synthesis is estimated to be the single largest consumer of SAM in the body, accounting for approximately 40% of all labile methyl groups derived from the methionine cycle.86
The Effect of Creatine Supplementation
When an individual supplements with exogenous creatine (e.g., creatine monohydrate), the body's need for endogenous synthesis is eliminated. This leads to a powerful feedback inhibition mechanism that suppresses the activity of the enzymes in the synthesis pathway, particularly AGAT and GAMT.87 In essence, taking creatine supplements tells the body to stop making its own.
The Metabolic Contradiction and a Critical Insight
The direct metabolic opposition between the glycine strategy and creatine supplementation becomes immediately apparent when their effects on the SAM pool are considered. This is summarized in Table 2.
Table 2: The Metabolic Tug-of-War: Glycine vs. Creatine Supplementation on the SAM Pool
Metabolic Intervention
Target Enzyme
Effect on Enzyme Activity
Effect on SAM Pool
Implication for MR Mimicry
High-Dose Glycine Supplementation
Glycine N-Methyltransferase (GNMT)
Upregulated (substrate-driven)
Depleted (consumed)
Promotes MR-mimetic state
Creatine Supplementation
Guanidinoacetate Methyltransferase (GAMT)
Downregulated (feedback inhibition)
Spared (consumption blocked)
Antagonizes MR-mimetic state
Combined Glycine + Creatine
GNMT & GAMT
Competing effects
Net effect is likely SAM-sparing or neutral, negating the glycine effect
Contradictory and Ineffective
The goal of the glycine strategy is to deplete the SAM pool by driving the GNMT reaction. The effect of creatine supplementation is to spare the SAM pool by shutting down the GAMT reaction, which is its largest consumer. These two interventions are pulling the SAM/SAH ratio in opposite directions. Attempting to do both simultaneously is biochemically futile; the SAM-sparing effect of creatine supplementation would likely overwhelm or completely negate the SAM-depleting effect of glycine.
This conclusion is powerfully supported by a crucial study published in PLOS Genetics. Researchers found that the unique transcriptional program activated by methionine deprivation in cells was completely dependent on an intact, functioning creatine biosynthesis pathway.88 When they blocked creatine synthesis (by co-depriving the cells of methionine and glycine/arginine), the cell was no longer able to effectively deplete its SAM pool, and the beneficial gene expression changes characteristic of the MR response were abolished. This provides a "smoking gun," demonstrating that the cell's natural response to low methionine relies on the SAM-consuming activity of the creatine synthesis pathway to achieve the proper signaling state. Therefore, creatine supplementation does not just fail to help; it actively sabotages the very mechanism that the glycine-mimetic strategy seeks to exploit.
The clear and unavoidable conclusion is that the goals of maximizing muscle creatine stores via supplementation and achieving an MR-mimetic state via glycine-induced SAM depletion are mutually exclusive. An individual must choose one or the other.
C. Practical Implementation: Dosages, Protocols, and Safety
Based on the available evidence, a practical protocol for implementing a glycine-based MR mimetic strategy can be formulated. This protocol must include appropriate dosages for glycine and its essential cofactors and acknowledge safety considerations.
Glycine Dosage
Human clinical trials have used a wide range of glycine doses, with the effective dose depending on the desired outcome.
For Sleep Improvement: Doses as low as 3 grams taken 30-60 minutes before bedtime have been shown to be effective.61
For Metabolic Benefits: Studies demonstrating improvements in metabolic markers in patients with metabolic syndrome or type 2 diabetes have typically used higher doses, on the order of 15 grams per day, usually divided into three 5-gram doses with meals.64
For MR Mimicry: To effectively drive the GNMT "sink" and deplete SAM, higher doses are likely necessary. Clinical trials in schizophrenia have safely used doses up to 0.8 g/kg of body weight per day.89 A reasonable starting point for an individual seeking MR-mimetic effects would be 10 grams per day, divided into two doses. This could be gradually titrated upwards towards 20-30 grams per day (or approximately 0.2-0.4 g/kg), depending on individual tolerance.
Cofactor Dosages
The dosages for cofactors should be sufficient to support the increased metabolic demand. Recommendations can be guided by the doses used in successful homocysteine-lowering trials.
B-Vitamins: A network meta-analysis of 16 randomized trials concluded that combinations are more effective than single vitamins.90 A highly effective combination identified was 1 mg of folic acid, 7.2 mg of vitamin B6, and 20 µg of vitamin B12.91 Other research suggests optimal daily doses for homocysteine management are in the range of
Folic Acid (as methylfolate): 800 mcg, Vitamin B12 (as methylcobalamin): 500-1000 mcg, and Vitamin B6 (as P5P): 20-50 mg.92 The use of activated (methylated) forms of B9 and B12 is preferable as it bypasses potential enzymatic impairments in individuals with common MTHFR genetic polymorphisms.Betaine (TMG): Doses of 1-6 grams per day have been used in clinical studies.94 A supplemental dose of
1-2 grams per day would be a rational addition to provide robust, folate-independent support for homocysteine remethylation.
Safety and Contraindications
Glycine is generally recognized as safe (GRAS) by the FDA and is well-tolerated even at high doses.63 The primary safety consideration for this protocol is not the glycine itself, but the potential for pathway dysregulation if cofactors are inadequate.
Homocysteine Monitoring: While ensuring adequate B-vitamin intake should prevent any significant rise in homocysteine, individuals embarking on this strategy, particularly those with known MTHFR variants, may consider baseline and follow-up testing of plasma homocysteine as a precautionary measure.
Creatine Avoidance: As detailed extensively, the primary contraindication is the concurrent use of creatine supplements.
Dietary Context: While the goal is to mimic MR via supplementation, adopting a diet that is naturally lower in methionine—such as a plant-predominant or vegan diet—would be synergistic with the glycine strategy, as it reduces the overall methionine load that needs to be cleared.42
V. Synthesis, Recommendations, and Future Outlook
The vast and growing body of scientific literature reviewed in this report converges on a clear and compelling narrative. Methionine restriction is a foundational and potent intervention that extends healthspan and lifespan in diverse organisms by reprogramming core metabolic and signaling pathways. It achieves this primarily by reducing the intracellular concentration of S-adenosylmethionine (SAM), which in turn inhibits the pro-aging mTORC1 and IGF-1 pathways, induces the beneficial hormone FGF21, and enhances cellular resilience through autophagy and reduced oxidative stress.
The central thesis of this report is that these profound benefits can be substantially and practically mimicked through high-dose glycine supplementation. The mechanism is elegant and direct: glycine acts as a substrate for the hepatic enzyme GNMT, creating a metabolic sink that consumes SAM and replicates the key biochemical signature of MR. This strategy moves the concept from the realm of extreme dietary discipline to that of a targeted, evidence-based nutritional protocol.
However, the success of this protocol is not guaranteed by glycine alone. It hinges on a nuanced understanding of the interconnected metabolic network. The increased flux through the methionine cycle necessitates robust support from B-vitamin cofactors (B6, B9, B12) and potentially betaine to ensure safe and efficient processing of metabolites like homocysteine. Most critically, the strategy is biochemically incompatible with creatine supplementation. The SAM-sparing effect of exogenous creatine directly antagonizes the SAM-depleting goal of the glycine protocol, rendering a combined approach ineffective and contradictory.
Actionable Protocol Summary
Based on a comprehensive synthesis of the available evidence, the following protocol represents a rational and actionable strategy for individuals seeking to implement a glycine-based methionine restriction mimetic program:
Primary Intervention:
Glycine: Initiate supplementation at 10 grams per day, divided into two 5-gram doses. Based on tolerance and goals, this may be titrated up to 20-30 grams per day (approximately 0.2-0.4 g/kg body weight). Taking a dose before sleep may confer additional benefits for sleep quality.
Essential Cofactors:
B-Vitamin Complex: Daily supplementation with a high-quality B-complex providing doses in the range of:
Folate (as L-5-Methyltetrahydrofolate): 800 - 1000 mcg
Vitamin B12 (as Methylcobalamin): 500 - 1000 mcg
Vitamin B6 (as Pyridoxal-5'-Phosphate): 20 - 50 mg
Betaine (Trimethylglycine - TMG): 1 - 2 grams per day to provide additional methyl donor support.
Critical Contraindication:
Creatine: Strict avoidance of all forms of creatine supplementation (e.g., creatine monohydrate) is essential for the protocol to be effective.
Knowledge Gaps and Future Research
Despite the strong mechanistic rationale and promising evidence, several key areas require further investigation to refine and validate this strategy for widespread human use.
Long-Term Human Trials: The most pressing need is for long-term, randomized controlled trials in healthy human populations. Such studies are necessary to confirm the projected healthspan and lifespan benefits, to establish optimal, evidence-based dosing for glycine and its cofactors, and to monitor for any unforeseen long-term effects.
Direct Testing of the Glycine-Creatine Interaction: A targeted clinical trial should be conducted to directly test the metabolic conflict between glycine and creatine. Such a study would measure SAM/SAH ratios and key downstream biomarkers (e.g., IGF-1, FGF21, mTORC1 activity) in groups receiving glycine alone, creatine alone, and the combination, to definitively quantify the antagonistic interaction in humans.
The Role of Sarcosine: The product of the GNMT reaction, sarcosine, is not merely a waste product. It has been shown to induce autophagy in its own right.51 Research is needed to determine if sarcosine itself acts as a beneficial signaling molecule, which would add another layer to the mechanism of the glycine strategy.
Genetic Variability: Individuals have significant genetic variability in key enzymes of one-carbon metabolism, such as GNMT and MTHFR. Future research should investigate how these genetic differences influence an individual's response to a glycine-based protocol, which could pave the way for personalized recommendations.
Exploration of Other MR Mimetics: The success of the MR paradigm should continue to drive the search for other mimetics. This includes further investigation into pharmacological agents like methioninase, a bacterial enzyme that degrades methionine and has shown promise in preclinical and early clinical studies.16
In conclusion, the strategy of mimicking methionine restriction with glycine supplementation represents a frontier in translational aging research. It is a prime example of how a deep understanding of fundamental biochemistry can be leveraged to develop targeted, rational interventions aimed at promoting human healthspan and longevity. While further research is needed, the existing evidence provides a strong foundation for a protocol that is safe, accessible, and mechanistically poised to deliver a significant portion of the benefits of one of nature's most powerful anti-aging interventions.
Works cited
B Vitamins and One-Carbon Metabolism: Implications in Human Health and Disease - PMC, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7551072/
B Vitamins and One-Carbon Metabolism: Implications in Human Health and Disease - MDPI, accessed on June 27, 2025, https://www.mdpi.com/2072-6643/12/9/2867
Vitamin B12 , folate, and the methionine remethylation cycle-biochemistry, pathways, and regulation - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/30693532/
Physiological and Molecular Mechanisms of Methionine Restriction - PMC - PubMed Central, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5945823/
Effect of Methionine Restriction on Aging: Its Relationship to ... - MDPI, accessed on June 27, 2025, https://www.mdpi.com/2227-9059/9/2/130
Glycine N-Methyltransferase and Regulation of S-Adenosylmethionine Levels - PMC, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2755656/
S-adenosylmethionine metabolism buffering is regulated by a decrease in glycine N-methyltransferase via the nuclear ubiquitin–proteasome system | PNAS, accessed on June 27, 2025, https://www.pnas.org/doi/10.1073/pnas.2417821122
Disrupted liver oxidative metabolism in glycine N-methyltransferase ..., accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8866067/
Dietary methionine restriction on cancer development and antitumor immunity - PMC, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11096033/
Effect of Methionine Restriction on Aging: Its Relationship to Oxidative Stress - PMC, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7911310/
Methionine restriction for cancer therapy: From preclinical studies to clinical trials, accessed on June 27, 2025, https://mednexus.org/doi/10.1016/j.cpt.2025.01.002
Effects of B Vitamins on Homocysteine Lowering and Thrombotic Risk Reduction—A Review of Randomized Controlled Trials Published Since January 1996 - MDPI, accessed on June 27, 2025, https://www.mdpi.com/2072-6643/17/7/1122
Methionine + cystine levels and vitamin B6 supplementation on performance and enzyme expression of methionine metabolism of gilts from 75 to 100 kg - SciELO, accessed on June 27, 2025, https://www.scielo.br/j/rbz/a/LJXdZ4TNx5ZjpYkYpf6vvHD/
Methylation and Homocysteine: A Brief Overview - Food for the Brain, accessed on June 27, 2025, https://foodforthebrain.org/nutrition-for-healthcare-professionals/methylation-and-homocysteine/
Extension of Cellular Lifespan by Methionine Restriction Involves Alterations in Central Carbon Metabolism and Is Mitophagy-Dependent - Frontiers, accessed on June 27, 2025, https://www.frontiersin.org/journals/cell-and-developmental-biology/articles/10.3389/fcell.2019.00301/full
A genetic model of methionine restriction extends Drosophila health- and lifespan - PNAS, accessed on June 27, 2025, https://www.pnas.org/doi/10.1073/pnas.2110387118
Methionine restriction activates the retrograde response and confers both stress tolerance and lifespan extension to yeast, mouse and human cells - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/24830393/
(PDF) Low methionine ingestion extends lifespan. - ResearchGate, accessed on June 27, 2025, https://www.researchgate.net/publication/256546320_Low_methionine_ingestion_extends_lifespan
Methionine Restriction and Cancer Biology - MDPI, accessed on June 27, 2025, https://www.mdpi.com/2072-6643/12/3/684
Methionine restriction extends lifespan of Drosophila melanogaster under conditions of low amino-acid status - Portal de Periódicos da CAPES, accessed on June 27, 2025, https://www.periodicos.capes.gov.br/index.php/acervo/buscador.html?task=detalhes&id=W2139669573
Early-adult methionine restriction reduces methionine sulfoxide and extends lifespan in Drosophila - PMC, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10698029/
Selenium supplementation inhibits IGF-1 signaling and confers methionine restriction-like healthspan benefits to mice | eLife, accessed on June 27, 2025, https://elifesciences.org/articles/62483
FGF21 Mediates the Thermogenic and Insulin-Sensitizing Effects of Dietary Methionine Restriction but Not Its Effects on Hepatic Lipid Metabolism | Request PDF - ResearchGate, accessed on June 27, 2025, https://www.researchgate.net/publication/312508875_FGF21_Mediates_the_Thermogenic_and_Insulin-Sensitizing_Effects_of_Dietary_Methionine_Restriction_but_Not_Its_Effects_on_Hepatic_Lipid_Metabolism
The effects of dietary methionine restriction on the function and metabolic reprogramming in the liver and brain - implications for longevity - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/30817309/
Effect of Methionine Restriction on Aging: Its Relationship to Oxidative Stress - MDPI, accessed on June 27, 2025, https://www.mdpi.com/2227-9059/9/2/130/review_report
Dietary methionine restriction started late in life promotes healthy aging in a sex-specific manner - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/40238871/
Longevity Briefs: Could Limiting Dietary Methionine Improve Health In Old Age?, accessed on June 27, 2025, https://www.gowinglife.com/longevity-briefs-could-limiting-dietary-methionine-improve-health-in-old-age/
An overview of mTOR and IGF-1 | Peter Attia - FoundMyFitness, accessed on June 27, 2025, https://www.foundmyfitness.com/episodes/overview-mtor-igf-1
Methionine Dependency and Restriction in Cancer: Exploring the Pathogenic Function and Therapeutic Potential - MDPI, accessed on June 27, 2025, https://www.mdpi.com/1424-8247/18/5/640
New Developments in Human Healthspan and Longevity - NYAS, accessed on June 27, 2025, https://www.nyas.org/shaping-science/events/ebriefing/new-developments-in-human-healthspan-and-longevity/
(PDF) Methionine Restriction beyond Lifespan Extension - ResearchGate, accessed on June 27, 2025, https://www.researchgate.net/publication/291153217_Methionine_Restriction_beyond_Lifespan_Extension
Intermittent methionine restriction reduces IGF-1 levels and produces similar healthspan benefits to continuous methionine restriction - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/35570387/
Methionine restriction alleviates diabetes-associated cognitive impairment via activation of FGF21 - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/39383602/
Methionine restriction alleviates age-associated cognitive decline via fibroblast growth factor 21 - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/33765615/
Dietary Methionine Restriction Reduces Inflammation Independent of FGF21 Action: Anti-Inflammatory Effects of MR and FGF21 - ResearchGate, accessed on June 27, 2025, https://www.researchgate.net/publication/333830222_Dietary_Methionine_Restriction_Reduces_Inflammation_Independent_of_FGF21_Action_Anti-Inflammatory_Effects_of_MR_and_FGF21
Effects of Dietary Methionine Restriction on Cognition in Mice - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/38068808/
Effect of Methionine Restriction on Aging: Its Relationship to Oxidative Stress - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/33572965/
Dietary Methionine Restriction Increases Fat Oxidation in Obese ..., accessed on June 27, 2025, https://academic.oup.com/jcem/article/96/5/E836/2834121
Methionine restriction for cancer therapy: From preclinical studies to clinical trials, accessed on June 27, 2025, https://mednexus.org/doi/abs/10.1016/j.cpt.2025.01.002
Methionine Restriction: Ready for Prime Time in the Cancer Clinic? - Anticancer Research, accessed on June 27, 2025, https://ar.iiarjournals.org/content/42/2/641
The Role of Methionine in Cancer Growth and Control | Natural Medicine Journal, accessed on June 27, 2025, https://www.naturalmedicinejournal.com/journal/role-methionine-cancer-growth-and-control
Starving Cancer with Methionine Restriction | NutritionFacts.org, accessed on June 27, 2025, https://nutritionfacts.org/video/starving-cancer-with-methionine-restriction/
Altering Diet to Enhance Cancer Treatment Response - NCI, accessed on June 27, 2025, https://www.cancer.gov/news-events/cancer-currents-blog/2019/targeting-cancer-metabolism-low-methionine-diet
Methionine Restriction and Cancer Biology - PMC - PubMed Central, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7146589/
Methionine-Restricted Diet to Potentiate The Effects of Radiation Therapy | ClinicalTrials.gov, accessed on June 27, 2025, https://www.clinicaltrials.gov/study/NCT03574194
Effects of age and dietary methionine restriction on cognitive and behavioural phenotypes in the rTg4510 model of frontotemporal dementia | bioRxiv, accessed on June 27, 2025, https://www.biorxiv.org/content/10.1101/2024.03.05.583295v1.full-text
Methionine restriction and lifespan control - PMC, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5008916/
Amino acid restriction, aging, and longevity: an update - Frontiers, accessed on June 27, 2025, https://www.frontiersin.org/journals/aging/articles/10.3389/fragi.2024.1393216/full
View of Alterations of methionine metabolism in hepatocarcinogenesis: the emergent role of glycine N-methyltransferase in liver injury | Annals of Gastroenterology, accessed on June 27, 2025, http://annalsgastro.gr/index.php/annalsgastro/article/view/4092/4443
Mutations in human glycine N-methyltransferase give insights into its role in methionine metabolism - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/11810299/
Glycine and aging: Evidence and mechanisms | Request PDF - ResearchGate, accessed on June 27, 2025, https://www.researchgate.net/publication/369721883_Glycine_and_aging_Evidence_and_mechanisms
Glycine and aging: Evidence and mechanisms - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/37004845/
Glycine supplementation extends lifespan of male and female mice - University of Michigan Library, accessed on June 27, 2025, https://deepblue.lib.umich.edu/bitstream/handle/2027.42/149346/acel12953_am.pdf
Glycine Supplementation as a Methionine Restriction Mimetic - Fight Aging!, accessed on June 27, 2025, https://www.fightaging.org/archives/2023/04/glycine-supplementation-as-a-methionine-restriction-mimetic/
Methionine-glycine interaction, a review of animal and human studies. - Reddit, accessed on June 27, 2025, https://www.reddit.com/r/ketoscience/comments/264upi/methionineglycine_interaction_a_review_of_animal/
Disrupted liver oxidative metabolism in glycine N-methyltransferase-deficient mice is mitigated by dietary methionine restriction - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/35121169/
Glycine supplementation extends lifespan of male and female mice ..., accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6516426/
Glycine supplementation extends lifespan of male and female mice - ResearchGate, accessed on June 27, 2025, https://www.researchgate.net/publication/332053384_Glycine_supplementation_extends_lifespan_of_male_and_female_mice
Glycine supplementation extends lifespan of male and female mice - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/30916479/
The Anti-Aging and Longevity Benefits of Glycine - NOVOS, accessed on June 27, 2025, https://novoslabs.com/novos-anti-aging-longevity-supplement/glycine-anti-aging-supplement-longevity/
The effect of glycine administration on the characteristics of physiological systems in human adults: A systematic review - PubMed Central, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10828290/
How to Get Less Cysteine and More Glycine - NutritionFacts.org, accessed on June 27, 2025, https://nutritionfacts.org/hnta-video/how-to-get-less-cysteine-and-more-glycine/
What is Glycine? - News-Medical.net, accessed on June 27, 2025, https://www.news-medical.net/health/What-is-Glycine.aspx
The Power of Balance and the Balance of Power - Earth Fed Muscle, accessed on June 27, 2025, https://www.earthfedmuscle.com/blogs/articles/the-power-of-balance-and-the-balance-of-power
The potential impact of glycine supplementation on the process of aging | Quality in Sport, accessed on June 27, 2025, https://apcz.umk.pl/QS/article/view/57686
Effects of an oral mixture containing glycine, glutamine and niacin on memory, GH and IGF-I secretion in middle-aged and elderly subjects - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/14609312/
Glycine: Health Benefits, Safety Information, Dosage, and More - WebMD, accessed on June 27, 2025, https://www.webmd.com/diet/health-benefits-glycine
Study Details | Glycine Supplement for Severe COVID-19 | ClinicalTrials.gov, accessed on June 27, 2025, https://www.clinicaltrials.gov/study/NCT04443673?term=AREA%5BBasicSearch%5D(GLYCINE)&rank=6
Glycine Metabolism and Its Alterations in Obesity and Metabolic Diseases - PMC, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6627940/
A Randomized Controlled Clinical Trial in Healthy Older ... - Frontiers, accessed on June 27, 2025, https://www.frontiersin.org/journals/aging/articles/10.3389/fragi.2022.852569/full
15 Powerful Insights from the Latest Research on Glycine - Avid Organics, accessed on June 27, 2025, https://www.avidorganics.net/15-powerful-insights-from-the-latest-research-on-glycine/
Mortality calculator - Dean Foster, accessed on June 27, 2025, http://deanfoster.net:3838/life_calculator/
Living To 100 Life Expectancy Calculator, accessed on June 27, 2025, https://www.livingto100.com/calculator
Lifespan Calculator – Test Your Life Expectancy || NM - Northwestern Mutual, accessed on June 27, 2025, https://media.nmfn.com/tnetwork/lifespan/index.html
The potential function and clinical application of FGF21 in metabolic diseases - Frontiers, accessed on June 27, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2022.1089214/full
Methionine vs. Glycine — Is Too Much Muscle Meat Bad? - Healthline, accessed on June 27, 2025, https://www.healthline.com/nutrition/methionine-vs-glycine
Effects of B Vitamins on Homocysteine Lowering and Thrombotic Risk Reduction-A Review of Randomized Controlled Trials Published Since January 1996 - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/40218880/
Natural treatments for hyperhomocysteinemia | EBSCO Research Starters, accessed on June 27, 2025, https://www.ebsco.com/research-starters/complementary-and-alternative-medicine/natural-treatments-hyperhomocysteinemia
Homocysteine, B-vitamin supplementation, and stroke prevention: from observational to interventional trials - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/15261610/
Insulin-like Growth Factor (IGF-1) - Alzheimer's Drug Discovery Foundation, accessed on June 27, 2025, https://www.alzdiscovery.org/uploads/cognitive_vitality_media/IGF1-Cognitive-Vitality-For-Researchers.pdf
8.14: Vitamins Important for Metabolism - Medicine LibreTexts, accessed on June 27, 2025, https://med.libretexts.org/Courses/American_Public_University/APUS%3A_Basic_Foundation_of_Nutrition_for_Sports_Performance_(Byerley)/08%3A_Vitamin_and_Mineral_Basics_-_Metabolic_Regulators/8.14%3A_Vitamins_Important_for_Metabolism
High Homocysteine | Linus Pauling Institute | Oregon State University, accessed on June 27, 2025, https://lpi.oregonstate.edu/mic/health-disease/high-homocysteine
Dietary Betaine Supplementation Increases Fgf21 Levels to Improve Glucose Homeostasis and Reduce Hepatic Lipid Accumulation in Mice - American Diabetes Association, accessed on June 27, 2025, https://diabetesjournals.org/diabetes/article/65/4/902/35096/Dietary-Betaine-Supplementation-Increases-Fgf21
creatine, accessed on June 27, 2025, https://www1.udel.edu/chem/C465/senior/fall00/Performance2/creatine
Creatine - Wikipedia, accessed on June 27, 2025, https://en.wikipedia.org/wiki/Creatine
The metabolic burden of creatine synthesis - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/21387089/
Study Details | Creatine Kinetics, Synthesis From Glycine and Response to Creatine and Amino Acid Supplement in Healthy Humans | ClinicalTrials.gov, accessed on June 27, 2025, https://clinicaltrials.gov/study/NCT01725503?term=AREA%5BConditionSearch%5D(Creatine%20Supplementation)%20AND%20AREA%5BOverallStatus%5D(COMPLETED)&rank=5
Comprehensive Profiling of Amino Acid Response Uncovers Unique Methionine-Deprived Response Dependent on Intact Creatine Biosynthesis | PLOS Genetics, accessed on June 27, 2025, https://journals.plos.org/plosgenetics/article?id=10.1371/journal.pgen.1005158
Acute Glycine Pharmacodynamic Study - ClinicalTrials.gov, accessed on June 27, 2025, https://clinicaltrials.gov/study/NCT01610011?term=AREA%5BConditionSearch%5D(Glycine)&rank=5
Effect of Nutritional Supplements for Reducing Homocysteine Levels in Healthy Adults: A Systematic Review and Network Meta-Analysis of Randomized Trials, accessed on June 27, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12166197/
Effect of Nutritional Supplements for Reducing Homocysteine Levels in Healthy Adults: A Systematic Review and Network Meta-Analysis of Randomized Trials - ResearchGate, accessed on June 27, 2025, https://www.researchgate.net/publication/389063644_Effect_of_Nutritional_Supplements_for_Reducing_Homocysteine_Levels_in_Healthy_Adults_A_Systematic_Review_and_Network_Meta-Analysis_of_Randomized_Trials
Lowering Homocysteine - learn how to prevent high homocysteine levels - Food for the Brain, accessed on June 27, 2025, https://foodforthebrain.org/nutrition-for-healthcare-professionals/homocysteine-b-vitamins/
Homocysteine-lowering B vitamins - Food for the Brain, accessed on June 27, 2025, https://foodforthebrain.org/homocysteine-lowering-b-vitamins/
Effect of Homocysteine-Lowering Nutrients on Blood Lipids: Results from Four Randomised, Placebo-Controlled Studies in Healthy Humans | PLOS Medicine, accessed on June 27, 2025, https://journals.plos.org/plosmedicine/article?id=10.1371/journal.pmed.0020135
Effects of low-dose B vitamins plus betaine supplementation on lowering homocysteine concentrations among Chinese adults with hyperhomocysteinemia: a randomized, double-blind, controlled preliminary clinical trial - PubMed, accessed on June 27, 2025, https://pubmed.ncbi.nlm.nih.gov/36717385/
Methionine Restriction as a Life-Extension Strategy - NutritionFacts.org, accessed on June 27, 2025, https://nutritionfacts.org/video/methionine-restriction-as-a-life-extension-strategy/