2,484
Views
0
CrossRef citations to date
0
Altmetric
Review Articles

Dietary protein and amino acid intakes for mitigating sarcopenia in humans

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon

Abstract

Adult humans generally experience a 0.5–1%/year loss in whole-body skeletal muscle mass and a reduction of muscle strength by 1.5–5%/year beginning at the age of 50 years. This results in sarcopenia (aging-related progressive losses of skeletal muscle mass and strength) that affects 10–16% of adults aged ≥ 60 years worldwide. Concentrations of some amino acids (AAs) such as branched-chain AAs, arginine, glutamine, glycine, and serine are reduced in the plasma of older than young adults likely due to insufficient protein intake, reduced protein digestibility, and increased AA catabolism by the portal-drained viscera. Acute, short-term, or long-term administration of some of these AAs or a mixture of proteinogenic AAs can enhance blood flow to skeletal muscle, activate the mechanistic target of rapamycin cell signaling pathway for the initiation of muscle protein synthesis, and modulate the metabolic activity of the muscle. In addition, some AA metabolites such as taurine, β-alanine, carnosine, and creatine have similar physiological effects on improving muscle mass and function in older adults. Long-term adequate intakes of protein and the AA metabolites can aid in mitigating sarcopenia in elderly adults. Appropriate combinations of animal- and plant-sourced foods are most desirable to maintain proper dietary AA balance.

Introduction

Skeletal muscle is a major organ in humans, representing approximately 40% and 32% of the total body mass in 30-year-old healthy men and women, respectively (Janssen et al. Citation2000). The whole body and skeletal muscle of adults contain approximately 15% and 20% protein, respectively (Duda et al. Citation2019). Thus, the 70-kg man and the 60-kg women have 10.5 and 9.0 kg protein, respectively, and their skeletal muscle accounts for 53% and 43% of the whole-body protein, respectively. Notably, skeletal muscle mass, which depends on the balance between the rates of protein synthesis and degradation, is relatively constant between 18 and 45 years of age in both men and women and thereafter decline (Janssen et al. Citation2000).

Sarcopenia is an aging-related progressive loss of skeletal muscle mass and function (Cruz-Jentoft et al. Citation2019) and affects 10–16% of adults aged 60 years or older worldwide (Yuan and Larsson Citation2023). This health problem can start as early as 27 years of age in sedentary men and women (Silva et al. Citation2010). Without preventative methods, adults generally experience a 0.5–1% loss/year in the whole-body skeletal muscle mass (including a loss of leg muscle by 1–2% per year) and a reduction of leg muscle strength by 1.5–5% per year) from the age of 50 years, with the rate of loss accelerating from 70 years of age onwards (Cruz-Jentoft et al. Citation2019; Lonnie et al. Citation2018). Notably, in individuals with limited physical activity, there is a 30–50% decrease in skeletal muscle mass and strength between the ages of 30 and 80 years (Daley and Spinks Citation2000). Sarcopenia results in physical weakness, disability, dependent living, and a diminished quality of life, while increasing the risks of falls, fractures, morbidity, and mortality, as well as health care expenditure, costly hospitalization, and extended rehabilitation (Larsson et al. Citation2019). With an increase in life expectancy due to improvements in living conditions and health management, the global aging population is increasing. For example, it has been estimated that by the year 2050, 20% of the U.S. population will be aged > 65 years (Mather, Jacobsen, and Pollard Citation2015). This will be associated with a dramatic increase in the prevalence of sarcopenia, which will reduce the longevity of humans and the quality of life. Preserving skeletal muscle mass and function is crucial for elderly adults to remain healthy and functionally independent.

Rates of protein synthesis and degradation in skeletal muscle are affected by physiological, pathological, and nutritional factors, including muscle contractility, insulin sensitivity, and stress as well as dietary intakes of energy, protein, amino acids (AAs), lipids, carbohydrates, vitamins, and minerals (Suryawan and Davis Citation2011; Wu Citation2022). A loss of muscle protein results from a lower rate of its synthesis than the rate of its degradation. Much evidence shows that an insufficient intake of protein, especially animal-sourced protein, contributes to the development of sarcopenia in men and women (Lancha et al. Citation2017; Putra et al. Citation2021). A national trend of nutrition is toward a limitation of consumption of animal-sourced protein in the U.S. (Lau et al. Citation2023; USDA Citation2021). For example, annual beef consumption per capita by American adults aged 19–59 years had declined by 20.8% from 21.2 kg in 2005 to 16.8 kg in 2015 (Lau et al. Citation2023). This is due, in part, to an inadequate understanding of aging-related nutrition and metabolism of protein and AAs in humans. The objective of this review article is to fill in this critical gap of knowledge to beneficially mitigate sarcopenia in humans and improve their health and well-being.

Anabolic resistance of protein synthesis in skeletal muscle with aging

There are reports that basal rates of mixed or myofibrillar protein synthesis in skeletal muscle, which are measured in the overnight (or 12-h)-fasted state, either do not differ between young (a mean age of 20 to 28 years) and older (a mean age of 61 to 75 years) adults (Cuthbertson et al. Citation2005; Fry and Rasmussen Citation2011) or are 15–25% lower in older than in young adults (Welle et al. Citation1993; Yarasheski, Welle, and Nair Citation2002), depending on nutritional and physiological states. Short et al. (Citation2004) reported that the basal rates of mixed protein synthesis in the skeletal muscle of untrained men and women aged 19 – 87 years declined with advancing age at 3.5% per decade. However, results of most, but not all, studies have revealed that the response of skeletal muscle to dietary protein intake or resistance (strength) exercise in terms of mixed or myofibrillar protein synthesis is diminished by 20–35% with aging (Dorrens and Rennie Citation2003; Farnfield et al. Citation2012; Nair Citation1995; Paddon‐Jones and Rasmussen Citation2009; Wall et al. Citation2015). This phenomenon is termed the anabolic resistance of skeletal muscle with aging, likely because of impaired mechanistic target of rapamycin (mTOR) cell signaling (Cuthbertson et al. Citation2005), as well as reductions in the availability of certain AAs in the plasma and the uptake of AAs (e.g., Phe) by skeletal muscle (Katsanos et al. Citation2005). Insulin insensitivity in elderly men and women (a mean age of 69 years) is positively associated with elevated plasma concentrations of methylarginines (Marliss et al. Citation2006), which are inhibitors of the synthesis of nitric oxide (NO, the major vasodilator) from L-arginine in vascular endothelial cells (Wu et al. Citation2021). Thus, due to anabolic resistance with aging, elderly adults (e.g., 71 years of age) require 140% more protein intake (0.60 vs 0.25 g/kg body weight [BW] in a meal) to stimulate muscle protein synthesis at the same rate as that in young adults (Moore et al. Citation2015). Nonetheless, older adults can respond to elevated dietary protein intake with increases in protein synthesis and protein accretion in their skeletal muscle. Bauer et al. (Citation2013) recommended a daily intake of 1.0–1.2 g and ≥ 1.2 g protein/kg BW for older people (> 65 years of age) with minimum physical activity and regular exercise (endurance or resistance), respectively, to maintain or regain lean body mass and function.

Intake and digestion of dietary protein by adult humans

Based on food intake data from 11,680 adults [51–60 years of age (n = 4,016), 61–70 years of age (n = 3,854), and 71 years of age and older (n = 3,810)] from the 2005– 2014 National Health and Nutrition Examination Survey (NHANES), Krok-Schoen et al. (Citation2019) reported that a large proportion of U.S. adults did not meet the current Institute of Medicine (IOM)-recommended protein intake of 0.8 g/kg BW/day. Specifically, across all age categories, dietary protein intakes were decreased with age, with up to 50% of the oldest adults not meeting the protein intake recommendation due, in part, to poor food quality (e.g., inadequate amounts of animal-sourced food; Krok-Schoen et al. Citation2019). The percentages of men consuming < 0.8 g protein/kg BW/day are as follows: 51–60-year-old, 31%; 61–70-year-old, 37%; and > 70-year-old, 42%, and the percentages of women consuming < 0.8 g protein/kg BW/day are as follows: 51–60-year-old, 45%; 61–70-year-old, 48%; and > 70-year-old, 50% (Krok-Schoen et al. Citation2019). Based on the protein intake of 1.0–1.2 g/kg BW/day currently proposed by nutrition experts, even more adults in the U.S. do not have sufficient protein intake for optimum health including skeletal muscle health (Krok-Schoen et al. Citation2019). Likewise, dietary protein intakes of 29%, 54%, and 76% of community-dwelling older adults in the European union are below 0.8, 1.0, and 1.2 g/kg BW/day, respectively (Hengeveld et al. Citation2020).

Proteases in the lumen of the stomach, as well as proteases and peptidases in the lumen of the small intestine and on the apical membrane of its enterocytes are responsible for the hydrolysis of dietary proteins into small (di- and tri-) peptides and free AAs (Moughan and Rutherfurd Citation2012; Wu Citation2022). Aged adults have a sufficient ability to digest dietary protein and absorb the small peptides and free AAs, but the catabolism of AAs (e.g., branched-chain AAs) by the splanchnic bed (the portal-drained viscera plus liver) is greater in older than young adults (Boirie, Gachon, and Beaufrère Citation1997). Thus, given the same amount of ingested protein, less dietary AAs are available for use by extra-intestinal organs (including skeletal muscle, heart, kidneys, and brain) in elderly than in young adults. In support of this notion, Gorissen et al. (Citation2020) reported that 45% and 51% of phenylalanine in dietary protein entered the blood circulation in 71- and 22-year-old adults, respectively, during a 5-h postprandial period. This should be taken into consideration when recommending protein intakes for elderly adults to meet their requirements for AAs and prevent the development of sarcopenia during the aging process.

Concentrations of free AAs in the plasma (or serum) of young vs older adult humans

Because of insufficient protein intake and the reduced entry of dietary AAs into the blood circulation in elderly adults (see above), concentrations of AAs in their serum or plasma would be expected to be reduced, compared with young adults. This is indeed the case for most AAs, because the circulating levels of many AAs (including arginine, glycine, serine, branched-chain AAs, methionine, threonine, and tryptophan) are lower in elderly than young adults (). A decrease in the circulating level of arginine in older than young adults results, in part, from a lower rate of endogenous synthesis (Deutz et al. Citation2018), as reported for aged mice (Wu et al. Citation2022). Note that the circulating levels of AAs are regulated by multiple factors, including the dietary intake of AAs, intestinal and extraintestinal catabolism and synthesis of AAs, and whole-body synthesis and degradation of protein. Thus, elevations in plasma or serum concentrations of BCAAs in some older than in young adults (Caballero, Gleason, and Wurtman Citation1991; Menni et al. Citation2013) may not necessarily indicate an adequate intake of these AAs but rather may reflect a net increase in whole-body protein degradation in the former. To our knowledge, previous investigators did not determine 4-hydroxyproline (a major AA in the whole body) in the serum or plasma of humans. Interestingly, studies involving rodents (mice and rats) also indicated lower concentrations of some AAs (including arginine, glycine, 4-hydroxyproline, serine, branched-chain AAs, methionine, and tryptophan) in elderly than in younger animals (Supplemental Table 1) (Gross et al. Citation1991; Hartman et al. Citation1997; Houtkooper et al. Citation2011; Jeon et al. Citation2018; Seo et al. Citation2016; Suárez et al. Citation2016; Uchitomi et al. Citation2019; Varshavi et al. CitationCitation2018; and Yeung and Friedman Citation1991). Thus, elderly mammals such as humans are likely deficient in some AAs [including so-called nutritionally nonessential AAs (NEAAs; i.e., the AAs that are synthesizable de novo)] that are crucial for optimum health including skeletal muscle health, increasing risk for sarcopenia during aging.

Table 1. Changes in the concentrations of amino acids (AAs) in the serum or plasma of young versus older adult humans.

There were conflicting reports regarding changes in plasma AA concentrations between young and older humans possibly due to differences in ages, food intakes, or both among free-living study participants. For example, compared with young men (the mean age of 25 years) and women (the mean age of 26 years), respectively, among all the so-called nutritionally essential AAs (EAAs) analyzed, the post-absorptive (over-night fasting) plasma concentration of only tryptophan in older men (the mean age of 71 years) was decreased by 13%, and the post-absorptive plasma concentrations of only branched-chain AAs in older women (the mean age of 72 years) were increased by 20–22% (Caballero, Gleason, and Wurtman Citation1991). There were no changes in other EAAs between young and older individuals (Achermann and Kheim Citation1964). Changes in plasma concentrations of NEAAs (including arginine, glutamine, and serine) in young vs older adults were not reported (Caballero, Gleason, and Wurtman Citation1991), possibly due to the previous ignorance of the nutritional and physiological roles of NEAAs in humans. By contrast, Achermann and Kheim (Citation1964) found that the concentrations of the following AAs were lower in older men and women (the mean ages of 67 to 69 years) than those in young men and women (the mean ages of 30 to 32 years): alanine, −11%; arginine, −14%; branched-chain AAs, −13%; glycine, −11%; lysine, −17%; phenylalanine, −15%; serine, −18%; and tyrosine, −14%). These AAs are essential substrates for the syntheses of not only proteins but also other molecules [e.g., NO from arginine; glutathione from glycine; phosphatidylserine (a signaling molecule in the cell membrane) from serine; catecholamines and thyroid hormones from tyrosine; and creatine from arginine and glycine] with enormous physiological importance (Wu Citation2022). For example, these substances promote microvascular perfusion, anti-oxidative responses, DNA synthesis, fatty acid and glucose oxidation, energy provision, and physical contractions in skeletal muscle (Wu Citation2022). Chronic reductions in plasma AA concentrations [that can affect intracellular AA concentrations (Wu Citation2022)] and tissue sensitivity to insulin (Moore et al. Citation2015) may contribute to sarcopenia in adults. Some investigators continue to ignore the dietary intake (Church et al. Citation2020; Park et al. Citation2021; Pinckaers et al. Citation2023) or plasma concentrations (e.g., Church et al. Citation2020; Park et al. Citation2021; Pinckaers et al. Citation2022a) of NEAAs in aged humans.

Concentrations of free AAs in the skeletal muscle of young vs older adult humans

At present, little information is available regarding concentrations of AAs in the skeletal muscle of elderly vs young adult humans, possibly because of the previous notion that muscle protein synthesis is regulated by extracellular, but not intracellular, AAs in healthy young adults (Bohé et al. Citation2003). However, neither dietary intakes nor plasma/serum concentration of NEAAs in humans were determined in the study of Bohé et al. (Citation2003). Results of studies over the past 20 years have shown that some traditionally classified NEAAs (e.g., arginine, glutamine, and glycine) can activate the mTOR cell signaling to stimulate protein synthesis and inhibit protein breakdown in skeletal muscle cells (Wu Citation2022). Of particular note, free AAs in the intracellular pool [with their concentrations being affected by their transmembrane uptake and release, intracellular synthesis and catabolism, and intracellular proteolysis (Wu Citation2022)], rather than free AAs in the plasma or serum, are the immediate precursors for protein synthesis. In addition, free NEAAs (Ala, Arg, Asn, Asp, Cys, Glu, Gln, Gly, Pro, Ser, and Tyr) are much more abundant than free EAAs (His, Ile, Leu, Lys, Met, Phe, Thr, Trp, and Val) in the skeletal muscle of healthy adult humans, with the ratio (mol/mol) of free NEAAs to free EAAs being approximately 10:1 (Bergstrӧm et al. Citation1974). Thus, it is imperative that the intramuscular concentrations of all AAs (including both NEAAs and EAAs) be determined to provide insights into the availability of AAs for muscle protein synthesis.

We are aware of only one report of concentrations of most proteinogenic AAs in the skeletal muscle of elderly vs young adults (Gheller et al. Citation2021). These authors found that among all the measured proteinogenic AAs (albeit an incomplete list), only serine exhibited a reduced concentration in 60–80-year-old humans (n = 15) than 20–45-year-old adults (n = 43; Gheller et al. Citation2021). By contrast, a limited number of studies revealed lower intramuscular concentrations of some AAs (including β-alanine, glycine, histidine, 4-hydroxyproline, serine, threonine, and tryptophan) in old than in young rodents (Supplemental Table 1). Because intracellular AAs participate in multiple metabolic pathways including protein synthesis and its regulation, more studies should be conducted to determine intramuscular AAs in elderly vs young adults.

Concentrations of free AAs in the plasma (or serum) of sarcopenic vs nonsarcopenic adult humans

There are large variations in reports regarding changes in concentrations of plasma (or serum) AAs in sarcopenic vs non-sarcopenic adult humans (), likely due to differences in age, food intake, and health status. For example, compared with nonsarcopenic adults, concentrations of asparagine in the plasma (or serum) of sarcopenic adults were either increased (Calvani et al. Citation2018), decreased (Duan et al. Citation2023), or unchanged (Marques et al. Citation2023); concentrations of glutamine in the plasma (or serum) of sarcopenic adults were either decreased (Duan et al. Citation2023) or unchanged (Marques et al. Citation2023); and concentrations of taurine in the plasma (or serum) of sarcopenic adults were either increased (Calvani et al. Citation2018) or unchanged (Yeung et al. Citation2022). Nonetheless, most studies revealed that concentrations of branched-chain AAs, lysine, methionine, phenylalanine, threonine, tryptophan, and tyrosine were lower in the plasma (or serum) of sarcopenic than in non-sarcopenic elderly adults (). Interestingly, some of these changes occurred in a gender-specific manner, possibly due to differences in food patterns, hormones, and AA metabolism. For example, compared with nonsarcopenic adults, concentrations of leucine and valine were reduced in the plasma (or serum) of sarcopenic men and women; concentrations of threonine and tryptophan were reduced only in the plasma (or serum) of sarcopenic men, whereas concentrations of glutamate, histidine, isoleucine, methionine, phenylalanine, and proline were reduced only in the plasma (or serum) of sarcopenic women (Lo et al. Citation2021). Of note, most of these AAs are present in much lower amounts in plant- than animal-sourced foods (Li, He, and Wu Citation2021).

Table 2. Changes in the concentrations of amino acids in the serum or plasma of sarcopenic versus non-sarcopenic adult humans.

There are reports that the development of sarcopenia in adults is associated with dietary AA intake. For example, in adult humans, a low risk for sarcopenia is correlated with a high intake of BCAAs and tryptophan, whereas gait speed (an indicator of skeletal muscle function) is positively correlated with dietary intakes of all EAAs (Ma et al. Citation2022). By contrast, Ebrahimi-Mousavi et al. (Citation2022) reported no association between dietary intakes of BCAAs and odds of sarcopenia in adults (Ebrahimi-Mousavi et al. Citation2022). This discrepancy may be explained by differences in the dietary intakes of total protein and other nutrients (e.g., lipids and carbohydrates). However, no attention has been paid to dietary intakes of NEAAs by sarcopenic adults or the roles of these AAs in either modulating muscle protein synthesis (Church et al. Citation2020) or mitigating muscle loss and dysfunction in adults (Ma et al. Citation2022; van Vliet et al. Citation2015).

Acute effects of single administration of AAs on muscle protein metabolism in adult humans

In nutrition research, effects of bolus AA ingestion on muscle protein metabolism in people are often determined in combination with resistance- or endurance-type exercise. The latter stimulates both protein synthesis and protein breakdown in skeletal muscle, with a net effect on protein balance depending on dietary AA intake (Phillips Citation2012; Yang et al. Citation2012a). Amino acids are the building blocks of proteins, and some AAs (e.g., arginine, glutamine, glycine, and leucine) are activators of the mTOR cell signaling pathway to promote the initiation of protein synthesis in skeletal muscle (Wu Citation2022). In addition, arginine enhances the flow of blood and therefore the uptake of nutrients into skeletal muscle. Thus, single administration of 4 g leucine, a mix of 6 g BCAAs or their α-ketoacids, 6.7–40 g EAAs, or a mixture of 13–40 g EAAs plus some EAAs to young or older adults increased the rates of synthesis of mixed and myofibrillar proteins by skeletal muscle at rest and following resistance training (). Specifically, compared with the isonitrogenous control (alanine), ingestion of 4 g leucine along with a complete liquid diet over a 5-h period increased the rate of myofibrillar protein synthesis in skeletal muscle by 56% in elderly men (Rieu et al. Citation2006). Likewise, single oral administration of 2 g leucine to young men activated the mTOR cell signaling and increased muscle myofibrillar protein synthesis (Holowaty et al. Citation2023). Of note, intravenous or oral administration of a group of EAAs (defined differently by various research groups) or a mixture of EAAs plus some NEAAs for 2–4 h increases muscle protein synthesis in both young and elderly adults (Biolo et al. Citation1997; Bohé et al. Citation2001; Cuthbertson et al. Citation2005; Mitchell et al. Citation2015b; Tipton et al. Citation1999b). However, little is known about the response of skeletal muscle to administration of individual or a group of NEAAs in young or elderly adults. In contrast to data on protein synthesis, available evidence shows little change in the rates of breakdown of intramuscular mixed or myofibrillar proteins with aging in humans when expressed per kg fat-free lean mass (Rennie et al. Citation2010). Furthermore, there are reports that the rates of degradation of mixed proteins in skeletal muscle do not appear to be affected by intravenous or oral administration of a group of EAAs (Biolo et al. Citation1997; Tipton et al. Citation1999b). Collectively, a reduction in intramuscular protein synthesis appears to be primarily responsible for the loss of skeletal muscle protein during aging and can be a major therapeutic target.

Table 3. Acute effects of single administration of amino acids (AAs) or metabolites on skeletal muscle protein metabolism and function in young and/or older adult humansTable Footnotea.

NEAAs are more abundant than EAAs in the proteins of human skeletal muscle, with their ratio being 55 to 45 (g/g; Wu Citation2023). Growing evidence indicates important roles for NEAAs (e.g., arginine, glutamine and glycine) in enhancing protein synthesis in mammalian skeletal muscle cells in vitro and protein accretion in the skeletal muscle of experimental animals (e.g., pigs; Wu Citation2022). However, based on a study with young adults who showed a net positive nitrogen balance across skeletal muscle after receiving 2-h oral administration of eight EAAs and no change in intramuscular concentrations of all measured NEAAs, Tipton et al. (Citation1999b) concluded that NEAAs are not necessary to stimulate net muscle protein synthesis in healthy individuals. This conclusion, however, lacked direct evidence and should be reevaluated. The AA solution used by Tipton et al. (Citation1999b) lacked Trp (an EAA that is not synthesized by mammalian cells) and all NEAAs but human skeletal muscle could still respond to the ingestion of the 8 EAAs with an increase in protein synthesis during a period of 2 h. Clearly, such a result should not be interpreted to indicate that the dietary intake of Trp or NEAAs is not essential for maximizing net muscle protein synthesis in a longer term. In the case of Trp, a decrease in its dietary intake by healthy humans from 0.2 to 0.1 g per day for 5 days resulted in a negative nitrogen balance (Rose, Lambert, and Coon Citation1954). Likewise, both young and elderly adults responded positively to 3-h oral administration of a mixture of EAAs lacking lysine with an increase in muscle protein synthesis (Cuthbertson et al. Citation2005). This result does not negate an essential role for dietary Lys in muscle or whole-body protein synthesis during a longer period. Indeed, a decrease in the dietary intake of lysine by healthy humans from 1.2 to 0.6 g per day for 4 days resulted in a negative nitrogen balance (Rose et al. Citation1955). Thus, an acute increase in muscle protein synthesis in an absence of NEAAs or even some EAAs from a mixture of administered AAs does not mean that these AAs are not nutritionally or physiologically essential for this metabolic event in humans during a long-term period (e.g., weeks, months, and years). In support of such a notion, dietary supplementation with intact protein [e.g., whey protein providing both EAAs and NEAAs (Park, Choi, and Hwang Citation2018)], but not EAAs alone (Kim et al. Citation2012), for 3 months increased skeletal muscle mass and strength in older adults without regular exercise training. These results indicate that a sufficient provision of dietary NEAAs (e.g., arginine, glutamine and glycine; ) are crucial for the optimal responses of skeletal muscle metabolism and function to dietary EAAs in elderly individuals.

Table 5. Effects of short- and long-term supplementation with amino acids or their metabolites in young or older adult humansTable Footnotea.

Acute effects of single ingestion of protein on muscle protein metabolism in adult humans

Dietary proteins are classified as either “fast digestible” (e.g., whey protein), “slow digestible” (e.g., casein and proteins in well-cooked beef), or “intermediate digestible” (e.g., egg, soy, pea, and rice proteins) based on the speed of their digestion to release small peptides and AAs in the small intestine (Dangin et al. Citation2001, Citation2003; Bax et al. Citation2013). Compared with “slow digestible” proteins, the ingestion of “fast digestible” proteins usually results in more rapid appearance of AAs in the blood but a shorter period of elevated AA concentrations. Nonetheless, Park et al. (Citation2021) reported that the bolus ingestion of 57 g beef, 57 g pork, or 2 eggs (equivalent to ∼11 g protein) increased whole-body protein synthesis and protein balance to a similar extent in healthy young adults. This result indicates that these animal proteins are equivalent for stimulating protein synthesis and inhibit protein breakdown in adults.

Much evidence has shown that, in young, middle-aged, and older adults, the rates of synthesis of mixed and myofibrillar proteins by skeletal muscle at rest and following resistance training are increased in response to a single ingestion of 10–40 g of animal (e.g., whey, casein, egg, or beef) proteins (). In young adults, a maximal increase in muscle protein synthesis occurred at the dietary intake of 20 g animal-sourced protein (Moore et al. Citation2009). There are reports that a 100% increase in the plasma concentrations of EAAs is associated with a 34% increase in the rate of muscle protein synthesis in young and older adults consuming animal-protein supplements (Church et al. Citation2020). Because of sustained increases in the circulating levels of all AAs, a single ingestion of 30 g milk protein (providing 6 g BCAAs) augmented muscle protein synthesis in older adults for a longer period of time than ingestion of 6 g BCAAs or their α-ketoacids (e.g., 5 vs 2 h; Fuchs et al. Citation2019). In addition, both young and older adults responded to a single ingestion of plant-sourced proteins, such as 20–40 g soy protein (Yang et al. Citation2012b; Mitchell et al. Citation2015a), 30 g potato protein (Pinckaers et al. Citation2022b), 60 g wheat protein, or 30 g plant protein blend (15 g wheat protein, 7.5 g corn protein, and 7.5 g pea protein; Pinckaers et al. Citation2022a), with an increase in plasma concentrations of EAAs as well as the synthesis of mixed and myofibrillar proteins by skeletal muscle. However, in contrast to 35 g casein, a single ingestion of 35 g wheat protein did not affect protein synthesis in the skeletal muscle of older adults (Gorissen et al. Citation2016), suggesting that the composition of dietary AAs plays a crucial role in this metabolic pathway. In addition, compared with 113 g soy, the bolus ingestion of 113 g beef (isonitrogenous to soy) had a greater effect on the synthesis of myofibrillar proteins in the skeletal muscle of middle-aged (∼55 years of age) men at both rest and after resistance exercise (Phillips Citation2012). Furthermore, Holwerda et al. (Citation2019) reported that bolus co-ingestion of 15 g milk protein with 1.5 g leucine enhanced muscle protein synthesis following resistance exercise in older men, when compared with the consumption of the protein alone. Most recently, Pinckaers et al. (2023) found that the bolus ingestion of a beef-based meal as the primary source of protein (0.45 g protein/kg BW) stimulated muscle protein synthesis in older adults (65–85 years of age) to a greater extent than the consumption of an isocaloric and isonitrogenous vegan meal. Thus, an adequate amount of either animal or plant protein in diets can provide AAs to enhance muscle protein synthesis, but the effect appears to be greater with the intake of animal- than plant-sourced proteins.

Table 4. Acute effects of single ingestion of protein on skeletal muscle protein metabolism and function in young and/or older adult humans.

A combination of animal and plant proteins in diets is effective in increasing muscle protein synthesis in humans. The rates of muscle protein synthesis did not differ in young adults within a 5-h period after the consumption of (a) 30 g milk protein or 30 g potato protein (Pinckaers et al. Citation2022b); and (b) 30 g milk protein or 30 g plant protein blend (15 g wheat protein plus 7.5 g corn protein plus 7.5 g pea protein; Pinckaers et al. Citation2022a). Likewise, rates of muscle protein synthesis in young adults (men and women) did not differ between the consumption of 19 g soy plus casein plus whey proteins and 18 g whey protein during the first 2 h after ingestion, but the effect of 19 g soy plus casein plus whey proteins was greater than that of 18 g whey protein during the last 2 h after ingestion (Reidy et al. Citation2013). Thus, a combination of whey, casein and soy protein can extend postprandial muscle protein synthesis for a longer period than whey protein alone, possibly due to a better balance of AAs (including arginine, methionine, and cysteine) in the mixture of animal and plant proteins. Interestingly, an adequate amount of animal and plant proteins has an equal efficacy in supporting protein synthesis for a few hours in the skeletal muscle of adult despite differences in the content of some AAs. For example, the content of the following EAAs was much lower in the plant protein blend than in milk protein (based on percentages of decrease): Ile, 33%; Lys, 65%; Met, 43%; Thr, 22%; and Val, 36% (Pinckaers et al. Citation2022a). The content of Trp is expected to differ substantially between the animal- and plant-sourced proteins (Li, He, and Wu Citation2021), but this AA was not analyzed by Pinckaers et al. (Citation2022b, Citation2022a). These results indicate that some EAAs in diets may not be the major factors affecting protein metabolism within hours in the skeletal muscle of humans.

Effects of short- and long-term supplementation with AAs or their metabolites on skeletal muscle protein metabolism and health in adult humans

Results of extensive research has documented the beneficial effects of AAs and other nutrients in improving skeletal muscle health in the elderly (). They include total EAAs, individual or a mixture of BCAAs (particularly leucine), some individual NEAAs (e.g., arginine, glutamine and citrulline), a mixture of NEAAs (e.g., arginine plus glutamine; glycine plus N-acetyl-cysteine), AA metabolites [e.g., β-hydroxy-β-methylbutyrate (HMB, a metabolite of leucine), branched-chain α-ketoacids, taurine, β-alanine, anserine, carnosine, glutathione, creatine and carnitine], vitamins (e.g., vitamins C, D and E), and minerals (e.g., calcium, magnesium and selenium). For example, as reported for a single ingestion, oral daily administration of a mix of 7.2 g BCAAs (5 wk), 6.7–22 g EAAs (8–18 wk), 3 g EAAs with 800 IU vitamin D plus 6 g medium-chain triglyceride (3 months), or 10 g glutamine (for 30 days) increased lean muscle mass and muscle function (indicated by handgrip strength) in older adults, including sarcopenic individuals (). Similar results were reported for dietary supplementation with 3 g HMB plus 14 g arginine plus 14 g glutamine; for 6 months); 0.1 g glycine plus 0.132 g N-acetylcysteine/kg BW/day (for 24 wk), 3 g arginine plus antioxidants per day (for 6 wk), 3–10 g citrulline malate/day (an immediate precursor of arginine; for 3–6 wk), 1.6–3.2 g β-alanine/day (a precursor of carnosine; for 4–12 wk), and AA metabolites (). The latter include creatine monohydrate (0.1 g/kg BW/day for 32 wk, 0.3 g/kg BW/day for 7 days, or 0.08 g/kg BW/day for 12 wk), 10–20 g/day creatine monohydrate (for 2 wk), 3 g/day calcium-HMB (for 10 days to 42 wk), 1.3–3 g/day HMB/day (for 24 wk), or 2–3 g HMB plus 5–7.5 g arginine plus 1.5–2.25 g lysine per day (for 12 months). There is evidence that the concentration of HMB in the plasma of elderly people is much lower than that for young adults due to reduced leucine catabolism (Duetz et al. 2018). Notably, oral administration of 1.5 g/day taurine to adult humans for 16 wk enhanced the antioxidant capacity and health of skeletal muscle (Abud et al. Citation2022). Similarly, acute or chronic ingestion of 1 to 6 g taurine/day improved skeletal muscle strength and overall endurance performance in adult humans (Waldron et al. Citation2018). Among the AA metabolites with beneficial effects on skeletal muscle, taurine and creatine are abundant in meat (e.g., beef) but are absent from plant-sourced foods, whereas β-alanine is abundant in meat but is negligible in plant-sourced foods (Wu Citation2020). This indicates a major difference in nutritional values between animal- and plant-sourced foods for humans.

As noted previously, the single ingestion of leucine acutely stimulated muscle protein synthesis in humans (including older adults; Rieu et al. Citation2006). Similar results have been reported for the oral administration of leucine daily (3 × 4 g/day) for 2 wk (Casperson et al. Citation2012). However, long-term dietary supplementation with leucine (3 × 2.5 g/day) to older men with or without type-2 diabetes for 3–6 months had no effect on skeletal muscle mass or strength, whole-body insulin sensitivity, or plasma lipid concentrations (Verhoeven et al. Citation2009; Leenders et al. Citation2011). This indicates that the acute effects of a supplemental AA may not predict its long-term impacts on metabolism or health because protein balance in skeletal muscle depends on not only the rate of its protein synthesis but also the rate of its protein degradation. Of particular note, the rates of intramuscular protein turnover are regulated by a plethora of factors, including endocrine status, dietary intakes of AAs and other nutrients, physical activity, and ambient temperatures (Wu Citation2022). Because no single experiment has examined the roles of all the proteinogenic AAs (individual or mixtures) and related metabolites in mitigating human sarcopenia, available evidence does not allow us to provide any weighting of their importance. Nonetheless, the published studies underscore the nutritional and physiological significance of consuming well-balanced diets [consisting of whole foods of both animal and plant origins] that provide adequate amounts of all the needed nutrients.

Effects of short- and long-term supplementation with protein on skeletal muscle protein metabolism and health in adult humans

A short-term (e.g., 6-day) dietary supplementation with protein (15–42 g/day) enhanced muscle protein synthesis in older adults (Devries et al. Citation2018) but was not effective in alleviating their muscle loss (Dirks et al. Citation2014). By contrast, long-term supplementation with whey or milk protein (10–50 g/day) or the combination of milk and soy proteins increased lean body mass, as well as skeletal muscle mass and strength, while reducing white-fat mass in older adults (). In addition, co-ingestion with milk proteins (13–22 g) with 1.2–4 g leucine plus 120–800 IU vitamin D3 per day, 20 g mixed proteins plus 800 IU vitamin D3 plus 300 mg calcium per day, or 11 g casein plus 11 g whey protein plus 7 g BCAAs plus 206 mg ursolic acid plus 432 IU vitamin D3 per day, for 30 days to 9 months, increased skeletal muscle mass and strength in older adults (including sarcopenic patients) (). Similar results were reported for sarcopenic women consuming milk plus EAA (–His) supplement for 3 months (Kim et al. Citation2012). Although whey protein had a greater effect on promoting muscle protein synthesis than casein (), these two types of protein had the same efficacy in increasing skeletal muscle mass and strength in adult humans during a period of 8 weeks (Wilborn et al. Citation2013). A meta-analysis of randomized controlled trials involving 1063 adults (≥ 18 years of age) revealed that in comparison with a low-fat diet with a standard amount of protein (providing 12–18% of energy), the consumption of a low-fat diet with a high amount of protein (providing 25–35% of energy) for a mean duration of 12.1 wk (≥ 4 wk) promoted the mobilization of fat tissue and reduced the loss of fat-free mass including muscle (Wycherley et al. Citation2012).

Table 6. Effects of short- and long-term supplementation with protein in young or older adult humansTable Footnotea.

Aubertin-Leheudre et al. (2009)

Table 7. Effects of short- and long-term supplementation with protein + amino acids + other nutrients on older adult humansTable Footnotea.

Several lines of evidence indicate that long-term supplementation with milk protein has a greater effect than soy protein to sustain skeletal muscle mass in adult humans (). First, compared with the same amount of soy protein, daily dietary supplementation with 17.5 g milk protein (including casein and whey) to young men during a 12-week resistance exercise program increased lean body mass by 1.1 kg (Hartman et al. Citation2007). Second, dietary supplementation with 24 g whey protein per day to young men during a 36-week resistance exercise program enhanced lean tissue gain by 1.5 kg than those consuming 24 g soy protein (Volek et al. Citation2013). Third, long-term consumption of diets containing both animal- and plant-sourced proteins for a mean of 12 years resulted in a 4.4 kg greater lean body mass in middle-aged women, compared with the consumption of plant-sourced foods without animal protein (Aubertin-Leheudre and Adlercreutz Citation2009). Fourth, consumption of meat-containing omnivorous diets contributed to greater gains in skeletal muscle mass in older men during a 12-week resistance training program, compared with meat-free lactoovovegetarian diets (Campbell et al. Citation1999). Fifth, in most of the home-bound elderly, consuming < 65% of total protein from animal-sourced foods results in the deficiency of at least one EAA (based on the recommended requirement of 0.8 g protein/kg BW/day) and therefore protein undernutrition (Dasgupta, Sharkey, and Wu Citation2005). Although there are differences in nutritional quality between animal- and plant-sourced proteins, they should be provided in appropriate amounts and proportions to meet the dietary requirements of humans for all AAs and to prevent an excessive intake of some AAs (e.g., methionine and cysteine) from animal protein alone.

Older adults have higher requirements for dietary protein than currently recommended to maintain muscle mass and mitigate sarcopenia

As noted previously, AAs not only activate the mTOR cell signaling required for the initiation of mRNA translation but also serve as the building blocks of proteins and the precursors of antioxidative molecules in tissues including skeletal muscle (Dillon Citation2013; Wu Citation2022). Multiple AA-related factors likely contribute to sarcopenia in adults, including reductions in protein intake, plasma AA concentrations, tissue sensitivity to insulin, muscle protein synthesis, the endothelial synthesis of NO from arginine, and blood flow to skeletal muscle (Campbell et al. Citation2001, Citation2002; Cereda et al. Citation2022; Cruz-Jentoft et al. Citation2019; Lonnie et al. Citation2018; Murton Citation2015). In support of this view, aging is associated with the impairment of NO-dependent blood flow into skeletal muscle (Delp et al. Citation2008; Dillon Citation2013) and that either physical exercise or dietary arginine supplementation enhances the flow of blood (and thus AAs and other nutrients) to skeletal muscle in humans including 70-year-old men (Mitchell et al.0.2017; Wu et al. Citation2021). In addition, there is a relationship between sarcopenia and physical activity-associated blood flow, as exercise (aerobic or resistance training) can improve microvascular perfusion in skeletal muscle and can alleviate sarcopenia (Zhang et al. Citation2022). Furthermore, compelling evidence shows that effective mitigation of sarcopenia in adult humans requires protein intake that is at least 50% greater than the current recommended value (i.e., 0.8 g protein/kg BW/day; Paddon-Jones and Rasmussen Citation2009). Thus, elderly individuals consuming 1.2 g protein/kg BW/day lost 40% less lean body mass over a 3-year period than those ingesting 0.8 g protein/kg BW/day (Houston et al. Citation2008). Likewise, older persons (≥ 65 years of age) with minimal physical activity required 1.2 g of high-quality protein/kg BW/day to improve the strength and function of skeletal muscle (Morais, Chevalier, and Gougeon Citation2006). For elderly persons who are capable of exercise, sufficient intakes of AAs have a synergistic effect with appropriate physical activity to maintain or increase skeletal muscle mass and strength, which can be an effective countermeasure to sarcopenia (Aas et al. Citation2020). Adults with physical frailty who are not able to perform regular resistance training may also benefit from an increased intake of high-quality protein to alleviate muscle loss (Morley et al. Citation2010). At present, it is unknown whether all or certain AAs are responsible for the beneficial effects of dietary protein in mitigating the progression of muscle loss in adults. This knowledge is important for designing precise nutrition for adults because a chronic high intake of dietary protein would increase the whole-body production of ammonia and homocysteine (risk factors for renal disorders, brain damage and cardiovascular disease).

High risk for sarcopenia in vegans and vegetarians

Worldwide, less than 10% of the population are vegans (consuming no animal-sourced foods) and vegetarians (consuming dairy products and eggs but no other animal-sourced foods), and more than 90% of the population are omnivores. Based on a survey of people (aged 18 to 64 years in Canada and the United States but 16 to 64 years in other countries) in 2018, globally 3% and 5% of them were vegans and vegetarians, respectively, and 97% of them consumed animal products; in the United States, the same (3%) percentages of adults were vegans and vegetarians, and 94% of adults consumed animal products (Ipsos Citation2018). A large-scale investigation involving 30,251 generally health-conscious adults (79% women and 21% men) in the United Kingdom revealed that dietary protein intakes by vegans and vegetarians were 23% and 19% lower, respectively than those for meat eaters (Sobiecki et al. Citation2016 Supplemental Table 2). There is evidence that the incidence of sarcopenia in vegetarians is much higher than that in meat eaters (20% vs 6.2%, respectively; Wang et al. Citation2023). Similarly, vegan diets may increase risks for sarcopenia in older adults (Domić et al. Citation2022). Consequently, compared with meat eaters, vegetarians had higher risks for hip (+25%) and arm (+25%) fractures, whereas vegans had much higher risks for not only hip (+231%) and arm (+56%) fractures but also leg (+205%) and vertebral (+59%) fractures (Tong et al. 2020).

Roles of animal vs plant protein intakes in maintaining skeletal muscle mass and alleviating sarcopenia in older humans

Results of a recent systematic review and meta-analysis reveal a positive association between healthy dietary patterns and the maintenance of gait speed with age (an intermediate marker of sarcopenia risk) in adult humans (Van Elswyk et al. Citation2022). There is also interest in the role of dietary animal- and plant-sourced foods in maintaining skeletal muscle mass and mitigating human sarcopenia (Alexandrov et al. Citation2018; Bradlee et al. Citation2018; FAO Citation2007; Mariotti and Gardner Citation2019; Valenzuela et al. Citation2019; Nunes et al. Citation2022; van Vliet et al. Citation2015). Recent articles have highlighted experimental evidence indicating that animal-sourced foods are more effective than plant-sourced foods in improving muscle health and reducing risk for sarcopenia in adults (Bradlee et al. Citation2018; Domić et al. Citation2022; Tong et al. Citation2020). For example, in studying the effect of animal vs plant proteins with or without physical activity in older adults (both men and women) over a mean duration of 9 years, Bradlee et al. (Citation2018) found that the intake of animal proteins (a mixture of beef, lamb, pork, poultry, fish, and dairy) resulted in a greater lean mass and a lower risk of functional decline (and thus sarcopenia), whereas the benefits of an equal amount of plant proteins (a mixture of legumes, soy, nuts, and seeds) were only visible in those who performed regular exercise.

Because eggs provide high quality protein which stimulates muscle protein synthesis, Smith and Gray (Citation2016) have proposed that consumption of eggs can help mitigate sarcopenia in older humans. In support of this view, results of a randomized trial indicated that the consumption of a high-protein diet (1.4 g protein/kg BW/day) consisting of three whole eggs per day for 12 wk preserved skeletal-muscle mass in older adults (a mean of 71 years of age) with overweight or obesity, when compared with an isocaloric diet (1800 kcal/day) providing 0.8 g protein/kg BW/day (Wright et al. Citation2018). In addition, dietary supplementation with dried egg white (20 g protein/day) for 6 months improved upper body physical function in elder women (a mean of 74 years of age), in comparison with the isocaloric maltodextrin supplement (Ullevig et al. Citation2022). Most recently, Okamura et al. (Citation2023) reported that consumption of ≥ 60 g egg/day resulted in a greater skeletal-muscle mass in elderly men with obesity and type-2 diabetes, as compared with control individuals ingesting < 60 g egg/day.

Consumption of fish has been suggested to improve muscle health and prevent in older people (Alexandrov et al. Citation2018; Leroy et al. Citation2023). This is likely but experimental data are lacking to date. Here, we use beef as a prototype of functional meat to reduce risk for sarcopenia in humans because of the following two reasons. First, among meats, beef is the only food with the known composition of all proteinogenic AAs and their functional metabolites (e.g., taurine, β-alanine, 4-hydroxyproline, anserine, carnosine, and glutathione; Wu et al. Citation2016). Dietary supplementation with these substances improves skeletal muscle strength and exercise performance in elderly adults (Waldron et al. Citation2019; Wu Citation2020). At present, there is no direct evidence for each of these AA derivatives in preventing or alleviating human sarcopenia. However, Eshima et al. (Citation2023) reported that dietary carnosine supplementation to aging mice for 2 wk (1 week of pretreatment and 1 week during hindlimb unloading) enhanced skeletal muscle mass and strength.

Second, there are published studies regarding the effects of beef consumption in mitigating sarcopenia in older people, but we are not aware of such work involving pork or poultry meat (). Specifically, beef is an abundant source of all proteinogenic AAs in both adequate amounts and balanced ratios, while also providing physiologically essential nonproteinogenic AAs (taurine and β-alanine; Wu et al. Citation2016) as well as vitamins (e.g., vitamins B6 and B12) and minerals (e.g., iron, zinc, and selenium; Daly et al. Citation2015; Leroy Citation2023; Klurfeld Citation2024; Wu et al. Citation2014). The content and bioavailability of proteinogenic AAs in beef are greater than those in plant-sourced foods. In most of the studies involving effects of meat (e.g., beef) consumption on human health, their authors did not take, into consideration, bioactive and protective compounds (e.g., taurine, 4-hydroxyproline, creatine, and glutathione) present in whole foods (Leroy et al. Citation2022; Sekhar et al. Citation2011; Turner and Lloyd Citation2017). Taurine (a potent antioxidant) is essential for the integrity and functions of tissues, including skeletal muscle, heart and eyes, whereas β-alanine is required for the production of antioxidative and neuromodulatory dipeptides (carnosine and anserine). Furthermore, beef contains a large amount of creatine (essential for energy metabolism in tissues, particularly skeletal muscle and brain), as well as 4-hydroxyproline (a major precursor of glycine; Wu et al. Citation2016). There are myths that plants provide all nutrients that are available in animal-sourced foods. However, taurine, vitamin B12, creatine, carnosine, and anserine are absent from plants, whereas β-alanine and 4-hydroxyproline are low or negligible in plants (Hou et al. Citation2019; Wu Citation2022). Thus, the bolus ingestion of ground beef (0, 57, 113, or 170 g) by middle-aged (59-year-old) men rapidly increased myofibrillar protein synthesis in their skeletal muscle in a dose-dependent manner, and the effect of beef was further enhanced with resistance exercise (Robinson et al. Citation2013).

Table 8. Effects of long-term supplementation with beef on maintaining skeletal muscle mass and alleviating sarcopenia in young and older humans.

Third, long-term supplementation with beef to adult humans enhances their nutritional status, lean body mass, lower-limb muscle strength, and the duration of moderate physical activity (). For example, daily dietary supplementation with 46 g beef protein to young adults for 8 wk along with regular resistance exercise increased lean body mass and skeletal muscle mass, while reducing whole-body fat mass (Sharp et al. Citation2018). Similarly, daily dietary supplementation with 45 g beef protein to older women for 4 months along with regular resistance exercise increased lean body mass and skeletal muscle mass, while reducing the circulating level of interleukin-6 (an indicator of inflammation) in older women (Daly et al. Citation2014). Likewise, in older men and women (60–88 years of age), there were positive correlations between beef intake (1.1 to 109 g raw beef/day) and skeletal muscle mass (Asp et al. Citation2012). Furthermore, a higher protein intake from beef (30 g lean beef three times/day) improved physical strength and function in frail and obese older adults (Porter Starr et al. Citation2016). Thus, increasing the consumption of beef may provide an effective nutritional means to mitigate sarcopenia in adults.

Some epidemiological studies raised concern that the consumption of red meat (including beef) might increase risks for obesity, type 2 diabetes mellitus, and cardiovascular disease in humans (Willett et al. Citation2019). Note that 130-g raw beef contains ∼7.7 g lipids (Wu et al. Citation2016), which provide only 3.5% of the daily energy (2000 kcal) needed by a 70-kg adult. Lipids are an important part of a healthy diet for humans, and the consumption of some fatty acids (e.g., oleic acid and conjugated fatty acids) in beef increases high-density lipoproteins (HDL; “good cholesterol”) and decrease low-density lipoproteins (LDL; “bad cholesterol”) in the blood of men and women (Smith et al. Citation2020). Recently, Lytle et al. (Citation2023) reported that the weekly consumption of five 115-g ground beef patties (containing either 5.6% or 23.6% fat, on a fresh basis; prepared from the beef pectoralis muscle) for 5 wk did not negatively affect vascular function in healthy adult men with a mean age of 40 years. Furthermore, compared with the 5.6%-fat patty group, study participants in the 23.6%-fat patty group had 11% greater flow-mediated dilation in the forearm and 3.7% lower resting systolic blood pressure (Lytle et al. Citation2023), possibly due to the enhanced production of NO by endothelial cells (Wu et al. Citation2021). Thus, an appropriate amount of meat (e.g., beef) can contribute to an AA-balanced, healthy diet for humans.

Conclusions

With the US population growing older, sarcopenia is increasing in prevalence. The older population suffers from an insufficient intake of protein (especially animal-sourced protein) and insulin resistance. Insufficient protein intake leads to lower concentrations of several AAs that are important for skeletal muscle metabolism and health. In this review, we systematically and comprehensively summarized, for the first time to our knowledge, nutritional research related to (1) concentrations of free AAs in the plasma (or serum) and skeletal muscle of young vs older adult humans, (2) acute effects of single ingestion of AAs or protein on muscle protein metabolism in adult humans, (3) effects of short- and long-term supplementation with AAs (or their functional metabolites) on skeletal muscle protein metabolism and health in adult humans, (4) higher requirements for dietary protein in older people than currently recommended to maintain muscle mass and mitigate sarcopenia, and (5) roles of animal vs plant protein intakes in maintaining skeletal muscle mass and alleviating sarcopenia in older humans. Specifically, BCAAs were found to be lower in sarcopenic adults compared to non-sarcopenic adults. High dietary AA intake is correlated with lower sarcopenia risk. Administration of a mixture of BCAAs and EAAs following resistance training was found to increase rates of protein synthesis by skeletal muscle. Rates of protein synthesis are also increased by ingesting 10–40 grams of animal proteins. Much evidence shows that long-term dietary supplementation with animal and plant sourced proteins can enhance muscle protein synthesis in older adults, but both should be provided appropriately. In addition, most of the work cited herein is based on single dietary protein in acute or short-term studies but humans normally do not consume a single source of dietary protein (certainly not for vegetarians) and the results of such studies may not indicate long-term outcomes. Overall, older adults may benefit from an increase in the consumption of high-quality protein such as beef to combat their risk of sarcopenia. Furthermore, we highlighted discrepancies in the measured variables among the published studies and the need to determine intramuscular concentrations of AAs in young vs older adults. Finally, we proposed that adult humans require both EAAs and NEAAs in diets for maintaining muscle protein mass and function and, therefore, for mitigating the age-related sarcopenia. The integrated findings and new thoughts of this article are expected to guide further research for improving muscle health in older individuals.

Authors' contributions

GW designed and supervised this writing project. WH, EDC, and GW wrote the manuscript. HRC and GW contributed to manuscript revisions. All authors read and approved the final manuscript.

Supplemental material

Supplemental Material

Download MS Word (35.8 KB)

Acknowledgments

We thank Dr. Shalene H. McNeill and Dr. Clara Lau-Rosenow at National Cattlemen's Beef Association (Centennial, CO) for helpful discussion on the impacts of dietary protein intakes on human nutrition and health.

Disclosure statement

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Additional information

Funding

This writing project was supported by the National Cattlemen's Beef Association. The funder had no influence on the preparation of the paper.

References

  • Aas, S. N., O. Seynnes, H. B. Benestad, and T. Raastad. 2020. Strength training and protein supplementation improve muscle mass, strength, and function in mobility-limited older adults: A randomized controlled trial. Aging Clinical and Experimental Research 32 (4):605–16. doi:10.1007/s40520-019-01234-2.
  • Abe, S., O. Ezaki, and M. Suzuki. 2016. Medium-chain triglycerides in combination with leucine and Vitamin D increase muscle strength and function in frail elderly adults in a randomized controlled trial. The Journal of Nutrition 146 (5):1017–26. doi:10.3945/jn.115.228965.
  • Abud, G. F., G. De Carvalho, S. G. Batitucci, C. R. Travieso, F. Bueno Junior, J. S. Barbosa Junior, E. C. Marchini, and de Freitas, F. G. 2022. Taurine as a possible antiaging therapy: A controlled clinical trial on taurine antioxidant activity in women ages 55 to 70. Nutrition 101:111706. doi:10.1016/j.nut.2022.111706.
  • Ackermann, P. G., and T. Kheim. 1964. Plasma amino acids in young and older adult human subjects. Clinical Chemistry 10 (1):32–40. doi:10.1093/clinchem/10.1.32.
  • Aguiar, A. F., R. S. B. Januário, R. P. Junior, A. M. Gerage, F. L. C. Pina, M. A. do Nascimento, C. R. Padovani, and E. S. Cyrino. 2013. Long-term creatine supplementation improves muscular performance during resistance training in older women. European Journal of Applied Physiology 113 (4):987–96. doi:10.1007/s00421-012-2514-6.
  • Alexandrov, N. V., C. Eelderink, C. M. Singh-Povel, G. J. Navis, S. J. L. Bakker, and E. Corpeleijn. 2018. Dietary protein sources and muscle mass over the life course: The lifelines cohort study. Nutrients 10 (10):1471. doi:10.3390/nu10101471.
  • Almeida, E. B., J. M. B. Santos, V. Paixão, J. B. Amaral, R. Foster, A. Sperandio, T. Roseira, M. Rossi, T. G. Cordeiro, F. R. Monteiro, et al. 2020. L-glutamine supplementation improves the benefits of combined-exercise training on oral redox balance and inflammatory status in elderly individuals. Oxidative Medicine and Cellular Longevity 2020:1–13. doi:10.1155/2020/2852181.
  • Amirato, G. R., J. O. Borges, D. L. Marques, J. M. B. Santos, C. A. F. Santos, M. S. Andrade, G. E. Furtado, M. Rossi, L. N. Luis, R. F. Zambonatto, et al. 2021. L-glutamine supplementation enhances strength and power of knee muscles and improves glycemia control and plasma redox balance in exercising elderly women. Nutrients 13 (3):1025. doi:10.3390/nu13031025.
  • Asp, M. L., J. R. Richardson, A. L. Collene, K. R. Droll, and M. A. Belury. 2012. Dietary protein and beef consumption predict for markers of muscle mass and nutrition status in older adults. Journal of Nutrition, Health & Aging 16 (9):784–90. doi:10.1007/s12603-012-0064-6.
  • Aubertin-Leheudre, M., and H. Adlercreutz. 2009. Relationship between animal protein intake and muscle mass index in healthy women. British Journal of Nutrition 102 (12):1803–10. doi:10.1017/S0007114509991310.
  • Baier, S., D. Johannsen, N. Abumrad, J. A. Rathmacher, S. Nissen, and P. Flakoll. 2009. Year-long changes in protein metabolism in elderly men and women supplemented with a nutrition cocktail of beta-hydroxy-beta-methylbutyrate (HMB), L-arginine, and L-lysine. Journal of Parenteral and Enteral Nutrition 33 (1):71–82. doi:10.1177/0148607108322403.
  • Barichella, M., E. Cereda, G. Pinelli, L. Iorio, D. Caroli, I. Masiero, V. Ferri, E. Cassani, C. Bolliri, S. Caronni, et al. 2019. Muscle-targeted nutritional support for rehabilitation in patients with parkinsonian syndrome. Neurology 93 (5):e485–96. doi:10.1212/WNL.0000000000007858.
  • Bauer, J., G. Biolo, T. Cederholm, M. Cesari, A. J. Cruz-Jentoft, J. E. Morley, S. Phillips, C. Sieber, P. Stehle, D. Teta, et al. 2013. Evidence-based recommendations for optimal dietary protein intake in older people: A position paper from the P ROT-AGE Study Group. Journal of the American Medical Directors Association 14 (8):542–59. doi: 10.1016/j.jamda.2013.05.021.
  • Bauer, J. M., S. Verlaan, I. Bautmans, K. Brandt, L. M. Donini, M. Maggio, M. E. McMurdo, T. Mets, C. Seal, S. L. Wijers, et al. 2015. Effects of a vitamin D and leucine-enriched whey protein nutritional supplement on measures of sarcopenia in older adults, the provide study: A randomized, double-blind, placebo-controlled trial. Journal of the American Medical Directors Association 16 (9):740–7. doi:10.1016/j.jamda.2015.05.021.
  • Bax, M. L., C. Buffière, N. Hafnaoui, C. Gaudichon, I. Savary-Auzeloux, D. Dardevet, V. Santé-Lhoutellier, and D. Rémond. 2013. Effects of meat cooking, and of ingested amount on protein digestion speed and entry of residual proteins into the colon: A study in minipigs. PLoS ONE. 8 (4):e61252. doi:10.1371/journal.pone.0061252.
  • Bergström, J., P. Fürst, L. O. Norée, and E. Vinnars. 1974. Intracellular free amino acid concentration in human muscle tissue. Journal of Applied Physiology 36 (6):693–7. doi:10.1152/jappl.1974.36.6.693.
  • Bernat, P., D. G. Candow, K. Gryzb, S. Butchart, B. J. Schoenfeld, and P. Bruno. 2019. Effects of high-velocity resistance training and creatine supplementation in untrained healthy aging males. Applied Physiology, Nutrition, and Metabolism 44 (11):1246–53. doi:10.1139/apnm-2019-0066.
  • Biolo, G., K. D. Tipton, S. Klein, and R. R. Wolfe. 1997. An abundant supply of amino acids enhances the metabolic effect of exercise on muscle protein. The American Journal of Physiology 273 (1 Pt 1):E122–9. doi:10.1152/ajpendo.1997.273.1.E122.
  • Bohé, J., J. F. A. Low, R. R. Wolfe, and M. J. Rennie. 2001. Latency and duration of stimulation of human muscle protein synthesis during continuous infusion of amino acids. The Journal of Physiology 532 (2):575–9. doi:10.1111/j.1469-7793.2001.0575f.x.
  • Bohé, J., A. Low, R. R. Wolfe, and M. J. Rennie. 2003. Human muscle protein synthesis is modulated by extracellular, not intramuscular amino acid availability: A dose-response study. The Journal of Physiology 552 (Pt 1):315–24. doi:10.1113/jphysiol.2003.050674.
  • Boirie, Y., P. Gachon, and B. Beaufrère. 1997. Splanchnic and whole-body leucine kinetics in young and elderly men. The American Journal of Clinical Nutrition 65 (2):489–95. doi:10.1093/ajcn/65.2.489.
  • Bologna, C., and E. Pone. 2022. Clinical study on the efficacy and safety of arginine administered orally in association with other active ingredients for the prevention and treatment of sarcopenia in patients with COVID-19-related pneumonia, hospitalized in a sub-intensive care unit. Healthcare 10 (1):162. doi:10.3390/healthcare10010162.
  • Borack, M. S., P. T. Reidy, S. H. Husaini, M. M. Markofski, R. R. Deer, A. B. Richison, B. S. Lambert, M. B. Cope, R. Mukherjea, K. Jennings, et al. 2016. Soy-dairy protein blend or whey protein isolate ingestion induces similar postexercise muscle mechanistic target of rapamycin complex 1 signaling and protein synthesis responses in older men. The Journal of Nutrition 146 (12):2468–75. doi:10.3945/jn.116.231159.
  • Børsheim, E., Q. U. Bui, S. Tissier, H. Kobayashi, A. A. Ferrando, and R. R. Wolfe. 2008. Effect of amino acid supplementation on muscle mass, strength and physical function. in elderly. Clinical Nutrition 27 (2):189–95. doi:10.1016/j.clnu.2008.01.001.
  • Bouillanne, O., J.-C. Melchior, C. Faure, M. Paul, F. Canouï-Poitrine, Y. Boirie, D. Chevenne, C. Forasassi, E. Guery, S. Herbaud, et al. 2019. Impact of 3-week citrulline supplementation on postprandial protein metabolism in malnourished older patients: The ciproage randomized controlled trial. Clinical Nutrition 38 (2):564–74. doi:10.1016/j.clnu.2018.02.017.
  • Bradlee, M. L., J. Mustafa, M. R. Singer, and L. L. Moore. 2018. High-protein foods and physical activity protect against age-related muscle loss and functional decline. The Journals of Gerontology: Series A 73 (1):88–94. doi:10.1093/gerona/glx070.
  • Burd, N. A., Y. Yang, D. R. Moore, J. E. Tang, M. A. Tarnopolsky, and S. M. Phillips. 2012. Greater stimulation of myofibrillar protein synthesis with ingestion of whey protein isolate v. micellar casein at rest and after resistance exercise in elderly men. British Journal of Nutrition 108 (6):958–62. doi:10.1017/S0007114511006271.
  • Caballero, B., R. E. Gleason, and R. J. Wurtman. 1991. Plasma amino acid concentrations in healthy elderly men and women. The American Journal of Clinical Nutrition 53 (5):1249–52. doi:10.1093/ajcn/53.5.1249.
  • Caballero-García, A., J. Pascual-Fernández, D. C. Noriega-González, H. J. Bello, A. Pons-Biescas, E. Roche, and A. Córdova-Martínez. 2021. L-citrulline supplementation and exercise in the management of sarcopenia. Nutrients 13 (9):3133. doi:10.3390/nu13093133.
  • Calvani, R., A. Picca, F. Marini, A. Biancolillo, J. Gervasoni, S. Persichilli, A. Primiano, H. Coelho-Junior, M. Bossola, A. Urbani, et al. 2018. A distinct pattern of circulating amino acids characterizes older persons with physical frailty and sarcopenia: Results from the BIOSPHERE study. Nutrients 10 (11):1691. doi:10.3390/nu10111691.
  • Campbell, W. W., M. L. Barton, Jr, D. Cyr-Campbell, S. L. Davey, J. L. Beard, G. Parise, and W. J. Evans. 1999. Effects of an omnivorous diet compared with a lactoovovegetarian diet on resistance-training-induced changes in body composition and skeletal muscle in older men. The American Journal of Clinical Nutrition 70 (6):1032–9. doi:10.1093/ajcn/70.6.1032.
  • Campbell, W. W., T. A. Trappe, R. R. Wolfe, and W. J. Evans. 2001. The recommended dietary allowance for protein may not be adequate for older people to maintain skeletal muscle. Journals of Gerontology A 56:M373–80.
  • Campbell, W. W., T. A. Trappe, A. C. Jozsi, L. J. Kruskall, R. R. Wolfe, and W. J. Evans. 2002. Dietary protein adequacy and lower body versus whole body resistive training in older humans. The Journal of Physiology 542 (2):631–42. doi:10.1113/jphysiol.2002.020685.
  • Candow, D. G., J. P. Little, P. D. Chilibeck, S. Abeysekara, G. A. Zello, M. Kazachkov, S. M. Cornish, and P. H. Yu. 2008. Low-dose creatine combined with protein during resistance training in older men. Medicine & Science in Sports & Exercise 40 (9):1645–52. doi:10.1249/MSS.0b013e318176b310.
  • Candow, D. G., E. Vogt, S. Johannsmeyer, S. C. Forbes, and J. P. Farthing. 2015. Strategic creatine supplementation and resistance training in healthy older adults. Applied Physiology, Nutrition, and Metabolism 40 (7):689–94. doi:10.1139/apnm-2014-0498.
  • Candow, D. G., P. D. Chilibeck, S. C. Forbes, C. M. Fairman, B. Gualano, and H. Roschel. 2022. Creatine supplementation for older adults: Focus on sarcopenia, osteoporosis, frailty and cachexia. Bone 162:116467. doi:10.1016/j.bone.2022.116467.
  • Casperson, S. L., M. Sheffield-Moore, S. J. Hewlings, and D. Paddon-Jones. 2012. Leucine supplementation chronically improves muscle protein synthesis in older adults consuming the RDA for protein. Clinical Nutrition 31 (4):512–9. doi:10.1016/j.clnu.2012.01.005.
  • Cereda, E., R. Pisati, M. Rondanelli, R. and Caccialanza. 2022. Whey protein, leucine- and Vitamin-D-enriched oral nutritional supplementation for the treatment of sarcopenia. Nutrients 14:1524.
  • Chak, C., M. Lacruz, J. Adam, S. Brandmaier, M. Covic, J. Huang, C. Meisinger, D. Tiller, C. Prehn, J. Adamski, et al. 2019. Ageing investigation using two-time-point metabolomics data from KORA and CARLA studies. Metabolites 9 (3):44. doi:10.3390/metabo9030044.
  • Chanet, A., S. Verlaan, J. Salles, C. Giraudet, V. Patrac, V. Pidou, C. Pouyet, N. Hafnaoui, A. Blot, N. Cano, et al. 2017. Supplementing breakfast with a Vitamin D and leucine-enriched whey protein medical nutrition drink enhances postprandial muscle protein synthesis and muscle mass in healthy older men. The Journal of Nutrition 147 (12):2262–71. doi:10.3945/jn.117.252510.
  • Church, D. D., K. R. Hirsch, S. Park, I.-Y. Kim, J. A. Gwin, S. M. Pasiakos, R. R. Wolfe, and A. A. Ferrando. 2020. Essential amino acids and protein synthesis: Insights into maximizing the muscle and whole-body response to feeding. Nutrients 12 (12):3717. doi:10.3390/nu12123717.
  • Córdova-Martínez, A., A. Caballero-García, H. J. Bello, A. Pons-Biescas, D. C. Noriega, and E. Roche. 2022. L-arginine and beetroot extract supplementation in the prevention of sarcopenia. Pharmaceuticals 15 (3):290. doi:10.3390/ph15030290.
  • Costa Riela, N. d A., M. M. Alvim Guimarães, D. Oliveira de Almeida, and E. M. Q. Araujo. 2021. Effects of beta-hydroxy-beta-methylbutyrate supplementation on elderly body composition and muscle strength: A review of clinical trials. Annals of Nutrition and Metabolism 77 (1):16–22. doi:10.1159/000514236.
  • Cruz-Jentoft, A. J., G. Bahat, J. Bauer, Y. Boirie, O. Bruyère, T. Cederholm, C. Cooper, F. Landi, Y. Rolland, A. A. Sayer, et al. 2019. Sarcopenia: Revised European consensus on definition and diagnosis. Age and Ageing 48 (1):16–31. doi:10.1093/ageing/afy169.
  • Cuthbertson, D., K. Smith, J. Babraj, G. Leese, T. Waddell, P. Atherton, H. Wackerhage, P. M. Taylor, and M. J. Rennie. 2005. Anabolic signaling deficits underlie amino acid resistance of wasting, aging muscle. The FASEB Journal 19 (3):1–22. doi:10.1096/fj.04-2640fje.
  • Daley, M. J., and W. L. Spinks. 2000. Exercise mobility and aging. Sports Medicine 29 (1):1–12. doi:10.2165/00007256-200029010-00001.
  • Daly, R. M., S. L. O'Connell, N. L. Mundell, C. A. Grimes, D. W. Dunstan, and C. A. Nowson. 2014. Protein-enriched diet, with the use of lean red meat, combined with progressive resistance training enhances lean tissue mass and muscle strength and reduces circulating IL-6 concentrations in elderly women: A cluster randomized controlled trial. The American Journal of Clinical Nutrition 99 (4):899–910. doi:10.3945/ajcn.113.064154.
  • Daly, R. M., J. Gianoudis, M. Prosser, D. Kidgell, K. A. Ellis, S. O'Connell, and C. A. Nowson. 2015. The effects of a protein enriched diet with lean red meat combined with a multi-modal exercise program on muscle and cognitive health and function in older adults: Study protocol for a randomised controlled trial. Trials 16 (1):339. doi:10.1186/s13063-015-0884-x.
  • Dangin, M., Y. Boirie, C. Garcia-Rodenas, P. Gachon, J. Fauquant, P. Callier, O. Ballèvre, and B. Beaufrère. 2001. The digestion rate of protein is an independent regulating factor of postprandial protein retention. American Journal of Physiology. Endocrinology and Metabolism 280 (2):E340–8. doi:10.1152/ajpendo.2001.280.2.E340.
  • Dangin, M., C. Guillet, C. Garcia-Rodenas, P. Gachon, C. Bouteloup-Demange, K. Reiffers-Magnani, J. Fauquant, O. Ballèvre, and B. Beaufrère. 2003. The rate of protein digestion affects protein gain differently during aging in humans. The Journal of Physiology 549 (2):635–44. doi:10.1113/jphysiol.2002.036897.
  • Darst, B. F., R. L. Koscik, K. J. Hogan, S. C. Johnson, and C. D. Engelman. 2019. Longitudinal plasma metabolomics of aging and sex. Aging 11 (4):1262–82. doi:10.18632/aging.101837.
  • Dasgupta, M., J. R. Sharkey, and G. Wu. 2005. Inadequate intakes of indispensable amino acids among homebound older adults. Journal of Nutrition for the Elderly 24 (3):85–99. doi:10.1300/J052v24n03_07.
  • del Favero, S., H. Roschel, M. Y. Solis, A. P. Hayashi, G. G. Artioli, M. C. Otaduy, F. B. Benatti, R. C. Harris, J. A. Wise, C. C. Leite, et al. 2012. Beta-alanine (CarnosynTM) supplementation in elderly subjects (60–80 years): Effects on muscle carnosine content and physical capacity. Amino Acids. 43 (1):49–56. doi:10.1007/s00726-011-1190-x.
  • Delp, M. D., B. J. Behnke, S. A. Spier, G. Wu, and J. M. Muller-Delp. 2008. Aging diminishes endothelium-dependent vasodilation and tetrahydrobiopterin content in rat skeletal muscle arterioles. The Journal of Physiology 586 (4):1161–8. doi:10.1113/jphysiol.2007.147686.
  • Deutz, N. E., S. L. Pereira, N. P. Hays, J. S. Oliver, N. K. Edens, C. M. Evans, and R. R. Wolfe. 2013. Effect of beta-hydroxy-beta-methylbutyrate (HMB) on lean body mass during 10 days of bed rest in older adults. Clinical Nutrition 32 (5):704–12. doi:10.1016/j.clnu.2013.02.011.
  • Deutz, N. E. P., J. J. Thaden, G. A. M. Ten Have, D. K. Walker, and M. P. K. J. Engelen. 2018. Metabolic phenotyping using kinetic measurements in young and older healthy adults. Metabolism 78:167–78. doi:10.1016/j.metabol.2017.09.015.
  • Devries, M. C., C. McGlory, D. R. Bolster, A. Kamil, M. Rahn, L. Harkness, S. K. Baker, and S. M. Phillips. 2018. Protein leucine content is a determinant of shorter- and longer-term muscle protein synthetic responses at rest and following resistance exercise in healthy older women: A randomized, controlled trial. The American Journal of Clinical Nutrition 107 (2):217–26. doi:10.1093/ajcn/nqx028.
  • Dillon, E. L. 2013. Nutritionally essential amino acids and metabolic signaling in aging. Amino Acids 45 (3):431–41. doi:10.1007/s00726-012-1438-0.
  • Dillon, E. L., M. Sheffield-Moore, D. Paddon-Jones, C. Gilkison, A. P. Sanford, S. L. Casperson, J. Jiang, D. L. Chinkes, and R. J. Urban. 2009. Amino acid supplementation increases lean body mass, basal muscle protein synthesis, and insulin-like growth factor-I expression in older women. The Journal of Clinical Endocrinology and Metabolism 94 (5):1630–7. doi:10.1210/jc.2008-1564.
  • Dimori, S., G. Leoni, L. Fior, and F. Gasparotto. 2018. Clinical nutrition and physical rehabilitation in a long-term care setting: Preliminary observations in sarcopenic older patients. Aging Clinical and Experimental Research 30 (8):951–8. doi:10.1007/s40520-017-0859-8.
  • Dirks, M. L., B. T. Wall, R. Nilwik, D. H. Weerts, L. B. Verdijk, and L. J. C. van Loon. 2014. Skeletal muscle disuse atrophy is not attenuated by dietary protein supplementation in healthy older men. The Journal of Nutrition 144 (8):1196–203. doi:10.3945/jn.114.194217.
  • Domić, J., P. Grootswagers, L. J. C. van Loon, and L. C. P. G. M. de Groot. 2022. Vegan diets for older adults? A perspective on the potential impact on muscle mass and strength. Advances in Nutrition 13 (3):712–25. doi:10.1093/advances/nmac009.
  • Dorrens, J., and M. J. Rennie. 2003. Effects of ageing and human whole body and muscle protein turnover. Scandinavian Journal of Medicine & Science in Sports 13 (1):26–33. doi:10.1034/j.1600-0838.2003.00306.x.
  • D'Souza, R. F., J. F. Markworth, V. C. Figueiredo, P. A. Della Gatta, A. C. Petersen, C. J. Mitchell, and D. Cameron-Smith. 2014. Dose-dependent increases in p70S6K phosphorylation and intramuscular branched-chain amino acids in older men following resistance exercise and protein intake. Physiological Reports 2 (8):e12112. doi:10.14814/phy2.12112.
  • Duan, Y., K. Tao, Z. Fang, and Y. Lu. 2023. Possible-sarcopenic screening with disturbed plasma amino acid profile in the elderly. BMC Geriatrics 23 (1):427. doi:10.1186/s12877-023-04137-0.
  • Duda, K., J. Majerczak, Z. Nieckarz, S. B. Heymsfield, and J. A. Zoladz. 2019., Human body composition and muscle mass. In Muscle and exercise physiology, J. A. Zoladz, 3–26. San Diego, CA: Academic Press.
  • Dunn, W. B., W. Lin, D. Broadhurst, P. Begley, M. Brown, E. Zelena, A. A. Vaughan, A. Halsall, N. Harding, J. D. Knowles, et al. 2014. Molecular phenotyping of a UK population: Defining the human serum metabolome. Metabolomics 11 (1):9–26. doi:10.1007/s11306-014-0707-1.
  • Ebrahimi-Mousavi, S., R. Hashemi, A. Bagheri, R. Heshmat, A. Dorosty-Motlagh, and A. Esmaillzadeh. 2022. Association between dietary intake of branched-chain amino acids and sarcopenia and its components: A cross-sectional study. Scientific Reports 12 (1):5666. doi:10.1038/s41598-022-07605-6.
  • Ellis, A., G. Hunter, A. Goss, and B. Gower. 2018. Effects of oral supplementation with beta-hydroxy-beta-methylbutyrate, arginine, and glutamine on lean body mass of healthy older adults. Journal of Dietary Supplements 19:1–13.
  • Eshima, H., J. L. Shahtout, P. Siripoksup, M. J. Pearson, Z. S. Mahmassani, P. J. Ferrara, A. W. Lyons, J. A. Maschek, A. D. Peterlin, A. R. P. Verkerke, et al. 2023. Lipid hydroperoxides promote sarcopenia through carbonyl stress. eLife 12:E85289. doi: 10.7554/eLife.85289.
  • FAO (Food and Agriculture Organization of the United Nations). 2007. Protein and amino acid requirements in human nutrition. Report of a Joint WHO/FAO/UNU Expert Consultation. Rome, Italy.
  • Farnfield, M. M., L. Breen, K. A. Carey, A. Garnham, and D. Cameron-Smith. 2012. Activation of mTOR signalling in young and old human skeletal muscle in response to combined resistance exercise and whey protein ingestion. Applied Physiology, Nutrition, and Metabolism 37 (1):21–30. doi:10.1139/h11-132.
  • Formica, M. B., J. Gianoudis, C. A. Nowson, S. L. O'Connell, C. Milte, K. A. Ellis, and R. M. Daly. 2020. Effect of lean red meat combined with a multicomponent exercise program on muscle and cognitive function in older adults: A 6-month randomized controlled trial. The American Journal of Clinical Nutrition 112 (1):113–28. doi:10.1093/ajcn/nqaa104.
  • Foroumandi, E., M. Alizadeh, and S. Kheirouri. 2018. Age-dependent changes in plasma amino acids contribute to alterations in glycoxidation products. Journal of Medical Biochemistry 37 (4):426–33. doi:10.1515/jomb-2017-0065.
  • Francaux, M., B. Demeulder, D. Naslain, R. Fortin, O. Lutz, G. Caty, and L. Deldicque. 2016. Aging reduces the activation of the mTORC1 pathway after resistance exercise and protein intake in human skeletal muscle: Potential role of REDD1 and impaired anabolic sensitivity. Nutrients 8 (1):47. doi:10.3390/nu8010047.
  • Fry, C. S., and B. B. Rasmussen. 2011. Skeletal muscle protein balance and metabolism in the elderly. Current Aging Sciencee 4 (3):260–8. doi:10.2174/1874609811104030260.
  • Fuchs, C. J., W. J. H. Hermans, A. M. Holwerda, J. S. J. Smeets, J. M. Senden, J. van Kranenburg, A. P. Gijsen, W. K. H. W. Wodzig, H. Schierbeek, L. B. Verdijk, et al. 2019. Branched-chain amino acid and branched-chain ketoacid ingestion increases muscle protein synthesis rates in vivo in older adults: A double-blind, randomized trial. The American Journal of Clinical Nutrition 110 (4):862–72. doi:10.1093/ajcn/nqz120.
  • Furst, T., A. Massaro, C. Miller, B. T. Williams, Z. M. LaMacchia, and P. J. Horvath. 2018. β-Alanine supplementation increased physical performance and improved executive function following endurance exercise in middle aged individuals. Journal of the International Society of Sports Nutrition 15 (1):32. doi:10.1186/s12970-018-0238-7.
  • Gheller, B. J., J. E. Blum, E. W. Lim, M. K. Handzlik, E. H. Hannah Fong, A. C. Ko, S. Khanna, M. E. Gheller, E. L. Bender, M. S. Alexander, et al. 2021. Extracellular serine and glycine are required for mouse and human skeletal muscle stem and progenitor cell function. Molecular Metabolism 43:101106. doi:10.1016/j.molmet.2020.101106.
  • Glynn, E. L., C. S. Fry, M. J. Drummond, K. L. Timmerman, S. Dhanani, E. Volpi, and B. B. Rasmussen. 2010. Excess leucine intake enhances muscle anabolic signaling but not net protein anabolism in young men and women. The Journal of Nutrition 140 (11):1970–6. doi:10.3945/jn.110.127647.
  • Gorissen, S. H., A. M. Horstman, R. Franssen, J. J. Crombag, H. Langer, J. Bierau, F. Respondek, and L. J. C. Van Loon. 2016. Ingestion of wheat protein increases in vivo muscle protein synthesis rates in healthy older men in a randomized trial. The Journal of Nutrition 146 (9):1651–9. doi:10.3945/jn.116.231340.
  • Gorissen, S. H. M., J. Trommelen, I. W. K. Kouw, I. W. K. Kouw, B. Pennings, B. Pennings, B. T. Wall, T. A. Churchward-Venne, A. M. H. Horstman, R. Koopman, et al. 2020. Protein type, protein dose, and age modulate dietary protein digestion and phenylalanine absorption kinetics and plasma phenylalanine availability in humans. The Journal of Nutrition 150 (8):2041–50. doi:10.1093/jn/nxaa024.
  • Gotshalk, L. A., J. S. Volek, R. S. Staron, C. R. Denegar, F. C. Hagerman, and W. J. Kraemer. 2002. Creatine supplementation improves muscular performance in older men. Medicine & Science in Sports & Exercise 34 (3):537–43. doi:10.1097/00005768-200203000-00023.
  • Gotshalk, L. A., W. J. Kraemer, M. A. G. Mendonca, J. L. Vingren, A. M. Kenny, B. A. Spiering, D. L. Hatfield, M. S. Fragala, and J. S. Volek. 2008. Creatine supplementation improves muscular performance in older women. European Journal of Applied Physiology 102 (2):223–31. doi:10.1007/s00421-007-0580-y.
  • Griffen, C., M. Duncan, J. Hattersley, M. O. Weickert, A. Dallaway, and D. Renshaw. 2022. Effects of resistance exercise and whey protein supplementation on skeletal muscle strength, mass, physical function, and hormonal and inflammatory biomarkers in healthy active older men: A randomised, double-blind, placebo-controlled trial. Experimental Gerontology 158:111651. doi:10.1016/j.exger.2021.111651.
  • Grootswagers, P., E. Smeets, A. B. Oteng, and L. Groot. 2021. A novel oral nutritional supplement improves gait speed and mitochondrial functioning compared to standard care in older adults with (or at risk of) undernutrition: Results from a randomized controlled trial. Aging 13 (7):9398–418. doi:10.18632/aging.202912.
  • Gross, K. L., W. J. Hartman, A. Ronnenberg, and R. L. Prior. 1991. Arginine-deficient diets alter plasma and tissue amino acids in young and aged rats. The Journal of Nutrition 121 (10):1591–9. doi:10.1093/jn/121.10.1591.
  • Hartman, W. J., E. Seyoum, R. Villalobos-Molina, J. A. Joseph, and R. L. Prior. 1997. Responses of circulating urea cycle and branched-chain amino acids to feeding in adult and aged Fischer-344 rats. Aging Clinical and Experimental Research 9 (3):198–206. doi:10.1007/BF03340150.
  • Hartman, J. W., J. E. Tang, S. B. Wilkinson, M. A. Tarnopolsky, R. L. Lawrence, A. V. Fullerton, and S. M. Phillips. 2007. Consumption of fat-free fluid milk after resistance exercise promotes greater lean mass accretion than does consumption of soy or carbohydrate in young, novice, male weightlifters. The American Journal of Clinical Nutrition 86 (2):373–81. doi:10.1093/ajcn/86.2.373.
  • Hengeveld, L. M., J. M. A. Boer, P. Gaudreau, M. W. Heymans, C. Jagger, N. Mendonça, M. C. Ocké, N. Presse, S. Sette, E. M. Simonsick, et al. 2020. Prevalence of protein intake below recommended in community-dwelling older adults: A meta-analysis across cohorts from the PROMISS consortium. Journal of Cachexia, Sarcopenia and Muscle 11 (5):1212–22. doi:10.1002/jcsm.12580.
  • Holowaty, M. N. H., M. J. Lees, S. Abou Sawan, K. J. M. Paulussen, R. Jäger, M. Purpura, S. A. Paluska, N. A. Burd, N. Hodson, and D. R. Moore. 2023. Leucine ingestion promotes mTOR translocation to the periphery and enhances total and peripheral RPS6 phosphorylation in human skeletal muscle. Amino Acids. 55 (2):253–61. doi:10.1007/s00726-022-03221-w.
  • Holwerda, A. M., K. J. M. Paulussen, M. Overkamp, J. P. B. Goessens, I. F. Kramer, W. K. W. H. Wodzig, L. B. Verdijk, L. C. P. G. M. de Groot, and L. J. C. van Loon. 2019. Leucine coingestion augments the muscle protein synthetic response to the ingestion of 15 g of protein following resistance exercise in older men. American Journal of Physiology 317:E473–82.
  • Hou, Y. Q., W. L. He, S. D. Hu, and G. Wu. 2019. Composition of polyamines and amino acids in plant-source foods for human consumption. Amino Acids. 51 (8):1153–65. doi:10.1007/s00726-019-02751-0.
  • Houston, D. K., B. J. Nicklas, J. Ding, T. B. Harris, F. A. Tylavsky, A. B. Newman, J. S. Lee, N. R. Sahyoun, M. Visser, and S. B. Kritchevsky. 2008. Dietary protein intake is associated with lean mass change in older, community-dwelling adults: The Health, Ageing, and Body Composition (Health ABC) Study. The American Journal of Clinical Nutrition 87 (1):150–5. doi:10.1093/ajcn/87.1.150.
  • Houtkooper, R. H., C. Argmann, S. M. Houten, C. Cantó, E. H. Jeninga, P. A. Andreux, C. Thomas, R. Doenlen, K. Schoonjans, and J. Auwerx. 2011. The metabolic footprint of aging in mice. Scientific Reports 1 (1):134. doi:10.1038/srep00134.
  • Ipsos. 2018. An exploration into diets around the world. https://www.ipsos.com/sites/default/files/ct/news/documents/2018-09/an_exploration_into_diets_around_the_world.pdf
  • Ispoglou, T., H. White, T. Preston, S. McElhone, J. McKenna, and K. Hind. 2016. Double-blind, placebo-controlled pilot trial of L-leucine-enriched amino-acid mixtures on body composition and physical performance in men and women aged 65-75 years. European Journal of Clinical Nutrition 70 (2):182–8. doi:10.1038/ejcn.2015.91.
  • Jackman, S. R., G. A. Wallis, J. Yu, A. Philp, K. Baar, K. D. Tipton, and O. C. Witard. 2023. Co-ingestion of branched-chain amino acids and carbohydrate stimulates myofibrillar protein synthesis following Resistance exercise in trained young men. International Journal of Sport Nutrition and Exercise Metabolism 33 (4):189–97. doi:10.1123/ijsnem.2023-0015.
  • Janssen, I., S. B. Heymsfield, Z. Wang, and R. Ross. 2000. Skeletal muscle mass and distribution in 468 men and women aged 18–88 yr. Journal of Applied Physiology 89 (1):81–8. doi:10.1152/jappl.2000.89.1.81.
  • Jeon, J. S., J. J. Oh, H. C. Kwak, H. Y. Yun, H. C. Kim, Y. M. Kim, S. J. Oh, and S. K. Kim. 2018. Age-related changes in sulfur amino acid metabolism in male C57BL/6 mice. Biomolecules & Therapeutics 26 (2):167–74. doi:10.4062/biomolther.2017.054.
  • Kang, Y., N. Kim, Y. J. Choi, Y. Lee, J. Yun, S. J. Park, H. S. Park, Y. S. Chung, and Y. K. Park. 2020. Leucine-enriched protein supplementation increases lean body mass in healthy korean adults aged 50 years and older: A randomized, double-blind, placebo-controlled trial. Nutrients 12 (6):1816. doi:10.3390/nu12061816.
  • Katsanos, C. S., H. Kobayashi, M. Sheffield-Moore, A. Aarsland, and R. R. Wolfe. 2005. Aging is associated with diminished accretion of muscle proteins after the ingestion of a small bolus of essential amino acids. The American Journal of Clinical Nutrition 82 (5):1065–73. doi:10.1093/ajcn/82.5.1065.
  • Katsanos, C. S., H. Kobayashi, M. Sheffield-Moore, A. Aarsland, and R. R. Wolfe. 2006. A high proportion of leucine is required for optimal stimulation of the rate of muscle protein synthesis by essential amino acids in the elderly. American Journal of Physiology 291:E381–7.
  • Kim, H. K., T. Suzuki, K. Saito, H. Yoshida, H. Kobayashi, H. Kato, and M. Katayama. 2012. Effects of exercise and amino acid supplementation on body composition and physical function in community-dwelling elderly Japanese sarcopenic women: A randomized controlled trial. Journal of the American Geriatrics Society 60 (1):16–23.
  • Kiskini, A., H. M. Hamer, B. T. Wall, B. B. Groen, A. de Lange, J. A. Bakker, J. M. Senden, L. B. Verdijk, and L. J. C. van Loon. 2013. The muscle protein synthetic response to the combined ingestion of protein and carbohydrate is not impaired in healthy older men. Age 35 (6):2389–98. doi:10.1007/s11357-013-9522-2.
  • Klurfeld, D. M. 2024. The whole food beef matrix is more than the sum of its parts. Critical Reviews in Food Science and Nutrition 64 (14):4523–31. doi: 10.1080/10408398.2022.2142931.
  • Ko, C. H., S. J. Wu, S. T. Wang, Y. F. Chang, C. S. Chang, T. S. Kuan, H. Y. Chuang, C. M. Chang, W. Chou, and C. H. Wu. 2020. Effects of enriched branched-chain amino acid supplementation on sarcopenia. Aging 12 (14):15091–103. doi:10.18632/aging.103576.
  • Koopman, R., L. Verdijk, R. J. F. Manders, A. P. Gijsen, M. Gorselink, E. Pijpers, A. J. M. Wagenmakers, and L. J. C. van Loon. 2006. Co-ingestion of protein and leucine stimulates muscle protein synthesis rates to the same extent in young and elderly lean men. The American Journal of Clinical Nutrition 84 (3):623–32. doi:10.1093/ajcn/84.3.623.
  • Kouchiwa, T., K. Wada, M. Uchiyama, N. Kasezawa, M. Niisato, H. Murakami, K. Fukuyama, and H. Yokogoshi. 2012. Age-related changes in serum amino acids concentrations in healthy individuals. Clinical Chemistry and Laboratory Medicine 50 (5):861–70. doi:10.1515/cclm-2011-0846.
  • Kramer, I. F., L. B. Verdijk, H. M. Hamer, S. Verlaan, Y. Luiking, I. W. K. Kouw, J. M. Senden, J. van Kranenburg, A. P. Gijsen, M. Poeze, et al. 2015. Impact of the macronutrient composition of a nutritional supplement on muscle protein synthesis rates in older men: A randomized, double blind, controlled trial. The Journal of Clinical Endocrinology & Metabolism 100 (11):4124–32. doi:10.1210/jc.2015-2352.
  • Kramer, I. F., L. B. Verdijk, H. M. Hamer, S. Verlaan, Y. C. Luiking, I. W. K. Kouw, J. M. Senden, J. van Kranenburg, A. P. Gijsen, J. Bierau, et al. 2017. Both basal and post-prandial muscle protein synthesis rates, following the ingestion of a leucine-enriched whey protein supplement, are not impaired in sarcopenic older males. Clinical Nutrition (Edinburgh, Scotland) 36 (5):1440–9. doi:10.1016/j.clnu.2016.09.023.
  • Krok-Schoen, J. L., A. Archdeacon Price, M. Luo, O. J. Kelly, and C. A. Taylor. 2019. Low dietary protein intakes and associated dietary patterns and functional limitations in an aging population: A NHANES analysis. Journal of Nutrition, Health & Aging 23 (4):338–47. doi:10.1007/s12603-019-1174-1.
  • Kumar, P., C. Liu, J. W. Hsu, S. Chacko, C. Minard, F. Jahoor, and R. Sekhar. 2021. Glycine and N-acetylcysteine (GlyNAC) supplementation in older adults improves glutathione deficiency, oxidative stress, mitochondrial dysfunction, inflammation, insulin resistance, endothelial dysfunction, genotoxicity, muscle strength, and cognition: Results of a pilot clinical trial. Clinical and Translational Medicine 11 (3):e372. doi:10.1002/ctm2.372.
  • Lancha, A. H., Jr, R. Zanella, Jr, S. G. Tanabe, M. Andriamihaja, and F. Blachier. 2017. Dietary protein supplementation in the elderly for limiting muscle mass loss. Amino Acids. 49 (1):33–47. doi:10.1007/s00726-016-2355-4.
  • Larsson, L., H. Degens, M. Li, L. Salviati, Y. I. Lee, W. Thompson, J. L. Kirkland, and M. Sandri. 2019. Sarcopenia: Aging-related loss of muscle mass and function. Physiological Reviews 99 (1):427–511.
  • Lau, C. S., V. L. Fulgoni, I. I. I. M. E. Van Elswyk, and S. H. McNeill. 2023. Trends in beef intake in the United States: Analysis of the National Health and Nutrition Examination Survey, 2001–2018. Nutrients 15 (18):2475–91. doi: 10.3390/nu15112475.
  • Leenders, M., L. B. Verdijk, L. van der Hoeven, J. van Kranenburg, F. Hartgens, W. K. W. H. Wodzig, W. H. M. Saris, and L. J. C. van Loon. 2011. Prolonged leucine supplementation does not augment muscle mass or affect glycemic control in elderly type 2 diabetic men. The Journal of Nutrition 141 (6):1070–6. doi:10.3945/jn.111.138495.
  • Leroy, F., F. Abraini, T. Beal, P. Dominguez-Salas, P. Gregorini, P. Manzano, J. Rowntree, and S. van Vliet. 2022. Animal source foods in healthy, sustainable, and ethical diets – An argument against drastic limitation of livestock in the food system. Animal 16 (3):100457. doi:10.1016/j.animal.2022.100457.
  • Leroy, F., N. W. Smith, A. T. Adesogan, T. Beal, L. Iannotti, P. J. Moughan, and N. Mann. 2023. The role of meat in the human diet: Evolutionary aspects and nutritional value. Animal Frontiers 13 (2):11–8. doi:10.1093/af/vfac093.
  • Liberman, K., R. Njemini, Y. Luiking, L. N. Forti, S. Verlaan, J. M. Bauer, R. Memelink, K. Brandt, L. M. Donini, M. Maggio, et al. 2019. Thirteen weeks of supplementation of vitamin D and leucine-enriched whey protein nutritional supplement attenuates chronic low-grade inflammation in sarcopenic older adults: The PROVIDE study. Aging Clinical and Experimental Research 31 (6):845–54. doi:10.1007/s40520-019-01208-4.
  • Li, P., W. L. He, and G. Wu. 2021. Composition of amino acids in foodstuffs for humans and animals. Advances in Experimental Medicine and Biology 1332:189–210.
  • Lin, C. C., M. H. Shih, C. D. Chen, and S. L. Yeh. 2021. Effects of adequate dietary protein with whey protein, leucine, and vitamin D supplementation on sarcopenia in older adults: An open-label, parallel-group study. Clinical Nutrition 40 (3):1323–9. doi:10.1016/j.clnu.2020.08.017.
  • Lo, C. J., Y. S. Ko, S. W. Chang, H. Y. Tang, C. Y. Huang, Y. C. Huang, H. Y. Ho, C. M. Lin, and M. L. Cheng. 2021. Metabolic signatures of muscle mass loss in an elderly Taiwanese population. Aging 13 (1):944–56. doi:10.18632/aging.202209.
  • Lonnie, M., E. Hooker, J. M. Brunstrom, B. M. Corfe, M. A. Green, A. W. Watson, E. A. Williams, E. J. Stevenson, S. Penson, and A. M. Johnstone. 2018. Protein for life: Review of optimal protein intake, sustainable dietary sources and the effect on appetite in ageing adults. Nutrients 10 (3):360. doi:10.3390/nu10030360.
  • Lu, Y., L. G. Karagounis, T. P. Ng, C. Carre, V. Narang, G. Wong, C. T. Y. Tan, M. S. Zin Nyunt, Q. Gao, B. Abel, et al. 2020. Systemic and metabolic signature of sarcopenia in community-dwelling older adults. Journals of Gerontology A 75:309–17.
  • Luiking, Y. C., N. E. P. Deutz, R. G. Memelink, S. Verlaan, and R. R. Wolfe. 2014. Postprandial muscle protein synthesis is higher after a high whey protein, leucine-enriched supplement than after a dairy-like product in healthy older people: A randomized controlled trial. Nutrition Journal 13 (1):9. doi:10.1186/1475-2891-13-9.
  • Lytle, J. R., S. T. Stanelle, S. E. Martin, S. B. Smith, D. R. Smith, and S. F. Crouse. 2023. Assessment of vascular function in response to high-fat and low-fat ground beef consumption in men. Nutrients 15 (6):1410. doi:10.3390/nu15061410.
  • Ma, W.-J., W. Zhang, J. Zhai, H. Li, T. Zhao, L. Han, Q. Wen, X. Song, and X.-F. Guo. 2022. Branched chain and other amino acid intakes are inversely associated with sarcopenia among community elders in Qingdao, China. Asia Pacific Journal of Clinical Nutrition 31:97–107.
  • Malafarina, V., F. Uriz-Otano, C. Malafarina, J. Martinez, and M. Zulet. 2017. Effectiveness of nutritional supplementation on sarcopenia and recovery in hip fracture patients. A Multi-Centre Randomized Trial. Maturitas 101:42–50.
  • Marcangeli, V., L. Youssef, M. Dulac, L. P. Carvalho, G. Hajj-Boutros, O. Reynaud, B. Guegan, F. Buckinx, P. Gaudreau, J. A. Morais, et al. 2022. Impact of high-intensity interval training with or without L-citrulline on physical performance, skeletal muscle, and adipose tissue in obese older adults. Journal of Cachexia, Sarcopenia and Muscle 13 (3):1526–40. doi:10.1002/jcsm.12955.
  • Mariotti, F., and C. D. Gardner. 2019. Dietary protein and amino acids in vegetarian diets—A review. Nutrients, 1111: 2661. doi:10.3390/nu11112661.
  • Marliss, E. B., S. Chevalier, R. Gougeon, J. A. Morais, M. Lamarche, O. A. J. Adegoke, and G. Wu. 2006. Elevations of plasma methylarginines in obesity and ageing are related to insulin sensitivity and rates of protein turnover. Diabetologia 49 (2):351–9. doi:10.1007/s00125-005-0066-6.
  • Marques, J., E. Shokry, O. Uhl, L. Baber, F. Hofmeister, S. Jarmusch, M. Bidlingmaier, U. Ferrari, B. Koletzko, and M. Drey. 2023. Sarcopenia: Investigation of metabolic changes and its associated mechanisms. Skeletal Muscle 13 (1):2–14. doi:10.1186/s13395-022-00312-w.
  • Mather, M., L. A. Jacobsen, and K. M. Pollard. 2015. Aging in the United States. Population Bulletin 70:1–18.
  • McCormack, W. P., J. R. Stout, N. S. Emerson, T. C. Scanlon, A. M. Warren, A. J. Wells, A. M. Gonzalez, G. T. Mangine, E. H. Robinson, IV, M. S. Fragala, et al. 2013. Oral nutritional supplement fortified with beta-alanine improves physical working capacity in older adults: A randomized, placebo-controlled study. Experimental Gerontology 48 (9):933–9. doi:10.1016/j.exger.2013.06.003.
  • Menni, C., G. Kastenmüller, A. K. Petersen, J. T. Bell, M. Psatha, P. C. Tsai, C. Gieger, H. Schulz, I. Erte, S. John, et al. 2013. Metabolomic markers reveal novel pathways of ageing and early development in human populations. International Journal of Epidemiology 42 (4):1111–9. doi:10.1093/ije/dyt094.
  • Mitchell, C. J., P. A. Della Gatta, A. C. Petersen, D. Cameron-Smith, and J. F. Markworth. 2015a. Soy protein ingestion results in less prolonged p70S6 kinase phosphorylation compared to whey protein after resistance exercise in older men. Journal of the International Society of Sports Nutrition 12 (1):6–9. doi:10.1186/s12970-015-0070-2.
  • Mitchell, W. K., B. E. Phillips, J. P. Williams, D. Rankin, J. N. Lund, D. J. Wilkinson, K. Smith, and P. J. Atherton. 2015b. The impact of delivery profile of essential amino acids upon skeletal muscle protein synthesis in older men. Clinical Efficacy of Pulse vs. bolus Supply. American Journal of Physiology 309:E450–7.
  • Mobley, C. B., C. T. Haun, P. A. Roberson, P. W. Mumford, M. A. Romero, W. C. Kephart, R. G. Anderson, C. G. Vann, S. C. Osburn, C. D. Pledge, et al. 2017. Effects of whey, soy or leucine supplementation with 12 weeks of resistance training on strength, body composition, and skeletal muscle and adipose tissue histological attributes in college-aged males. Nutrients 9 (9):972. doi:10.3390/nu9090972.
  • Moore, D. R., M. J. Robinson, J. L. Fry, J. E. Tang, E. I. Glover, S. B. Wilkinson, T. Prior, M. A. Tarnopolsky, and S. M. Phillips. 2009. Ingested protein dose response of muscle and albumin protein synthesis after resistance exercise in young men. The American Journal of Clinical Nutrition 89 (1):161–8. doi:10.3945/ajcn.2008.26401.
  • Moore, D. R., T. A. Churchward-Venne, O. Witard, L. Breen, N. A. Burd, K. D. Tipton, and S. M. Phillips. 2015. Protein ingestion to stimulate myofibrillar protein synthesis requires greater relative protein intakes in healthy older versus younger men. Journals of Gerontology A 70:57–62.
  • Morais, J. A., S. Chevalier, and R. Gougeon. 2006. Protein turnover and requirements in the healthy and frail elderly. Journal of Nutrition, Health & Aging 10:272–83.
  • Morley, J. E., J. M. Argiles, W. J. Evans, S. Bhasin, D. Cella, N. E. Deutz, W. Doehner, K. C. Fearon, L. Ferrucci, M. K. Hellerstein, et al. 2010. Nutritional recommendations for the management of sarcopenia. Journal of the American Medical Directors Association 11 (6):391–6. doi:10.1016/j.jamda.2010.04.014.
  • Moughan, P. J., and S. M. Rutherfurd. 2012. True ileal amino acid and protein digestibility (%) for selected human foods. In Report of a Sub-Committee of the 2011 FAO Consultation on “Protein Quality Evaluation in Human Nutrition”. Rome: FAO.
  • Murton, A. J. 2015. Muscle protein turnover in the elderly and its potential contribution to the development of sarcopenia. Proceedings of the Nutrition Society 74:387–96.
  • Naclerio, F., M. Seijo, E. Larumbe-Zabala, and C. P. Earnest. 2017. Carbohydrates alone or mixing with beef or whey protein promote similar training outcomes in resistance training males: A double-blind, randomized controlled clinical trial. International Journal of Sport Nutrition and Exercise Metabolism 27 (5):408–20. doi:10.1123/ijsnem.2017-0003.
  • Nair, K. S. 1995. Muscle protein-turnover - methodological issues and the effect of aging. Journals of Gerontology A 50:107–12.
  • Nakayama, K., Y. Saito, C. Sanbongi, K. Murata, and T. Urashima. 2021. Effects of low-dose milk protein supplementation following low-to moderate intensity exercise training on muscle mass in healthy older adults: A randomized placebo-controlled trial. European Journal of Nutrition 60 (2):917–28. doi:10.1007/s00394-020-02302-4.
  • Norton, C., C. Toomey, W. G. McCormack, P. Francis, J. Saunders, E. Kerin, and P. Jakeman. 2016. Protein supplementation at breakfast and lunch for 24 weeks beyond habitual intakes increases whole-body lean tissue mass in healthy older adults. The Journal of Nutrition 146 (1):65–9. doi:10.3945/jn.115.219022.
  • Nunes, E. A., L. Colenso‐Semple, S. R. McKellar, T. Yau, M. U. Ali, D. Fitzpatrick‐Lewis, D. Sherifali, C. Gaudichon, D. Tomé, P. J. Atherton, et al. 2022. Systematic review and meta‐analysis of protein intake to support muscle mass and function in healthy adults. Journal of Cachexia, Sarcopenia and Muscle 13 (2):795–810. doi:10.1002/jcsm.12922.
  • Okamura, T., Y. Hasegawa, M. Hamaguchi, R. Sasano, and M. Fukui. 2023. The role of chicken eggs in modulating sarcopenic obesity and gut microbiota in db/db mice. Frontiers in Microbiology 14:1281217. doi:10.3389/fmicb.2023.1281217.
  • Ottestad, I., S. M. Ulven, L. K. L. Øyri, K. S. Sandvei, G. O. Gjevestad, A. Bye, N. A. Sheikh, A. S. Biong, L. F. Andersen, and K. B. Holven. 2018. Reduced plasma concentration of branched-chain amino acids in sarcopenic older subjects: A cross-sectional study. British Journal of Nutrition 120 (4):445–53. doi:10.1017/S0007114518001307.
  • Paddon-Jones, D., and B. B. Rasmussen. 2009. Dietary protein recommendations and the prevention of sarcopenia. Current Opinion in Clinical Nutrition & Metabolic Care 12 (1):86–90. doi:10.1097/MCO.0b013e32831cef8b.
  • Park, Y., J. E. Choi, and H. S. Hwang. 2018. Protein supplementation improves muscle mass and physical performance in undernourished prefrail and frail elderly subjects: A randomized, double-blind, placebo-controlled trial. The American Journal of Clinical Nutrition 108 (5):1026–33. doi:10.1093/ajcn/nqy214.
  • Park, S., D. D. Church, S. E. Schutzler, G. Azhar, I. Y. Kim, A. A. Ferrando, and R. R. Wolfe. 2021. Metabolic evaluation of the dietary guidelines' ounce equivalents of protein food sources in young adults: A randomized controlled trial. The Journal of Nutrition 151 (5):1190–6. doi:10.1093/jn/nxaa401.
  • Pennings, B., Y. Boirie, J. M. Senden, A. P. Gijsen, H. Kuipers, and L. J. C. van Loon. 2011a. Whey protein stimulates postprandial muscle protein accretion more effectively than do casein and casein hydrolysate in older men. The American Journal of Clinical Nutrition 93 (5):997–1005.
  • Pennings, B., R. Koopman, M. Beelen, J. M. Senden, W. H. Saris, and L. J. C. van Loon. 2011b. Exercising before protein intake allows for greater use of dietary protein-derived amino acids for de novo muscle protein synthesis in both young and elderly men. The American Journal of Clinical Nutrition 93 (2):322–31. doi:10.3945/ajcn.2010.29649.
  • Pennings, B., B. Groen, A. de Lange, A. P. Gijsen, A. H. Zorenc, J. M. Senden, and L. J. C. van Loon. 2012. Amino acid absorption and subsequent muscle protein accretion following graded intakes of whey protein in elderly men. American Journal of Physiology 302:E992–9.
  • Pérez-Guisado, J., and P. M. Jakeman. 2010. Citrulline malate enhances athletic anaerobic performance and relieves muscle soreness. Journal of Strength and Conditioning Research 24 (5):1215–22. doi:10.1519/JSC.0b013e3181cb28e0.
  • Phillips, S. M. 2012. Nutrient-rich meat proteins in offsetting age-related muscle loss. Meat Science 92 (3):174–8. doi:10.1016/j.meatsci.2012.04.027.
  • Pinckaers, P. J. M., F. K. Hendriks, W. J. H. Hermans, J. P. B. Goessens, J. M. Senden, J. M. X. Van Kranenburg, W. K. H. W. Wodzig, T. Snijders, and L. J. C. Van Loon. 2022b. Potato protein ingestion increases muscle protein synthesis rates at rest and during recovery from exercise in humans. Medicine and Science in Sports and Exercise 54 (9):1572–81. doi:10.1249/MSS.0000000000002937.
  • Pinckaers, P. J. M., I. W. K. Kouw, S. H. M. Gorissen, L. H. P. Houben, J. M. Senden, W. K. H. W. Wodzig, L. C. P. G. M. de Groot, L. B. Verdijk, T. Snijders, and L. J. C. van Loon. 2022a. The muscle protein synthetic response to the ingestion of a plant-derived protein blend does not differ from an equivalent amount of milk protein in healthy young males. The Journal of Nutrition 152 (12):2734–43. doi:10.1093/jn/nxac222.
  • Pinckaers, P. J., J. Domić, H. L. Petrick, A. M. Holwerda, J. Trommelen, F. K. Hendriks, L. H. Houben, J. P. Goessens, J. M. van Kranenburg, J. M. Senden, et al. 2023. Higher muscle protein synthesis rates following ingestion of an omnivorous meal compared with an isocaloric and isonitrogenous vegan meal in healthy, older adults. The Journal of Nutrition doi: 10.1016/j.tjnut.2023.11.004.
  • PitkäNen, H. T., S. S. Oja, K. Kemppainen, J. M. Seppä, and A. A. Mero. 2003. Serum amino acid concentrations in aging men and women. Amino Acids. 24 (4):413–21. doi:10.1007/s00726-002-0338-0.
  • Porter Starr, K. N., C. F. Pieper, M. C. Orenduff, S. R. McDonald, L. B. McClure, R. Zhou, M. E. Payne, and C. W. Bales. 2016. Improved function with enhanced protein intake per meal: A pilot study of weight reduction in frail, obese older adults. Journal of Gerontology A 71:1369–75.
  • Putra, C., N. Konow, M. Gage, C. G. York, and K. M. Mangano. 2021. Protein source and muscle health in older adults: A literature review. Nutrients 13 (3):743. doi:10.3390/nu13030743.
  • Reidy, P. T., D. K. Walker, J. M. Dickinson, D. M. Gundermann, M. J. Drummond, K. L. Timmerman, C. S. Fry, M. S. Borack, M. B. Cope, R. Mukherjea, et al. 2013. Protein blend ingestion following resistance exercise promotes human muscle protein synthesis. The Journal of Nutrition 143 (4):410–6. doi:10.3945/jn.112.168021.
  • Rennie, M. J., A. Selby, P. Atherton, K. Smith, V. Kumar, E. L. Glover, and S. M. Philips. 2010. Facts, noise and wishful thinking: Muscle protein turnover in aging and human disuse atrophy. Scandinavian Journal of Medicine & Science in Sports 20 (1):5–9. doi:10.1111/j.1600-0838.2009.00967.x.
  • Rieu, I., M. Balage, C. Sornet, C. Giraudet, E. Pujos, J. Grizard, L. Mosoni, and D. Dardevet. 2006. Leucine supplementation improves muscle protein synthesis in elderly men independently of hyperaminoacidaemia. The Journal of Physiology 575 (1):305–15. doi:10.1113/jphysiol.2006.110742.
  • Robinson, M. J., N. A. Burd, L. Breen, T. Rerecich, Y. Yang, A. J. Hector, S. K. Baker, and S. M. Phillips. 2013. Dose-dependent responses of myofibrillar protein synthesis with beef ingestion are enhanced with resistance exercise in middle-aged men. Applied Physiology, Nutrition, and Metabolism 38 (2):120–5. doi:10.1139/apnm-2012-0092.
  • Rondanelli, M., A. Opizzi, N. Antoniello, F. Boschi, P. Iadarola, E. Pasini, R. Aquilani, and F. S. Dioguardi. 2011. Effect of essential amino acid supplementation on quality of life, amino acid profile and strength in institutionalized elderly patients. Clinical Nutrition 30 (5):571–7. doi:10.1016/j.clnu.2011.04.005.
  • Rondanelli, M., C. Klersy, G. Terracol, J. Talluri, R. Maugeri, D. Guido, M. A. Faliva, B. S. Solerte, M. Fioravanti, H. Lukaski, et al. 2016. Whey protein, amino acids, and vitamin D supplementation with physical activity increases fat-free mass and strength, functionality, and quality of life and decreases inflammation in sarcopenic elderly. The American Journal of Clinical Nutrition 103 (3):830–40. doi:10.3945/ajcn.115.113357.
  • Rondanelli, M., E. Cereda, C. Klersy, M. A. Faliva, G. Peroni, M. Nichetti, C. Gasparri, G. Iannello, D. Spadaccini, V. Infantino, et al. 2020. Improving rehabilitation in sarcopenia (IRIS) by muscle-targeted food for special medical purposes: A randomized, double-blind, controlled trial. Journal of Cachexia, Sarcopenia and Muscle 11 (6):1535–47. doi:10.1002/jcsm.12532.
  • Rose, W. C., G. F. Lambert, and M. J. Coon. 1954. The amino acid requirement of man. VII General procedures: The tryptophan requirement. The Journal of Biological Chemistry 211 (2):815–27.
  • Rose, W. C., A. Borman, M. J. Coon, and G. F. Lambert. 1955. The amino acid requirement of man. The Journal of Biological Chemistry 214 (2):579–87.
  • Saka, B., S. Şahin, D. Kılıç, L. Öztürk, H. Yılmaz, M. Doğanay, S. Türkyılmaz, S. Menteş, O. Usluer, K. Arıkan, et al. 2019. Effects of beta-hydroxy-beta-methylbutyrate, arginine, and glutamine supplementation on the body composition and muscle strength in the elderly. Clinical Science of Nutrition 1 (2):67–74. doi:10.5152/ClinSciNutr.2019.892.
  • Scognamiglio, R., R. Piccolotto, C. Negut, A. Tiengo, and A. Avogaro. 2005. Oral amino acids in elderly subjects: Effect on myocardial function and walking capacity. Gerontology 51 (5):302–8.
  • Sekhar, R. V., S. G. Patel, A. P. Guthikonda, M. Reid, A. Balasubramanyam, G. E. Taffet, and F. Jahoor. 2011. Deficient synthesis of glutathione underlies oxidative stress in aging and can be corrected by dietary cysteine and glycine supplementation. The American Journal of Clinical Nutrition 94 (3):847–53. doi:10.3945/ajcn.110.003483.
  • Seo, C., Y. H. Hwang, Y. Kim, B. S. Joo, S. T. Yee, C. M. Kim, and M. J. Paik. 2016. Metabolomic study of aging in mouse plasma by gas chromatography–mass spectrometry. Journal of Chromatography. B, Analytical Technologies in the Biomedical and Life Sciences 1025:1–6.
  • Sharp, M. H., R. P. Lowery, K. A. Shields, J. R. Lane, J. L. Gray, J. M. Partl, D. W. Hayes, G. J. Wilson, C. A. Hollmer, J. R. Minivich, et al. 2018. The effects of beef, chicken, or whey protein post-workout on body composition and muscle performance. Journal of Strength and Conditioning Research 2:2233–42.
  • Short, K. R., J. L. Vittone, M. L. Bigelow, D. N. Proctor, and K. S. Nair. 2004. Age and aerobic exercise training effects on whole body and muscle protein metabolism. American Journal of Physiology. Endocrinology and Metabolism 286 (1):E92–101.
  • Silva, A. M., W. Shen, M. Heo, D. Gallagher, Z. Wang, L. B. Sardinha, and S. B. Heymsfield. 2010. Ethnicity-related skeletal muscle differences across the lifespan. American Journal of Human Biology 22 (1):76–82. doi:10.1002/ajhb.20956.
  • Smith, A., and J. Gray. 2016. Considering the benefits of egg consumption for older people at risk of sarcopenia. British Journal of Community Nursing 21 (6):305–9. doi:10.12968/bjcn.2016.21.6.305.
  • Smith, S. B., D. K. Lunt, D. R. Smith, and R. L. Walzem. 2020. Producing high-oleic acid beef and the impact of ground beef consumption on risk factors for cardiovascular disease: A review. Meat Science 163:108076. doi:10.1016/j.meatsci.2020.108076.
  • Sobiecki, J. G., P. N. Appleby, K. E. Bradbury, and T. J. Key. 2016. High compliance with dietary recommendations in a cohort of meat eaters, fish eaters, vegetarians, and vegans: Results from the European Prospective Investigation into Cancer and Nutrition-Oxford study. Nutrition Research 36 (5):464–77. doi:10.1016/j.nutres.2015.12.016.
  • Solerte, S. B., C. Gazzaruso, R. Bonacasa, M. Rondanelli, M. Zamboni, C. Basso, E. Locatelli, N. Schifino, A. Giustina, and M. Fioravanti. 2008. Nutritional supplements with oral amino acid mixtures increases whole-body lean mass and insulin sensitivity in elderly subjects with sarcopenia. American Journal of Cardiology 101:E69–77.
  • Stout, J. R., B. S. Graves, J. T. Cramer, E. R. Goldstein, P. B. Costa, A. E. Smith, and A. A. Walter. 2007. Effects of creatine supplementation on the onset of neuromuscular fatigue threshold and muscle strength in elderly men and women (64–86 years). Journal of Nutrition, Health & Aging 11:459–64.
  • Suárez, L. M., M.-D. Muñoz, R. M. del Río, and J. M. Solís. 2016. Taurine content in different brain structures during ageing: Effect on hippocampal synaptic plasticity. Amino Acids. 48 (5):1199–208. doi:10.1007/s00726-015-2155-2.
  • Suryawan, A., and T. A. Davis. 2011. Regulation of protein synthesis by amino acids in muscle of neonates. Frontiers in Bioscience 16 (1):1445–60. doi:10.2741/3798.
  • Symons, T., S. E. Schutzler, T. L. Cocke, D. L. Chinkes, R. R. Wolfe, and D. Paddon-Jones. 2007. Aging does not impair the anabolic response to a protein-rich meal. The American Journal of Clinical Nutrition 86 (2):451–6. doi:10.1093/ajcn/86.2.451.
  • Symons, T. B., M. Sheffield-Moore, R. R. Wolfe, and D. Paddon-Jones. 2009. Moderating the portion size of a protein-rich meal improves anabolic efficiency in young and elderly. Journal of the American Dietetic Association 109 (9):1582–6. doi:10.1016/j.jada.2009.06.369.
  • Takada, A., F. Shimizu, Y. Ishii, M. Ogawa, and T. Takao. 2020. Glucose or sucrose intakes and plasma levels of essential and nonessential amino acids. In New insights into metabolic syndrome, ed. T. Takada, 1–15. London: IntechOpen.
  • Tang, J. E., D. R. Moore, G. W. Kujbida, M. A. Tarnopolsky, and S. M. Phillips. 2009. Ingestion of whey hydrolysate, casein, or soy protein isolate: Effects on mixed muscle protein synthesis at rest and following resistance exercise in young men. Journal of Applied Physiology (Bethesda, Md.: 1985) 107 (3):987–92. doi:10.1152/japplphysiol.00076.2009.
  • Ten Haaf, D. S. M., T. M. H. Eijsvogels, C. C. W. G. Bongers, A. M. H. Horstman, S. Timmers, L. C. P. G. M. de Groot, and M. T. E. Hopman. 2019. Protein supplementation improves lean body mass in physically active older adults: A randomized placebo-controlled trial. Journal of Cachexia, Sarcopenia and Muscle 10 (2):298–310. doi:10.1002/jcsm.12394.
  • Tieland, M., O. van de Rest, M. L. Dirks, N. van der Zwaluw, M. Mensink, L. J. C. van Loon, and L. C. de Groot. 2012a. Protein supplementation improves physical performance in frail elderly people: A randomized, double-blind, placebo-controlled trial. Journal of the American Medical Directors Association 13 (8):720–6. doi:10.1016/j.jamda.2012.07.005.
  • Tieland, M., M. L. Dirks, N. van der Zwaluw, L. B. Verdijk, O. van de Rest, L. C. de Groot, and L. J. C. van Loon. 2012b. Protein supplementation increases muscle mass gain during prolonged resistance-type exercise training in frail elderly people: A randomized, double-blind, placebo-controlled trial. Journal of the American Medical Directors Association 13 (8):713–9. doi:10.1016/j.jamda.2012.05.020.
  • Tipton, K. D., A. A. Ferrando, S. M. Phillips, D. Doyle, Jr, and R. R. Wolfe. 1999a. Postexercise net protein synthesis in human muscle from orally administered amino acids. The American Journal of Physiology 276 (4):E628–34. doi:10.1152/ajpendo.1999.276.4.E628.
  • Tipton, K. D., B. E. Gurkin, S. Matin, and R. R. Wolfe. 1999b. Nonessential amino acids are not necessary to stimulate net muscle protein synthesis in healthy volunteers. The Journal of Nutritional Biochemistry 10 (2):89–95. doi:10.1016/s0955-2863(98)00087-4.
  • Tong, T. Y. N., P. N. Appleby, M. E. G. Armstrong, G. K. Fensom, A. Knuppel, K. Papier, A. Perez-Cornago, R. C. Travis, and T. J. Ke. 2020. Vegetarian and vegan diets and risks of total and site-specific fractures: Results from the prospective EPIC-Oxford study. BMC Medicine 18 (1):353. doi:10.1186/s12916-020-01815-3.
  • Toyoshima, K., M. Nakamura, Y. Adachi, A. Imaizumi, T. Hakamada, Y. Abe, E. Kaneko, S. Takahashi, and K. Shimokado. 2017. Increased plasma proline concentrations are associated with sarcopenia in the elderly. Plos One 12 (9):e0185206. doi:10.1371/journal.pone.0185206.
  • Turner, N. D., and S. K. Lloyd. 2017. Association between red meat consumption and colon cancer: A systematic review of experimental results. Experimental Biology and Medicine (Maywood, N.J.) 242 (8):813–39. doi:10.1177/1535370217693117.
  • USDA. 2021. Economic research service. Washington, D.C.: United States Department of Agriculture.
  • Uchitomi, R., Y. Hatazawa, N. Senoo, K. Yoshioka, M. Fujita, T. Shimizu, S. Miura, Y. Ono, and Y. Kamei. 2019. Metabolomic analysis of skeletal muscle in aged mice. Scientific Reports 9 (1):10425. doi:10.1038/s41598-019-46929-8.
  • Ullevig, S. L., K. Zuniga, C. A. Lobitz, A. Santoyo, and Z. Yin. 2022. Egg protein supplementation improved upper body muscle strength and protein intake in community-dwelling older adult females who attended congregate meal sites or adult learning centers: A pilot randomized controlled trial. Nutrition and Health 28 (4):611–20. doi:10.1177/02601060211051592.
  • Valenzuela, P. L., F. Mata, J. S. Morales, A. Castillo-García, and A. Lucia. 2019. Does beef protein supplementation improve body composition and exercise performance? A systematic review and meta-analysis of randomized controlled trials. Nutrients 11 (6):1429. doi:10.3390/nu11061429.
  • Van Elswyk, M. E., L. Teo, C. S. Lau, and C. J. Shanahan. 2022. Dietary patterns and the risk of sarcopenia: A systematic review and meta-analysis. Current Developments in Nutrition 6 (5):nzac001. doi:10.1093/cdn/nzac001.
  • Varshavi, D., F. H. Scott, D. Varshavi, S. Veeravalli, I. R. Phillips, K. Veselkov, N. Strittmatter, Z. Takats, E. A. Shephard, and J. R. Everett. 2018. Metabolic biomarkers of ageing in C57BL/6J wild-type and flavin-containing monooxygenase 5 (FMO5)-knockout mice. Frontiers in Molecular Biosciences 5:28. doi:10.3389/fmolb.2018.00028.
  • van Vliet, S., N. A. Burd, and L. J. C. van Loon. 2015. The skeletal muscle anabolic response to plant- versus animal-based protein consumption. The Journal of Nutrition 145 (9):1981–91. doi:10.3945/jn.114.204305.
  • Verhoeven, S., K. Vanschoonbeek, L. B. Verdijk, R. Koopman, W. K. Wodzig, P. Dendale, and L. J. C. van Loon. 2009. Long-term leucine supplementation does not increase muscle mass or strength in healthy elderly men. The American Journal of Clinical Nutrition 89 (5):1468–75. doi:10.3945/ajcn.2008.26668.
  • Verlaan, S., A. B. Maier, J. M. Bauer, I. Bautmans, K. Brandt, L. M. Donini, M. Maggio, M. E. T. McMurdo, T. Mets, C. Seal, et al. 2018. Sufficient levels of 25-hydroxyvitamin D and protein intake required to increase muscle mass in sarcopenic older adults—The PROVIDE study. Clinical Nutrition 37 (2):551–7. doi:10.1016/j.clnu.2017.01.005.
  • Verreijen, A. M., S. Verlaan, M. F. Engberink, S. Swinkels, J. de Vogel-van den Bosch, and P. J. M. Weijs. 2015. A high whey protein-, leucine-, and vitamin D-enriched supplement preserves muscle mass during intentional weight loss in obese older adults: A double-blind randomized controlled trial. The American Journal of Clinical Nutrition 101 (2):279–86. doi:10.3945/ajcn.114.090290.
  • Volek, J. S., B. M. Volk, A. L. Gómez, L. J. Kunces, B. R. Kupchak, D. J. Freidenreich, J. C. Aristizabal, C. Saenz, C. Dunn-Lewis, K. D. Ballard, et al. 2013. Whey protein supplementation during resistance training augments lean body mass. Journal of the American College of Nutrition 32 (2):122–35. doi:10.1080/07315724.2013.793580.
  • Volpi, E., B. Mittendorfer, B. B. Rasmussen, and R. R. Wolfe. 2000. The response of muscle protein anabolism to combined hyperaminoacidemia and glucose-induced hyperinsulinemia is impaired in the elderly. The Journal of Clinical Endocrinology and Metabolism 85 (12):4481–90. doi:10.1210/jcem.85.12.7021.
  • Waldron, M., S. D. Patterson, J. Tallent, and O. Jeffries. 2018. The effects of an oral taurine dose and supplementation period on endurance exercise performance in humans: A meta-analysis. Sports Medicine 48 (5):1247–53. doi:10.1007/s40279-018-0896-2.
  • Waldron, M., S. D. Patterson, and O. Jeffries. 2019. Oral taurine improves critical power and severe-intensity exercise tolerance. Amino Acids. 51 (10-12):1433–41. doi:10.1007/s00726-019-02775-6.
  • Wall, B. T., S. H. Gorissen, B. Pennings, R. Koopman, B. B. Groen, L. B. Verdijk, and L. J. C. van Loon. 2015. Aging is accompanied by a blunted muscle protein synthetic response to protein ingestion. PloS One 10 (11):e0140903.
  • Walrand, S., C. Gryson, J. Salles, C. Giraudet, C. Migné, C. Bonhomme, P. L. Ruyet, and Y. Boirie. 2016. Fast-digestive protein supplement for ten days overcomes muscle anabolic resistance in healthy elderly men. Clinical Nutrition 35 (3):660–8. doi:10.1016/j.clnu.2015.04.020.
  • Wang, B., Y. Wei, L. Shao, M. Li, X. Zhang, W. Li, S. Zhao, X. Xia, and P. Liu. 2023. Association of dietary patterns and sarcopenia in the elderly population: A crosssectional study. Frontiers in Aging 4:1239945. doi:10.3389/fragi.2023.1239945.
  • Welle, S., C. Thornton, R. Jozefowicz, and M. Statt. 1993. Myofibrillar protein synthesis in young and old men. American Journal of Physiology 264:E693–8.
  • Wilborn, C. D., L. W. Taylor, J. Outlaw, L. Williams, B. Campbell, C. A. Foster, A. Smith-Ryan, S. Urbina, and S. Hayward. 2013. The effects of pre- and post-exercise whey vs. casein protein consumption on body composition and performance measures in collegiate female athletes. Journal of Sports Science and Medicine 12:74–9.
  • Wilkinson, S. B., M. A. Tarnopolsky, M. J. MacDonald, J. R. MacDonald, D. Armstrong, and S. M. Phillips. 2007. Consumption of fluid skim milk promotes greater muscle protein accretion after resistance exercise than does consumption of an isonitrogenous and isoenergetic soy-protein beverage. The American Journal of Clinical Nutrition 85 (4):1031–40. doi:10.1093/ajcn/85.4.1031.
  • Willett, W., J. Rockström, B. Loken, M. Springmann, T. Lang, S. Vermeulen, T. Garnett, D. Tilman, F. DeClerck, A. Wood, et al. 2019. Food in the anthropocene: The EAT-Lancet Commission on healthy diets from sustainable food systems. The Lancet 393 (10170):447–92. doi:10.1016/S0140-6736(18)31788-4.
  • Wright, C. S., J. Zhou, R. D. Sayer, J. E. Kim, and W. W. Campbell. 2018. Effects of a high-protein diet including whole eggs on muscle composition and indices of cardiometabolic health and systemic inflammation in older adults with overweight or obesity: A randomized controlled trial. Nutrients 10 (7):946. doi:10.3390/nu10070946.
  • Wu, G. 2020. Important roles of dietary taurine, creatine, carnosine, anserine and 4-hydroxyproline in human nutrition and health. Amino Acids 52 (3):329–60. doi:10.1007/s00726-020-02823-6.
  • Wu, G. 2022. Amino acids: Biochemistry and nutrition. 2nd ed. Boca Raton, FL: CRC Press.
  • Wu, G. 2023. Amino acids: Chemistry and classification. In Encyclopedia of human nutrition, ed. Benjamin Caballero, 4th ed, 10–22. New York, NY: Elsevier, New York.
  • Wu, G., J. Fanzo, D. D. Miller, P. Pingali, M. Post, J. L. Steiner, and A. E. Thalacker-Mercer. 2014. Production and supply of high-quality food protein for human consumption: Sustainability, challenges and innovations. Annals of the New York Academy of Sciences 1321 (1):1–19.
  • Wu, G., H. R. Cross, K. B. Gehring, J. W. Savell, A. N. Arnold, and S. H. McNeill. 2016. Composition of free and peptide-bound amino acids in beef chuck, loin, and round cuts. Journal of Animal Science 94 (6):2603–13. doi:10.2527/jas.2016-0478.
  • Wu, G., C. J. Meininger, C. J. McNeal, F. W. Bazer, and J. M. Rhoads. 2021. Role of L-arginine in nitric oxide synthesis and health in humans. Advances in Experimental Medicine and Biology 1332:167–87.
  • Wu, G., F. W. Bazer, M. C. Satterfield, K. R. Gilbreath, E. A. Posey, and Y. X. Sun. 2022. L-arginine nutrition and metabolism in ruminants. Advances in Experimental Medicine and Biology 1354:177–206. doi:10.1007/978-3-030-85686-1_10.
  • Wycherley, T. P., L. J. Moran, P. M. Clifton, M. Noakes, and G. D. Brinkworth. 2012. Effects of energy-restricted high-protein, low-fat compared with standard-protein, low-fat diets: A meta-analysis of randomized controlled trials. The American Journal of Clinical Nutrition 96 (6):1281–98. doi:10.3945/ajcn.112.044321.
  • Yang, Y., L. Breen, N. A. Burd, A. J. Hector, T. A. Churchward-Venne, A. R. Josse, M. A. Tarnopolsky, and S. M. Phillips. 2012a. Resistance exercise enhances myofibrillar protein synthesis with graded intakes of whey protein in older men. British Journal of Nutrition 108 (10):1780–8. doi:10.1017/S0007114511007422.
  • Yang, Y., T. A. Churchward-Venne, N. A. Burd, L. Breen, M. A. Tarnopolsky, and S. M. Phillips. 2012b. Myofibrillar protein synthesis following ingestion of soy protein isolate at rest and after resistance exercise in elderly men. Nutrition & Metabolism 9 (1):57. doi:10.1186/1743-7075-9-57.
  • Yarasheski, K. E., S. Welle, and K. S. Nair. 2002. Muscle protein synthesis in younger and older men. Jama 287 (3):317–8.
  • Yeung, J. M., and E. Friedman. 1991. Effect of aging and diet restriction on monoamines and amino acids in cerebral cortex of Fischer-344 rats. Growth, Development, and Aging: GDA 55 (4):275–83.
  • Yeung, S. S. Y., Z. L. Y. Zhu, T. Kwok, and J. Woo. 2022. Serum amino acids patterns and 4-year sarcopenia risk in community-dwelling Chinese older adults. Gerontology 68 (7):736–45. doi:10.1159/000518412.
  • Yu, Z., G. Zhai, P. Singmann, Y. He, T. Xu, C. Prehn, W. Römisch-Margl, E. Lattka, C. Gieger, N. Soranzo, et al. 2012. Human serum metabolic profiles are age dependent. Aging Cell 11 (6):960–7. doi:10.1111/j.1474-9726.2012.00865.x.
  • Yuan, S., and S. C. Larsson. 2023. Epidemiology of sarcopenia: Prevalence, risk factors, and consequences. Metabolism 144:155533. doi:10.1016/j.metabol.2023.155533.
  • Zhang, X.-Z., W.-Q. Xie, L. Chen, G.-D. Xu, L. Wu, Y.-S. Li, and Y.-X. Wu. 2022. Blood flow restriction training for the intervention of Sarcopenia: Current stage and future perspective. Frontiers of Medicine 9:894996.