Hostname: page-component-76fb5796d-dfsvx Total loading time: 0 Render date: 2024-04-26T00:52:53.543Z Has data issue: false hasContentIssue false

Potential benefits of phytochemicals against Alzheimer's disease

Published online by Cambridge University Press:  01 February 2017

Emma L. Wightman*
Affiliation:
Brain, Performance and Nutrition Research Centre, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
*
Corresponding author: E. L. Wightman, tel (+44) 0191 2437253, email emma.l.wightman@northumbria.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Our current therapeutic drugs for Alzheimer's disease are predominantly derived from the alkaloid class of plant phytochemicals. These drugs, such as galantamine and rivastigmine, attenuate the decline in the cholinergic system but, as the alkaloids occupy the most dangerous end of the phytochemical spectrum (indeed they function as feeding deterrents and poisons to other organisms within the plant itself), they are often associated with unpleasant side effects. In addition, these cholinesterase inhibiting alkaloids target only one system in a disorder, which is typified by multifactorial deficits. The present paper will look at the more benign terpene (such as Ginkgo biloba, Ginseng, Melissa officinalis (lemon balm) and Salvia lavandulaefolia (sage)) and phenolic (such as resveratrol) phytochemicals; arguing that they offer a safer alternative and that, as well as demonstrating efficacy in cholinesterase inhibition, these phytochemicals are able to target other salient systems such as cerebral blood flow, free radical scavenging, anti-inflammation, inhibition of amyloid-β neurotoxicity, glucoregulation and interaction with other neurotransmitters (such as γ-aminobutyric acid) and signalling pathways (e.g. via kinase enzymes).

Type
Conference on ‘Phytochemicals and health: new perspectives on plant-based nutrition’
Copyright
Copyright © The Author 2017 

Background

To date, research into the psychophysiological effects of nutritional supplements in human subjects has predominantly focused on young, healthy, cognitively intact individuals. The premise underlying this body of research is that the supplementation of these compounds will, via a multitude of mechanisms, enhance some aspect/s of cognitive function, mood and/or physical performance. Naturally these studies produce varied results with some robust results evinced from compounds such as caffeine( Reference Haskell, Kennedy and Wesnes 1 ), the neural substrates oxygen( Reference Moss, Scholey and Wesnes 2 , Reference Scholey, Moss and Neave 3 ) and glucose( Reference Scholey, Laing and Kennedy 4 ) and, more recently, supplementation of the water-soluble vitamins( Reference Kennedy, Stevenson and Jackson 5 ). However, other supplemented compounds appear almost to elicit no cognitive benefit to the young, healthy cohorts utilised; the polyphenol resveratrol, for example( Reference Kennedy, Wightman and Reay 6 Reference Wightman, Haskell-Ramsay and Reay 8 ). This has led to the conclusion that some supplements may have limited cognitive benefit in those who are within the cognitive peak age-range (i.e. 18–35 years)( Reference Rönnlund, Nyberg and Bäckman 9 ) and that the mechanism underpinning their purported activity might be of more interest and benefit to those who are experiencing natural and pathological neurocognitive decline. Currently, pharmacological treatment options for pathological neurocognitive disorders such as Alzheimer's disease (AD) are derived from the alkaloid class of plant phytochemical compounds and this report will outline the disadvantages of this group and present an argument for, instead, looking at the potential benefit that taking these drugs from the more benign terpene and phenolic class of phytochemicals could provide in terms of safety and clinical benefit.

Alzheimer's disease and current treatment options from the alkaloid secondary metabolites

AD is the most common form of dementia. This is a global, progressive neurocognitive disorder typified by amyloid-β protein plaques and tau protein tangles outside and inside, respectively, of the neural cell body. These insults ultimately disrupt all cognitive processes and result in death( 10 ). The World Alzheimer Report 2015( 11 ) estimates that, worldwide, 46·8 million people live with a dementia and that this number will double every 20 years. The main risk factor for developing AD, and other dementias, is age but this is a multifactorial disease, which is also influenced (positively and negatively) by genetics (specifically the ApoE gene has received much recent attention)( Reference Karch and Goate 12 ), diet( Reference Luchsinger and Mayeux 13 ), nicotine( Reference Lee 14 , Reference Durazzo, Mattsson and Weiner 15 ) and alcohol( Reference Mukamal, Kuller and Fitzpatrick 16 ) consumption, free radical damage( Reference Markesbery 17 ), glucose regulation( Reference Vignini, Giulietti and Nanetti 18 ), cerebral blood flow( Reference Kalaria 19 ), inflammation( Reference Matrone, Djelloul and Taglialatela 20 ), ferrous metals( Reference Adlard and Bush 21 ), hormones( Reference Vest and Pike 22 ), socioeconomic status( Reference Sattler, Toro and Schönknecht 23 ) and many more known and unknown variables.

The morphological changes to neurons that the afore-mentioned risk factors mediate are seen to predominantly disrupt the cholinergic neurotransmitter system and, in turn, the cognitive processes that the ubiquitous neurotransmitter acetylcholine sub-serves. Hence, current pharmaceutical drugs for AD solely target this cholinergic decline( Reference Mufson, Counts and Perez 24 ). These drugs include galantamine and rivastigmine and, as a group of drugs defined as cholinesterase inhibitors (preventing the deamination of acetylcholine), these are currently the only approved first line pharmacologic treatment for AD in the UK( 25 ). A recent Cochrane review reported that these drugs attenuate the decline in cognition, daily living and behaviour in AD when compared with placebo( Reference Birks and Craig 26 ) but, interestingly, highlighted that none of the treatment effects were large. Cholinesterase inhibiting drugs also lack efficacy in some stages of AD and here use of the antipsychotic drug risperidone is often turned to in order the mediate challenging behaviour( 27 ). Cholinesterase drugs are also associated with some quite unpleasant side effects (including gastrointestinal problems( Reference Birks and Craig 26 )) and this is likely related to their current derivation from the alkaloid spectrum of plant secondary metabolites (hereafter referred to as phytochemicals) in that they occupy relatively more dangerous roles within the plant.

Phytochemicals exist to mediate communication and protection of the static plant and, in doing so, increase its survivability( Reference Kennedy and Wightman 28 ). These compounds fall into one of three categories; the alkaloids, terpenes and phenolics, with this order denoting their potency from dangerous to relatively benign. Within the plant, although there is some overlap, each category of phytochemical appears to have a particular function. Here the alkaloids are broadly expressed as poisons to deter the encroachment of other plants and potentially destructive insects. The terpenes also play a role in defence and deterrence but their provision of attractive colours and smells within the plant also demonstrates their role in attraction to facilitate pollination. Finally, the phenolics occupy the most benign ground in terms of safety and their role appears to be one of protection; expressed as they are when the plant comes under some kind of stress( Reference Kennedy and Wightman 28 ). Of interest here, many phenolic and terpene phytochemicals have also demonstrated efficacy against cholinergic decline and, beyond this, many of the other factors contributing to AD; which the current alkaloid-based drugs do not. Added to this, their relatively benign ecological roles mean that they may also represent a safer way of attenuating neurocognitive decline in AD. The following discusses those terpenes and phenolics, which represent the current most promising phytochemicals in this regard.

Potential benefit of terpene phytochemicals against Alzheimer's disease

Terpenes are a diverse group of more than 30 000 lipid-soluble compounds and exhibit a range of toxicity from deadly to entirely edible. This is in keeping with their broad range of ecological roles, which include antimicrobial properties and a range of measures, which attract symbiotes for the purposes of pollination, seed dispersal and secondary protective roles. This complex communication with insects requires the ability to interact directly with the central nervous system (CNS), including hormones and the γ-aminobutyric acid and cholinergic neurotransmitter systems; interactions, which should also translate to the human CNS and, as a result, provide benefit to AD (for review( Reference Kennedy and Wightman 28 )).

Ginkgo biloba

Extracts of G. biloba leaf contain a number of bioactive components, which include diterpenes, ginkgolides A, B, C, J and M, the sesquiterpene bilobalide and a range of flavonoids. The synergistic effects of these phytochemicals results in interactions with a number of CNS systems, which would be expected to attenuate neurocognitive decline. These include an up-regulation of the vasorelaxatory neurotransmitter nitric oxide and a resulting increase in cerebral blood flow, a down-regulation in the enzymatic deamination of monoaminergic neurotransmitters, free radical scavenging and neuroprotection, which includes reduced amyloid-β neurotoxicity( Reference Fehske, Leuner and Müller 29 Reference DeFeudis and Drieu 31 ). These interactions support the prescription of ginkgo for millennia in traditional Eastern forms of medicine for disorders of old age including AD( Reference Berger 32 ) and the beneficial effects seen in vitro and in vivo animal models of AD; where attenuation of cognitive decline has been observed in the AD mouse( Reference Stackman, Eckenstein and Frei 33 ); often reported to be due to protection against amyloid-β-induced oxidative stress( Reference Smith and Luo 34 ). Modern, controlled intervention trials in human subjects have also yielded some positive results.

In 2002, a Cochrane review concluded that ‘overall there is promising evidence of improvement in cognition and function associated with ginkgo’( Reference Birks and Grimley Evans 35 ) but, in 2009, this message had changed to one blighted by ‘inconsistent’ and ‘unconvincing’ results( Reference Birks and Evans 36 ). This is despite a study conducted in the same year where cognitive decline, as assessed by the Alzheimer's disease assessment scale (ADAS-cog), was attenuated by ginkgo( Reference Snitz, O'Meara and Carlson 37 ) but perhaps represents the influence of several small, heterogeneous studies on a research area still in its infancy. Nevertheless, since this review, a handful of larger scale reviews have reported more promising results of ginkgo. In 2010, a review of nine studies, comprising 2372 patients with various dementias, found that ginkgo attenuated declines in cognitive performance across all dementia groups tested and additional improvements in activities of daily living were seen in the AD groups( Reference Weinmann, Roll and Schwarzbach 38 ). In the same year a review of six studies found that 6 months administration of ginkgo resulted in significant improvements on the ADAS-cog( Reference Wang, Wang and Song 39 ). Importantly, this result was evinced when baseline risk was taken into account and might represent an important methodological consideration in AD research. In support of this, a separate review( Reference Janßen, Sturtz and Skipka 40 ) found that improvements seen in daily living, cognitive function and amelioration of neuropsychiatric symptoms (such as psychosis, agitation, aggression, anxiety, euphoria/dysphoria or disordered motor behaviour), in a review of six studies comprising 1800 participants with AD, were most striking in those suffering significant levels of neuropsychiatric symptoms; thus individual differences in risk levels and severity of symptoms likely has an impact on response to ginkgo and overall study findings; especially if small cohorts are utilised in individual trials.

Ginseng

Ginseng has a 5000-year history of medicinal consumption( Reference Yun 41 ) and comprises forty or more bioactive saponins (known as ginsenosides), which exert anti-fungal/viral/bacterial/feeding effects within the plant( Reference Osbourn 42 , Reference Sparg, Light and van Staden 43 ). Again, this terpene-derived nutritional supplement demonstrates efficacy in interacting with numerous physiological systems, including acting as an antioxidant, stimulating nitric oxide production and acting as a ligand for glucocorticoid and androgen receptors; interactions which, among others, are seen to increase immune function, enhance CNS function and prevent CVD and other diseases in animal models( Reference Lu, Yao and Chen 44 ). Specific neurocognitive interactions with neurotransmitter function and the processes of neurogenesis and long-term potentiation are also observed to exert anti-stress, antidepressant, and anxiolytic effects, to moderate fatigue and improve memory in impaired rodents( Reference Attele, Wu and Yuan 45 , Reference Dang, Chen and Liu 46 ).

Research in young healthy participants is still in its infancy and buoyed by heterogeneous methodology but, on the whole, provides promise in terms of cognitive enhancement( Reference Kennedy, Scholey and Wesnes 47 Reference Reay, Scholey and Kennedy 50 ). In vitro and animal data supports the potential for ginseng to be of specific benefit to AD-induced cognitive decline where ginsenosides have been observed to minimise the inhibitory effect of amyloid-β protein on cholinergic transmission( Reference Lee, Shiao and Chen 51 ) and, in turn, prevent the resulting amnesiac effects in rats( Reference Wang, Wang and Ng 52 ). To the best of current knowledge, however, only two trials exist, which investigate whether these cognitive benefits also extend to AD in human subjects. The first of these reports on the 12-week consumption of 9 g/d Korean ginseng in fifteen patients with dementia where scores on the ADAS-cog and clinical dementia rating were significantly improved( Reference Heo, Lee and Chu 53 ). The second trial is a follow-up of patients in this same trial after 24 weeks where a significant improvement on the Korean Mini Mental State Exam was evinced following 4·5 and 9 g/d ginseng and maintained at 48 and 96 weeks( Reference Heo, Lee and Oh 54 ).

Melissa officinalis (lemon balm)

Melissa is another terpene with a centuries-long history for treating disorders, which modern research has confirmed efficacy for; including as a memory and mood enhancer( Reference Kennedy and Scholey 55 ). The bioactives underpinning these effects include monoterpenes and sesquiterpenes; which include 1, 8 cineole( Reference Tittel, Wagner and Bos 56 ), and the CNS-relevant effects of these compounds includes antioxidant activity( Reference Ferreira, Proença and Serralheiro 57 , Reference Pereira, Fachinetto and de Souza Prestes 58 ), activation of the cholinergic system (including cholinesterase inhibition)( Reference Ferreira, Proença and Serralheiro 57 , Reference Perry, Court and Bidet 59 Reference Dastmalchi, Ollilainen and Lackman 61 ) and up-regulation of γ-aminobutyric acid (GABA) ergic neurons( Reference Awad, Muhammad and Durst 62 ).

These interactions would suggest benefit to AD sufferers and, indeed, one of the only two controlled trails, which has investigated Melissa here observed reduced agitation and improved cognitive (ADAS-cog) and behavioural function (as assessed by the Cognitive Drug Research test battery) following 16 weeks administration of an alcoholic-Melissa tincture in a group of mild–moderate sufferers( Reference Akhondzadeh, Noroozian and Mohammadi 63 ). The other of the two studies, however, failed to find statistically significant differences in AD symptoms with Melissa( Reference Ballard, O'Brien and Reichelt 64 ). This study, though, administered Melissa in the form of an aromatherapy spray (dispersed once in the morning and afternoon in patient rooms) or essential oil hand massage (with a third group receiving a combination), which also contained lavender. This novel approach to administration presents an unknown quantity in terms of subsequent plasma levels of Melissa and time needed for the bioactives to reach the CNS and, as such, makes it difficult to compare with the above study and related studies, which administer phytochemicals orally. It could also be the case that the alcoholic matrix in the initial study in some way enhanced, or indeed was solely responsible for, the significant effects seen there. Nevertheless, it is important to note that the latter study did observe clinical benefit to some participants and this may indicate the very important role of individual differences in response to terpene phytochemicals; a consideration also noted with ginkgo studies described earlier. Here too it may be the case that pre-AD differences and current symptom severity influence the role that terpenes play and, with the Melissa essential oil study specifically, it could be that the response to scent (including lavender; which contains the active terpene linalool) and the pleasant sensation of being massaged, interact to produce effects which are of benefit to some and not others.

Salvia lavandulaefolia and Salvia officinalis (sage)

Sage has a history stretching back as far as the ancient Greeks where it was used as a cognitive enhancer and to prevent age-related decline; hence the derivation of the word sage in relation to wisdom. The two most abundant bioactive monoterpenes in sage are 1, 8 cineole and camphor and, of interest here, these monoterpenes have demonstrated potent cholinesterase-inhibiting properties( Reference Savelev, Okello and Perry 65 Reference Perry, Houghton and Jenner 68 ); with 1, 8 cineole alone evincing the greatest effects( Reference Savelev, Okello and Perry 65 ). These CNS effects produce enhanced secondary memory, accuracy and attention in healthy aged (over 65 years) participants( Reference Scholey, Tildesley and Ballard 69 ) and consumption of this terpene, in the form of an essential oil, is reportedly well tolerated in a small group (n 11) of patients aged 76–95 years with mild–moderate AD following 6 weeks of 50–150 µl daily consumption of S. officinalis ( Reference Perry, Bollen and Perry 70 ). The latter study did not observe any statistically significant cognitive benefit but this was not the a priori aim of the study and this is reflected in the sample size. Nevertheless the authors do report positive indications on the cognitive test battery used (Cognitive Drug Research) and this is in line with the only other trial investigating the benefit of sage in AD( Reference Akhondzadeh, Noroozian and Mohammadi 71 ). Here nineteen participants (65–80 years), with mild–moderate AD, consumed an S. officinalis-alcoholic tincture for 16 weeks and better outcomes on the ADAS-cog, compared with the placebo controls, was observed. This study also demonstrated a trend towards reduced agitation in the S. officinalis group.

Potential benefit of phenolic phytochemicals against Alzheimer's disease

Currently about 10 000 compounds have been classified as polyphenols and this large class comprises both flavonoid and non-flavonoid forms. The former comprise the largest grouping and these can be further sub-divided into isoflavones (found in soya and soya products), flavones (found, for example, in sweet pepper), flavanones (found in citrus fruits), flavanols (which can be further sub-categorised into flavan-3-ols (found in tea) and proanthocyanidins (found in fruits)), flavonols (fruits and vegetables; specifically onions) and anthocyanins (specifically found in berries)( Reference Scalbert and Williamson 72 ).

Epidemiological data have established links between the consumption of polyphenol-rich diets, and specific polyphenols, and reduced incidence of AD in human populations. Consumption of fruits and vegetables and total levels of flavonoids are associated with protection against, or slowed progression of, AD and other dementias( Reference Barberger-Gateau, Raffaitin and Letenneur 73 Reference Letenneur, Proust-Lima and Le Gouge 75 ). Large cohort studies have also evidenced links between neurocognitive protection (as indexed in all cases by scores on the Mini Mental State Exam) and tea consumption in elderly cohorts( Reference Ng, Feng and Niti 76 , Reference Kuriyama, Hozawa and Ohmori 77 ) as well as chocolate and red wine( Reference Nurk, Refsum and Drevon 78 ).

Resveratrol

Resveratrol derives from a sub-class of non-flavonoid polyphenols termed stilbenes and is found in limited sources, which include grapes and, as a result, wine. Resveratrol has received much research attention regarding its potential to benefit a number of disease states, including CVD( Reference Zordoky, Robertson and Dyck 79 ), cancer( Reference Carter, D'Orazio and Pearson 80 ) and even life extension in a range of animal models( Reference Hector, Lagisz and Nakagawa 81 ). The many and varied health effects attributed to resveratrol are likely underpinned by the multifarious biological targets that it interacts with. These include, but are not limited to, cyclooxygenase 1 and 2, the sirtuins and various kinases and DNA/RNA and lipoproteins. These specific interactions explain how resveratrol is able to exert anti-inflammatory effects, to interact directly with cell signalling and its links to cardiovascular health, respectively( Reference Britton, Kovoor and Brown 82 ). Interaction with these targets, and others such as up-regulation of cerebral blood flow ( Reference Kennedy, Wightman and Reay 6 , Reference Wightman, Reay and Haskell 7 ), and the ability of resveratrol to attenuate amyloid-β-induced cell death in vitro ( Reference Han, Zheng and Bastianetto 83 ), suggests that this polyphenol should be capable of beneficial therapeutic potential in AD. Indeed, results from animal models supports the function of resveratrol here with reduced markers of pathology, e.g. amyloid-β plaques( Reference Karuppagounder, Pinto and Xu 84 ), and behavioural deficits, e.g. improved learning and memory( Reference Ma, Sun and Liu 85 ), in response to resveratrol exposure and consumption (25 mg/kg per d) of resveratrol, respectively.

However, to the best of current knowledge, only one study exists, which investigates resveratrol in human volunteers with AD. Here a phase-2 randomised, placebo-controlled, double-blind 12-month trial of 500 mg/d (escalating to 1000 mg twice daily) resveratrol was conducted in participants with mild–moderate AD( Reference Turner, Thomas and Craft 86 ). Unfortunately the therapeutic measures of the present study were limited and, whilst amyloid-β markers were reduced by resveratrol, this was not more significant than in the placebo group, and brain volume loss was not attenuated. Resveratrol consumption was generally well tolerated, but participants did report significant gastrointestinal problems and weight loss, which is likely due to the high dose being received after escalation as these side effects are not seen often in the literature with doses at or lower than 500 mg.

Conclusions

This review began with the assertion that our current alkaloid phytochemical-derived AD pharmaceutical treatments, such as galantamine and rivastigmine, produce unpleasant side effects and, ultimately, target only one of the multifactorial deficits of this progressive neurocognitive disorder. Attenuating cholinergic decline is arguably the most important and easily influenced of the AD deficits, within our current capabilities, but this report argued that the terpene and phenolic groupings of plant phytochemicals might offer an equally efficacious and safer alternative for AD drugs, relative to the alkaloids, which also target multiple deficits.

The terpene and phenolic studies presented here are few and a clear, overall view is hindered by heterogeneous trials where sample size, method of assessment, trial length, route of administration and individual differences associated with pre-AD status and current severity of symptoms vary or are not considered. Another area which future studies should focus, and something which resonated from several talks at the Nutrition Society spring conference, is the concept of ‘responders’ and ‘non-responders’ in phytochemical research. These terms refer to individuals who experience an anticipated pharmacokinetic response to consumption of drugs, and those who do not, respectively; with this phenomenon based on a whole host of known and unknown factors. This likely includes the speed of gut transit, the microbiotic profile of the gastrointestinal tract and the functionality of efflux pumps, alongside multiple other variables, and these factors will be unique to each participant. It is likely that the impact of these individual differences will be diluted in large cohorts but, apart from the meta-analyses discussed, one common factor across terpene and phenolic research trials is relatively small sample sizes. Studies with these phytochemicals undoubtedly hold promise but robust and replicable outcomes will not be evinced until the above methodological constraints are addressed.

Acknowledgements

I would like to thank Professor David Kennedy for his help in preparing the Nutrition Society spring conference presentation of the same title and, therefore, this manuscript.

Financial Support

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

Conflict of Interest

None.

Authorship

Dr E Wightman devised and investigated this research question and takes sole ownership of the written content.

References

1. Haskell, CF, Kennedy, DO, Wesnes, KA et al. (2005) Cognitive and mood improvements of caffeine in habitual consumers and habitual non-consumers of caffeine. Psychopharmacology 179, 813825.Google Scholar
2. Moss, M, Scholey, A, Wesnes, K (1998) Oxygen administration selectively enhances cognitive performance in healthy young adults: a placebo-controlled double-blind crossover study. Psychopharmacology (Berl) 138, 2733.Google Scholar
3. Scholey, A, Moss, M, Neave, N et al. (1999) Cognitive performance, hyperoxia, and heart rate following oxygen administration in healthy young adults. Physiol Behav 67, 783789.Google Scholar
4. Scholey, A, Laing, S, Kennedy, D (2006) Blood glucose changes and memory: effects of manipulating emotionality and mental effort. Biol Psychol 71, 1219.Google Scholar
5. Kennedy, D, Stevenson, E, Jackson, P 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, 1.Google Scholar
6. Kennedy, D, Wightman, EL, Reay, JL et al. (2010) Effects of resveratrol on cerebral blood flow variables and cognitive performance in humans: a double-blind, placebo-controlled, crossover investigation. Am J Clin Nutr 91, 15901597.Google Scholar
7. Wightman, E, Reay, J, Haskell, C et al. (2014) Effects of resveratrol alone or in combination with piperine on cerebral blood flow parameters and cognitive performance in humans: a randomised, double-blind, placebo-controlled, crossover investigation. Br J Nutr 112, 203213.Google Scholar
8. Wightman, E, Haskell-Ramsay, C, Reay, J et al. (2015) The effects of chronic trans-resveratrol supplementation on aspects of cognitive function, mood, sleep, health and cerebral blood flow in healthy, young humans. Br J Nutr 114, 14271437.Google Scholar
9. Rönnlund, M, Nyberg, L, Bäckman, L et al. (2005) Stability, growth, and decline in adult life span development of declarative memory: cross-sectional and longitudinal data from a population-based study. Psychol Aging 20, 318.Google Scholar
10. Association As (2016) 2016 Alzheimer's disease facts and figures. Alzheimer's Demen 12, 459509.Google Scholar
11. International AsD (2015) World Alzheimer Report 2015: The Global Impact of Dementia. An Analysis of Prevalence, Incidence, Cost and Trends. Available at www.alz.co.uk/research/world-report-2015 (Accessed February 2016).Google Scholar
12. Karch, CM, Goate, AM (2015) Alzheimer's disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry 77, 4351.Google Scholar
13. Luchsinger, JA, Mayeux, R (2004) Dietary factors and Alzheimer's disease. Lancet Neurol 3, 579587.Google Scholar
14. Lee, P (1994) Smoking and Alzheimer's disease: a review of the epidemiological evidence. Neuroepidemiology 13, 131144.Google Scholar
15. Durazzo, TC, Mattsson, N, Weiner, MW et al. (2014) Smoking and increased Alzheimer's disease risk: a review of potential mechanisms. Alzheimer's Demen 10, S122S145.Google Scholar
16. Mukamal, KJ, Kuller, LH, Fitzpatrick, AL et al. (2003) Prospective study of alcohol consumption and risk of dementia in older adults. JAMA 289, 14051413.Google Scholar
17. Markesbery, WR (1997) Oxidative stress hypothesis in Alzheimer's disease. Free Radic Biol Med 23, 134147.Google Scholar
18. Vignini, A, Giulietti, A, Nanetti, L et al. (2013) Alzheimer's disease and diabetes: new insights and unifying therapies. Curr Diab Rev 9, 218227.Google Scholar
19. Kalaria, RN (2000) The role of cerebral ischemia in Alzheimer's disease. Neurobiol Aging 21, 321330.Google Scholar
20. Matrone, C, Djelloul, M, Taglialatela, G et al. (2015) Inflammatory risk factors and pathologies promoting Alzheimer's disease progression: is RAGE the key. Histol Histopathol 30, 125139.Google Scholar
21. Adlard, PA, Bush, AI (2006) Metals and Alzheimer's disease. J Alzheimer's Dis 10, 145163.Google Scholar
22. Vest, RS, Pike, CJ (2013) Gender, sex steroid hormones, and Alzheimer's disease. Horm Behav 63, 301307.Google Scholar
23. Sattler, C, Toro, P, Schönknecht, P et al. (2012) Cognitive activity, education and socioeconomic status as preventive factors for mild cognitive impairment and Alzheimer's disease. Psychiatry Res 196, 9095.Google Scholar
24. Mufson, EJ, Counts, SE, Perez, SE et al. (2008) Cholinergic system during the progression of Alzheimer's disease: therapeutic implications. Expert Rev Neurother 8, 17031718.Google Scholar
25. NICE (2009) Donepezil, Galantamine, Rivastigmine (review) and Memantine for the Treatment of Alzheimer's Disease. UK: National Institute of Clinical Excellence.Google Scholar
26. Birks, J, Craig, D (2006) No Consistent Evidence of Efficacy of Galantamine in Vascular Cognitive Impairment. Health. Cochrane library. Available at onlinelibrary.wiley.com/doi/10.1002/14651858.CD004746.pub2full (Accessed February 2016).Google Scholar
27. Society As (2016) Treating behavioural and psychological symptoms of dementia. https://www.alzheimers.org.uk/site/scripts/documents_info.php?documentID=1191&pageNumber=2 (accessed May 2016).Google Scholar
28. Kennedy, D, Wightman, E (2011) Herbal extracts and phytochemicals: plant secondary metabolites and the enhancement of human brain function. Adv Nutr: Int Rev J 2, 3250.Google Scholar
29. Fehske, CJ, Leuner, K, Müller, WE (2009) Ginkgo biloba extract (EGb761®) influences monoaminergic neurotransmission via inhibition of NE uptake, but not MAO activity after chronic treatment. Pharmacol Res 60, 6873.Google Scholar
30. Chan, PC, Xia, QS, Fu, PP (2007) Ginkgo biloba leave extract: biological, medicinal, and toxicological effects. J Environ Sci Health C: Environ Carcinog Ecotoxicol Rev 25, 211244.Google Scholar
31. DeFeudis, FV, Drieu, K (2004) “Stress-alleviating” and “vigilance-enhancing” actions of Ginkgo biloba extract (EGb 761). Drug Dev Res 62, 125.Google Scholar
32. Berger, P (2001) Ginkgo leaf extract. Med Herbal. 2, 56.Google Scholar
33. Stackman, RW, Eckenstein, F, Frei, B et al. (2003) Prevention of age-related spatial memory deficits in a transgenic mouse model of Alzheimer's disease by chronic Ginkgo biloba treatment. Exp Neurol 184, 510520.Google Scholar
34. Smith, JV, Luo, Y (2003) Elevation of oxidative free radicals in Alzheimer's disease models can be attenuated by Ginkgo biloba extract EGb 761. J Alzheimer's Dis 5, 287300.Google Scholar
35. Birks, J, Grimley Evans, J (2002) Ginkgo biloba for cognitive impairment and dementia. The Cochrane Library. Available at onlinelibrary.wiley.com/doi/10.1002/14651858.CD003120 (Accessed February 2016).Google Scholar
36. Birks, J, Evans, JG (2009) Ginkgo biloba for cognitive impairment and dementia. Cochrane Database Syst Rev. Available at onlinelibrary.wiley.com/doi/10.1002/14651858.CD003120.PUB3/FULL (Accessed February 2016).Google Scholar
37. Snitz, BE, O'Meara, ES, Carlson, MC et al. (2009) Ginkgo biloba for preventing cognitive decline in older adults: a randomized trial. JAMA 302, 2663.Google Scholar
38. Weinmann, S, Roll, S, Schwarzbach, C et al. (2010) Effects of Ginkgo biloba in dementia: systematic review and meta-analysis. BMC Geriatr 10, 1.Google Scholar
39. Wang, B, Wang, H, Song, Y et al. (2010) Effectiveness of standardized ginkgo biloba extract on cognitive symptoms of dementia with a six-month treatment: a bivariate random effect meta-analysis. Pharmacopsychiatry 43, 8691.Google Scholar
40. Janßen, IM, Sturtz, S, Skipka, G et al. (2010) Ginkgo biloba in Alzheimer's disease: a systematic review. Wiener Medizinische Wochenschrift 160, 539546.Google Scholar
41. Yun, TK (2001) Brief introduction of Panax ginseng CA Meyer. J Korean Med Sci 16, 35.Google Scholar
42. Osbourn, A (1996) Saponins and plant defence – a soap story. Trends Plant Sci 1, 49.Google Scholar
43. Sparg, SG, Light, ME, van Staden, J (2004) Biological activities and distribution of plant saponins. J Ethnopharmacol 94, 219243.Google Scholar
44. Lu, JM, Yao, QZ, Chen, CY (2009) Ginseng compounds: an update on their molecular mechanisms and medical applications. Curr Vasc Pharmacol 7, 293302.Google Scholar
45. Attele, AS, Wu, JA, Yuan, C-S (1999) Ginseng pharmacology: multiple constituents and multiple actions. Biochem Pharmacol 58, 16851693.Google Scholar
46. Dang, HX, Chen, Y, Liu, XM et al. (2009) Antidepressant effects of ginseng total saponins in the forced swimming test and chronic mild stress models of depression. Progr Neuro-Psychopharmacol Biol Psychiatry 33, 14171424.Google Scholar
47. Kennedy, D, Scholey, AB, Wesnes, KA (2001) Differential, dose dependent changes in cognitive performance following acute administration of a Ginkgo biloba/Panax ginseng combination to healthy young volunteers. Nutr Neurosci 4, 399412.Google Scholar
48. Kennedy, D, Scholey, AB, Wesnes, KA (2002) Modulation of cognition and mood following administration of single doses of Ginkgo biloba, ginseng, and a ginkgo/ginseng combination to healthy young adults. Physiol Behav 75, 739751.Google Scholar
49. Reay, JL, Kennedy, DO, Scholey, AB (2005) Single doses of Panax ginseng (G115) reduce blood glucose levels and improve cognitive performance during sustained mental activity. J Psychopharmacol 19, 357365.Google Scholar
50. Reay, JL, Scholey, AB, Kennedy, DO (2010) Panax ginseng (G115) improves aspects of working memory performance and subjective ratings of calmness in healthy young adults. Human Psychopharmacol: Clin Exp 25, 462471.Google Scholar
51. Lee, T, Shiao, Y-J, Chen, C-F et al. (2001) Effect of ginseng saponins on b-amyloid-suppressed acetylcholine release from rat hippocampal slices. Planta Med 67, 634637.Google Scholar
52. Wang, LC, Wang, B, Ng, S-Y et al. (2006) Effects of ginseng saponins on β-amyloid-induced amnesia in rats. J Ethnopharmacol 103, 103108.Google Scholar
53. Heo, JH, Lee, ST, Chu, K et al. (2008) An open-label trial of Korean red ginseng as an adjuvant treatment for cognitive impairment in patients with Alzheimer's disease. Eur J Neurol 15, 865868.Google Scholar
54. Heo, J-H, Lee, S-T, Oh, M-J et al. (2011) Improvement of cognitive deficit in Alzheimer's disease patients by long term treatment with Korean red ginseng. J Ginseng Res 35, 457461.Google Scholar
55. Kennedy, D, Scholey, AB (2006) The psychopharmacology of European herbs with cognition-enhancing properties. Curr Pharm Des 12, 46134623.Google Scholar
56. Tittel, G, Wagner, H, Bos, R (1982) Chemical-composition of the essential oil from Melissa. Planta Med 46, 9198.Google Scholar
57. Ferreira, A, Proença, C, Serralheiro, M et al. (2006) The in vitro screening for acetylcholinesterase inhibition and antioxidant activity of medicinal plants from Portugal. J Ethnopharmacol 108, 3137.Google Scholar
58. Pereira, RP, Fachinetto, R, de Souza Prestes, A et al. (2009) Antioxidant effects of different extracts from Melissa officinalis, Matricaria recutita and Cymbopogon citratus . Neurochem Res 34, 973983.Google Scholar
59. Perry, N, Court, G, Bidet, N et al. (1996) European herbs with cholinergic activities: potential in dementia therapy. Int J Geriatr Psychiatry 11, 10631069.3.0.CO;2-1>CrossRefGoogle Scholar
60. Wake, G (2000) CNS acetylcholine receptor activity in European medicinal plants traditionally used to improve failing memory. J Ethnopharmacol 69, 105114.Google Scholar
61. Dastmalchi, K, Ollilainen, V, Lackman, P et al. (2009) Acetylcholinesterase inhibitory guided fractionation of Melissa officinalis L. Bioorg Med Chem 17, 867871.Google Scholar
62. Awad, R, Muhammad, A, Durst, T et al. (2009) Bioassay-guided fractionation of Lemon balm (Melissa officinalis L.) using an in Vitro measure of GABA transaminase activity. Phytother Res 23, 10751081.Google Scholar
63. Akhondzadeh, S, Noroozian, M, Mohammadi, M et al. (2003) Melissa officinalis extract in the treatment of patients with mild to moderate Alzheimer's disease: a double blind, randomised, placebo controlled trial. J Neurol Neurosurg Psychiatry 74, 863866.Google Scholar
64. Ballard, CG, O'Brien, JT, Reichelt, K et al. (2002) Aromatherapy as a safe and effective treatment for the management of agitation in severe dementia: the results of a double-blind, placebo-controlled trial with Melissa. J Clin Psychiatry 63, 553558.Google Scholar
65. Savelev, S, Okello, E, Perry, N et al. (2003) Synergistic and antagonistic interactions of anticholinesterase terpenoids in Salvia lavandulaefolia essential oil. Pharmacol Biochem Behav 75, 661668.Google Scholar
66. Savelev, S, Okello, E, Perry, E (2004) Butyryl- and acetyl-cholinesterase inhibitory activities in essential oils of Salvia species and their constituents. Phytother Res 18, 315324.Google Scholar
67. Perry, N, Houghton, P, Theobald, A et al. (2000) In-vitro inhibition of human erythrocyte acetylcholinesterase by Salvia lavandulaefolia essential oil and constituent terpenes. J Pharm Pharmacol 52, 895902.Google Scholar
68. Perry, N, Houghton, P, Jenner, P et al. (2002) Salvia lavandulaefolia essential oil inhibits cholinesterase in vivo . Phytomed: Int J Phytother Phytopharmacol 9, 4851.Google Scholar
69. Scholey, A, Tildesley, N, Ballard, C et al. (2008) An extract of Salvia (sage) with anticholinesterase properties improves memory and attention in healthy older volunteers. Psychopharmacology 198, 127139.Google Scholar
70. Perry, N, Bollen, C, Perry, E et al. (2003) Salvia for dementia therapy: review of pharmacological activity and pilot tolerability clinical trial. Pharmacol Biochem Behav 75, 651659.Google Scholar
71. Akhondzadeh, S, Noroozian, M, Mohammadi, M et al. (2003) Salvia officinalis extract in the treatment of patients with mild to moderate Alzheimer's disease: a double blind, randomized and placebo-controlled trial. J Clin Pharm Ther 28, 5359.Google Scholar
72. Scalbert, A, Williamson, G (2000) Dietary intake and bioavailability of polyphenols. J Nutr 130, 2073S2085S.Google Scholar
73. Barberger-Gateau, P, Raffaitin, C, Letenneur, L et al. (2007) Dietary patterns and risk of dementia the three-city cohort study. Neurology 69, 19211930.Google Scholar
74. Commenges, D, Scotet, V, Renaud, S et al. (2000) Intake of flavonoids and risk of dementia. Eur J Epidemiol 16, 357363.Google Scholar
75. 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.Google Scholar
76. Ng, T, Feng, L, Niti, M et al. (2008) Tea consumption and cognitive impairment and decline in older Chinese adults. Am J Clin Nutr 88, 224231.Google Scholar
77. Kuriyama, S, Hozawa, A, Ohmori, K et al. (2006) Green tea consumption and cognitive function: a cross-sectional study from the Tsurugaya Project 1. Am J Clin Nutr 83, 355.Google Scholar
78. 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.Google Scholar
79. Zordoky, BN, Robertson, IM, Dyck, JR (2015) Preclinical and clinical evidence for the role of resveratrol in the treatment of cardiovascular diseases. Biochim Biophys Acta 1852, 11551177.Google Scholar
80. Carter, LG, D'Orazio, JA, Pearson, KJ (2014) Resveratrol and cancer: focus on in vivo evidence. Endocr-Relat Cancer 21, R209R225.Google Scholar
81. Hector, KL, Lagisz, M, Nakagawa, S (2012) The effect of resveratrol on longevity across species: a meta-analysis. Biol Lett 8, 790793.Google Scholar
82. Britton, RG, Kovoor, C, Brown, K (2015) Direct molecular targets of resveratrol: identifying key interactions to unlock complex mechanisms. Ann N Y Acad Sci 1348, 124133.Google Scholar
83. Han, YS, Zheng, WH, Bastianetto, S et al. (2004) Neuroprotective effects of resveratrol against β-amyloid-induced neurotoxicity in rat hippocampal neurons: involvement of protein kinase C. Br J Pharmacol 141, 9971005.Google Scholar
84. Karuppagounder, SS, Pinto, JT, Xu, H et al. (2009) Dietary supplementation with resveratrol reduces plaque pathology in a transgenic model of Alzheimer's disease. Neurochem Int 54, 111118.Google Scholar
85. Ma, X, Sun, Z, Liu, Y et al. (2013) Resveratrol improves cognition and reduces oxidative stress in rats with vascular dementia. Neural Regen Res 8, 2050.Google Scholar
86. Turner, R, Thomas, R, Craft, S et al. (2015) Resveratrol is safe and well-tolerated in individuals with mild–moderate dementia due to Alzheimer's disease. Neurology 84, Supplement 33009.Google Scholar