Hostname: page-component-8448b6f56d-jr42d Total loading time: 0 Render date: 2024-04-23T21:38:53.148Z Has data issue: false hasContentIssue false

Nutrients for neurocognition in health and disease: measures, methodologies and mechanisms

Published online by Cambridge University Press:  11 December 2017

Andrew Scholey*
Affiliation:
Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
*
Correspondence author: Andrew Scholey, email andrew@scholeylab.com
Rights & Permissions [Opens in a new window]

Abstract

Effective pharmaceutical treatments for age-related cognitive decline have proved elusive. There is, however, compelling evidence that nutritional status and supplementation could play crucial roles in modifying the expression of cognitive change through the lifespan. Subjective memory impairment and mild cognitive impairment can be harbingers of dementia but this is by no means inevitable. Neurocognitive change is influenced by a variety of processes, many of which are involved in other aspects of systemic health, including cardiovascular function. Importantly, many of these processes are governed by mechanisms which may be modified by specific classes of bioactive nutrients. There is increasing, converging evidence from controlled trials that nutritional interventions can improve mood and cognitive function in both clinical and healthy populations. Specific examples include selected botanical extracts such as the flavonoids. Some nutritional supplements (e.g. broad-spectrum micronutrient supplementation) appear to support improved cognitive function, possibly through redressing insufficient nutrient status (i.e. suboptimal but above the threshold for frank deficiency). Recent mechanistic research has unveiled physiologically plausible, modifiable, cognition-relevant targets for nutrition and nutraceuticals. These include processes involved in both systemic and central vascular function, inflammation, metabolism, central activation, improved neural efficiency and angiogenesis. The advent and development of human neuroimaging methodology have greatly aided our understanding of the core central mechanisms of cognitive change. Different imaging modalities can provide insights into modifiable central mechanisms which may be targeted by bioactive nutrients. The latter may contribute to slowing age-related decline through supporting neurocognitive scaffolding mechanisms.

Type
Conference on ‘Diet, nutrition and mental health and wellbeing’
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author 2017

The dramatic increase in life expectancy during the twentieth century surely ranks as one of the greatest achievements of medical science. Most babies born in 1900 did not live past the age of 50, today those born in Westernised countries can expect to live into their 70s and 80s and beyond( Reference Suzman and Beard 1 ), a trend which is likely to continue at least for the next decade or so( Reference Kontis, Bennett and Mathers 2 ). Nevertheless, population-level, increased longevity brings with it unexpected challenges in the form of age-related diseases. As well as placing a heavy economic burden on healthcare systems, these engender serious human costs. Amongst such disorders, perhaps the most pernicious is dementia which shares many mechanisms with less severe conditions such as subjective memory impairment (SMI; more recently termed subjective cognitive decline), mild cognitive impairment (MCI) and non-clinical age-related cognitive decline.

The World Health Organisation estimates that there is a new case of dementia every 4 s( Reference Prince, Guerchet and Prina 3 ), a figure which illustrates the urgency of the global health crisis associated with cognitive decline. Such decline, however, is far from inevitable. While there is a genetic influence in dementia (a three-generation family history roughly doubles the lifetime probability of a dementia diagnosis), it is estimated that <2 % of Alzheimer's disease (AD) cases are due to single-gene mutations with approximately 13 % of cases having a clear autosomal-dominant pattern of inheritance( Reference Bekris, Yu and Bird 4 ). Higher estimates of genetic influences on non-pathological age-related cognitive decline suggest that approximately 50 % of the variance may be explained by genetic factors( Reference Deary, Yang and Davies 5 ), with the remainder assumed to be attributable to environmental and lifestyle influences. This has driven the search for achievable ways to modify the expression of age-related neurocognitive decline and progression to AD. There is now increasing evidence that candidate modifying factors include exercise( Reference Kontis, Bennett and Mathers 2 ) and, importantly for this review, nutritional processes. There is also growing evidence that many of these modifiable influences on dementia risk also modify non-clinical cognitive decline and may be set in motion many years before the manifestation of impaired cognitive function.

The most common form of dementia is AD, accounting for some 60–80 % of dementia cases( 6 ). Distinguishing pathological from non-pathological cognitive ageing is challenging, partly because many of the markers for AD are also observed to some degree in normal ageing, as well in other cognitive phenotypes, such as SMI and MCI. Individuals with SMI perceive their memory to be declining but perform as well as their peers on objective cognitive tests. Nevertheless, they have reduced grey matter volume in the temporal lobe( Reference Jessen, Feyen and Freymann 7 ), which houses structures important for memory processing, and increased levels of brain activation during cognitive tasks( Reference Rodda, Dannhauser and Cutinha 8 ). This suggests that there may be central compensatory mechanisms during early phases of cognitive decline, with greater activation required to maintain previous levels of cognitive performance. Compared with SMI, MCI is more severe, associated with significantly reduced cognitive functions but, unlike AD, the ability to conduct activities of daily living is spared. Both SMI and MCI are associated with increased risk of future dementia( Reference Jessen, Wolfsgruber and Wiese 9 ) with 30–40 % of those with MCI converting to AD over a given 5-year period( Reference Ward, Tardiff and Dye 10 ). SMI also increases the risk of later dementia, especially in those who go on to be classified as having MCI( Reference Jessen, Wolfsgruber and Wiese 9 ), leading to the suggestion of a three-stage model. This risk is particularly high when SMI co-presents with worry( Reference Jessen, Wolfsgruber and Wiese 9 ), raising the intriguing possibility of experiential concern about declining memory being linked to a more severe organic, prodromal state. Conversely, while both conditions can be harbingers of dementia, some individuals stabilise and do not progress to more severe stages. This suggests that MCI and, especially, SMI may be viable targets for preventative interventions including from nutritional interventions.

The first-line pharmacotherapy for AD is dominated by the cholinesterase inhibitor (ChEI) family of drugs, introduced in 1997. Two decades later, there is a general consensus that the ChEI have not realised their initial promise. The drugs can treat the symptomatology of AD, but the impact on the disease itself has been modest. As the name suggests, their common mode of action is to inhibit the enzyme cholinesterase, resulting in elevated levels of the neurotransmitter acetylcholine, which is important for memory and attentional functions and is depleted in the AD brain. ChEI offer moderate respite at best and are effective only at earlier stages of the disorder. They have a relatively small therapeutic window due to low tolerance in many patients( Reference Deardorff, Feen and Grossberg 11 ), with a 30 % drop out rate in clinical trials. Interestingly there are certain herbs with ChEI properties which appear to be better tolerated than their pharmaceutical counterparts, including species of Salvia (sage). Extracts of Salvia have been shown to acutely improve memory and attention in healthy young and older populations( Reference Kennedy, Dodd and Robertson 12 Reference Tildesley, Kennedy and Perry 15 ), with two small trials in cognitively impaired populations also showing promise( Reference Miroddi, Navarra and Quattropani 16 ).

The relative contribution of the ChEI properties of Salvia to these effects is not known, as the herb affects many cognition-relevant processes( Reference Miroddi, Navarra and Quattropani 16 ). Indeed, one reason for the relative lack of success of pharmaceutical ChEI in the treatment of AD is their restricted mode of action. Like other degenerative diseases, AD can be considered to be product of a pathological cascade, in this case involving progressively accelerating, reciprocal, neurotoxic interactions between oxidative stress, inflammatory responses, compromised cerebral metabolism, neurofibrillary tangle generation, β-amyloid deposition, amongst other processes which include damage to the cholinergic system( Reference Jack, Knopman and Jagust 17 ). The relative contribution of each of these is unknown and likely differs in idiosyncratic ways from person to person. Given the multifaceted nature of AD, unsurprisingly ‘magic bullet’ approaches to halting cognitive decline have been relatively unsuccessful. In part this is because they do not match well with our present understanding of central nervous system function, where our growth in knowledge is characterised by increased complexity; it seems unlikely that there will be a ‘double helix’ of brain function. The lack of success of central nervous system pharmaceutical pipelines in the development of treatments for brain senescence has led to investigations into the modifiable processes which underlie neurocognitive decline. These include (but are not restricted to) cognitive activity and education, exercise and nutritional factors. Some examples of the latter will be outlined in following sections.

Factors influencing age-related cognitive decline

There is increasing evidence that many of the risk factors for cognitive decline may be detectable in mid-life, with several of these overlapping with those for CVD( Reference Qiu and Fratiglioni 18 ). Such processes include poorer glycaemic control, compromised vascular function, oxidative stress, inflammation, perturbations of the hypothalamic–pituitary–adrenocortical axis and microbiotic dysbiosis (Fig. 1).

Fig. 1. Influences on neurocognitive ageing including therapeutic targets for delaying age-related neurocognitive decline. Pharmaceutical approaches are largely restricted to effects on neurotransmitter systems (grey panel). Processes within the green-shaded area represent realistic nutritional targets. Age and genetics (unshaded area) remain non-modifiable factors. HPA, hypothalamic–pituitary–adrenal axis.

The interactions between these factors and cognitive decline may be reciprocal or even reflect reverse causality. Childhood cognitive ability is a strong predictor of both age-related cognitive decline and many later life health indices( Reference Jessen, Wolfsgruber and Wiese 9 ) suggesting that both may be manifestations of some core mechanism(s). Irrespective of the underlying influences, the cluster of processes depicted in Fig. 1 is known to be modifiable by dietary interventions. Indeed, it has been suggested that, despite a projected 3-fold increase in dementia prevalence by 2050, the age-specific risk of dementia may be decreasing in some Western countries due in part to ‘[rising] levels of education and more widespread and successful treatment of key cardiovascular risk factors’( Reference Langa 19 ). Such treatment includes better nutrition advice, again supporting the concept of dietary interventions positively modifying the trajectory of cognitive decline in senescence. The following sections present a selective review of nutritional/dietary interventions, which have potential to modify cognitive function.

Various epidemiological studies have shown an association between certain dietary (patterns and specific bioactive nutrients) and protection against cognitive decline. These include the Mediterranean dietary pattern( Reference Hardman, Kennedy and Macpherson 20 ) and specific dietary components such as the flavonoids( Reference Nurk, Refsum and Drevon 21 ), a class of approximately 5000 compounds believed to contribute to the health benefits of fruit and vegetable intake. Several epidemiological studies have identified an association between flavonoid intake and better cognitive function( Reference Nurk, Refsum and Drevon 21 Reference Camfield, Stough and Farrimond 24 ). Although useful, such studies may have missed ‘third-factor’ influences on both cognition and diet which, sometimes by definition, are presently unknown. These include genetic polymorphisms and microbiome differences. In the latter case, there is presently intense research focus on the so-called gut–brain axis which has become something of a new frontier for brain research in health and disease. Increasing evidence points to bidirectional cross-signalling between the gut microbiota and brain including via microbiotic metabolites, the immune system and the vagus nerve( Reference Sandhu, Sherwin and Schellekens 25 ). The intestinal microbiome is a rich source of signalling molecules and can be rapidly modified by diet( Reference Albenberg and Wu 26 ) raising the possibility of another nutritional target which influences brain function (e.g. using pre- or probiotics). Despite huge potential and compelling evidence from animal studies, results from early controlled human trials have been mixed. It is unclear to what extent probiotic supplementation achieves the primary goal of altering the microbiota composition( Reference Kristensen, Bryrup and Allin 27 ). Interventions specifically aimed at redressing microbiomic dysbiosis enhanced cognition in a cohort with dementia( Reference Akbari, Asemi and Kakhaki 28 ), but did not improve stress or cognitive function in healthy volunteers( Reference Kelly, Allen and Temko 29 ). A recent systematic review reported positive effects on anxiety and depression in five out of ten included studies( Reference Pirbaglou, Katz and de Souza 30 ). Some of these mixed results may be due to methodological issues. Better understanding of optimal treatment characteristics, including duration and type of intervention will doubtless emerge in the coming years and may herald an era of ‘psychobiotics’.

Trajectories of age-related cognitive change

Many of the same processes involved in cognitive pathology are integral mechanisms in non-clinical ageing and clearly contribute to population level, age-associated cognitive decline. There is however enormous variation in the trajectory of cognitive decline both across individuals and cognitive domains( Reference Wilson, Beckett and Barnes 31 ). Broadly, age-related decline is observed in ‘fluid’ cognitive abilities including memory (especially working memory), processing speed and executive functioning. Conversely, so-called ‘crystallised’ abilities, e.g. general knowledge and some verbal skills, remain relatively unscathed by the ageing process( Reference Ward, Tardiff and Dye 10 , Reference Deary, Corley and Gow 32 ). It should be noted that, even though domains such as working memory and processing speed decline with age, there is large individual variability, with some people maintaining high functioning well into old age( Reference Wilson, Beckett and Barnes 31 ). Again, while some of this variance can be explained by genetic factors, there remains a great deal which is influenced by environmental and lifestyle processes including nutrition.

As with cognitive abilities, there are substantial individual differences in age-related changes to the neural substrates which underpin cognitive performance. Longitudinal studies are rare but indicate an adult global cerebral atrophy rate of approximately 0·8 % per year( Reference Crivello, Tzourio-Mazoyer and Tzourio 33 ). Nevertheless some structures such as the visual and entorhinal cortices are relatively spared, while others such as the cerebellum and hippocampus show a more striking decline( Reference Park and Reuter-Lorenz 34 ), again with marked variability across individuals( Reference Callaghan, Freund and Draganski 35 ). The hippocampus is particularly important in the context of modifiable structure–function relationships. It is critically involved in binding together disparate elements of experience (time, location, environment, actions, specific stimuli, etc.) into specific memories( Reference Konkel and Cohen 36 ). Hippocampal size is reduced in a cluster of diet-modifiable disorders, including diabetes, obesity and hypertension( Reference Monti, Baym and Cohen 37 ). It is one of two mammalian brain structures which have been identified as having the capacity for adult neurogenesis (the birth of new neurons). Crucially there is increasing evidence that lifestyle factors such as exercise can increase human hippocampal volume( Reference Erickson, Voss and Prakash 38 ). In animals, this increased volume is accompanied by elevated rates of neurogenesis following exercise( Reference Pereira, Huddleston and Brickman 39 ) and by nutritional interventions such as diets rich in flavanols( Reference Van Praag, Lucero and Yeo 40 ) or n-3 fatty acids( Reference Luchtman and Song 41 ). This again supports the notion that there are plastic neural substrates of cognitive performance, which are viable targets for nutritional interventions in human subjects.

Neuroimaging and nutritional interventions

Unlike pharmaceuticals, the effects of dietary interventions can be subtle (although standardised extracts of the herbs Ginseng and Bacopa improve cognitive performance with effect sizes comparable with those of the US Food and Drug Administration-approved, cognitive enhancer du jour, Modafinil( Reference Neale, Camfield and Reay 42 ). The advent of computerised cognitive tests has helped to advance the field. Compared with paper-and-pencil psychometric assessment, these are capable of exquisite sensitivity in detecting slight changes in accuracy as well as resolving changes in response times at the millisecond level. This is particularly important when considering that speed of processing declines with ageing.

It is beyond the scope of the present paper to cover all randomised controlled trials in this area; the following sections, therefore, give a flavour of the results of such studies, focusing on those which have included the application of neuroimaging methodology.

Using neuroimaging to detect structural/functional changes associated with dietary interventions can be important for a number of reasons. If nothing else such studies can confirm that the treatment is centrally active, that is, that some component of the ingested substance is bioavailable and, directly or indirectly, changes the pattern of activation during cognitive processing.

Neuroimaging can also provide important insights into the mechanism of action of nutritional interventions. There are various neuroimaging modalities, each with its own advantages and disadvantages. Structural imaging, often performed using MRI can discern the size of neuroanatomical regions, or specifically grey matter (reflecting most structures other than the myelin sheath). The method is used to quantify changes in whole brain volume or specific neuroanatomical loci such as the hippocampus and its subfields. Other sophisticated measures have been developed, such as diffusion tensor imaging which can visualise white matter (essentially myelin), an important constituent of the tracts which convey electrochemical traffic between brain structures. Age-related declines in white matter integrity have been shown to mediate perceptual slowing in ageing( Reference Bucur, Madden and Spaniol 43 ). White matter integrity is related to vitamin D status( Reference Karakis, Pase and Beiser 44 ) and can be improved by nutritional change including n-3 fatty acid supplementation( Reference Janssen and Kiliaan 45 , Reference Witte, Kerti and Hermannstädter 46 ), suggesting it may be a viable target for nutrient interventions.

As well as structural changes, neuroimaging can be used to detect changes in functional activation using several methods. Each has advantages and can be viewed in terms of its spatial and temporal resolution, that is, how well the method can measure where and when, respectively, neural events occur.

Electroencephalography (EEG) was developed in the first half of the twentieth century and successfully detects changes in electrical activity associated with neuronal firing. It has a temporal resolution at the millisecond scale (reflecting the timescale of activity underpinning actual cognitive events). It has, however, poor spatial resolution since the signal is smeared by the skull. Continuous EEG recordings are typically divided into traditional frequency bands of δ (<4 Hz), θ (4–7 Hz), α (8–15 Hz), β (15–30 Hz) and γ (30+ Hz) with lower frequencies dominating during a relaxation, transitioning to higher frequencies during cognitive processing.

Applying EEG methodology to nutrient interventions have revealed effects of several nutritional interventions( Reference Camfield and Scholey 47 ). Acute administration of the green tea polyphenol epigallocatechin gallate reduced subjective stress and increased α, β and θ wave activity in healthy young volunteers( Reference Scholey, Downey and Ciorciari 48 ). There is evidence of relaxant properties of other tea components from human EEG activity( Reference Kobayashi, Nagato and Aoi 49 , Reference Juneja, Chu and Okubo 50 ). Two hundred milligrams of the green tea amino acid l-theanine, but not 50 mg, led to increased α-wave activity in the occipital and parietal regions of the brain within 40 min of ingestion when administered to resting participants. A different study found evidence of a decrease in α-wave activity following 250 mg l-theanine when measured during performance of an attention task( Reference Gomez-Ramirez, Higgins and Rycroft 51 ). While these findings may appear to be contradictory, it may also be indicative of differing EEG effects of l-theanine when administered during attentional processing (requiring focus) as opposed to at rest. In particular, these data can be explained by the fact that, as well as the classic relaxation-associated ‘tonic’ α, there is also a phasic α wave. This is believed to be associated with brain areas which are inhibited as, if they were active they would render this type of processing less efficient; a so-called distracter suppression mechanism. This interpretation was confirmed in a follow-up study where theanine again facilitated the EEG component of attentional switching, this time when the cue signalled a stimulus to the left or right visual field rather than between visual and auditory modalities( Reference Gomez-Ramirez, Kelly and Montesi 52 ).

More recently, neuroimaging using magnetoencephalography (MEG) has been developed. Rather than changes in electrical activity, MEG detects fluctuations in the magnetic field of neuronal populations. As the signal is relatively unchanged by bone, compared with EEG the spatial resolution from MEG is also very high. The brain's magnetic field is approximately 50 femtotesla (i.e. 10−9 that of the earth's magnetic field) making MEG methodologically challenging. The technique requires shielded housing and arrays of recording magnetometors (superconducting quantum interference devices) which are bathed in liquid helium.

Pharmaco-MEG is a developing discipline and MEG studies into central effects of nutritional interventions are lacking. One exception is an application of MEG to the study of the acute anti-stress effects of a drink containing 200 mg l-theanine( Reference White, de Klerk and Woods 53 ). Compared with a placebo, multitasking-evoked workload stress was reduced in the theanine condition 1 h post-drink only, with cortisol following a different pattern, being reduced at 3 h only. Additionally, MEG revealed that resting state α oscillatory activity was significantly greater in posterior sensors after active treatment 2 h post-dose. This effect was only apparent for those higher in trait anxiety, emphasising the need for control and/or measurement of initial situational and dispositional factors in this type of study. The change in resting state α oscillatory activity was not correlated with the change in subjective anxiety, stress or cortisol response, suggesting further research is required to assess the functional relevance of these treatment-related changes in resting α activity( Reference White, de Klerk and Woods 53 ).

Many neuroimaging studies utilise functional MRI (fMRI) which reflects neuronal activation by measuring the amount and ratio between oxygenated and deoxygenated haemoglobin (the blood oxygen level dependent or BOLD signal) in target regions of interest. fMRI was developed over 25 years ago( Reference Belliveau, Kennedy and McKinstry 54 ) and has undergone significant advances in the past decades. While the spatial resolution of fMRI is relatively good, to the level of a cubic millimetre or so (representing perhaps tens of thousands of neurons), the method has poor temporal resolution compared with EEG and MEG. Activation is typically measured over a second or two and averaged over many trials. Many cognitive tasks involve perceptual and motor responses (e.g. attending to stimuli and pressing a button) which themselves produce region-specific central activation. Therefore, fMRI methodologies attempt to isolate the signal associated with cognitive processing only by subtracting the activation associated with a control task, which includes similar perceptual and motor elements but not the cognitive process (see Fig. 2).

Fig. 2. Rapid visual information processing (RVIP) as example functional MRI task used to capture effects of nutritional intervention studies. Panel A shows the active task requiring the subject to respond to three consecutive odd or three consecutive even digits when single digits are presented every 600 ms. In the control task (B), matched for perceptual and motor activity, subjects see a similar stimulus stream but respond to the presentation of the ‘0’ digit only (i.e. there is minimal cognitive load). When activation measured during B is subtracted from that during A, there is consistent activation in a well-characterised working memory circuit consisting of more anterior (ant.) frontal and supplementary motor areas and more posterior (post.) parietal and cerebellar regions linked with attention and working memory. C and D illustrate this pattern of activation from ( Reference White, Cox and Hughes 77 , Reference Neale, Johnston and Hughes 90 ) with warmer colours depicting greater activation. Increased activation within this circuit has been found following multivitamin mineral supplementation( Reference White, Cox and Hughes 77 ), see Fig. 3.

The methodology is developing increasing sophistication and there are several well-controlled fMRI studies that have shown changes in neural activity associated with nutritional interventions. These include multivitamins, caffeine, n-3 fatty acids glucose and flavanols (including from tea and cocoa). Each is dealt with in detail in( Reference Kobayashi, Nagato and Aoi 49 ) and covered in the following sections.

Cocoa flavanols (CF) have a number of potential health benefits including in the neurocognitive domain( Reference Scholey and Owen 55 ), where the most effective dose appears to be approximately 400–500 mg CF. Acute administration enhances mental function, particularly during mentally effortful processing( Reference Massee, Ried and Pase 56 , Reference Scholey, French and Morris 57 ). The differential effects according to mental effort suggest that increased cognitive load is itself associated with greater central activation, in the form of recruitment of more neural tissue. Neurovascular coupling dictates that this will signal greater localised blood flow and oxygen utilisation as reflected by a higher BOLD signal.

In addition to localising patterns of activation associated with specific cognitive tasks, MRI can be used to measure other aspects of brain function. Arterial spin labelling allows quantification of cerebral blood flow or perfusion. Two studies have reported increased cerebral blood flow 2 h following CF administration; the first examining whole brain cerebral blood flow( Reference Francis, Head and Morris 58 ), with a more recent study resolving specific regions with greater perfusion( Reference Lamport, Pal and Moutsiana 59 ). The latter reported increased activation in areas including the anterior cingulate cortex and left parietal lobe. The former is associated with numerous cognitive processes including goal-directed behaviour and attentional processes, while the latter is activated during working memory tasks.

CF consistently improve aspects of vascular functioning including those which are directly related to cognition-relevant neural processes. The dentate gyrus subfield of the hippocampus differentially atrophies during ageing. It is particularly important for specific aspects of memory including pattern separation, which reflects the ability to correctly discriminate similar overlapping sets of stimuli as specific memories. Twelve-week CF supplementation resulted in increased dentate gyrus cerebral blood volume which correlated with scores on a pattern recognition test( Reference Brickman, Khan and Provenzano 60 ). The dentate gyrus is one of the few areas of the adult brain capable of neurogenesis (the birth of new neurons) and in rodents cerebral blood volume is a marker of neurogenesis( Reference Pereira, Huddleston and Brickman 39 ). This finding could have significant implications, if CF (or other flavonoid) supplementation can promote neurogenesis, there are obvious applications in delaying cognitive decline. Conversely, it should also be noted that this was a small trial (n 37, parallel groups), half the cohort underwent an exercise regimen and there was a relatively high dropout rate. A more recent trial has confirmed that both 520 and 993 mg CF can improve cognitive functions, including working memory in older individuals as well as some of the processes depicted in Fig. 1, including indices of glucose utilisation( Reference Mastroiacovo, Kwik-Uribe and Grassi 61 ).

As well as detecting regional activation during task performance, fMRI can be used to examine structural and functional connections between brain regions (the latter is the focus of the human connectome project). The human brain is disproportionately metabolically active, typically contributing to 2 % of body weight but requiring some 20 % of metabolic resources in the form of oxygen and glucose. Unlike other tissue, the brain stores negligible amounts of glucose. It follows that neurocognitive function is exquisitely sensitive to fluctuations in glucose availability, with poor glucose control observed in ageing and dementia (to the extent that AD has been termed type 3 diabetes( Reference Kandimalla, Thirumala and Reddy 62 )). Cognitive function can be reliably improved by simply administering a glucose drink. Clearly there are health issues associated with the administration of sugar; nevertheless this manipulation has provided a useful prototype for examining acute cognitive enhancement( Reference Scholey, Macpherson and Sunram-Lea 63 Reference Scholey, Sunram-Lea and Greer 65 ). In the context of nutritional interventions during an emotional episodic memory task, glucose improved both task performance and increased fMRI signal intensity in structures known to encode episodic memories( Reference Parent, Krebs-Kraft and Ryan 66 ). Additionally, increased connectivity was observed between the same brain regions that showed enhanced activation during task performance.

Magnetic resonance can also be used to measure the global cerebral rate of oxygen consumption. This has not yet been applied to dietary interventions, save for a single publication reporting reduced cerebral rate of oxygen consumption following glucose loading( Reference Xu, Liu and Pascual 67 ). The sensitivity to glucose suggests that the method may have applications in nutrition intervention studies.

More recently, magnetic resonance spectroscopy (MRS) has been successfully applied to the detection and quantification of various brain molecules. Proton MRS is the most commonly used methodology in human subjects (although other methods, including 31P, 13C and 19F MRS, have also been used). Proton MRS uses the signals emitted from hydrogen protons to measure central metabolite concentrations( Reference Novotny, Fulbright and Pearl 68 ). Many central molecules are invisible to MRS, with the most readily detectable resonance peaks being from N-acetyl aspartate, choline, creatine (Cr), glutamate/glutamine, and myo-inositol( Reference Rae 69 ). N-acetyl aspartate is a putative marker of neuronal density or viability and may index neurogenesis. Choline is enriched in membranes and is a marker of membrane turnover. Cr plays a key role in energy metabolism, while glutamate/glutamine represents the major excitatory neurotransmitter family and also correlates with metabolic activity. Myo-inositol is a second messenger involved in cell signalling and is also a marker of glial density. A small number of studies have examined age-related changes in these molecules with somewhat inconsistent findings( Reference Maudsley, Domenig and Govind 70 , Reference Chiu, Mak and Yau 71 ). Generally, levels of N-acetyl aspartate and glutamate/glutamine decline (possibly reflecting reduced neuronal number), while Cr and choline increase with age( Reference Maudsley, Domenig and Govind 70 , Reference Marjańska, McCarten and Hodges 72 ). Two small studies have used MRS to examine nutritional interventions. Six weeks Cr supplementation improved cognitive performance and increased central Cr levels( Reference Rae, Digney and McEwan 73 ). Central glucose can be measured using MRS, and one study( Reference Haley, Knight-Scott and Simnad 74 ) reported that oral glucose ingestion results in a measurable increase in the glucose MRS peak in eight AD patients compared with fourteen healthy young and fourteen age-matched controls. This reinforces the notion that the ability to effectively utilise central glucose contributes to the pathology of AD.

Several studies have examined the effects of vitamin and mineral supplementation on brain structure and function. These have focused on either single vitamins or broad-spectrum multivitamin mineral (MVM) preparations. Changes in patterns of activation were associated with MVM supplementation both acutely( Reference Scholey, Bauer and Neale 75 , Reference White, Camfield and Maggini 76 ) and following 4 weeks supplementation( Reference White, Cox and Hughes 77 ). One three-arm study compared placebo with MVM both with and without added guaraná. Focusing here on the MVM only arm, pilot fMRI data (n 6 in a three-arm crossover trial) indicated increased activation in a neural circuit associated with executive attentional functioning in healthy young adults in the hour following a single dose of an MVM( Reference Scholey, Bauer and Neale 75 ). In the same study, a specialised EEG methodology, steady-state visually evoked potential, was applied to a larger cohort (n 20) and found increased excitation in frontal regions coupled with inhibition in posterior regions during an attentional task( Reference White, Camfield and Maggini 76 ); see Fig. 3.

Fig. 3. Increased activation of a working memory network following administration of a broad-spectrum B vitamin and mineral (MVM) preparation compared with placebo (MVM > placebo), using two neuroimaging modalities (with warmer colour indicate greater difference over placebo). A and B show increased activation measured using functional MRI (fMRI) during the Rapid visual information processing task (as depicted in Fig. 2). Significantly increased activation is shown 30 min (A) and 28 d (B) following supplementation. Specifically A depicts activation of cerebellar regions shown in posterior (i), lateral (ii) and superior (iii) view, while B depicts clusters of increased activation following 28 d MVM supplementation, including parietal structures (precuneus and superior and inferior parietal lobes) and frontal regions (supplementary motor area). Graphs present levels of activation at baseline (BL) and following 28 d for MVM and placebo. C shows a similar network of activation to B during a spatial working memory task as measured using an electrophysiological measure Steady-state visual evoked potential (SSVEP). Effects follow the same 28-d intervention, and illustrate the more diffuse visualisation using this technique. Data are adapted from( Reference Scholey, Bauer and Neale 75 , Reference White, Cox and Hughes 77 ).

Acute neurocognitive effects of MVM administration, while not frequently reported, are not without precedence and, in this context, the absence of evidence should not be taken as evidence of absence. For example, acute mood and cognitive effects have been reported in children( Reference Haskell, Scholey and Jackson 78 ) and older adults( Reference Macpherson, Rowsell and Cox 79 ). While in healthy young adults, MVM administration was associated with acute increases in central blood flow, energy expenditure and fat oxidation( Reference Kennedy, Stevenson and Jackson 80 ). Clearly further work is needed to better characterise acute effects of MVM and the underlying mechanism of action.

The fact that these effects were evident in healthy young adults raises the question of what role initial nutrient status may play in the observed effects. In the case of the 4-week MVM trial, the neurocognitive benefits were accompanied by positive shifts in mood and also blood B vitamin levels( Reference White, Cox and Peters 81 ). This suggests that it is possible to optimise nutrient status and improve function in individuals who have sub-optimal levels or ‘insufficiency’ rather than frank nutrient deficiency (see Fig. 4). Clearly this challenges the dogma that benefits of MVM supplementation will only be observed in individuals with a clinical nutrient deficiency. Indeed, a recent report from >10 000 adults in the US National Health and Nutrition Examination Surveys study reveals that any MVM supplementation lowers the odds ratio of deficiency of four out of five nutrients with recognised markers of deficiency, and reduces insufficiency of fifteen/seventeen nutrients examined( Reference Blumberg, Frei and Fulgoni 82 ).

Fig. 4. Representation of relationship between nutrient status and functional status (adapted from( Reference Morris and Tangney 91 )). Frank nutrient deficiency (blue) manifests itself with clinical symptoms (y-axis) and these may be treated by optimising nutrient status (arrow a). There is growing evidence that suboptimal nutrient status (also termed marginal nutrient deficiency or nutrient insufficiency( Reference Popper 85 )) may be associated with subclinical functional deficits. These may be reversed by nutrient supplementation (arrow b).

Indeed, there is growing evidence for neurocognitive benefits of MVM supplementation in non-clinical populations. For example, in a 16-week trial of a MVM supplementation in healthy young adults, improved attentional performance in males was significantly correlated with increased levels of vitamin B6 ( Reference Pipingas, Camfield and Stough 83 ). This strongly suggests a functional role for changes in B6, despite the fact that the group was not deficient in B6 (see( Reference Kennedy 84 ) for an overview of the role of B vitamins in this context). This begs the question as to what proportion of the population have suboptimal levels of nutrients which may have functional behavioural consequences( Reference Popper 85 ). While frank clinical deficiencies may be found in up to 10–20 % of the population in Westernised countries (depending on the nutrient in question). The figure is much higher for those with marginal deficiencies, with estimates for vitamin B12 insufficiency as high as 40–50 % or more of the US and UK adult populations( Reference Tucker, Rich and Rosenberg 86 , Reference Moat, Ashfield-Watt and Powers 87 ).

More recently, B vitamin supplementation has been applied to clinical cognitive decline in the individuals with MCI( Reference de Jager 88 ). In the VITACOG trial, 156 over 70-year olds with MCI underwent structural MRI at baseline and at 24 months following either placebo or a B vitamin preparation. Those in the vitamin group showed reduced grey matter atrophy (0·5 % v. 3·7 % in the placebo group); this effect was more marked in those with higher baseline homocysteine (0·6 % v. 5·2 %). It should be noted that high and low homocysteine groups were derived by median splits and no participant met the criterion for clinically elevated homocysteine (150 pmol/l). Again this supports the potential for improved neurocognitive outcomes following supplementation in non-deficient, with the effects being more pronounced in those with suboptimal nutrient status (Fig. 4).

Nutrition and neurocognitive scaffolding

What can these methodologies tell us about the mechanisms which might underlie cognitive enhancement from nutritional interventions? One clue might come from the Scaffolding theory of aging and cognition (STAC). In its original form, the STAC model proposed that ageing is characterised by a balance between negative and positive neural plasticity( Reference Park and Reuter-Lorenz 34 ), and that cognitive decline represents a preponderance of the former over the latter. Negative plasticity includes neural challenges and functional deterioration. Neural challenges include the previously described structural corrosion of grey and white matter which occurs with ageing. Functional deterioration describes dysregulation of neural activity, including reduced task-related recruitment of hippocampal and related structures during memory encoding, and maladaptive patterns in the default mode network. The STAC model argues that, at the same time, positive plasticity in the form of compensatory scaffolding occurs which acts to counter these negative structural and functional changes. In brain imaging studies, this compensatory scaffolding manifests itself as greater activation (i.e. more recruitment) of frontal and parietal regions and increased bilateral recruitment in tasks which may be lateralised in younger individuals. It is notable that the neuroimaging examples described earlier include the possibility of frontal recruitment being increased by multivitamin/nutrient supplementation and neurogenesis being facilitated by flavanols and n-3 supplementation. Later revisions of the model speculate that neurogenesis may also be part of scaffolding mechanisms( Reference Reuter-Lorenz and Park 89 ). Importantly, the STAC model suggests that it is possible to promote neural scaffolding, including via ‘various lifestyle activities including exercise, intellectual engagement and new learning, as well as more formal cognitive training interventions’( Reference Reuter-Lorenz and Park 89 ). The select neuroimaging examples described earlier suggest that nutritional interventions may also be added to this list.

In conclusion, converging evidence from epidemiology, mechanistic studies and clinical trials strongly suggest that nutritional interventions may offer a realistic option for offsetting neurocognitive decline. Such interventions include whole dietary change, select food components, well-characterised botanical extracts and nutrient supplementation. Since cognitive processes are influenced by multiple biological systems, positive effects of nutrition are likely to be pleiotropic. Recent studies employing a range of biomarkers and neuroimaging methodologies have shed light on which changes in neural substrates, including neurocognitive scaffolding, underlie benefits of nutrient interventions. In turn, such studies are helping to identify biologically plausible targets for nutrients. Thus, there is presently a realistic opportunity for a ‘virtuous cycle’, where evidence-led interventions and mechanistic studies reciprocally inform each other to further develop nutritional interventions for neurocognition. It should be noted that for many potentially beneficial, centrally active nutrients, there is a history of safe consumption. Compared with traditional pharmaceutical drug development pipelines, where many candidate treatments fail at early stages, this may allow more rapid translation of research findings into benefits for neurocognitive health.

Acknowledgements

The author is grateful to Dr David White who provided images used in Figs 2 and 3.

Financial Support

None.

Conflicts of Interest

The author has received research funding, consultancy, honoraria and travel costs from various nutrition and supplement industry bodies.

Authorship

The author had sole responsibility for all aspects of preparation of this paper.

References

1. Suzman, R & Beard, J (2011) Global health and aging. US Department of State, pp. 132. Available at: http://www.who.int/ageing/publications/global_health.pdf Google Scholar
2. Kontis, V, Bennett, JE, Mathers, CD et al. (2017) Future life expectancy in 35 industrialised countries: projections with a Bayesian model ensemble. Lancet 389, 13231335.CrossRefGoogle ScholarPubMed
3. Prince, M, Guerchet, M & Prina, M (2016) The epidemiology and impact of dementia: current state and future trends 2015. http://wdelword_wwwhoint/mental_health/neurology/dementia/dementia_thematicbrief_epidemiologypdf (accessed December 2016) [WebCite Cache].Google Scholar
4. Bekris, LM, Yu, C-E, Bird, TD et al. (2010) Review article: genetics of Alzheimer disease. J Geriatr Psychiatry Neurol 23, 213227.CrossRefGoogle ScholarPubMed
5. Deary, IJ, Yang, J, Davies, G et al. (2012) Genetic contributions to stability and change in intelligence from childhood to old age. Nature 482, 212215.CrossRefGoogle ScholarPubMed
6. Association As (2016) 2016 Alzheimer's disease facts and figures. Alzheimer's Dementia 12, 459509.CrossRefGoogle Scholar
7. Jessen, F, Feyen, L, Freymann, K et al. (2006) Volume reduction of the entorhinal cortex in subjective memory impairment. Neurobiol Aging 27, 17511756.CrossRefGoogle ScholarPubMed
8. Rodda, J, Dannhauser, T, Cutinha, D et al. (2011) Subjective cognitive impairment: functional MRI during a divided attention task. Eur Psychiatry 26, 457462.CrossRefGoogle ScholarPubMed
9. Jessen, F, Wolfsgruber, S, Wiese, B et al. (2014) AD dementia risk in late MCI, in early MCI, and in subjective memory impairment. Alzheimer's Dementia 10, 7683.CrossRefGoogle ScholarPubMed
10. Ward, A, Tardiff, S, Dye, C et al. (2013) Rate of conversion from prodromal Alzheimer's disease to Alzheimer's dementia: a systematic review of the literature. Dementia Geriatric Cogn Disorders Extra 3, 320332.CrossRefGoogle ScholarPubMed
11. Deardorff, WJ, Feen, E & Grossberg, GT (2015) The use of cholinesterase inhibitors across all stages of Alzheimer's disease. Drugs Aging 32, 537547.CrossRefGoogle ScholarPubMed
12. Kennedy, DO, Dodd, FL, Robertson, BC et al. (2011) Monoterpenoid extract of sage (Salvia lavandulaefolia) with cholinesterase inhibiting properties improves cognitive performance and mood in healthy adults. J Psychopharmacol 25, 10881100.CrossRefGoogle ScholarPubMed
13. Scholey, AB, Tildesley, NTJ, Ballard, CG et al. (2008) An extract of Salvia (sage) with anticholinesterase properties improves memory and attention in healthy older volunteers. Psychopharmacology 198, 127139.CrossRefGoogle ScholarPubMed
14. Tildesley, NTJ, Kennedy, DO, Perry, EK et al. (2003) Salvia lavandulaefolia (Spanish Sage) enhances memory in healthy young volunteers. Pharmacol Biochem Behav 75, 669674.CrossRefGoogle ScholarPubMed
15. Tildesley, NTJ, Kennedy, DO, Perry, EK et al. (2005) Positive modulation of mood and cognitive performance following administration of acute doses of Salvia lavandulaefolia essential oil to healthy young volunteers. Physiol Behav 83, 699709.CrossRefGoogle ScholarPubMed
16. Miroddi, M, Navarra, M, Quattropani, MC et al. (2014) Systematic review of clinical trials assessing pharmacological properties of Salvia species on memory, cognitive impairment and Alzheimer's disease. CNS Neurosci Therapeutics 20, 485495.CrossRefGoogle ScholarPubMed
17. Jack, CR, Knopman, DS, Jagust, WJ et al. (2010) Hypothetical model of dynamic biomarkers of the Alzheimer's pathological cascade. Lancet Neurol 9, 119128.CrossRefGoogle ScholarPubMed
18. Qiu, C & Fratiglioni, L (2015) A major role for cardiovascular burden in age-related cognitive decline. Nat Rev Cardiol 12, 267277.CrossRefGoogle Scholar
19. Langa, KM (2015) Is the risk of Alzheimer's disease and dementia declining? Alzheimer's Res Ther 7, 34.CrossRefGoogle ScholarPubMed
20. Hardman, RJ, Kennedy, G, Macpherson, H et al. (2016) Adherence to a Mediterranean-style diet and effects on cognition in adults: a qualitative evaluation and systematic review of longitudinal and prospective trials. Front Nutr 3. Available at: https://www.frontiersin.org/articles/10.3389/fnut.2016.00022/full CrossRefGoogle ScholarPubMed
21. Nurk, E, Refsum, H, Drevon, CA et al. (2009) Intake of flavonoid-rich wine, tea, and chocolate by elderly men and women is associated with better cognitive test performance. J Nutr 139, 120127.CrossRefGoogle ScholarPubMed
22. Cox, KH & Scholey, A (2016) Polyphenols for brain and cognitive health. Recent Adv Polyphenol Res 5, 259.Google Scholar
23. Letenneur, L, Proust-Lima, C, Le Gouge, A et al. (2007) Flavonoid intake and cognitive decline over a 10-year period. Am J Epidemiol 165, 13641371.CrossRefGoogle ScholarPubMed
24. Camfield, DA, Stough, C, Farrimond, J et al. (2014) Acute effects of tea constituents L-theanine, caffeine, and epigallocatechin gallate on cognitive function and mood: a systematic review and meta-analysis. Nutr Rev 72, 507522.CrossRefGoogle Scholar
25. Sandhu, KV, Sherwin, E, Schellekens, H et al. (2017) Feeding the microbiota-gut-brain axis: diet, microbiome, and neuropsychiatry. Transl Res 179, 223244.CrossRefGoogle ScholarPubMed
26. Albenberg, LG & Wu, GD (2014) Diet and the intestinal microbiome: associations, functions, and implications for health and disease. Gastroenterology 146, 15641572.CrossRefGoogle ScholarPubMed
27. Kristensen, NB, Bryrup, T, Allin, KH et al. (2016) Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: a systematic review of randomized controlled trials. Genome Med 8, 52.CrossRefGoogle ScholarPubMed
28. Akbari, E, Asemi, Z, Kakhaki, RD et al. (2016) Effect of probiotic supplementation on cognitive function and metabolic status in Alzheimer's disease: a randomized, double-blind and controlled trial. Front Aging Neurosci 8. Available at: https://www.frontiersin.org/articles/10.3389/fnagi.2016.00256/full CrossRefGoogle ScholarPubMed
29. Kelly, JR, Allen, AP, Temko, A et al. (2017) Lost in translation? The potential psychobiotic Lactobacillus rhamnosus (JB-1) fails to modulate stress or cognitive performance in healthy male subjects. Brain Behav Immun 61, 5059.CrossRefGoogle ScholarPubMed
30. Pirbaglou, M, Katz, J, de Souza, RJ et al. (2016) Probiotic supplementation can positively affect anxiety and depressive symptoms: a systematic review of randomized controlled trials. Nutr Res 36, 889898.CrossRefGoogle ScholarPubMed
31. Wilson, RS, Beckett, LA, Barnes, LL et al. (2002) Individual differences in rates of change in cognitive abilities of older persons. Psychol Aging 17, 179.CrossRefGoogle ScholarPubMed
32. Deary, IJ, Corley, J, Gow, AJ et al. (2009) Age-associated cognitive decline. Br Med Bull 92, 135152.CrossRefGoogle ScholarPubMed
33. Crivello, F, Tzourio-Mazoyer, N, Tzourio, C et al. (2014) Longitudinal assessment of global and regional rate of grey matter atrophy in 1,172 healthy older adults: modulation by sex and age. PLoS ONE 9, e114478.CrossRefGoogle Scholar
34. Park, DC & Reuter-Lorenz, P (2009) The adaptive brain: aging and neurocognitive scaffolding. Annu Rev Psychol 60, 173196.CrossRefGoogle ScholarPubMed
35. Callaghan, MF, Freund, P, Draganski, B et al. (2014) Widespread age-related differences in the human brain microstructure revealed by quantitative magnetic resonance imaging. Neurobiol Aging 35, 18621872.CrossRefGoogle ScholarPubMed
36. Konkel, A & Cohen, NJ (2009) Relational memory and the hippocampus: representations and methods. Front Neurosci 3, 23.CrossRefGoogle ScholarPubMed
37. Monti, JM, Baym, CL & Cohen, NJ (2014) Identifying and characterizing the effects of nutrition on hippocampal memory. Adv Nutr: Int Rev J 5, 337S343S.CrossRefGoogle ScholarPubMed
38. Erickson, KI, Voss, MW, Prakash, RS et al. (2011) Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci USA 108, 30173022.CrossRefGoogle ScholarPubMed
39. Pereira, AC, Huddleston, DE, Brickman, AM et al. (2007) An in vivo correlate of exercise-induced neurogenesis in the adult dentate gyrus. Proc Natl Acad Sci USA 104, 56385643.CrossRefGoogle Scholar
40. Van Praag, H, Lucero, MJ, Yeo, GW et al. (2007) Plant-derived flavanol (−) epicatechin enhances angiogenesis and retention of spatial memory in mice. J Neurosci 27, 58695878.CrossRefGoogle ScholarPubMed
41. Luchtman, DW & Song, C (2013) Cognitive enhancement by omega-3 fatty acids from child-hood to old age: findings from animal and clinical studies. Neuropharmacology 64, 550565.CrossRefGoogle ScholarPubMed
42. Neale, C, Camfield, D, Reay, J et al. (2013) Cognitive effects of two nutraceuticals Ginseng and Bacopa benchmarked against modafinil: a review and comparison of effect sizes. Br J Clin Pharmacol 75, 728737.CrossRefGoogle ScholarPubMed
43. Bucur, B, Madden, DJ, Spaniol, J et al. (2008) Age-related slowing of memory retrieval: contributions of perceptual speed and cerebral white matter integrity. Neurobiol Aging 29, 10701079.CrossRefGoogle ScholarPubMed
44. Karakis, I, Pase, MP, Beiser, A et al. (2016) Association of serum vitamin D with the risk of incident dementia and subclinical indices of brain aging: the Framingham Heart Study. J Alzheimer's Dis 51, 451461.CrossRefGoogle ScholarPubMed
45. Janssen, CI & Kiliaan, AJ (2014) Long-chain polyunsaturated fatty acids (LCPUFA) from genesis to senescence: the influence of LCPUFA on neural development, aging, and neurodegeneration. Prog Lipid Res 53, 117.CrossRefGoogle ScholarPubMed
46. Witte, AV, Kerti, L, Hermannstädter, HM et al. (2013) Long-chain omega-3 fatty acids improve brain function and structure in older adults. Cereb Cortex 24, 30593068.CrossRefGoogle ScholarPubMed
47. Camfield, DA & Scholey, A (2015). Use of neuroimaging techniques in the assessment of nutraceuticals for cognitive enhancement: Methodological and interpretative issues.CrossRefGoogle Scholar
48. Scholey, A, Downey, LA, Ciorciari, J et al. (2012) Acute neurocognitive effects of epigallocatechin gallate (EGCG). Appetite 58, 767770.CrossRefGoogle ScholarPubMed
49. Kobayashi, K, Nagato, Y, Aoi, N et al. (1998) Effects of L-theanine on the release of α-brain waves in human volunteers. Nippon Nōgei Kagakukaishi 72, 153157.CrossRefGoogle Scholar
50. Juneja, LR, Chu, DC, Okubo, T et al. (1999) L-theanine – a unique amino acid of green tea and its relaxation effect in humans. Trends Food Sci Technol 10, 199204.CrossRefGoogle Scholar
51. Gomez-Ramirez, M, Higgins, BA, Rycroft, JA et al. (2007) The deployment of intersensory selective attention: a high-density electrical mapping study of the effects of theanine. Clin Neuropharmacol 30, 25.CrossRefGoogle ScholarPubMed
52. Gomez-Ramirez, M, Kelly, SP, Montesi, JL et al. (2009) The effects of L-theanine on alpha-band oscillatory brain activity during a visuo-spatial attention task. Brain Topogr 22, 4451.CrossRefGoogle ScholarPubMed
53. White, DJ, de Klerk, S, Woods, W et al. (2016) Anti-stress, behavioural and magnetoencephalography effects of an L-theanine-based nutrient drink: a randomised, double-blind, placebo-controlled, crossover trial. Nutrients 8, 5373.CrossRefGoogle ScholarPubMed
54. Belliveau, J, Kennedy, D, McKinstry, R et al. (1991) Functional mapping of the human visual cortex by magnetic resonance imaging. Science 254, 716719.CrossRefGoogle ScholarPubMed
55. Scholey, A & Owen, L (2013) Effects of chocolate on cognitive function and mood: a systematic review. Nutr Rev 71, 665681.CrossRefGoogle ScholarPubMed
56. Massee, LA, Ried, K, Pase, M et al. (2015) The acute and sub-chronic effects of cocoa flavanols on mood, cognitive and cardiovascular health in young healthy adults: a randomized, controlled trial. Front Pharmacol 6. Available at: https://www.frontiersin.org/articles/10.3389/fphar.2015.00093/full CrossRefGoogle ScholarPubMed
57. Scholey, AB, French, SJ, Morris, PJ et al. (2010) Consumption of cocoa flavanols results in acute improvements in mood and cognitive performance during sustained mental effort. J Psychopharmacol 24, 15051514.CrossRefGoogle ScholarPubMed
58. Francis, S, Head, K, Morris, P et al. (2006) The effect of flavanol-rich cocoa on the fMRI response to a cognitive task in healthy young people. J Cardiovasc Pharmacol 47, S215S220.CrossRefGoogle ScholarPubMed
59. Lamport, DJ, Pal, D, Moutsiana, C et al. (2015) The effect of flavanol-rich cocoa on cerebral perfusion in healthy older adults during conscious resting state: a placebo controlled, crossover, acute trial. Psychopharmacology 232, 32273234.CrossRefGoogle Scholar
60. Brickman, AM, Khan, UA, Provenzano, FA et al. (2014) Enhancing dentate gyrus function with dietary flavanols improves cognition in older adults. Nat Neurosci 17, 17981803.CrossRefGoogle ScholarPubMed
61. Mastroiacovo, D, Kwik-Uribe, C, Grassi, D et al. (2015) Cocoa flavanol consumption improves cognitive function, blood pressure control, and metabolic profile in elderly subjects: the Cocoa, Cognition, and Aging (CoCoA) Study – a randomized controlled trial. Am J Clin Nutr 101, 538548.CrossRefGoogle ScholarPubMed
62. Kandimalla, R, Thirumala, V & Reddy, PH (2017) Is Alzheimer's disease a type 3 diabetes? A critical appraisal. Biochimica Biophysica Acta (BBA)-Mol. Basis Dis 1863, 10781089.CrossRefGoogle ScholarPubMed
63. Scholey, A, Macpherson, H, Sunram-Lea, S et al. (2013) Glucose enhancement of recognition memory: differential effects on effortful processing but not aspects of ‘remember-know’ responses. Neuropharmacology 64, 544549.CrossRefGoogle Scholar
64. Scholey, AB, Harper, S & Kennedy, DO (2001) Cognitive demand and blood glucose. Physiol Behav 73, 585592.CrossRefGoogle ScholarPubMed
65. Scholey, AB, Sunram-Lea, SI, Greer, J et al. (2009) Glucose administration prior to a divided attention task improves tracking performance but not word recognition: evidence against differential memory enhancement? Psychopharmacology 202, 549558.CrossRefGoogle Scholar
66. Parent, MB, Krebs-Kraft, DL, Ryan, JP et al. (2011) Glucose administration enhances fMRI brain activation and connectivity related to episodic memory encoding for neutral and emotional stimuli. Neuropsychologia 49, 10521066.CrossRefGoogle ScholarPubMed
67. Xu, F, Liu, P, Pascual, JM et al. (2015) Acute effect of glucose on cerebral blood flow, blood oxygenation, and oxidative metabolism. Hum Brain Mapp 36, 707716.CrossRefGoogle ScholarPubMed
68. Novotny, EJ, Fulbright, RK, Pearl, PL et al. (2003) Magnetic resonance spectroscopy of neurotransmitters in human brain. Ann Neurol 54, S25S31.CrossRefGoogle ScholarPubMed
69. Rae, CD (2014) A guide to the metabolic pathways and function of metabolites observed in human brain 1H magnetic resonance spectra. Neurochem Res 39, 136.CrossRefGoogle Scholar
70. Maudsley, AA, Domenig, C, Govind, V et al. (2009) Mapping of brain metabolite distributions by volumetric proton MR spectroscopic imaging (MRSI). Magn Reson Med 61, 548559.CrossRefGoogle ScholarPubMed
71. Chiu, P-W, Mak, HK-F, Yau, KK-W et al. (2014) Metabolic changes in the anterior and posterior cingulate cortices of the normal aging brain: proton magnetic resonance spectroscopy study at 3T. Age 36, 251264.CrossRefGoogle Scholar
72. Marjańska, M, McCarten, JR, Hodges, J et al. (2017) Region-specific aging of the human brain as evidenced by neurochemical profiles measured noninvasively in the posterior cingulate cortex and the occipital lobe using 1 H magnetic resonance spectroscopy at 7T. Neuroscience 354, 168177.CrossRefGoogle Scholar
73. Rae, C, Digney, AL, McEwan, SR et al. (2003) Oral creatine monohydrate supplementation improves brain performance: a double-blind, placebo-controlled, cross-over trial. Proc R Soc B 270, 21472150.CrossRefGoogle ScholarPubMed
74. Haley, AP, Knight-Scott, J, Simnad, VI et al. (2006) Increased glucose concentration in the hippocampus in early Alzheimer's disease following oral glucose ingestion. Magn Reson Imaging 24, 715720.CrossRefGoogle ScholarPubMed
75. Scholey, A, Bauer, I, Neale, C et al. (2013) Acute effects of different multivitamin mineral preparations with and without Guarana on mood, cognitive performance and functional brain activation. Nutrients 5, 35893604.CrossRefGoogle ScholarPubMed
76. White, DJ, Camfield, DA, Maggini, S et al. (2017) The effect of a single dose of multivitamin and mineral combinations with and without guaraná on functional brain activity during a continuous performance task. Nutr Neurosci 20, 822.CrossRefGoogle ScholarPubMed
77. White, DJ, Cox, KH, Hughes, ME et al. (2016) Functional brain activity changes after 4 weeks supplementation with a multi-vitamin/mineral combination: a randomized, double-blind, placebo-controlled trial exploring functional magnetic resonance imaging and steady-state visual evoked potentials during working memory. Front Aging Neurosci 8. Available at: https://www.frontiersin.org/articles/10.3389/fnagi.2016.00288/full CrossRefGoogle ScholarPubMed
78. Haskell, CF, Scholey, AB, Jackson, PA et al. (2008) Cognitive and mood effects in healthy children during 12 weeks’ supplementation with multi-vitamin/minerals. Br J Nutr 100, 10861096.CrossRefGoogle ScholarPubMed
79. Macpherson, H, Rowsell, R, Cox, KHM et al. (2015) Acute mood but not cognitive improvements following administration of a single multivitamin and mineral supplement in healthy women aged 50 and above: a randomised controlled trial. Age 37, 3848.CrossRefGoogle Scholar
80. Kennedy, DO, Stevenson, EJ, Jackson, PA et al. (2016) Multivitamins and minerals modulate whole-body energy metabolism and cerebral blood-flow during cognitive task performance: a double-blind, randomised, placebo-controlled trial. Nutr Metab 13, 11.CrossRefGoogle ScholarPubMed
81. White, DJ, Cox, KHM, Peters, R et al. (2015) Effects of four-week supplementation with a multi-vitamin/mineral preparation on mood and blood biomarkers in young adults: a randomised, double-blind, placebo-controlled trial. Nutrients 7, 90059017.CrossRefGoogle ScholarPubMed
82. Blumberg, JB, Frei, BB, Fulgoni, VL et al. (2017) Impact of frequency of multi-vitamin/multi-mineral supplement intake on nutritional adequacy and nutrient deficiencies in US adults. Nutrients 9, 849.CrossRefGoogle Scholar
83. Pipingas, A, Camfield, DA, Stough, C et al. (2014) Effects of multivitamin, mineral and herbal supplement on cognition in younger adults and the contribution of B group vitamins. Hum Psychopharmacol-Clin Exp 29, 7382.CrossRefGoogle ScholarPubMed
84. Kennedy, DO (2016) B vitamins and the brain: mechanisms, dose and efficacy – a review. Nutrients 8, 68.CrossRefGoogle ScholarPubMed
85. Popper, CW (2014) Single-micronutrient and broad-spectrum micronutrient approaches for treating mood disorders in youth and adults. Child Adolesc Psychiatr Clin N Am 23, 591672.CrossRefGoogle Scholar
86. Tucker, KL, Rich, S, Rosenberg, I et al. (2000) Plasma vitamin B-12 concentrations relate to intake source in the Framingham Offspring study. Am J Clin Nutr 71, 514522.CrossRefGoogle ScholarPubMed
87. Moat, SJ, Ashfield-Watt, PA, Powers, HJ et al. (2003) Effect of riboflavin status on the homocysteine-lowering effect of folate in relation to the MTHFR (C677T) genotype. Clin Chem 49, 295302.CrossRefGoogle Scholar
88. de Jager, CA (2014) Critical levels of brain atrophy associated with homocysteine and cognitive decline. Neurobiol Aging 35, S35SS9.CrossRefGoogle ScholarPubMed
89. Reuter-Lorenz, PA & Park, DC (2014) How does it STAC up? Revisiting the scaffolding theory of aging and cognition. Neuropsychol Rev 24, 355370.CrossRefGoogle ScholarPubMed
90. Neale, C, Johnston, P, Hughes, M et al. (2015) Functional activation during the rapid visual information processing task in a middle aged cohort: an fMRI study. PLoS ONE 10, e0138994.CrossRefGoogle Scholar
91. Morris, MC & Tangney, CC (2011) A potential design flaw of randomized trials of vitamin supplements. JAMA 305, 13481349.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Influences on neurocognitive ageing including therapeutic targets for delaying age-related neurocognitive decline. Pharmaceutical approaches are largely restricted to effects on neurotransmitter systems (grey panel). Processes within the green-shaded area represent realistic nutritional targets. Age and genetics (unshaded area) remain non-modifiable factors. HPA, hypothalamic–pituitary–adrenal axis.

Figure 1

Fig. 2. Rapid visual information processing (RVIP) as example functional MRI task used to capture effects of nutritional intervention studies. Panel A shows the active task requiring the subject to respond to three consecutive odd or three consecutive even digits when single digits are presented every 600 ms. In the control task (B), matched for perceptual and motor activity, subjects see a similar stimulus stream but respond to the presentation of the ‘0’ digit only (i.e. there is minimal cognitive load). When activation measured during B is subtracted from that during A, there is consistent activation in a well-characterised working memory circuit consisting of more anterior (ant.) frontal and supplementary motor areas and more posterior (post.) parietal and cerebellar regions linked with attention and working memory. C and D illustrate this pattern of activation from (77,90) with warmer colours depicting greater activation. Increased activation within this circuit has been found following multivitamin mineral supplementation(77), see Fig. 3.

Figure 2

Fig. 3. Increased activation of a working memory network following administration of a broad-spectrum B vitamin and mineral (MVM) preparation compared with placebo (MVM > placebo), using two neuroimaging modalities (with warmer colour indicate greater difference over placebo). A and B show increased activation measured using functional MRI (fMRI) during the Rapid visual information processing task (as depicted in Fig. 2). Significantly increased activation is shown 30 min (A) and 28 d (B) following supplementation. Specifically A depicts activation of cerebellar regions shown in posterior (i), lateral (ii) and superior (iii) view, while B depicts clusters of increased activation following 28 d MVM supplementation, including parietal structures (precuneus and superior and inferior parietal lobes) and frontal regions (supplementary motor area). Graphs present levels of activation at baseline (BL) and following 28 d for MVM and placebo. C shows a similar network of activation to B during a spatial working memory task as measured using an electrophysiological measure Steady-state visual evoked potential (SSVEP). Effects follow the same 28-d intervention, and illustrate the more diffuse visualisation using this technique. Data are adapted from(75,77).

Figure 3

Fig. 4. Representation of relationship between nutrient status and functional status (adapted from(91)). Frank nutrient deficiency (blue) manifests itself with clinical symptoms (y-axis) and these may be treated by optimising nutrient status (arrow a). There is growing evidence that suboptimal nutrient status (also termed marginal nutrient deficiency or nutrient insufficiency(85)) may be associated with subclinical functional deficits. These may be reversed by nutrient supplementation (arrow b).