Hostname: page-component-76fb5796d-x4r87 Total loading time: 0 Render date: 2024-04-25T14:20:51.341Z Has data issue: false hasContentIssue false

Selected B vitamins and their possible link to the aetiology of age-related sarcopenia: relevance of UK dietary recommendations

Published online by Cambridge University Press:  09 March 2018

N. Aytekin
Affiliation:
School of Applied Sciences, London South Bank University, 103 Borough Road, London SE1 0AA, UK
K. N. Mileva
Affiliation:
School of Applied Sciences, London South Bank University, 103 Borough Road, London SE1 0AA, UK
A. D. Cunliffe*
Affiliation:
School of Applied Sciences, London South Bank University, 103 Borough Road, London SE1 0AA, UK
*
*Corresponding author: Dr Adam Cunliffe, email cunliffa@lsbu.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

The possible roles of selected B vitamins in the development and progression of sarcopenia are reviewed. Age-related declines in muscle mass and function are associated with huge and increasing costs to healthcare providers. Falls and loss of mobility and independence due to declining muscle mass/function are associated with poor clinical outcomes and their prevention and management are attractive research targets. Nutritional status appears a key modifiable and affordable intervention. There is emerging evidence of sarcopenia being the result not only of diminished anabolic activity but also of declining neurological integrity in older age, which is emerging as an important aspect of the development of age-related decline in muscle mass/function. In this connection, several B vitamins can be viewed as not only cofactors in muscle synthetic processes, but also as neurotrophic agents with involvements in both bioenergetic and trophic pathways. The B vitamins thus selected are examined with respect to their relevance to multiple aspects of neuromuscular function and evidence is considered that requirements, intakes or absorption may be altered in the elderly. In addition, the evidence base for recommended intakes (UK recommended daily allowance) is examined with particular reference to original datasets and their relevance to older individuals. It is possible that inconsistencies in the literature with respect to the nutritional management of sarcopenia may, in part at least, be the result of compromised micronutrient status in some study participants. It is suggested that in order, for example, for intervention with amino acids to be successful, underlying micronutrient deficiencies must first be addressed/eliminated.

Type
Review Article
Copyright
© The Authors 2018 

Context

The term ‘sarcopenia’ was coined in 1989 to describe an age-associated process characterised by a decline in human skeletal muscle mass( Reference Rosenberg 1 ). This definition has been extended to include the associated loss of muscle function (strength and power), which represents the primary qualitative concern due to its link with frailty( Reference Cruz-Jentoft, Baeyens and Bauer 2 Reference Landi, Calvani and Cesari 4 ). Sarcopenia is also described by a measure of total lean mass ≥2 sd below that of a comparable young adult, distinguished by a reduction in muscle fibre number and cross-sectional area( Reference Cruz-Jentoft, Baeyens and Bauer 2 ). This loss is often described as irreversible and associated with a significant functional deficit, leading to a loss of physical independence( Reference Landi, Calvani and Cesari 4 , Reference dos Santos, Cyrino and Antunes 5 ). Sarcopenia is associated with an increased risk of falls and related fractures( Reference Landi, Liperoti and Russo 6 , Reference Clynes, Edwards and Buehring 7 ).

The population in the UK is ageing; the elderly sector of the population is increasing independent of the overall increase in population( 8 ). Similar shifts are being observed in other European Union (EU) countries. Taken together, 18–21 % of the European population were found to be aged 65 years or over and this range is expected to rise to 19–31 % by 2035, which translates to >25 % of all Europeans being aged 65 years or over by that date( 8 ). There are at present few initiatives or therapeutic protocols in place to manage sarcopenia at the level of the general population. Research is emerging that indicates that the lean tissue decrements and increased frailty associated with ageing may be relatively manipulable phenomena, with nutrition as a key modifiable variable.

Nutritional interventions, for example with essential amino acids, have produced rather variable results, with some interventions appearing effective and others of limited value( Reference Paddon-Jones, Sheffield-Moore and Zhang 9 Reference Cuthbertson, Smith and Babraj 11 ). In such research, elderly sarcopenic subjects frequently present with multiple, potentially confounding factors. In this connection, the present discussion will examine the potential role of selected B vitamins in normal and pathophysiological modes of neuromuscular function. Several key themes will be explored, notably issues associated with the adequacy of the knowledge base regarding older adults’ requirements and intakes for B vitamins, the similarities between certain deficiency and frailty states associated with older age and finally the potential for suboptimal micronutrient status to mimic/mask features of sarcopenia.

Nutritional challenges in later life

Age-related features underlying compromise of intake or processing of nutrients are manifold and range from the psychosocial through to the physiological. In later life, issues of reduced income, mobility and socialisation may conflate to produce a reduction in energy and nutrient intake per se ( Reference Kant and Graubard 12 , Reference Saka, Kaya and Ozturk 13 ). Problems with dental health may impair intake regardless of food availability( Reference Han and Kim 14 , Reference Kagawa, Ikebe and Inomata 15 ), while changes in taste perception can also occur with ageing( Reference Mojet 16 ). In addition, the elderly may experience difficulty interpreting labelling information( Reference Kim, Oh and No 17 ), which, combined with economic limitations, may present the older adult with fewer appetising meal options. Many elderly regularly take medications and these can alter the uptake, metabolism and/or excretion of key nutrients( Reference Valuck and Ruscin 18 , Reference Keller 19 ). Disease states such as cancer, diabetes and gastrointestinal inflammatory disorders also make a direct impact on nutritional status. There is considerable evidence that the elderly are at significant risk of two or more micronutrient deficiencies( Reference Sharkey, Branch and Zohoori 20 Reference Marshall, Stumbo and Warren 23 ) and it has been established that there is a clear association between multiple micronutrient deficiencies and frailty( Reference Bartali, Frongillo and Bandinelli 24 , Reference Semba, Bartali and Zhou 25 ). The elderly therefore represent an ‘at-risk’ group for malnutrition and deficiency states that may have causal or aggravating connections to sarcopenia. With respect to the very elderly (aged 85+ years), a study in the UK found lower than estimated average requirements for energy and highlighted the lack of dietary intake data and the uncertainties about dietary reference values for very old adults( Reference Mendonça, Hill and Granic 26 ).

B vitamins: rationale for review

The rationale for selecting B vitamins is as follows. First, B vitamins are involved in multiple aspects of energy and protein metabolism, and also in multiple aspects of neural integrity and function. Second, deficiencies of B vitamins may manifest in obvious neuromuscular problems (for example, beri beri) and/or neurological symptoms (for example, pellagra, peripheral neuropathies). Some of these signs and symptoms could mask or exaggerate key features of a number of age-related syndromes such as sarcopenia. Finally, in older adults, possible suboptimal dietary intakes of B vitamins, issues of impaired absorption and age-related anorexia potentially combine to produce a sector of the population for which B vitamin deficency is a reality. The long-term effects of subclinical deficiencies remain poorly understood.

The aim of the present text therefore is to review known functions of selected B vitamins in connection with sarcopenia and to assess the likelihood of an interactive or additive effect of suboptimal B vitamin status in the initiation, progression or extent of age-related decrements in muscle mass and function. The B vitamins that will be examined were selected on the basis of their known roles in relevant physiological and metabolic processes and their potential for suboptimal intake or status in older adults.

With respect to determination of sufficiency or otherwise, issues of intake are considered alongside biochemical indices where available, and contextualised through the lens of UK recommended daily allowances (RDA), which are the values that form the basis of advice and evaluation by most of the UK-based science, health and public health organisations. Differences exist between the UK RDA and the European Food Safety Authority (EFSA) recommended daily intakes of B vitamins for the elderly, with EFSA values being higher in almost all cases. Differences also exist between the values cited by EFSA and several European national guidelines, including those set by the partner institutions of the German Nutrition Society (Die Deutsche Gesellschaft für Ernährung e. V. (DGE)), the Austrian Nutrition Society (AGE) and the Swiss Nutrition Society (SGE). The present review therefore explores the rationale for the current UK RDA in particular.

The UK National Diet and Nutrition Survey (NDNS)( 27 ), with respect to the elderly, presents varying degrees of detail in terms of B vitamin status, which is in some cases described by intake, in other cases by plasma levels and in others by functional assays. While the latest data appear to suggest a generally positive picture in terms of B vitamin sufficiency for those living in the UK, there are some important caveats. First, while the NDNS provides important, up-to-date insight into the nutritional status of the UK population, its findings are given meaning by national dietary guidelines, which are often at considerable variance with estimated requirements for nearby, comparable, modern industrial societies. Second, whilst the NDNS groups ‘the elderly’ within the single age bracket of ≥65+ years, in many instances the authors acknowledge age-related declines in the nutritional status of older adults, i.e. from 65 years onwards. As such, the stated guidelines for older adults would appear to lose applicability with advancing age. In this connection, the latest survey (2014/2015) does not include samples from non-community dwelling (i.e. institutionalised) elderly populations; the data presented for this survey are only for free-living individuals. This may in part explain the discrepancies that exist between the latest results and the results of the previous study (which indicated a greater prevalence of deficiency), since the latter included individuals living in institutions as well as the free-living. As there is an increasing number of elderly individuals in care, and since the non-community-dwelling elderly represent a group at high risk of malnutrition, this may well constitute an important oversight; one that may distort the perceived landscape of nutritional health in the UK elderly population.

More generally, assessment of nutritional status in the elderly is for the most part based on nutritional intake data, for example, from FFQ, food diaries and food purchasing data. Whilst such methods have utility in assessing patterns of consumption and estimating energy intake, their application in determining micronutrient status in the elderly is limited as follows. In general terms, these methods are not reflective of intra-population requirements or rates of utilisation, do not account for the significant changes in micronutrient availability due to variation in storage, processing or cooking methods, nor for antagonisms between different nutrients and/or non-nutritive substances in food. These methods may also be limited in terms of accuracy, since they may rely either on recall (FFQ), present a high respondent burden (food diaries) or represent an estimation (purchase data). In addition, dietary records and questionnaires do not accurately capture supplement use in the elderly( Reference Bates, Prentice and van der Pols 28 ), which, research suggests, may be substantial( Reference Fabian, Bogner and Kickinger 29 Reference Bates, Lennox and Prentice 31 ). Thus it is possible that reported average B vitamin intake, at least in some studies, may be misleading.

More important perhaps is the conceptual framework by which the reference guidelines seek to specify deficiency prophylaxis. There may well be important differentials between levels of intake associated with deficit symptom prevention and optimal metabolic status and function. Analyses using additional biochemical measures have shown nutrient intake data to lack validity in determining micronutrient status in elderly populations( Reference Bailey, Maisey and Southon 32 ).

Sarcopenia appears to be a multi-factorial pathological process, which, although associated with age, is not a de facto outcome of ageing. Since B vitamins are directly and indirectly involved in the operation of an array of biological systems, and since these systems appear to be sensitive to even relatively short-term deficiencies, vitamin contribution to the complex aetiology of sarcopenia is likely to be significant and may be modifiable in relation to its progression.

Thiamin (vitamin B1)

Functions and the effects of deficiency

Important food sources of vitamin B1 include yeasts, whole-grain cereals (B vitamin content being concentrated in the outer germ layer of grains), nuts and legumes, pork meat and organ meat (particularly the liver, kidneys and heart)( Reference Tanphaicitr 33 , Reference Scheid, Bennett and Schweigert 34 ). Vitamin B1 is present in the human body in different forms. The best characterisation of vitamin B1 is as thiamin pyrophosphate (TPP) or thiamin diphosphate (ThDP) in its twice-phosphorylated, activated, cofactor form. Poorly characterised forms include adenosine thiamin triphosphate (AThTP), thiamin monophosphate (ThMP) and thiamin triphosphate (ThTP). As TPP, vitamin B1 is principally regarded as a key enzymic cofactor in oxidative metabolism( Reference Martin, Singleton and Hiller-Sturmhöfel 35 ). More specifically, vitamin B1 is known to function in twenty-four enzymic reactions, most importantly pyruvate dehydrogenase (for energy production via the Krebs cycle), transketolase (for lipid and glucose metabolism, production of branched-chain amino acids, and production and maintenance of the myelin sheath) and 2-oxoglutarate dehydrogenase (for synthesis of acetylcholine, γ-aminobutyric acid (GABA) and glutamate)( Reference Thomson and Pratt 36 ).

Vitamin B1 is known to be directly associated with nervous system function, primarily as a result of observation of rapid structural and functional declines in deficiency states and in alcoholism( Reference Williams 37 , Reference Phillips, Victor and Adams 38 ). Whilst the sensitivity of the nervous system to vitamin B1 is largely attributed to the heavy reliance of this system on oxidative metabolism( Reference Rindi 39 ), region-specific sensitivity to vitamin B1 deficiency has been observed in neuronal tissues that have otherwise comparable metabolic profiles( Reference Calingasan, Chun and Park 40 ). This sensitivity may in part be explained by the additional, non-coenzyme functions of vitamin B1, particularly as an antioxidant, i.e. in relation to varying regional susceptibility/exposure to oxidative stress( Reference Lukienko, Mel’nichenko and Zverinskii 41 , Reference Gibson and Zhang 42 ).

Chronic vitamin B1 deficiency is associated with several potentially life-threatening neurological disorders. Whilst vitamin B1 deficiency is frequently associated with the central nervous system (CNS), the most advanced neurological changes have been shown to occur in the periphery – particularly in the lower limbs( 43 ). This finding mirrors an aetiological feature of sarcopenia, for which muscle mass and function in the lower limbs are relatively more compromised( Reference Janssen, Heymsfield and Wang 44 , Reference Nogueira, Libardi and Vechin 45 ). Since muscle fibres function within the context of motor units, such an overlap is unsurprising.

Early signs of vitamin B1 deficiency are cognitive decline, loss of appetite, weight loss/loss of lean mass, reduced walking speed, abnormal gait and muscle weakness/tremors( 46 ). Though poorly delineated, these symptoms are relevant and represent the least easily detected (by clinical appraisal) and most likely manifestations of vitamin B1 deficiency in otherwise healthy elderly. Vitamin B1 deficiency is being increasingly associated with loss of vibratory sensation in the lower extremities( Reference Wilkinson, Hanger and George 47 ), a higher incidence of falls over time( Reference Pepersack, Garbusinski and Robberecht 48 ) and depression( Reference Zhang, Ding and Chen 49 ). A low vitamin B1 status is also increasingly implicated in other age-related neurodegenerative disorders such as Alzheimer’s disease( Reference Gold, Hauser and Chen 50 , Reference Pan, Fei and Lu 51 ).

A number of different mechanisms have been put forward to explain neuronal loss/damage during thiamin deficiency (TD). The mechanisms proposed relate chiefly to the reduced activity of vitamin B1-dependent enzymes in mitochondria and include impairments in oxidative metabolism( Reference Aikawa, Watanabe and Furuse 52 ), and mitochondrial function( Reference Bettendorff, Sluse and Goessens 53 ), resulting in inflammatory responses associated with microglial activation( Reference Ke, DeGiorgio and Volpe 54 , Reference Todd and Butterworth 55 ), increased production of reactive oxygen species (ROS)( Reference Langlais, Anderson and Guo 56 ), and later, glutamate receptor-mediated excitotoxicity( Reference Todd and Butterworth 57 , Reference Langlais and Mair 58 ). These mechanisms have been elucidated through investigations using cell lines, following/leading to observations of CNS damage during experimental vitamin B1 depletion in animals.

In addition to providing mechanistic insight, these studies have elucidated important temporal dimensions in TD, and suggest a need for further research into the effects of episodic and cumulative suboptimal vitamin B1 status in human subjects. For example, Ke et al. ( Reference Ke, DeGiorgio and Volpe 54 ) found that a moderate-low vitamin B1 intake in mice triggered an immune response, activating microglia and resulting in widespread loss of neural tissue (29 %) in vitamin B1-sensitive regions within 9 d of TD. Whilst intervention with vitamin B1 on day 8 of TD was shown to prevent further damage, the commitment of neuronal cells to apoptotic pathways appeared to be irreversible by day 10–11, when a 90 % loss of neural tissue was observed( Reference Ke, DeGiorgio and Volpe 54 ).

Prevalence of deficiency

The UK NDNS reports that the average intake of vitamin B1 exceeds the reference nutrient intake (RNI) across all age groups( 27 ). Audits in other European countries have largely produced similar findings. However, recent large-scale, cross-European investigations have indicated important regional variations( Reference Olsen, Halkjaer and van Gils 59 , Reference Fabian and Elmadfa 60 ). Other large-scale and smaller-scale studies of free-living elderly populations demonstrate significant numbers who have inadequate intake (up to 60 %)( Reference Jyväkorpi, Pitkälä and Puranen 61 Reference ter Borg, Verlaan and Hemsworth 64 ), possibly due to more rigorous methodological approaches or biases resulting from the age ranges selected, socio-economic or geographical factors( Reference Lim and Cho 65 ). Inadequate intake in non-free-living elderly populations is reported to be widespread in industrialised nations: between 33 and 94 %( Reference Lengyel, Whiting and Zello 66 Reference McCabe-Sellers, Sharkey and Browne 70 ). Where subject age is reported as a continuum, vitamin B1 intake appears to be negatively correlated with this variable( Reference Wilkinson, Hanger and George 47 , Reference Wakimoto and Block 71 ). Interestingly, supplemental intake, which has been reported to make a major (>50 %) contribution to overall intake( Reference Sette, Le Donne and Piccinelli 72 ) is often not included in dietary intake studies.

As with other B vitamins, variation in the reported prevalence of deficiency is partly due to the use of different referencing guidelines, for example, minimum requirements v. two-thirds of the RDA( Reference De Groot, Van Den Broek and Van Staveren 21 ). Furthermore, assessments of intake frequently do not take into account a potential relative deficit of vitamin B1 induced by consumption of refined carbohydrates, which require vitamin B1 for their metabolism( Reference Rindi 39 , Reference Elmadfa, Majchrzak and Rust 73 ). Storage, cooking methods and co-ingestion of foodstuffs such as alcohol, sulfites, tannins and o-diphenols (from coffee), and widely used prescription drugs such as diuretics( Reference Suter, Haller and Hany 74 ), that reduce vitamin B1 content and bioavailability( Reference Severi, Bedogni and Manzieri 75 Reference Butterworth, Kril and Harper 77 ) are seldom considered. It is unsurprising therefore that studies comparing assessments of intake with biochemical markers find little correlation, with one study demonstrating biochemical deficiency (TPP effect >14 %) in more than 50 % of a sample who had an intake exceeding guideline requirements (RNI)( Reference Nichols and Basu 78 ).

Biochemical data from elderly populations are scant and mainly related to specific pathological states. Results from the UK NDNS indicate that less than 1·2 % of the elderly are deficient in vitamin B1, based on a measure of transketolase activity (TKA); erythrocyte transketolase activation coefficient of >1·25( 27 ). This cut-off point is less conservative than that suggested by other authorities, including EFSA (>1·15)( Reference Turck, Bresson and Burlingame 79 ). Studies on free-living elderly populations, which employ a cut-off point of >1·15, have indicated a greater prevalence of vitamin B1 deficiency: between 10 and 47 %( Reference Fabian, Bogner and Kickinger 29 , Reference Bailey, Maisey and Southon 32 , Reference Pepersack, Garbusinski and Robberecht 48 , Reference Andrade Juguan, Lukito and Schultink 80 , Reference Wolters, Hermann and Hahn 81 ). The reported range does not appear to be greater in non-free-living elderly populations. In view of these findings, it is interesting to note that whilst most reports, including that of the UK NDNS, consider ‘the elderly’ as a single age bracket having reduced requirements, biochemical data indicate a significant age-related decline in the vitamin B1 status of even apparently ‘healthy elderly’( Reference Wilkinson, Hanger and George 47 ).

UK dietary recommendations

Guidelines for vitamin B1 intake in the UK currently indicate a requirement of 0·9 and 0·8 mg/d for men and women aged ≥50 years, respectively( 82 ). These represent the RNI, and have largely been determined according to measures of minimum vitamin B1 requirements/4184 kJ (1000 kcal) energy intake( 82 ). Whilst these recommendations are safeguarded by established minimum vitamin B1 requirements, associated urinary output measures for the prevention of beri beri and/or signs of clinical TD, not enough consideration appears to have been given as yet to the elderly in this respect, and only one of the studies referred to appears to have included elderly participants. More importantly perhaps was the nature of this study: experimental depletion/repletion of a small sample (n 21) of institutionalised male psychiatric patients( Reference Horwitt, Liebert and Kreisler 83 ). Given the known association of neuropsychiatric illness with vitamin B1 status, a history of mental illness at study baseline may limit the broader contemporary relevance of this work. On the other hand, this research does present some interesting suggestions in that while small age-related differences in glucose absorption rates were found, functional differences in vitamin B1 status (via lactate and pyruvate accumulation) were in evidence according to younger–older subject status. The study does not, however, go on to discuss any precise mechanistic role for vitamin B1 status to explain these observations. In the absence of such, and given the limited scope of the measurements, derivation of age-specific dietary requirements may be in need of review. Finally, it is interesting to note that the study found the association between vitamin status and the urinary output of vitamin B1 to be aberrant( Reference Horwitt, Liebert and Kreisler 83 ).

Recommendations made by other major industrialised countries, including Germany, Switzerland, Australia, New Zealand and the USA, are between 10 and 25 % higher than the UK guidelines( 46 , 84 , 85 ). Part of this difference may be accounted for by use of different paradigms, whereby guidelines are developed in relation to points of saturation rather than minimum requirements for disease prevention. There is some epidemiological evidence to indicate that an intake of vitamin B1 at a level of about 0·9 mg/d appears to be sufficient to sustain a good level of health in elderly Italian women( Reference Bolzetta, Veronese and De Rui 86 ), an observation-based indication of vitamin B1 sufficiency at a level 12·5 % higher than the UK RNI.

With respect to the use of the TKA assay as a reflection of vitamin B1 sufficiency it should be noted that in the elderly: (a) age-related reductions in TKA activity are known to occur (probably due to depressions in apoenzyme levels and potentially confounding the interpretation( Reference O’Rourke, Bunkner and Thomas 87 ); (b) prolonged vitamin B1 deficiency also induces a lowering of basal and stimulated transketolase activities, possibly through the same mechanism( Reference Schrijver 88 ). Therefore the absence of baseline TKA data may be misleading with regard to sufficiency. It is of note that syndromes of polyneuritis, as commonly observed in the sarcopenic elderly( Reference Kjosen and Seim 89 ), are also associated with reduced TKA, potentially masking deficiency states.

Niacin (vitamin B3)

Function and effects of deficiency

Important food sources of vitamin B3 include: yeasts, teas and coffees, whole-grain cereals, dark-green leafy vegetables, poultry and meats, fish (especially varieties which have ‘red’ meat, for example, tuna), nuts and legumes and organ meat (in particular, liver)( Reference Scheid, Bennett and Schweigert 34 , Reference Jacob and Swenseid 90 ).

Niacin (nicotinic acid), and its derivative nicotinamide, are principally understood as components of the coenzymes NAD+ and NADP+, which have related functions. NAD+ and NADP+, which may be reversibly reduced to NADH and NADPH, are known for their role in energy metabolism: the transfer of hydride ions (H) within dehydrogenase–reductase systems. These two co-enzymes mediate, and have impact on, a wide range of processes within the body. These include, but are not limited to, Ca homeostasis( Reference Guse 91 ), gene expression( Reference Imai, Armstrong and Kaeberlein 92 ), mitochondrial function( Reference Zoratti and Szabò 93 , Reference La Piana, Marzulli and Gorgoglione 94 ), anti-oxidation( Reference Jaeschke, Kleinwaechter and Wendel 95 ) and immune function( Reference Aswad, Kawamura and Dennert 96 ).

Potential relationship with sarcopenia

Chronic vitamin B3 deficiency (niacin deficiency; ND) is known to result in pellagra( Reference Hegyi, Schwartz and Hegyi 97 ). Although the neurodegenerative symptomology in pellagra is well established, the neuropathological component of pellagra (per se) has had relatively little attention. Neuropathological observations from anatomical studies in human subjects in the 1930s include the chromatolysis of motor neurons, beginning at the level of the CNS (for example, Betz cells in motor cortex)( Reference Langworthy 98 , Reference Zimmerman 99 ), with clear implications for neuromuscular function. With regard to disease progression, human depletion studies have indicated that the clinical symptoms of pellagra can manifest in subjects on a diet low in vitamin B3 (about 4·3–5·7 mg/d) and tryptophan (178–230 mg/d) in 4–6 weeks, and that it is possible to reach a critical state within a further 4–8 weeks( Reference Goldsmith, Sarett and Register 100 , Reference Goldsmith, Gibbens and Unglaub 101 ).

Symptoms associated with vitamin B3 deficiency include neuromuscular deficits such as muscle weakness and wasting, gait and truncal ataxia, peripheral neuritis, limb areflexia and myoclonus( Reference Sakai, Nakajima and Fukuhara 102 Reference Brown 104 ). Although non-specific, our understanding of the early/less acute clinical manifestations of ND largely derives from behavioural changes associated with a diminished metabolism and nervous system dysfunction, for example, anorexia, weakness, inactivity, a decline in nerve transmission velocities, fatigue, anxiety, irritability and depression( Reference Buzina 105 , Reference Ronthal and Adler 106 ). It is interesting to note a degree of similarity between the neurological and neuromuscular deficits observed in ND states and the frailty of sarcopenia. In this connection, it is important to be aware that sub-acute ND is poorly characterised in the literature, seemingly due to the variable and non-specific nature of associated symptoms.

Potential mechanisms for neuromuscular damage can be discerned from experimental depletion of cellular NAD+ that induces oxidative damage and mitochondrial instability. Such models may be of particular relevance since they mirror key processes underlying cellular senescence and which are associated with the pathogenesis of sarcopenia( Reference Sakellariou, Pearson and Lightfoot 107 ).

NAD+ is a cofactor for poly (ADP-ribose) polymerases (PARP), which carry out DNA base excision repair processes and/or mediate cell death in response to oxidative damage, ischaemia and excitotoxicity( Reference Fomby and Cherlin 108 , Reference Kim, Mauro and Gévry 109 ). When DNA damage is increased following ROS insult, cytosolic NAD+ is rapidly depleted. Such events have been demonstrated to initiate PARP-mediated apoptosis in myocytes in vitro, as well as in neurons, if NAD+ status is not restored within hours( Reference Pillai, Isbatan and Imai 110 , Reference Alano, Garnier and Ying 111 ). In terms of the precise concentration–effect relationship, it is noteworthy that PARP complex formation appears to cease with only a 50 % reduction in cellular NAD+ ( Reference Jacobson, Nunbhakdi-Craig and Smith 112 ). Importantly, the administration of ‘supraphysiological’ doses of vitamin B3 (≥500 mg/kg), to allow for a surplus of NAD+/NADP+, has been demonstrated to prevent these specific events and the subsequent loss of neuronal tissue during experimentally induced ROS insult in vivo ( Reference Klaidman, Mukherjee and Adams 113 ). Following on from this line of enquiry, and since ROS production is known to increase with age along with mitochondrial dysfunction, investigations aimed at re-examining requirements according to such parameters as the maintenance of cellular NAD+ seem warranted.

A number of additional mechanisms by which low levels of NAD+ may contribute to cell death have been identified and it appears that the apoptotis-inducing events following PARP-induced NAD+ depletion can manifest independent of PARP activation, when NAD+ levels are depleted( Reference Alano, Garnier and Ying 111 ). With regard to the latter point, mitochondria are known to require relatively high concentrations of NAD+ to drive ATP production, and to re-coup NAD+ from reduced NADH formed during glycolysis (in cytosol). Subsequently, low levels of NAD+ have been shown to result in a ‘cellular energy crisis’ caused by mitochondrial dysfunction. This dysfunction has been further shown to result in mitochondrial depolarisation and the release of mitochondrial apoptosis-inducing factor, as in PARP-1-mediated cell death following NAD+ depletion( Reference Alano, Garnier and Ying 111 ). Though these effects have been observed in neurons, which are highly glycolytic, they may also apply to muscle tissue, which periodically enter phases of high glycolytic production of ATP.

In addition, Gomes et al. ( Reference Gomes, Price and Ling 114 ) demonstrated in animal models that the age-related decline in nuclear NAD+ induced pseudohypoxia by disrupting mitochondrial homeostasis through a shift in PGC-1α/β-independent pathways associated with nuclear–mitochondrial communication (and secondary to hypoxia-inducible factor 1-α (HIF-1α) accumulation). This communication change alters mitochondrial function through loss of mitochondrial DNA coding for specific subunits required for oxidative phosphorylation. Since mitochondrial dysfunction is now well established as one of the hallmarks of sarcopenia and ageing in general, the maintenance of mitochondrial function should represent a fundamental consideration in the establishment of nutritional requirements.

Gomes et al. ( Reference Gomes, Price and Ling 114 ) demonstrated that normal mitochondrial function can be restored in ageing mice, relative to young mice, simply by increasing cellular concentrations of NAD+ via normal dietary means. This effect appears to be sirtuin (SIRT) 1 mediated. Since the SIRT family of enzymes are also NAD+ dependent, low levels of NAD+ have been shown to have a negative influence on their activities( Reference Liu, Gharavi and Pitta 115 , Reference Narayan, Lee and Borenstein 116 ). The better-characterised of these, SIRT1 and SIRT2, have been implicated in cell survival (and human longevity) due to their role in the regulation of programmed cell death( Reference Hall, Dominy and Lee 117 ). Importantly, recent experiments in mice have indicated that when NAD+ levels within the cell are limited, the stimulation of SIRT1 in neuronal tissue can hasten action in apoptotic pathways. However, when NAD+ levels are high, or with increasing concentrations of nicotinamide (a SIRT1 inhibitor), neuronal tissue has been observed to be protected from apoptotis-inducing events( Reference Liu, Gharavi and Pitta 115 ). Similarly, there is now good evidence to support the importance of maintaining cellular nicotinamide and NAD+ concentrations in order to prevent SIRT2-mediated apoptosis( Reference Skoge, Dölle and Ziegler 118 ).

There is evidence that age is an important determninant of cellular NAD+ levels. A recent study on Wistar rats demonstrated that, starting in adulthood, ageing is associated with a significant decrease in intracellular NAD+ levels (P≤0·01), and a decrease in the NAD+:NADH ratio (P≤0·01)( Reference Braidy, Guillemin and Mansour 119 ). These changes have been shown to be consistent with increasing ROS production and DNA damage, resulting in decreased SIRT1 (P≤0·01) and increased PARP (P≤0·01) activity, as well as a decrease in ATP production (P≤0·01), indicating impaired mitochondrial function. This finding appears to translate to humans. Massudi et al. ( Reference Massudi, Grant and Braidy 120 ) have shown a significant age-related decline in cellular NAD+ levels in both men (P=0·001; r –0·706) and women (P=0·01; r –0·537). Interestingly, Massudi et al. ( Reference Massudi, Grant and Braidy 120 ) found that the change in cellular NAD+ concentration over time was consistent with the increased activation of PARP in men (P≤0·0003; r –0·639) and a decline in SIRT1 activity (P≤0·007), but not in women( Reference Massudi, Grant and Braidy 120 ). These results demonstrate an increasing requirement for NAD+ with age, and whilst the mechanisms for NAD+ depletion may appear to differ between the sexes, this only strengthens the need to review vitamin B3 requirements in the elderly in particular.

Prevalence of deficiency

There is a paucity of recent biochemical data on vitamin B3 status in the elderly (UK) population, and from elsewhere in Europe and North America. This may be attributable to the relative difficulty in assessment, with the urinary output of associated metabolites representing the only universally accepted method. Subsequently, assessments of vitamin B3 status are widely based on intake.

The 2014 update to the UK NDNS suggests a mean intake of vitamin B3 above the RNI for all age groups( 27 ). Recent large-scale European and North American studies have recently suggested that ND is not widespread in free-living elderly populations( Reference Deierlein, Morland and Scanlin 121 , Reference Troesch, Hoeft and McBurney 122 ). Interpretation of such is difficult as larger-scale intake studies suggesting adequacy are often presented in terms of mean intake for an entire cohort. However, intake studies specifically focused on the elderly have indicated that dietary intake of vitamin B3 in free-living elderly populations decreases significantly, typically between the ages of 50 and 90+ years( Reference Wakimoto and Block 71 , Reference Sahyoun 123 , Reference Woo, Ho and Mak 124 ). Intake data from institutionalised elderly populations in the Western world indicate a broad range of inadequate intakes, between 0 and 26·7 %( Reference Aghdassi, McArthur and Liu 125 Reference Engelheart and Akner 127 ). It is important to note, however, that intake data frequently do not correlate with biochemical determinants of status( Reference Dhonukshe-Rutten, de Vries and de Bree 128 ). Numerous factors, including genetic variability( Reference Shelnutt, Kauwell and Chapman 129 ), have been indicated in this regard. In addition, it should be noted that the primary source of vitamin B3 for most populations is mature staple cereal grains, in particular wheat and corn, which contain high concentrations of glycosides. These bond with vitamin B3 in a manner which significantly reduces its bioavailability( Reference Gregory 130 ).

UK dietary recommendations

The UK RDA (RNI) for vitamin B3 is 16 and 12 mg/d, for men and women aged 50+ years, respectively( 82 ). This determination is based on data from an early human depletion study, indicating mean vitamin B3 intake requirements (a) to alleviate the clinical signs of pellagra, and (b) to ‘normalise’ urinary output of N-methyl-nicotinamide and its downstream metabolite methyl-pyridone carboxamide, whilst in energy balance( 82 ). This recommendation, apparently derived from the one study referenced( Reference Hokwitt, Harvey and Rothwell 131 ), is 5·5 mg/4184 kJ (1000 kcal). The UK RNI is inclusive of a 10 % CV and yields an RNI of 16 and 12 mg/d for men and women, respectively (assuming set levels of energy expenditure in individuals in energy balance). Also included in this RNI is the contribution of niacin derived from the endogenous conversion of tryptophan to niacin in the ratio 60:1, giving a stated requirement for niacin or nicotinic acid equivalents. Whilst it is suggested that tryptophan to niacin conversion alone may satisfy vitamin B3 requirements, based on assessments of dietary protein intakes( 82 ), the conversion ratio is dependent on the intake, and relative availability of, multiple nutrient factors, including the availability of all other relevant cofactors (for example, pyridoxine)( Reference Nakagawa, Takahashi and Sasaki 132 ). The study of Hokwitt et al. ( Reference Hokwitt, Harvey and Rothwell 131 ) included only three elderly subjects who were reported as sedentary. Taken together, this gives limited scope to interpret requirements in the elderly and further research appears warranted.

Pyridoxine (vitamin B6)

Function and effects of deficiency

Vitamin B6 is naturally abundant in meat and poultry, including lean muscle meat, and can also be found in nuts and legumes and in fortified foods( 133 ). Vitamin B6 refers to a class of N-containing compounds, which comprise pyridoxine, pyridoxamine, pyridoxal and their phosphate derivatives. In physiological terms, vitamin B6 is principally understood to function in amino acid metabolism( Reference Bender 134 ) as the cofactor pyridoxal 5′-phosphate (PLP) and is associated with a wide range of physiological processes, most of which are biosynthetic, being (for example) involved in the synthesis of haeme( Reference Vogler and Mingioli 135 ), several neurotransmitters, including serotonin( Reference Hartvig, Lindner and Bjurling 136 ), purines( Reference Lamers, Williamson and Ralat 137 ), several hormones, including steroid hormones( Reference Allgood and Cidlowski 138 ) and fatty acids( Reference Mueller and Iacono 139 ). Clinical vitamin B6 deficiency (pyridoxine deficiency; PD) may therefore manifest as a diverse symptom-set, affecting multiple systems.

Potential relationship with sarcopenia

Clinical indicators of marked PD in adults are largely neurological and epithelial/mucosal aberrations, which mimic deficiency of other vitamins within the B complex. The majority of neurological symptoms (particularly in cases of mild PD, which is more commonplace) appear to affect the peripheral nervous system (PNS) and are associated with loss of motor function( Reference Zaoui, Abdelghani and Ben Salem 140 ). These include numbness/paresthesia in the extremities which may progress to a loss of distal sensation, motor ataxia, weakness and loss of deep tendon reflexes( Reference Vilter, Mueller and Glazer 141 Reference Devadatta, Gangadharam and Andrews 143 ). Non-specific symptoms such as nausea, gastrointestinal disturbances, vomiting and mood/behavioural changes (for example, anorexia, apathy, depression and fatigue) have also been described.

Work in this area has been carried out with rats, with one seminal investigation demonstrating axonal degeneration akin to Wallerian degeneration, as primary in the development of peripheral neuropathy characterised by ultrastructural changes, including swelling of local (axonal) mitochondria, the disruption of axoplasmic ground substance, and the accumulation of dense bodies within neurons( Reference Schalaepfer and Hager 144 ). Pathological disturbances have also been noted in and around Schwann cells and within the nucleus( Reference Jacobs, Miller and Whittle 145 ). Motor neurons, and in particular those which feed more distal muscles, appear most affected by this process, which leads to an increasing denervation of muscle fibres( Reference Cavanagh 146 ). These findings are consistent with case studies of isoniazid (isonicotinic acid hydrazide, a vitamin B6 antagonist) toxicity in humans, in which motor symptoms are reportedly predominant( Reference Zaoui, Abdelghani and Ben Salem 140 ). Interestingly, however, the morphological changes discussed earlier have been shown to be present in cases without any clinical signs of neuromuscular dysfunction. Therefore, studies in which isoniazid has been administered to animals with the aim of developing an understanding of early changes in neuropathies may provide valuable insight. Noting from previous studies that functional disturbances take 7–8 months to manifest( Reference Zbinden and Studer 147 ), Hildebrand et al. ( Reference Hildebrand, Joffroy and Coërs 148 ) administered isoniazid to rats for 3–13 d and found a significant loss of neural tissue, a reduction in conduction velocity and wide-spread muscle atrophy (by day 3), followed by extensive myelin disruption, fragmentation of intramuscular nerve fibres and a reduction in muscle action potential (by day 10). Whilst reactive collateral branching of motor fibres appeared to be sufficient to normalise most of the assessment parameters 37 d after ceasing the intervention, a significant proportion of the damage to peripheral nerve fibres was still apparent and the pattern of atrophy in large-group muscle fibres showed no improvement( Reference Hildebrand, Joffroy and Coërs 148 ). At present a mechanistic model for these observations has not been established, and whilst it is difficult to exclude the potential contribution of direct toxicity from isoniazid on nerve axons, it seems probable that vitamin B6 deficiency is indeed implicated. With regard to muscle atrophy, studies elsewhere have observed reduction in skeletal muscle protein synthesis in rats with a marginal vitamin B6 intake( Reference Sampson, Young and Kretsch 149 ).

The effects of a chronic, suboptimal vitamin B6 status in the elderly are poorly understood. Epidemiological associations between low vitamin B6 intake and frailty/disability are, however, widely reported – particularly in women. Amongst these, a prospective (3 years) study of 643 free-living elderly (≥65 years) women, into the possible relationship between specific micronutrient status (biochemical) and risk of developing disability, found that those with a plasma PLP level of >4·4 ng/ml (17·8 nmol/l) had a significantly greater risk of developing disability (hazard ratio (HR) 1·31; 95 % CI 1·03, 1·67; P=0·02) than those with PLP levels exceeding this threshold( Reference Bartali, Semba and Frongillo 150 ). In direct relation to sarcopenia, the Maastricht Sarcopenia Study, which comprised 227 free-living elderly (>60 years) individuals indicated that intakes of vitamin B6 are reduced by 10–18 % in individuals with sarcopenia, relative to non-sarcopenic controls( Reference Ter Borg, de Groot and Mijnarends 151 ).

In this regard, it is interesting to note that inadequate vitamin B6 intake has been linked with an increased risk of osteoporotic fractures and related falls. Since osteoporotic fractures are associated with concurrent sarcopenia and since the pathogenic mechanisms underlying both sarcopenia and osteoporosis may be inter-related( Reference Cederholm, Cruz-Jentoft and Maggi 152 ), this association should be considered in view of a more holistic approach. With this in mind, it is important to note the likelihood of sex-based differences here, since the risk/progression of osteoporosis is more marked in older women. This is attributed to the relative deficit in bone mineral homeostasis incurred as a result of the decline in the oestrogen axis post-menopause( Reference Richelson, Wahner and Melton 153 ). To illustrate, a large-scale (n 63 257) prospective investigation (follow-up time about 13·4 years) into the association between dietary B vitamin intake and the risk of hip fracture in men and women aged 45–74 years found a statistically significant negative correlation between the incidence of hip fractures and energy-adjusted vitamin B6 intake in women (P=0·002)( Reference Dai, Wang, Ang and Yuan 154 ). Women whose vitamin B6 intake represented the lowest quartile (mean pyridoxine ≤1·30 mg/d) had a 22 % increased risk of hip fracture, relative to those in the highest quartile (mean pyridoxine ≥2·03 mg/d) (HR=0·78; 95 % CI 0·66, 0·93). This is consistent with other similar investigations, including the Rotterdam Study, which found women in the highest quartile to have a significantly reduced risk of fragility fractures (HR=0·55; 95 % CI 0·40, 0·77; P=0·004) and non-vertebral fractures (HR=0·77; 95 % CI 0·65, 0·92; P=0·005)( Reference Yazdanpanah, Zillikens and Rivadeneira 155 ). Importantly, the same study identified a significant risk reduction following supplementation with vitamin B6, which took place independent to any change in BMD. Whilst the specific mechanism(s) underlying this effect are at unclear at present, it is known that vitamin B6 is required for the maintenance of the collagen matrix supporting bones( Reference Bird and Levene 156 ). Subsequently, an increased rate of collagen synthesis has been suggested as one potential mechanism for this effect( Reference Yazdanpanah, Zillikens and Rivadeneira 155 ).

Elsewhere, studies which have observed a correlation between hyperhomocysteinaemia and a decline in BMD, as well as deleterious architectural changes, have put forward the B vitamin deficiency (vitamins B6, B9 and B12)-induced build up of this metabolite as another possible mechanism, in addition to the direct role these B vitamins play in bone metabolism( Reference Herrmann, Widmann and Herrmann 157 , Reference van Wijngaarden, Doets and Szczecińska 158 ). A recent review of this area suggests that while these and other B vitamins (for example, vitamin B1) are clearly involved in bone homeostasis, clinical trials are needed to determine the efficacy of supplementation protocols( Reference Fratoni and Brandi 159 ).

Whilst such data help inform the relationship between poor vitamin B6 nutrient status and frailty, evidence for optimal status/intake remains incomplete.

Prevalence of deficiency

The UK NDNS suggests a mean intake of vitamin B6 above the RNI for all age groups( 27 ). However, the UK RNI is lower than those established by other authorities such as EFSA( 160 ). Data from other industrialised nations suggest considerable variation in intakes. Whilst some large-scale investigations report adequate intake of vitamin B6 amongst free-living elderly populations, these often contradict smaller-scale studies, which typically employ more time-intensive methods and suggest a high prevalence of inadequate intake( Reference De Groot, Van Den Broek and Van Staveren 21 , Reference Dewolfe and Millan 161 , Reference Szponar and Rychlik 162 ). Outside the UK, PD was suggested in 1990 to have reached epidemic proportions in the total US population (affecting 71 and 90 % of men and women, respectively) and this was significantly associated with ageing (P<0·001)( Reference Kant and Block 163 ). In fact, a significant age-related decline in intake is almost always observed where an extended age range is considered( Reference Bates, Lennox and Prentice 31 , Reference Sette, Le Donne and Piccinelli 72 , Reference Bechthold, Albrecht and Leschik-Bonnet 164 Reference Power, Jeffery and Ross 166 ). Inadequate intake is more consistently reported in non-free-living elderly populations( Reference Lengyel, Whiting and Zello 66 , Reference Paker-Eichelkraut, Bai-Habelski and Overzier 167 , Reference Buell, Arsenault and Scott 168 ). This includes an extensive, cross-sectional assessment of vitamin B6 intake across eleven long-term residential facilities in Canada, which reported inadequate intake in more than 50 % of the total participant group( Reference Aghdassi, McArthur and Liu 125 ).

As with other B vitamins, data on intake are typically revealed to provide a more optimistic picture than biochemical data( Reference Morris, Picciano and Jacques 169 ). Amongst these, Kjeldby et al. ( Reference Kjeldby, Fosnes and Ligaarden 170 ) were able to identify a biochemical deficiency (using PLP values) in up to 49 % of an elderly population with an ‘adequate’ (mean) intake. Past NDNS (1994–1995) had indicated PD (plasma PLP<30 nmol/l) to be endemic in the UK elderly population, being 49 % in community-dwelling and 75 % in the institutionalised elderly (>65 years)( Reference Bates, Pentieva and Prentice 171 ). The 2014 NDNS report – the first since 1999 to include biochemical data on the elderly – suggests no indication of deficiency despite using the same measures( 27 ). Only the mean value for the entire cohort is provided, however. Elsewhere, the range of reported prevalence of PD according to biochemical indices is typically between 26–59 % in free-living( Reference Fabian, Bogner and Kickinger 29 , Reference Bailey, Maisey and Southon 32 , Reference Wolters, Hermann and Hahn 81 , Reference Huang, Yan and Wong 172 ) and 5·3–49 %( Reference Paulionis, Kane and Meckling 126 , Reference Kjeldby, Fosnes and Ligaarden 170 ) in non-free-living elderly populations in industrialised nations.

Assessment according to functional biomarkers, such as erythrocyte aspartate aminotransferase activity (EAAA), and levels of circulating metabolites, such as cystathionine and taurine, which are known to build up in the deficient state, appear to indicate an even greater prevalence than PLP measures( Reference Lamers, Williamson and Ralat 137 , Reference Guilland, Bereksi-Reguig and Lequeu 173 ). Review data suggest that a combination of functional biomarkers are required for assessments in the elderly, due to confounders such as changes in renal function, increased inflammation, serum albumin and inorganic phosphate( Reference Ueland, Ulvik and Rios-Avila 174 ).

UK dietary recommendations

The RDA (RNI) for vitamin B6 intake in the UK is set at 1·4 and 1·2 mg/d for men and women aged ≥50 years, respectively( 82 ). This determination is based on a small number of studies focused on functional parameters related to protein metabolism: namely, changes in tryptophan and methionine metabolism in response to variation in protein intake, and during depletion( 82 ). The RDA (RNI) is based on an assessment of the quantity of vitamin B6 required to metabolise recommended (or slightly greater) levels of protein intake (about 50–150 g protein/d) whilst maintaining protein homeostasis, according to select outcome measures (for example, urinary output of xanthurenic acid). The RDA is therefore not in fact absolute, and is delineated as 15 µg/g protein( 82 ). Even if these parameters are sufficient in specifying intake requirements (according to protein intake), there may be issues with respect to their wider application, since determinations are based on small sample groups (n 6–13) comprised of young men (≤31 years)( Reference Canham, Baker and Harding 175 Reference Kelsay, Miller and Linkswiler 178 ).

Whilst it is true that elderly populations are widely reported to have a reduced intake of protein, an increased requirement for protein in elderly individuals (1–1·2 g/kg per d) is now being recognised( Reference Campbell, Crim and Dallal 179 , Reference Phillips, Chevalier and Leidy 180 ). Subsequently, there is a growing impetus to increase protein intake in this population. On this basis alone, the existing guidelines for vitamin B6 (although not absolute) may be outdated. The extent of this is perhaps best elucidated by the fact that the 25–50 % increase in protein requirements not reflected in the RNI for vitamin B6 represents more than the difference between the current estimated average requirement and RNI, implying that >50 % of elderly individuals would not be able to match vitamin B6 requirements with protein intake. It should also be noted that recent investigations do not in fact support any association between protein intake and vitamin B6 status in both elderly men and women( Reference Huang, Yan and Wong 172 ).

Data from more recent depletion/repletion studies in the healthy elderly indicate an extended physiological range of function for vitamin B6 that is observed in response to increased levels of intake (15 µg/kg body weight per d – 50 mg/d). Evidence for this includes a significant decrease in urinary output of xanthurenic acid post-tryptophan loading (5 g) in elderly women and other direct outcomes in immune system function, including lymphocyte proliferation in response to peripheral mitogens( Reference Meydani, Ribaya-Mercado and Russell 181 ). Another study examined requirements in the elderly in relation to normalising multiple associated physiological outcomes, including EAAA and output of xanthurenic acid( Reference Ribaya-Mercado, Russell and Sahyoun 182 ). These authors took into account different levels of protein intake and suggested that the minimum requirement for vitamin B6 in the elderly is 1·96 and 1·90 mg/d, for men and women, respectively( Reference Ribaya-Mercado, Russell and Sahyoun 182 ). Several noteworthy studies indicate still greater requirements. Amongst these, Morris et al. ( Reference Morris, Picciano and Jacques 169 ) differentially examined (for the first time) the correlation between vitamin B6 intake and plasma PLP levels, using data from a US population-representative sample of over 6000 participants in the National Health and Nutrition Examination Survey (NHANES; 2003–2004). Their study also examined the possible relationship between plasma PLP and homocysteine (hcy). The findings indicated that the elderly have a markedly increased requirement for vitamin B6, an intake of 3–4·9 mg of vitamin B6/d was needed simply to avoid a low plasma PLP (<20 nmol/l) and hyperhomocysteinaemia( Reference Morris, Picciano and Jacques 169 ). More importantly perhaps, vitamin B6 requirements were not found to be reduced in elderly individuals with a low protein intake (54 g/d)( Reference Ribaya-Mercado, Russell and Sahyoun 182 ), which may suggest that the parallel with protein intake may have limited utility in the assessment of vitamin B6 requirements in the elderly.

Folate (vitamin B9)

Function and effects of deficiency

Important sources of vitamin B9 include green leafy vegetables, citrus fruits, nuts and legumes, and fortified foods( 133 ). Folate content in lean muscle meat is typically low, but high in organ meats, particularly the kidneys and in the liver( Reference Williams 183 ). Folate (vitamin B9) describes collectively both natural folates and the synthetic folic acid/pteroylmonoglutamic acid (PteGlu), which are functional within the body as the cofactor tetrahydrofolate (THF). THF has important roles in one-carbon metabolism: as a methyl group donor. The first step in the methylation cycle, in which THF is transformed into 5,10-methylenetetrahydrofolate (5,10-MTHF), is catalysed by the PLP-dependent enzyme, serine hydroxymethyltransferase( Reference Paiardini, Contestabile and Buckle 184 ). This highlights the functional interdependence of B vitamins. Viamin B12 is of particular interest in this regard, since there is considerable functional overlap with vitamin B9 – such that one being in sufficient supply may mask a functional deficiency of the other, for example, prevent macrocytosis( Reference Wyckoff and Ganji 185 ). This can have implications for elderly populations which are not routinely screened, since the neurological effects of vitamin B12 deficiency are irreversible at an advanced stage when clinical symptoms are manifest. It is also well established that vitamin B12 deficiency leads to a functional vitamin B9 deficiency (folate deficiency; FD)( Reference Hoffbrand and Jackson 186 ). Whilst the precise mechanism is as yet unclear, the subsequent reduction in the activity of (the vitamin B12-dependent enzyme) methionine synthase is known to result in a build up of 5-methyltetrahydrofolate (5-MTHF)( Reference Banerjee and Matthews 187 ), and therefore represents a popular hypothesis( Reference Nijhout, Reed and Budu 188 ). In all, the assessment of vitamin B9 status should include where possible an estimation of vitamin B12 status also.

Potential relationship with sarcopenia

Whilst anaemia is the traditional association, FD may compromise the function of any system dependent on one-carbon metabolism, or affected by concentrations of associated metabolites. Subsequently, vitamin B9 deficiency may contribute toward a range of pathologies, particularly if prolonged and in the presence of aggravating factors such as inadequate levels of vitamin B12. Determination of the extended role of vitamin B9 in human health may be difficult in clinical terms since the effects of marginal deficiency may be subtle or so indirect as not to be obviously tied to vitamin B9 status.

One of the established pathophysiological outcomes of vitamin B9 deficiency, with possible pertinence to sarcopenia, is the build up of hcy: hyperhomocysteinaemia. Hyperhomocysteinaemia, which is prevalent in the UK elderly population( Reference Bates, Mansoor and Pentieva 189 ), has been shown both epidemiologically and experimentally to be tied more closely to FD than to other methyl group donors such as vitamin B12 ( Reference Carmel 190 , Reference Pietrzik and Bronstrup 191 ).

With regard to possible long-term effects, large-scale longitudinal investigations (>15 years) have identified plasma hcy as an independent risk factor for future incidence of major, age-related, neurodegenerative disease( Reference Zylberstein, Lissner and Björkelund 192 , Reference Seshadri, Beiser and Selhub 193 ). Similarly, studies into the long-term relationship between hcy and physical function have identified significant associations between modest elevations in plasma hcy and a decline in important aspects of neuromuscular function in elderly individuals( Reference Kado, Bucur and Selhub 194 , Reference Ng, Aung and Feng 195 ). Whilst effects on skeletal muscle are relatively unexplored, using both cross-sectional and prospective data obtained from 1301 elderly (aged >65 years) participants in the Longitudinal Ageing Study in Amsterdam (LASA), Swart et al. ( Reference Swart, Van Schoor and Heymans 196 ) have demonstrated an association between elevated hcy and reduced grip strength in men, and increased functional limitations in both men and women. Elevated (total) hcy (≥15 µmol/l) and vitamin B9 (but not vitamin B12) deficiency (<3 ng/ml) have also been associated with markedly increased risk of brain atrophy( Reference Yang, Wong and Wu 197 ).

Whilst the authors of these studies suggest hcy to be a causal factor and not a marker in neuromuscular decline, there is a dearth of mechanistic enquiry into this topic, particularly in relation to neuromuscular/musculoskeletal decline. Several plausible mechanistic links have, however, been demonstrated, for example, hcy induction of neurotoxicity in animals. It is notable that neural tissue appears to be more sensitive to elevations in hcy than endothelial smooth muscle cells( Reference Stanojlovic, Hrncic and Rasic-Markovic 198 ), on which most research in this field has been focused. Relevant mechanisms in the CNS include: the inhibition of acetyl-cholinesterase activity (involved in neurotransmitter breakdown) in rats, N-methyl-d-aspartate (NMDA; glutamate) receptor interaction-induced excitotoxicity in neurons, and region-specific inhibition of Na+/K+-ATPase activity in the brain (resulting in inability to maintain cell polarity)( Reference Stanojlovic, Hrncic and Rasic-Markovic 198 ). In the PNS, hcy–NMDA receptor interaction has additionally been linked (in mice) to increased sensitivity to oxidative stress-induced inhibition of neurotransmitter release and associated loss of cell integrity at the level of the neuromuscular junction( Reference Bukharaeva, Shakirzyanova and Khuzakhmetova 199 ), as in motorneurone disease. Motorneurone disease is loss of motor neurons, so naturally there is degeneration of the neuromuscular junction.

Other mechanisms include the progressive depletion of NAD+ in rats (secondary to DNA strand break-induced increases in PARP activity), subsequent mitochondrial dysfunction, and the activation of caspase-induced apoptosis( Reference Kruman, Culmsee and Chan 200 ). This is in accord with findings elsewhere (in rats), which demonstrate that the selective inhibition of PARP not only prevents hcy-induced damage, but improves the condition of other tissue types( Reference Tasatargil, Dalaklioglu and Sadan 201 ). Vitamin B9 supplementation has proven efficacy in this regard: in addition to reducing hcy-associated PARP activity and inhibiting caspase-induced apoptotic pathways, treatment with vitamin B9 but not vitamin B12 has been shown to increase expression of the anti-apoptotic B-cell lymphoma protein Bcl-2. In addition, enhanced vitamin B9 supply inhibits expression of TNF-α, induces NO synthase and supresses the activation of glial cells in the CNS( Reference Zhang, Chen and Li 202 ). These effects have been demonstrated in knock-out models in mice that mimic action of a common genetic variant in humans that results in amyotrophic lateral sclerosis, a neurodegenerative condition in which elevated hcy is associated with the loss of motor neurons.

Stable isotope studies have provided valuable insights into the metabolism and relative bioavailability of synthetic PteGlu, compared with naturally occurring folates. In humans, as well as in other mammals, the metabolic path to which a high proportion of PteGlu is committed begins with conversion to the naturally occurring THF( Reference Rogers, Pfeiffer and Bailey 203 , Reference Wright, Finglas and Dainty 204 ). In this connection, it is important to note that the enzyme dihydrofolate reductase, which is expressed in the liver, and which is required for the conversion (reduction and methylation) of PteGlu into THF, is relatively limited in its activity( Reference Whitehead, Kamen and Beaulieu 205 ). This has led some to suggest that PteGlu is therefore less biologically active than naturally occurring folates. Also, due to the low rate of conversion, PteGlu supplementation can result in high concentrations of this circulating metabolite and potentially distort the accuracy of the assessment of vitamin B9 status. Certainly the use of PteGlu as a ‘reference folate’ in studies of folate bioavailability has been questioned( Reference Wright, Finglas and Dainty 204 ).

Vitamin B9 has both a direct and an indirect role in DNA synthesis, methylation and repair processes, principally due to its function as a cofactor for specific enzymes known to be involved in the relevant pathways. Most notably, the (5, 10-MTHF) folate-dependent thymidylate synthases are known to catalyse the conversion of uracil into thymine( Reference Carreras and Santi 206 ). Correspondingly, low intracellular concentrations of 5,10-MTHF have been shown to result in a build up of uracil, an imbalance of the cellular deoxyuridine monophosphate:deoxythmidine monophosphate ratio, and the DNA polymerase-associated, mass misincorporation of uracil into cellular DNA( Reference Goulian, Bleile and Tseng 207 Reference Luzzatto, Falusi and Joju 209 ). Emerging evidence suggests that this frequently overlooked effect of vitamin B9 deficiency is implicated in increased DNA deletions, chromosomal instability and damage to factors maintaining nuclear integrity( Reference Wickramasinghe and Fida 208 , Reference Blount, Mack and Wehr 210 Reference Duthie 212 ). Although currently this phenomenon has primarily been studied in relation to carcinogenesis, tissue-wide chromosomal instability has the potential to have a negative impact multiple systems, including neuromuscular systems.

The tissue-specific effects of this phenomenon have not yet been comprehensively delineated and the possible impact on the nervous system and, in particular, the musculoskeletal/neuromuscular system has not been explored in human subjects. This may represent an oversight, since animal studies suggest potentially severe consequences. With regard to the nervous system, where more research is in evidence, the dysregulation of the aforementioned pathway has been associated with neurodegenerative and neuropsychiatric dysfunction( Reference Kronenberg, Harms and Sobol 213 ). Knock-out models in mice have indicated impaired uracil repair and impaired DNA glycosylase activity, secondary to vitamin B9 depletion that resulted in moderate deficiency in this vitamin. Overall in this situation, there is promotion of neurodegenerative processes resulting in neuronal death and an inhibition of neurogenesis, associated with a region-specific reduction in brain-derived neurotrophic factor and glutathione( Reference Kronenberg, Harms and Sobol 213 ), and nerve growth factor( Reference Eckart, Hörtnagl and Kronenberg 214 ). Duthie( Reference Duthie 212 ) has further highlighted the relative contribution of vitamin B9 deficiency, having observed that (moderate) FD, and not an inadequate status of other methyl-donors with overlapping functions (for example, choline and methionine), leads to progressive strand-breaking, DNA damage in lymphocytes (100 % after 8 weeks). Finally, vitamin B9 deficiency has been shown to result in an increased frequency of large-scale deletions in mitochondrial DNA in older rats (1 year), compared with young weanling controls( Reference Crott, Choi and Branda 215 ). Interestingly, whilst this effect appears to be modulated by age, the association is lost following vitamin B9 repletion; uracil levels in DNA are normalised upon restoring optimal vitamin B9 status, and this is consistent with findings in other animal studies.

Animal studies into the musculoskeletal effects of the mass misincorporation of uracil into DNA are in shortfall. In addition to non-tissue-specific risks to myocytes, chromosomal instability would probably interfere with the formation of new myonuclei, which are required in the repair of skeletal muscle cells( Reference Hikida, Staron and Hagerman 216 ) and to facilitate muscle hypertrophy in animals( Reference Allen, Monke and Talmadge 217 ) and humans( Reference Kadi and Thornell 218 , Reference Sinha-Hikim, Roth and Lee 219 ). The latter association may be interpreted within the context of the myonuclear domain theory, namely that additional mynucleii are required to facilitate hypertrophy( Reference Teixeira and Duarte 220 ). In addition to compromising muscle repair processes, for example, following injury/extended bed rest, such a phenomenon may also compromise the prophylactic/therapeutic benefit of exercise/movement as a stimulus for increased muscle mass/strength. Damage to mitochondrial DNA within muscle fibres/cells is also likely to represent a limitation for maintaining muscle strength and function.

Prevalence of deficiency

The UK NDNS suggests a mean intake of vitamin B9 close to or above the RNI for all age groups( 27 ). Data from other industrialised countries typically suggest a high prevalence of inadequate intake, with some reports ranging as high as 37–100 % in both free-living( Reference Marshall, Stumbo and Warren 23 , Reference Ortega, Mañas and Andrés 221 Reference Volkert, Kreuel and Heseker 224 ) and non-free-living elderly populations. Over time, there seems to be a reduction in reported prevalence of inadequate vitamin B9 intake in community-dwelling elderly populations, probably due to the emergence of functional foods and food-fortification initiatives, but also in part due to the popularisation of vitamin supplementation in the general population( Reference Morris, Evans and Bienias 225 ), particularly in Europe. Data from the European Nutrition and Health Report 2004 indicate vitamin B9 intake to be below acceptable levels across Europe( Reference Fabian and Elmadfa 60 ).

The reported prevalence of biochemically determined FD typically ranges from 0 to 23·5 %( Reference Ramos, Allen and Mungas 226 Reference Pfeiffer, Caudill and Gunter 229 ) in free-living elderly populations and from 5 to 68 % in non-free-living/sick elderly populations( Reference Aghdassi, McArthur and Liu 125 , Reference Paker-Eichelkraut, Bai-Habelski and Overzier 167 , Reference O’Leary, Flood and Petocz 230 , Reference Dimopoulos, Piperi and Salonicioti 231 ) in industrialised nations. It is again difficult to extrapolate from these findings due to the differences (unknown or otherwise) in sample populations, reference parameters and methods of assessment. In particular, the percentage of participants with polymorphisms affecting the methylene THF reductase (MTHFR) gene is almost always unknown. The 2015 supplementary report on the UK NDNS presents an interpretation of serum and erythrocyte folate levels according to thresholds used by the WHO (<10 nmol/l and <340 nmol/l, respectively)( 232 ). This assessment finds the prevalence of FD in the UK elderly population (>65 years) to be 8·5 % in men and 12·4 % in women, according to serum total folate, and 7·3 % in men and 10·8 % in women, when determined by erythrocyte folate levels( 232 ). Whilst these data do present evidence for/against potential age-related differences in vitamin B9 status for those aged over 65 years, similar investigations, which differentially assessed the over 65s, suggest that the prevalence of deficiency in this age group increases significantly with age( Reference Clarke, Grimley Evans and Schneede 233 ). It should be noted that supplement users were not excluded from the NDNS sample population, and as with other B vitamins, supplemental vitamin B9 intake is often found in sample populations with a low prevalence of FD( Reference Paulionis, Kane and Meckling 126 , Reference Koehler, Baumgartner and Garry 234 ) and this probably distorts perception of dietary adequacy across a given population.

Metabolic markers of vitamin B9 status (for example, hcy) are rarely employed in the assessment of prevalence, despite indications of greater sensitivity in terms of detecting deficiency and typically translating into marked increases in reported deficiency prevalence( Reference Lindenbaum, Rosenberg and Wilson 235 ). It should be noted that past national surveys, such as the 1994–1995 sample assessed by the UK Ministry of Agriculture, Fisheries and Food (MAFF), reported hyperhomocysteinaemia to be endemic in the British population( Reference Finch, Doyle and Lowe 236 ). The same report, which distinguished between free-living and institutional elderly populations, found serum vitamin B9 to be relatively lower in those living in residential care, despite intakes exceeding requirements( Reference Finch, Doyle and Lowe 236 ).

UK dietary recommendations

The RDA (RNI) for vitamin B9 in the UK is 200 µg/d for both men and women aged 11+ years( 82 ). As such, the UK RNI for vitamin B9 represents one of the lowest amongst EU member states( Reference Dhonukshe-Rutten, de Vries and de Bree 128 ). The UK RDA is based on several considerations. First, the lower reference range is based on the quantities of PteGlu shown simply to reverse the signs of acute FD (megaloblastic anaemia), following experimental depletion in (a) an undefined sample( Reference Herbert 237 ) and (b) in a sample of seven moribund cancer patients( Reference Gailani, Carey and Holland 238 ). Whilst it is not even possible to comment on the sample population for the first study, determining requirements through study of advanced-stage cancer patients is problematic as derangements in cell turnover are pathognomonic. Further work in understanding the lower threshold for vitamin B9 requirements seems merited.

The determination of the upper reference ranges (RNI) for vitamin B9 are based on findings of adequate liver vitamin B9 stores (>3 µg) following post-mortem analysis of 560 Canadian samples, which represent a heterogeneous sample( Reference Hoppner and Lampi 239 ). Since dietary intake is not included, the results are contextualised (by the Committee on Medical Aspects of Food and Nutrition Policy (COMA)) by allusion to an altogether separate investigation reporting a mean dietary vitamin B9 intake of 150–200 µg/d, and adequate erythrocyte vitamin B9 levels (>150 µg/ml), in about 90 % of a 1978 sample of the Canadian population( Reference Cooper 240 ). COMA attempts to establish the relevance of these data to the UK population through reference to ‘comparable’ median intakes in 1990( Reference Gregory, Foster and Tyler 241 ). This is a rather indirect estimate of requirements. Additionally, it was noted in the said report, and in a subsequent analysis of the same dataset, that FD, as it was then defined (erythrocyte vitamin B9 <170 µg/l (0·37 µmol/l)), was observed in 35 and 47 % of men and women, and so represented the most widespread deficiency of a nutrient known( 242 ).

Elderly individuals have been shown to be more sensitive to marginal FD and relatively less responsive to repletion than younger adults( Reference Kauwell, Lippert and Wilsky 243 ), particularly when more rapidly responding biochemical parameters such as changes in DNA methylation are used in assessment( Reference Rampersaud, Kauwell and Hutson 244 ). Other such parameters indicate even higher requirements still. Amongst these, hcy has been observed not to reach its nadir until an intake of 400 µg/d has been achieved( Reference Selhub, Jacques and Wilson 245 ).

Finally, and as COMA later acknowledges, intake is typically observed to decline with age and vitamin B9 status is further affected by age-associated medical conditions, and associated medications( 82 ). Common examples include: non-steroidal anti-inflammatory drugs (NSAIDs)( Reference Baggott, Morgan and Ha 246 ), hydrogen receptor antagonists, antacids, proton pump inhibitors( Reference Roe 247 ), insulin mimetics (biguanides, for example, metformin)( Reference Corominas-Faja, Quirantes-Piné and Oliveras-Ferraros 248 ), a wide-range of commonly prescribed antibiotics (for example, amoxicillin)( Reference Neu and Gootz 249 ), bile acid sequestrants (cholestyramine and colestipol)( Reference Leonard, Desager and Beckers 250 ) and K-sparing diuretics (for example, triamterenes)( Reference Corcino, Waxman and Herbert 251 ). Taken together, there appears to be a strong case for careful reappraisal of the area with the aim of developing population-specific guidelines.

Cyanocobalamin (vitamin B12)

Function and effects of deficiency

Since vitamin B12 is synthesised by bacteria living in the gastrointestinal tracts of animals( Reference LeBlanc, Milani and de Giori 252 ), sources include all animal produce, to a lesser extent eggs and dairy produce, and fortified foods( Reference Brody 253 ). Unlike other B vitamins, vitamin B12 is stored in relatively large quantities in the liver( Reference Williams 183 ).

Cyanocobalamin, or cobalamin (vitamin B12), forms the central component of the two corrinoid coenzymes, methylcobalamin and S-adenosyl-cobalamin (cobamamide), which are unusual in that they are thought to have limited functions within the body. Methylcobalamin is known to be required for the methylation of hcy to produce methionine by way of methionine synthase( Reference Goulding, Postigo and Matthews 254 ). S-adenosyl-cobalamin is required for the catabolic isomerisation of methylmalonyl-CoA into succinyl-coA by l-methylmalonyl-CoA mutase( Reference Cooper and Rosenblatt 255 ). Inadequate vitamin B12 intake therefore results in a build up of hcy and methylmalyonyl-CoA. Finally, low levels of vitamin B12 are also known to result in a build up of glycine, since vitamin B12 is required for the synthesis of porphyrin rings from glycine and succinyl-CoA( Reference Neuberger 256 ). The build up of these metabolites results in a shortfall in methionine and related upstream products such as S-adenosyl methionine, which carry out a broader range of functions. Therefore, despite being involved in a limited number of reactions, the effects of vitamin B12 deficiency are extensive, affecting multiple systems. The manifestations of deficiency may be categorised as gastrointestinal disturbances, neuropathies and megaloblastic anaemia. Pernicious anaemia, which typically affects the elderly, is aetiologically distinct from megaloblastic anaemia, in that it refers to instances in which vitamin B12 absorption has been compromised( Reference Toh, van Driel and Gleeson 257 ).

Potential relationship with sarcopenia

In terms of neuromuscular dysfunction, vitamin B12 deficiency (cobalamin deficiency; CD) is associated with sub-acute combined degeneration of the spinal cord and polyneuropathies. Histopathological findings from post-mortem examinations indicate that CD results in extensive demyelination in the CNS – most prominently in the spinal cord – although focal demyelination is also notably apparent in the white matter of the brain( Reference Silva, Cavalcanti and Moreira 258 Reference Pant, Asbury and Richardson 260 ). Demyelination has also been observed in the PNS, along with signs of significant damage to nerve fibres, most notable of which is axonal degeneration in distal afferent fibres of dorsal root ganglion neurons( Reference Greenfield and Carmichael 261 , Reference McCombe and McLeod 262 ). The mechanism by which demyelination occurs is at present unclear.

Clinically, vitamin B12 deficiency is associated with an extensive range of neuromuscular symptoms, which become more severe as deficiency progresses, including paraesthesia, numbness in the trunk, muscle weakness, abnormal reflexes, tendon jerks, spasticity, gait ataxia, myelopathies and myeloneuropathies( Reference Chand and Maller 263 Reference Hin, Clarke and Sherliker 266 ). Although CD is primarily associated with macrocytic anaemia, it is noteworthy that neuromuscular symptoms, such as gait ataxia, represent common clinical associations and mark the earlier stages of deficiency. These symptoms are primarily connected with the loss of sensory rather than motor-unit function( Reference Healton, Savage and Brust 264 , Reference Savage and Lindenbaum 267 ), specifically a decline in proprioceptive, vibratory, tactile and nociceptive sensation. CD has also been shown to have a significant negative impact on nerve conduction velocity( Reference Leishear, Boudreau and Studenski 268 ), implying a direct detrimental effect on peripheral motor function.

Given the extent of neuromuscular dysfunction observed in CD, it is unsurprising that recent studies have found significant associations between vitamin B12 status and frailty in old age. Amongst these, Matteini et al. ( Reference Matteini, Walston and Fallin 269 ) recently observed a 1·66–2·33 times greater risk of frailty (P≤0·02) in elderly Caucasian women with elevated methylmalonic acid (MMA). Whilst not all studies report such an association, this again appears to be explained largely by use of different parameters and cut-off points, as indicated in a recent study by Oberlin et al. ( Reference Oberlin, Tangney and Gustashaw 270 ). Oberlin et al. ( Reference Oberlin, Tangney and Gustashaw 270 ) carried out a retrospective examination of data on 3105 elderly individuals (≥60 years) from early NHANES (1999–2002), for which multiple measures of vitamin B12 status were employed. They found that CD was significantly associated with a much greater range of disabilities (for example, in social and leisure activity, in mobility, etc.) when based on functional biomarkers and/or higher cut-off points for serum cobalamin( Reference Oberlin, Tangney and Gustashaw 270 ). The inconsistency between cobalamin values and the functional outcomes of vitamin B12 status have been noted in seminal investigations into CD as discussed below( Reference Lindenbaum, Healton and Savage 271 , Reference Carmel 272 ).

Although investigations into the long-term effects of vitamin B12 supplementation in the elderly do not appear to have produced clinically significant outcomes that directly relate to sarcopenia (for example, time to/number of falls, muscle mass and strength), there has been indication of improvements in more general measures of physical function, such as walking speed (cumulative OR 1·3; 95 % CI 1·1, 1·5), which may indicate improved neuromuscular interactions( Reference Swart, Ham and van Wijngaarden 273 ). A realistic approach to future research may utilise vitamin B12 data in conjunction with the many other known factors and cofactors involved in neuromuscular structure and function – particularly amino acids associated with muscle anabolism, such as leucine.

Indirect negative outcomes for neuromuscular function are also apparent. Amongst these, vitamin B12 status (as well as that of vitamin B6) has been significantly associated with length of stay in rehabilitation following injury( Reference O’Leary, Flood and Petocz 230 ), which in turn was related to a decline in both muscle mass and function – muscle cachexia( Reference Paddon-Jones, Sheffield-Moore and Zhang 9 , Reference Kortebein, Ferrando and Lombeida 274 ). The relationship between mood disturbances (for example, depression)/neurocognitive decline related to CD( Reference Hin, Clarke and Sherliker 266 , Reference Bell, Edman and Miller 275 , Reference Tangney, Aggarwal and Li 276 ) and the development and progression of sarcopenia requires further investigation since these are increasingly being established as significant contributors( Reference Kim, Kim and Eun 277 , Reference Hsu, Liang and Chou 278 ).

Finally, it is interesting to note that a recent study by Verlaan et al. ( Reference Verlaan, Aspray and Bauer 279 ) found intakes of vitamin B12 to be 22 % lower and serum cobalamin to be 15 % lower in a sample of individuals with sarcopenia relative to non-sarcopenic controls.

Prevalence of deficiency

The UK NDNS suggests a mean intake of vitamin B12 above the RNI for all age groups( 27 ). This is consistent with other large-scale pan-European studies on free-living populations which report mean intake( Reference Olsen, Halkjaer and van Gils 59 ). However, as demonstrated in a recent systematic review and pooled analysis of observational cohort and prospective studies of intakes in the elderly (>65 years), even though mean reported intakes may be above the RNI, a large percentage of individuals (16–19 % in the case of vitamin B12) had an intake below the UK RNI/EU estimated average requirement( Reference ter Borg, Verlaan and Hemsworth 64 ). Inadequate intake is reportedly even greater in non-free-living elderly populations (>65 years) across Europe and other industrialised countries, particularly in the lower socio-economic strata and amongst migrants( Reference Fabian, Bogner and Kickinger 29 , Reference Paker-Eichelkraut, Bai-Habelski and Overzier 167 ). There is indication that a further 20–30 % would be deficient if not for supplementation( Reference Fabian, Bogner and Kickinger 29 , Reference Bor, Lydeking-Olsen and Møller 280 ). In any case, intake in the elderly is unlikely to translate to metabolic sufficiency in many cases due to poor absorption associated with the combined issues of hypochlorhydria( Reference King, Leibach and Toskes 281 ), gastritis and diminished intrinsic factor production( Reference Suter, Golner and Goldin 282 Reference Kong, Yi and Dai 285 ), as well as the widespread use of antacids( Reference Valuck and Ruscin 18 , Reference Lam, Schneider and Zhao 286 ).

As for other B vitamins, vitamin B12 status has been shown to decline significantly with advancing age( Reference Wolters, Hermann and Hahn 81 , Reference Clarke, Grimley Evans and Schneede 233 ). This finding is confirmed with respect to vitamin B12 in a study of an elderly population aged 85+ years in New Zealand( Reference Wham, Teh and Moyes 287 ).

By measure of serum cobalamin, the UK NDNS( 27 ) has indicated that 5·9 % of elderly men and women are deficient (<150 pmol/l) in vitamin B12. Although varied, estimates of the prevalence of CD from elsewhere in Europe tend to be much higher, ranging from 10 to 40 % for both community-dwelling and institutionalised elderly populations( Reference Hin, Clarke and Sherliker 266 , Reference van Oijen, Sipponen and Laheij 284 , Reference Bianchetti, Rozzini and Carabellese 288 Reference Gonzalez-Gross, Sola and Albers 292 ). These typically use higher cut-off points and other/additional, functional measures that have been shown to demonstrate greater sensitivity( Reference Gonzalez-Gross, Sola and Albers 292 ). To illustrate the extent of the potential discrepancy, 12 % of community-dwelling elderly in the Framingham Study were identified as vitamin B12 deficient according to serum cobalamin; however, follow-on investigations identified a further 50 % of the sample (having elevated serum MMA) to be deficient metabolically( Reference Lindenbaum, Rosenberg and Wilson 235 ). Similarly, Morris et al. ( Reference Morris, Jacques and Rosenberg 293 ) identified a further 15 % of the NHANES III sample population as being deficient when MMA was used as the criterion instead of serum cobalamin. Also holo-transcobalamin (holoTC) has been shown to demonstrate even greater sensitivity than serum cobalamin( Reference Heil, de Jonge and de Rotte 294 , Reference Obeid and Herrmann 295 ) and MMA( Reference Remacha, Sardà and Canals 296 ), particularly in older populations( Reference Valente, Scott and Ueland 297 ). Due to the lack of clear reference standards it is, however, unclear whether these differences are made up of false positives.

UK dietary recommendations

Vitamin B12 is distinct from other B vitamins in that, under normal conditions, there is a physiologically significant bodily store, with estimates ranging from 2 to 3·9 mg, most of which appears to be in the liver( Reference Reizenstein, Ek and Matthews 298 , Reference Grasbeck, Nyberg and Reizenstein 299 ). For this reason, and since the neurological effects of CD cannot be reversed, human depletion studies have never been carried out.

The RDA (RNI) for vitamin B12 in the UK is 1·5 µg/d for both men and women aged 15+ years( 82 ). Excluding Ireland, this value represents the lowest RNI in Europe( Reference Dhonukshe-Rutten, de Vries and de Bree 128 ), and stands in sharp contrast to the German Nutrition Society guideline, for which the RNI is set at 3 µg/d( 300 ), and the EFSA guideline of 4 µg/d( 301 ). EFSA, which has based its recommendation on mean intakes associated with normal ranges of circulating functional markers (MMA, total hcy, serum cobalamin and holoTC), in fact reports that it is intakes between 4·3 and 8·6 µg/d that are associated with these values( 301 ). This is consistent with a recent, comprehensive analysis of vitamin B12 status alongside intake in Danish postmenopausal women, which considered serum cobalamin, MMA, holoTC, transcobalamin (TC) saturation (holoTC/total TC) and total hcy, as well as potential absorptive complications( Reference Bor, Lydeking-Olsen and Møller 280 ). It was found that an intake of 6 µg/d was required to normalise all parameters in individuals with normal absorptive capacity( Reference Bor, Lydeking-Olsen and Møller 280 ). Follow-up investigations into requirements in adulthood (18–50 years) have likewise suggested an increased requirement of between 4 and 7 µg/d, even in healthy individuals( Reference Bor, von Castel-Roberts and Kauwell 302 ).

The UK recommendation is based on an assessment of the requirements for the prevention of megaloblastic anaemia as indicated by: (a) a small number of intake studies on narrowly defined groups of vegan/vegetarians in Sweden, Australia and South Asia; and (b) the haematological response to parenteral vitamin B12 administration in patients with pernicious anaemia( 82 ).

There appear to be several limitations to the appropriateness of these data in determining the RDA for elderly populations. First, whilst it is widely held that haematological abnormalities represent the first symptom of CD, it has long been understood that neurological deficits often present first( Reference Elsborg, Lung and Bastrup-Madsen 291 , Reference Gross, Weintraub and Neufeld 303 ). Moreover, experimental investigations from the 1980s and 1990s suggest that the absence of macrocytic anaemia is commonplace, relating to about one-third of cases, whilst the absence of neurological symptoms appears to be much less common( Reference Healton, Savage and Brust 264 , Reference Lindenbaum, Healton and Savage 271 , Reference Carmel 272 ). Interestingly, severe incidence for one set of symptoms (for example, neurological symptoms) has been associated with absence of the other (for example, haematological symptoms)( Reference Healton, Savage and Brust 264 , Reference Savage and Lindenbaum 267 ), and this inverse relationship is reflected in measures of vitamin B12 status( Reference Oberlin, Tangney and Gustashaw 270 , Reference Lindenbaum, Healton and Savage 271 , Reference Carmel, Karnaze and Weiner 304 ) and vitamin B12 analogues( Reference Carmel, Karnaze and Weiner 304 ). This suggests that vitamin B12 utilisation in the deficient state is subject to antagonistic or compensatory mechanisms which are determined at the level of the individual. The relative proportion of individuals affected by each or both sets of symptoms is unclear from the literature, since there is a shortfall of relevant and epidemiologically representative data. With regard to the timeline of symptom development, it is again interesting to note that the great majority of those with only neurological symptoms (about 80 %) do not present until sometime between the fifth and the seventh decade( Reference Healton, Savage and Brust 264 , Reference Lindenbaum, Healton and Savage 271 , Reference Pennypacker, Allen and Kelly 290 ).

Conclusions

Emerging perspectives on sarcopenia, in which declines in muscle mass and function in older age are seen as neuromuscular rather than simply muscular, have raised important questions in terms of research into potential therapies. Nutritional intervention strategies for sarcopenia have a strong theoretical basis and much work in terms of protein and individual amino acids is evident in the literature, with varying success reported. Elderly subjects frequently present with multiple, potentially confounding factors in terms of research, many of which may be nutritional in nature. In this connection the present discussion has examined the potential role of selected B vitamins in normal and pathophysiological modes of neuromuscular function. Several key themes emerge, notably those surrounding the adequacy of the knowledge base in terms of older adults’ requirements for B vitamins and the clear similarities between certain deficiency and disease/frailty states associated with older age. It is possible therefore that compromised intake or function of the B vitamins discussed may induce, mimic or aggravate key aspects of neuromuscular compromise as observed in age-related sarcopenia.

The available evidence regarding the B vitamins considered and their potential role in the development of sarcopenia presents an interesting mix of clinical observations and biophysiological mechanisms. TD, for example, has long been characterised by reduced appetite, weight loss and neural and neuromuscular compromise mirroring aetiological features of age-related sarcopenia (including disproportionate effects in the lower limbs). Niacin deficiency has known associations with anorexia and with reductions in hydrochloric acid in gastric secretions, leading to specific and general nutrient deficiencies. Furthermore, low niacin intake can lead to motor neuron chromatolysis. With respect to vitamin B6 deficiency, axonal degeneration and pathological disturbances in Schwann cells has been described as affecting distal motor neurons and is associated with loss of motor function. In FD, in addition to the correlation between hyperhomocysteinaemia and decline in neuromuscular function, it is possible that the induction of mass misincorporation of uracil into DNA as a result of low folate status and subsequent chromosomal instability could inhibit muscle synthetic and repair processes. Similarly, compromised vitamin B12 status is a known risk for muscle weakness, abnormal reflexes, spasticity, gait ataxia and myelopathies, probably due to demyelination in both the CNS and PNS.

It is concluded therefore that for any meaningful investigation of the development or treatment of sarcopenia, it is important to consider, and where possible eliminate, deficiencies of B vitamins, the lack of which may impinge on muscle, nerve or the neuromuscular nexus. While it is not possible to conclude that one or more B vitamin deficiencies are causative of sarcopenia, within the mélange of known and surmised risk factors, such deficiencies warrant serious consideration. This seems especially true given the ease and relatively low cost of addressing such nutritional inadequacy. The evidence base for recommended daily quantities of micronutrients could usefully be contemporised in terms of adequacy for an ageing demographic, especially where there is a paucity of (functional) metabolic data available. Finally, given the increasing number of individuals reaching their ninth decade or beyond, it is worth considering future recommendations for older adults nuanced beyond a 65-years-and-over category.

Acknowledgements

This research received no specific grant from any funding agency, commercial or not-for-profit sectors. N. A.’s work was supported by a PhD scholarship from London South Bank University, UK.

N. A. and A. D. C. performed the database searches and the initial interpretation of the literature data. N. A., A. D. C. and K. N. M. contributed to the planning and writing of the paper. All authors critically reviewed the manuscript.

None of the authors has any financial or non-financial interests to declare that may conflict with the provision of their scientific input to this paper.

References

1. Rosenberg, I (1989) Summary comments: epidemiological and methodological problems in determining nutritional status of older persons. Am J Clin Nutr 50, 12311233.Google Scholar
2. Cruz-Jentoft, AJ, Baeyens, JP, Bauer, JM, et al. (2010) Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 39, 412423.Google Scholar
3. Goodpaster, BH, Park, SW, Harris, TB, et al. (2006) The loss of skeletal muscle strength, mass, and quality in older adults: The Health, Aging and Body Composition Study. J Gerontol A Biol Sci Med Sci 61, 10591064.Google Scholar
4. Landi, F, Calvani, R, Cesari, M, et al. (2015) Sarcopenia as the biological substrate of physical frailty. Clin Geriatr Med 31, 367374.Google Scholar
5. dos Santos, L, Cyrino, ES, Antunes, M, et al. (2016) Sarcopenia and physical independence in older adults: the independent and synergic role of muscle mass and muscle function. J Cachexia Sarcopenia Muscle 8, 245250.Google Scholar
6. Landi, F, Liperoti, R, Russo, A, et al. (2012) Sarcopenia as a risk factor for falls in elderly individuals: results from the ilSIRENTE study. Clin Nutr 31, 652658.Google Scholar
7. Clynes, MA, Edwards, MH, Buehring, B, et al. (2015) Definitions of sarcopenia: associations with previous falls and fracture in a population sample. Calcif Tissue Int 97, 445452.Google Scholar
8. Office for National Statistics (2012) Population Ageing in the United Kingdom, its Constituent Countries and the European Union. http://webarchive.nationalarchives.gov.uk/20160105160709/http://www.ons.gov.uk/ons/dcp171776_258607.pdf (accessed June 2016).Google Scholar
9. Paddon-Jones, D, Sheffield-Moore, M, Zhang, X-J, et al. (2004) Amino acid ingestion improves muscle protein synthesis in the young and elderly. Am J Physiol Endocrinol Metab 286, E321E328.Google Scholar
10. Katsanos, CS (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. AJP Endocrinol Metab 291, E381E387.Google Scholar
11. Cuthbertson, D, Smith, K, Babraj, J, et al. (2005) Anabolic signaling deficits underlie amino acid resistance of wasting, aging muscle. FASEB J 19, 422424.Google Scholar
12. Kant, AK & Graubard, BI (2007) Secular trends in the association of socio-economic position with self-reported dietary attributes and biomarkers in the US population: National Health and Nutrition Examination Survey (NHANES) 1971–1975 to NHANES 1999–2002. Public Health Nutr 10, 158167.Google Scholar
13. Saka, B, Kaya, O, Ozturk, GB, et al. (2010) Malnutrition in the elderly and its relationship with other geriatric syndromes. Clin Nutr 29, 745748.Google Scholar
14. Han, SY & Kim, CS (2016) Does denture-wearing status in edentulous South Korean elderly persons affect their nutritional intakes? Gerodontology 33, 169176.Google Scholar
15. Kagawa, R, Ikebe, K, Inomata, C, et al. (2012) Effect of dental status and masticatory ability on decreased frequency of fruit and vegetable intake in elderly Japanese subjects. Int J Prosthodont 25, 368375.Google Scholar
16. Mojet, J (2003) Taste perception with age: generic or specific losses in supra-threshold intensities of five taste qualities? Chem Senses 28, 397413.Google Scholar
17. Kim, H-S, Oh, C & No, J-K (2016) Can nutrition label recognition or usage affect nutrition intake according to age? Nutrition 32, 5660.Google Scholar
18. Valuck, RJ & Ruscin, JM (2004) A case–control study on adverse effects: H2 blocker or proton pump inhibitor use and risk of vitamin B12 deficiency in older adults. J Clin Epidemiol 57, 422428.Google Scholar
19. Keller, HH (1993) Malnutrition in institutionalized elderly: how and why? J Am Geriatr Soc 41, 12121218.Google Scholar
20. Sharkey, JR, Branch, LG, Zohoori, N, et al. (2002) Inadequate nutrient intakes among homebound elderly and their correlation with individual characteristics and health-related factors. Am J Clin Nutr 76, 14351445.Google Scholar
21. De Groot, CPGM, Van Den Broek, T & Van Staveren, W (1999) Energy intake and micronutrient intake in elderly Europeans: seeking the minimum requirement in the SENECA study. Age Ageing 28, 469474.Google Scholar
22. Jamieson, CP, Obeid, OA & Powell-Tuck, J (1999) The thiamin, riboflavin and pyridoxine status of patients on emergency admission to hospital. Clin Nutr 18, 8791.Google Scholar
23. Marshall, TA, Stumbo, PJ, Warren, JJ, et al. (2001) Inadequate nutrient intakes are common and are associated with low diet variety in rural, community-dwelling elderly. J Nutr 131, 21922196.Google Scholar
24. Bartali, B, Frongillo, EA, Bandinelli, S, et al. (2006) Low nutrient intake is an essential component of frailty in older persons. J Gerontol Ser A Biol Sci Med Sci 61, 589593.Google Scholar
25. Semba, RD, Bartali, B, Zhou, J, et al. (2006) Low serum micronutrient concentrations predict frailty among older women living in the community. J Gerontol A Biol Sci Med Sci 61, 594599.Google Scholar
26. Mendonça, N, Hill, TR, Granic, A, et al. (2016) Macronutrient intake and food sources in the very old: analysis of the Newcastle 85+ Study. Br J Nutr 115, 21702180.Google Scholar
27. Public Health England (2014) National Diet and Nutrition Survey Results from Years 1, 2, 3 and 4 (Combined) of the Rolling Programme (2008/2009–2011/2012). London: HM Stationery Office.Google Scholar
28. Bates, CJ, Prentice, A, van der Pols, JC, et al. (1998) Estimation of the use of dietary supplements in the National Diet and Nutrition Survey: people aged 65 years and over. An observed paradox and a recommendation. Eur J Clin Nutr 52, 917923.Google Scholar
29. Fabian, E, Bogner, M, Kickinger, A, et al. (2012) Vitamin status in elderly people in relation to the use of nutritional supplements. J Nutr Health Aging 16, 206212.Google Scholar
30. Schwarzpaul, S, Strassburg, A, Lührmann, PM, et al. (2006) Intake of vitamin and mineral supplements in an elderly German population. Ann Nutr Metab 50, 155162.Google Scholar
31. Bates, B, Lennox, A, Prentice, A, et al. (2012) National Diet and Nutrition Survey: Headline Results from Years 1, 2 and 3 (Combined) of the Rolling Programme (2008/2009–2010/11). London: HM Stationery Office.Google Scholar
32. Bailey, AL, Maisey, S, Southon, S, et al. (1997) Relationships between micronutrient intake and biochemical indicators of nutrient adequacy in a ‘free-living’ elderly UK population. Br J Nutr 77, 225242.Google Scholar
33. Tanphaicitr, V (1999) Modern Nutrition in Health and Disease, 9th ed., pp. 381389 [M Shils, JA Olson, M Shike et al., editors]. Baltimore, MD: Williams & Wilkins.Google Scholar
34. Scheid, HE, Bennett, BA & Schweigert, BS (1953) Thiamine, riboflavin, and niacin content of organ meats. J Food Sci 18, 109112.Google Scholar
35. Martin, P, Singleton, C & Hiller-Sturmhöfel, S (2003) The role of thiamine deficiency in alcoholic brain disease. Alcohol Res Heal 27, 174181.Google Scholar
36. Thomson, AD & Pratt, OE (1992) Interaction of nutrients and alcohol: absorption, transport, utilization and metabolism. In Nutrition and Alcohol, 2nd ed., pp. 7599 [RR Watson and B Watzl, editors]. Boca Raton, FL: CRC Press.Google Scholar
37. Williams, RD (1940) Observations on induced thiamine (vitamin B1) deficiency in man. Arch Intern Med 66, 785799.Google Scholar
38. Phillips, GB, Victor, M, Adams, RD, et al. (1952) A study of the nutritional defect in Wernicke’s syndrome; the effect of a purified diet, thiamine, and other vitamins on the clinical manifestations. J Clin Invest 31, 859871.Google Scholar
39. Rindi, G (1996) Thiamine. In Present Knowledge in Nutrition, 7th ed., pp. 160166 [EE Ziegler and LJ Filer, editors]. Washington, DC: ILSI Press.Google Scholar
40. Calingasan, NY, Chun, WJ, Park, LC, et al. (1999) Oxidative stress is associated with region-specific neuronal death during thiamine deficiency. J Neuropathol Exp Neurol 58, 946958.Google Scholar
41. Lukienko, PI, Mel’nichenko, NG, Zverinskii, IV, et al. (2000) Antioxidant properties of thiamine. Bull Exp Biol Med 130, 874876.Google Scholar
42. Gibson, GE & Zhang, H (2002) Interactions of oxidative stress with thiamine homeostasis promote neurodegeneration. Neurochem Int 40, 493504.Google Scholar
43. World Health Organization (1999) Thiamine deficiency and its prevention and control in major emergencies. http://www.who.int/nutrition/publications/emergencies/WHO_NHD_99.13/en/ (accessed March 2015).Google Scholar
44. Janssen, I, Heymsfield, SB, Wang, ZM, et al. (2000) Skeletal muscle mass and distribution in 468 men and women aged 18–88 yr. J Appl Physiol 89, 8188.Google Scholar
45. Nogueira, FRD, Libardi, CA, Vechin, FC, et al. (2013) Comparison of maximal muscle strength of elbow flexors and knee extensors between younger and older men with the same level of daily activity. Clin Interv Aging 8, 401407.Google Scholar
46. Institute of Medicine (US) Standing Committee on the Scientific Evaluation of Dietary Reference Intakes and its Panel on Folate, Other B Vitamins, and Choline (1998) Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B 6 , Folate, Vitamin B 12 , Pantothenic Acid, Biotin and Choline . Washington, DC: National Academies Press.Google Scholar
47. Wilkinson, TJ, Hanger, HC, George, PM, et al. (2000) Is thiamine deficiency in elderly people related to age or co-morbidity? Age Ageing 29, 111116.Google Scholar
48. Pepersack, T, Garbusinski, J, Robberecht, J, et al. (1999) Clinical relevance of thiamine status amongst hospitalized elderly patients. Gerontology 45, 96101.Google Scholar
49. Zhang, G, Ding, H, Chen, H, et al. (2013) Thiamine nutritional status and depressive symptoms are inversely associated among older Chinese adults. J Nutr 143, 5358.Google Scholar
50. Gold, M, Hauser, RA & Chen, MF (1998) Plasma thiamine deficiency associated with Alzheimer’s disease but not Parkinson’s disease. Metab Brain Dis 13, 4353.Google Scholar
51. Pan, X, Fei, G, Lu, J, et al. (2016) Measurement of blood thiamine metabolites for Alzheimer’s disease diagnosis. EBioMedicine 3, 155162.Google Scholar
52. Aikawa, H, Watanabe, IS, Furuse, T, et al. (1984) Low energy levels in thiamine-deficient encephalopathy. J Neuropathol Exp Neurol 43, 276287.Google Scholar
53. Bettendorff, L, Sluse, F, Goessens, G, et al. (1995) Thiamine deficiency – induced partial necrosis and mitochondrial uncoupling in neuroblastoma cells are rapidly reversed by addition of thiamine. J Neurochem 65, 21782184.Google Scholar
54. Ke, Z-J, DeGiorgio, LA, Volpe, BT, et al. (2003) Reversal of thiamine deficiency-induced neurodegeneration. J Neuropathol Exp Neurol 62, 195207.Google Scholar
55. Todd, KG & Butterworth, RF (1999) Early microglial response in experimental thiamine deficiency: an immunohistochemical analysis. Glia 25, 190198.Google Scholar
56. Langlais, PJ, Anderson, G, Guo, SX, et al. (1997) Increased cerebral free radical production during thiamine deficiency. Metab Brain Dis 12, 137143.Google Scholar
57. Todd, KG & Butterworth, RF (1998) Evaluation of the role of NMDA-mediated excitotoxicity in the selective neuronal loss in experimental Wernicke encephalopathy. Exp Neurol 149, 130138.Google Scholar
58. Langlais, PJ & Mair, RG (1990) Protective effects of the glutamate antagonist MK-801 on pyrithiamine-induced lesions and amino acid changes in rat brain. J Neurosci 10, 16641674.Google Scholar
59. Olsen, A, Halkjaer, J, van Gils, CH, et al. (2009) Dietary intake of the water-soluble vitamins B1, B2, B6, B12 and C in 10 countries in the European Prospective Investigation into Cancer and Nutrition. Eur J Clin Nutr 63, Suppl. 4, S122S149.Google Scholar
60. Fabian, E & Elmadfa, I (2008) Nutritional situation of the elderly in the European Union: data of the European Nutrition and Health Report (2004). Ann Nutr Metab 52, Suppl. 1, 5761.Google Scholar
61. Jyväkorpi, SK, Pitkälä, KH, Puranen, TM, et al. (2015) Low protein and micronutrient intakes in heterogeneous older population samples. Arch Gerontol Geriatr 61, 464471.Google Scholar
62. Toffanello, ED, Inelmen, EM, Minicuci, N, et al. (2011) Ten-year trends in vitamin intake in free-living healthy elderly people: the risk of subclinical malnutrition. J Nutr Health Aging 15, 99103.Google Scholar
63. Tur, J, Colomer, M, Bonnin, T, et al. (2005) Dietary intake and nutritional risk among free-living elderly people in Palma de Mallorca. J Nutr Health Aging 9, 390396.Google Scholar
64. ter Borg, S, Verlaan, S, Hemsworth, J, et al. (2015) Micronutrient intakes and potential inadequacies of community-dwelling older adults: a systematic review. Br J Nutr 113, 11951206.Google Scholar
65. Lim, YS & Cho, KJ (2000) A comparative study of nutrient intakes and factors to influence on nutrient intake between low-income elderly living in urban and rural areas. J Korean Soc . Food Sci Nutr 29, 257267.Google Scholar
66. Lengyel, CO, Whiting, SJ & Zello, GA (2008) Nutrient inadequacies among elderly residents of long-term care facilities. Can J Diet Pract Res 69, 8288.Google Scholar
67. Löwik, MR, Schneijder, P, Hulshof, KF, et al. (2013) Institutionalized elderly women have lower food intake than do those living more independently (Dutch Nutrition Surveillance System). J Am Coll Nutr 11, 432440.Google Scholar
68. Gorelik, O, Almoznino-Sarafian, D, Feder, I, et al. (2003) Dietary intake of various nutrients in older patients with congestive heart failure. Cardiology 99, 177181.Google Scholar
69. Berner, YN, Stern, F, Polyak, Z, et al. (2002) Dietary intake analysis in institutionalized elderly: a focus on nutrient density. J Nutr Health Aging 6, 237242.Google Scholar
70. McCabe-Sellers, B, Sharkey, J & Browne, B (2005) Diuretic medication therapy use and low thiamin intake in homebound older adults. J Nutr Elder 24, 5771.Google Scholar
71. Wakimoto, P & Block, G (2001) Dietary intake, dietary patterns, and changes with age: an epidemiological perspective. J Gerontol A Biol Sci Med Sci 56, 6580.Google Scholar
72. Sette, S, Le Donne, C, Piccinelli, R, et al. (2011) The third Italian National Food Consumption Survey, INRAN-SCAI 2005-06 – part 1: nutrient intakes in Italy. Nutr Metab Cardiovasc Dis 21, 922932.Google Scholar
73. Elmadfa, I, Majchrzak, D, Rust, P, et al. (2001) The thiamine status of adult humans depends on carbohydrate intake. Int J Vitam Nutr Res 71, 217221.Google Scholar
74. Suter, PM, Haller, J, Hany, A, et al. (2000) Diuretic use: a risk for subclinical thiamine deficiency in elderly patients. J Nutr Health Aging 4, 6971.Google Scholar
75. Severi, S, Bedogni, G, Manzieri, AM, et al. (1997) Effects of cooking and storage methods on the micronutrient content of foods. Eur J Cancer Prev 6, Suppl. 1, S21S24.Google Scholar
76. UK Food Standards Agency: Expert Group on Vitamins and Minerals (2002) Revised Review of Thiamine. London: Food Standards Agency.Google Scholar
77. Butterworth, RF, Kril, JJ & Harper, CG (1993) Thiamine-dependent enzyme changes in the brains of alcoholics: relationship to the Wernicke–Korsakoff syndrome. Alcohol Clin Exp Res 17, 10841088.Google Scholar
78. Nichols, HK & Basu, TK (1994) Thiamin status of the elderly: dietary intake and thiamin pyrophosphate response. J Am Coll Nutr 13, 5761.Google Scholar
79. Turck, D, Bresson, J, Burlingame, B, et al. (2016) Dietary reference values for thiamin. EFSA J 14, 4653.Google Scholar
80. Andrade Juguan, J, Lukito, W & Schultink, W (1999) Thiamine deficiency is prevalent in a selected group of urban Indonesian elderly people. J Nutr 129, 366371.Google Scholar
81. Wolters, M, Hermann, S & Hahn, A (2003) B vitamin status and concentrations of homocysteine and methylmalonic acid in elderly German women. Am J Clin Nutr 78, 765772.Google Scholar
82. Committee on Medical Aspects of Food Policy (1991) Dietary Reference Values for Food Energy and Nutrients for the United Kingdom. Report on Health and Social Subjects, no. 41. London: HM Stationery Office.Google Scholar
83. Horwitt, MK, Liebert, E, Kreisler, O, et al. (1948) Investigations of Human Requirements for B-complex Vitamins. Bulletin of the National Research Council, no. 116. Washington, DC: National Academies of Sciences.Google Scholar
84. Die Deutsche Gesellschaft für Ernährung e. V. (DGE) (2015) Thiamin (vitamin B1) [in German]. https://www.dge.de/wissenschaft/referenzwerte/thiamin/ (accessed November 2015).Google Scholar
85. National Health and Medical Council (2005) Nutrient reference values for Australia and New Zealand including recommended dietary intakes: evidence appendix. https://www.nhmrc.gov.au/guidelines-publications/n35-n36-n37 (accessed March 2015).Google Scholar
86. Bolzetta, F, Veronese, N, De Rui, M, et al. (2015) Are the recommended dietary allowances for vitamins appropriate for elderly people? J Acad Nutr Diet 115, 17891797.Google Scholar
87. O’Rourke, N, Bunkner, V, Thomas, A, et al. (1990) Thiamine status of healthy and institutionalized elderly subjects: analysis of dietary and biochemical indices. Age Ageing 19, 325329.Google Scholar
88. Schrijver, J (1991) Biochemical markers for micronutrient status and their interpretation. In Modern Lifestyles, Lower Energy Intake and Micronutrient Status. ILSI Human Nutrition Reviews, pp. 5585 [K Pietrzik, editor]. London: Springer.Google Scholar
89. Kjosen, B & Seim, SH (1977) The transketolase assay of thiamine in some diseases. Am J Clin Nutr 30, 15911596.Google Scholar
90. Jacob, R & Swenseid, M (1996) Niacin. In Present Knowledge in Nutrition, 7th ed., pp. 185190 [EE Ziegler and L Filer, editors]. Washington, DC: ILSI Press.Google Scholar
91. Guse, AH (2005) Second messenger function and the structure–activity relationship of cyclic adenosine diphosphoribose (cADPR). FEBS J 272, 45904597.Google Scholar
92. Imai, S, Armstrong, CM, Kaeberlein, M, et al. (2000) Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403, 795800.Google Scholar
93. Zoratti, M & Szabò, I (1995) The mitochondrial permeability transition. Biochim Biophys Acta 1241, 139176.Google Scholar
94. La Piana, G, Marzulli, D, Gorgoglione, V, et al. (2005) Porin and cytochrome oxidase containing contact sites involved in the oxidation of cytosolic NADH. Arch Biochem Biophys 436, 91100.Google Scholar
95. Jaeschke, H, Kleinwaechter, C & Wendel, A (1992) NADH-dependent reductive stress and ferritin-bound Fe in allyl alcohol-induced lipid peroxidation in vivo: the protective effect of vitamin E. Chem Biol Interact 81, 5768.Google Scholar
96. Aswad, F, Kawamura, H & Dennert, G (2005) High sensitivity of CD4+CD25+ regulatory T cells to extracellular metabolites nicotinamide adenine dinucleotide and ATP: a role for P2X7 receptors. J Immunol 175, 30753083.Google Scholar
97. Hegyi, J, Schwartz, RA & Hegyi, V (2004) Pellagra: dermatitis, dementia, and diarrhea. Int J Dermatol 43, 15.Google Scholar
98. Langworthy, OR (1931) Lesions of the central nervous system characteristic of pellagra. Brain 54, 291302.Google Scholar
99. Zimmerman, HM (1934) Pellagra in association with chronic alcoholism. Arch Neurol Psychiatry 31, 290309.Google Scholar
100. Goldsmith, GA, Sarett, HP, Register, UD, et al. (1952) Studies of niacin requirement in man. I. Experimental pellagra in subjects on corn diets low in niacin and tryptophan. J Clin Invest 31, 533542.Google Scholar
101. Goldsmith, GA, Gibbens, J, Unglaub, WG, et al. (1956) Studies of niacin requirement in man: III. Comparative effects of diets containing lime-treated and untreated corn in the production of experimental pellagra. Am J Clin Nutr 4, 151160.Google Scholar
102. Sakai, K, Nakajima, T & Fukuhara, N (2006) A suspected case of alcoholic pellagra encephalopathy with marked response to niacin showing myoclonus and ataxia as chief complaints. Brain Nerve 58, 141144.Google Scholar
103. Terada, N, Kinoshita, K, Taguchi, S, et al. (2015) Wernicke encephalopathy and pellagra in an alcoholic and malnourished patient. BMJ Case Rep 2015, bcr2015209412.Google Scholar
104. Brown, TM (2010) Pellagra: an old enemy of timeless importance. Psychosomatics 51, 9397.Google Scholar
105. Buzina, R (1976) Early signs of niacin deficiency. Bibl Nutr Dieta 23, 8894.Google Scholar
106. Ronthal, M & Adler, H (1969) Motor nerve conduction velocity and the electromyograph in pellagra. South African Med J 43, 642644.Google Scholar
107. Sakellariou, GK, Pearson, T, Lightfoot, AP, et al. (2016) Mitochondrial ROS regulate oxidative damage and mitophagy but not age-related muscle fiber atrophy. Sci Rep 6, 33944.Google Scholar
108. Fomby, P & Cherlin, AJ (2011) Role of poly(ADP-ribose) polymerase 1 (PARP-1) in CVDs: the therapeutic potential of PARP inhibitors. Cardiovasc Drug Rev 72, 181204.Google Scholar
109. Kim, MY, Mauro, S, Gévry, N, et al. (2004) NAD+-dependent modulation of chromatin structure and transcription by nucleosome binding properties of PARP-1. Cell 119, 803814.Google Scholar
110. Pillai, JB, Isbatan, A, Imai, S-I, et al. (2005) Poly(ADP-ribose) polymerase-1-dependent cardiac myocyte cell death during heart failure is mediated by NAD+ depletion and reduced Sir2α deacetylase activity. J Biol Chem 280, 4312143130.Google Scholar
111. Alano, CC, Garnier, P, Ying, W, et al. (2010) NAD+ depletion is necessary and sufficient for poly(ADP-ribose) polymerase-1-mediated neuronal death. J Neurosci 30, 29672978.Google Scholar
112. Jacobson, EL, Nunbhakdi-Craig, V, Smith, DG, et al. (1992) ADP-ribose polymer metabolism: implications for human nutrition. In ADP-Ribosylation Reactions, 1st ed., pp. 153162 [G Poirer and P Moreau, editors]. New York: Academic Press.Google Scholar
113. Klaidman, LK, Mukherjee, SK & Adams, JD Jr (2001) Oxidative changes in brain pyridine nucleotides and neuroprotection using nicotinamide. Biochim Biophys Acta 1525, 136148.Google Scholar
114. Gomes, AP, Price, NL, Ling, AJY, et al. (2013) Declining NAD+ induces a pseudohypoxic state disrupting nuclear–mitochondrial communication during aging. Cell 155, 16241638.Google Scholar
115. Liu, D, Gharavi, R, Pitta, M, et al. (2009) Nicotinamide prevents NAD+ depletion and protects neurons against excitotoxicity and cerebral ischemia: NAD+ consumption by sirt1 may endanger energetically compromised neurons. NeuroMolecular Med 11, 2842.Google Scholar
116. Narayan, N, Lee, IH, Borenstein, R, et al. (2012) The NAD-dependent deacetylase SIRT2 is required for programmed necrosis. Nature 492, 199204.Google Scholar
117. Hall, JA, Dominy, JE, Lee, Y, et al. (2013) The sirtuin family’s role in aging and age-associated pathologies. J Clin Invest 123, 973979.Google Scholar
118. Skoge, RH, Dölle, C & Ziegler, M (2014) Regulation of SIRT2-dependent α-tubulin deacetylation by cellular NAD levels. DNA Repair (Amst) 23, 3338.Google Scholar
119. Braidy, N, Guillemin, GJ, Mansour, H, et al. (2011) Age related changes in NAD+ metabolism oxidative stress and Sirt1 activity in Wistar rats. PLoS ONE 6, e19194.Google Scholar
120. Massudi, H, Grant, R, Braidy, N, et al. (2012) Age-associated changes in oxidative stress and NAD+ metabolism in human tissue. PLOS ONE 7, e42357.Google Scholar
121. Deierlein, AL, Morland, KB, Scanlin, K, et al. (2014) Diet quality of urban older adults age 60 to 99 years: The Cardiovascular Health of Seniors and Built Environment Study. J Acad Nutr Diet 114, 279287.Google Scholar
122. Troesch, B, Hoeft, B, McBurney, M, et al. (2012) Dietary surveys indicate vitamin intakes below recommendations are common in representative Western countries. Br J Nutr 108, 692698.Google Scholar
123. Sahyoun, N (1992) Nutrition in the elderly: the Boston Nutritional Status Survey. In Nutritent Intake by the NSS Elderly Population, 1st ed., pp. 3144 [S Hartz S, IH Rosenberg and RM Russell, editors]. London: Smith-Gordon and Co. Ltd.Google Scholar
124. Woo, J, Ho, SC, Mak, YT, et al. (1988) Nutritional status of the water-soluble vitamins in an active Chinese elderly population in Hong Kong. Eur J Clin Nutr 42, 415424.Google Scholar
125. Aghdassi, E, McArthur, M, Liu, B, et al. (2007) Dietary intake of elderly living in Toronto long-term care facilities: comparison to the dietary reference intake. Rejuvenation Res 10, 301309.Google Scholar
126. Paulionis, L, Kane, S-L & Meckling, KA (2005) Vitamin status and cognitive function in a long-term care population. BMC Geriatr 5, 16.Google Scholar
127. Engelheart, S & Akner, G (2015) Dietary intake of energy, nutrients and water in elderly people living at home or in nursing home. J Nutr Health Aging 19, 265272.Google Scholar
128. Dhonukshe-Rutten, RAM, de Vries, JHM, de Bree, A, et al. (2009) Dietary intake and status of folate and vitamin B12 and their association with homocysteine and cardiovascular disease in European populations. Eur J Clin Nutr 63, 1830.Google Scholar
129. Shelnutt, KP, Kauwell, GPA, Chapman, CM, et al. (2003) Folate status response to controlled folate intake is affected by the methylenetetrahydrofolate reductase 677C→T polymorphism in young women. J Nutr 133, 41074111.Google Scholar
130. Gregory, J (1998) Nutritional properties and significance of vitamin glycosides. Annu Rev Nutr 18, 277296.Google Scholar
131. Hokwitt, MK, Harvey, CC, Rothwell, WS, et al. (1956) Tryptophan–niacin relationships in man. J Nutr 60, 143.Google Scholar
132. Nakagawa, I, Takahashi, T, Sasaki, A, et al. (1973) Efficiency of conversion of tryptophan to niacin in humans. J Nutr 103, 11951199.Google Scholar
133. Institute of Medicine (US) Standing Committee on the Scientific Evaluation of Dietary Reference Intakes and its Panel on Folate, Other B Vitamins, and Choline and Subcommittee on Upper Reference Levels of Nutrients (1998) ) Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B 6 , Folate, Vitamin B 12 , Pantothenic Acid, Biotin, and Choline . Washington, DC: National Academies Press.Google Scholar
134. Bender, DA (1989) Vitamin B6 requirements and recommendations. Eur J Clin Nutr 43, 289309.Google Scholar
135. Vogler, WR & Mingioli, ES (1965) Heme synthesis in pyridoxine-responsive anemia. N Engl J Med 273, 347353.Google Scholar
136. Hartvig, P, Lindner, KJ, Bjurling, P, et al. (1995) Pyridoxine effect on synthesis rate of serotonin in the monkey brain measured with positron emission tomography. J Neural Transm Gen Sect 102, 9197.Google Scholar
137. Lamers, Y, Williamson, J, Ralat, M, et al. (2009) Moderate dietary vitamin B-6 restriction raises plasma glycine and cystathionine concentrations while minimally affecting the rates of glycine turnover and glycine cleavage in healthy men and women. J Nutr 139, 452460.Google Scholar
138. Allgood, V & Cidlowski, J (1992) Vitamin B6 modulates transcriptional activation by multiple members of the steroid hormone receptor superfamily. J Biol Chem 267, 38193824.Google Scholar
139. Mueller, JF & Iacono, JM (1963) Effect of desoxypyridoxine-induced vitamin B6 deficiency on polyunsaturated fatty acid metabolism in human beings. Am J Clin Nutr 12, 358367.Google Scholar
140. Zaoui, A, Abdelghani, A, Ben Salem, H, et al. (2012) Early-onset severe isoniazid-induced motor-dominant neuropathy: a case report. East Mediterr Health J 18, 298299.Google Scholar
141. Vilter, RW, Mueller, JF, Glazer, HS, et al. (1953) The effect of vitamin B6 deficiency induced by desoxypyridoxine in human beings. J Lab Clin Med 42, 335357.Google Scholar
142. Steichen, O, Martinez-Almonya, L & de Broucker, T (2006) Isoniazid induced neuropathy: consider prevention. Rev Mal Respir 23, 157160.Google Scholar
143. Devadatta, S, Gangadharam, PR, Andrews, RH, et al. (1960) Peripheral neuritis due to isoniazid. Bull World Health Organ 23, 587598.Google Scholar
144. Schalaepfer, WW & Hager, H (1964) Ultrastructural studies of INH-induced neuropathy in rats. I. Early axonal changes. Am J Pathol 45, 209219.Google Scholar
145. Jacobs, JM, Miller, RH, Whittle, A, et al. (1979) Studies on the early changes in acute isoniazid neuropathy in the rat. Acta Neuropathol 47, 8592.Google Scholar
146. Cavanagh, JB (1967) On the pattern of change in peripheral nerves produced by isoniazid intoxication in rats. J Neurol Neurosurg Psychiatry 30, 2633.Google Scholar
147. Zbinden, G & Studer, A (1955) Experimental contribution to the question of isoniazid neuritis and the effects of pyridoxin on it. Z Tuberk 107, 97107.Google Scholar
148. Hildebrand, J, Joffroy, A & Coërs, C (1968) Myoneural changes in experimental isoniazid neuropathy. Arch Neurol 19, 6070.Google Scholar
149. Sampson, DA, Young, LA & Kretsch, MJ (1988) Marginal intake of vitamin B-6: effects on protein synthesis in liver, kidney and muscle of the rat. Nutr Res 8, 309319.Google Scholar
150. Bartali, B, Semba, RD, Frongillo, EA, et al. (2006) Low micronutrient levels as a predictor of incident disability in older women. Arch Intern Med 166, 23352340.Google Scholar
151. Ter Borg, S, de Groot, LCPGMPGM, Mijnarends, DM, et al. (2016) Differences in nutrient intake and biochemical nutrient status between sarcopenic and nonsarcopenic older adults – results from the Maastricht Sarcopenia Study. J Am Med Dir Assoc 17, 393401.Google Scholar
152. Cederholm, T, Cruz-Jentoft, AJ & Maggi, S (2013) Sarcopenia and fragility fractures. Eur J Phys Rehabil Med 49, 111117.Google Scholar
153. Richelson, LS, Wahner, HW, Melton, LJ, et al. (1984) Relative contributions of aging and estrogen deficiency to postmenopausal bone loss. N Engl J Med 311, 12731275.Google Scholar
154. Dai, Z, Wang, R, Ang, LW, Yuan, J-M, et al. (2013) Dietary B vitamin intake and risk of hip fracture: the Singapore Chinese Health Study. Osteoporos Int 24, 20492059.Google Scholar
155. Yazdanpanah, N, Zillikens, MC, Rivadeneira, F, et al. (2007) Effect of dietary B vitamins on BMD and risk of fracture in elderly men and women: the Rotterdam Study. Bone 41, 987994.Google Scholar
156. Bird, TA & Levene, CI (1982) Lysyl oxidase: evidence that pyridoxal phosphate is a cofactor. Biochem Biophys Res Commun 108, 11721180.Google Scholar
157. Herrmann, M, Widmann, T & Herrmann, W (2005) Homocysteine – a newly recognised risk factor for osteoporosis. Clin Chem Lab Med 43, 11111117.Google Scholar
158. van Wijngaarden, JP, Doets, EL, Szczecińska, A, et al. (2013) Vitamin B12, folate, homocysteine, and bone health in adults and elderly people: a systematic review with meta-analyses. J Nutr Metab 2013, 486186.Google Scholar
159. Fratoni, V & Brandi, ML (2015) B vitamins, homocysteine and bone health. Nutrients 7, 21762192.Google Scholar
160. European Food Safety Authority (EFSA) Panel on Dietetic Products, Nutrition and Allergies (2016) Scientific opinion on dietary reference values for vitamin B6 . EFSA J 14, 4485.Google Scholar
161. Dewolfe, J & Millan, K (2003) Dietary intake of older adults in the Kingston area. Can J Diet Pract Res 64, 1624.Google Scholar
162. Szponar, L & Rychlik, E (2002) Dietary intake elderly subjects in rural and urban area in Poland. Pol Merkur Lekarski 13, 490496.Google Scholar
163. Kant, AK & Block, G (1990) Dietary vitamin B-6 intake and food sources in the US population: NHANES II, 1976–1980. Am J Clin Nutr 52, 707716.Google Scholar
164. Bechthold, A, Albrecht, V, Leschik-Bonnet, E, et al. (2012) Statement: evaluation of vitamin supplies in Germany. Data on vitamin intake. Ernährungs Umschau 59, 324336.Google Scholar
165. Waern, RVR, Cumming, RG, Blyth, F, et al. (2015) Adequacy of nutritional intake among older men living in Sydney, Australia: findings from the Concord Health and Ageing in Men Project (CHAMP). Br J Nutr 114, 812821.Google Scholar
166. Power, SE, Jeffery, IB, Ross, RP, et al. (2014) Food and nutrient intake of Irish community-dwelling elderly subjects: who is at nutritional risk? J Nutr Health Aging 18, 561572.Google Scholar
167. Paker-Eichelkraut, HS, Bai-Habelski, JC, Overzier, S, et al. (2013) Nutritional status and related factors in elderly nursing home residents: comparative cross-sectional study in migrants and native Germans. J Nutr Gerontol Geriatr 32, 330342.Google Scholar
168. Buell, JS, Arsenault, LN, Scott, TM, et al. (2007) Multivitamin use and B vitamin status in a homebound elderly population. J Nutr Health Aging 11, 299303.Google Scholar
169. Morris, MS, Picciano, MF, Jacques, PF, et al. (2008) Plasma pyridoxal 5’-phosphate in the US population: the National Health and Nutrition Examination Survey, 2003–2004. Am J Clin Nutr 87, 14461454.Google Scholar
170. Kjeldby, IK, Fosnes, GS, Ligaarden, SC, et al. (2013) Vitamin B6 deficiency and diseases in elderly people – a study in nursing homes. BMC Geriatr 13, 13.Google Scholar
171. Bates, CJ, Pentieva, KD, Prentice, A, et al. (1999) Plasma pyridoxal phosphate and pyridoxic acid and their relationship to plasma homocysteine in a representative sample of British men and women aged 65 years and over. Br J Nutr 81, 191201.Google Scholar
172. Huang, YC, Yan, YY, Wong, Y, et al. (2001) Vitamin B6 intakes and status assessment of elderly men and women in Taiwan. Int J Vitam Nutr Res 71, 313318.Google Scholar
173. Guilland, JC, Bereksi-Reguig, B, Lequeu, B, et al. (1984) Evaluation of pyridoxine intake and pyridoxine status among aged institutionalised people. Int J Vitam Nutr Res 54, 185193.Google Scholar
174. Ueland, PM, Ulvik, A, Rios-Avila, L, et al. (2015) Direct and functional biomarkers of vitamin B6 status. Annu Rev Nutr 35, 3370.Google Scholar
175. Canham, JE, Baker, EM, Harding, RS, et al. (1969) Dietary protein – its relationship to vitamin B6 requirements and function. Ann N Y Acad Sci 166, 1629.Google Scholar
176. Miller, LT & Linkswiler, H (1967) Effect of protein intake on the development of abnormal tryptophan metabolism by men during vitamin B6 depletion. J Nutr 93, 5359.Google Scholar
177. Kelsay, J, Baysal, A & Linkswiler, H (1968) Effect of vitamin B6 depletion on the pyridoxal, pyridoxamine and pyridoxine content of the blood and urine of men. J Nutr 94, 490494.Google Scholar
178. Kelsay, J, Miller, LT & Linkswiler, H (1968) Effect of protein intake on the excretion of quinolinic acid and niacin metabolites by men during vitamin B6 depletion. J Nutr 94, 2731.Google Scholar
179. Campbell, W, Crim, M, Dallal, G, et al. (1994) Increased protein requirements in elderly people: new data and retrospective reassessments. Am J Clin Nutr 60, 501509.Google Scholar
180. Phillips, SM, Chevalier, S & Leidy, HJ (2016) Protein “requirements” beyond the RDA: implications for optimizing health. Appl Physiol Nutr Metab 41, 565572.Google Scholar
181. Meydani, SN, Ribaya-Mercado, JD, Russell, RM, et al. (1991) Vitamin B-6 deficiency impairs interleukin 2 production and lymphocyte proliferation in elderly adults. Am J Clin Nutr 53, 12751280.Google Scholar
182. Ribaya-Mercado, JD, Russell, RM, Sahyoun, N, et al. (1991) Vitamin B-6 requirements of elderly men and women. J Nutr 121, 10621074.Google Scholar
183. Williams, P (2007) Nutritional composition of red meat. Nutr Diet 64, S113S119.Google Scholar
184. Paiardini, A, Contestabile, R, Buckle, AM, et al. (2014) PLP-dependent enzymes. Biomed Res Int 2014, 856076.Google Scholar
185. Wyckoff, KF & Ganji, V (2007) Proportion of individuals with low serum vitamin B-12 concentrations without macrocytosis is higher in the post folic acid fortification period than in the pre folic acid fortification period. Am J Clin Nutr 86, 11871192.Google Scholar
186. Hoffbrand, AV & Jackson, BF (1993) Correction of the DNA synthesis defect in vitamin B12 deficiency by tetrahydrofolate: evidence in favour of the methyl-folate trap hypothesis as the cause of megaloblastic anaemia in vitamin B12 deficiency. Br J Haematol 83, 643647.Google Scholar
187. Banerjee, RV & Matthews, RG (1990) Cobalamin-dependent methionine synthase. FASEB J 4, 14501459.Google Scholar
188. Nijhout, HF, Reed, MC, Budu, P, et al. A mathematical model of the folate cycle: new insights into folate homeostasis. J Biol Chem 279, 5500855016.Google Scholar
189. Bates, CJ, Mansoor, MA, Pentieva, KD, et al. (2010) Biochemical risk indices, including plasma homocysteine, that prospectively predict mortality in older British people: the National Diet and Nutrition Survey of People Aged 65 Years and Over. Br J Nutr 104, 893899.Google Scholar
190. Carmel, R (2001) Folate deficiency. In Homocysteine in Health and Disease, 2nd ed., pp. 271288 [R Carmel and D Jacobsen, editors]. Cambridge: Cambridge University Press.Google Scholar
191. Pietrzik, K & Bronstrup, A (1998) Vitamins B12, B6 and folate as determinants of homocysteine concentration in the healthy population. Eur J Pediatr 157, 135138.Google Scholar
192. Zylberstein, DE, Lissner, L, Björkelund, C, et al. (2011) Midlife homocysteine and late-life dementia in women. A prospective population study. Neurobiol Aging 32, 380386.Google Scholar
193. Seshadri, S, Beiser, A, Selhub, J, et al. (2002) Plasma homocysteine as a risk factor for dementia and Alzheimer’s disease. N Engl J Med 346, 476483.Google Scholar
194. Kado, DM, Bucur, A, Selhub, J, et al. (2002) Homocysteine levels and decline in physical function: MacArthur Studies of Successful Aging. Am J Med 113, 537542.Google Scholar
195. Ng, T-P, Aung, KCY, Feng, L, et al. (2012) Homocysteine, folate, vitamin B-12, and physical function in older adults: cross-sectional findings from the Singapore Longitudinal Ageing Study. Am J Clin Nutr 96, 13621368.Google Scholar
196. Swart, KMA, Van Schoor, NM, Heymans, MW, et al. (2013) Elevated homocysteine levels are associated with low muscle strength and functional limitations in older persons. J Nutr Health Aging 17, 578584.Google Scholar
197. Yang, L-K, Wong, K-C, Wu, M-Y, et al. (2007) Correlations between folate, B12, homocysteine levels, and radiological markers of neuropathology in elderly post-stroke patients. J Am Coll Nutr 26, 272278.Google Scholar
198. Stanojlovic, O, Hrncic, D, Rasic-Markovic, A, et al. (2011) Homocysteine: neurotoxicity and mechanisms of induced hyperexcitability. Serbian J Exp Clin Res 12, 39.Google Scholar
199. Bukharaeva, E, Shakirzyanova, A, Khuzakhmetova, V, et al. (2015) Homocysteine aggravates ROS-induced depression of transmitter release from motor nerve terminals: potential mechanism of peripheral impairment in motor neuron diseases associated with hyperhomocysteinemia. Front Cell Neurosci 9, 391.Google Scholar
200. Kruman, II, Culmsee, C, Chan, SL, et al. (2000) Homocysteine elicits a DNA damage response in neurons that promotes apoptosis and hypersensitivity to excitotoxicity. J Neurosci 20, 69206926.Google Scholar
201. Tasatargil, A, Dalaklioglu, S & Sadan, G (2004) Poly(ADP-ribose) polymerase inhibition prevents homocysteine-induced endothelial dysfunction in the isolated rat aorta. Pharmacology 72, 99105.Google Scholar
202. Zhang, X, Chen, S, Li, L, et al. (2008) Folic acid protects motor neurons against the increased homocysteine, inflammation and apoptosis in SOD1 G93A transgenic mice. Neuropharmacology 54, 11121119.Google Scholar
203. Rogers, LM, Pfeiffer, CM, Bailey, LB, et al. (1997) A dual-label stable-isotopic protocol is suitable for determination of folate bioavailability in humans: evaluation of urinary excretion and plasma folate kinetics of intravenous and oral doses of [13C5] and [2H2]folic acid. J Nutr 127, 23212327.Google Scholar
204. Wright, AJA, Finglas, PM, Dainty, JR, et al. (2005) Differential kinetic behavior and distribution for pteroylglutamic acid and reduced folates: a revised hypothesis of the primary site of PteGlu metabolism in humans. J Nutr 135, 619623.Google Scholar
205. Whitehead, VM, Kamen, BA & Beaulieu, D (1987) Levels of dihydrofolate reductase in livers of birds, animals, primates, and man. Cancer Drug Deliv 4, 185189.Google Scholar
206. Carreras, CW & Santi, DV (1995) The catalytic mechanism and structure of thymidylate synthase. Annu Rev Biochem 64, 721762.Google Scholar
207. Goulian, M, Bleile, B & Tseng, BY (1980) Methotrexate-induced misincorporation of uracil into DNA. Proc Natl Acad Sci U S A 77, 19561960.Google Scholar
208. Wickramasinghe, S & Fida, S (1994) Bone marrow cells from vitamin B12- and folate-deficient patients misincorporate uracil into DNA. Blood 83, 16561661.Google Scholar
209. Luzzatto, L, Falusi, AO & Joju, EA (1981) Uracil in DNA in megaloblastic anemia. N Engl J Med 305, 11561157.Google Scholar
210. Blount, BC, Mack, MM, Wehr, CM, et al. (1997) Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci U S A 94, 32903295.Google Scholar
211. Duthie, SJ, Narayanan, S, Blum, S, et al. (2000) Folate deficiency in vitro induces uracil misincorporation and DNA hypomethylation and inhibits DNA excision repair in immortalized normal human colon epithelial cells. Nutr Cancer 37, 245251.Google Scholar
212. Duthie, SJ (2000) Increased uracil misincorporation in lymphocytes from folate-deficient rats. Br J Cancer 83, 15321537.Google Scholar
213. Kronenberg, G, Harms, C, Sobol, RW, et al. (2008) Folate deficiency induces neurodegeneration and brain dysfunction in mice lacking uracil DNA glycosylase. J Neurosci 28, 72197230.Google Scholar
214. Eckart, S, Hörtnagl, H, Kronenberg, G, et al. (2013) Reduced nerve growth factor levels in stress-related brain regions of folate-deficient mice. Neuroscience 245, 129135.Google Scholar
215. Crott, JW, Choi, S-W, Branda, RF, et al. (2005) Accumulation of mitochondrial DNA deletions is age, tissue and folate-dependent in rats. Mutat Res Mol Mech Mutagen 570, 6370.Google Scholar
216. Hikida, RS, Staron, RS, Hagerman, FC, et al. (2000) Effects of high-intensity resistance training on untrained older men. II. Muscle fiber characteristics and nucleo-cytoplasmic relationships. J Gerontol 55A, B347B354.Google Scholar
217. Allen, DL, Monke, SR, Talmadge, RJ, et al. (1995) Plasticity of myonuclear number in hypertrophied and atrophied mammalian skeletal muscle fibers. J Appl Physiol 78, 19691976.Google Scholar
218. Kadi, F & Thornell, L-E (2000) Concomitant increases in myonuclear and satellite cell content in female trapezius muscle following strength training. Histochem Cell Biol 113, 99103.Google Scholar
219. Sinha-Hikim, I, Roth, SM, Lee, MI, et al. (2003) Testosterone-induced muscle hypertrophy is associated with an increase in satellite cell number in healthy, young men. Am J Physiol Endocrinol Metab 285, E197E205.Google Scholar
220. Teixeira, CE & Duarte, JA (2011) Myonuclear domain in skeletal muscle fibers. A critical review. Arch Exerc Health Dis 2, 92101.Google Scholar
221. Ortega, RM, Mañas, LR, Andrés, P, et al. (1996) Functional and psychic deterioration in elderly people may be aggravated by folate deficiency. J Nutr 126, 19921999.Google Scholar
222. Jiménez-Redondo, S, Beltrán de Miguel, B, Gavidia Banegas, J, et al. (2014) Influence of nutritional status on health-related quality of life of non-institutionalized older people. J Nutr Health Aging 18, 359364.Google Scholar
223. Kamphuis, MH, Geerlings, MI, Grobbee, DE, et al. (2008) Dietary intake of B6–9–12 vitamins, serum homocysteine levels and their association with depressive symptoms: the Zutphen Elderly Study. Eur J Clin Nutr 62, 939945.Google Scholar
224. Volkert, D, Kreuel, K, Heseker, H, et al. (2004) Energy and nutrient intake of young-old, old-old and very-old elderly in Germany. Eur J Clin Nutr 58, 11901200.Google Scholar
225. Morris, MC, Evans, DA, Bienias, JL, et al. (2005) Dietary folate and vitamin B12 intake and cognitive decline among community-dwelling older persons. Arch Neurol 62, 641645.Google Scholar
226. Ramos, MI, Allen, LH, Mungas, DM, et al. (2005) Low folate status is associated with impaired cognitive function and dementia in the Sacramento Area Latino Study on Aging. Am J Clin Nutr 82, 13461352.Google Scholar
227. Planells, E, Sánchez, C, Montellano, MA, et al. (2003) Vitamins B6 and B12 and folate status in an adult Mediterranean population. Eur J Clin Nutr 57, 777785.Google Scholar
228. Polito, A, Intorre, F, Andriollo-Sanchez, M, et al. (2005) Estimation of intake and status of vitamin A, vitamin E and folate in older European adults: the ZENITH. Eur J Clin Nutr 59, S42S47.Google Scholar
229. Pfeiffer, CM, Caudill, SP, Gunter, EW, et al. (2005) Biochemical indicators of B vitamin status in the US population after folic acid fortification: results from the National Health and Nutrition Examination Survey 1999–2000. Am J Clin Nutr 82, 442450.Google Scholar
230. O’Leary, F, Flood, VM, Petocz, P, et al. (2011) B vitamin status, dietary intake and length of stay in a sample of elderly rehabilitation patients. J Nutr Health Aging 15, 485489.Google Scholar
231. Dimopoulos, N, Piperi, C, Salonicioti, A, et al. (2007) Correlation of folate, vitamin B12 and homocysteine plasma levels with depression in an elderly Greek population. Clin Biochem 40, 604608.Google Scholar
232. Public Health England (2015) National Diet and Nutrition Survey Rolling Programme (NDNS RP). Supplementary Report: Blood Folate Results for the UK as a Whole, Scotland, Northern Ireland (Years 1 to 4 Combined) and Wales (Years 2 to 5 Combined). London: Public Health England.Google Scholar
233. Clarke, R, Grimley Evans, J, Schneede, J, et al. (2003) Vitamin B12 and folate deficiency in later life. Age Ageing 33, 3441.Google Scholar
234. Koehler, KM, Baumgartner, RN, Garry, PJ, et al. (2001) Association of folate intake and serum homocysteine in elderly persons according to vitamin supplementation and alcohol use. Am J Clin Nutr 73, 628637.Google Scholar
235. Lindenbaum, J, Rosenberg, IH, Wilson, PW, et al. (1994) Prevalence of cobalamin deficiency in the Framingham elderly population. Am J Clin Nutr 60, 211.Google Scholar
236. Finch, SA, Doyle, W, Lowe, C, et al. (1998) National Diet and Nutrition Survey: People Aged 65 Years and Over. Volume 1: Report of the Diet and Nutrition Survey. London: The Stationery Office.Google Scholar
237. Herbert, V (1962) Experimental nutritional folate deficiency. Trans Assoc Am Physicians 75, 307320.Google Scholar
238. Gailani, SD, Carey, RW, Holland, JF, et al. (1970) Studies of folate deficiency in patients with neoplastic diseases. Cancer Res 30, 327333.Google Scholar
239. Hoppner, K & Lampi, B (1980) Folate levels in human liver from autopsies in Canada. Am J Clin Nutr 33, 862864.Google Scholar
240. Cooper, BA (1978) Reassessment of folic acid requirements. In Nutrition in Transition: Proceedings of the Western Hemisphere Congress, no. 5, pp. 281–288 [P White and N Selvey, editors]. Monroe, WI: American Medical Association.Google Scholar
241. Gregory, J, Foster, K & Tyler, H (1990) The Dietary and Nutritional Survey of British Adults. London: HM Stationery Office.Google Scholar
242. Ministry of Agriculture, Fisheries and Food (1994) The Dietary Survey of British Adults: Further Analysis. London: HM Stationery Office.Google Scholar
243. Kauwell, GPA, Lippert, BL, Wilsky, CE, et al. (2000) Folate status of elderly women following moderate folate depletion responds only to a higher folate intake. J Nutr 130, 15841590.Google Scholar
244. Rampersaud, GC, Kauwell, GP, Hutson, AD, et al. (2000) Genomic DNA methylation decreases in response to moderate folate depletion in elderly women. Am J Clin Nutr 72, 9981003.Google Scholar
245. Selhub, J, Jacques, PF, Wilson, PW, et al. (1993) Vitamin status and intake as primary determinants of homocysteinemia in an elderly population. JAMA 270, 26932698.Google Scholar
246. Baggott, JE, Morgan, SL, Ha, T, et al. (1992) Inhibition of folate-dependent enzymes by non-steroidal anti-inflammatory drugs. Biochem J 282, 197202.Google Scholar
247. Roe, DA (1989) Diet and Drug Interactions, 1st ed., pp. 83105. New York: Van Nostrand Reinhold.Google Scholar
248. Corominas-Faja, B, Quirantes-Piné, R, Oliveras-Ferraros, C, et al. (2012) Metabolomic fingerprint reveals that metformin impairs one-carbon metabolism in a manner similar to the antifolate class of chemotherapy drugs. Aging 4, 480498.Google Scholar
249. Neu, HC & Gootz, TD (1996) Antimicrobial chemotherapy. In Medical Microbiology, 4th ed., chapter 11 [S Baron, editor]. Galveston, TX: University of Texas Medical Branch at Galveston.Google Scholar
250. Leonard, JP, Desager, JP, Beckers, C, et al. (1979) In vitro binding of various biological substances by two hypocholesterolaemic resins. Cholestyramine and colestipol. Arzneimittelforschung 29, 979981.Google Scholar
251. Corcino, J, Waxman, S & Herbert, V (1970) Mechanism of triamterene-induced megaloblastosis. Ann Intern Med 73, 419424.Google Scholar
252. LeBlanc, JG, Milani, C, de Giori, GS, et al. (2013) Bacteria as vitamin suppliers to their host: a gut microbiota perspective. Curr Opin Biotechnol 24, 160168.Google Scholar
253. Brody, T (1998) Nutritional Biochemistry, 2nd ed., pp. 516524. San Diego, CA: Academic Press.Google Scholar
254. Goulding, CW, Postigo, D & Matthews, RG (1997) Cobalamin-dependent methionine synthase is a modular protein with distinct regions for binding homocysteine, methyltetrahydrofolate, cobalamin, and adenosylmethionine. Biochemistry 36, 80828091.Google Scholar
255. Cooper, BA & Rosenblatt, DS (1987) Inherited defects of vitamin B12 metabolism. Annu Rev Nutr 7, 291320.Google Scholar
256. Neuberger, A (1961) Aspects of the metabolism of glycine and of porphyrins. Biochem J 78, 110.Google Scholar
257. Toh, BH, van Driel, IR & Gleeson, PA (1997) Pernicious anemia. N Engl J Med 337, 14411448.Google Scholar
258. Silva, MT, Cavalcanti, JL & Moreira, DM (2000) Neuroradiological features of the brain in subacute combined spinal cord degeneration: case report [Article in Portuguese]. Arq Neuropsiquiatr 58, 752755.Google Scholar
259. Adams, RD & Kubik, CS (1944) Subacute degeneration of the brain in pernicious anemia. N Engl J Med 231, 19.Google Scholar
260. Pant, SS, Asbury, AK & Richardson, EP (1968) The myelopathy of pernicious anemia. A neuropathological reappraisal. Acta Neurol Scand 44, Suppl. 5, 136.Google Scholar
261. Greenfield, JG & Carmichael, EA (1935) The peripheral nerves in cases of subacute combined degeneration of the cord. Brain 58, 483491.Google Scholar
262. McCombe, PA & McLeod, JG (1984) The peripheral neuropathy of vitamin B12 deficiency. J Neurol Sci 66, 117126.Google Scholar
263. Chand, G & Maller, V (2008) Subacute combined degeneration of the spinal cord. Internet J Radiol 10, 1.Google Scholar
264. Healton, EB, Savage, DG, Brust, JC, et al. (1991) Neurologic aspects of cobalamin deficiency. Medicine 70, 229245.Google Scholar
265. Ralapanawa, DMPUK, Jayawickreme, KP, Ekanayake, EMM, et al. (2015) B12 deficiency with neurological manifestations in the absence of anaemia. BMC Res Notes 8, 458.Google Scholar
266. Hin, H, Clarke, R, Sherliker, P, et al. (2006) Clinical relevance of low serum vitamin B12 concentrations in older people: the Banbury B12 Study. Age Ageing 35, 416422.Google Scholar
267. Savage, DG & Lindenbaum, J (1995) Neurological complications of acquired cobalamin deficiency: clinical aspects. Baillieres Clin Haematol 8, 657678.Google Scholar
268. Leishear, K, Boudreau, RM, Studenski, SA, et al. (2012) Relationship between vitamin B12 and sensory and motor peripheral nerve function in older adults. J Am Geriatr Soc 60, 10571063.Google Scholar
269. Matteini, AM, Walston, JD, Fallin, MD, et al. (2008) Markers of B-vitamin deficiency and frailty in older women. J Nutr Health Aging 12, 303308.Google Scholar
270. Oberlin, BS, Tangney, CC, Gustashaw, KA, et al. (2013) Vitamin B12 deficiency in relation to functional disabilities. Nutrients 5, 44624475.Google Scholar
271. Lindenbaum, J, Healton, EB, Savage, DG, et al. (1988) Neuropsychiatric disorders caused by cobalamin deficiency in the absence of anemia or macrocytosis. N Engl J Med 318, 17201728.Google Scholar
272. Carmel, R (1988) Pernicious anemia. The expected findings of very low serum cobalamin levels, anemia, and macrocytosis are often lacking. Arch Intern Med 148, 17121714.Google Scholar
273. Swart, KMA, Ham, AC, van Wijngaarden, JP, et al. (2005) A randomized controlled trial to examine the effect of 2-year vitamin B12 and folic acid supplementation on physical performance, strength, and falling: additional findings from the B-PROOF Study. Calcif Tissue Int 98, 1827.Google Scholar
274. Kortebein, P, Ferrando, A, Lombeida, J, et al. (2007) Effect of 10 days of bed rest on skeletal muscle in healthy older adults. JAMA 297, 17721774.Google Scholar
275. Bell, IR, Edman, JS, Miller, J, et al. (1990) Relationship of normal serum vitamin B12 and folate levels to cognitive test performance in subtypes of geriatric major depression. J Geriatr Psychiatry Neurol 3, 98105.Google Scholar
276. Tangney, CC, Aggarwal, NT, Li, H, et al. (2011) Vitamin B12, cognition, and brain MRI measures: a cross-sectional examination. Neurology 77, 12761282.Google Scholar
277. Kim, NH, Kim, HS, Eun, CR, et al. (2011) Depression is associated with sarcopenia, not central obesity, in elderly Korean men. J Am Geriatr Soc 59, 20622068.Google Scholar
278. Hsu, Y-H, Liang, C-K, Chou, M-Y, et al. (2014) Association of cognitive impairment, depressive symptoms and sarcopenia among healthy older men in the veterans retirement community in southern Taiwan: a cross-sectional study. Geriatr Gerontol Int 14, 102108.Google Scholar
279. Verlaan, S, Aspray, TJ, Bauer, JM, et al. (2015) Nutritional status, body composition, and quality of life in community-dwelling sarcopenic and non-sarcopenic older adults: a case–control study. Clin Nutr 36, 267274.Google Scholar
280. Bor, MV, Lydeking-Olsen, E, Møller, J, et al. (2006) A daily intake of approximately 6 microg vitamin B-12 appears to saturate all the vitamin B-12-related variables in Danish postmenopausal women. Am J Clin Nutr 83, 5258.Google Scholar
281. King, CE, Leibach, J & Toskes, PP (1979) Clinically significant vitamin B12 deficiency secondary to malabsorption of protein-bound vitamin B12 . Dig Dis Sci 24, 397402.Google Scholar
282. Suter, PM, Golner, BB, Goldin, BR, et al. (1991) Reversal of protein-bound vitamin B12 malabsorption with antibiotics in atrophic gastritis. Gastroenterology 101, 10391045.Google Scholar
283. Krasinski, SD, Russell, RM, Samloff, IM, et al. (1986) Fundic atrophic gastritis in an elderly population. Effect on hemoglobin and several serum nutritional indicators. J Am Geriatr Soc 34, 800806.Google Scholar
284. van Oijen, MGH, Sipponen, P, Laheij, RJF, et al. (2007) Gastric status and vitamin B12 levels in cardiovascular patients. Dig Dis Sci 52, 21862189.Google Scholar
285. Kong, Y-J, Yi, H-G, Dai, J-C, et al. (2014) Histological changes of gastric mucosa after Helicobacter pylori eradication: a systematic review and meta-analysis. World J Gastroenterol 20, 59035911.Google Scholar
286. Lam, JR, Schneider, JL, Zhao, W, et al. (2013) Proton pump inhibitor and histamine 2 receptor antagonist use and vitamin B12 deficiency. JAMA 310, 24352442.Google Scholar
287. Wham, C, Teh, R, Moyes, SA, et al. (2016) Micronutrient intake in advanced age: Te Puāwaitanga o Ngā Tapuwae Kia ora Tonu, Life and Living in Advanced Age: A Cohort Study in New Zealand (LiLACS NZ). Br J Nutr 116, 17541769.Google Scholar
288. Bianchetti, A, Rozzini, R, Carabellese, C, et al. (1990) Nutritional intake, socioeconomic conditions, and health status in a large elderly population. J Am Geriatr Soc 38, 521526.Google Scholar
289. Penninx, BW, Guralnik, JM, Ferrucci, L, et al. (2000) Vitamin B12 deficiency and depression in physically disabled older women: epidemiologic evidence from the Women’s Health and Aging Study. Am J Psychiatry 157, 715721.Google Scholar
290. Pennypacker, LC, Allen, RH, Kelly, JP, et al. (1992) High prevalence of cobalamin deficiency in elderly outpatients. J Am Geriatr Soc 40, 11971204.Google Scholar
291. Elsborg, L, Lung, V & Bastrup-Madsen, P (1976) Serum vitamin B12 levels in the aged. Acta Med Scand 200, 309314.Google Scholar
292. Gonzalez-Gross, M, Sola, R, Albers, U, et al. (2007) B-vitamins and homocysteine in Spanish institutionalized elderly. Int J Vitam Nutr Res 77, 2233.Google Scholar
293. Morris, MS, Jacques, PF, Rosenberg, IH, et al. (2002) Elevated serum methylmalonic acid concentrations are common among elderly Americans. J Nutr 132, 27992803.Google Scholar
294. Heil, SG, de Jonge, R, de Rotte, MCFJ, et al. (2012) Screening for metabolic vitamin B12 deficiency by holotranscobalamin in patients suspected of vitamin B12 deficiency: a multicentre study. Ann Clin Biochem 49, 184189.Google Scholar
295. Obeid, R & Herrmann, W (2007) Holotranscobalamin in laboratory diagnosis of cobalamin deficiency compared to total cobalamin and methylmalonic acid. Clin Chem Lab Med 45, 17461750.Google Scholar
296. Remacha, AF, Sardà, MP, Canals, C, et al. (2014) Role of serum holotranscobalamin (holoTC) in the diagnosis of patients with low serum cobalamin. Comparison with methylmalonic acid and homocysteine. Ann Hematol 93, 565569.Google Scholar
297. Valente, E, Scott, JM, Ueland, P-M, et al. (2011) Diagnostic accuracy of holotranscobalamin, methylmalonic acid, serum cobalamin, and other indicators of tissue vitamin B12 status in the elderly. Clin Chem 57, 856863.Google Scholar
298. Reizenstein, P, Ek, G & Matthews, CME (1966) Vitamin B12 kinetics in man. Implications on total-body-B12-determinations, human requirements, and normal and pathological cellular B12 uptake. Phys Med Biol 11, 295306.Google Scholar
299. Grasbeck, R, Nyberg, W & Reizenstein, P (1958) Biliary and fecal vit. B12 excretion in man. an isotope study. Exp Biol Med 97, 780784.Google Scholar
300. Die Deutsche Gesellschaft für Ernährung e. V. (DGE) (2015) Vitamin B12 (cobalamine) [in German]. https://www.dge.de/wissenschaft/referenzwerte/vitamin-b12/ (accessed July 2016).Google Scholar
301. European Food Safety Authority (EFSA) Panel on Dietetic Products, Nutrition and Allergies (2015) Scientific opinion on dietary reference values for cobalamin (vitamin B12). EFSA J 13, 4150.Google Scholar
302. Bor, MV, von Castel-Roberts, KM, Kauwell, GP, et al. (2010) Daily intake of 4 to 7 g dietary vitamin B-12 is associated with steady concentrations of vitamin B-12-related biomarkers in a healthy young population. Am J Clin Nutr 91, 571577.Google Scholar
303. Gross, JS, Weintraub, NT, Neufeld, RR, et al. (1986) Pernicious anemia in the demented patient without anemia or macrocytosis. A case for early recognition. J Am Geriatr Soc 34, 612614.Google Scholar
304. Carmel, R, Karnaze, DS & Weiner, JM (1988) Neurologic abnormalities in cobalamin deficiency are associated with higher cobalamin “analogue” values than are hematologic abnormalities. J Lab Clin Med 111, 5762.Google Scholar