You are on page 1of 20

Translational Medicine of Aging 3 (2019) 70e89

Contents lists available at ScienceDirect

Translational Medicine of Aging


journal homepage: www.keaipublishing.com/TMA

Amino acids in the regulation of aging and aging-related diseases


Clare-Ann Canfield a, Patrick C. Bradshaw b, *
a
Department of Biomedical Sciences, Keiser University Tampa, Tampa, FL, USA
b
Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA

a r t i c l e i n f o a b s t r a c t

Article history: Amino acids are the building blocks of protein, but also play important cellular signaling roles. The
Received 8 July 2019 mechanisms through which altered levels of many amino acids are sensed and the signals transmitted
Received in revised form are still largely unknown. Increasing evidence is showing that these signals may influence the aging
30 August 2019
process. In this regard, methionine restriction appears to be an evolutionary conserved mechanism to
Accepted 3 September 2019
Available online 4 September 2019
delay aging and supplementation with glycine can mimic methionine restriction to extend lifespan in
rodents. Tryptophan restriction may also activate specific anti-aging pathways, but it could interfere with
cognitive function. With exercise the consumption of dietary branched chain amino acids (BCAAs) may
Keywords:
Amino acids
be beneficial in building muscle mass, but high levels of BCAAs as well as tyrosine and phenylalanine in
Aging the bloodstream are associated with metabolic disease such as insulin resistance. Individual supple-
Lifespan mentation or restriction of several different amino acids has shown promise in the treatment of insulin
Yeast resistance in rodents. Much progress regarding the effects of amino acids on longevity has been made
C. elegans using yeast, nematodes, and fruit flies. Clearly, much more research is needed to understand the
signaling pathways activated by amino acid imbalance before efficacious and well-tolerated dietary in-
terventions can be developed for human aging and aging-related disorders. In this review the mecha-
nisms through which altered dietary and cellular levels of the twenty proteogenic amino acids affect
aging or aging-related disorders are discussed.
© 2020 The Authors. Production and hosting by Elsevier B.V. on behalf of KeAi Communication Co., Ltd.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).

1. Introduction piriform cortex region of the brain and will become anorexic on
such a diet [1].
There are 20 proteogenic amino acids used as the building There are mechanisms present to detect uncharged tRNAs and
blocks of protein synthesis. Plasma amino acid imbalance can result activate the eukaryotic initiation factor 2a (eIF2a) kinase general
from liver or kidney disease and may lead to some of the pheno- amino acid control non-derepressible 2 (GCN2) to signal the
types of those diseases [1]. Roughly 40 years ago amino acid solu- decreased level of a specific amino acid so the cell can decrease the
tions were formulated to treat such imbalances [1]. However, an rate of protein synthesis accordingly [7]. The other major pathway
imbalance of amino acids can also lead to the activation of stress for sensing the level of single amino acids and adjusting the rate of
response pathways and lifespan extension in model organisms protein synthesis is the target of rapamycin (TOR) pathway [8].
[2e4]. Dietary intake of large amounts of a single amino acid can be Modulation of either of these pathways in C. elegans can extend
toxic [5,6]. Single amino acids have been administered to humans lifespan in part due to increases in proteostasis [9], as decreased
without severe side effects in the 3e40 g per dose range with protein synthesis was found to correlate with increased lifespan
cysteine and methionine being the most toxic [1]. However, rodents under several experimental conditions [10,11]. But other mecha-
showed a slowed rate of growth when large amounts of a single nisms such as activation of the PHA-4/FoxA transcriptional regu-
amino acid were given with a low protein diet [1]. Rodents can lator [12] and the prevention of aging-induced mitochondrial
sense the lack of a single amino acid in their food using the anterior fission [13] to maintain energy levels [14] also likely play an
important role in lifespan regulation by TOR and GCN2. Since
rapamycin, a TOR inhibitor, can extend the lifespan of mice [15],
* Corresponding author. these pathways are also important targets for the regulation of
E-mail addresses: cedwardscanfield@keiseruniversity.edu (C.-A. Canfield), mammalian longevity.
bradshawp@etsu.edu (P.C. Bradshaw).

https://doi.org/10.1016/j.tma.2019.09.001
2468-5011/© 2020 The Authors. Production and hosting by Elsevier B.V. on behalf of KeAi Communication Co., Ltd. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 71

When amino acid intake exceeds the requirements for protein content matches the in silico translated codon usage of the
synthesis they are deaminated and catabolized into pyruvate, expressed genome (the exome) enhances the rate of growth and
acetyl-CoA, or mitochondrial citric acid cycle intermediates for development without affecting lifespan [32]. Therefore, the levels
energy generation [16]. In the nematode C. elegans, individual of amino acids in the diet may also one day be optimized for other
supplementation at an optimal concentration with the vast ma- purposes including increased stress resistance or a decreased rate
jority of amino acids or citric acid cycle intermediates extended of aging. This could be particularly useful for the treatment of
lifespan [3,17]. Higher concentrations frequently decreased lifespan diabetes where the plasma levels of all three BCAAs and phenyl-
suggesting a hormetic dose response. Supplementation with alanine and tyrosine positively associate with the disease [33]. This
peptone, a balanced source of amino acids decreased lifespan when may somehow be related to the finding that the level of these same
C. elegans was cultured in liquid media [3,18], but not when five amino acids plus glutamate increased in brain when rats were
cultured on agar plates [19], suggesting that the effects of high fed diets of altered protein content, while the levels of other amino
amino acid levels depend upon the culture conditions. acids changed little to none [34]. More research is needed on the
Like mammals, C. elegans evolved to utilize several D-amino ability of amino acid supplementation to alter levels of free amino
acids as signaling molecules and can metabolize a subset of these D- acids in mammalian brain regions.
amino acids for energy generation [20,21]. Individual supplemen- When examining the fibroblasts taken from 15 primate species,
tation to C. elegans with each of the three D-amino acids, D-alanine, 33 bird species, and 13 rodent species, free levels of 11 of the 20
D-aspartate, or D-glutamate, which are metabolized by the worms amino acids including arginine, glutamate, histidine, leucine,
and present in the E. coli that the C. elegans standardly consume as lysine, methionine, phenylalanine, proline, tryptophan, tyrosine,
the major portion of their diet [21], also extended lifespan [3], while and valine were found to have a positive association with species
supplementation with D-proline or D-serine, which are not readily lifespan [35]. Individual supplementation with 18 of the 20 amino
metabolized by the four characterized C. elegans D-amino acid acids extended lifespan in C. elegans (Table 1). These data suggest
oxidizing enzymes [20,21], did not extend lifespan [3]. This data that free amino acids have an important role in the regulation of
suggests metabolism as an important component of the longevity aging. Interestingly 8 of the 20 amino acids, including 5 of the 11
effects of supplemented amino acids. However, other data suggest listed above, were able to protect Caco-2 intestinal epithelial cells
that metabolism is not always required for amino acid-mediated from oxidative stress including alanine, cysteine, histidine, isoleu-
longevity in C. elegans as increasing the intracellular levels of cine, leucine, lysine, tryptophan, and valine [36]. Five of the amino
tryptophan [22] or branched chain amino acids (BCAAs) (isoleu- acids where the abundance associates with longevity that did not
cine, leucine, and valine) [23] by preventing their catabolism show an antioxidant effect in Caco-2 cells including arginine [37],
increased lifespan. So, clearly different signaling pathways are glutamate [38], methionine [39,40], proline [41,42], and tyrosine
activated by different amino acids in the regulation of lifespan. [43,44] have shown antioxidant effects using in vitro assays or have
Whether or not individual amino acid supplementation at an been shown to stimulate the expression of antioxidant enzymes in
optimal dose can influence the lifespan of other multicellular or- other cell types or tissues. These data suggest that decreased
ganisms such as fruit flies, rodents, or humans is mostly unknown. oxidative stress may play a role in the positive association between
But recently it was shown that glycine supplementation increased free amino acid levels and longevity. In the sections below we
the lifespan of mice [24] and the molecular mechanism is likely outline the current state of knowledge regarding how altered levels
similar to that induced by methionine restriction [25]. of each of the 20 proteogenic amino acids affect aging, aging-
Calorie restriction (CR) or dietary restriction (DR) is an evolu- related diseases, and the associated signaling pathways.
tionary conserved mechanism to delay aging resulting in lifespan
extension. In fruit flies the lifespan extending effects of DR have 2. Amino acids
been shown to be regulated by the protein to carbohydrate ratio.
Adding back essential amino acids to dietary restricted flies pre- 2.1. Alanine
vented lifespan extension [4]. This area has been extensively
reviewed [26] and will not be covered here in detail. Decreased Alanine is a non-essential amino acid and also one of 13
levels of single amino acids in the culture media also extended exclusively glucogenic amino acids [45]. Following its uptake and
replicative [27] or chronological (Table 1) lifespan in yeast. Repli- entry into the hepatic circulation, alanine is taken up by the liver,
cative aging in yeast appears to be due to a loss of vacuole acidifi- where much of the alanine pool is converted to pyruvate, which
cation that leads to the vacuolar release of neutral amino acids that enters into gluconeogenesis. In a fasted state, skeletal muscle re-
reduce mitochondrial membrane potential. This aging-induced loss leases large amounts of alanine and glutamine into the circulation
of vacuolar acidification is delayed by CR [28]. One large study using where they are taken up by the liver and kidney and used for
a Geometric Framework of Nutrition approach in mice also impli- gluconeogenesis [46]. Increased alanine consumption may be
cated decreased protein levels as having an important role in the beneficial for metabolic disorders such as type 2 diabetes as its
longevity effects of CR [29]. But these results could be due to the supplementation to a cultured pancreatic beta cell line increased
unique method (using cellulose to fill the void of restricted carbo- both non-oxidative and oxidative glucose metabolism and the
hydrate, lipid, or protein food mass) of implementing the restricted secretion of insulin [47]. Consistent with this, alanine infusion into
diets, as many older studies using rodents have shown that protein hyperglycemic dogs increased insulin secretion without much
restriction is not responsible for the large (~30%) increase in me- change in glucagon levels, but in the fasting state when glucose
dian lifespan of the typical 30%e40% CR diet [30]. But protein re- levels were low, alanine infusion stimulated glucagon secretion,
striction of 50%e85% can also increase lifespan up to roughly 15%, while having little effect on insulin levels [48]. In mice, dietary
apparently independently of the effects of CR. Many beneficial alanine was shown to prevent obesity due to a high fat diet [49].
disease-delaying effects of protein restriction have been found [31]. Alanine supplementation has also been shown to stimulate the
Since amino acids signal through distinct pathways to modulate proliferation of lymphocytes [50] and thymocytes [51], which may
lifespan in model organisms, consuming an optimized ratio of decrease the aging-related loss of immune system function. A diet
amino acids in the diet may be a viable approach for delaying aging containing 10% alanine administered to rats was also able to pre-
in humans as well. Along these lines, research using fruit flies and vent the formation or dissolve preexisting kidney stones [52].
mice has shown that consuming a diet where the amino acid Alanine levels have been shown to decline with aging in mouse
72 C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89

Table 1
Changes in lifespan caused by increased or decreased levels of specific amino acids.

Amino acid C. elegans % change in mean lifespan [3] Yeasta Fruit flies Rodents

1 mM in media 5 mM in media 10 mM in media Incr. amino acid Decr. Incr. amino acid Decr. amino acid Incr. amino acid Decr. amino acid
Amino acid

Ala þ8 ns þ11 Y [56] ↑[56]


Arg þ8 þ11 þ15 [[83]
Asn ns þ5 25 [[93]
Asp ns ns 6 [[106]
Cys þ9 þ16 ns
Gln þ15 þ16 6 [[93]
Glu þ14 þ11 8 [[106] [[93]
Gly þ10 ns ns [[24]
His ns þ9 þ12 ns[200]
Ile ns ns þ3 [[209] ns[273]
Leu þ16 þ6 þ7 [[209] [[238]
Lys þ7 þ8 þ6 [[93] ns[273]
Met ns þ8 þ14 [[273] [[4] [[245]
[[106]
Phe ns 8 12
Pro þ17 þ19 þ18
Ser þ8 þ18 þ22 [[321]
[[322]
Thr ns ns þ8 [[209] ns[273]
[[322]
Trp þ14 þ6 ns [[356] ns[4] [[337]
[[379]
Tyr þ10 þ5 þ2
Val þ13 þ8 ns [[209] ns[273][[322]

ns ¼ not significantly different than the control. A plus sign (þ) or up arrow ([) indicates increased lifespan, while a minus sign () or down arrow (Y) refers to decreased
lifespan. Incr. ¼ Increased and Decr. ¼ Decreased.
a
Data indicates yeast chronological lifespan, except for that for arginine and tryptophan, where it indicates replicative lifespan.

plasma [53] and muscle [54]. system [71]. Arginine supplementation has been shown to increase
Alanine, leucine, and glutamine levels were shown to decline in the secretion of insulin-like growth factor-1 (IGF-1) [72] and
aged parasitoid wasps [55]. In yeast, deletion of the Alt1 alanine growth hormone [73], both of which have been shown to have pro-
transaminase gene increased alanine levels and decreased chro- aging functions in mice [74,75]. Arginine is hydrolyzed by the
nological lifespan, while longevity extending CR decreased alanine enzyme arginase to form ornithine and urea in the liver during urea
levels [56]. In C. elegans supplementation with 1 mM or 10 mM cycle function [76] and in the liver and other tissues for polyamine
alanine extended lifespan [3], while supplementation with 5 mM synthesis from ornithine. The role of arginase in aging has been
did not [3,23] (Table 1). The metabolism of alanine to pyruvate may reviewed [77].
play a role in the extended longevity of nematodes as pyruvate Arginine stimulates mechanistic target of rapamycin (mTORC1)
supplementation also extended lifespan [57]. kinase activity by two different mechanisms [78,79]. First cyto-
plasmic arginine binds the CASTOR1 (cytosolic arginine sensor for
2.2. Arginine mTORC1 subunit 1) protein to stimulate mTORC1. Second the
SLC38A9 lysosomal amino acid transporter (that transports leucine,
Arginine is a semi-essential amino acid and the anti-aging ef- phenylalanine, isoleucine, tryptophan, methionine, valine, and
fects of arginine supplementation have been reviewed [58]. Argi- tyrosine [80]) interacts with the Ragulator protein complex that
nine catabolism is altered in learning and memory centers in the signals to mTORC1 dependent upon high (likely lysosomal) argi-
aged brain [59]. Arginine has been shown to stimulate the prolif- nine levels [81]. The activation of mTORC1 leads to increased pro-
eration of young human T lymphocytes to aid in wound healing tein synthesis and inhibition of autophagy that may exert a pro-
[60,61], but it did not stimulate the proliferation of young or aged aging effect in many cellular contexts.
rat lymphocytes [62]. Increased arginine levels resulted in a switch Increased arginine levels were found in aged Drosophila [82]. In
from glycolysis to oxidative phosphorylation in activated human T yeast deletion of the arginine permease CAN1 extended replicative
lymphocytes and an upregulated anti-tumor response directly lifespan depending upon the transcriptional regulator GCN4 and
associated with their enhanced survival [63]. Arginine was shown translational activation of stress response genes [83]. Arginine
to reverse the aging-related loss of acetylcholine-induced vasodi- supplementation potently increased the lifespan of C. elegans
lation of coronary endothelial cells in humans [64] and to improve (Table 1) [3]. Although C. elegans lacks a CASTOR1 homolog, its
depressed macrophage and wound immune function following genome does contain homologs to the lysosomal amino acid
hemorrhage in rats [65,66]. Long term arginine supplementation transporter SLC38A9 and the Ragulator complex protein Lamtor2
prevented the aging-related decrease in renal function [67]. The that may play a role in signaling cellular arginine levels.
protective effects of arginine may be due in part to its metabolism
to polyamines that are known to have anti-aging effects [68]. 2.3. Asparagine
Consistent with this mechanism, anti-aging DR has been shown to
increase the levels of polyamine synthesis enzymes through a post- Asparagine is a non-essential amino acid except when gluta-
transcriptional mechanism [69]. mine levels are very low [84]. Asparagine synthetase catalyzes the
Arginine is used for nitric oxide synthesis [70] and decreased cytoplasmic synthesis of asparagine from aspartate and ammonia.
nitric oxide synthesis contributes to the aging of the cardiovascular Older subjects showing physical frailty and sarcopenia were shown
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 73

to possess increased plasma levels of asparagine, aspartate, and ratio in human plasma were shown to increase with aging [109],
glutamate [85]. Asparagine plays a role in the proliferation of while the levels of some amino acids were shown to decline [110].
cancer cells via activation of mTOR during tumor development [86]. The plasma cystine/cysteine oxidation state is an important
Consistent with this role, a diet low in asparagine has been shown determinant of systemic inflammation [111], which likely contrib-
to be protective against breast cancer metastasis [87]. Asparagine utes to aging [112]. A reduced extracellular cystine/cysteine
and glutamine metabolism play essential roles in blood vessel oxidation state was also shown to restore free mitochondrial NADH
formation [88] and tumor growth [89]. Asparagine and glutamine levels in isolated neurons from aged mice and the 3xTg-AD Alz-
also play anti-inflammatory roles to protect intestinal integrity and heimer's disease mouse model [113]. Mitochondrial cysteine
stimulate intestinal epithelial cell division to protect against lipo- oxidation may play a role in the aging process as long-lived animals
polysaccharide (LPS) treatment [90]. Treatment with asparagine have evolved to encode less cysteines in the genomes of their
and aspartate together decreased fatigue and increased time to mitochondria, where ROS production is the highest [114]. Cysteine
exhaustion during moderate or intense exercise [91,92]. In yeast supplementation has shown promise as an adjuvant for type 2
removal of asparagine from the media extended the chronological diabetes [115]. When rats were fed a low protein diet cysteine
lifespan by inhibiting TOR kinase [93]. The asparagine catabolizing supplementation was shown to decrease the plasma levels of ho-
enzyme asparaginase was shown to be selectively retained in yeast mocysteine [116], a risk factor for neurodegenerative and cardio-
mother cells following the budding of daughter cells [94], largely vascular diseases [117]. However, high levels of plasma cysteine
explaining the decrease in asparagine levels in replicatively aged have been associated with many disorders including coronary heart
yeast [95]. Asparagine supplementation only slightly extended disease, Alzheimer's and Parkinson's diseases [118], rheumatoid
mean lifespan in C. elegans at a 5 mM dose and decreased lifespan arthritis [119], and systemic lupus erythematosus [120]. Systems
to the greatest extent of any amino acid at a 10 mM dose (Table 1) biology experiments in yeast identified that high cysteine levels
[3]. inhibit protein synthesis to cause toxicity and that supplementa-
tion with leucine and pyruvate prevented this toxicity. The pro-
2.4. Aspartate tective effect of leucine relied upon its transamination to alpha-
ketoisocaproate (KIC), which depended upon the function of the
The non-essential amino acid, aspartate, participates in a range tRNA methyltransferase NCL1 [121].
of important cellular functions such as the urea cycle and the The antioxidant glutathione is composed of cysteine, glutamate,
malate-aspartate shuttle, which transports nicotinamide adenine and glycine [122]. The oxidized glutathione disulfide (GSSG) to
dinucleotide (NADH) reducing equivalents between the cytoplasm reduced glutathione (GSH) ratio increases with aging, but is not in
and mitochondrial matrix. The shuttle plays an essential role in CR- strict equilibrium with the cystine/cysteine ratio [123]. When GSH
mediated longevity in yeast by increasing the nucleocytoplasmic is consumed orally by humans it is hydrolyzed in the intestine by g-
NADþ/NADH ratio to activate sirtuin deacetylases [96]. Aspartate is glutamyltransferase and plasma GSH was not altered [124]. Sup-
synthesized from oxaloacetate by aspartate aminotransferase. plementing with cysteine and glycine together has shown more
Mitochondrial electron transport chain function declines with ag- promise and led to enhanced plasma GSH levels in aged subjects
ing [97] and is required for aspartate and pyrimidine synthesis [98]. [125]. This elevation corresponded with a marked decrease in
Metals such as Mn2þ, Cu2þ, Zn2þ, and Mg2þ bind to aspartate to oxidative damage. N-acetyl-L-cysteine (NAC) is a membrane
form complexes with antioxidant function [99]. Aspartate (as well permeable prodrug form of cysteine that releases cysteine through
as proline and serine) supplementation was effective at decreasing the action of intracellular esterases. NAC supplementation has been
reactive oxygen species (ROS) production and increasing ATP levels shown to extend the lifespan of mice [126], fruit flies [127], and
in M17 neuroblastoma cells expressing alpha-synuclein treated nematodes [128,129]. But the effects were gender-specific and not
with ferrous sulfate [100]. Aspartate or glutamate was effective in concentration dependent suggesting hormesis [127]. The negative
promoting blood antioxidant levels when male pigs [101] or effects at higher doses may also suggest reductive stress [130].
weaned piglets [38] were challenged with hydrogen peroxide. Studies using NAC have been reviewed [131]. Consistent with the
However, glutamate, but not aspartate was effective at reducing effects of NAC, cysteine supplementation potently increased life-
diquat-mediated oxidative stress in piglets [102]. Aspartate sup- span in C. elegans at two lower doses, but not at the highest 10 mM
plementation also decreased ethanol-induced hepatotoxicity and dose administered (Table 1) [3].
oxidative stress in the testes of rats [103].
Within the brain, aspartate acts as an agonist of N-methyl-D- 2.6. Glutamate
aspartate (NMDA) receptors to regulate neurotransmission [104].
Therefore increased amounts of aspartate may contribute to aging- Glutamate is a non-essential amino acid most well-known for
related cognitive impairment by facilitating excitotoxicity [105]. its role as an excitatory neurotransmitter in the brain. Therefore,
Restriction of aspartate was shown to increase the chronological glutamate plays an essential role in learning and cognition, but at
lifespan of yeast [106], likely by inhibiting TOR kinase activity [107]. high levels glutamate induces neuronal excitotoxicity, contributing
However, the replicative lifespan of many yeast gene deletion to neuronal injury in neurodegenerative disorders including Alz-
strains positively correlated with increased levels of aspartate, heimer's disease (AD) [132] and amyotrophic lateral sclerosis (ALS)
methionine, proline, threonine, isoleucine, histidine, and glutamine [133]. Glutamate levels have been shown to decline in human brain
[108]. So some amino acids, such as aspartate, may show opposite during young adulthood [134]. Glutamate is only very slightly
effects on the chronological and replicative lifespan of yeast. permeable through the blood brain barrier (BBB), so relatively high
Aspartate was one of two amino acids that did not extend lifespan levels of dietary glutamate are non-toxic in healthy subjects. But it
when supplemented in the 1e10 mM range to C. elegans (Table 1) is not recommended to consume membrane permeable glutamate
[3]. esters that may permeate the BBB. Patients with neurodegenerative
disorders or who have had a stroke may also choose to limit their
2.5. Cysteine glutamate consumption as their BBB may be more permeable [135]
and lower plasma glutamate levels may stimulate limited release of
Cysteine is a non-essential amino acid. Total cysteine (including glutamate from the brain to decrease excitotoxicity [136,137]. The
reduced cysteine and oxidized cystine) and the cystine to cysteine crucial balance in the brain between low extracellular glutamate
74 C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89

concentrations and high intracellular glutamate concentrations is supplementation was shown to protect against Alzheimer's
kept in check by plasma membrane glutamate transporters. As- amyloid-beta toxicity in cultured neurons [157]. The role of amino
trocytes take up glutamate, convert it to glutamine, and release the acids in Alzheimer's disease brain has been reviewed [158,159]. In
glutamine to the extracellular space where it can be taken up by yeast inhibition of glutamine synthesis decreased glutamine levels
neurons that convert it back to glutamate for release at synaptic [160] and increased chronological longevity [93]. During chrono-
terminals [138]. logical aging the intracellular levels of 16 of the 20 amino acids
One potential therapy for lowering systemic glutamate levels in were found to decrease with glutamine levels declining to the
neurodegenerative disorders and stroke is the infusion of oxalo- greatest extent followed by glutamate, lysine, histine, methionine,
acetate and the aspartate transaminase enzyme [136,137]. How- and cysteine [161]. Supplementation of glutamine to C. elegans had
ever, glutamate plays important roles in the body such as binding an almost identical effect on lifespan as glutamate at all three doses
free ammonia, which is especially toxic to neurons [139], and it administered (Table 1) suggesting that the glutamine synthetase
serves as an important anaplerotic source of the anti-aging citric and glutaminase enzymes readily interconvert these amino acids
acid cycle intermediate alpha-ketoglutarate [140]. Higher levels of when they are consumed in the diet [3].
glutamate were found in the plasma of long-lived naked mole rats
compared to mice, while lower levels of many other amino acids 2.8. Glycine
including glycine, methionine, leucine, tyrosine, phenylalanine, and
tryptophan were found [141]. Another group found glutamate, The amino acid glycine is considered conditionally essential as it
alanine, aspartate, isoleucine, leucine, serine, and threonine levels can be synthesized from serine, but not always at the levels needed.
to be higher in the plasma of naked mole rats than mice [142]. One The known effects of dietary glycine supplementation have been
study found that high glutamate levels increased the chronological reviewed [162]. As mentioned earlier glycine is the one amino acid
lifespan of yeast [106]. However, another group using different studied thus far in which supplementation has been shown to
experimental conditions found removal of glutamate from the extend the lifespan of mice, albeit moderately [24]. Dietary glycine
media to extend chronological lifespan [93]. In C. elegans studies, supplementation also extended the lifespan of Fisher 344 rats
glutamate was one of the most potent lifespan extending amino through a mechanism mimicking methionine restriction to in-
acids when supplemented at the 1 and 5 mM doses, but it crease the hepatic clearance of methionine [163]. Glycine supple-
decreased lifespan when supplemented at the 10 mM dose mentation has been show to restore T cell activation, T cell one-
(Table 1) [3]. Knockout of the glutamate transporter glt-4 decreased carbon metabolism, and mitochondrial function in aged mice
nematode lifespan, while knockout of the glutamate transporter [164]. Administration of glycine was also shown to decrease
glt-5 increased lifespan [143]. oxidative stress and inflammation by decreasing the levels of
advanced glycation endproducts (AGEs) by inducing the expression
2.7. Glutamine of glyoxalase 1 [165].
By comparing young and old human fibroblasts it was found
Glutamine is an essential amino acid and the most abundant that epigenetic downregulation of the glycine-C-acetyltransferase
amino acid in the serum, CSF, and muscle. Its anti-aging roles have (GCAT) and serine hydroxymethyltransferase 2 (SHMT2) genes
been reviewed [144]. Glutamine is an important energy source, involved in mitochondrial glycine synthesis (see Fig. 1) are involved
especially in rapidly dividing cells such as epithelial cells and im- in the aging-related loss of cellular respiration. Adding glycine to
mune cells, due to its ability to be readily broken down and feed the culture media restored the phenotype of the aged cells back to
carbon into alpha-ketoglutarate in the citric acid cycle [145]. that of the young cells [166]. Supplemental glycine can be trans-
Increased glutamine levels can inhibit fatty acid oxidation, decrease ported across the mitochondrial inner membrane by the mito-
plasma glucose, and attenuate weight gain in mice prone to obesity chondrial carrier family member SLC25A38 [167]. Another study
[146]. A mitochondrial glutamine transport activity has been did not find much change in human whole body glycine meta-
characterized, but its molecular identity is still unknown [147]. bolism with aging, but found large changes following altered pro-
Following prolonged exercise by athletes supplemented with tein consumption [168]. Glycine is an inhibitory neurotransmitter
glutamine, an increased T-helper/T-suppressor cell ratio was that plays especially important roles in the spinal cord, brainstem,
measured, which corresponded with a decrease in the rate of and retina [169]. Increased glycine levels can induce hypothermia
infection [148]. Glutamine regulates the expression of genes and sleep by binding NMDA receptors in the suprachiasmatic nu-
involved with cellular proliferation and survival, as well as de- cleus of the brain [170]. Glycine supplementation protected the
creases the expression of pro-inflammatory genes such as NFҠb colon wall during radiotherapy [171]. Glycine infusion led to
[149]. Supplementation with glutamine increased the levels of anti- vasodilation in the rat kidney, but this effect was greatly reduced in
inflammatory monocytes and regulatory T lymphocytes in the aged animals [172]. N-arachidonoyl glycine and other N-acyl amino
serum of diabetic mice contributing to enhanced muscle regener- acids play important anti-inflammatory and analgesic functions
ation following limb ischemia [150]. Alanine facilitates glutamine [173,174]. Therefore, therapies using these compounds could be
metabolism. The combination of glutamine and alanine was shown tested for improving inflammation in aging-related disorders.
to reduce the glutathione couple (GSSG/GSH) [151] to increase Mild mitochondrial uncoupling is associated with increased
antioxidant function and had anti-inflammatory and cytoprotective longevity in nematodes [175] and fruit flies [176]. Overexpression of
effects in rats undergoing resistance exercise [152]. Long-term uncoupling protein 1 (UCP1) in skeletal muscle most greatly
supplementation with glutamine was shown to be beneficial for induced the expression of genes involved in glycine, serine, and
maintaining intestinal immunity in the elderly [153]. asparagine synthesis, and one-carbon metabolism [177], while the
Glutamine synthetase is highly expressed in the mammalian expression of pro-longevity cytoplasmic phosphoenolpyruvate
brain [154] where it may play a neuroprotective role in healthy carboxykinase (PEPCKc) [178] was also induced. Overexpression of
young animals [155]. Glutamine levels and glutamine synthetase glycine-N-methyltransferase (gnmt), which converts glycine and S-
activity are decreased in Alzheimer's disease brain, but glutamine adenosylmethionine to sarcosine and S-adenosylhomocysteine
synthetase was increased in a fraction of hippocampal neurons extended lifespan in Drosophila and knockdown of gnmt prevented
where it can play a maladaptive, cytotoxic role when glutamine DR-mediated longevity [179]. In mice plasma sarcosine levels
levels are low [156]. Consistent with this, glutamine declined with age and increased with anti-aging CR [180].
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 75

Fig. 1. Serine, glycine, threonine, methionine, cysteine, and histidine metabolism influence one-carbon and NADPH redox metabolism. TDH and GLY1 enzymes are not present in
humans. Metabolites shown on the mitochondrial (inner) membrane can be transported into and out of the matrix space. Glycine catabolism by the mitochondrial glycine cleavage
system is not shown. This results in the conversion of THF to 5,10-meTHF for additional one-carbon metabolism and is accompanied by the reduction of NADþ to NADH. THF,
tetrahydrofolate; 5-meTHF, 5-methyltetrahydrofolate; 5,10-meTHF, 5,10-methylenetetrahydrofolate; coA, coenzyme A; NADþ, nicotinamide adenine dinucleotide (oxidized); NADPþ,
nicotinamide adenine dinucleotide phosphate (oxidized); NADH, nicotinamide adenine dinucleotide (reduced); NADPH, nicotinamide adenine dinucleotide phosphate (reduced);
ATP, adenosine triphosphate; PPi, inorganic pyrophosphate; CO2, carbon dioxide; SHMT1, Serine Hydroxymethyltransferase 1; SHMT2, Serine Hydroxymethyltransferase 2;
MTHFD2L, Methylenetetrahydrofolate Dehydrogenase (NADPþ Dependent) 2 Like; MTHFD1L, Methylenetetrahydrofolate Dehydrogenase (NADPþ Dependent) 1 Like; MTHFD1,
Methylenetetrahydrofolate Dehydrogenase, Cyclohydrolase And Formyltetrahydrofolate Synthetase 1; TDH, Threonine Dehydrogenase; GLY1, Threonine Aldolase; GCAT, Glycine C-
Acetyltransferase; ALDH1L1, Aldehyde Dehydrogenase 1 Family Member L1; ALDH1L2, Aldehyde Dehydrogenase 1 Family Member L2; HAL, Histidine Ammonia-Lyase; UROC1,
Urocanate Hydratase 1; AMDHD1, Amidohydrolase Domain Containing 1; FTCD, Formimidoyltransferase Cyclodeaminase; MTHFR, Methylenetetrahydrofolate Reductase; MTR, 5-
Methyltetrahydrofolate-Homocysteine Methyltransferase; MAT1A, Methionine Adenosyltransferase 1A; MAT2A, Methionine Adenosyltransferase 2A; AHCY, Adenosylhomocys-
teinase; CBS, Cystathionine-Beta-Synthase; CTH, Cystathionine Gamma-Lyase; SDS, Serine Dehydratase; GCLC, Glutamate-Cysteine Ligase Catalytic Subunit; GSS, Glutathione
Synthetase; PHGDH, Phosphoglycerate Dehydrogenase; PSAT1, Phosphoserine Aminotransferase 1; PSPH, Phosphoserine Phosphatase; AGXT2, Alanine–Glyoxylate And Serine–
Pyruvate Aminotransferase 2; GRHPR, Glyoxylate And Hydroxypyruvate Reductase; GLYCTK, Glycerate Kinase.

Metabolomics studies of aging using C. elegans have yielded alanine, is found at high levels in muscle and brain, and has
fairly consistent data showing decreased levels of the vast majority similar physiological properties as histidine. But carnosine was
of amino acids with aging [181,182]. One group found glycine and shown to be less toxic in cell culture studies, so is a more promising
aspartate levels increased in C. elegans with aging, while the levels therapeutic [187]. Much data in model systems supports the po-
of the other amino acids peaked in the late larval stages or first few tential use of carnosine for the treatment of aging-related disorders
days of adulthood and declined thereafter [183]. Another group [191]. But we will focus on histidine as the anti-aging effects of
found glycine and alanine as the only amino acids to increase in carnosine have been previously reviewed [192]. Histidine supple-
abundance with aging [184], and a third group found glycine and mentation increased insulin secretion and glycemic control,
glutamine to be the only amino acids that increased in abundance increased glutathione peroxidase activity, and decreased pro-
with aging (see Table 2) [184]. DR resulting from the reduced inflammatory cytokine levels in diabetic mice [193]. Histidine
pharyngeal pumping in eat-2 mutants prevented these increases in supplementation increased GSH levels and enhanced catalase ac-
glycine and glutamine as well as prevented the decreased levels of tivity to decrease liver injury induced by chronic alcohol con-
arginine, valine, phenylalanine, and isoleucine, but not the sumption [194]. Individual administration of histidine, cysteine, or
decreased levels of the ketogenic amino acids lysine and leucine glycine, but not alanine, inhibited NF-kB activation in cultured
that occurred with aging [185]. Glycine and serine supplementation coronary endothelial cells [195]. Fig. 1 shows that catabolism of
have been shown to extend lifespan in C. elegans through stimu- histidine, cysteine, and glycine influence one-carbon and redox
lating one carbon metabolism including the methionine cycle [186]. metabolism.
Glycine supplementation only extended lifespan at concentrations Histidine can scavenge singlet oxygen and hydroxyl radicals
of 1 mM and below (Table 1) [3,186]. Knockdown of mel-32, the [196]. Low plasma histidine levels have been associated with
C. elegans homolog of serine hydroxymethyltransferase 1 and 2 (see inflammation, oxidative stress, and mortality in chronic kidney
Fig. 1), increased glycine levels and extended lifespan by preventing disease patients [197]. A metabolomics study of human plasma
glycine conversion to serine and instead funneled glycine to the found histidine levels to decline with aging [198]. A metabolomics
glycine cleavage system for one-carbon metabolism including the study of 647 human subjects linked high histidine levels (and two
methionine cycle. Consistent with this, knockdown of one of the other amino acids) with longevity as defined by attaining at least 80
four glycine cleavage system subunits, gcst-1, increased glycine years [199]. In studies using Drosophila histidine supplementation
levels and decreased lifespan [186]. showed no effect on the mean lifespan, while an equivalent dose of
carnosine extended the lifespan of male flies [200]. However, his-
2.9. Histidine tidine administration increased the lifespan of C. elegans at the two
higher concentrations tested, but not at a lower 1 mM dose
Histidine is an essential amino acid that acts as an anti-glycating (Table 1) [3]. The lifespan extension observed at the higher doses
agent [187], free radical scavenger [188], and metal chelator may have been due to its activation of the pro-longevity hypoxia
[189,190]. The dipeptide carnosine contains histidine and beta- inducible factor-1 (HIF-1) and SKN-1/Nrf2 stress response
76 C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89

Table 2
Changes in endogenous amino acid levels with aging.

Amino acid C. elegans Yeasta Fruit flies Mice or rats Humans

Tissue Effect Tissue Effect

Ala Y [183] ns [95] muscle Y [54]


Y [185] plasma Y [53]
[ [184]
Arg Y [185] ns [95] [[82] plasma Y [380]
Asn Y [183] Y [95]
Asp [ [183] ns [95]
[ [182]
Cys
Gln [ [185] ns [95] muscle [ [54] plasma [ [359]
Y [183] brain [ [59]
Glu Y [183] Y [95]
Y [185]
Gly [ [183] Y [95] muscle Y [54]
[ [185]
[ [184]
Y [182]
His Y [95] plasma Y [198, 359]
Ile Y [183] Y [95] brain [ [205] plasma Y [202]
Y [185]
Y [184]
Leu Y [183] Y [95] plasma [ [381] plasma Y [359]
Y [185] plasma Y [202]
Lys Y [185] [ [95]
Met Y [183] Y [95] plasma Y [53]
Y [184]
Phe Y [183] Y [95] [[82] muscle [ [54]
Y [185]
Y [184]
Pro Y [183] ns [95] plasma Y [53]
Ser Y [183] ns [95] plasma Y [53] plasma Y [359]
plasma [ [380]
Thr Y [184] ns [95] plasma Y [205] plasma Y [359]
Trp Y [183] Y [95] plasma Y [198, 359]
Y [184]
Tyr Y [183] Y [95] plasma Y [53] plasma [ [359]
Y [185] brain Y [205] plasma [ [380]
Val Y [183] Y [95] plasma [ [381]
Y [185]
Y [184]

ns ¼ not significantly different than the control. An up arrow ([) refers to increased amino acid level with aging, while a down arrow (Y) refers to decreased amino acid level
with aging.
a
Data from yeast was taken from results of replicative lifespan experiments.

pathways. isoleucine levels in the culture media extended chronological


longevity in yeast [209] and just slightly increased lifespan in
C. elegans [3,23]. In C. elegans, DAF-16, a homolog of mammalian
2.10. Isoleucine FOXO proteins, is a pro-longevity transcriptional regulator activated
when insulin signaling is blocked. Metabolomics analysis of worms
Isoleucine, an essential amino acid that is both glucogenic and found that many amino acids, including isoleucine, glutamine, and
ketogenic, is one of three branched-chain amino acids (BCAAs), valine were increased in long-lived daf-2 insulin receptor mutant
with leucine and valine being the other two [201]. A metabolomics worms, while glutamate levels declined [210]. Isoleucine and valine
study showed that isoleucine and leucine levels decreased in the levels returned to wild-type levels in daf-2; daf-16 double mutants
blood of aged human subjects [202]. Mice undergoing fasting, as did lifespan, suggesting a tight connection between these amino
which is associated with anti-aging and anti-cancer properties acids and longevity. However, isoleucine supplementation to
[203], showed increased plasma levels of all three BCAAs [204]. In a C. elegans only increased lifespan at the highest 10 mM dose of
metabolomics study of young and aged rat brains, isoleucine levels three doses tested and this effect was small (Table 1) [3].
increased, while tyrosine levels declined with aging [205]. Much
conflicting data has been obtained regarding the role of BCAAs on
metabolic health, insulin resistance, and glucose homeostasis 2.11. Leucine
[206]. In one study using rats, oral isoleucine was shown to
decrease plasma glucose levels and stimulate glucose uptake into Like isoleucine, leucine is an essential BCAA. Leucine is one of
C2C12 myotubes in a phosphatidylinositol 3-kinase (PI3K)-depen- two exclusively ketogenic amino acids and so is metabolized into
dent manner, while equivalent doses of leucine or valine did not acetyl-CoA and potentially into ketone bodies if blood glucose
[207]. Consistent with this, high blood levels of isoleucine have levels are low. A diet high in BCAAs has been shown to be associated
been associated with hypoglycemia in rats [208]. with increased mean lifespan of male mice [210]. However, a diet
Altered isoleucine/leucine (indistinguishable by mass spec- low in BCAAs [211,212], or only low in leucine [213], increased
trometry) levels were found in aged Drosophila [82]. Increased glucose homeostasis and enhanced metabolic health in mice. High
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 77

levels of the alpha-ketoacid of leucine, alpha-ketoisocaproic acid, restored specifically in neurons, but not when mTORC1 activity was
has been shown to cause mitochondrial dysfunction in neurons specifically restored in the intestine [13]. This data indicates that
[214]. A review of 34 reports in the literature has identified leucine neuronal TOR has a pro-aging function. Perhaps decreased function
supplementation to mice in a narrow range of 90e140 mg per day of both mTORC1 and mTORC2 by TOR kinas/let-363 knockdown in
consistently protected metabolic health against diet-induced the first example above explains the divergent results. It has been
obesity, while BCAA administration had no effect [215]. However, shown that specific mTORC1 deficiency compared to concurrent
BCAA administration was protective in models of type 1 diabetes mTORC1 and mTORC2 deficiency led to the activation of different
[215]. BCAA levels increased with aging in mouse skeletal muscle stress response pathways [239]. The inability to restore normal
[216]. Leucine levels were found to decrease by 14% in the urine lifespan by restoring mTORC1 activity in the intestine at first glance
from aged rats and increase by 6% with exercise [217]. appears to be inconsistent with other data where specific inhibition
Leucine strongly activates TOR kinase [218,219] by binding of mTORC1 in the intestine extended lifespan [239]. But it is
Sestrin1 and Sestrin2 [220e222] and by binding leucyl-tRNA syn- possible that specific knockdown of mTORC1 activity in the intes-
thetase [223,224]. Since inhibition of TOR kinase extends lifespan tine generates a signal inactivating mTORC1 activity in neurons that
across eukaryotic species, one may expect leucine supplementation leads to lifespan extension, which is consistent with the other
to decrease lifespan due to the downstream effects of TOR kinase findings.
activation including stimulation of protein synthesis and inhibition
of autophagy [225]. However, strikingly, to the best of our knowl- 2.12. Lysine
edge leucine supplementation has yet to result in decreased life-
span in any animal model yet tested, although a mouse diet high in Lysine is an essential ketogenic amino acid. Therefore both
BCAAs, and compensatorily lower in other amino acids, has been leucine and lysine can be supplemented while a subject is on the
shown to decrease lifespan by decreasing the ratio of threonine and anti-aging ketogenic diet [240,241] without stimulating gluco-
tryptophan to BCAAs, which led to decreased serotonin synthesis, neogenesis to raise glucose levels. A metabolomics study of 647
hyperphagia, and obesity [226]. human subjects linked high lysine levels (and two other amino
Leucine is a major amino acid regulating the rate of protein acids) with longevity as defined by attaining at least 80 years [199].
synthesis by the ribosome in many tissues such as skeletal muscle In humans lysine supplementation was shown to mimic protein
[227,228]. Activation of TOR kinase increased levels of the YY1- supplementation to increase plasma insulin and glucagon levels
PGC-1a complex in skeletal muscle to stimulate mitochondrial and when lysine was administered with glucose it decreased blood
oxidative function to match energy supply with energy demand glucose levels [242]. Lysine supplementation also decreased blood
[229]. Administration of leucine has been shown to decrease glucose levels and prevented cataract formation when adminis-
muscle degeneration in vitro [230] and may increase overall muscle tered to diabetic rats. An equivalent amount of a mixed amino acid
anabolism in young to middle-aged healthy humans [231,232]. supplement was also beneficial for both of these disease pheno-
Leucine supplementation has been shown to promote fatty acid types, but not to the extent of lysine [243]. Lysine supplementation
oxidation, stimulate mitochondrial biogenesis, increase NADþ induced an angiogenic response and increased the rate of wound
levels, and upregulate SIRT1 and AMP kinase (AMPK) activity in healing [244]. In yeast deletion of the lysine biosynthesis gene
skeletal myotube cells [233]. Although leucine is commonly Lys12 increased chronological lifespan [93]. Lysine was the only
consumed by young or middle aged athletes to facilitate an increase amino acid to increase in abundance in replicatively aged yeast,
in muscle mass, there is little evidence to support the long-term use while the abundance of roughly half of the amino acids declined
of leucine supplementation without exercise in the elderly for the [95]. In C. elegans lysine supplementation yielded small increases in
amelioration of sarcopenia [234,235]. lifespan at all three doses tested (Table 1) [3].
A study in Drosophila showed that lowering BCAAs in the diet
extended lifespan, but not more than a diet lowering three other 2.13. Methionine
essential amino acids threonine, histidine, and lysine [236]. So at
least in fruit flies, the BCAAs do not suppress longevity more than Reducing dietary levels of methionine, an essential amino, in
other essential amino acids. Restriction of BCAAs did not further rodents was shown to partially mimic the effects of CR and induce
extend lifespan of an already low amino acid diet suggesting that lifespan extension [245]. This topic has been extensively reviewed
BCAA restriction and general protein/amino acid restriction acti- [246e251]. Many of the protective effects of DR are blocked by
vate the same molecular mechanisms. One group showed that in- adding back methionine to the diet [252]. Some of the protective
dividual supplementation with leucine could extend chronological effects of CR and methionine restriction are due to increased
longevity in yeast [209] and that leucine availability was important transsulfuration pathway flux to increase the expression of the
for maximal lifespan extension during CR [237]. However, another cystathionine gamma lyase (CTH) gene and the production of
group under different conditions showed that blocking leucine hydrogen sulfide (H2S) [252,253]. Not only can CTH catabolize
synthesis in yeast stimulated chronological longevity [238]. cystathionine to cysteine, but it can catabolize cysteine to H2S
Leucine supplementation led to a prominent increase in lifespan [253]. Cysteine supplementation can inhibit some of the beneficial
in C. elegans that was slightly greater than valine and much greater effects of methionine restriction [254] perhaps through restoring
than isoleucine (Table 1) [3,23]. The extended longevity could be glutathione levels that are decreased during methionine restriction
mimicked by knocking down the bcat-1 BCAA aminotransferase [255]. The decreased glutathione levels led to the activation of
[23]. Surprisingly, knockdown of bcat-1 in neurons also extended protein kinase Relike endoplasmic reticulum kinase (PERK) and the
lifespan that was reduced by concurrent neuronal knockdown of integrated stress response (ISR) including transcription of the ATF4
TOR kinase (let-363) [23]. The lifespan extension resulting from transcriptional regulator [256]. Increased hepatic ATF4 levels
bcat-1 knockdown or leucine supplementation could also be miti- stimulated release of fibroblast growth factor 21 (FGF21) leading to
gated by ablation of ASI neurons (perform some functions of the increased expression of uncoupling protein 1 (UCP1) in white adi-
mammalian hypothalamus). This data suggests that neuronal TOR pose tissue (WAT) and a browning of WAT [256], which results in
has a pro-longevity function. In contrast to these results, long-lived increased energy expenditure. FGF21 has been shown to be trans-
global mTORC1 pathway-deficient worms lost their enhanced ported into the brain and signal to alter metabolism during a low
longevity and became normal-lived when mTORC1 activity was protein diet [257]. It also likely plays a similar role during
78 C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89

methionine restriction. The Nrf2 transcriptional regulator was also hydroxylase. Phenylalanine and glutamine levels were found to be
activated by methionine restriction, likely due to the decreased higher in the plasma of centenarians compared to other elderly
glutathione levels. Nrf2 increases transcription from antioxidant individuals [279]. Using a longitudinal approach phenylalanine/
response elements (AREs) in gene promoters. tyrosine metabolism, tryptophan metabolism, and methionine
Dietary methionine restriction inhibits mTORC1 activity to metabolism were shown to be altered with aging in the plasma of
activate autophagy by lowering tissue levels of S-adenosylme- marmosets, a primate aging model [280,281]. Specifically, phenyl-
thionine, which normally binds the SAMTOR protein to stimulate alanine levels decreased with age [281]. Phenylalanine binds hy-
mTORC1 activity [258]. In yeast increased methionine and S-ade- droxyl radicals and prevented hydroxyl radical-mediated inhibition
nosylmethionine levels also inhibit autophagy by stimulating the S- of acetylcholinesterase activity in brain homogenates [282].
adenosylmethionine-dependent methylation of the catalytic sub- Elevated serum phenylalanine has been linked to telomere loss
unit of the PP2A phosphatase by the Ppm1p methyltransferase in men [283], inflammatory disease [284], and type 2 diabetes [33].
leading to the dephosphorylation of the Npr2p regulator of auto- Dietary protein restriction in rats increased lifespan and decreased
phagy [259]. Therefore, decreased S-adenosylmethionine levels, phenylalanine levels in liver [285]. Increased phenylalanine levels
inhibition of mTORC1, and activation of autophagy likely explain were found in aged Drosophila [82]. Increased dietary protein as
the lifespan extension observed following knockdown of the S- well as individual supplementation with phenylalanine, methio-
adenosylmethionine synthetase-1 (sams-1) gene in C. elegans [260], nine, serine, or threonine greatly decreased the lifespan of the
even though stimulation of the methionine cycle is also associated Argentine ant, while individual supplementation with most other
with longevity under other conditions [186]. amino acids slightly decreased lifespan and supplementation with
Methionine restriction can also occur during a vegan diet glutamate, tyrosine, or tryptophan did not affect lifespan [286].
[261,262] or a strict ketogenic diet [263] due to the low protein Phenylalanine was one of two amino acids that did not extend the
intake prescribed to these diets, and the addition of methionine to a lifespan of C. elegans when added in the 1e10 mM range and was
ketogenic diet resulted in reversal of weight loss in mice [263]. A the only amino acid that decreased lifespan at the 5 mM dose
vegan diet is also typically lower in lysine content [264]. A diet (Table 1) [3]. Phenylalanine can be oxidized to the toxic metabolite
deficient in methionine is associated with many positive effects meta-tyrosine (m-tyrosine), which decreases C. elegans lifespan. So
such as enhanced fatty acid oxidation, increased energy expendi- during oxidative stress C. elegans upregulates the expression of
ture, decreased ROS production, reduced oxidative damage [265], tyrosine aminotransferase (tatn-1) to deaminate excess tyrosine,
and inhibition of tumorigenesis [266]. Most notably, methionine which likely decreases product inhibition of phenylalanine hy-
restriction extended lifespan in mice by nearly 7% [267], rats by 44% droxylase to stimulate the conversion of phenylalanine to tyrosine
[268], and Drosophila by 36% [269]. Similar to CR, methionine re- and decrease phenylalanine and m-tyrosine levels [287].
striction produced a lifelong reduction in fatty body mass [245]. A
diet that restricts methionine by 83% in humans, similar to the 2.15. Proline
extent that produces the positive longevity effect in rodents, has
been established [266]. Glycine supplementation to rats mimicked Proline is a non-essential amino acid that is synthesized from
the longevity effects of methionine restriction though stimulating glutamate in a two-step NADPH-requiring pathway. A metab-
the clearance of hepatic methionine [270]. Methionine restriction olomics study of aging mice found proline, alanine, serine, tyrosine
had greater benefits to the metabolic health of mice than equivalent and methionine to decrease in plasma with aging [53]. Increased
leucine restriction [271]. plasma proline concentrations have also been associated with
When fruit flies were placed on a protein-deficient diet that sarcopenia in the elderly [288]. Dietary proline supplementation
extended lifespan, the addition of methionine could restore had an immunostimulatory effect to protect mice from Pasteurella
fecundity without decreasing lifespan, suggesting that it is possible multocida infection [289]. The topical application of proline to
to design a modified amino acid diet that offers the positive effects cutaneous wounds led to an acceleration of healing in rats [290].
of DR without the negative effects [4]. Methionine restriction also Proline acts as a metal chelator, supports osmotic balance, and
extended lifespan in Drosophila [4], but only on a low amino acid prevents oxidative damage [291]. Proline supplementation miti-
diet [272]. In one study in yeast, chronological lifespan was gated the early stage of liver injury in rats by decreasing oxidative
extended by methionine restriction, but not by lysine, isoleucine, stress. It also improved redox status to improve nitric oxide avail-
threonine, or valine restriction [273], and this increased longevity ability and prevent a pathological increase in blood pressure [292].
required autophagy-dependent vacuolar acidification [274]. Proline was the second most potent amino acid at extending
Methionine restriction extended the chronological lifespan of yeast lifespan in C. elegans (Table 1) and the most potent for increasing
even when H2S production was inhibited [275]. Methionine sup- thermotolerance [3] suggesting that it stimulates pathways medi-
plementation increased mitochondrial oxidative metabolism in ating proteostasis. It has been shown to extend the lifespan by
yeast [276] and also increased oxidative stress resistance by transiently increasing ROS levels, which then stimulate stress
increasing pentose phosphate pathway flux and cellular reduced response pathways that induce antioxidant gene expression [293].
nicotinamide adenine dinucleotide phosphate (NADPH) levels Proline is hydroxylated to form hydroxyproline, a prominent amino
[277]. Methionine supplementation to fasted mice and catabolism acid in collagen, a major protein of the C. elegans cuticle [294].
of the methionine through the little studied methionine trans- Collagen remodeling was shown to be required for C. elegans life-
amination pathway decreased blood sugar levels by inhibiting he- span extension due to daf-2 insulin receptor mutation [295].
patic gluconeogenesis through GCN5 acetyltransferase and PGC-
1a-dependent mechanisms [278]. Methionine supplementation 2.16. Serine
was shown to extend the lifespan of C. elegans at the two highest
doses administered, but not at the lowest dose (Table 1) [3]. Serine is a non-essential amino acid that is synthesized in a 3
step pathway from the glycolytic intermediate 3-phosphoglycerate
2.14. Phenylalanine (Fig. 1). Serine supplementation showed many protective effects on
the phenotypes induced by a high fat diet in mice including
Phenylalanine is an essential amino acid that is both glucogenic increasing glucose tolerance and insulin sensitivity, decreasing
and ketogenic and is converted to tyrosine by phenylalanine hepatic lipid levels, and preserving reduced glutathione (GSH)
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 79

levels by preventing the hypermethylation of promoters of GSH ketogenic. Threonine supplementation in humans can greatly in-
synthesis genes [296]. Serine supplementation also activated AMPK crease its levels in plasma [323]. Threonine is a precursor for
in palmitic acid-treated primary hepatocytes to decrease ROS glycine synthesis in most mammals and threonine supplementa-
production [296e299]. Although short term serine supplementa- tion has been shown to increase plasma glycine levels in pigs [324]
tion did not reduce weight gain on a high fat diet [296], long term and increase glycine neurotransmitter levels in the brain of rats
serine supplementation to a normal chow diet was shown to [325]. An excess or deficiency in dietary threonine decreased pro-
decrease food consumption and weight gain [300]. Serine supple- tein synthesis rates in pigs [324]. Threonine levels were shown to
mentation increased Sirt1 protein deacetylase expression and decline in the plasma of aged rats [205]. A metabolomics study of
decreased ROS levels in the hypothalamus, and decreased levels of 647 human subjects linked high threonine levels (and two other
phosphorylated NFkB to decrease the levels of pro-inflammatory amino acids) with longevity as defined by attaining at least 80 years
cytokines [300]. Serine has been shown to be neuroprotective [199]. In human [326] and mouse embryonic stem cells, threonine
[301,302] in part through upregulation of the expression of ER levels can regulate the cell cycle G1/S phase transition and prolif-
protein disulfide isomerase to increase proteostasis [303,304]. With eration [327,328]. Threonine deficiency in mammals has been
aging, microglia synthesize and release increased amounts of D- associated with depression and neurological dysfunction [329].
serine, synthesized from L-serine. D-serine is a co-agonist for NMDA Threonine supplementation to chickens led to an improved im-
receptors [305] and therefore this increased extracellular D-serine mune response [330] and protection from LPS-induced inflamma-
may contribute to the excitotoxicity induced by neuronal NMDA tion and intestinal barrier damage [331]. Threonine
receptor activation in aged animals [306]. A phase I clinical trial has supplementation to pigs led to increased protection against a viral
been conducted testing the effects of serine supplementation on challenge [332]. Supplementation with threonine has been shown
amyotrophic lateral sclerosis (ALS) patients [307,308]. A 15 g dose to extend chronological longevity in yeast [209]. However, a
of serine twice a day was well tolerated with only mild intestinal different group using different conditions found threonine re-
discomfort in a few subjects. It has been postulated that the long striction (as well as valine restriction) to increase yeast chrono-
life of Ogimi village Okinawans may be due in part to the high logical lifespan [322,333,334] by inhibiting TOR kinase [322].
content of serine in their diet [309]. Threonine supplementation only extended C. elegans lifespan
Serine metabolism can increase cellular antioxidant function slightly at the highest (10 mM) dose of three doses applied
through several different mechanisms. In studies with human (Table 1). Threonine can be converted to glycine in in one step by
endothelial cells, serine supplementation increased antioxidant threonine aldolase and in two steps by the threonine dehydroge-
gene expression through upregulation of Nrf2 and its downstream nase and GCAT enzymes (Fig. 1). Therefore threonine could extend
effector heme oxygenase-1 [310]. Serine is a direct precursor for the lifespan by a similar stimulation of the methionine cycle as shown
synthesis of glycine and cysteine, which are limiting for the syn- for glycine or serine supplementation [186]. However, another
thesis of GSH. See Fig. 1. Once GSH is oxidized, the GSSG is reduced mechanism could be involved. The 2-amino-3-ketobutyrate prod-
by NADPH and recycled for use. One-carbon units derived from uct of the threonine dehydrogenase reaction is unstable and
serine can also be added to homocysteine for methionine synthesis. spontaneously decarboxylates into aminoacetone, which is further
Serine is transported into the mitochondrial matrix by the SFXN1 broken down producing hydrogen peroxide and methylglyoxal.
transporter [311]. Catabolism in mitochondria, initiated by the Knockdown of the C. elegans GCAT homolog T25B9.1 extended
enzyme SHMT2, increased mitochondrial NADPH levels to increase lifespan through increasing 2-amino-3-ketobutyrate and the pro-
antioxidant function [312]. Data has emerged that mitochondrial duction of hydrogen peroxide and methylglyoxal, which then
serine can also be catabolized in a three step pathway to 2- activated the SKN-1 and HSF-1 transcriptional regulators. So life-
phosphoglycerate (Fig. 1), which can then be exported to the span was extended by a hormetic effect [335]. Therefore, increased
cytoplasm for gluconeogenic or glycolytic metabolism [313,314], production of these reactive products could also contribute to the
while synthesis of serine from glycolytic 3-phosphoglycerate oc- lifespan extension that occurs during threonine supplementation.
curs in the cytoplasm [315,316]. Serine is also an essential building
block of lipids such as phosphatidylserine [317], sphingolipids, and 2.18. Tryptophan
ceramides. Serine deficiency results in mitochondrial fragmenta-
tion and dysfunction [318]. Tryptophan is an essential amino acid that is both glucogenic
Anti-aging CR was shown to increase serine dehydratase levels and ketogenic. Dietary tryptophan restriction in rats delayed
in the liver of aged mice to increase the catabolism of serine to reproductive aging [336] and extended lifespan [337,338]. How-
pyruvate to fuel gluconeogenesis [319]. Subjects with fibromyalgia ever, the rats were switched to a normal diet later in life, so more
showed lower plasma levels of serine and histidine [320]. In yeast experiments need to be done to confirm these lifespan findings
inhibition of serine synthesis from 3-phosphoglycerate (see Fig. 1) [339]. Tryptophan is a precursor for the neurotransmitter serotonin
increased chronological lifespan by stimulating glyoxylate shunt and for the hormone melatonin that regulates circadian rhythms.
activity and trehalose synthesis [321]. In other studies with yeast, Tryptophan is degraded by the kynurenine pathway to nicotin-
serine was further shown to promote aging by activating a phos- amide adenine dinucleotide (NADþ) and other end products with
phorylation signaling cascade using the Pkh1/2, Sch9 (homolog of several of the intermediates being toxic such as quinolinic acid, due
mammalian S6 kinase), and Rim15 protein kinases [322]. In to it being an agonist of the neuronal NMDA receptor. The inter-
contrast to the results with yeast, in C. elegans serine supplemen- mediate kynurenic acid is neuroprotective because it is an antag-
tation increased lifespan to the greatest extent of any amino acid onist of the NMDA receptor. Tryptophan levels were shown to
(Table 1) [3]. Serine supplementation activated the HIF-1, DAF-16, decrease in human serum with aging [198], while toxic tryptophan
and SKN-1 stress response pathways and lifespan extension was catabolites were shown to increase [340]. This increase in trypto-
dependent upon the function of AAK-2/AMPK and sirtuin SIR-2.1 phan catabolism with aging is likely due to increased levels of the
[3]. enzyme indoleamine-2,3-dioxygenase (IDO) [340]. IDO breaks
down tryptophan to kynurenine in tissues outside the liver, while
2.17. Threonine tryptophan-2,3-dioxygenase performs this task in the liver. IDO is
induced by pro-inflammatory cytokines and superoxide [340],
Threonine is an essential amino acid that is both glucogenic and which increase with aging.
80 C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89

Tryptophan plasma levels have been measured or tryptophan been shown to enhance dopaminergic neurotransmission in Par-
supplementation studies have been performed for many disorders kinson's disease patients [366]. However, high levels of tyrosine in
as recently reviewed [341]. In brief, studies have been performed the plasma increase the risk of type 2 diabetes to a similar extent as
focusing on tryptophan in the following aging-related disorders: increased levels of each of the three BCAAs [33]. In C. elegans
cardiovascular disease [342e344], chronic kidney disease [345], tyrosine supplementation increased lifespan moderately at the
diabetes [346], depression [347,348], inflammatory bowel disease lowest 1 mM dose, but only slightly at the two higher doses
[349], and multiple sclerosis [350]. Tryptophan levels were most (Table 1) [3].
frequently lower in the disease state and supplemental tryptophan
most often decreased disease phenotypes. There were two excep- 2.20. Valine
tions. Limiting tryptophan was beneficial for chronic kidney disease
and increased plasma tryptophan levels were found to predict Valine is an essential glucogenic BCAA. A breakdown product of
future type 2 diabetes in a Chinese population [346]. However, this valine, 3-hydroxyisobutyrate (3-HIB) was shown to drive vascular
association with type 2 diabetes has yet to be found in other ethnic fatty acid transport and insulin resistance [367]. However, valine
groups. In fact, several tryptophan catabolites have been found to supplementation to rats reduced fatigue during swim exercise and
be increased in the plasma of subjects with type 2 diabetes [351] partly prevented the drop in blood glucose and liver glycogen
suggesting increased tryptophan catabolism. In rats tryptophan following exercise, while leucine or isoleucine supplementation did
supplementation was shown to lower blood glucose levels, and not [368]. Valine supplementation also protected against paraquat-
delay disease progression in hereditary diabetic rats [352]. A induced toxicity in rats [369]. Valine supplementation, more than
decrease in plasma tryptophan levels with aging was associated the supplementation of any other amino acid, increased ATP levels
with a loss of cognitive function in non-demented women [340]. and protected against oxidative stress in M17 neuroblastoma cells
Supplemental tryptophan was also shown to improve the memory overexpressing alpha-synuclein [100]. This could be due to an in-
of aged rats [353], while tryptophan-enriched cereals were shown crease in mitochondrial biogenesis induced by BCAAs [210], as
to improve the sleep/wake cycle of elderly humans [354]. isoleucine showed a similar, but smaller effect [100]. The mito-
Altered tryptophan levels were found in aged Drosophila [82] chondrial carrier family member SLC25A44 has recently been
and long-lived flies showed higher tryptophan levels [355]. identified as a mitochondrial BCAA transporter that plays an
Decreasing tryptophan uptake by deleting the tryptophan especially important role in brown adipose tissue metabolism
permease Tat2p increased replicative lifespan in yeast [356]. [370]. Using yeast, one group showed that supplemental valine
Blocking tryptophan catabolism extended lifespan and decreased extended chronological longevity [209]. However, another group
aging-related proteotoxicity in C. elegans [22]. Tryptophan supple- using different conditions found valine restriction to increase yeast
mentation increased the lifespan of C. elegans at two lower doses, chronological lifespan by inhibiting TOR kinase [322,333,334].
but not at the highest 10 mM dose tested (Table 1) [3]. Tryptophan Replicative aging in yeast has been shown to lead to an increase in
was the only lifespan-extending amino acid tested out of ten tested the expression of amino acid catabolism genes, a decrease in the
that extended the lifespan in daf-16 mutant worms. Also trypto- expression of amino acid biosynthesis genes, and a decrease in
phan and cysteine were the only two lifespan-promoting amino most amino acid levels, especially all three BCAAs but also aspar-
acids out of ten tested that did not activate a transcriptional re- agine, glutamate, glycine, histidine, and methionine (see Table 2)
porter for SKN-1/Nrf2 activity, suggesting novel mechanisms of [95]. In C. elegans valine supplementation increased lifespan
lifespan extension [3]. Tryptophan and a few of its catabolites were moderately at the two lower 1 mM and 5 mM doses, but not at the
shown to activate the mitochondrial unfolded protein response highest dose (Table 1) [3].
(UPRmt) and the ER stress response pathways in C. elegans [3]. The
UPRmt is frequently, but not always associated with nematode 3. Conclusions
longevity [357,358].
In the last decade major strides have been made in the under-
2.19. Tyrosine standing that amino acids are signaling molecules and can be used
as a relatively non-toxic treatments to alter the rate of aging in
Tyrosine, a non-essential amino acid, is both glucogenic and experimental models. Important molecular details of how specific
ketogenic and is synthesized from phenylalanine by the phenylal- amino acids such as leucine and arginine activate TOR to decrease
anine hydroxylase enzyme. Tyrosine is the precursor for the neu- lifespan have been elucidated. But the molecular mechanisms
rotransmitters dopamine and norepinephrine. In a metabolomics through which increased levels of many of the amino acids provide
study of human plasma, tyrosine and glutamine levels increased health benefits remain largely unknown. The ability of glycine to
with age, while histidine, threonine, tryptophan, leucine, and extend longevity in rodents is an important breakthrough as amino
serine decreased with age [359]. Supplementation with tyrosine in acid-restricted diets, such as methionine-restricted diets, are
both hypertensive humans and rats led to a decrease in blood expensive and can be relatively unpalatable resulting in difficulties
pressure [360]. In one study of human subjects, increased dietary in subject retention in clinical trials. Another important goal should
tyrosine levels were linked with increased cognitive performance be the determination of the effects of serine and threonine on the
[361]. However, another study found that supplementation of lifespan of mice, since these amino acids can be metabolized to
moderate tyrosine levels had no effect, while high dietary tyrosine glycine but also have their own unique properties. In support of
levels decreased cognitive performance in older adults [362]. Au- these unique properties, serine or glycine administration to rats
thors of a recent review suggest that tyrosine supplementation altered brain amino acid levels in different ways [371]. Women
likely only increases cognition during times of stress when either have been shown to have higher plasma levels of serine and glycine
dopamine or norepinephrine levels are depleted [363]. There is no than men, while 13 of the other 18 amino acids were lower [359].
consistent data that tyrosine supplementation can decrease the Whether these amino acid level differences contribute to the
effects of stress on cognitive or physical performance [364]. Tyro- extended longevity of women compared to men remains to be
sine supplementation has been shown to increase the vasocon- determined. Serine supplementation in humans has been shown to
striction response to body cooling in older adults, so it may aid lower plasma homocysteine levels, a marker of metabolic and
thermoregulatory function [365]. Tyrosine supplementation has cardiovascular disease [372]. This may occur through stimulation of
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 81

cystathionine-beta synthase (CBS) activity (See Fig. 1). Serine Funding


metabolism in mitochondria leads to NADPH production that pro-
tects mitochondria from ROS-mediated damage, but at the expense This research was funded by The National Institutes of Health
of NADPH oxidation in the cytoplasm [373]. However, some of these grant numbers AG059096 and AG046769 awarded to PB.
lost cytoplasmic reducing equivalents can be restored if increased
serine levels stimulate CBS activity of the transsulfuration pathway Acknowledgements
to increase cysteine levels, which decreases the plasma membrane
import of oxidized cystine and its subsequent reduction to cysteine, We would like to thank John Canfield, Neil Copes, Vedad Delic,
which is indirectly coupled to cytoplasmic NADPH oxidation [374]. and Jeddidiah Griffin for helpful discussion.
The human threonine dehydrogenase [375] and threonine
aldolase [376] genes have mutated so functional proteins are not
References
expressed, while the expressed proteins are functional in rodents
for the conversion of threonine to glycine (Fig. 1) [373]. So threo- [1] E. Roth, W. Druml, Plasma amino acid imbalance: dangerous in chronic
nine supplementation to humans would not likely yield similar diseases? Curr. Opin. Clin. Nutr. Metab. Care 14 (1) (2011) 67e74.
effects to those of glycine. Several amino acids from the diet are [2] J.P. Aris, L.K. Fishwick, M.L. Marraffini, A.Y. Seo, C. Leeuwenburgh,
W.A. Dunn Jr., Amino acid homeostasis and chronological longevity in
catabolized to a large extent by first pass splanchnic [377] and Saccharomyces cerevisiae, Sub Cell. Biochem. 57 (2012) 161e186.
hepatic [378] metabolism and amino acid supplementation stra- [3] C. Edwards, J. Canfield, N. Copes, A. Brito, M. Rehan, D. Lipps, J. Brunquell,
tegies may fail to raise amino acid levels to a large extent in pe- S.D. Westerheide, P.C. Bradshaw, Mechanisms of amino acid-mediated life-
span extension in Caenorhabditis elegans, BMC Genet. 16 (2015) 8.
ripheral tissues due to this issue. Since the hypothalamus is a major [4] R.C. Grandison, M.D. Piper, L. Partridge, Amino-acid imbalance explains
regulator of the aging process, amino acids may need to cross the extension of lifespan by dietary restriction in Drosophila, Nature 462 (7276)
BBB to exert their anti-aging effects and in doing so they must (2009) 1061e1064.
[5] P.J. Garlick, The nature of human hazards associated with excessive intake of
compete with other amino acids for transport [158]. So it may be amino acids, J. Nutr. 134 (6 Suppl) (2004) 1633Se1639S, discussion 1664S-
difficult to raise the level of many amino acids in the brain. To 1666S, 1667S-1672S.
address these issues, studies could be performed with membrane [6] A.E. Harper, N.J. Benevenga, R.M. Wohlhueter, Effects of ingestion of
disproportionate amounts of amino acids, Physiol. Rev. 50 (3) (1970)
permeable forms of amino acids such as N-acetylated forms and 428e558.
ethyl ester forms that may more readily permeate throughout the [7] J. Gallinetti, E. Harputlugil, J.R. Mitchell, Amino acid sensing in dietary-
body. restriction-mediated longevity: roles of signal-transducing kinases GCN2
and TOR, Biochem. J. 449 (1) (2013) 1e10.
Another challenge that needs to be addressed is determining
[8] D.M. Sabatini, Twenty-five years of mTOR: uncovering the link from nutri-
the correct amount of an amino acid to supplement to obtain a ents to growth, Proc. Natl. Acad. Sci. U. S. A. 114 (45) (2017) 11818e11825.
therapeutic effect as a hormetic dose response is often observed [9] A. Vlanti, A. Rousakis, P. Syntichaki, GCN2 and TOR converge on aging, Aging
[3]. The presence of transaminases and alpha-ketoacids in the (Albany NY) 5 (8) (2013) 584e585.
[10] M. Kaeberlein, B.K. Kennedy, Protein translation, 2007, Aging Cell 6 (6)
blood and many tissues including the liver lead to the relatively (2007) 731e734.
quick transamination of one amino acid and synthesis of another. [11] P. Syntichaki, K. Troulinaki, N. Tavernarakis, Protein synthesis is a novel
Many of the common transaminases such as aspartate trans- determinant of aging in Caenorhabditis elegans, Ann. N. Y. Acad. Sci. 1119
(2007) 289e295.
aminase and alanine transaminase use alpha-ketoglutarate as an [12] A. Rousakis, A. Vlassis, A. Vlanti, S. Patera, G. Thireos, P. Syntichaki, The
acceptor of an amino group to form glutamate. Therefore, the ef- general control nonderepressible-2 kinase mediates stress response and
fects of supplementation with certain amino acids may be a result longevity induced by target of rapamycin inactivation in Caenorhabditis
elegans, Aging Cell 12 (5) (2013) 742e751.
of increased glutamate or decreased alpha-ketoglutarate levels [13] Y. Zhang, A. Lanjuin, S.R. Chowdhury, M. Mistry, C.G. Silva-Garcia, H.J. Weir,
instead of from a direct effect of the supplemented amino acid. In C.L. Lee, C.C. Escoubas, E. Tabakovic, W. Mair, Neuronal TORC1 modulates
this regard, alpha-ketoglutarate has anti-aging properties [140] longevity via AMPK and cell nonautonomous regulation of mitochondrial
dynamics in C. elegans, eLife 8 (2019).
even at low doses [3] and so supplementation with high levels of
[14] S.N. Chaudhari, E.T. Kipreos, Increased mitochondrial fusion allows the sur-
certain amino acids may shorten lifespan through alpha- vival of older animals in diverse C. elegans longevity pathways, Nat. Com-
ketoglutarate depletion. Amino acid transamination likely ex- mun. 8 (1) (2017) 182.
[15] A. Bitto, T.K. Ito, V.V. Pineda, N.J. LeTexier, H.Z. Huang, E. Sutlief, H. Tung,
plains why 18 of the 20 amino acids extended lifespan in C. elegans
N. Vizzini, B. Chen, K. Smith, D. Meza, M. Yajima, R.P. Beyer, K.F. Kerr,
and why many of the amino acids demonstrated a similar hormetic D.J. Davis, C.H. Gillespie, J.M. Snyder, P.M. Treuting, M. Kaeberlein, Transient
dose response [3]. At high doses of supplementation a common rapamycin treatment can increase lifespan and healthspan in middle-aged
mechanism of toxicity may be occurring even though a different mice, eLife 5 (2016).
[16] E.M. Jones, Amino acid metabolism, Annu. Rev. Biochem. 34 (1) (1965)
amino acid was supplemented. Heavy isotope amino acid tracer 381e418.
studies will be useful to follow the metabolism of the supple- [17] C.B. Edwards, N. Copes, A.G. Brito, J. Canfield, P.C. Bradshaw, Malate and
mented amino acids to determine if common downstream pro- fumarate extend lifespan in Caenorhabditis elegans, PLoS One 8 (3) (2013),
e58345.
longevity and pro-aging catabolites can be identified as causative [18] T. Yamaguchi, A. Onodera, K. Yasuda, Y. Nishio, M. Arai, M. Tsuda,
longevity modulators. Therefore, much work remains to harness M. Miyazawa, P.S. Hartman, A low cost and quick assay system using the
the great potential of altering amino acid levels for the treatment free-living nematode Caenorhabditis elegans to determine the effects of
Kampo medicines on life span, AATEX 13 (2008) 1e10.
of aging and aging-related diseases. [19] D. Chandler-Brown, H. Choi, S. Park, B.R. Ocampo, S. Chen, A. Le, G.L. Sutphin,
L.S. Shamieh, E.D. Smith, M. Kaeberlein, Sorbitol treatment extends lifespan
and induces the osmotic stress response in Caenorhabditis elegans, Front.
Genet. 6 (2015) 316.
Conflict of interest [20] M. Katane, Y. Saitoh, Y. Seida, M. Sekine, T. Furuchi, H. Homma, Comparative
characterization of three D-aspartate oxidases and one D-amino acid oxidase
The authors declare there is no conflict of interest. from Caenorhabditis elegans, Chem. Biodivers. 7 (6) (2010) 1424e1434.
[21] Y. Saitoh, M. Katane, T. Kawata, K. Maeda, M. Sekine, T. Furuchi, H. Kobuna,
T. Sakamoto, T. Inoue, H. Arai, Y. Nakagawa, H. Homma, Spatiotemporal
localization of D-amino acid oxidase and D-aspartate oxidases during
Declarations of interest development in Caenorhabditis elegans, Mol. Cell. Biol. 32 (10) (2012)
1967e1983.
[22] A.T. van der Goot, W. Zhu, R.P. Vazquez-Manrique, R.I. Seinstra, K. Dettmer,
Declarations of interest: none. The funders had no role in the H. Michels, F. Farina, J. Krijnen, R. Melki, R.C. Buijsman, M. Ruiz Silva,
writing of the manuscript or in the decision to publish. K.L. Thijssen, I.P. Kema, C. Neri, P.J. Oefner, E.A. Nollen, Delaying aging and
82 C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89

the aging-associated decline in protein homeostasis by inhibition of tryp- Diabetes 51 (6) (2002) 1714e1721.
tophan degradation, Proc. Natl. Acad. Sci. U. S. A. 109 (37) (2012) [48] W.A. Muller, G.R. Faloona, R.H. Unger, The effect of alanine on glucagon
14912e14917. secretion, J. Clin. Investig. 50 (10) (1971) 2215e2218.
[23] J. Mansfeld, N. Urban, S. Priebe, M. Groth, C. Frahm, N. Hartmann, J. Gebauer, [49] A. Freudenberg, K.J. Petzke, S. Klaus, Dietary L-leucine and L-alanine sup-
M. Ravichandran, A. Dommaschk, S. Schmeisser, D. Kuhlow, plementation have similar acute effects in the prevention of high-fat diet-
S. Monajembashi, S. Bremer-Streck, P. Hemmerich, M. Kiehntopf, induced obesity, Amino Acids 44 (2) (2013) 519e528.
N. Zamboni, C. Englert, R. Guthke, C. Kaleta, M. Platzer, J. Suhnel, O.W. Witte, [50] K. Nordlind, U. Ernstrom, V. Mutt, L-alanine–an essential amino acid for
K. Zarse, M. Ristow, Branched-chain amino acid catabolism is a conserved growth of lymphocytes in vitro, Int. Arch. Allergy Appl. Immunol. 59 (2)
regulator of physiological ageing, Nat. Commun. 6 (2015) 10043. (1979) 215e221.
[24] R.A. Miller, D.E. Harrison, C.M. Astle, M.A. Bogue, J. Brind, E. Fernandez, [51] B. Hagglund, G. Sandberg, Effect of L-alanine and some other amino acids on
K. Flurkey, M. Javors, W. Ladiges, C. Leeuwenburgh, F. Macchiarini, J. Nelson, thymocyte proliferation in vivo, Immunobiology 188 (1e2) (1993) 62e69.
A.G. Ryazanov, J. Snyder, T.M. Stearns, D.E. Vaughan, R. Strong, Glycine [52] F.C. Chow, M.I. Dysart, D.W. Hamar, R.H. Udall, Control of oxalate urolithiasis
supplementation extends lifespan of male and female mice, Aging Cell 18 (3) by DL-alanine, Investig. Urol. 13 (2) (1975) 113e116.
(2019), e12953. [53] R.H. Houtkooper, C. Argmann, S.M. Houten, C. Canto, E.H. Jeninga,
[25] J. Brind, V. Malloy, I. Augie, N. Caliendo, J.H. Vogelman, J.A. Zimmerman, P.A. Andreux, C. Thomas, R. Doenlen, K. Schoonjans, J. Auwerx, The metabolic
N. Orentreich, Dietary glycine supplementation mimics lifespan extension by footprint of aging in mice, Sci. Rep. 1 (2011) 134.
dietary methionine restriction in Fisher 344 rats, FASEB J. 25 (1_supplement) [54] R. Uchitomi, Y. Hatazawa, N. Senoo, K. Yoshioka, M. Fujita, T. Shimizu,
(2011), 528.2-528.2. S. Miura, Y. Ono, Y. Kamei, Metabolomic analysis of skeletal muscle in aged
[26] K.M. Hoedjes, M.A. Rodrigues, T. Flatt, Amino acid modulation of lifespan and mice, Sci. Rep. 9 (1) (2019) 10425.
reproduction in Drosophila, Curr. opin. insect. sci. 23 (2017) 118e122. [55] A. Kapranas, C.J. Snart, H. Williams, I.C. Hardy, D.A. Barrett, Metabolomics of
[27] J.C. Jiang, E. Jaruga, M.V. Repnevskaya, S.M. Jazwinski, An intervention aging assessed in individual parasitoid wasps, Sci. Rep. 6 (2016) 34848.
resembling caloric restriction prolongs life span and retards aging in yeast, [56] S.L. Yu, Y.J. An, H.J. Yang, M.S. Kang, H.Y. Kim, H. Wen, X. Jin, H.N. Kwon,
FASEB J. 14 (14) (2000) 2135e2137. K.J. Min, S.K. Lee, S. Park, Alanine-metabolizing enzyme Alt1 is critical in
[28] A.L. Hughes, D.E. Gottschling, An early age increase in vacuolar pH limits determining yeast life span, as revealed by combined metabolomic and ge-
mitochondrial function and lifespan in yeast, Nature 492 (7428) (2012) netic studies, J. Proteome Res. 12 (4) (2013) 1619e1627.
261e265. [57] L. Mouchiroud, L. Molin, P. Kasturi, M.N. Triba, M.E. Dumas, M.C. Wilson,
[29] S.M. Solon-Biet, A.C. McMahon, J.W. Ballard, K. Ruohonen, L.E. Wu, re, L. Se
A.P. Halestrap, D. Roussel, I. Masse, N. Dallie galat, M. Billaud, F. Solari,
V.C. Cogger, A. Warren, X. Huang, N. Pichaud, R.G. Melvin, R. Gokarn, Pyruvate imbalance mediates metabolic reprogramming and mimics lifespan
M. Khalil, N. Turner, G.J. Cooney, D.A. Sinclair, D. Raubenheimer, D.G. Le extension by dietary restriction in Caenorhabditis elegans, Aging Cell 10 (1)
Couteur, S.J. Simpson, The ratio of macronutrients, not caloric intake, dictates (2011) 39e54.
cardiometabolic health, aging, and longevity in ad libitum-fed mice, Cell [58] M. Gad, Anti-aging Effects of L-Arginine, 2010.
Metabol. 19 (3) (2014) 418e430. [59] M. Mazlan, H.S. Hamezah, N.M. Taridi, Y. Jing, P. Liu, H. Zhang, W.Z. Wan
[30] J.R. Speakman, S.E. Mitchell, M. Mazidi, Calories or protein? The effect of Ngah, H.A. Damanhuri, Effects of aging and tocotrienol-rich fraction sup-
dietary restriction on lifespan in rodents is explained by calories alone, Exp. plementation on brain arginine metabolism in rats, Oxid Med Cell Longev
Gerontol. 86 (2016) 28e38. 2017 (2017) 6019796.
[31] M. Kitada, Y. Ogura, I. Monno, D. Koya, The impact of dietary protein intake [60] A. Barbul, S.A. Lazarou, D.T. Efron, H.L. Wasserkrug, G. Efron, Arginine en-
on longevity and metabolic health, EBioMedicine 43 (2019) 632e640. hances wound healing and lymphocyte immune responses in humans,
[32] M.D.W. Piper, G.A. Soultoukis, E. Blanc, A. Mesaros, S.L. Herbert, P. Juricic, Surgery 108 (2) (1990) 331e336, discussion 336-7.
X. He, I. Atanassov, H. Salmonowicz, M. Yang, S.J. Simpson, C. Ribeiro, [61] S.J. Kirk, M. Hurson, M.C. Regan, D.R. Holt, H.L. Wasserkrug, A. Barbul, Argi-
L. Partridge, Matching dietary amino acid balance to the in silico-translated nine stimulates wound healing and immune function in elderly human be-
exome optimizes growth and reproduction without cost to lifespan, Cell ings, Surgery 114 (2) (1993) 155e159, discussion 160.
Metabol. 25 (3) (2017) 610e621. [62] A.G. Ronnenberg, K.L. Gross, W.J. Hartman, S.N. Meydani, R.L. Prior, Dietary
[33] M. Guasch-Ferre, A. Hruby, E. Toledo, C.B. Clish, M.A. Martinez-Gonzalez, arginine supplementation does not enhance lymphocyte proliferation or
J. Salas-Salvado, F.B. Hu, Metabolomics in prediabetes and diabetes: a sys- interleukin-2 production in young and aged rats, J. Nutr. 121 (8) (1991)
tematic review and meta-analysis, Diabetes Care 39 (5) (2016) 833e846. 1270e1278.
[34] J.P. Colombo, H. Cervantes, M. Kokorovic, U. Pfister, R. Perritaz, Effect of [63] R. Geiger, J.C. Rieckmann, T. Wolf, C. Basso, Y. Feng, T. Fuhrer, M. Kogadeeva,
different protein diets on the distribution of amino acids in plasma, liver and P. Picotti, F. Meissner, M. Mann, N. Zamboni, F. Sallusto, A. Lanzavecchia, T. L-
brain in the rat, Ann. Nutr. Metab. 36 (1) (1992) 23e33. Arginine Modulates, Cell metabolism and enhances survival and anti-tumor
[35] S. Ma, A. Upneja, A. Galecki, Y.M. Tsai, C.F. Burant, S. Raskind, Q. Zhang, activity, Cell 167 (3) (2016) 829e842, e13.
Z.D. Zhang, A. Seluanov, V. Gorbunova, C.B. Clish, R.A. Miller, V.N. Gladyshev, [64] A. Chauhan, R.S. More, P.A. Mullins, G. Taylor, C. Petch, P.M. Schofield, Aging-
Cell culture-based profiling across mammals reveals DNA repair and meta- associated endothelial dysfunction in humans is reversed by L-arginine,
bolism as determinants of species longevity, eLife 5 (2016). J. Am. Coll. Cardiol. 28 (7) (1996) 1796e1804.
[36] S. Katayama, Y. Mine, Antioxidative activity of amino acids on tissue [65] M.K. Angele, S.M. Nitsch, R.A. Hatz, P. Angele, T. Hernandez-Richter,
oxidative stress in human intestinal epithelial cell model, J. Agric. Food M.W. Wichmann, I.H. Chaudry, F.W. Schildberg, L-arginine, A unique amino
Chem. 55 (21) (2007) 8458e8464. acid for improving depressed wound immune function following hemor-
[37] L. Shan, B. Wang, G. Gao, W. Cao, Y. Zhang, l-Arginine supplementation rhage, European surgical research. Europaische chirurgische Forschung,
improves antioxidant defenses through l-arginine/nitric oxide pathways in Rech. chirurgicales Euro. 34 (1e2) (2002) 53e60.
exercised rats, J. Appl. Physiol. 115 (8) (2013) 1146e1155. [66] M.K. Angele, N. Smail, A. Ayala, W.G. Cioffi, K.I. Bland, I.H. Chaudry, L-argi-
[38] J. Duan, J. Yin, W. Ren, T. Liu, Z. Cui, X. Huang, L. Wu, S.W. Kim, G. Liu, X. Wu, nine, A unique amino acid for restoring the depressed macrophage functions
G. Wu, T. Li, Y. Yin, Dietary supplementation with l-glutamate and l-aspar- after trauma-hemorrhage, J. Trauma 46 (1) (1999) 34e41.
tate alleviates oxidative stress in weaned piglets challenged with hydrogen [67] J.F. Reckelhoff, J.A. Kellum Jr., L.C. Racusen, D.A. Hildebrandt, Long-term di-
peroxide, Amino Acids 48 (1) (2016) 53e64. etary supplementation with L-arginine prevents age-related reduction in
[39] Y. Jie, L. Tiejun, Y. Yulong, Methionine and antioxidant potential, J. Antioxid. renal function, Am. J. Physiol. 272 (6 Pt 2) (1997) R1768eR1774.
Activ. 1 (2) (2016) 17e22. [68] F. Madeo, D. Carmona-Gutierrez, O. Kepp, G. Kroemer, Spermidine delays
[40] J.-H. Kim, H.-J. Jang, W.-Y. Cho, S.-J. Yeon, C.-H. Lee, In vitro antioxidant ac- aging in humans, Aging (Albany NY) 10 (8) (2018) 2209e2211.
tions of sulfur-containing amino acids, Arabian J. Chem. (2018). [69] J.A. Rollins, D. Shaffer, S.S. Snow, P. Kapahi, A.N. Rogers, Dietary restriction
[41] N. Krishnan, M.B. Dickman, D.F. Becker, Proline modulates the intracellular induces posttranscriptional regulation of longevity genes, Life science alli-
redox environment and protects mammalian cells against oxidative stress, ance 2 (4) (2019).
Free Radic. Biol. Med. 44 (4) (2008) 671e681. [70] R.M. Palmer, D.S. Ashton, S. Moncada, Vascular endothelial cells synthesize
[42] X. Liang, L. Zhang, S.K. Natarajan, D.F. Becker, Proline mechanisms of stress nitric oxide from L-arginine, Nature 333 (6174) (1988) 664e666.
survival, Antioxidants Redox Signal. 19 (9) (2013) 998e1011. [71] A.L. Sverdlov, D.T.M. Ngo, W.P.A. Chan, Y.Y. Chirkov, J.D. Horowitz, Aging of
[43] I. Gulcin, Comparison of in vitro antioxidant and antiradical activities of L- the nitric oxide system: are we as old as our NO? J Am Heart Assoc 3 (4)
tyrosine and L-Dopa, Amino Acids 32 (3) (2007) 431e438. (2014) e000973.
[44] F.W. van Overveld, G.R. Haenen, J. Rhemrev, J.P. Vermeiden, A. Bast, Tyrosine [72] Y. Tsugawa, H. Handa, T. Imai, Arginine induces IGF-1 secretion from the
as important contributor to the antioxidant capacity of seminal plasma, endoplasmic reticulum, Biochem. Biophys. Res. Commun. 514 (4) (2019)
Chem. Biol. Interact. 127 (2) (2000) 151e161. 1128e1132.
[45] N. Kraus-Friedmann, Hormonal regulation of hepatic gluconeogenesis, [73] J. Alba-Roth, O.A. Muller, J. Schopohl, K. von Werder, Arginine stimulates
Physiol. Rev. 64 (1) (1984) 170e259. growth hormone secretion by suppressing endogenous somatostatin secre-
[46] T.W. Chang, A.L. Goldberg, The origin of alanine produced in skeletal muscle, tion, J. Clin. Endocrinol. Metab. 67 (6) (1988) 1186e1189.
J. Biol. Chem. 253 (10) (1978) 3677e3684. [74] M.H. Aguiar-Oliveira, A. Bartke, Growth hormone deficiency: health and
[47] L. Brennan, A. Shine, C. Hewage, J.P. Malthouse, K.M. Brindle, longevity, Endocr. Rev. 40 (2) (2019) 575e601.
N. McClenaghan, P.R. Flatt, P. Newsholme, A nuclear magnetic resonance- [75] M. Holzenberger, J. Dupont, B. Ducos, P. Leneuve, A. Geloen, P.C. Even,
based demonstration of substantial oxidative L-alanine metabolism and L- P. Cervera, Y. Le Bouc, IGF-1 receptor regulates lifespan and resistance to
alanine-enhanced glucose metabolism in a clonal pancreatic beta-cell line: oxidative stress in mice, Nature 421 (6919) (2003) 182e187.
metabolism of L-alanine is important to the regulation of insulin secretion, [76] P.A. Wright, Nitrogen excretion: three end products, many physiological
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 83

roles, J. Exp. Biol. 198 (Pt 2) (1995) 273e281. production in neuronal cells overexpressing alpha-synuclein, Neuro-
[77] J. Moretto, C. Girard, C. Demougeot, The role of arginase in aging: a sys- Molecular Med. 19 (2e3) (2017) 322e344.
tematic review, Exp. Gerontol. 116 (2019) 54e73. [101] H. Ni, L. Lu, J. Deng, W. Fan, T. Li, J. Yao, Effects of glutamate and aspartate on
[78] B. Carroll, D. Maetzel, O.D. Maddocks, G. Otten, M. Ratcliff, G.R. Smith, serum antioxidative enzyme, sex hormones, and genital inflammation in
E.A. Dunlop, J.F. Passos, O.R. Davies, R. Jaenisch, A.R. Tee, S. Sarkar, boars challenged with hydrogen peroxide, Mediat. Inflamm. (2016) 10.
V.I. Korolchuk, Control of TSC2-Rheb signaling axis by arginine regulates [102] J. Yin, M. Liu, W. Ren, J. Duan, G. Yang, Y. Zhao, R. Fang, L. Chen, T. Li, Y. Yin,
mTORC1 activity, eLife 5 (2016). Effects of dietary supplementation with glutamate and aspartate on diquat-
[79] T. Weichhart, mTOR as regulator of lifespan, Aging, and Cellular Senescence: induced oxidative stress in piglets, PLoS One 10 (4) (2015) e0122893.
A Mini-Review, Gerontology 64 (2) (2018) 127e134. [103] S.I. Oh, M.S. Lee, C.I. Kim, K.Y. Song, S.C. Park, Aspartate modulates the
[80] M. Abu-Remaileh, G.A. Wyant, C. Kim, N.N. Laqtom, M. Abbasi, S.H. Chan, ethanol-induced oxidative stress and glutathione utilizing enzymes in rat
E. Freinkman, D.M. Sabatini, Lysosomal metabolomics reveals V-ATPase- and testes, Exp. Mol. Med. 34 (1) (2002) 47e52.
mTOR-dependent regulation of amino acid efflux from lysosomes, Science [104] B.E. Herring, K. Silm, R.H. Edwards, R.A. Nicoll, Is aspartate an excitatory
(New York, N.Y.) 358 (6364) (2017) 807e813. neurotransmitter? J. Neurosci. 35 (28) (2015) 10168e10171.
[81] L. Chantranupong, S.M. Scaria, R.A. Saxton, M.P. Gygi, K. Shen, G.A. Wyant, [105] R. Hu, D. Huang, J. Tong, Q. Liao, Z. Hu, W. Ouyang, Aspartic acid in the
T. Wang, J.W. Harper, S.P. Gygi, D.M. Sabatini, The CASTOR proteins are hippocampus: a biomarker for postoperative cognitive dysfunction, Neural
arginine sensors for the mTORC1 pathway, Cell 165 (1) (2016) 153e164. regeneration research 9 (2) (2014) 143e152.
[82] J.M. Hoffman, Q.A. Soltow, S. Li, A. Sidik, D.P. Jones, D.E. Promislow, Effects of [106] Z. Wu, L. Song, S.Q. Liu, D. Huang, Independent and additive effects of glu-
age, sex, and genotype on high-sensitivity metabolomic profiles in the fruit tamic acid and methionine on yeast longevity, PLoS One 8 (11) (2013),
fly, Drosophila melanogaster, Aging Cell 13 (4) (2014) 596e604. e79319.
[83] C. Beaupere, B.M. Wasko, J. Lorusso, B.K. Kennedy, M. Kaeberlein, [107] K. Mohammad, P. Dakik, Y. Medkour, M. McAuley, D. Mitrofanova,
V.M. Labunskyy, CAN1 arginine permease deficiency extends yeast replica- V.I. Titorenko, Some metabolites act as second messengers in yeast chro-
tive lifespan via translational activation of stress response genes, Cell Rep. 18 nological aging, Int. J. Mol. Sci. 19 (3) (2018).
(8) (2017) 1884e1892. [108] R. Yoshida, T. Tamura, C. Takaoka, K. Harada, A. Kobayashi, Y. Mukai,
[84] N.N. Pavlova, S. Hui, J.M. Ghergurovich, J. Fan, A.M. Intlekofer, R.M. White, E. Fukusaki, Metabolomics-based systematic prediction of yeast lifespan and
J.D. Rabinowitz, C.B. Thompson, J. Zhang, As extracellular glutamine levels its application for semi-rational screening of ageing-related mutants, Aging
decline, asparagine becomes an essential amino acid, Cell Metabol. 27 (2) Cell 9 (4) (2010) 616e625.
(2018) 428e438, e5. [109] D.P. Jones, V.C. Mody Jr., J.L. Carlson, M.J. Lynn, P. Sternberg Jr., Redox analysis
[85] R. Calvani, A. Picca, F. Marini, A. Biancolillo, J. Gervasoni, S. Persichilli, of human plasma allows separation of pro-oxidant events of aging from
A. Primiano, H.J. Coelho-Junior, M. Bossola, A. Urbani, F. Landi, R. Bernabei, decline in antioxidant defenses, Free Radic. Biol. Med. 33 (9) (2002)
E. Marzetti, A distinct pattern of circulating amino acids characterizes older 1290e1300.
persons with physical frailty and sarcopenia: results from the BIOSPHERE [110] H.T. Pitkanen, S.S. Oja, K. Kemppainen, J.M. Seppa, A.A. Mero, Serum amino
study, Nutrients 10 (11) (2018). acid concentrations in aging men and women, Amino Acids 24 (4) (2003)
[86] A.S. Krall, S. Xu, T.G. Graeber, D. Braas, H.R. Christofk, Asparagine promotes 413e421.
cancer cell proliferation through use as an amino acid exchange factor, Nat. [111] S.S. Iyer, C.J. Accardi, T.R. Ziegler, R.A. Blanco, J.D. Ritzenthaler, M. Rojas,
Commun. 7 (2016) 11457. J. Roman, D.P. Jones, Cysteine redox potential determines pro-inflammatory
[87] S.R.V. Knott, E. Wagenblast, S. Khan, S.Y. Kim, M. Soto, M. Wagner, IL-1beta levels, PLoS One 4 (3) (2009) e5017.
M.O. Turgeon, L. Fish, N. Erard, A.L. Gable, A.R. Maceli, S. Dickopf, [112] T.N. Akbaraly, M. Hamer, J.E. Ferrie, G. Lowe, G.D. Batty, G. Hagger-Johnson,
E.K. Papachristou, C.S. D'Santos, L.A. Carey, J.E. Wilkinson, J.C. Harrell, A. Singh-Manoux, M.J. Shipley, M. Kivim€ aki, Chronic inflammation as a
C.M. Perou, H. Goodarzi, G. Poulogiannis, G.J. Hannon, Asparagine bioavail- determinant of future aging phenotypes, CMAJ (Can. Med. Assoc. J.) 185 (16)
ability governs metastasis in a model of breast cancer, Nature 554 (7692) (2013) E763eE770.
(2018) 378e381. [113] Y. Dong, S. Sameni, M.A. Digman, G.J. Brewer, Reversibility of age-related
[88] H. Huang, S. Vandekeere, J. Kalucka, L. Bierhansl, A. Zecchin, U. Bruning, oxidized free NADH redox states in alzheimer's disease neurons by
A. Visnagri, N. Yuldasheva, J. Goveia, B. Cruys, K. Brepoels, S. Wyns, imposed external Cys/CySS redox shifts, Sci. Rep. 9 (1) (2019) 11274.
S. Rayport, B. Ghesquiere, S. Vinckier, L. Schoonjans, R. Cubbon, [114] B. Moosmann, C. Behl, Mitochondrially encoded cysteine predicts animal
M. Dewerchin, G. Eelen, P. Carmeliet, Role of glutamine and interlinked lifespan, Aging Cell 7 (1) (2008) 32e46.
asparagine metabolism in vessel formation, EMBO J. 36 (16) (2017) [115] S.K. Jain, L-Cysteine supplementation as an adjuvant therapy for type-2
2334e2352. diabetes, Can. J. Physiol. Pharmacol. 90 (8) (2012) 1061e1064.
[89] M. Luo, M. Brooks, M.S. Wicha, Asparagine and glutamine: Co-conspirators [116] H. Okawa, T. Morita, K. Sugiyama, Cysteine supplementation decreases
fueling metastasis, Cell Metabol. 27 (5) (2018) 947e949. plasma homocysteine concentration in rats fed on a low-casein diet in rats,
[90] S. Chen, Y. Liu, X. Wang, H. Wang, S. Li, H. Shi, H. Zhu, J. Zhang, D. Pi, C.A. Hu, Biosci. Biotechnol. Biochem. 71 (1) (2007) 91e97.
X. Lin, J. Odle, Asparagine improves intestinal integrity, inhibits TLR4 and [117] T. Huang, J. Ren, J. Huang, D. Li, Association of homocysteine with type 2
NOD signaling, and differently regulates p38 and ERK1/2 signaling in diabetes: a meta-analysis implementing Mendelian randomization
weanling piglets after LPS challenge, Innate Immun. 22 (8) (2016) 577e587. approach, BMC Genomics 14 (2013), 867-867.
[91] A.H. Lancha Jr., M.B. Recco, D.S. Abdalla, R. Curi, Effect of aspartate, aspara- [118] M.T. Heafield, S. Fearn, G.B. Steventon, R.H. Waring, A.C. Williams,
gine, and carnitine supplementation in the diet on metabolism of skeletal S.G. Sturman, Plasma cysteine and sulphate levels in patients with motor
muscle during a moderate exercise, Physiol. Behav. 57 (2) (1995) 367e371. neurone, Parkinson's and Alzheimer's disease, Neurosci. Lett. 110 (1e2)
[92] M.L. Marquezi, H.A. Roschel, A. dos Santa Costa, L.A. Sawada, A.H. Lancha Jr., (1990) 216e220.
Effect of aspartate and asparagine supplementation on fatigue determinants [119] H. Bradley, A. Gough, R.S. Sokhi, A. Hassell, R. Waring, P. Emery, Sulfate
in intense exercise, Int. J. Sport Nutr. Exerc. Metab. 13 (1) (2003) 65e75. metabolism is abnormal in patients with rheumatoid arthritis. Confirmation
[93] R.W. Powers 3rd, M. Kaeberlein, S.D. Caldwell, B.K. Kennedy, S. Fields, by in vivo biochemical findings, J. Rheumatol. 21 (7) (1994) 1192e1196.
Extension of chronological life span in yeast by decreased TOR pathway [120] C. Gordon, H. Bradley, R.H. Waring, P. Emery, Abnormal sulphur oxidation in
signaling, Genes Dev. 20 (2) (2006) 174e184. systemic lupus erythematosus, Lancet 339 (8784) (1992) 25e26.
[94] M. Okada, S. Kusunoki, Y. Ishibashi, K. Kito, Proteomics analysis for asym- [121] A. Bhat, R. Chakraborty, K. Adlakha, G. Agam, K. Chakraborty, S. Sengupta,
metric inheritance of preexisting proteins between mother and daughter Ncl1-mediated metabolic rewiring critical during metabolic stress, Life sci-
cells in budding yeast, Genes Cells 22 (6) (2017) 591e601. ence alliance 2 (4) (2019).
[95] Y. Kamei, Y. Tamada, Y. Nakayama, E. Fukusaki, Y. Mukai, Changes in tran- [122] R.F. Grimble, A.A. Jackson, C. Persaud, M.J. Wride, F. Delers, R. Engler,
scription and metabolism during the early stage of replicative cellular Cysteine and glycine supplementation modulate the metabolic response to
senescence in budding yeast, J. Biol. Chem. 289 (46) (2014) 32081e32093. tumor necrosis factor alpha in rats fed a low protein diet, J. Nutr. 122 (11)
[96] E. Easlon, F. Tsang, C. Skinner, C. Wang, S.J. Lin, The malate-aspartate NADH (1992) 2066e2073.
shuttle components are novel metabolic longevity regulators required for [123] Y.M. Go, D.P. Jones, Redox compartmentalization in eukaryotic cells, Bio-
calorie restriction-mediated life span extension in yeast, Genes Dev. 22 (7) chim. Biophys. Acta 1780 (11) (2008) 1273e1290.
(2008) 931e944. [124] J. Allen, R.D. Bradley, Effects of oral glutathione supplementation on systemic
[97] A. Navarro, A. Boveris, The mitochondrial energy transduction system and oxidative stress biomarkers in human volunteers, J. Altern. Complement.
the aging process, Am. J. Physiol. Cell Physiol. 292 (2) (2007) C670eC686. Med. 17 (9) (2011) 827e833.
[98] K. Birsoy, T. Wang, W.W. Chen, E. Freinkman, M. Abu-Remaileh, [125] R.V. Sekhar, S.G. Patel, A.P. Guthikonda, M. Reid, A. Balasubramanyam,
D.M. Sabatini, An essential role of the mitochondrial electron transport chain G.E. Taffet, F. Jahoor, Deficient synthesis of glutathione underlies oxidative
in cell proliferation is to enable aspartate synthesis, Cell 162 (3) (2015) stress in aging and can be corrected by dietary cysteine and glycine sup-
540e551. plementation, Am. J. Clin. Nutr. 94 (3) (2011) 847e853.
[99] I.B. Afanas'ev, T.B. Suslova, Z.P. Cheremisina, N.E. Abramova, L.G. Korkina, [126] K. Flurkey, C.M. Astle, D.E. Harrison, Life extension by diet restriction and N-
Study of antioxidant properties of metal aspartates, Analyst 120 (3) (1995) acetyl-L-cysteine in genetically heterogeneous mice, J Gerontol A Biol Sci
859e862. Med Sci 65 (12) (2010) 1275e1284.
[100] V. Delic, J.W.D. Griffin, S. Zivkovic, Y. Zhang, T.A. Phan, H. Gong, D. Chaput, [127] M.V. Shaposhnikov, N.V. Zemskaya, L.A. Koval, E.V. Schegoleva,
C. Reynes, V.B. Dinh, J. Cruz, E. Cvitkovic, D. Placides, E. Frederic, H. Mirzaei, A. Zhavoronkov, A.A. Moskalev, Effects of N-acetyl-L-cysteine on lifespan,
S.M. Stevens Jr., U. Jinwal, D.C. Lee, P.C. Bradshaw, Individual amino acid locomotor activity and stress-resistance of 3 Drosophila species with
supplementation can improve energy metabolism and decrease ROS different lifespans, Aging 10 (9) (2018) 2428e2458.
84 C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89

[128] S.I. Oh, J.K. Park, S.K. Park, Lifespan extension and increased resistance to [154] J. Albrecht, U. Sonnewald, H.S. Waagepetersen, A. Schousboe, Glutamine in
environmental stressors by N-acetyl-L-cysteine in Caenorhabditis elegans, the central nervous system: function and dysfunction, Front. Biosci. : J. Vis.
Clinics (Sao Paulo, Brazil) 70 (5) (2015) 380e386. Lit. 12 (2007) 332e343.
[129] A. Shibamura, T. Ikeda, Y. Nishikawa, A method for oral administration of [155] R. Gorovits, N. Avidan, N. Avisar, I. Shaked, L. Vardimon, Glutamine synthe-
hydrophilic substances to Caenorhabditis elegans: effects of oral supple- tase protects against neuronal degeneration in injured retinal tissue, Proc.
mentation with antioxidants on the nematode lifespan, Mech. Ageing Dev. Natl. Acad. Sci. U.S.A. 94 (13) (1997) 7024e7029.
130 (9) (2009) 652e655. [156] H. Tumani, G. Shen, J.B. Peter, W. Bruck, Glutamine synthetase in cerebro-
[130] H. Zhang, P. Limphong, J. Pieper, Q. Liu, C.K. Rodesch, E. Christians, spinal fluid, serum, and brain: a diagnostic marker for Alzheimer disease?
I.J. Benjamin, Glutathione-dependent reductive stress triggers mitochondrial Arch. Neurol. 56 (10) (1999) 1241e1246.
oxidation and cytotoxicity, FASEB J. : off. publi. Fed. Am. Soc. Exp. Biol. 26 (4) [157] J. Chen, K. Herrup, Glutamine acts as a neuroprotectant against DNA damage,
(2012) 1442e1451. beta-amyloid and H2O2-induced stress, PLoS One 7 (3) (2012), e33177.
[131] V. Mokhtari, P. Afsharian, M. Shahhoseini, S.M. Kalantar, A. Moini, A review [158] J.W. Griffin, P.C. Bradshaw, Amino acid catabolism in alzheimer's disease
on various uses of N-acetyl cysteine, Cell J 19 (1) (2017) 11e17. brain: friend or foe? Oxid Med Cell Longev 2017 (2017) 5472792.
[132] D. Schubert, D. Piasecki, Oxidative glutamate toxicity can be a component of [159] E. Socha, M. Koba, P. Koslinski, Amino acid profiling as a method of discov-
the excitotoxicity cascade, J. Neurosci. : Off. J. Soc. Neurosci. 21 (19) (2001) ering biomarkers for diagnosis of neurodegenerative diseases, Amino Acids
7455e7462. 51 (3) (2019) 367e371.
[133] L.I. Bruijn, T.M. Miller, D.W. Cleveland, Unraveling the mechanisms involved [160] J.L. Crespo, T. Powers, B. Fowler, M.N. Hall, The TOR-controlled transcription
in motor neuron degeneration in ALS, Annu. Rev. Neurosci. 27 (2004) activators GLN3, RTG1, and RTG3 are regulated in response to intracellular
723e749. levels of glutamine, Proc. Natl. Acad. Sci. U. S. A. 99 (10) (2002) 6784e6789.
[134] A. Marsman, R.C.W. Mandl, M.P. van den Heuvel, V.O. Boer, J.P. Wijnen, [161] X. Li, M.P. Snyder, Yeast longevity promoted by reversing aging-associated
D.W.J. Klomp, P.R. Luijten, H.P. Hilleke E, Glutamate changes in healthy decline in heavy isotope content, NPJ Aging Mech. Dis. 2 (2016) 16004.
young adulthood, Eur. Neuropsychopharmacol. 23 (11) (2013) 1484e1490. [162] M.A. Razak, P.S. Begum, B. Viswanath, S. Rajagopal, Multifarious beneficial
[135] M.D. Sweeney, Z. Zhao, A. Montagne, A.R. Nelson, B.V. Zlokovic, Blood-brain effect of nonessential amino acid, Glycine: a review, Oxidative Med. Cell.
barrier: from physiology to disease and back, Physiol. Rev. 99 (1) (2019) Longev. 2017 8 (2017).
21e78. [163] J. Brind, V. Malloy, I. Augie, N. Caliendo, J.H. Vogelman, J.A. Zimmerman,
[136] J. Castillo, M.I. Loza, D. Mirelman, J. Brea, M. Blanco, T. Sobrino, F. Campos, N. Orentreich, Dietary glycine supplementation mimics lifespan extension by
A novel mechanism of neuroprotection: blood glutamate grabber, J. Cereb. dietary methionine restriction in Fisher 344 rats, FASEB J. 25 (2011).
Blood Flow Metab. 36 (2) (2016) 292e301. [164] N. Ron-Harel, G. Notarangelo, J.M. Ghergurovich, J.A. Paulo, P.T. Sage,
[137] A. Zhumadilov, M. Boyko, S.E. Gruenbaum, E. Brotfain, F. Bilotta, A. Zlotnik, D. Santos, F.K. Satterstrom, S.P. Gygi, J.D. Rabinowitz, A.H. Sharpe,
Extracorporeal methods of blood glutamate scavenging: a novel therapeutic M.C. Haigis, Defective respiration and one-carbon metabolism contribute to
modality, Expert Rev. Neurother. 15 (5) (2015) 501e508. impaired naive T cell activation in aged mice, Proc. Natl. Acad. Sci. U. S. A. 115
[138] Y. Daikhin, M. Yudkoff, Compartmentation of brain glutamate metabolism in (52) (2018) 13347e13352.
neurons and glia, J. Nutr. 130 (4S Suppl) (2000), 1026s-31s. [165] Z. Wang, J. Zhang, L. Chen, J. Li, H. Zhang, X. Guo, Glycine suppresses AGE/
[139] A.R. Jayakumar, M.D. Norenberg, Glutamine synthetase: role in neurological RAGE signaling pathway and subsequent oxidative stress by restoring Glo1
disorders, Adv. Neurobiol. 13 (2016) 327e350. function in the aorta of diabetic rats and in HUVECs, Oxid Med Cell Longev
[140] R.M. Chin, X. Fu, M.Y. Pai, L. Vergnes, H. Hwang, G. Deng, S. Diep, B. Lomenick, 2019 (2019) 4628962.
V.S. Meli, G.C. Monsalve, E. Hu, S.A. Whelan, J.X. Wang, G. Jung, G.M. Solis, [166] O. Hashizume, S. Ohnishi, T. Mito, A. Shimizu, K. Ishikawa, K. Nakada,
F. Fazlollahi, C. Kaweeteerawat, A. Quach, M. Nili, A.S. Krall, H.A. Godwin, M. Soda, H. Mano, S. Togayachi, H. Miyoshi, K. Okita, J. Hayashi, Epigenetic
H.R. Chang, K.F. Faull, F. Guo, M. Jiang, S.A. Trauger, A. Saghatelian, D. Braas, regulation of the nuclear-coded GCAT and SHMT2 genes confers human age-
H.R. Christofk, C.F. Clarke, M.A. Teitell, M. Petrascheck, K. Reue, M.E. Jung, associated mitochondrial respiration defects, Sci. Rep. 5 (2015) 10434.
A.R. Frand, J. Huang, The metabolite alpha-ketoglutarate extends lifespan by [167] P. Lunetti, F. Damiano, G. De Benedetto, L. Siculella, A. Pennetta, L. Muto,
inhibiting ATP synthase and TOR, Nature 510 (7505) (2014) 397e401. E. Paradies, C.M. Marobbio, V. Dolce, L. Capobianco, Characterization of hu-
[141] K.N. Lewis, N.D. Rubinstein, R. Buffenstein, A window into extreme man and yeast mitochondrial Glycine carriers with implications for heme
longevity; the circulating metabolomic signature of the naked mole-rat, a biosynthesis and anemia, J. Biol. Chem. 291 (38) (2016) 19746e19759.
mammal that shows negligible senescence, GeroScience 40 (2) (2018) [168] M. Gersovitz, D. Bier, D. Matthews, J. Udall, H.N. Munro, V.R. Young, Dynamic
105e121. aspects of whole body glycine metabolism: influence of protein intake in
[142] M. Viltard, S. Durand, M. Perez-Lanzon, F. Aprahamian, D. Lefevre, C. Leroy, young adult and elderly males, Metabolism 29 (11) (1980) 1087e1094.
F. Madeo, G. Kroemer, G. Friedlander, The metabolomic signature of extreme [169] N.G. Bowery, T.G. Smart, GABA and glycine as neurotransmitters: a brief
longevity: naked mole rats versus mice, Aging (Albany NY) 11 (14) (2019) history, Br. J. Pharmacol. 147 (Suppl 1) (2006) S109eS119.
4783e4800. [170] N. Kawai, N. Sakai, M. Okuro, S. Karakawa, Y. Tsuneyoshi, N. Kawasaki,
[143] M. Muldowney, The Effects of Glutamate Transporter Deletions on Lifespan T. Takeda, M. Bannai, S. Nishino, The sleep-promoting and hypothermic ef-
in Caenorhabditis elegans, Honors Thesis Collection Wellesley College, 2018. fects of glycine are mediated by NMDA receptors in the suprachiasmatic
[144] D. Meynial-Denis, Glutamine metabolism in advanced age, Nutr. Rev. 74 (4) nucleus, Neuropsychopharmacol.: off. publi. Am. Coll. Neuro-
(2016) 225e236. psychopharmacol. 40 (6) (2015) 1405e1416.
[145] M. Yuneva, N. Zamboni, P. Oefner, R. Sachidanandam, Y. Lazebnik, Deficiency [171] E.D. Picanco, F. Lopes-Paulo, R.G. Marques, C.F. Diestel, C.E.R. Caetano,
in glutamine but not glucose induces MYC-dependent apoptosis in human M.V.M. de Souza, G.M. Moscoso, H.M.F. Pazos, L-arginine and glycine sup-
cells, J. Cell Biol. 178 (1) (2007) 93e105. plementation in the repair of the irradiated colonic wall of rats, Int. J.
[146] E.C. Opara, A. Petro, A. Tevrizian, M.N. Feinglos, R.S. Surwit, L-glutamine Colorectal Dis. 26 (5) (2011) 561e568.
supplementation of a high fat diet reduces body weight and attenuates [172] C. Baylis, M. Fredericks, C. Wilson, K. Munger, R. Collins, Renal vasodilatory
hyperglycemia and hyperinsulinemia in C57BL/6J mice, J. Nutr. 126 (1) response to intravenous glycine in the aging rat kidney, Am. J. Kidney Dis. :
(1996) 273e279. Off. J. Natl. Kidney Found 15 (3) (1990) 244e251.
[147] M. Scalise, L. Pochini, M. Galluccio, C. Indiveri, Glutamine transport. From [173] S.H. Burstein, N-acyl amino acids (elmiric acids): endogenous signaling
energy supply to sensing and beyond, Biochim. Biophys. Acta 1857 (8) molecules with therapeutic potential, Mol. Pharmacol. 93 (3) (2018)
(2016) 1147e1157. 228e238.
[148] L.M. Castell, E.A. Newsholme, The effects of oral glutamine supplementation [174] D.P. Waluk, F. Sucharski, L. Sipos, J. Silberring, M.C. Hunt, Reversible lysine
on athletes after prolonged, exhaustive exercise, Nutrition 13 (7e8) (1997) acetylation regulates activity of human glycine N-acyltransferase-like 2
738e742. (hGLYATL2): implications for production of glycine-conjugated signaling
[149] C. Brasse-Lagnel, A. Lavoinne, A. Husson, Control of mammalian gene molecules, J. Biol. Chem. 287 (20) (2012) 16158e16167.
expression by amino acids, especially glutamine, FEBS J. 276 (7) (2009) [175] B.D. Lemire, M. Behrendt, A. DeCorby, D. Gaskova, C. elegans longevity
1826e1844. pathways converge to decrease mitochondrial membrane potential, Mech.
[150] M.H. Pai, C.S. Lei, S.T. Su, S.L. Yeh, Y.C. Hou, Effects of dietary glutamine Ageing Dev. 130 (7) (2009) 461e465.
supplementation on immune cell polarization and muscle regeneration in [176] V.I. Padalko, Uncoupler of oxidative phosphorylation prolongs the lifespan of
diabetic mice with limb ischemia, Eur. J. Nutr. (2019). Drosophila, Biochemistry, Biokhimiia 70 (9) (2005) 986e989.
[151] J.S. Leite, R. Raizel, T.M. Hypolito, T.D. Rosa, V.F. Cruzat, J. Tirapegui, l- [177] M. Ost, S. Keipert, E.M. van Schothorst, V. Donner, I. van der Stelt, A.P. Kipp,
glutamine and l-alanine supplementation increase glutamine-glutathione K.J. Petzke, M. Jove, R. Pamplona, M. Portero-Otin, J. Keijer, S. Klaus, Muscle
axis and muscle HSP-27 in rats trained using a progressive high-intensity mitohormesis promotes cellular survival via serine/glycine pathway flux,
resistance exercise, Appl. physiol. nutr. metabol. ¼ Physiologie appliquee, FASEB J. 29 (4) (2015) 1314e1328.
nutrition et metabolisme 41 (8) (2016) 842e849. [178] R.W. Hanson, P. Hakimi, Born to run; the story of the PEPCK-Cmus mouse,
[152] R. Raizel, J.S. Leite, T.M. Hypolito, A.Y. Coqueiro, P. Newsholme, V.F. Cruzat, Biochimie 90 (6) (2008) 838e842.
J. Tirapegui, Determination of the anti-inflammatory and cytoprotective ef- [179] F. Obata, M. Miura, Enhancing S-adenosyl-methionine catabolism extends
fects of l-glutamine and l-alanine, or dipeptide, supplementation in rats Drosophila lifespan, Nat. Commun. 6 (2015), 8332-8332.
submitted to resistance exercise, Br. J. Nutr. 116 (3) (2016) 470e479. [180] R.O. Walters, E. Arias, A. Diaz, E.S. Burgos, F. Guan, S. Tiano, K. Mao,
[153] M. Mignon, A.M. Beaufrere, L. Combaret, D. Meynial-Denis, Does long-term C.L. Green, Y. Qiu, H. Shah, D. Wang, A.D. Hudgins, T. Tabrizian, V. Tosti,
intermittent treatment with glutamine improve the well-being of fed and D. Shechter, L. Fontana, I.J. Kurland, N. Barzilai, A.M. Cuervo,
fasted very old rats? JPEN - J. Parenter. Enter. Nutr. 31 (6) (2007) 456e462. D.E.L. Promislow, D.M. Huffman, Sarcosine is uniquely modulated by aging
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 85

and dietary restriction in rodents and humans, Cell Rep. 25 (3) (2018) S. Schroder, I. Treise, R. Bekeredjian, D.H. Busch, J. Graw, G. Ehninger,
663e676, e6. M. Klingenspor, T. Klopstock, M. Ollert, M. Sandholzer, C. Schmidt-Weber,
[181] J. Hastings, A. Mains, B. Virk, N. Rodriguez, S. Murdoch, J. Pearce, M. Weiergraber, E. Wolf, W. Wurst, A. Zimmer, V. Gailus-Durner, H. Fuchs,
S. Bergmann, N. Le Novere, O. Casanueva, Multi-omics and genome-scale M. Hrabe de Angelis, D. Ehninger, Every-other-day feeding extends lifespan
modeling reveal a metabolic shift during C. elegans aging, Front. mol. bio- but fails to delay many symptoms of aging in mice, Nat. Commun. 8 (1)
sci. 6 (2019) 2. (2017) 155.
[182] Q.L. Wan, X. Shi, J. Liu, A.J. Ding, Y.Z. Pu, Z. Li, G.S. Wu, H.R. Luo, Metabolomic [204] T. Teruya, R. Chaleckis, J. Takada, M. Yanagida, H. Kondoh, Diverse metabolic
signature associated with reproduction-regulated aging in Caenorhabditis reactions activated during 58-hr fasting are revealed by non-targeted
elegans, Aging (Albany NY) 9 (2) (2017) 447e474. metabolomic analysis of human blood, Sci. Rep. 9 (1) (2019), 854-854.
[183] A.W. Gao, I.A. Chatzispyrou, R. Kamble, Y.J. Liu, K. Herzog, R.L. Smith, H. van [205] U.V. Wesley, V.J. Bhute, J.F. Hatcher, S.P. Palecek, R.J. Dempsey, Local and
Lenthe, M.A.T. Vervaart, A. van Cruchten, A.C. Luyf, A. van Kampen, M.L. Pras- systemic metabolic alterations in brain, plasma, and liver of rats in response
Raves, F.M. Vaz, R.H. Houtkooper, A sensitive mass spectrometry platform to aging and ischemic stroke, as detected by nuclear magnetic resonance
identifies metabolic changes of life history traits in C. elegans, Sci. Rep. 7 (1) (NMR) spectroscopy, Neurochem. Int. 127 (2019) 113e124.
(2017) 2408. [206] Z. Arany, M. Neinast, Branched chain amino acids in metabolic disease, Curr.
[184] N. Copes, C. Edwards, D. Chaput, M. Saifee, I. Barjuca, D. Nelson, A. Paraggio, Diabetes Rep. 18 (10) (2018) 76.
P. Saad, D. Lipps, S.M. Stevens Jr., P.C. Bradshaw, Metabolome and proteome [207] M. Doi, I. Yamaoka, T. Fukunaga, M. Nakayama, Isoleucine, a potent plasma
changes with aging in Caenorhabditis elegans, Exp. Gerontol. 72 (2015) glucose-lowering amino acid, stimulates glucose uptake in C2C12 myotubes,
67e84. Biochem. Biophys. Res. Commun. 312 (4) (2003) 1111e1117.
[185] C. Pontoizeau, L. Mouchiroud, L. Molin, A. Mergoud-dit-Lamarche, [208] O. Ikehara, N. Kawasaki, K. Maezono, M. Komatsu, A. Konishia, Acute and
N. Dalliere, P. Toulhoat, B. Elena-Herrmann, F. Solari, Metabolomics analysis chronic treatment of L-isoleucine ameliorates glucose metabolism in
uncovers that dietary restriction buffers metabolic changes associated with glucose-intolerant and diabetic mice, Biol. Pharm. Bull. 31 (3) (2008)
aging in Caenorhabditis elegans, J. Proteome Res. 13 (6) (2014) 2910e2919. 469e472.
[186] Y.J. Liu, G.E. Janssens, R.L. McIntyre, M. Molenaars, R. Kamble, A.W. Gao, [209] A.L. Alvers, L.K. Fishwick, M.S. Wood, D. Hu, H.S. Chung, W.A. Dunn Jr.,
A. Jongejan, M.V. Weeghel, A.W. MacInnes, R.H. Houtkooper, Glycine pro- J.P. Aris, Autophagy and amino acid homeostasis are required for chrono-
motes longevity in Caenorhabditis elegans in a methionine cycle-dependent logical longevity in Saccharomyces cerevisiae, Aging Cell 8 (4) (2009)
fashion, PLoS Genet. 15 (3) (2019) e1007633. 353e369.
[187] M.A. Freund, B. Chen, E.A. Decker, The inhibition of advanced glycation end [210] A. Valerio, G. D'Antona, E. Nisoli, Branched-chain amino acids, mitochondrial
products by carnosine and other natural dipeptides to reduce diabetic and biogenesis, and healthspan: an evolutionary perspective, Aging (Albany NY)
age-related complications, Compr. Rev. Food Sci. Food Saf. 17 (5) (2018) 3 (5) (2011) 464e478.
1367e1378. [211] N.E. Cummings, E.M. Williams, I. Kasza, E.N. Konon, M.D. Schaid,
[188] H. Ukeda, Y. Hasegawa, Y. Harada, M. Sawamura, Effect of carnosine and B.A. Schmidt, C. Poudel, D.S. Sherman, D. Yu, S.I. Arriola Apelo, S.E. Cottrell,
related compounds on the inactivation of human Cu,Zn-superoxide dis- G. Geiger, M.E. Barnes, J.A. Wisinski, R.J. Fenske, K.A. Matkowskyj,
mutase by modification of fructose and glycolaldehyde, Biosci. Biotechnol. M.E. Kimple, C.M. Alexander, M.J. Merrins, D.W. Lamming, Restoration of
Biochem. 66 (1) (2002) 36e43. metabolic health by decreased consumption of branched-chain amino acids,
[189] J.T. Murphy, J.J. Bruinsma, D.L. Schneider, S. Collier, J. Guthrie, A. Chinwalla, J. Physiol. 596 (4) (2018) 623e645.
J.D. Robertson, E.R. Mardis, K. Kornfeld, Histidine protects against zinc and [212] L. Fontana, N.E. Cummings, S.I. Arriola Apelo, J.C. Neuman, I. Kasza,
nickel toxicity in Caenorhabditis elegans, PLoS Genet. 7 (3) (2011) e1002013. B.A. Schmidt, E. Cava, F. Spelta, V. Tosti, F.A. Syed, E.L. Baar, N. Veronese,
[190] M.C. Udechukwu, S.A. Collins, C.C. Udenigwe, Prospects of enhancing dietary S.E. Cottrell, R.J. Fenske, B. Bertozzi, H.K. Brar, T. Pietka, A.D. Bullock,
zinc bioavailability with food-derived zinc-chelating peptides, Food & R.S. Figenshau, G.L. Andriole, M.J. Merrins, C.M. Alexander, M.E. Kimple,
function 7 (10) (2016) 4137e4144. D.W. Lamming, Decreased consumption of branched-chain amino acids
[191] A.R. Hipkiss, E. Baye, B. de Courten, Carnosine and the processes of ageing, improves metabolic health, Cell Rep. 16 (2) (2016) 520e530.
Maturitas 93 (2016) 28e33. [213] F. Xiao, Z. Huang, H. Li, J. Yu, C. Wang, S. Chen, Q. Meng, Y. Cheng, X. Gao, J. Li,
[192] J.H. Cararo, E.L. Streck, P.F. Schuck, C. Ferreira Gda, Carnosine and related Y. Liu, F. Guo, Leucine deprivation increases hepatic insulin sensitivity via
peptides: therapeutic potential in age-related disorders, Aging Dis 6 (5) GCN2/mTOR/S6K1 and AMPK pathways, Diabetes 60 (3) (2011) 746e756.
(2015) 369e379. [214] A.U. Amaral, G. Leipnitz, C.G. Fernandes, B. Seminotti, P.F. Schuck, M. Wajner,
[193] Y.T. Lee, C.C. Hsu, M.H. Lin, K.S. Liu, M.C. Yin, Histidine and carnosine delay Alpha-ketoisocaproic acid and leucine provoke mitochondrial bioenergetic
diabetic deterioration in mice and protect human low density lipoprotein dysfunction in rat brain, Brain Res. 1324 (2010) 75e84.
against oxidation and glycation, Eur. J. Pharmacol. 513 (1e2) (2005) [215] H.S. Brunetta, C.Q. de Camargo, E.A. Nunes, Does L-leucine supplementation
145e150. cause any effect on glucose homeostasis in rodent models of glucose intol-
[194] W.H. Liu, T.C. Liu, M.C. Yin, Beneficial effects of histidine and carnosine on erance? A systematic review, Amino Acids 50 (12) (2018) 1663e1678.
ethanol-induced chronic liver injury, Food Chem. Toxicol. : an international [216] J. Zhou, S.Y. Chong, A. Lim, B.K. Singh, R.A. Sinha, A.B. Salmon, P.M. Yen,
journal published for the British Industrial Biological Research Association Changes in macroautophagy, chaperone-mediated autophagy, and mito-
46 (5) (2008) 1503e1509. chondrial metabolism in murine skeletal and cardiac muscle during aging,
[195] S. Hasegawa, T. Ichiyama, I. Sonaka, A. Ohsaki, S. Okada, H. Wakiguchi, Aging (Albany NY) 9 (2) (2017) 583e599.
K. Kudo, S. Kittaka, M. Hara, S. Furukawa, Cysteine, histidine and glycine [217] O. Deda, H.G. Gika, I. Taitzoglou, N. Raikos, G. Theodoridis, Impact of exercise
exhibit anti-inflammatory effects in human coronary arterial endothelial and aging on rat urine and blood metabolome. An LC-MS based metab-
cells, Clin. Exp. Immunol. 167 (2) (2012) 269e274. olomics longitudinal study, Metabolites 7 (1) (2017) 10.
[196] O.I. Pisarenko, Mechanisms of myocardial protection by amino acids: facts [218] H.L. Fox, P.T. Pham, S.R. Kimball, L.S. Jefferson, C.J. Lynch, Amino acid effects
and hypotheses, Clin. Exp. Pharmacol. Physiol. 23 (8) (1996) 627e633. on translational repressor 4E-BP1 are mediated primarily by L-leucine in
[197] M. Watanabe, M.E. Suliman, A.R. Qureshi, E. Garcia-Lopez, P. Barany, isolated adipocytes, Am. J. Physiol. 275 (5) (1998) C1232eC1238.
O. Heimburger, P. Stenvinkel, B. Lindholm, Consequences of low plasma [219] C.J. Lynch, H.L. Fox, T.C. Vary, L.S. Jefferson, S.R. Kimball, Regulation of amino
histidine in chronic kidney disease patients: associations with inflammation, acid-sensitive TOR signaling by leucine analogues in adipocytes, J. Cell.
oxidative stress, and mortality, Am. J. Clin. Nutr. 87 (6) (2008) 1860e1866. Biochem. 77 (2) (2000) 234e251.
[198] Z. Yu, G. Zhai, P. Singmann, Y. He, T. Xu, C. Prehn, W. Ro € misch-Margl, [220] R.A. Saxton, K.E. Knockenhauer, R.L. Wolfson, L. Chantranupong, M.E. Pacold,
E. Lattka, C. Gieger, N. Soranzo, J. Heinrich, M. Standl, E. Thiering, T. Wang, T.U. Schwartz, D.M. Sabatini, Structural basis for leucine sensing by
K. Mittelstraß, H.-E. Wichmann, A. Peters, K. Suhre, Y. Li, J. Adamski, the Sestrin2-mTORC1 pathway, Science 351 (6268) (2016) 53e58.
T.D. Spector, T. Illig, R. Wang-Sattler, Human serum metabolic profiles are [221] R.L. Wolfson, L. Chantranupong, R.A. Saxton, K. Shen, S.M. Scaria, J.R. Cantor,
age dependent, Aging Cell 11 (6) (2012) 960e967. D.M. Sabatini, Sestrin2 is a leucine sensor for the mTORC1 pathway, Science
[199] S. Cheng, M.G. Larson, E.L. McCabe, J.M. Murabito, E.P. Rhee, J.E. Ho, 351 (6268) (2016) 43e48.
P.F. Jacques, A. Ghorbani, M. Magnusson, A.L. Souza, A.A. Deik, K.A. Pierce, [222] D. Xu, K.L. Shimkus, H.A. Lacko, L. Kutzler, L.S. Jefferson, S.R. Kimball, Evi-
K. Bullock, C.J. O'Donnell, O. Melander, C.B. Clish, R.S. Vasan, R.E. Gerszten, dence for a role for Sestrin1 in mediating leucine-induced activation of
T.J. Wang, Distinct metabolomic signatures are associated with longevity in mTORC1 in skeletal muscle, Am. J. Physiol. Endocrinol. Metab. 316 (5) (2019)
humans, Nat. Commun. 6 (2015) 6791. E817ee828.
[200] A.O. Yuneva, G.G. Kramarenko, T.V. Vetreshchak, S. Gallant, A.A. Boldyrev, [223] J.M. Han, S.J. Jeong, M.C. Park, G. Kim, N.H. Kwon, H.K. Kim, S.H. Ha, S.H. Ryu,
Effect of carnosine on Drosophila melanogaster lifespan, Bull. Exp. Biol. Med. S. Kim, Leucyl-tRNA synthetase is an intracellular leucine sensor for the
133 (6) (2002) 559e561. mTORC1-signaling pathway, Cell 149 (2) (2012) 410e424.
[201] M.G. Buse, D.A. Weigand, Studies concerning the specificity of the effect of [224] M. Lee, J.H. Kim, I. Yoon, C. Lee, M. Fallahi Sichani, J.S. Kang, J. Kang, M. Guo,
leucine on the turnover of proteins in muscles of control and diabetic rats, K.Y. Lee, G. Han, S. Kim, J.M. Han, Coordination of the leucine-sensing Rag
Biochim. Biophys. Acta 475 (1) (1977) 81e89. GTPase cycle by leucyl-tRNA synthetase in the mTORC1 signaling pathway,
[202] R. Chaleckis, I. Murakami, J. Takada, H. Kondoh, M. Yanagida, Individual Proc. Natl. Acad. Sci. U. S. A. 115 (23) (2018) E5279ee5288.
variability in human blood metabolites identifies age-related differences, [225] J. Boultwood, B.H. Yip, C. Vuppusetty, A. Pellagatti, J.S. Wainscoat, Activation
Proc. Natl. Acad. Sci. U. S. A. 113 (16) (2016) 4252e4259. of the mTOR pathway by the amino acid (L)-leucine in the 5q- syndrome and
[203] K. Xie, F. Neff, A. Markert, J. Rozman, J.A. Aguilar-Pimentel, O.V. Amarie, other ribosomopathies, Adv. Biol. Regul. 53 (1) (2013) 8e17.
L. Becker, R. Brommage, L. Garrett, K.S. Henzel, S.M. Holter, D. Janik, [226] S.M. Solon-Biet, V.C. Cogger, T. Pulpitel, D. Wahl, X. Clark, E.E. Bagley,
I. Lehmann, K. Moreth, B.L. Pearson, I. Racz, B. Rathkolb, D.P. Ryan, G.C. Gregoriou, A.M. Senior, Q.-P. Wang, A.E. Brandon, R. Perks, J. O'Sullivan,
86 C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89

Y.C. Koay, K. Bell-Anderson, M. Kebede, B. Yau, C. Atkinson, G. Svineng, [250] R.S. McIsaac, K.N. Lewis, P.A. Gibney, R. Buffenstein, From yeast to human:
T. Dodgson, J.A. Wali, M.D.W. Piper, P. Juricic, L. Partridge, A.J. Rose, exploring the comparative biology of methionine restriction in extending
D. Raubenheimer, G.J. Cooney, D.G. Le Couteur, S.J. Simpson, Branched-chain eukaryotic life span, Ann. N. Y. Acad. Sci. 1363 (2016) 155e170.
amino acids impact health and lifespan indirectly via amino acid balance and [251] D. Mladenovic, T. Radosavljevic, D. Hrncic, A. Rasic-Markovic, O. Stanojlovic,
appetite control, Nat. Metab. 1 (5) (2019) 532e545. The effects of dietary methionine restriction on the function and metabolic
[227] J.C. Anthony, T.G. Anthony, S.R. Kimball, L.S. Jefferson, Signaling pathways reprogramming in the liver and brain - implications for longevity, Rev.
involved in translational control of protein synthesis in skeletal muscle by Neurosci. 30 (6) (2019) 581e593.
leucine, J. Nutr. 131 (3) (2001) 856se860s. [252] S. Yoshida, K. Yamahara, S. Kume, D. Koya, M. Yasuda-Yamahara, N. Takeda,
[228] J.C. Anthony, F. Yoshizawa, T.G. Anthony, T.C. Vary, L.S. Jefferson, S.R. Kimball, N. Osawa, M. Chin-Kanasaki, Y. Adachi, K. Nagao, H. Maegawa, S.I. Araki, Role
Leucine stimulates translation initiation in skeletal muscle of postabsorptive of dietary amino acid balance in diet restriction-mediated lifespan extension,
rats via a rapamycin-sensitive pathway, J. Nutr. 130 (10) (2000) 2413e2419. renoprotection, and muscle weakness in aged mice, Aging Cell 17 (4) (2018),
[229] J.T. Cunningham, J.T. Rodgers, D.H. Arlow, F. Vazquez, V.K. Mootha, e12796.
P. Puigserver, mTOR controls mitochondrial oxidative function through a [253] C. Hine, J.R. Mitchell, Calorie restriction and methionine restriction in control
YY1-PGC-1alpha transcriptional complex, Nature 450 (7170) (2007) of endogenous hydrogen sulfide production by the transsulfuration
736e740. pathway, Exp. Gerontol. 68 (2015) 26e32.
[230] R.M. Fulks, J.B. Li, A.L. Goldberg, Effects of insulin, glucose, and amino acids [254] A. Gomez, J. Gomez, M. Lopez Torres, A. Naudi, N. Mota-Martorell,
on protein turnover in rat diaphragm, J. Biol. Chem. 250 (1) (1975) 290e298. R. Pamplona, G. Barja, Cysteine dietary supplementation reverses the
[231] W.M. Bennet, A.A. Connacher, C.M. Scrimgeour, K. Smith, M.J. Rennie, In- decrease in mitochondrial ROS production at complex I induced by methi-
crease in anterior tibialis muscle protein synthesis in healthy man during onine restriction, J. Bioenerg. Biomembr. 47 (3) (2015) 199e208.
mixed amino acid infusion: studies of incorporation of [1-13C]leucine, Clin. [255] K.P. Stone, D. Wanders, M. Orgeron, C.C. Cortez, T.W. Gettys, Mechanisms of
Sci. 76 (4) (1989) 447e454. increased in vivo insulin sensitivity by dietary methionine restriction in
[232] K. Smith, J.M. Barua, P.W. Watt, C.M. Scrimgeour, M.J. Rennie, Flooding with mice, Diabetes 63 (11) (2014) 3721e3733.
L-[1-13C]leucine stimulates human muscle protein incorporation of [256] D. Wanders, K.P. Stone, L.A. Forney, C.C. Cortez, K.N. Dille, J. Simon, M. Xu,
continuously infused L-[1-13C]valine, Am. J. Physiol. 262 (3 Pt 1) (1992) E.C. Hotard, I.A. Nikonorova, A.P. Pettit, T.G. Anthony, T.W. Gettys, Role of
E372eE376. GCN2-independent signaling through a noncanonical PERK/NRF2 pathway in
[233] C. Liang, B.J. Curry, P.L. Brown, M.B. Zemel, Leucine modulates mitochondrial the physiological responses to dietary methionine restriction, Diabetes 65
biogenesis and SIRT1-AMPK signaling in C2C12 myotubes, J. Nutr. Metabol. (6) (2016) 1499e1510.
2014 (2014) 239750. [257] C.M. Hill, T. Laeger, M. Dehner, D.C. Albarado, B. Clarke, D. Wanders,
[234] M. Bonnefoy, T. Gilbert, O. Bruyere, E. Paillaud, A. Raynaud-Simon, O. Guerin, S.J. Burke, J.J. Collier, E. Qualls-Creekmore, S.M. Solon-Biet, S.J. Simpson,
C. Jeandel, B. Le Sourd, M. Haine, M. Ferry, Y. Rolland, G. Berrut, Protein H.R. Berthoud, H. Munzberg, C.D. Morrison, FGF21 signals protein status to
supplementation to prevent loss in muscle mass and strength in frail older the brain and adaptively regulates food choice and metabolism, Cell Rep. 27
patients: a review, Geriatrie et psychol. neuropsychiatrie vieillissement 17 (10) (2019) 2934e2947, e3.
(2) (2019) 137e143. [258] X. Gu, J.M. Orozco, R.A. Saxton, K.J. Condon, G.Y. Liu, P.A. Krawczyk,
[235] S. Verhoeven, K. Vanschoonbeek, L.B. Verdijk, R. Koopman, W.K. Wodzig, S.M. Scaria, J.W. Harper, S.P. Gygi, D.M. Sabatini, SAMTOR is an S-adeno-
P. Dendale, L.J. van Loon, Long-term leucine supplementation does not in- sylmethionine sensor for the mTORC1 pathway, Science 358 (6364) (2017)
crease muscle mass or strength in healthy elderly men, Am. J. Clin. Nutr. 89 813e818.
(5) (2009) 1468e1475. [259] B.M. Sutter, X. Wu, S. Laxman, B.P. Tu, Methionine inhibits autophagy and
[236] €nke, L. Partridge, Branched-chain amino acids have equiva-
P. Juricic, S. Gro promotes growth by inducing the SAM-responsive methylation of PP2A, Cell
lent effects to other essential amino acids on lifespan and aging-related traits 154 (2) (2013) 403e415.
in Drosophila, J. Gerontol.: Series A (2019). [260] F. Cabreiro, C. Au, K.Y. Leung, N. Vergara-Irigaray, H.M. Cocheme, T. Noori,
[237] J.P. Aris, A.L. Alvers, R.A. Ferraiuolo, L.K. Fishwick, A. Hanvivatpong, D. Hu, D. Weinkove, E. Schuster, N.D. Greene, D. Gems, Metformin retards aging in
C. Kirlew, M.T. Leonard, K.J. Losin, M. Marraffini, A.Y. Seo, V. Swanberg, C. elegans by altering microbial folate and methionine metabolism, Cell 153
J.L. Westcott, M.S. Wood, C. Leeuwenburgh, W.A. Dunn Jr., Autophagy and (1) (2013) 228e239.
leucine promote chronological longevity and respiration proficiency during [261] M.F. McCarty, J. Barroso-Aranda, F. Contreras, The low-methionine content of
calorie restriction in yeast, Exp. Gerontol. 48 (10) (2013) 1107e1119. vegan diets may make methionine restriction feasible as a life extension
[238] C.M. Gorospe, S.-L. Yu, M.-S. Kang, S.-K. Lee, Chronological lifespan regulation strategy, Med. Hypotheses 72 (2) (2009) 125e128.
of Saccharomyces cerevisiae by leucine biosynthesis pathway genes via [262] J.A. Schmidt, S. Rinaldi, A. Scalbert, P. Ferrari, D. Achaintre, M.J. Gunter,
TOR1 and COX2 expression regulation, Mol. Cell. Toxicol. 15 (1) (2019) P.N. Appleby, T.J. Key, R.C. Travis, Plasma concentrations and intakes of
65e73. amino acids in male meat-eaters, fish-eaters, vegetarians and vegans: a
[239] S. Robida-Stubbs, K. Glover-Cutter, D.W. Lamming, M. Mizunuma, cross-sectional analysis in the EPIC-Oxford cohort, Eur. J. Clin. Nutr. 70 (3)
S.D. Narasimhan, E. Neumann-Haefelin, D.M. Sabatini, T.K. Blackwell, TOR (2016) 306e312.
signaling and rapamycin influence longevity by regulating SKN-1/Nrf and [263] P. Pissios, S. Hong, A.R. Kennedy, D. Prasad, F.F. Liu, E. Maratos-Flier,
DAF-16/FoxO, Cell Metabol. 15 (5) (2012) 713e724. Methionine and choline regulate the metabolic phenotype of a ketogenic
[240] J.C. Newman, A.J. Covarrubias, M. Zhao, X. Yu, P. Gut, C.P. Ng, Y. Huang, diet, Mol. Metabol. 2 (3) (2013) 306e313.
S. Haldar, E. Verdin, Ketogenic diet reduces midlife mortality and improves [264] S.H.M. Gorissen, J.J.R. Crombag, J.M.G. Senden, W.A.H. Waterval, J. Bierau,
memory in aging mice, Cell Metabol. 26 (3) (2017) 547e557, e8. L.B. Verdijk, L.J.C. van Loon, Protein content and amino acid composition of
[241] M.N. Roberts, M.A. Wallace, A.A. Tomilov, Z. Zhou, G.R. Marcotte, D. Tran, commercially available plant-based protein isolates, Amino Acids 50 (12)
G. Perez, E. Gutierrez-Casado, S. Koike, T.A. Knotts, D.M. Imai, S.M. Griffey, (2018) 1685e1695.
K. Kim, K. Hagopian, F.G. Haj, K. Baar, G.A. Cortopassi, J.J. Ramsey, J.A. Lopez- [265] I. Sanchez-Roman, A. Gomez, J. Gomez, H. Suarez, C. Sanchez, A. Naudi,
Dominguez, A ketogenic diet extends longevity and healthspan in adult V. Ayala, M. Portero-Otin, M. Lopez-Torres, R. Pamplona, G. Barja, Forty
mice, Cell Metabol. 26 (3) (2017) 539e546, e5. percent methionine restriction lowers DNA methylation, complex I ROS
[242] D. Kalogeropoulou, L. LaFave, K. Schweim, M.C. Gannon, F.Q. Nuttall, Lysine generation, and oxidative damage to mtDNA and mitochondrial proteins in
ingestion markedly attenuates the glucose response to ingested glucose rat heart, J. Bioenerg. Biomembr. 43 (6) (2011) 699e708.
without a change in insulin response, Am. J. Clin. Nutr. 90 (2) (2009) [266] X. Gao, S.M. Sanderson, Z. Dai, M.A. Reid, D.E. Cooper, M. Lu, J.P. Richie,
314e320. A. Ciccarella, A. Calcagnotto, P.G. Mikhael, S.J. Mentch, J. Liu, G. Ables,
[243] K.N. Sulochana, R. Punitham, S. Ramakrishnan, Beneficial effect of lysine and D.G. Kirsch, D.S. Hsu, S.N. Nichenametla, J.W. Locasale, Dietary methionine
amino acids on cataractogenesis in experimental diabetes through possible restriction targets one carbon metabolism in humans and produces broad
antiglycation of lens proteins, Exp. Eye Res. 67 (5) (1998) 597e601. therapeutic responses in cancer, bioRxiv (2019) 627364.
[244] D. Datta, A. Bhinge, V. Chandran, Lysine: is it worth more? Cytotechnology 36 [267] R.A. Miller, G. Buehner, Y. Chang, J.M. Harper, R. Sigler, M. Smith-Wheelock,
(1e3) (2001) 3e32. Methionine-deficient diet extends mouse lifespan, slows immune and lens
[245] N. Orentreich, J.R. Matias, A. DeFelice, J.A. Zimmerman, Low methionine aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte
ingestion by rats extends life span, J. Nutr. 123 (2) (1993) 269e274. MIF levels and stress resistance, Aging Cell 4 (3) (2005) 119e125.
[246] Z. Dong, R. Sinha, J.P. Richie Jr., Disease prevention and delayed aging by [268] J.P. Richie Jr., Y. Leutzinger, S. Parthasarathy, V. Malloy, N. Orentreich,
dietary sulfur amino acid restriction: translational implications, Ann. N. Y. J.A. Zimmerman, Methionine restriction increases blood glutathione and
Acad. Sci. 1418 (1) (2018) 44e55. longevity in F344 rats, FASEB J. : off. publi. Feder. Am. Soc. Exp. Biol. 8 (15)
[247] L.A. Forney, K.P. Stone, D. Wanders, T.W. Gettys, Sensing and signaling (1994) 1302e1307.
mechanisms linking dietary methionine restriction to the behavioral and [269] A.M. Troen, E.E. French, J.F. Roberts, J. Selhub, J.M. Ordovas, L.D. Parnell,
physiological components of the response, Front. Neuroendocrinol. 51 C.Q. Lai, Lifespan modification by glucose and methionine in Drosophila
(2018) 36e45. melanogaster fed a chemically defined diet, Age 29 (1) (2007) 29e39.
[248] W.O. Jonsson, N.S. Margolies, T.G. Anthony, Dietary sulfur amino acid re- [270] J. Brind, V. Malloy, I. Augie, N. Caliendo, J.H. Vogelman, J.A. Zimmerman,
striction and the integrated stress response: mechanistic insights, Nutrients N. Orentreich, Dietary glycine supplementation mimics lifespan extension by
11 (6) (2019). dietary methionine restriction in Fisher 344 rats, FASEB (Fed. Am. Soc. Exp.
[249] M.N. Latimer, K.W. Freij, B.M. Cleveland, P.R. Biga, Physiological and mo- Biol.) J. 25 (2011), 528.2.
lecular mechanisms of methionine restriction, Front. Endocrinol. 9 (2018) [271] E.K. Lees, R. Banks, C. Cook, S. Hill, N. Morrice, L. Grant, N. Mody,
217. M. Delibegovic, Direct comparison of methionine restriction with leucine
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 87

restriction on the metabolic health of C57BL/6J mice, Sci. Rep. 7 (1) (2017) M. Platzer, C.R. Kahn, M. Ristow, Impaired insulin/IGF1 signaling extends life
9977. span by promoting mitochondrial L-proline catabolism to induce a transient
[272] B.C. Lee, A. Kaya, S. Ma, G. Kim, M.V. Gerashchenko, S.H. Yim, Z. Hu, ROS signal, Cell Metabol. 15 (4) (2012) 451e465.
L.G. Harshman, V.N. Gladyshev, Methionine restriction extends lifespan of [294] I.L. Johnstone, Cuticle collagen genes. Expression in Caenorhabditis elegans,
Drosophila melanogaster under conditions of low amino-acid status, Nat. Trends Genet. : TIG (Trends Genet.) 16 (1) (2000) 21e27.
Commun. 5 (2014) 3592. [295] C.Y. Ewald, J.N. Landis, J. Porter Abate, C.T. Murphy, T.K. Blackwell, Dauer-
[273] J.E. Johnson, F.B. Johnson, Methionine restriction activates the retrograde independent insulin/IGF-1-signalling implicates collagen remodelling in
response and confers both stress tolerance and lifespan extension to yeast, longevity, Nature 519 (7541) (2015) 97e101.
mouse and human cells, PLoS One 9 (5) (2014), e97729. [296] X. Zhou, L. He, S. Zuo, Y. Zhang, D. Wan, C. Long, P. Huang, X. Wu, C. Wu,
[274] C. Ruckenstuhl, C. Netzberger, I. Entfellner, D. Carmona-Gutierrez, G. Liu, Y. Yin, Serine prevented high-fat diet-induced oxidative stress by
T. Kickenweiz, S. Stekovic, C. Gleixner, C. Schmid, L. Klug, A.G. Sorgo, activating AMPK and epigenetically modulating the expression of gluta-
T. Eisenberg, S. Buttner, G. Marino, R. Koziel, P. Jansen-Durr, K.U. Frohlich, thione synthesis-related genes, Biochimica et biophysica acta, Mol Basis Dis.
G. Kroemer, F. Madeo, Lifespan extension by methionine restriction requires 1864 (2) (2018) 488e498.
autophagy-dependent vacuolar acidification, PLoS Genet. 10 (5) (2014) [297] G. Cao, F. Tao, L. Xin, Z. Li, X. Zhou, Effects of maternal serine supplemen-
e1004347. tation on high-fat diet-induced oxidative stress and epigenetic changes in
[275] K.M. Choi, S. Kim, S. Kim, H.M. Lee, A. Kaya, B.H. Chun, Y.K. Lee, T.S. Park, promoters of glutathione synthesis-related genes in offspring, J. Function.
C.K. Lee, S.I. Eyun, B.C. Lee, Sulfate assimilation regulates hydrogen sulfide Foods 47 (2018) 316e324.
production independent of lifespan and reactive oxygen species under [298] X. Zhou, L. He, C. Wu, Y. Zhang, X. Wu, Y. Yin, Serine alleviates oxidative
methionine restriction condition in yeast, Aging (Albany NY) 11 (12) (2019) stress via supporting glutathione synthesis and methionine cycle in mice,
4254e4273. Mol. Nutr. Food Res. 61 (11) (2017).
[276] F. Tripodi, A. Castoldi, R. Nicastro, V. Reghellin, L. Lombardi, C. Airoldi, [299] X. Zhou, Y. Zhang, L. He, D. Wan, G. Liu, X. Wu, Y. Yin, Serine prevents LPS-
E. Falletta, E. Maffioli, P. Scarcia, L. Palmieri, L. Alberghina, G. Agrimi, induced intestinal inflammation and barrier damage via p53-dependent
G. Tedeschi, P. Coccetti, Methionine supplementation stimulates mitochon- glutathione synthesis and AMPK activation, J. Function. Foods 39 (2017)
drial respiration, Biochimica et biophysica acta, Mol. cell Res. 1865 (12) 225e232.
(2018) 1901e1913. [300] X. Zhou, H. Zhang, L. He, X. Wu, Y. Yin, Long-term l-serine administration
[277] K. Campbell, J. Vowinckel, M.A. Keller, M. Ralser, Methionine metabolism reduces food intake and improves oxidative stress and sirt1/NFkappaB
alters oxidative stress resistance via the pentose phosphate pathway, Anti- signaling in the hypothalamus of aging mice, Front. Endocrinol. 9 (2018) 476.
oxidants Redox Signal. 24 (10) (2016) 543e547. [301] Y. Hirabayashi, S. Furuya, Roles of l-serine and sphingolipid synthesis in
[278] C.D. Tavares, K. Sharabi, J.E. Dominy, Y. Lee, M. Isasa, J.M. Orozco, brain development and neuronal survival, Prog. Lipid Res. 47 (3) (2008)
M.P. Jedrychowski, T.M. Kamenecka, P.R. Griffin, S.P. Gygi, P. Puigserver, The 188e203.
methionine transamination pathway controls hepatic glucose metabolism [302] J.S. Metcalf, R.A. Dunlop, J.T. Powell, S.A. Banack, P.A. Cox, L-Serine,
through regulation of the GCN5 acetyltransferase and the PGC-1alpha A naturally-occurring amino acid with therapeutic potential, Neurotox. Res.
transcriptional coactivator, J. Biol. Chem. 291 (20) (2016) 10635e10645. 33 (1) (2018) 213e221.
[279] I. Montoliu, M. Scherer, F. Beguelin, L. DaSilva, D. Mari, S. Salvioli, F.P. Martin, [303] R.A. Dunlop, J. Powell, G.J. Guillemin, P.A. Cox, Mechanisms of L-serine
M. Capri, L. Bucci, R. Ostan, P. Garagnani, D. Monti, E. Biagi, P. Brigidi, neuroprotection in vitro include ER proteostasis regulation, Neurotox. Res.
M. Kussmann, S. Rezzi, C. Franceschi, S. Collino, Serum profiling of healthy 33 (1) (2018) 123e132.
aging identifies phospho- and sphingolipid species as markers of human [304] R.A. Dunlop, J.T. Powell, J.S. Metcalf, G.J. Guillemin, P.A. Cox, L-Serine-
longevity, Aging (Albany NY) 6 (1) (2014) 9e25. Mediated neuroprotection includes the upregulation of the ER stress chap-
[280] J.M. Hoffman, C. Ross, V. Tran, D.E.L. Promislow, S. Tardif, D.P. Jones, The erone protein disulfide isomerase (PDI), Neurotox. Res. 33 (1) (2018)
metabolome as a biomarker of mortality risk in the common marmoset, Am. 113e122.
J. Primatol. 81 (2) (2019), e22944. [305] H. Wolosker, D-serine regulation of NMDA receptor activity, Sci. STKE :
[281] J.M. Hoffman, V. Tran, L.M. Wachtman, C.L. Green, D.P. Jones, signal transduction knowledge environment 2006 356 (2006) pe41.
D.E.L. Promislow, A longitudinal analysis of the effects of age on the blood [306] S. Beltran-Castillo, J. Eugenin, R. von Bernhardi, Impact of aging in microglia-
plasma metabolome in the common marmoset, Callithrix jacchus, Exp. mediated D-serine balance in the CNS, Mediat. Inflamm. 2018 (2018)
Gerontol. 76 (2016) 17e24. 7219732.
[282] S. Tsakiris, P. Angelogianni, K.H. Schulpis, J.C. Stavridis, Protective effect of L- [307] W.G. Bradley, R.X. Miller, T.D. Levine, E.W. Stommel, P.A. Cox, Studies of
phenylalanine on rat brain acetylcholinesterase inhibition induced by free environmental risk factors in amyotrophic lateral sclerosis (ALS) and a phase
radicals, Clin. Biochem. 33 (2) (2000) 103e106. I clinical trial of l-serine, Neurotox. Res. 33 (1) (2018) 192e198.
[283] J.G. Eriksson, M.A. Guzzardi, P. Iozzo, E. Kajantie, H. Kautiainen, M.K. Salonen, [308] T.D. Levine, R.G. Miller, W.G. Bradley, D.H. Moore, D.S. Saperstein, L.E. Flynn,
Higher serum phenylalanine concentration is associated with more rapid J.S. Katz, D.A. Forshew, J.S. Metcalf, S.A. Banack, P.A. Cox, Phase I clinical trial
telomere shortening in men, Am. J. Clin. Nutr. 105 (1) (2017) 144e150. of safety of L-serine for ALS patients, Amyotrophic Lateral Sclerosis and
[284] G. Neurauter, K. Schrocksnadel, S. Scholl-Burgi, B. Sperner-Unterweger, Frontotemporal Degeneration 18 (1e2) (2017) 107e111.
C. Schubert, M. Ledochowski, D. Fuchs, Chronic immune stimulation corre- [309] P.A. Cox, J.S. Metcalf, Traditional food items in Ogimi, okinawa: l-serine
lates with reduced phenylalanine turnover, Curr. Drug Metabol. 9 (7) (2008) content and the potential for neuroprotection, Curr. Nutri. Rep. 6 (1) (2017)
622e627. 24e31.
[285] B. Medovar, K.J. Petzke, T.G. Semesko, V. Albrecht, G. Grigorov Ju, [The effect [310] M.N. Maralani, A. Movahedian, H. Javanmard Sh, Antioxidant and cytopro-
of different protein diets on longevity and various biochemical parameters of tective effects of L-Serine on human endothelial cells, Res. pharm. sci. 7 (4)
aged rats], Nahrung 35 (9) (1991) 961e967. (2012) 209e215.
[286] S. Arganda, S. Bouchebti, S. Bazazi, S. Le Hesran, C. Puga, G. Latil, S.J. Simpson, [311] N. Kory, G.A. Wyant, G. Prakash, J. Uit de Bos, F. Bottanelli, M.E. Pacold,
A. Dussutour, Parsing the life-shortening effects of dietary protein: effects of S.H. Chan, C.A. Lewis, T. Wang, H.R. Keys, Y.E. Guo, D.M. Sabatini, SFXN1 is a
individual amino acids, Proc Biol Sci 284 (2017) 1846. mitochondrial serine transporter required for one-carbon metabolism, Sci-
[287] B.R. Ipson, R.A. Green, J.T. Wilson, J.N. Watson, K.F. Faull, A.L. Fisher, Tyrosine ence 362 (6416) (2018).
aminotransferase is involved in the oxidative stress response by metabo- [312] J. Ye, J. Fan, S. Venneti, Y.W. Wan, B.R. Pawel, J. Zhang, L.W. Finley, C. Lu,
lizing meta-tyrosine in Caenorhabditis elegans, J. Biol. Chem. 294 (24) (2019) T. Lindsten, J.R. Cross, G. Qing, Z. Liu, M.C. Simon, J.D. Rabinowitz,
9536e9554. C.B. Thompson, Serine catabolism regulates mitochondrial redox control
[288] K. Toyoshima, M. Nakamura, Y. Adachi, A. Imaizumi, T. Hakamada, Y. Abe, during hypoxia, Cancer Discov. 4 (12) (2014) 1406e1417.
E. Kaneko, S. Takahashi, K. Shimokado, Increased plasma proline concen- [313] P. Guda, C. Guda, S. Subramaniam, Reconstruction of pathways associated
trations are associated with sarcopenia in the elderly, PLoS One 12 (9) (2017) with amino acid metabolism in human mitochondria, Genom. Proteom.
e0185206-e0185206. Bioinform. 5 (3e4) (2007) 166e176.
[289] W. Ren, L. Zou, Z. Ruan, N. Li, Y. Wang, Y. Peng, G. Liu, Y. Yin, T. Li, Y. Hou, [314] Y. Kitagawa, E. Sugimoto, Possibility of mitochondrial-cytosolic cooperation
G. Wu, Dietary L-proline supplementation confers immunostimulatory ef- in gluconeogenesis from serine via hydroxypyruvate, Biochim. Biophys. Acta
fects on inactivated Pasteurella multocida vaccine immunized mice, Amino 582 (2) (1979) 276e282.
Acids 45 (3) (2013) 555e561. [315] S.C. Kalhan, R.W. Hanson, Resurgence of serine: an often neglected but
[290] T. Ponrasu, S. Jamuna, A. Mathew, K.N. Madhukumar, M. Ganeshkumar, indispensable amino Acid, J. Biol. Chem. 287 (24) (2012) 19786e19791.
K. Iyappan, L. Suguna, Efficacy of L-proline administration on the early re- [316] K. Snell, D.A. Fell, Metabolic control analysis of mammalian serine meta-
sponses during cutaneous wound healing in rats, Amino Acids 45 (1) (2013) bolism, Adv. Enzym. Regul. 30 (1990) 13e32.
179e189. [317] L. Freysz, R. Bieth, P. Mandel, Kinetics of the biosynthesis of phospholipids in
[291] S. Hayat, Q. Hayat, M.N. Alyemeni, A.S. Wani, J. Pichtel, A. Ahmad, Role of neurons and glial cells isolated from rat brain cortex, J. Neurochem. 16 (10)
proline under changing environments: a review, Plant Signal. Behav. 7 (11) (1969) 1417e1424.
(2012) 1456e1466. [318] X. Gao, K. Lee, M.A. Reid, S.M. Sanderson, C. Qiu, S. Li, J. Liu, J.W. Locasale,
[292] J. Leal, L. Teixeira-Santos, D. Pinho, J. Afonso, J. Carvalho, M. de Lourdes Serine availability influences mitochondrial dynamics and function through
Bastos, A. Albino-Teixeira, S. Fraga, T. Sousa, l-proline supplementation im- lipid metabolism, Cell Rep. 22 (13) (2018) 3507e3520.
proves nitric oxide bioavailability and counteracts the blood pressure rise [319] K. Hagopian, J.J. Ramsey, R. Weindruch, Serine utilization in mouse liver:
induced by angiotensin II in rats, Nitric Oxide 82 (2019) 1e11. influence of caloric restriction and aging, FEBS Lett. 579 (9) (2005)
[293] K. Zarse, S. Schmeisser, M. Groth, S. Priebe, G. Beuster, D. Kuhlow, R. Guthke, 2009e2013.
88 C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89

[320] M.B. Yunus, J.W. Dailey, J.C. Aldag, A.T. Masi, P.C. Jobe, Plasma tryptophan [346] T. Chen, X. Zheng, X. Ma, Y. Bao, Y. Ni, C. Hu, C. Rajani, F. Huang, A. Zhao,
and other amino acids in primary fibromyalgia: a controlled study, W. Jia, W. Jia, Tryptophan predicts the risk for future type 2 diabetes, PLoS
J. Rheumatol. 19 (1) (1992) 90e94. One 11 (9) (2016) e0162192.
[321] P.P. Jung, Z. Zhang, N. Paczia, C. Jaeger, T. Ignac, P. May, C.L. Linster, Natural [347] G. Lindseth, B. Helland, J. Caspers, The effects of dietary tryptophan on af-
variation of chronological aging in the Saccharomyces cerevisiae species fective disorders, Arch. Psychiatr. Nurs. 29 (2) (2015) 102e107.
reveals diet-dependent mechanisms of life span control, NPJ Aging Mec. Dis. [348] B. Strasser, B. Sperner-Unterweger, D. Fuchs, J.M. Gostner, Mechanisms of
4 (2018) 3. inflammation-associated depression: immune influences on tryptophan and
[322] M.G. Mirisola, G. Taormina, P. Fabrizio, M. Wei, J. Hu, V.D. Longo, Serine- and phenylalanine metabolisms, Current topics in behav. neurosci. 31 (2017)
threonine/valine-dependent activation of PDK and tor orthologs converge on 95e115.
Sch9 to promote aging, PLoS Genet. 10 (2) (2014) e1004113. [349] S. Nikolaus, B. Schulte, N. Al-Massad, F. Thieme, D.M. Schulte, J. Bethge,
[323] X. H Zhao, Z.M. Wen, C. N Meredith, D. Matthews, D. M Bier, V. R Young, A. Rehman, F. Tran, K. Aden, R. Hasler, N. Moll, G. Schutze, M.J. Schwarz,
Threonine kinetics at graded thre-onine intakes in young men, Am. J. Clin. G.H. Waetzig, P. Rosenstiel, M. Krawczak, S. Szymczak, S. Schreiber, Increased
Nutr. 43 (1986) 795e802. tryptophan metabolism is associated with activity of inflammatory bowel
[324] X. Wang, S. Qiao, Y. Yin, L. Yue, Z. Wang, G. Wu, A deficiency or excess of diseases, Gastroenterology 153 (6) (2017) 1504e1516, e2.
dietary threonine reduces protein synthesis in jejunum and skeletal muscle [350] C.K. Lieben, A. Blokland, N.E. Deutz, W. Jansen, G. Han, R.M. Hupperts, Intake
of young pigs, J. Nutr. 137 (6) (2007) 1442e1446. of tryptophan-enriched whey protein acutely enhances recall of positive
[325] G. Boehm, H. Cervantes, G. Georgi, J. Jelinek, G. Sawatzki, B. Wermuth, loaded words in patients with multiple sclerosis, Clin. Nutr. 37 (1) (2018)
J.P. Colombo, Effect of increasing dietary threonine intakes on amino acid 321e328.
metabolism of the central nervous system and peripheral tissues in growing [351] A. Takada, F. Shimizu, J. Masuda, K. Matsuoka, Chapter 17 - plasma levels of
rats, Pediatr. Res. 44 (6) (1998) 900e906. tryptophan metabolites in patients of type 2 diabetes mellitus, in: R.R.
[326] L.J. Van Winkle, V. Galat, P.M. Iannaccone, Threonine appears to be essential Watson, V.R. Preedy (Eds.), Bioactive Food as Dietary Interventions for Dia-
for proliferation of human as well as mouse embryonic stem cells, Front. Cell. betes (second ed.), Academic Press2019, pp. 265-276.
Dev. Biol. 2 (18) (2014). [352] T. Inubushi, N. Kamemura, M. Oda, J. Sakurai, Y. Nakaya, N. Harada,
[327] J.M. Ryu, H.J. Han, L-threonine regulates G1/S phase transition of mouse M. Suenaga, Y. Matsunaga, K. Ishidoh, N. Katunuma, L-tryptophan suppresses
embryonic stem cells via PI3K/Akt, MAPKs, and mTORC pathways, J. Biol. rise in blood glucose and preserves insulin secretion in type-2 diabetes
Chem. 286 (27) (2011) 23667e23678. mellitus rats, J. Nutr. Sci. Vitaminol. 58 (6) (2012) 415e422.
[328] G. Chen, J. Wang, Threonine metabolism and embryonic stem cell self- [353] S. Esteban, C. Garau, S. Aparicio, D. Moranta, P. Barcelo, M.A. Fiol, R. Rial,
renewal, Curr. Opin. Clin. Nutr. Metab. Care 17 (1) (2014) 80e85. Chronic melatonin treatment and its precursor L-tryptophan improve the
[329] C.A. Titchenal, Q.R. Rogers, R.J. Indrieri, J.G. Morris, Threonine imbalance, monoaminergic neurotransmission and related behavior in the aged rat
deficiency and neurologic dysfunction in the Kitten, J. Nutr. 110 (12) (1980) brain, J. Pineal Res. 48 (2) (2010) 170e177.
2444e2459. [354] R. Bravo, S. Matito, J. Cubero, S.D. Paredes, L. Franco, M. Rivero,
[330] M.M.M. Azzam, X.Y. Dong, P. Xie, C. Wang, X.T. Zou, The effect of supple- A.B. Rodriguez, C. Barriga, Tryptophan-enriched cereal intake improves
mental L-threonine on laying performance, serum free amino acids, and nocturnal sleep, melatonin, serotonin, and total antioxidant capacity levels
immune function of laying hens under high-temperature and high-humidity and mood in elderly humans, Age 35 (4) (2013) 1277e1285.
environmental climates, J. Appl. Poult. Res. 20 (3) (2011) 361e370. [355] A.S. Avanesov, S. Ma, K.A. Pierce, S.H. Yim, B.C. Lee, C.B. Clish, V.N. Gladyshev,
[331] Y. Chen, H. Zhang, Y. Cheng, Y. Li, C. Wen, Y. Zhou, Dietary l-threonine Age- and diet-associated metabolome remodeling characterizes the aging
supplementation attenuates lipopolysaccharide-induced inflammatory re- process driven by damage accumulation, eLife 3 (2014), e02077.
sponses and intestinal barrier damage of broiler chickens at an early age, Br. [356] C. He, S.K. Tsuchiyama, Q.T. Nguyen, E.N. Plyusnina, S.R. Terrill, S. Sahibzada,
J. Nutr. 119 (11) (2018) 1254e1262. B. Patel, A.R. Faulkner, M.V. Shaposhnikov, R. Tian, M. Tsuchiya,
[332] X. Mao, X. Lai, B. Yu, J. He, J. Yu, P. Zheng, G. Tian, K. Zhang, D. Chen, Effects of M. Kaeberlein, A.A. Moskalev, B.K. Kennedy, M. Polymenis, Enhanced
dietary threonine supplementation on immune challenge induced by swine longevity by ibuprofen, conserved in multiple species, occurs in yeast
Pseudorabies live vaccine in weaned pigs, Arch. Anim. Nutr. 68 (1) (2014) through inhibition of tryptophan import, PLoS Genet. 10 (12) (2014)
1e15. e1004860.
[333] V.D. Longo, Programmed longevity, youthspan, and juventology, Aging Cell [357] C.F. Bennett, M. Kaeberlein, The mitochondrial unfolded protein response
18 (1) (2019), e12843. and increased longevity: cause, consequence, or correlation? Exp. Gerontol.
[334] H. Mirzaei, J.A. Suarez, V.D. Longo, Protein and amino acid restriction, aging 56 (2014) 142e146.
and disease: from yeast to humans, Trends Endocrinol. Metab. 25 (11) (2014) [358] C.F. Bennett, H. Vander Wende, M. Simko, S. Klum, S. Barfield, H. Choi,
558e566. V.V. Pineda, M. Kaeberlein, Activation of the mitochondrial unfolded protein
[335] M. Ravichandran, S. Priebe, G. Grigolon, L. Rozanov, M. Groth, B. Laube, response does not predict longevity in Caenorhabditis elegans, Nat. Com-
R. Guthke, M. Platzer, K. Zarse, M. Ristow, Impairing L-threonine catabolism mun. 5 (2014) 3483.
promotes healthspan through methylglyoxal-mediated proteohormesis, Cell [359] B.F. Darst, R.L. Koscik, K.J. Hogan, S.C. Johnson, C.D. Engelman, Longitudinal
Metabol. 27 (4) (2018) 914e925, e5. plasma metabolomics of aging and sex, Aging (Albany NY) 11 (4) (2019)
[336] P.E. Segall, P.S. Timiras, J.R. Walton, Low tryptophan diets delay reproductive 1262e1282.
aging, Mech. Ageing Dev. 23 (3e4) (1983) 245e252. [360] A.F. Sved, J.D. Fernstrom, R.J. Wurtman, Tyrosine administration reduces
[337] H. Ooka, P.E. Segall, P.S. Timiras, Histology and survival in age-delayed low- blood pressure and enhances brain norepinephrine release in spontaneously
tryptophan-fed rats, Mech. Ageing Dev. 43 (1) (1988) 79e98. hypertensive rats, Proc. Natl. Acad. Sci. U.S.A. 76 (7) (1979) 3511e3514.
[338] P.E. Segall, P.S. Timiras, Patho-physiologic findings after chronic tryptophan [361] S. Kühn, S. Düzel, L. Colzato, K. Norman, J. Gallinat, A.M. Brandmaier,
deficiency in rats: a model for delayed growth and aging, Mech. Ageing Dev. U. Lindenberger, K.F. Widaman, Food for Thought: Association between Di-
5 (2) (1976) 109e124. etary Tyrosine and Cognitive Performance in Younger and Older Adults,
[339] H.M. Brown-Borg, R. Buffenstein, Cutting back on the essentials: can Psychological Research, 2017.
manipulating intake of specific amino acids modulate health and lifespan? [362] O. van de Rest, M. Bloemendaal, R. de Heus, E. Aarts, Dose-dependent effects
Ageing Res. Rev. 39 (2017) 87e95. of oral tyrosine administration on plasma tyrosine levels and cognition in
[340] L.A. Ramos-Chavez, G. Roldan-Roldan, B. Garcia-Juarez, D. Gonzalez-Esquivel, aging, Nutrients 9 (12) (2017) 1279.
G. Perez de la Cruz, B. Pineda, D. Ramirez-Ortega, I. Garcia Munoz, B. Jimenez [363] B.J. Jongkees, B. Hommel, S. Kuhn, L.S. Colzato, Effect of tyrosine supple-
Herrera, C. Rios, S. Gomez-Manzo, J. Marcial-Quino, L. Sanchez Chapul, mentation on clinical and healthy populations under stress or cognitive
P. Carrillo Mora, V. Perez de la Cruz, Low serum tryptophan levels as an demands–A review, J. Psychiatr. Res. 70 (2015) 50e57.
indicator of global cognitive performance in nondemented women over 50 [364] S. Attipoe, S.A. Zeno, C. Lee, C. Crawford, R. Khorsan, A.R. Walter, P.A. Deuster,
Years of age, Oxid Med Cell Longev 2018 (2018) 8604718. Tyrosine for mitigating stress and enhancing performance in healthy adult
[341] M. Friedman, Analysis, nutrition, and health benefits of tryptophan, Int. J. humans, a rapid evidence assessment of the literature, Mil. Med. 180 (7)
Tryptophan Res. 11 (2018), 1178646918802282-1178646918802282. (2015) 754e765.
[342] H. Mangge, I. Stelzer, E.Z. Reininghaus, D. Weghuber, T.T. Postolache, [365] J.A. Lang, K.A. Smaller, Oral l-tyrosine supplementation augments the vaso-
D. Fuchs, Disturbed tryptophan metabolism in cardiovascular disease, Curr. constriction response to whole-body cooling in older adults, Exp. Physiol.
Med. Chem. 21 (17) (2014) 1931e1937. 102 (7) (2017) 835e844.
[343] C. Murr, T.B. Grammer, M.E. Kleber, A. Meinitzer, W. Marz, D. Fuchs, Low [366] J.H. Growdon, E. Melamed, M. Logue, F. Hefti, R.J. Wurtman, Effects of oral L-
serum tryptophan predicts higher mortality in cardiovascular disease, Eur. J. tyrosine administration on CSF tyrosine and homovanillic acid levels in pa-
Clin. Investig. 45 (3) (2015) 247e254. tients with Parkinson's disease, Life Sci. 30 (10) (1982) 827e832.
[344] E. Yu, M. Ruiz-Canela, M. Guasch-Ferre, Y. Zheng, E. Toledo, C.B. Clish, J. Salas- [367] C. Jang, S.F. Oh, S. Wada, G.C. Rowe, L. Liu, M.C. Chan, J. Rhee, A. Hoshino,
Salvado, L. Liang, D.D. Wang, D. Corella, M. Fito, E. Gomez-Gracia, J. Lapetra, B. Kim, A. Ibrahim, L.G. Baca, E. Kim, C.C. Ghosh, S.M. Parikh, A. Jiang, Q. Chu,
R. Estruch, E. Ros, M. Cofan, F. Aros, D. Romaguera, L. Serra-Majem, J.V. Sorli, D.E. Forman, S.H. Lecker, S. Krishnaiah, J.D. Rabinowitz, A.M. Weljie, J.A. Baur,
F.B. Hu, M.A. Martinez-Gonzalez, Increases in Plasma Tryptophan Are D.L. Kasper, Z. Arany, A branched-chain amino acid metabolite drives
Inversely Associated with Incident Cardiovascular Disease in the Prevencion vascular fatty acid transport and causes insulin resistance, Nat. Med. 22 (4)
con Dieta Mediterranea (PREDIMED) Study, J. Nutr. 147 (3) (2017) 314e322. (2016) 421e426.
[345] G. Schulman, A nexus of progression of chronic kidney disease: tryptophan, [368] Y. Tsuda, K. Iwasawa, M. Yamaguchi, Acute supplementation of valine re-
profibrotic cytokines, and charcoal, J. Ren. Nutr. : Off. J. Counc. Renal Nutri. duces fatigue during swimming exercise in rats, Biosc. Biotech. Biochem. 82
Natl. Kidney Found. 22 (1) (2012) 107e113. (5) (2018) 856e861.
C.-A. Canfield, P.C. Bradshaw / Translational Medicine of Aging 3 (2019) 70e89 89

[369] T. Chanyachukul, K. Yoovathaworn, W. Thongsaard, S. Chongthammakun, [375] A.J. Edgar, The human L-threonine 3-dehydrogenase gene is an expressed
P. Navasumrit, J. Satayavivad, Attenuation of paraquat-induced motor pseudogene, BMC Genet. 3 (2002) 18.
behavior and neurochemical disturbances by L-valine in vivo, Toxicol. Lett. [376] A.J. Edgar, Mice have a transcribed L-threonine aldolase/GLY1 gene, but the
150 (3) (2004) 259e269. human GLY1 gene is a non-processed pseudogene, BMC Genomics 6 (2005),
[370] T. Yoneshiro, Q. Wang, K. Tajima, M. Matsushita, H. Maki, K. Igarashi, Z. Dai, 32-32.
P.J. White, R.W. McGarrah, O.R. Ilkayeva, Y. Deleye, Y. Oguri, M. Kuroda, [377] B. Stoll, J. Henry, P.J. Reeds, H. Yu, F. Jahoor, D.G. Burrin, Catabolism domi-
K. Ikeda, H. Li, A. Ueno, M. Ohishi, T. Ishikawa, K. Kim, Y. Chen, C.H. Sponton, nates the first-pass intestinal metabolism of dietary essential amino acids in
R.N. Pradhan, H. Majd, V.J. Greiner, M. Yoneshiro, Z. Brown, milk protein-fed piglets, J. Nutr. 128 (3) (1998) 606e614.
M. Chondronikola, H. Takahashi, T. Goto, T. Kawada, L. Sidossis, F.C. Szoka, [378] T.J. Wester, G. Kraft, D. Dardevet, S. Polakof, I. Ortigues-Marty, D. Remond,
M.T. McManus, M. Saito, T. Soga, S. Kajimura, BCAA catabolism in brown fat I. Savary-Auzeloux, Nutritional regulation of the anabolic fate of amino acids
controls energy homeostasis through SLC25A44, Nature (2019). within the liver in mammals: concepts arising from in vivo studies, Nutr. Res.
[371] K. Shigemi, S. Tomonaga, N. Uotsu, M. Denbow D, M. Furuse, Oral adminis- Rev. 28 (1) (2015) 22e41.
tration of L-serine modifies amino acid metabolism in the brain of rats, [379] M.L. De Marte, H.E. Enesco, Influence of low tryptophan diet on survival and
J. Anim. Res. Nutr. 01 (2016). organ growth in mice, Mech. Ageing Dev. 36 (2) (1986) 161e171.
[372] P. Verhoef, G.R. Steenge, E. Boelsma, T. van Vliet, M.R. Olthof, M.B. Katan, [380] C.M. Chak, M.E. Lacruz, J. Adam, S. Brandmaier, M. Covic, J. Huang,
Dietary serine and cystine attenuate the homocysteine-raising effect of di- C. Meisinger, D. Tiller, C. Prehn, J. Adamski, U. Berger, C. Gieger, A. Peters,
etary methionine: a randomized crossover trial in humans, Am. J. Clin. Nutr. A. Kluttig, R. Wang-Sattler, Ageing investigation using two-time-point
80 (3) (2004) 674e679. metabolomics data from KORA and CARLA studies, Metabolites 9 (3) (2019).
[373] G.S. Ducker, J.D. Rabinowitz, One-carbon metabolism in health and disease, [381] D. Varshavi, F.H. Scott, D. Varshavi, S. Veeravalli, I.R. Phillips, K. Veselkov,
Cell Metabol. 25 (1) (2017) 27e42. N. Strittmatter, Z. Takats, E.A. Shephard, J.R. Everett, Metabolic biomarkers of
[374] S. Eriksson, J.R. Prigge, E.A. Talago, E.S. Arner, E.E. Schmidt, Dietary methio- ageing in C57BL/6J wild-type and flavin-containing monooxygenase 5
nine can sustain cytosolic redox homeostasis in the mouse liver, Nat. Com- (FMO5)-Knockout mice, Front. mol. biosci. 5 (2018) 28.
mun. 6 (2015) 6479.

You might also like