Hostname: page-component-7c8c6479df-r7xzm Total loading time: 0 Render date: 2024-03-28T15:26:11.581Z Has data issue: false hasContentIssue false

Muscle protein turnover in the elderly and its potential contribution to the development of sarcopenia

Published online by Cambridge University Press:  31 March 2015

Andrew J. Murton*
Affiliation:
MRC/ARUK Centre for Musculoskeletal Ageing Research, Division of Nutritional Sciences, School of Biosciences, Sutton Bonington Campus, The University of Nottingham, LE12 5RD, UK
*
Corresponding author: Andrew J. Murton, email andrew.murton@nottingham.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

The underlying aetiology of sarcopenia appears multifaceted and not yet fully defined, but ultimately involves the gradual loss of muscle protein content over time. The present evidence suggests that the loss of lean tissue in the elderly is exacerbated by low dietary protein intake. Moreover, acute stable-isotope-based methodologies have demonstrated that the muscle anabolic response to a given amount of protein may decline with age, a phenomenon that has been termed anabolic resistance. Although the mechanism responsible for the inability of muscle to mount a satisfactory anabolic response to protein provision with increasing age is presently unknown, it does not appear due to impaired digestion or absorption of dietary protein. Rather, the issue could reside with any combination of: a diminished delivery of amino acids to peripheral tissues, impaired uptake of amino acids into muscle cells, or an inability of amino acids to elicit intracellular events pivotal for anabolism to occur. Despite the presence of anabolic resistance to dietary protein, present evidence suggests that protein supplementation may be able to overcome these issues, particularly when combined with resistance exercise programmes. As such, protein supplementation may prove to be an effective approach to delay the loss of muscle mass with age and has led to calls for the recommended daily intake of protein to be increased for the elderly population.

Type
Conference on ‘Nutrition and age-related muscle loss, sarcopenia and cachexia’
Copyright
Copyright © The Author 2015 

Muscle mass and contractile function are intricately associated with perceived quality of life, and outcome following subsequent illness, disease or surgery( Reference Rantanen, Harris and Leveille 1 Reference Lee, Boyko and Nielson 4 ). As such, when a significant loss of muscle mass occurs, individuals are at a heightened risk of fall-related fractures and their ability to live an independent life is severely compromised. Of concern, the loss of muscle mass and strength are natural hallmarks of the ageing process, being termed sarcopenia and dynapenia, respectively. As early as age 40 years, leg muscle mass and strength start to decline, with the rate of loss accelerating from 70 years onwards( Reference Kitamura, Koda and Otsuka 5 , Reference Lindle, Metter and Lynch 6 ). In the USA, where figures are readily available, it is estimated that sarcopenia is indirectly responsible for US$18·5 billion in annual health care expenditure( Reference Sayer 7 ). With the projected increase in size of the global elderly population anticipated in subsequent decades, healthcare resources required for the prevention and treatment of sarcopenia are only set to grow. It is therefore imperative that an understanding of the underlying scientific basis for the loss of muscle mass with age is established so that effective and acceptable pharmaceutical, nutraceutical and lifestyle modifications can be developed and instigated in at risk populations.

There are numerous factors that could contribute to the development of sarcopenia, including but not limited to: reduced habitual food intake, increase in sedentary behaviour, poor vitamin D status, loss of motorneurones, development of chronic low-grade inflammation and increased oxidative stress. In reality, it is likely that the aetiology of sarcopenia is multifactorial and moreover, the underlying causative factors could differ between sarcopenic individuals. Regardless of the mechanism(s) involved, the insipid loss of muscle mass observed during sarcopenia will necessitate the net loss of muscle myofibrillar contractile protein, thereby representing a key target for therapeutic intervention. Thus, strategies that reverse or limit the age related decline in muscle protein content are likely to be advantageous. This review will focus on the mechanisms responsible for regulating muscle mass in human subjects and discuss how they are perturbed as part of the ageing process. Furthermore, the use of protein and amino acids as strategies to enhance muscle anabolism will be considered and possible challenges in their adoption highlighted.

The role of protein turnover in the regulation of muscle mass

Muscle mass, being typically composed of 88 % protein by dry weight( Reference Heymsfield, Stevens and Noel 8 ), is largely dependent upon the intricate balance between the rate at which muscle proteins are synthesised and degraded. Where an imbalance in the magnitude of these two opposing processes occurs, a net change in muscle protein content ensues and when sustained over chronic periods, is reflected by a reciprocal change in muscle mass. During sarcopenia, and in other conditions characterised by cachexia (e.g. cancer, sepsis, acquired immune deficiency syndrome and chronic obstructive pulmonary disease), rates of muscle proteolysis exceeds those of muscle protein synthesis, often leading to the debilitating loss of muscle mass over time.

Crucially, the processes of muscle protein synthesis and muscle protein breakdown are dynamic, changing in response to nutritional, hormonal and contractile cues. Importantly, following protein consumption, muscle protein synthesis is enhanced( Reference Pennings, Boirie and Senden 9 ); an effect that can be recreated solely through the administration of the essential amino acids( Reference Volpi, Kobayashi and Sheffield-Moore 10 ). In particular, the branched chain amino acid leucine appears particularly effective at stimulating muscle protein synthesis (see( Reference Garlick 11 )). Viewed in conjunction with observations that hyperinsulinaemia stimulates muscle protein synthesis in human subjects under conditions of increased amino acid availability( Reference Fujita, Rasmussen and Cadenas 12 ), it highlights the important roles played by both insulin and leucine in the induction of muscle protein anabolism post-meal consumption. The manner in which insulin and amino acids collectively elicit these effects within the muscle cell appears through stimulation of the mammalian target of rapamycin complex 1 (mTORc1) signalling cascade. Recent evidence has provided the notion that mTORc1 acts as a nutrient sensor, integrating signals on amino acid availability, energy status via feedback from 5′ adenosine monophosphate-activated protein kinase, and status of the insulin-signalling pathway through AKT-mediated phosphorylation of mTORc1 residue Ser2448 (Fig. 1; see ( Reference Frost and Lang 13 )). As such, mTORc1 co-ordinates the promotion of translation initiation in the postprandial period when substrates (amino acids) are readily available and ATP generation is sufficient to support the high-energy demands of protein synthesis. This is achieved by mTORc1 eliciting two main effects. Firstly, when active, mTORc1 phosphorylates residue Thr389 of p70 S6 K, promoting the increased translation of mRNA containing a 5′ oligopyrimidine tract, typically represented in mRNA encoding ribsosomal proteins and translation elongation factors. Secondly, mTORc1 phosphorylates and thereby inactivates eukaryotic initiation factor 4E binding protein 1 (also known as PHAS-1), a protein that when hypophosphorylated is known to repress translation by preventing the binding of the 7-methylguanylate cap, a structural feature of most mRNA transcripts, to the forming ribosome complex (see( Reference Nader 14 )). The demonstration that administration of the potent mTORc1 inhibitor rapamycin to healthy human volunteers blunts the ability of amino acid feeding to stimulate muscle protein synthesis( Reference Dickinson, Fry and Drummond 15 ), highlights the essential requirement of this kinase in eliciting feeding gains in muscle protein content.

Fig. 1. A simplified diagram representing the role of mammalian target of rapamycin complex 1 (mTORc1) signalling in regulating muscle protein synthesis in response to extracellular cues. SNAT, sodium-coupled neutral amino acid transporter; LAT1, L-type amino acid transporter 1; IRS-1, insulin receptor substrate-1; Gln, glutamine; Leu, leucine; 4EBP1, eukaryotic initiation factor 4E binding protein 1.

While the postprandial stimulation of muscle protein synthesis has historically received the most attention when events responsible for the net loss of muscle protein content in human subjects have been investigated, this is not to suggest that muscle protein breakdown also plays an essential, if yet to be fully defined, role. Indeed, in rodents enhanced muscle proteolysis has been repeatedly demonstrated to represent a major contributor to the loss of muscle mass in a number of atrophy-inducing conditions( Reference Lecker, Jagoe and Gilbert 16 ), including sarcopenia( Reference Altun, Besche and Overkleeft 17 ). The paucity of data in human subjects is more a reflection of the experimental challenges and technical requirements faced when trying to obtain accurate surrogate measures of muscle protein breakdown. However, in reports where attempts have been made to obtain an index of muscle protein breakdown by examining arteriovenous dilution of isotopically labelled amino acids across the leg, it has been consistently observed that during conditions of hyperinsulinaemia, as observed in the postprandial period, muscle protein breakdown is suppressed( Reference Wilkes, Selby and Atherton 18 ). Collectively therefore, the consumption of food plays an essential role in dictating the diurnal pattern of muscle protein synthesis and muscle protein breakdown.

As mentioned earlier, the maintenance of protein intake and its effectiveness at stimulating muscle protein synthesis are central to the maintenance of muscle mass in the elderly population. However, several aspects from the process of meal consumption to the eventual stimulation of muscle protein synthesis by amino acids have been proposed as negatively impacted as part of the ageing process. These include decreased food intake, impaired digestion and absorption of nutrients, altered splanchnic uptake of amino acids, insufficient delivery of nutrients through the microvascular circulation, blunted uptake of amino acids into the muscle cell and perturbed cellular response to increased amino acid availability. Each of these potential contributory factors will be discussed in turn.

Changes in protein intake requirements with age

It has been suggested that decreased protein intake with age, set in the wider context of reduced meal consumption, plays a major role in the aetiology of sarcopenia( Reference Morley, Argiles and Evans 19 ). Certainly, for a subset of individuals dental issues, blunted olfactory and taste perception, combined with social and economic factors contribute to reduced meal consumption or removal of high-quality animal protein from the diet( Reference Boyce and Shone 20 ), and likely compound the loss of muscle experienced by these individuals. However, for the majority of the elderly population the issue appears to be far greater than just insufficient protein intake alone. Observational studies performed in Western populations have demonstrated that a significant proportion (60–85 %) of older men and women meet the present adult recommendations for protein intake of 0·8 g/kg per d (see( Reference Morley, Argiles and Evans 19 )). However, of concern, a study by Campbell et al. in older adults reported that a 14-week carefully controlled and monitored eucaloric diet containing 0·8 g protein/kg per d was insufficient to stave off the 1·5 % decline in the thigh muscle cross-sectional area seen during the experimental period( Reference Campbell, Trappe and Wolfe 21 ). Similarly, in a preceding smaller study by the same researchers, they demonstrated that older healthy adults consuming the protein RDA for 10 d resulted in a negative nitrogen balance( Reference Campbell, Crim and Dallal 22 ). A 3-year longitudinal study conducted in 2066 North American septuagenarians provided further evidence that weight-losing and weight-stable individuals meeting or exceeding current protein recommendations still experienced the loss of appendicular lean mass( Reference Houston, Nicklas and Ding 23 ). Collectively, these important observations have led to the suggestion that the current RDA for adults aged over 18 years is insufficient to maintain muscle mass in the elderly, with the higher recommendations of 1–1·5 g/kg per d suggested as an appropriate intake to reduce risk of sarcopenia( Reference Morley, Argiles and Evans 19 ). Moreover, it identifies that the ability to elicit muscle anabolism per unit dietary protein is reduced in the elderly( Reference Morley, Argiles and Evans 19 ), potential reasons for which are discussed later.

Protein absorption and digestion

Independent of any purported change in food intake in the elderly, the maintenance of adequate protein digestion and absorption is essential. An inability to retain comparable digestion and absorption kinetics to young adults could underpin the failure of dietary protein to maintain muscle mass in the elderly. Given the highly complex nature of the gastrointestinal tract, involving the coordinated action of a wide variety of specialised cells, the exact impact of ageing per se on gastrointestinal function remains unclear (see( Reference Saffrey 24 )). However, delayed gastric emptying, increased colonic transit time and increased incidence of chronic constipation have all been reported( Reference Saffrey 24 ). Likewise, clinical disorders of the gastrointestinal tract, including susceptibility to gut infections, gastroesophageal reflux disease, irritable bowel syndrome, peptic ulcer formation and diverticulosis, all increase in incidence with age. Therefore, the impaired digestion and absorption of food constituents in the elderly is a genuine concern.

Typically, the direct study of protein digestion and absorption kinetics in human subjects is fraught with technical challenges, and as such rarely attempted. However, in a recent series of detailed experiments, the absorption and digestion characteristics of animal-derived protein in older adults were investigated. By intravenously administering [1–13C]phenylalanine to a cow over a 48 h period followed by milking and slaughter of the animal, Pennings et al. were able to generate intrinsically labelled milk protein and meat( Reference Pennings, Pellikaan and Senden 25 ). Utilising the casein fraction of the intrinsically labelled milk, they were able to demonstrate through oral administration of a 35 g bolus to young and elderly individuals that the rate of appearance of the labelled phenylalanine in the circulation was equivalent between the age groups studied( Reference Koopman, Walrand and Beelen 26 ). This demonstrated that in healthy elderly men the digestion and absorption of casein protein is not impaired. Moreover, despite previous suggestions that splanchnic uptake of absorbed amino acids may increase with age( Reference Volpi, Mittendorfer and Wolf 27 , Reference Boirie, Gachon and Beaufrere 28 ), the authors were able to demonstrate that splanchnic uptake was largely equivalent between the two age groups studied, at about 72 % each. This latter observation is noteworthy, if splanchnic uptake was found to be enhanced with age; this could impair the availability of dietary-consumed amino acids for utilisation in muscle protein synthesis.

It could be argued that the administration of a large casein bolus in beverage form is not representative of the types of foods ingested by elderly adults or the manner in which they typically consume meals. As such, the relevance of these findings to the real-world situation could be questioned. In a subsequent report, the same authors demonstrate that the manner in which food is processed can impact significantly on protein digestion and absorption kinetics. By comparing 135 g cooked minced beef with beef steak, both intrinsically labelled with [1–13C]phenylalanine and consumed by elderly male volunteers, they reported that minced beef is more rapidly digested and absorbed than its steak counterpart( Reference Pennings, Groen and van Dijk 29 ). Likewise, small-animal-based studies have demonstrated that protein digestibility is considerably lower in older animals compared with their young counterparts when protein is consumed in the presence of anti-nutritional factors such as phytases( Reference Gilani and Sepehr 30 ); an issue of particular concern in developing countries where there is a large reliance on crop-based protein sources high in concentrations of such anti-nutritional factors (see( Reference Gilani, Cockell and Sepehr 31 )). Collectively, this suggests that a more holistic approach reflecting the standard eating practices in the elderly would be beneficial when considering questions relating to digestion and absorption.

Given that the work of Koopman et al. tentatively suggests that the digestion and absorption of protein is not impaired in the elderly( Reference Koopman, Walrand and Beelen 26 ), it would thereby propose that the basis for the inability of dietary protein to maintain muscle mass in the elderly must reside elsewhere. In support of this notion, the intravenous administration of mixed amino acids to young and elderly volunteers under hyperinsulinaemic euglycaemic conditions, a strategy that thereby circumvents any potential issues associated with altered digestion, absorption or splanchnic uptake of orally administered protein, elicits greater increases in muscle protein synthesis in young compared with elderly individuals( Reference Guillet, Prod'homme and Balage 32 ). The blunting of the stimulatory effect of amino acids on muscle protein synthesis in elderly individuals has since been reported by others, following either intravenous or oral feeding( Reference Cuthbertson, Smith and Babraj 33 ) of amino acids, with the phenomenon subsequently termed anabolic resistance( Reference Cuthbertson, Smith and Babraj 33 ).

Impact of age on postprandial hyperaemia

The enhanced delivery of circulating amino acids to muscle tissue is an essential requisite for postprandial elevations in muscle protein synthesis to occur, with delivery dependent on both arterial amino acid concentration and arterial blood flow to the muscle. The administration of insulin to produce physiological hyperinsulinaemia has been repeatedly shown to stimulate enhanced blood flow to muscle tissue( Reference Fujita, Rasmussen and Cadenas 12 , Reference Vollenweider, Tappy and Randin 34 , Reference Laakso, Edelman and Brechtel 35 ) and therefore, the incretin effect of food consumption acts to prime the system to assist in the delivery of glucose and amino acids to peripheral tissues. As such, linear relationships have been described in healthy young subjects between the stimulation of muscle protein synthesis during graded localised insulin administration and the change seen in femoral arterial blood flow and amino acid delivery to the leg( Reference Fujita, Rasmussen and Cadenas 12 ). Given the progressive development of insulin resistance in the elderly population( Reference Karakelides, Irving and Short 36 ), it would appear to follow that an impairment of insulin-mediated increases in amino acid delivery under postprandial conditions may contribute to anabolic resistance.

In support of this notion, leg blood flow under hyperinsulinaemic euglycaemic clamp conditions, appears impaired in old volunteers compared with their young healthy counterparts( Reference Meneilly, Elliot and Bryer-Ash 37 ). The same finding has also been observed when performed in conjunction with exogenously administered amino acids( Reference Volpi, Mittendorfer and Rasmussen 38 ) or in response to intermittent feeding of an oral mixed-feed over a 2 h period( Reference Phillips, Williams and Atherton 39 ). Thus, with the stated requirement to enhance amino acid delivery to sufficiently increase muscle protein synthesis, it would be judicious to employ strategies that safely stimulate blood flow or improve the vasodilatory effects of insulin. In this regard, the principal mechanism by which insulin appears to impart is vasodilatory effects via stimulating the synthesis and release of endothelin-derived nitric oxide (NO)( Reference Steinberg, Brechtel and Johnson 40 ), thereby promoting a potent relaxation of arterial smooth muscle. Therefore, pharmacological stimulation of increased NO synthesis may represent one such strategy. In support, Dillon et al. were able to demonstrate that sodium nitroprusside, a known NO donor with potent vasodilatory effects, was able to enhance leg blood flow and stimulate muscle protein synthesis to similar degrees in young and elderly subjects when administered in combination with amino acids( Reference Dillon, Casperson and Durham 41 ). However, their decision not to include a control group who did not receive the nitroprusside treatment means caution has to be applied as to the significance of these findings.

In addition to pharmacological approaches, the use of nutrition to promote increased NO formation in the elderly is an area of current investigation. Although the primary focus in the majority of the studies performed to date has been to improve vascular function so as to reduce the incidence of CVD, an unintended benefit may be an improvement in the hyperaemic response to food consumption. Common strategies employed have included increasing dietary nitrate intake through the consumption of high-nitrate containing foods or by high-nitrate containing oral supplements, often in the form of beetroot concentrate. Although the number of studies conducted to date is limited, the acute (3-d) consumption of beetroot supplements by elderly subjects has been shown to increase plasma nitrite concentrations 4-fold and result in a 3 mm Hg decrease in mean arterial blood pressure with no change in resting heart rate( Reference Kelly, Fulford and Vanhatalo 42 ); changes consistent with increased NO-mediated smooth muscle relaxation. While the ability of a nitrate supplement to enhance a postprandial rise in blood flow has yet to be demonstrated in the elderly, reports of concentrated beetroot juice successfully enhancing brachial artery endothelial function in overweight and obese men 2 h post-meal consumption is promising( Reference Joris and Mensink 43 ). Separately, while the administration of the NO precursor, l-arginine, has been proposed as an alternative mechanism by which to enhance NO species generation( Reference Paddon-Jones, Borsheim and Wolfe 44 ), the inability of large bolus doses of l-arginine to increase plasma nitrate and nitrite concentrations in healthy young individuals( Reference Tang, Lysecki and Manolakos 45 ) suggests that this may not represent a viable strategy. Likewise, administration of citrulline, the endogenous precursor for the synthesis of arginine, does not appear to increase the responsiveness of muscle blood flow or muscle protein synthesis to protein feeding in elderly men( Reference Churchward-Venne, Cotie and MacDonald 46 ). Therefore, on the basis of current evidence, it would appear that nitrate supplements might represent the most viable strategy to reverse age-related impairments in hyperaemia following feeding. However, a crucial requirement before its adoption would be the need to understand the potential side effect of chronic high-nitrate intake in this population.

A second important modulator of muscle blood flow is exercise. While elderly men display a blunted hyperaemic response to both resistance-based and aerobic exercise across a spectrum of workloads( Reference Donato, Uberoi and Wray 47 , Reference Poole, Lawrenson and Kim 48 ), exercise appears to retain its ability to improve the hyperaemic response to feeding. Specifically, it has been demonstrated in elderly men that a 20-week whole-body resistance exercise training programme is able to restore the hyperaemic response to both feeding and acute bouts of resistance exercise( Reference Phillips, Williams and Atherton 39 ). This capacity of resistance exercise, combined with its widely known function to act as a potent stimulator of muscle growth, reinforces the need to consider resistance exercise as a central treatment to prevent and/or limit sarcopenia. While concerns may exist as to the suitability of this form of exercise in frail populations, work by Evans and co-workers has shown high-intensity resistance exercise to be both feasible in frail very elderly populations (age 72–98 years), and moreover, able to increase muscle mass and strength( Reference Fiatarone, O'Neill and Ryan 49 ).

Alterations in the muscle cell response to hyperaminoacidaemia with age

Regardless of purported impairments in the hyperaemic response to food consumption in the elderly, it is also feasible that the uptake of, or responsiveness of the muscle cell to, amino acids is no longer satisfactory to stimulate mTORc1 activity and initiate translation initiation. Certainly, it has been demonstrated that even after the consumption of large doses (≥40 g) of essential amino acids, phosphorylation of mTORc1 is blunted in elderly muscle v. the young( Reference Cuthbertson, Smith and Babraj 33 ). Moreover, the blunted response of muscle protein synthesis to amino acid provision in the elderly remains even when intramuscular concentrations of leucine are increased to a greater extent than that observed in the young( Reference Cuthbertson, Smith and Babraj 33 ), suggesting that impediments at the muscle cell level exist.

Leucine, the predominant amino acid that results in increased mTORc1 phosphorylation( Reference Atherton, Smith and Etheridge 50 ), and stimulates muscle protein synthesis( Reference Garlick 11 ), is unable to freely traverse the muscle cell membrane. Instead, in common with the other branched-chain amino acids, uptake into the muscle cell is regulated via System L transport through a protein complex consisting of a heterodimer between amino acid transporters L-type amino acid transporter 1 (LAT1) and CD98, with leucine import requiring the bidirectional transport of glutamine out of the muscle cell (Fig. 1; see( Reference Frost and Lang 13 )). Therefore, the maintenance of a glutamine concentration gradient is essential to retain the cell's ability to import leucine, achieved through the concerted action of the System A sodium-coupled neutral amino acid transporter (SNAT2) and System N amino acid transporters (SNAT3). As a result, intracellular concentrations of essential amino acids can be maintained under conditions of hypoaminoacidaemia( Reference Borsheim, Kobayashi and Traber 51 ). Likewise, except when concentrations of plasma amino acids are in excess of the amount required to maximally stimulate muscle protein synthesis( Reference Bohe, Low and Wolfe 52 ), intracellular concentrations of essential amino acids are maintained at relatively constant concentrations( Reference Borsheim, Kobayashi and Traber 51 , Reference Tipton, Ferrando and Phillips 53 ). Proton-assisted amino acid transporter 1, an intracellular transporter primarily located on the late endosome and lysosomes within cells has also been implicated in the activation of mTORc1 in response to amino acid availability( Reference Ogmundsdottir, Heublein and Kazi 54 ); deletion in Drosophila flies is known to impair the ability of amino acids to activate mTORc1 and as a consequence their overall growth is retarded( Reference Heublein, Kazi and Ogmundsdottir 55 ). As such, it collectively suggests that amino acid transporters may play a role in regulating mTORc1 activity and by virtue, muscle protein metabolism. Although the simplest scenario would be for the amino acid transporters to directly regulate intracellular leucine concentrations ultimately leading to the activation of mTORc1, this does not appear to be the case. By examining the dose–response relationship between muscle protein synthesis and extracellular and intracellular amino acid concentrations, Bohé et al. were able to demonstrate that the anabolic response to amino acids is not responsive to intracellular amino acid availability( Reference Bohe, Low and Wolfe 52 ). Subsequent cell culture experiments using methylaminoisobutyric acid, a non-metabolisable System A amino acid analogue, have demonstrated that SNAT2 appears able to induce p70 S6 K phosphorylation in a rapacmycin-sensitive manner, in spite of reduced intracellular amino acid concentrations( Reference Pinilla, Aledo and Cwiklinski 56 ). This potential ability of amino acid transporters to elicit intracellular signalling events has led to them being termed transceptors (transporter + receptor) and provides credence to their study in the relation to sarcopenia. In particular, an inability of the amino acid transporters to allow sufficient essential amino acids import or initialise signalling events leading to the activation of mTORc1 appear plausible areas of interest.

Although the study of the amino acid transporters in human skeletal muscle is limited, the evidence suggests that LAT1, SNAT2, CD98 and proton-assisted amino acid transporter 1 are transcriptionally up-regulated rapidly (1 h) following essential amino acids ingestion, and that this is paralleled by increases in intracellular leucine concentrations and muscle protein synthesis( Reference Drummond, Glynn and Fry 57 ). However, in the same study, increases in muscle intracellular leucine concentrations and muscle protein synthesis were found to precede detectable increases in LAT1 and SNAT2 protein levels, suggesting that increased transporter protein expression is not involved in the chain of events responsible for increased leucine uptake into the muscle cell or in enhanced muscle protein anabolism post-amino acid consumption. Indeed, the clearance of blood amino acids has been shown to be complete 3 h following meal consumption, suggesting that the changes in transporter protein expression would need to be rapid( Reference Bergstrom, Furst and Vinnars 58 ). Subsequent work in murine muscle cells has demonstrated SNAT2, proton-assisted amino acid transporter 1 and LAT1 to be transcriptionally up-regulated in response to insulin administration, with LAT1 increased in an apparent mTORc1-dependent manner( Reference Walker, Drummond and Dickinson 59 ), providing further evidence that transporter expression may lag behind the stimulation of muscle protein synthesis. Therefore, the changes observed in transporter expression in human muscle following essential amino acids ingestion may merely be a reflection of the known incretin effect of amino acids rather than any direct stimulatory effect on transporter expression to drive anabolism, although this remains to be determined. However, given that intracellular amino acid concentrations are thought to be regulated though the integration of protein synthesis, breakdown, movement into and out of the extracellular pool and where applicable, oxidation( Reference Borsheim, Kobayashi and Traber 51 , Reference Wolfe and Miller 60 ), a role for changes in amino acid transporter expression post-stimulation of muscle protein synthesis may prove important for the maintenance of intracellular amino acid concentrations.

Notably, while the constitutive activation of the insulin-signalling kinase AKT, results in substantial muscle growth in rodents( Reference Bodine, Stitt and Gonzalez 61 ), the effects of insulin and amino acids on human muscle protein synthesis appear distinct. The provision of a high-dose mixed-amino acid infusion concomitant to an insulin clamp designed to maintain serum insulin concentrations at post-absorptive levels has been shown to significantly stimulate muscle protein synthesis( Reference Greenhaff, Karagounis and Peirce 62 ). Moreover, no further enhancement in muscle protein synthesis rates are observed with stepwise increases in serum insulin concentrations despite elevated AKT phosphorylation. Therefore, the ability of amino acids to stimulate muscle protein synthesis appears independent of insulin action. Given the reported role of insulin in stimulating amino acid transporter expression, this would provide further evidence that transcriptional up-regulation of the amino acid transporters may not be a key process in the anabolic response to amino acid feeding. However, caution has to be applied to current findings; transporters by virtue of their function are localised to cellular membranes and in the case of specific transport systems (e.g. system L), have to form heterodimers before they are able to perform transport functions. Measures restricted to ‘readouts’ of mRNA and protein levels for the amino acid transporters are unable to take these key variables into account.

Although the role of altered amino acid transporter expression as a manner in which the cell regulates the stimulation of muscle anabolism remains unclear, impaired amino acid transporter expression may remain a contributor to sarcopenia and/or muscle disuse atrophy. Seven day bed rest in elderly individuals has been shown to prevent the stimulatory effect of essential amino acids on SNAT2 and LAT1 protein expression, concomitant to a partial blunting of mTORc1 phosphorylation and failure to enhance muscle protein synthesis rates over the period examined( Reference Drummond, Dickinson and Fry 63 ). Further work is required to place these findings into the context of sarcopenia; however, studies are currently limited by the availability of experimental tools for the study of amino acid transporter function in human subjects. Until such challenges are overcome the contribution of such findings to the aetiology of sarcopenia will remain unclear.

Impact of inactivity on muscle protein turnover

Like sarcopenia itself, the presence of anabolic resistance has not been universally observed in all elderly individuals. Indeed, some researchers have failed to observe anabolic resistance in their recruited elderly volunteers( Reference Koopman, Walrand and Beelen 26 , Reference Symons, Schutzler and Cocke 64 ), leading some to question the applicability of acute stable-isotope-based methods to detect such changes( Reference Burd, Wall and van Loon 65 ). The crux of their argument centres on two main ideas. Firstly, that the insipid loss of muscle mass with age, predicted to be 1–2 % per year( Reference Frontera, Hughes and Fielding 66 ), is unlikely to be of sufficient magnitude to be detectable by commonly employed stable-isotope approaches. Secondly, they argue that the short timeframes often examined with stable-isotope approaches (≤3 h) fail to take account for the possibility of a delayed rather than reduced anabolic response to amino acid provision. Certainly, a delay in the stimulation of muscle protein synthesis to amino acids has been reported in the elderly, with the cumulative protein synthetic response found to be equivalent between young and elderly subjects when long (5 h) postprandial periods are considered( Reference Drummond, Dreyer and Pennings 67 ) (a process that by its own virtue will diminish the sensitivity required to detect differences). While renewed vigour in the use of deuterium-labelled water to allow the chronic assessment of muscle protein synthesis( Reference Robinson, Turner and Hellerstein 68 , Reference Wilkinson, Franchi and Brook 69 ) should overcome both of these limitations and permit the habitual examination of elderly individuals, an alternative explanation for this discord in the literature may exist. Notably, the vast majority of studies interested in the impact of ageing on muscle protein turnover tend to recruit healthy, non-sarcopenic and active elderly individuals. As such, the anabolic resistance observed in such studies could be primarily a reflection of the activity status of the recruited individuals.

Muscle disuse, induced by bed-rest or limb immobilisation, is widely reported to result in a decline in insulin sensitivity and an impaired anabolic response of muscle to feeding in human subjects( Reference Glover, Phillips and Oates 70 , Reference Wall, Snijders and Senden 71 ). In stark contrast to small-animal models where protein turnover rates are significantly greater than human subjects( Reference Rennie, Selby and Atherton 72 ), current evidence would suggest that the overall contribution of muscle protein breakdown is likely small and transient, probably confined to the first few days post-immobilisation( Reference Glover, Yasuda and Tarnopolsky 73 ); albeit this remains an area of significant controversy( Reference Phillips and McGlory 74 , Reference Reid, Judge and Bodine 75 ). While such gross declines in activity would not be evident in a healthy recruited population, recent evidence has demonstrated that a decline and not necessarily cessation of daily activity is sufficient at inducing significant anabolic resistance in the elderly( Reference Breen, Stokes and Churchward-Venne 76 ). Reducing daily step-count by 76 % for 14 d, from a habitual level of about 5900 to about 1400 steps daily, was shown to reduce leg fat-free mass by 3·9 % and attenuate the rise in postprandial muscle protein synthesis rates by 26 %, independent of mTORc1 signalling. This highlights the need to carefully consider and monitor habitual physical activity levels in recruited subjects, especially as periods of reduced ambulatory activity are common in the elderly. It also highlights the utility of physical activity to maintain muscle mass and provides further credence to its use in this population.

Possible non-pharmacological treatment strategies

While the underlying aetiology for sarcopenia remains to be resolved, the current research has indicated several non-pharmacological approaches that may be applicable to stave off the age-dependent loss of muscle. A major area of focus has been on the use of protein and/or amino acid supplements to promote increased muscle anabolism. While there has been comprehensive discussion of the effectiveness of such approaches to impact on muscle mass, human metabolic studies have revealed several aspects that warrant consideration when developing effective supplementation approaches.

First and foremost, while protein supplementation may be beneficial for individuals not meeting protein intake requirements through habitual means, simply providing large single doses of protein with the premise that you can ‘game the system’ and stimulate anabolism above and beyond the response to standard protein intake is unrealistic. In response to prolonged intravenous infusions of amino acids, muscle protein synthesis is only elevated for the first 2 h of the infusion in young individuals( Reference Bohe, Low and Wolfe 77 ). Somewhat remarkably, rates of muscle protein synthesis subsequently return to post-absorptive values even in the face of persistently elevated blood amino acid concentrations( Reference Bohe, Low and Wolfe 77 ). This has led to the concept of the muscle full hypothesis, a ceiling upon which delivered amino acids are no longer incorporated into muscle protein but rather diverted towards oxidation( Reference Millward and Pacy 78 ). Of concern, the ceiling at which this effect takes place appears equivalent in healthy young and older individuals, with 10 g essential amino acids able to maximally stimulate muscle protein synthesis in both groups, with lower absolute rates of myofibrillar protein synthesis seen in the elderly volunteers( Reference Cuthbertson, Smith and Babraj 33 ). This highlights the potential futility of administering protein in large single doses to elderly individuals. The known properties of protein to impact on satiety are also of concern and it is perhaps not surprising that the use of nutritional supplements in the elderly has been associated with compromising subsequent food intake( Reference Fiatarone Singh, Bernstein and Ryan 79 ). Therefore, the dose at which a protein supplement is administered requires consideration of the balance between providing sufficient substrate to avoid negative nitrogen balance while avoiding the potential to impact on food intake or promote the oxidation of administered amino acids.

At the same time, a certain level of hyperaminoacidaemia is required to adequately stimulate muscle protein synthesis. Pennings et al., by orally administering to elderly men 20 g of either casein, casein hydrosylate, or whey protein on three separate occasions, with the known differences in digestion kinetics between the three proteins utilised, were able to demonstrate a correlation between the peak hyperacidaemia elicited by the test drink and the relative increase in muscle protein synthesis observed( Reference Pennings, Boirie and Senden 9 ). Collectively, this suggests that an optimal window for the quantity of protein consumed in a single sitting exists, within which muscle protein synthesis is meaningfully stimulated while avoiding issues associated with overconsumption (Fig. 2).

Fig. 2. (a) An illustrative example of the benefit of maintaining postprandial hyperleucinaemia within a set threshold. (b) The anticipated consequences of a skewed diurnal pattern of protein intake (- - -) v. a balanced approach to protein intake across meals (___) on blood leucine concentrations.

Such a scenario also has ramifications for the effectiveness of the diurnal pattern in which protein is consumed in the Western world. With standard food habits, protein intake is typically orientated towards the evening meal with examination of the North American diet revealing breakfast containing approximately one-third of the amount of protein contained in the evening meal (13 v. 38 g protein, respectively( Reference Mamerow, Mettler and English 80 )). The skewed manner in which protein is typically consumed may not be optimal, and as a consequence, impinge on the ability of elderly individuals to maintain sufficient anabolic potential to maintain muscle mass. When recreated experimentally over a 7-d period, protein intake spread equally throughout the day in three sets of about 30 g doses results in significantly greater 24 h muscle protein synthesis than 90 g split into 11, 16 and 63 g for the three daily meals( Reference Mamerow, Mettler and English 80 ). This has potential implications for the timing of any protein supplementation strategy, suggesting that they are best consumed around low protein intake meals, most likely breakfast. Utilising such an approach would allow postprandial increase in blood amino acid concentrations to be within the proposed optimum operating window for muscle protein synthesis.

While the utility of resistance exercise to increase muscle mass in the elderly has already been introduced, one aspect where resistance exercise may prove particularly useful is in sensitising the muscle to subsequent amino acid ingestion. The incorporation of resistance exercise during periods of excess amino acid supply has been shown in healthy young individuals to result in greater net muscle protein balance than either resistance exercise or amino acid ingestion alone( Reference Wolfe 81 ). Moreover, a recent meta-analysis of the literature has demonstrated that protein supplementation is able to enhance both muscle mass and strength in response to chronic (>6 weeks) resistance exercise training( Reference Cermak, Res and de Groot 82 ). Therefore, resistance exercise would appear to represent a useful strategy to raise the threshold before which circulating amino acids are no longer incorporated into muscle protein as described in the muscle full hypothesis. In support, Yang et al. by instructing elderly men to perform an acute bout of one-legged resistance exercise prior to the consumption of whey protein, demonstrated that muscle protein synthesis was greater in the exercised leg v. the non-exercised contralateral limb across a range of protein doses( Reference Yang, Breen and Burd 83 ). Moreover, when frail elderly subjects were supplemented with 15 g protein consumed twice daily during a 24-week progressive resistance exercise programme, significant improvements were observed in lean body mass, strength and physical performance( Reference Tieland, Dirks and van der Zwaluw 84 ). Most notably, elderly adults administered with a placebo for the 24-week period in place of the protein supplement saw no detectable gains in muscle mass, albeit similar improvements in strength and physical performance to the protein group were recorded( Reference Tieland, Dirks and van der Zwaluw 84 ).

Reduced mobility and joint pain is a common complaint among the elderly, particularly in frail individuals. As a result, the muscle contractile forces that they can comfortably generate could be potentially compromised and not operating near the typically high-intensity (>70 % of 1-repitition maximum) contractions employed in resistance exercise programmes for the young. Of relevance to affected individuals, recent evidence suggests that high-volume low-intensity resistance exercise is just as effective at eliciting the same degree of muscle protein synthesis as low-volume high-intensity exercise. In particular, young subjects performing leg extension exercise at 30 % of 1-repitition maximum until failure were able to acutely stimulate muscle protein synthesis to the same degree as that observed when exercise was performed at the much higher intensity of 90 % 1-repitition maximum ( Reference Burd, West and Staples 85 ). Likewise, comparable observations have been made in older individuals( Reference Kumar, Selby and Rankin 86 ), thereby suggesting that increasing volume rather than intensity is an effective process by which to stimulate muscle protein synthesis in the elderly and would likely assist in encouraging compliance in this population.

Conclusions

The inability of habitual protein intake to maintain muscle mass in the elderly is potentially the culmination of a number of events impairing the muscle protein synthetic response to food. Current evidence suggests that protein supplementation may be able to overcome these issues, particularly when combined with resistance exercise programmes. The role that inactivity plays in the aetiology of sarcopenia remains unclear, but could transpire to be a major contributor and should be the focus of future work.

Financial Support

This research received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.

Conflicts of Interest

None.

Authorship

A. J. M. was responsible for all aspects related to the authoring of this paper.

References

1. Rantanen, T, Harris, T, Leveille, SG et al. (2000) Muscle strength and body mass index as long-term predictors of mortality in initially healthy men. J Gerontol A Biol Sci Med Sci 55, M168M173.Google Scholar
2. Streja, E, Molnar, MZ, Kovesdy, CP et al. (2011) Associations of pretransplant weight and muscle mass with mortality in renal transplant recipients. Clin J Am Soc Nephrol 6, 14631473.Google Scholar
3. Han, SS, Kim, KW, Kim, KI et al. (2010) Lean mass index: a better predictor of mortality than body mass index in elderly Asians. J Am Geriatr Soc 58, 312317.CrossRefGoogle ScholarPubMed
4. Lee, CG, Boyko, EJ, Nielson, CM et al. (2011) Mortality risk in older men associated with changes in weight, lean mass, and fat mass. J Am Geriatr Soc 59, 233240.Google Scholar
5. Kitamura, I, Koda, M, Otsuka, R et al. (2014) Six-year longitudinal changes in body composition of middle-aged and elderly Japanese: age and sex differences in appendicular skeletal muscle mass. Geriatr Gerontol Int 14, 354361.Google Scholar
6. Lindle, RS, Metter, EJ, Lynch, NA et al. (1997) Age and gender comparisons of muscle strength in 654 women and men aged 20–93 yr. J Appl Physiol 83, 15811587.Google Scholar
7. Sayer, AA (2010) Sarcopenia. BMJ 341, c4097.Google Scholar
8. Heymsfield, SB, Stevens, V, Noel, R et al. (1982) Biochemical composition of muscle in normal and semistarved human subjects: relevance to anthropometric measurements. Am J Clin Nutr 36, 131142.Google Scholar
9. Pennings, B, Boirie, Y, Senden, JM et al. (2011) Whey protein stimulates postprandial muscle protein accretion more effectively than do casein and casein hydrolysate in older men. Am J Clin Nutr 93, 9971005.Google Scholar
10. Volpi, E, Kobayashi, H, Sheffield-Moore, M et al. (2003) Essential amino acids are primarily responsible for the amino acid stimulation of muscle protein anabolism in healthy elderly adults. Am J Clin Nutr 78, 250258.Google Scholar
11. Garlick, PJ (2005) The role of leucine in the regulation of protein metabolism. J Nutr 135, 1553S1556S.Google Scholar
12. Fujita, S, Rasmussen, BB, Cadenas, JG et al. (2006) Effect of insulin on human skeletal muscle protein synthesis is modulated by insulin-induced changes in muscle blood flow and amino acid availability. Am J Physiol Endocrinol Metab 291, E745E754.Google Scholar
13. Frost, RA, Lang, CH (2011) mTor signaling in skeletal muscle during sepsis and inflammation: where does it all go wrong? Physiology 26, 8396.Google Scholar
14. Nader, GA (2005) Molecular determinants of skeletal muscle mass: getting the “AKT” together. Int J Biochem Cell Biol 37, 19851996.Google Scholar
15. Dickinson, JM, Fry, CS, Drummond, MJ et al. (2011) Mammalian target of rapamycin complex 1 activation is required for the stimulation of human skeletal muscle protein synthesis by essential amino acids. J Nutr 141, 856862.Google Scholar
16. Lecker, SH, Jagoe, RT, Gilbert, A et al. (2004) Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression. FASEB J 18, 3951.Google Scholar
17. Altun, M, Besche, HC, Overkleeft, HS et al. (2010) Muscle wasting in aged, sarcopenic rats is associated with enhanced activity of the ubiquitin proteasome pathway. J Biol Chem 285, 3959739608.Google Scholar
18. Wilkes, EA, Selby, AL, Atherton, PJ et al. (2009) Blunting of insulin inhibition of proteolysis in legs of older subjects may contribute to age-related sarcopenia. Am J Clin Nutr 90, 13431350.Google Scholar
19. Morley, JE, Argiles, JM, Evans, WJ et al. (2010) Nutritional recommendations for the management of sarcopenia. J Am Med Dir Assoc 11, 391396.Google Scholar
20. Boyce, JM, Shone, GR (2006) Effects of ageing on smell and taste. Postgrad Med J 82, 239241.Google Scholar
21. Campbell, WW, Trappe, TA, Wolfe, RR et al. (2001) The recommended dietary allowance for protein may not be adequate for older people to maintain skeletal muscle. J Gerontol A Biol Sci Med Sci 56, M373M380.Google Scholar
22. Campbell, WW, Crim, MC, Dallal, GE et al. (1994) Increased protein requirements in elderly people: new data and retrospective reassessments. Am J Clin Nutr 60, 501509.Google Scholar
23. Houston, DK, Nicklas, BJ, Ding, J et al. (2008) Dietary protein intake is associated with lean mass change in older, community-dwelling adults: the health, aging, and body composition (Health ABC) study. Am J Clin Nutr 87, 150155.Google Scholar
24. Saffrey, MJ (2014) Aging of the mammalian gastrointestinal tract: a complex organ system. Age 36, 9603.Google Scholar
25. Pennings, B, Pellikaan, WF, Senden, JM et al. (2011) The production of intrinsically labeled milk and meat protein is feasible and provides functional tools for human nutrition research. J Dairy Sci 94, 43664373.Google Scholar
26. Koopman, R, Walrand, S, Beelen, M et al. (2009) Dietary protein digestion and absorption rates and the subsequent postprandial muscle protein synthetic response do not differ between young and elderly men. J Nutr 139, 17071713.Google Scholar
27. Volpi, E, Mittendorfer, B, Wolf, SE et al. (1999) Oral amino acids stimulate muscle protein anabolism in the elderly despite higher first-pass splanchnic extraction. Am J Physiol 277, E513E520.Google Scholar
28. Boirie, Y, Gachon, P, Beaufrere, B (1997) Splanchnic and whole-body leucine kinetics in young and elderly men. Am J Clin Nutr 65, 489495.Google Scholar
29. Pennings, B, Groen, BB, van Dijk, JW et al. (2013) Minced beef is more rapidly digested and absorbed than beef steak, resulting in greater postprandial protein retention in older men. Am J Clin Nutr 98, 121128.Google Scholar
30. Gilani, GS, Sepehr, E (2003) Protein digestibility and quality in products containing antinutritional factors are adversely affected by old age in rats. J Nutr 133, 220225.Google Scholar
31. Gilani, GS, Cockell, KA, Sepehr, E (2005) Effects of antinutritional factors on protein digestibility and amino acid availability in foods. J AOAC Int 88, 967987.Google Scholar
32. Guillet, C, Prod'homme, M, Balage, M et al. (2004) Impaired anabolic response of muscle protein synthesis is associated with S6K1 dysregulation in elderly humans. FASEB J 18, 15861587.Google Scholar
33. Cuthbertson, D, Smith, K, Babraj, J et al. (2005) Anabolic signaling deficits underlie amino acid resistance of wasting, aging muscle. FASEB J 19, 422424.CrossRefGoogle ScholarPubMed
34. Vollenweider, P, Tappy, L, Randin, D et al. (1993) Differential effects of hyperinsulinemia and carbohydrate metabolism on sympathetic nerve activity and muscle blood flow in humans. J Clin Invest 92, 147154.Google Scholar
35. Laakso, M, Edelman, SV, Brechtel, G et al. (1990) Decreased effect of insulin to stimulate skeletal muscle blood flow in obese man. A novel mechanism for insulin resistance. J Clin Invest 85, 18441852.Google Scholar
36. Karakelides, H, Irving, BA, Short, KR et al. (2010) Age, obesity, and sex effects on insulin sensitivity and skeletal muscle mitochondrial function. Diabetes 59, 8997.Google Scholar
37. Meneilly, GS, Elliot, T, Bryer-Ash, M et al. (1995) Insulin-mediated increase in blood flow is impaired in the elderly. J Clin Endocrinol Metab 80, 18991903.Google Scholar
38. Volpi, E, Mittendorfer, B, Rasmussen, BB et al. (2000) The response of muscle protein anabolism to combined hyperaminoacidemia and glucose-induced hyperinsulinemia is impaired in the elderly. J Clin Endocrinol Metab 85, 44814490.Google Scholar
39. Phillips, B, Williams, J, Atherton, P et al. (2012) Resistance exercise training improves age-related declines in leg vascular conductance and rejuvenates acute leg blood flow responses to feeding and exercise. J Appl Physiol 112, 347353.Google Scholar
40. Steinberg, HO, Brechtel, G, Johnson, A et al. (1994) Insulin-mediated skeletal muscle vasodilation is nitric oxide dependent. A novel action of insulin to increase nitric oxide release. J Clin Invest 94, 11721179.Google Scholar
41. Dillon, EL, Casperson, SL, Durham, WJ et al. (2011) Muscle protein metabolism responds similarly to exogenous amino acids in healthy younger and older adults during NO-induced hyperemia. Am J Physiol Regul Integr Comp Physiol 301, R1408R1417.Google Scholar
42. Kelly, J, Fulford, J, Vanhatalo, A et al. (2013) Effects of short-term dietary nitrate supplementation on blood pressure, O2 uptake kinetics, and muscle and cognitive function in older adults. Am J Physiol Regul Integr Comp Physiol 304, R73R83.Google Scholar
43. Joris, PJ, Mensink, RP (2013) Beetroot juice improves in overweight and slightly obese men postprandial endothelial function after consumption of a mixed meal. Atherosclerosis 231, 7883.Google Scholar
44. Paddon-Jones, D, Borsheim, E, Wolfe, RR (2004) Potential ergogenic effects of arginine and creatine supplementation. J Nutr 134, 2888S2894S; discussion 95S.Google Scholar
45. Tang, JE, Lysecki, PJ, Manolakos, JJ et al. (2011) Bolus arginine supplementation affects neither muscle blood flow nor muscle protein synthesis in young men at rest or after resistance exercise. J Nutr 141, 195200.Google Scholar
46. Churchward-Venne, TA, Cotie, LM, MacDonald, MJ et al. (2014) Citrulline does not enhance blood flow, microvascular circulation, or myofibrillar protein synthesis in elderly men at rest or following exercise. Am J Physiol Endocrinol Metab 307, E71E83.Google Scholar
47. Donato, AJ, Uberoi, A, Wray, DW et al. (2006) Differential effects of aging on limb blood flow in humans. Am J Physiol Heart Circ Physiol 290, H272H278.Google Scholar
48. Poole, JG, Lawrenson, L, Kim, J et al. (2003) Vascular and metabolic response to cycle exercise in sedentary humans: effect of age. Am J Physiol Heart Circ Physiol 284, H1251H1259.Google Scholar
49. Fiatarone, MA, O'Neill, EF, Ryan, ND et al. (1994) Exercise training and nutritional supplementation for physical frailty in very elderly people. N Engl J Med 330, 17691775.Google Scholar
50. Atherton, PJ, Smith, K, Etheridge, T et al. (2010) Distinct anabolic signalling responses to amino acids in C2C12 skeletal muscle cells. Amino Acids 38, 15331539.Google Scholar
51. Borsheim, E, Kobayashi, H, Traber, DL et al. (2006) Compartmental distribution of amino acids during hemodialysis-induced hypoaminoacidemia. Am J Physiol Endocrinol Metab 290, E643E652.Google Scholar
52. Bohe, J, Low, A, Wolfe, RR et al. (2003) Human muscle protein synthesis is modulated by extracellular, not intramuscular amino acid availability: a dose–response study. J Physiol 552, 315324.Google Scholar
53. Tipton, KD, Ferrando, AA, Phillips, SM et al. (1999) Postexercise net protein synthesis in human muscle from orally administered amino acids. Am J Physiol 276, E628E634.Google Scholar
54. Ogmundsdottir, MH, Heublein, S, Kazi, S et al. (2012) Proton-assisted amino acid transporter PAT1 complexes with Rag GTPases and activates TORC1 on late endosomal and lysosomal membranes. PLoS ONE 7, e36616.Google Scholar
55. Heublein, S, Kazi, S, Ogmundsdottir, MH et al. (2010) Proton-assisted amino-acid transporters are conserved regulators of proliferation and amino-acid-dependent mTORC1 activation. Oncogene 29, 40684079.CrossRefGoogle ScholarPubMed
56. Pinilla, J, Aledo, JC, Cwiklinski, E et al. (2011) SNAT2 transceptor signalling via mTOR: a role in cell growth and proliferation? Front Biosci 3, 12891299.Google Scholar
57. Drummond, MJ, Glynn, EL, Fry, CS et al. (2010) An increase in essential amino acid availability upregulates amino acid transporter expression in human skeletal muscle. Am J Physiol Endocrinol Metab 298, E1011E1018.Google Scholar
58. Bergstrom, J, Furst, P, Vinnars, E (1990) Effect of a test meal, without and with protein, on muscle and plasma free amino acids. Clin Sci (Lond) 79, 331337.Google Scholar
59. Walker, DK, Drummond, MJ, Dickinson, JM et al. (2014) Insulin increases mRNA abundance of the amino acid transporter SLC7A5/LAT1 via an mTORC1-dependent mechanism in skeletal muscle cells. Physiol Rep 2, e00238.Google Scholar
60. Wolfe, RR, Miller, SL (1999) Amino acid availability controls muscle protein metabolism. Diab Nutr Metab 12, 322328.Google Scholar
61. Bodine, SC, Stitt, TN, Gonzalez, M et al. (2001) Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol 3, 10141019.Google Scholar
62. Greenhaff, PL, Karagounis, L, Peirce, N et al. (2008) Disassociation between the effects of amino acids and insulin on signalling, ubiquitin-ligases and protein turnover in human muscle. Am J Physiol Endocrinol Metab 295, E595E604.Google Scholar
63. Drummond, MJ, Dickinson, JM, Fry, CS et al. (2012) Bed rest impairs skeletal muscle amino acid transporter expression, mTORC1 signaling, and protein synthesis in response to essential amino acids in older adults. Am J Physiol Endocrinol Metab 302, E1113E1122.Google Scholar
64. Symons, TB, Schutzler, SE, Cocke, TL et al. (2007) Aging does not impair the anabolic response to a protein-rich meal. Am J Clin Nutr 86, 451456.Google Scholar
65. Burd, NA, Wall, BT, van Loon, LJ (2012) The curious case of anabolic resistance: old wives’ tales or new fables? J Appl Physiol 112, 12331235.Google Scholar
66. Frontera, WR, Hughes, VA, Fielding, RA et al. (2000) Aging of skeletal muscle: a 12-yr longitudinal study. J Appl Physiol 88, 13211326.Google Scholar
67. Drummond, MJ, Dreyer, HC, Pennings, B et al. (2008) Skeletal muscle protein anabolic response to resistance exercise and essential amino acids is delayed with aging. J Appl Physiol 104, 14521461.Google Scholar
68. Robinson, MM, Turner, SM, Hellerstein, MK et al. (2011) Long-term synthesis rates of skeletal muscle DNA and protein are higher during aerobic training in older humans than in sedentary young subjects but are not altered by protein supplementation. FASEB J 25, 32403249.Google Scholar
69. Wilkinson, DJ, Franchi, MV, Brook, MS et al. (2014) A validation of the application of D(2)O stable isotope tracer techniques for monitoring day-to-day changes in muscle protein subfraction synthesis in humans. Am J Physiol Endocrinol Metab 306, E571E579.Google Scholar
70. Glover, EI, Phillips, SM, Oates, BR et al. (2008) Immobilization induces anabolic resistance in human myofibrillar protein synthesis with low and high dose amino acid infusion. J Physiol 586, 60496061.Google Scholar
71. Wall, BT, Snijders, T, Senden, JM et al. (2013) Disuse impairs the muscle protein synthetic response to protein ingestion in healthy men. J Clin Endocrinol Metab 98, 48724881.Google Scholar
72. Rennie, MJ, Selby, A, Atherton, P et al. (2010) Facts, noise and wishful thinking: muscle protein turnover in aging and human disuse atrophy. Scand J Med Sci Sports 20, 59.Google Scholar
73. Glover, EI, Yasuda, N, Tarnopolsky, MA et al. (2010) Little change in markers of protein breakdown and oxidative stress in humans in immobilization-induced skeletal muscle atrophy. Appl Physiol Nutr Metab 35, 125133.Google Scholar
74. Phillips, SM, McGlory, C (2014) Rebuttal from Stuart M. Phillips and Chris McGlory. J Physiol 592, 5349.Google Scholar
75. Reid, MB, Judge, AR, Bodine, SC (2014) CrossTalk opposing view: the dominant mechanism causing disuse muscle atrophy is proteolysis. J Physiol 592, 53455347.Google Scholar
76. Breen, L, Stokes, KA, Churchward-Venne, TA et al. (2013) Two weeks of reduced activity decreases leg lean mass and induces ‘anabolic resistance’ of myofibrillar protein synthesis in healthy elderly. J Clin Endocrinol Metab 98, 26042612.Google Scholar
77. Bohe, J, Low, JF, Wolfe, RR et al. (2001) Latency and duration of stimulation of human muscle protein synthesis during continuous infusion of amino acids. J Physiol 532, 575579.Google Scholar
78. Millward, DJ, Pacy, PJ (1995) Postprandial protein utilization and protein quality assessment in man. Clin Sci (Lond) 88, 597606.Google Scholar
79. Fiatarone Singh, MA, Bernstein, MA, Ryan, AD et al. (2000) The effect of oral nutritional supplements on habitual dietary quality and quantity in frail elders. J Nutr Health Aging 4, 512.Google Scholar
80. Mamerow, MM, Mettler, JA, English, KL et al. (2014) Dietary protein distribution positively influences 24-h muscle protein synthesis in healthy adults. J Nutr 144, 876880.Google Scholar
81. Wolfe, RR (2006) Skeletal muscle protein metabolism and resistance exercise. J Nutr 136, 525S528S.Google Scholar
82. Cermak, NM, Res, PT, de Groot, LC et al. (2012) Protein supplementation augments the adaptive response of skeletal muscle to resistance-type exercise training: a meta-analysis. Am J Clin Nutr 96, 14541464.Google Scholar
83. Yang, Y, Breen, L, Burd, NA et al. (2012) Resistance exercise enhances myofibrillar protein synthesis with graded intakes of whey protein in older men. Br J Nutr 108, 17801788.CrossRefGoogle ScholarPubMed
84. Tieland, M, Dirks, ML, van der Zwaluw, N et al. (2012) Protein supplementation increases muscle mass gain during prolonged resistance-type exercise training in frail elderly people: a randomized, double-blind, placebo-controlled trial. J Am Med Dir Assoc 13, 713719.Google Scholar
85. Burd, NA, West, DW, Staples, AW et al. (2010) Low-load high volume resistance exercise stimulates muscle protein synthesis more than high-load low volume resistance exercise in young men. PLoS ONE 5, e12033.Google Scholar
86. Kumar, V, Selby, A, Rankin, D et al. (2009) Age-related differences in the dose-response relationship of muscle protein synthesis to resistance exercise in young and old men. J Physiol 587, 211217.Google Scholar
Figure 0

Fig. 1. A simplified diagram representing the role of mammalian target of rapamycin complex 1 (mTORc1) signalling in regulating muscle protein synthesis in response to extracellular cues. SNAT, sodium-coupled neutral amino acid transporter; LAT1, L-type amino acid transporter 1; IRS-1, insulin receptor substrate-1; Gln, glutamine; Leu, leucine; 4EBP1, eukaryotic initiation factor 4E binding protein 1.

Figure 1

Fig. 2. (a) An illustrative example of the benefit of maintaining postprandial hyperleucinaemia within a set threshold. (b) The anticipated consequences of a skewed diurnal pattern of protein intake (- - -) v. a balanced approach to protein intake across meals (___) on blood leucine concentrations.