Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jul 7.
Published in final edited form as: Nat Rev Neurosci. 2012 Jan 18;13(3):209–216. doi: 10.1038/nrn3151

Recruiting Adaptive Cellular Stress Responses for Successful Brain Aging

Alexis M Stranahan 1, Mark P Mattson 2,*
PMCID: PMC4084510  NIHMSID: NIHMS596597  PMID: 22251954

Abstract

Successful aging is determined in part by genetic background, but also by experiential factors associated with lifestyle and culture. Dietary, behavioral and pharmacological interventions have been identified as potential means to slow brain aging and forestall neurodegenerative disease. Many of these interventions recruit adaptive cellular stress responses to strengthen neuronal networks and enhance plasticity. In this Perspective, we describe several determinants of healthy and pathological brain aging, with insights into how these processes are accelerated or prevented. We also describe the mechanisms underlying the neuroprotective actions of exercise and nutritional interventions, with the goal of recruiting these molecular targets for the treatment and prevention of neurodegenerative disease.

Introduction

Aging is associated with numerous physiological alterations across multiple organ systems, including the brain. Central nervous system (CNS) consequences of aging occur in part due to intrinsic responses to accumulated ‘wear and tear’, but may also be secondary to aging in other organ systems, including reproductive aging1 (Behl, 2002) and muscle loss2 (Burns et al., 2010). Some changes that occur in the context of aging are adaptive. For example, despite increased physical ailments, the elderly self-report higher levels of happiness relative to middle-aged and younger populations3, and presumably, the increased rate of contentment in older adults has some neuronal correlate. Other age-related alterations, such as declining executive function and motor impairment, are more pernicious; ideally, these changes could be delayed or prevented by targeted CNS interventions. Many of these interventions, including exercise and dietary moderation, resemble the dictates of common sense. In this article we describe mechanisms underlying neuroprotective ‘common sense’ manipulations, with the goal of harnessing these mechanisms to protect against the deleterious consequences of brain aging.

Salient features of brain aging

The aging process leads to an increase in the variability associated with cognitive and motor capabilities in humans4, and rodent models5. Just as some humans maintain cognitive function into their eighth decade and beyond, a subset of aged rodents retains the capacity for performance across a range of cognitive tasks57. In this regard, it is possible to distinguish between healthy aging with preserved cognition, age-related cognitive impairment, and neuropathology (Figure 1). As described in the remaining sections of this article, these general functional features of brain aging are underlain by impaired adaptive cellular stress responses, defined as the ability of neurons to rebound from potentially damaging alterations of the local environment and the endocrine milieu.

Figure 1. Intrinsic features of normal and pathological aging.

Figure 1

Normal aging is accompanied by alterations in neuronal calcium handling and changes in lipid peroxidation, leading to increased generation of reactive oxygen species and damage to mitochondria. These changes are permissive or instructive for the suppression of adult neurogenesis beginning in middle age. Successful aging is characterized by implementation of alternative plasticity mechanisms to compensate for changes in the local microenvironment. Age-related pathologies such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and Huntington’s disease (HD) arise from a combination of genetic and environmental factors, but each disease shares a common feature in that age is a risk factor for disease onset. In this respect, aging sets the stage for the onset of pathology. REST, repressor element 1-silencing transcription factor; ROS, reactive oxygen species, P, phosphorylation site; Aβ, amyloid β-peptide; Ca, calcium; BDNF, brain-derived neurotrophic factor; DJ1, Parkinson protein 7.

Regardless of genetic background, there are several major cellular and molecular changes that occur in the CNS during normal aging that are shared, more-or-less, with age-related alterations in other organ systems. These fundamental aspects of brain aging, common across the spectrum of cognitive performance include: increased oxidative stress; impaired cellular energy metabolism; perturbed cellular calcium signaling; and the abnormal accumulation of damaged proteins and organelles8. Superoxide anion radical, hydroxyl radical, nitric oxide and peroxynitrite are major oxygen free radicals that have been implicated in normal brain aging and cognitive impairment9. Protein oxidation and modification by lipid peroxidation products and peroxynitrite increases progressively during aging, and is particularly prominent in the neurons that degenerate in Alzheimer’s disease (AD). Likewise, normal aging was associated with increased immunoreactivity for indices of oxidative stress in humans10, and pathological aging further exacerbated this effect11. Oxidative damage to membrane lipids occurs more severely in aged, cognitively impaired animals12, and to an even greater extent in experimental models of neurodegenerative diseases1314. For example, membrane-associated oxidative stress results in neuronal dysfunction and degeneration by a mechanism involving the lipid peroxidation product 4-hydroxynonenal which covalently modifies proteins critical for cellular ion homeostasis including ion (Na+ and Ca2+) motive ATPases, and glucose and glutamate transport proteins15. Such aberrant oxidative damage likely contributes to an excitatory imbalance that may presage the onset of age-related cognitive impairment and AD16.

With advancing age neurons may suffer from reduced production of the molecular energy couriers ATP and NAD+, with impaired mitochondrial function being a principle reason for such a cellular energy deficit17. Changes that lead to cellular energy deficits include oxidative damage to mitochondrial DNA and electron transport chain proteins18, and the plasma membrane redox system19. Disease-specific factors may also compromise energy metabolism including Aβ in AD, α-synuclein in Parkinson’s disease (PD), huntingtin in Huntington’s disease (HD) and Cu/Zn-SOD in amyotrophic lateral sclerosis (ALS)17. Aging is also accompanied by accumulation of damaged proteins, nucleic acids and organelles2021. Deficits in proteasomal degradation and molecular repair processes are increasingly recognized as central to age-related CNS dysfunction, particularly following physiological challenges to homeostasis2223. As with oxidative stress, cellular energy deficits and accumulation of damaged molecules and organelles tips the balance of activity in neuronal circuits towards uncontrolled excitation24.

Dietary energy intake has a major impact on brain aging

Studies of human subjects and animal models support a bidirectional relationship between energy intake and brain function; excessive intake impairs function, while dietary energy restriction enhances function25. The underlying mechanisms for this relationship converge on changes in synaptic plasticity and neurogenesis. Diabetes and obesity are common consequences of excessive energy intake that accelerate brain aging by compromising hippocampal neurogenesis and long-term potentiation (LTP), a candidate cellular model for the synaptic changes underlying learning and memory26. Excessive caloric intake also increases the risk for neurodegenerative diseases and stroke, possibly by compromising the integrity of the blood-brain barrier27 resulting in deficient neurovascular coupling. Through these mechanisms and others, conditions arising from excessive energy intake have an impact on brain regions implicated in various facets of cognition.

Perturbation of the levels and rhythmicity of a number of hormones in the context of obesity and the metabolic syndrome (Box 1) leads to maladaptive alterations in circuit function that are not restricted to regions traditionally associated with feeding and metabolism. Ghrelin, a hormone produced in the stomach, is one example of a metabolically-relevant peptide that influences cognition and neuroplasticity. Ghrelin acts on the hypothalamic arcuate nucleus to stimulate feeding and promote deposition of adipose tissue28. Ghrelin administration enhances hippocampal synaptic density, while loss-of-function results in decreased synapse numbers and impaired Schaffer collateral LTP29. Synaptic compromise is associated with behavioral deficits in learning and memory following targeted disruption of the ghrelin gene29. In addition to regulating synaptic density, LTP and learning, ghrelin also promotes adult neurogenesis in rats30. These findings support a role for ghrelin in hippocampal plasticity, and open the possibility that disruption of ghrelin signaling in the context of obesity could lead to impairment of hippocampal function.

Box 1: Metabolic syndrome.

The metabolic syndrome is a constellation of risk factors that increase the probability of developing diabetes, stroke and cardiovascular diseases. Individuals with the metabolic syndrome have: a high waist-to-hip ratio indicative of central adiposity; elevated fasting glucose levels that, while they may not meet the criteria for type 2 diabetes, indicate that insulin sensitivity may be compromised; elevated triglycerides; and increased blood pressure. The most effective treatment for the metabolic syndrome is weight loss. The endocrine consequences of the metabolic syndrome are not restricted to insulin signaling, but also involve perturbation of leptin levels and sensitivity, changes in corticosteroid synthesis and rhythmicity, and alterations in ghrelin secretion. Because these endocrine factors have each been shown to influence neuronal function26, 7576, particularly in the hippocampus, it is likely that the metabolic syndrome could accelerate age-related cognitive deficits. Indeed, recent studies in animal models and human subjects have demonstrated improvements in cognitive performance in response to interventions that increase insulin sensitivity including dietary energy restriction, exercise and pharmacological agents such as glucagon-like peptide 1 analogs7780.

Because pre-existing societal prejudice against obese individuals, studies on the mechanisms related to cognition in obesity should be interpreted cautiously. The perception of obesity as a condition arising from lack of discipline, rather than a disease that, like certain psychiatric conditions, arises from a mixture of environmental pressures and genetic predisposition, often facilitates negative bias among the general public, and could potentially impact opinions among the scientific and communities. It is also important to recognize that experimental observations in genetically homogeneous rodent populations may not always translate to humans. Given these precautions, it should be noted that, for both humans and nonhuman animal models, there is evidence for and against cognitive dysfunction in the context of obesity (Table 1).

Table 1.

Recent studies of cognitive function in relationship to obesity.

Human Studies Age group Cognitive outcome
Roriz-Cruz et al. (2007)87 Aged
Gunstad et al. (2007)88 Adult
van den Berg et al. (2007)89 Aged
Sturman et al. (2008)90 Aged
Li et al. (2008)91 Young
Huizinga et al (2008)92 Adult
Sabia et al., (2009)93 Adult
Volkow et al., (2009)94 Adult
Fergenbaum et al. (2009)95 Adult

Animal models

Morrison et al. (2010)36 Aged
McNay et al. (2010)35 Adult
Granholm et al. (2008)96 Adult
Kanoski et al. (2007)38 Adult
Mielke et al. (2006)39 Aged

The results of human studies reveal a critical window in which obesity, and the metabolic syndrome more generally, has the potential to impact cognitive aging. Exposure to the direct oxidative and indirect endocrine consequences of excessive energy intake during the fourth and fifth decade of life increases the likelihood of cognitive decline during later years31, while higher body mass indices may not cause cognitive impairment later in life32. One interpretation is that the middle-aged brain is most susceptible to the endocrine adversities of the metabolic syndrome, while another hypothesis would suggest that other factors, such as socioeconomic status, that are correlated with the prevalence of obesity, exert their effects on cognition during this same window. Because some, but not all studies examining cognitive changes in relationship to obesity control for external variables such as socioeconomic status and level of education, the degree to which obesity has an impact on cognition remains an open question.

Research in animal models demonstrates more consistent impairments on various measures associated with learning and memory in rodents exposed to excessive caloric intake. Diets high in fats and simple sugars impair spatial learning in the water maze3334, spatial recognition in the Y-maze35, complex maze learning36, and operant discrimination3738. However, the time course for memory impairment in rats fed western diets varies. Some studies report memory impairment after two to three months of diet exposure33, 35, 37, using diets that range from 20% fat to 45% fat; by contrast, another study did not detect cognitive impairment using a diet that was 45% fat until after eight months of diet exposure34. Other researchers have failed to detect any impairment at all using diets comparably enriched with fats and simple sugars in mice39. Understanding the time course, and underlying cellular and molecular mechanisms, of memory impairment caused by diets that elicit features of the metabolic syndrome is critical in light of evidence for a critical window in humans; however, the question of when, and how, cognitive impairment develops in the context of the metabolic syndrome remains unanswered.

Dietary energy restriction can protect the brain against aging. Early studies showed that lifelong energy restriction through an alternate day fasting regimen improves learning in a complex maze40. Caloric restriction also improves memory and Schaffer collateral LTP in young mice41, and enhances neurogenesis through a brain-derived neurotrophic factor (BDNF)-dependent mechanism42. Dietary moderation is also positively associated with successful aging in human populations25. Interestingly, age-related impairments in cellular energy metabolism can be reversed by dietary energy restriction19, as can pathological deficits in learning and memory in AD model mice4344. Dietary energy restriction was also effective in protecting dopaminergic neurons and improving motor function in mouse45 and monkey46 models of PD, and protected striatal and cortical neurons and extended survival in a mouse model of HD47. A major mechanism by which energy restriction protects neurons against aging and disease is by engaging adaptive cellular stress response pathways that result in the production of a range of cytoprotective proteins including neurotrophic factors, protein chaperones, DNA repair enzymes and mitochondrial uncoupling proteins, among others4850 (Figure 2).

Figure 2. Adaptive cellular stress response signaling mediates beneficial effects of environmental challenges on neuroplasticity and vulnerability to degeneration.

Figure 2

A typical glutamatergic neuron in the hippocampus is depicted receiving excitatory inputs (red) from neurons activated in response to exercise, cognitive challenges and dietary energy restriction. In the cell body of the postsynaptic neuron are depicted a mitochondrion (top), the endoplasmic reticulum (left) and the nucleus (right). Examples of seven different adaptive stress response signaling pathways that protect neurons against degeneration and promote synaptic plasticity are shown. During exercise and cognitive challenges postsynaptic receptors for glutamate (1, 2), serotonin (3) and acetylcholine (4) are activated to engage intracellular signaling cascades and transcription factors that induce the expression of neuroprotective proteins including BDNF, mitochondrial uncoupling proteins (UCPs) and anti-apoptotic proteins (e.g., Bcl2). BDNF promotes neuronal growth, in part, by activating mTOR. Mild cellular stress resulting from reduced energy substrates (5) and reactive oxygen species (ROS) (6) engage adaptive stress response pathways including those that up-regulate antioxidant enzymes (AOE) and protein chaperones. Release of γ-aminobutyric acid (GABA) from interneurons (7), in response to activity in excitatory circuits (as occurs during exercise and cognitive challenges), hyperpolarizes the excitatory neurons which can protect them from Ca2+ overload and excitotoxicity. BDNF, brain-derived neurotrophic factor; CREB, cyclic AMP response element-binding protein; DAG, diacylglycerol; HO1, heme oxygenase 1; HSF1, heat shock factor 1; MnSOD, manganese superoxide dismutase; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor κB; NQO1, NADH-quinone oxidoreductase 1; Nrf2, nuclear regulatory factor 2; Ph2E, phase 2 enzyme; PKC, protein kinase C; PMRS, plasma membrane redox system; SIRT, sirtuin.

Specific dietary components may influence brain aging

Epidemiological data from human populations and mechanistic data from animal models support a protective role for certain dietary phytochemicals51 and a deleterious effect of saturated fats and refined sugars52. Specific macro- and micro-nutrients counteract brain aging by protecting against impairment and promoting adaptive plasticity, or by acting in a permissive or instructive fashion for deficits in neuronal function. For example, regular consumption of refined sugars and saturated fats accelerates age-related deficits in spatial learning34, whereas consuming relatively high amounts of vegetables and fruits may counteract the aging process53. Emerging evidence suggests that some neuroprotective phytochemicals engage adaptive cellular stress response pathways in neurons to upregulate the expression of neurotrophic factors, antioxidant enzymes, and proteins involved in cellular energy metabolism51. In this regard, the biological framework for nutritional neuroprotection conforms to the theory of hormesis (Box 2). Phytochemicals that have been shown to protect neurons against dysfunction and degeneration in animal models of neurodegenerative disorders include folic acid54, thiamine55, curcumin56, epigallocatechin-3-gallate57, plumbagin58, and resveratrol59. Signaling pathways activated by these phytochemicals include the Nrf2 – antioxidant response element (ARE) pathway, the Ca2+ - cyclic AMP response element-binding protein (CREB) pathway, and the sirtuin – FOXO pathway (Figure 2). Neuroprotective genes induced by the latter pathways include those encoding neurotrophic factors (e.g., BDNF), protein chaperones (e.g., heat shock protein-70 and glucose-regulated protein-78), antioxidant enzymes (e.g., manganese superoxide dismutase, heme oxygenase-1 and NADH quinone oxidoreductase 1), energy-regulating proteins (e.g., mitochondrial uncoupling proteins), and DNA repair proteins (e.g., AP endonuclease 1). Based on these natural products, medicinal chemists may be able to develop therapeutic agents with greater specificity for a stress response pathway and reduced toxicity.

Box 2: Adaptive cellular stress response pathways promote successful brain aging through hormesis.

Intermittent exposure to mild cognitive or physiological stress increases neuronal resistance to the degenerative disorders of aging48. Hormesis – the process through which exposure to low levels of a stressor activates biological repair mechanisms81 - occurs during exercise, in response to dietary energy restriction and when neurons are stimulated with certain phytochemicals. Hormetic stress stimulates signaling pathways that enhance the abilities of neurons to resist oxidative stress, DNA damage, mitochondrial impairment, and protein misfolding and aggregation. Some of the major proteins involved in such adaptive stress responses include neurotrophic factors, protein chaperones, and proteins involved in mitochondrial biogenesis, protein degradation and autophagy (Figure 2). The cellular signal pathways that mediate hermetic responses in neurons are being identified. For example, cognitive stimulation and exercise involve the activation of glutamate receptors resulting in Ca2+ influx, Ca2+/calmodulin-dependent kinases, the transcription factor CREB (cyclic AMP response element binding protein) and induction of genes encoding BDNF and the DNA repair enzyme APE1, among others6061. Dietary energy restriction has been reported to activate genes encoding proteins involved in synaptic plasticity, cellular energy metabolism and cell survival82. It has been proposed that there is an evolutionarily conserved network of genes that orchestrate hormetic responses of cells and organisms to various stressful challenges. These so-called ‘vitagenes’ encode proteins critical for the control of protein quality, redox balance, ion homeostasis, membrane integrity and energy metabolism83. Examples of phytochemicals that act via hormesis are: sulforaphane (present in relatively high amounts in broccoli) and plumbagin which activate the Nrf2 – antioxidant response element (ARE) pathway resulting in the expression of the antioxidant enzymes HO-1 and NQO184; curcumin (curry spice) stimulates adult hippocampal neurogenesis by inducing a mild cellular stress response involving the extracellular signal-regulated kinases (ERKs)85; flavonoids from fruits such as grapes and blueberries enhance learning and memory, possibly by activating sirtuins and CREB86. A better understanding of the mechanisms by which phytochemicals stimulate/stress neurons in ways that enhance CNS function and resilience may lead to novel interventions for neurodegenerative disorders.

Physical and cognitive exercise

Evidence in favor of experiential neuroprotection is compelling, and emerging data suggest that lifestyle factors can be harnessed to prevent or treat neurological disease. Physical and mental activities can protect neurons against age-related dysfunction and disease, through a variety of mechanisms including up-regulation of neurotrophic factors, enhanced synaptic plasticity and stimulation of neurogenesis. Lifestyle factors such as cognitive stimulation60 and aerobic exercise61 engage a variety of signaling cascades to maintain transcriptional profiles that are more commonly observed in the structurally dynamic developing brain. Such lifestyle factors could, in principle, enhance the efficacy of pharmacological treatments designed to forestall neurodegenerative diseases.

One example of a synergistic interaction between exercise and pharmacological neuroprotection involves the neurotransmitter noradrenaline (Figure 3). Noradrenergic innervation of the hippocampus is necessary for exercise-induced upregulation of BDNF expression62. The locus coeruleus (LC) is the origin of noradrenergic fibers, and depletion of LC noradrenaline exacerbates amyloid plaque deposition and cognitive impairment in AD model mice63. Both of these studies used loss-of-function pharmacological approaches, but it remains to be seen whether synergistic gain-of-function could be detected through concomitant treatment of AD model mice with exercise and selective noradrenaline reuptake inhibitors (SNRIs; Figure 3).

Figure 3. Potential for mechanistic synergy between exercise and pharmacological treatments designed to maintain cognition in an aging population.

Figure 3

The locus coeruleus (LC) projection to the hippocampus is essential for exercise-induced brain-derived neurotrophic factor (BDNF) expression. Exercise enhances hippocampal BDNF synthesis, leading to activation of the extracellular signal-related kinase (ERK) pathway, which converges on the transcription factor cyclic adenosine monophosphate (cAMP) response element binding protein (CREB). Treatment with drugs that block norepinephrine (NE) reuptake enhance noradrenergic neurotransmission, leading to activation of adenyl cyclase and cAMP, which also activates CREB, leading to transcription of a number of different genes associated with synaptic plasticity and neurogenesis. Concurrent exercise and norepinephrine reuptake inhibitor treatment could additively enhance hippocampal BDNF production and be neuroprotective. AMPA, {alpha}-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; NMDA, N-methyl-D-aspartate; TrkB, high-affinity BDNF receptor; CamKII, calcium calmodulin kinase II; MEK1/2, mitogen-activated protein kinase kinase 2; ERK, extracellular-signal related kinase; P, phosphorylation site.

While healthy aging is accompanied by increased variability in cognitive performance 57, aged rats are uniformly more susceptible to memory deficits following an immune challenge64 (Barrientos et al., 2011). Increased vulnerability to infection-induced learning deficits is accompanied by exacerbation of hippocampal inflammatory responses. Although aged rats run far less than their younger counterparts, even a limited amount of voluntary wheel running was sufficient to prevent both impaired learning following E. coli infection, and attenuate neuroinflammation in the hippocampus64. While this study demonstrates that exercise is indeed neuroprotective, no studies to date have addressed whether late-onset physical activity can ameliorate cognitive deficits in a naturally occurring model of age-related cognitive impairment, such as the aged-impaired rat model5. Possible interactions between exercise and aging of the noradrenergic system in healthy individuals also remains an open question.

Late-life resilience: it’s never too late to make new connections

During the past few decades there has been increased interest in regenerative medicine, largely as the result of major advances in research on stem cells and neurotrophic factors. Here we briefly describe the mechanisms that regulate neurogenesis, and advances in the development of therapeutic interventions that either stimulate endogenous neural stem cells, or involve transplantation of neural progenitors derived from stem cells in embryos or autologous induced pluripotent stem (iPS) cells. Several environmental factors and chemical agents have been shown to stimulate neurogenesis in one or both of the two major populations of neural progenitors in the adult brain, which are located in the hippocampal dentate gyrus and the subventricular zone. Exercise, dietary energy restriction and enriched environments enhance neurogenesis by a mechanism involving upregulation of BDNF expression65. The ability of these lifestyle-related factors to enhance neurogenesis is retained during aging, although their efficacy may be attenuated. On the other hand, chronic adverse stressors (e.g., psychosocial stress, depression) and metabolic derangements (e.g., diabetes and insulin resistance) impair neurogenesis by elevating glucocorticoid levels and reducing BDNF expression6668. Unfortunately, because there may not be neural progenitors capable of supplying new neurons to the vast majority of neuronal circuits in the brain, it is unlikely that stimulating endogenous progenitors will be sufficient to repair all damaged circuits in neurodegenerative disorders.

Neuronal replacement by transplantation of neural progenitors, embryonic stem cells or autologous iPS cells is being pursued in preclinical and translational research projects. Numerous studies have documented restorative effects of transplanted stem or progenitor cells in animal models of brain injury or neurodegenerative disorders69. Human studies in which human fetal neural progenitors have been transplanted into PD patients have yielded mixed results70. Nevertheless, emerging findings with iPS cells generated from fibroblasts or other cell types suggest that transplantation of these cells has beneficial effects in a rodent model of PD71. Some of the hurdles encountered in transplantation studies (immune rejection, insufficient trophic support, an ‘unfriendly’ cellular niche, etc.) might be circumvented by inducing endogenous glial cells to become multipotent neural progenitors which could then be coaxed towards neuronal phenotypes using one or more of the environmental factors described above, or by co-treatment with a neurotrophic factor (see below). For example, glial cells could be infected with an innocuous virus expressing two or three genes that encode proteins that change their cellular phenotype to that of iPS cells or neurons.

Neurotrophic factor replacement via infusion or gene therapy is another promising approach for halting and reversing age-related CNS dysfunction72. A long and rich history of preclinical research in animal models, together with considerable clinical experience in AD and PD, suggests that neurotrophic factor-based therapies are likely to counteract disease processes in patients. However, to be successful it is likely that the treatment must be initiated early in the disease process. In addition, such neurotrophic factor therapy assumes that neurons will be responsive (i.e., express functional neurotrophic factor receptors) which in some cases (e.g., insulin-like growth factor/insulin receptors) may not be so73. Because neurotrophic factors may not readily cross the blood-brain barrier, there are efforts to develop low molecular weight agonists of neurotrophic factors with high specificity and potency74, although the potential for adverse effects of such agents on peripheral neurons may hinder their development.

Adherence to lifestyle-based neuroprotection in a deleterious societal milieu

A major implication of the research described above is that successful brain aging is possible for most individuals if they maintain healthy diets and lifestyles throughout their adult life. Unfortunately, this is currently one of the major conundrums in modern societies where high energy food is readily available, exercise is unnecessary in daily routines, and preventative medicine is being suppressed by the actions of the food and pharmaceutical industries. Healthy lifestyle choices should be implemented and incentivized through the coordinated efforts of government, medical schools and health care providers. Moreover, additional research into the mechanisms underlying nutritional and experiential neuroprotection is warranted in order to understand mechanisms which can then be harnessed via pharmacomimetic strategies. Greater awareness of the extent to which physiological aging conforms to a hormetic framework will aid in this effort.

References

  • 1.Behl C. Oestrogen as a neuroprotective hormone. Nat. Rev. Neurosci. 2002;3:433–442. doi: 10.1038/nrn846. [DOI] [PubMed] [Google Scholar]
  • 2.Burns JM, Johnson DK, Watts A, Swerdlow RH, Brooks WM. Reduced lean mass in early Alzheimer disease and its association with brain atrophy. Arch. Neurol. 2010;67:428–433. doi: 10.1001/archneurol.2010.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Stone AA, Schwartz JE, Broderick JE, Deaton A. A snapshot of the age distribution of psychological well-being in the United States. Proc. Natl. Acad. Sci. U. S. A. 2010;107:9985–9990. doi: 10.1073/pnas.1003744107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Albert MS. The ageing brain: normal and abnormal memory. Philos Trans R Soc Lond B Biol Sci. 1997;352:1703–1709. doi: 10.1098/rstb.1997.0152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Gallagher M, Burwell R, Burchinal M. Severity of spatial learning impairment in aging: development of a learning index for performance in the Morris water maze. Behav. Neurosci. 1993;107:618–626. doi: 10.1037//0735-7044.107.4.618. [DOI] [PubMed] [Google Scholar]
  • 6.Robitsek RJ, Fortin NJ, Koh MT, Gallagher M, Eichenbaum H. Cognitive aging: a common decline of episodic recollection and spatial memory in rats. J. Neurosci. 2008;28:8945–8954. doi: 10.1523/JNEUROSCI.1893-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zyzak DR, Otto T, Eichenbaum H, Gallagher M. Cognitive decline associated with normal aging in rats: a neuropsychological approach. Learn Mem. 1995;2:1–16. doi: 10.1101/lm.2.1.1. [DOI] [PubMed] [Google Scholar]
  • 8.Mattson MP, Magnus T. Ageing and neuronal vulnerability. Nat. Rev. Neurosci. 2006;7:278–294. doi: 10.1038/nrn1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Floyd RA, Hensley K. Oxidative stress in brain aging. Implications for therapeutics of neurodegenerative diseases. Neurobiol. Aging. 2002;23:795–807. doi: 10.1016/s0197-4580(02)00019-2. [DOI] [PubMed] [Google Scholar]
  • 10.Dei R, et al. Lipid peroxidation and advanced glycation end products in the brain in normal aging and in Alzheimer's disease. Acta Neuropathol. 2002;104:113–122. doi: 10.1007/s00401-002-0523-y. [DOI] [PubMed] [Google Scholar]
  • 11.Lovell MA, Ehmann WD, Mattson MP, Markesbery WR. Elevated 4-hydroxynonenal in ventricular fluid in Alzheimer's disease. Neurobiol. Aging. 1997;18:457–461. doi: 10.1016/s0197-4580(97)00108-5. [DOI] [PubMed] [Google Scholar]
  • 12.Nicolle MM, Gonzalez J, Sugaya K, Baskerville KA, Bryan D, Lund K, Gallagher M, McKinney M. Signatures of hippocampal oxidative stress in aged spatial learning-impaired rodents. Neuroscience. 2001;107:415–431. doi: 10.1016/s0306-4522(01)00374-8. [DOI] [PubMed] [Google Scholar]
  • 13.Mattson MP. Modification of ion homeostasis by lipid peroxidation: roles in neuronal degeneration and adaptive plasticity. Trends Neurosci. 1998;21:53–57. doi: 10.1016/s0166-2236(97)01188-0. [DOI] [PubMed] [Google Scholar]
  • 14.Guo Q, Fu W, Sopher BL, Miller MW, Ware CB, Martin GM, Mattson MP. Increased vulnerability of hippocampal neurons to excitotoxic necrosis in presenilin-1 mutant knock-in mice. Nat. Med. 1999;5:101–106. doi: 10.1038/4789. [DOI] [PubMed] [Google Scholar]
  • 15.Mattson MP. Roles of the lipid peroxidation product 4-hydroxynonenal in obesity, the metabolic syndrome, and associated vascular and neurodegenerative disorders. Exp Gerontol. 2009;44:625–633. doi: 10.1016/j.exger.2009.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kapogiannis D, Mattson MP. Disrupted energy metabolism and neuronal circuit dysfunction in cognitive impairment and Alzheimer's disease. Lancet Neurol. 2011;10:187–198. doi: 10.1016/S1474-4422(10)70277-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mattson MP, Gleichmann M, Cheng A. Mitochondria in neuroplasticity and neurological disorders. Neuron. 2008;60:748–766. doi: 10.1016/j.neuron.2008.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gibson GE, Starkov A, Blass JP, Ratan RR, Beal MF. Cause and consequence: mitochondrial dysfunction initiates and propagates neuronal dysfunction, neuronal death and behavioral abnormalities in age-associated neurodegenerative diseases. Biochim Biophys Acta. 2010;1802:122–134. doi: 10.1016/j.bbadis.2009.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hyun DH, Emerson SS, Jo DG, Mattson MP, de Cabo R. Calorie restriction up-regulates the plasma membrane redox system in brain cells and suppresses oxidative stress during aging. Proc Natl Acad Sci U S A. 2006;103:19908–19912. doi: 10.1073/pnas.0608008103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Weissman L, Jo DG, Sørensen MM, de Souza-Pinto NC, Markesbery WR, Mattson MP, Bohr VA. Defective DNA base excision repair in brain from individuals with Alzheimer's disease and amnestic mild cognitive impairment. Nucleic Acids Res. 2007;35:5545–5555. doi: 10.1093/nar/gkm605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wong E, Cuervo AM. Autophagy gone awry in neurodegenerative diseases. Nat Neurosci. 2010;13:805–811. doi: 10.1038/nn.2575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Naidoo N, Ferber M, Master M, Zhu Y, Pack AI. Aging impairs the unfolded protein response to sleep deprivation and leads to proapoptotic signaling. J. Neurosci. 2008;28:6539–6548. doi: 10.1523/JNEUROSCI.5685-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bingol B, Sheng M. Deconstruction for reconstruction: the role of proteolysis in neural plasticity and disease. Neuron. 2011;69:22–32. doi: 10.1016/j.neuron.2010.11.006. [DOI] [PubMed] [Google Scholar]
  • 24.Gleichmann M, Chow VW, Mattson MP. Homeostatic disinhibition in the aging brain and Alzheimer's disease. J. Alzheimers Dis. 2011;24:15–24. doi: 10.3233/JAD-2010-101674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Stranahan AM, Mattson MP. Impact of energy intake and expenditure on neuronal plasticity. Neuromolecular Med. 2008;10:209–218. doi: 10.1007/s12017-008-8043-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Stranahan AM, Arumugam TV, Cutler RG, Lee K, Egan JM, Mattson MP. Diabetes impairs hippocampal function through glucocorticoid-mediated effects on new and mature neurons. Nat. Neurosci. 2008;11:309–317. doi: 10.1038/nn2055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kanoski SE, Zhang Y, Zheng W, Davidson TL. The effects of a high-energy diet on hippocampal function and blood-brain barrier integrity in the rat. J. Alzheimers Dis. 2010;21:207–219. doi: 10.3233/JAD-2010-091414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Inui A. Ghrelin: an orexigenic and somatotrophic signal from the stomach. Nat. Rev. Neurosci. 2001;2:551–560. doi: 10.1038/35086018. [DOI] [PubMed] [Google Scholar]
  • 29.Diano S, Farr SA, Benoit SC, McNay EC, da Silva I, Horvath B, Gaskin FS, Nonaka N, Jaeger LB, Banks WA, Morley JE, Pinto S, Sherwin RS, Xu L, Yamada KA, Sleeman MW, Tschöp MH, Horvath TL. Ghrelin controls hippocampal spine synapse density and memory performance. Nat. Neurosci. 2006;9:381–388. doi: 10.1038/nn1656. [DOI] [PubMed] [Google Scholar]
  • 30.Johansson I, Destefanis S, Aberg ND, Aberg MA, Blomgren K, Zhu C, Ghè C, Granata R, Ghigo E, Muccioli G, Eriksson PS, Isgaard J. Proliferative and protective effects of growth hormone secretagogues on adult rat hippocampal progenitor cells. Endocrinology. 2008;149:2191–2199. doi: 10.1210/en.2007-0733. [DOI] [PubMed] [Google Scholar]
  • 31.Debette S, Seshadri S, Beiser A, Au R, Himali JJ, Palumbo C, Wolf PA, Decarli C. Midlife vascular risk factor exposure accelerates structural brain aging and cognitive decline. Neurology. 2011;77:461–468. doi: 10.1212/WNL.0b013e318227b227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sturman MT, de Leon CF, Bienias JL, Morris MC, Wilson RS, Evans DA. Body mass index and cognitive decline in a biracial community population. Neurology. 2008;70:360–367. doi: 10.1212/01.wnl.0000285081.04409.bb. [DOI] [PubMed] [Google Scholar]
  • 33.Molteni R, Barnard RJ, Ying Z, Roberts CK, Gómez-Pinilla F. A high-fat, refined sugar diet reduces hippocampal brain-derived neurotrophic factor, neuronal plasticity, and learning. Neuroscience. 2002;112:803–814. doi: 10.1016/s0306-4522(02)00123-9. [DOI] [PubMed] [Google Scholar]
  • 34.Stranahan AM, Norman ED, Lee K, Cutler RG, Telljohann RS, Egan JM, Mattson MP. Diet-induced insulin resistance impairs hippocampal synaptic plasticity and cognition in middle-aged rats. Hippocampus. 2008;18:1085–1088. doi: 10.1002/hipo.20470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.McNay EC, Ong CT, McCrimmon RJ, Cresswell J, Bogan JS, Sherwin RS. Hippocampal memory processes are modulated by insulin and high-fat-induced insulin resistance. Neurobiol. Learn. Mem. 2010;93:546–553. doi: 10.1016/j.nlm.2010.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Morrison CD, Pistell PJ, Ingram DK, Johnson WD, Liu Y, Fernandez-Kim SO, White CL, Purpera MN, Uranga RM, Bruce-Keller AJ, Keller JN. High fat diet increases hippocampal oxidative stress and cognitive impairment in aged mice: implications for decreased Nrf2 signaling. J. Neurochem. 2010;114:1581–1589. doi: 10.1111/j.1471-4159.2010.06865.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Greenwood CE, Winocur G. Learning and memory impairment in rats fed a high saturated fat diet. Behav. Neural. Biol. 1990;53:74–87. doi: 10.1016/0163-1047(90)90831-p. [DOI] [PubMed] [Google Scholar]
  • 38.Kanoski SE, Meisel RL, Mullins AJ, Davidson TL. The effects of energy-rich diets on discrimination reversal learning and on BDNF in the hippocampus and prefrontal cortex of the rat. Behav. Brain Res. 2007;182:57–66. doi: 10.1016/j.bbr.2007.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Mielke JG, Nicolitch K, Avellaneda V, Earlam K, Ahuja T, Mealing G, Messier C. Longitudinal study of the effects of a high-fat diet on glucose regulation, hippocampal function, and cerebral insulin sensitivity in C57BL/6 mice. Behav. Brain Res. 2006;175:374–382. doi: 10.1016/j.bbr.2006.09.010. [DOI] [PubMed] [Google Scholar]
  • 40.Goodrick CL. Effects of lifelong restricted feeding on complex maze performance in rats. Age. 1984;7:1–2. [Google Scholar]
  • 41.Fontán-Lozano A, Sáez-Cassanelli JL, Inda MC, de los Santos-Arteaga M, Sierra-Domínguez SA, López-Lluch G, Delgado-García JM, Carrión AM. Caloric restriction increases learning consolidation and facilitates synaptic plasticity through mechanisms dependent on NR2B subunits of the NMDA receptor. J. Neurosci. 2007;27:10185–10195. doi: 10.1523/JNEUROSCI.2757-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lee J, Duan W, Mattson MP. Evidence that brain-derived neurotrophic factor is required for basal neurogenesis and mediates, in part, the enhancement of neurogenesis by dietary restriction in the hippocampus of adult mice. J. Neurochem. 2002;82:1367–1375. doi: 10.1046/j.1471-4159.2002.01085.x. [DOI] [PubMed] [Google Scholar]
  • 43.Halagappa VK, Guo Z, Pearson M, Matsuoka Y, Cutler RG, Laferla FM, Mattson MP. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease. Neurobiol Dis. 2007;26:212–220. doi: 10.1016/j.nbd.2006.12.019. [DOI] [PubMed] [Google Scholar]
  • 44.Wu P, Shen Q, Dong S, Xu Z, Tsien JZ, Hu Y. Calorie restriction ameliorates neurodegenerative phenotypes in forebrain-specific presenilin-1 and presenilin-2 double knockout mice. Neurobiol. Aging. 2008;29:1502–1511. doi: 10.1016/j.neurobiolaging.2007.03.028. [DOI] [PubMed] [Google Scholar]
  • 45.Duan W, Mattson MP. Dietary restriction and 2-deoxyglucose administration improve behavioral outcome and reduce degeneration of dopaminergic neurons in models of Parkinson's disease. J. Neurosci. Res. 1999;57:195–206. doi: 10.1002/(SICI)1097-4547(19990715)57:2<195::AID-JNR5>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 46.Maswood N, Young J, Tilmont E, Zhang Z, Gash DM, Gerhardt GA, Grondin R, Roth GS, Mattison J, Lane MA, Carson RE, Cohen RM, Mouton PR, Quigley C, Mattson MP, Ingram DK. Caloric restriction increases neurotrophic factor levels and attenuates neurochemical and behavioral deficits in a primate model of Parkinson's disease. Proc. Natl. Acad. Sci. U.S.A. 2004;101:18171–18176. doi: 10.1073/pnas.0405831102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Duan W, Guo Z, Jiang H, Ware M, Li XJ, Mattson MP. Dietary restriction normalizes glucose metabolism and BDNF levels, slows disease progression, and increases survival in huntingtin mutant mice. Proc. Natl. Acad. Sci. U.S.A. 2003;100:2911–2916. doi: 10.1073/pnas.0536856100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mattson MP. Hormesis defined. Ageing Res. Rev. 2008;7:1–7. doi: 10.1016/j.arr.2007.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Arumugam TV, Phillips TM, Cheng A, Morrell CH, Mattson MP, Wan R. Age and energy intake interact to modify cell stress pathways and stroke outcome. Ann. Neurol. 2010;67:41–52. doi: 10.1002/ana.21798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yang JL, Tadokoro T, Keijzers G, Mattson MP, Bohr VA. Neurons efficiently repair glutamate-induced oxidative DNA damage by a process involving CREB-mediated up-regulation of apurinic endonuclease 1. J. Biol. Chem. 2010;285:28191–28199. doi: 10.1074/jbc.M109.082883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Son TG, Camandola S, Mattson MP. Hormetic dietary phytochemicals. Neuromolecular Med. 2008;10:236–246. doi: 10.1007/s12017-008-8037-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kanoski SE, Davidson TL. Western diet consumption and cognitive impairment: links to hippocampal dysfunction and obesity. Physiol Behav. 2011;103:59–68. doi: 10.1016/j.physbeh.2010.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Parachikova A, Green KN, Hendrix C, LaFerla FM. Formulation of a medical food cocktail for Alzheimer's disease: beneficial effects on cognition and neuropathology in a mouse model of the disease. PLoS One. 2010;5:e14015. doi: 10.1371/journal.pone.0014015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kruman II, Kumaravel TS, Lohani A, Pedersen WA, Cutler RG, Kruman Y, Haughey N, Lee J, Evans M, Mattson MP. Folic acid deficiency and homocysteine impair DNA repair in hippocampal neurons and sensitize them to amyloid toxicity in experimental models of Alzheimer's disease. J. Neurosci. 2002;22:1752–1762. doi: 10.1523/JNEUROSCI.22-05-01752.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Karuppagounder SS, Xu H, Shi Q, Chen LH, Pedrini S, Pechman D, Baker H, Beal MF, Gandy SE, Gibson GE. Thiamine deficiency induces oxidative stress and exacerbates the plaque pathology in Alzheimer's mouse model. Neurobiol. Aging. 2009;30:1587–1600. doi: 10.1016/j.neurobiolaging.2007.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ma QL, Yang F, Rosario ER, Ubeda OJ, Beech W, Gant DJ, Chen PP, Hudspeth B, Chen C, Zhao Y, Vinters HV, Frautschy SA, Cole GM. Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: suppression by omega-3 fatty acids and curcumin. J. Neurosci. 2009;29:9078–9089. doi: 10.1523/JNEUROSCI.1071-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Obregon DF, Rezai-Zadeh K, Bai Y, Sun N, Hou H, Ehrhart J, Zeng J, Mori T, Arendash GW, Shytle D, Town T, Tan J. ADAM10 activation is required for green tea (−)-epigallocatechin-3-gallate-induced alpha-secretase cleavage of amyloid precursor protein. J. Biol. Chem. 2006;281:16419–16427. doi: 10.1074/jbc.M600617200. [DOI] [PubMed] [Google Scholar]
  • 58.Son TG, et al. Plumbagin, a novel Nrf2/ARE activator, protects against cerebral ischemia. J. Neurochem. 2010;112:1316–1326. doi: 10.1111/j.1471-4159.2009.06552.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Vingtdeux V, Giliberto L, Zhao H, Chandakkar P, Wu Q, Simon JE, Janle EM, Lobo J, Ferruzzi MG, Davies P, Marambaud P. AMP-activated protein kinase signaling activation by resveratrol modulates amyloid-beta peptide metabolism. J. Biol. Chem. 2010;285:9100–9113. doi: 10.1074/jbc.M109.060061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Park SS, Stranahan AM, Chadwick W, Zhou Y, Wang L, Martin B, Becker KG, Maudsley S. Cortical gene transcription response patterns to water maze training in aged mice. BMC Neurosci. 2011;12:63. doi: 10.1186/1471-2202-12-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Stranahan AM, Lee K, Becker KG, Zhang Y, Maudsley S, Martin B, Cutler RG, Mattson MP. Hippocampal gene expression patterns underlying the enhancement of memory by running in aged mice. Neurobiol. Aging. 2010;31:1937–1949. doi: 10.1016/j.neurobiolaging.2008.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Garcia C, Chen MJ, Garza AA, Cotman CW, Russo-Neustadt A. The influence of specific noradrenergic and serotonergic lesions on the expression of hippocampal brain-derived neurotrophic factor transcripts following voluntary physical activity. Neuroscience. 2003;119:721–732. doi: 10.1016/s0306-4522(03)00192-1. [DOI] [PubMed] [Google Scholar]
  • 63.Heneka MT, Ramanathan M, Jacobs AH, Dumitrescu-Ozimek L, Bilkei-Gorzo A, Debeir T, Sastre M, Galldiks N, Zimmer A, Hoehn M, Heiss WD, Klockgether T, Staufenbiel M. Locus ceruleus degeneration promotes Alzheimer pathogenesis in amyloid precursor protein 23 transgenic mice. J. Neurosci. 2006;26:1343–1354. doi: 10.1523/JNEUROSCI.4236-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Barrientos RM, Frank MG, Crysdale NY, Chapman TR, Ahrendsen JT, Day HE, Campeau S, Watkins LR, Patterson SL, Maier SF. Little exercise, big effects: reversing aging and infection-induced memory deficits, and underlying processes. J. Neurosci. 2011;31:11578–11586. doi: 10.1523/JNEUROSCI.2266-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Lazarov O, Mattson MP, Peterson DA, Pimplikar SW, van Praag H. When neurogenesis encounters aging and disease. Trends Neurosci. 2010;33:569–579. doi: 10.1016/j.tins.2010.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Bremner JD. Stress and brain atrophy. CNS Neurol. Disord. Drug Targets. 2006;5:503–512. doi: 10.2174/187152706778559309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Stranahan AM, Khalil D, Gould E. Social isolation delays the positive effects of running on adult neurogenesis. Nat. Neurosci. 2006;9:526–533. doi: 10.1038/nn1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Rothman SM, et al. 3×TgAD mice exhibit altered behavior and elevated Aβ after chronic mild social stress. Neurobiol. Aging. 2011 doi: 10.1016/j.neurobiolaging.2011.07.005. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lindvall O, Kokaia Z. Stem cells in human neurodegenerative disorders--time for clinical translation? J. Clin. Invest. 2010;120:29–40. doi: 10.1172/JCI40543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Olanow CW, Kordower JH, Lang AE, Obeso JA. Dopaminergic transplantation for Parkinson's disease: current status and future prospects. Ann. Neurol. 2009;66:591–596. doi: 10.1002/ana.21778. [DOI] [PubMed] [Google Scholar]
  • 71.Hargus G, et al. Differentiated Parkinson patient-derived induced pluripotent stem cells grow in the adult rodent brain and reduce motor asymmetry in Parkinsonian rats. Proc. Natl. Acad. Sci. U. S. A. 2010;107:15921–15926. doi: 10.1073/pnas.1010209107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nagahara AH, Tuszynski MH. Potential therapeutic uses of BDNF in neurological and psychiatric disorders. Nat. Rev. Drug Discov. 2011;10:209–219. doi: 10.1038/nrd3366. [DOI] [PubMed] [Google Scholar]
  • 73.Moloney AM, Griffin RJ, Timmons S, O'Connor R, Ravid R, O'Neill C. Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer's disease indicate possible resistance to IGF-1 and insulin signalling. Neurobiol. Aging. 2010;31:224–243. doi: 10.1016/j.neurobiolaging.2008.04.002. [DOI] [PubMed] [Google Scholar]
  • 74.Massa SM, Yang T, Xie Y, Shi J, Bilgen M, Joyce JN, Nehama D, Rajadas J, Longo FM. Small molecule BDNF mimetics activate TrkB signaling and prevent neuronal degeneration in rodents. J. Clin. Invest. 2010;120:1774–1785. doi: 10.1172/JCI41356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Harvey J. Leptin regulation of neuronal excitability and cognitive function. Curr. Opin. Pharmacol. 2007;7:643–647. doi: 10.1016/j.coph.2007.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Mirescu C, Gould E. Stress and adult neurogenesis. Hippocampus. 2006;16:233–238. doi: 10.1002/hipo.20155. [DOI] [PubMed] [Google Scholar]
  • 77.Mattson MP, Perry T, Greig NH. Learning from the gut. Nat. Med. 2003;9:1113–1115. doi: 10.1038/nm0903-1113. [DOI] [PubMed] [Google Scholar]
  • 78.Stranahan AM, Lee K, Martin B, Maudsley S, Golden E, Cutler RG, Mattson MP. Voluntary exercise and caloric restriction enhance hippocampal dendritic spine density and BDNF levels in diabetic mice. Hippocampus. 2009;19:951–961. doi: 10.1002/hipo.20577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Witte AV, Fobker M, Gellner R, Knecht S, Flöel A. Caloric restriction improves memory in elderly humans. Proc. Natl. Acad. Sci. U.S.A. 2009;106:1255–1260. doi: 10.1073/pnas.0808587106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Porter DW, Kerr BD, Flatt PR, Holscher C, Gault VA. Four weeks administration of Liraglutide improves memory and learning as well as glycaemic control in mice with high fat dietary-induced obesity and insulin resistance. Diabetes Obes. Metab. 2010;12:891–899. doi: 10.1111/j.1463-1326.2010.01259.x. [DOI] [PubMed] [Google Scholar]
  • 81.Calabrese EJ, Bachmann KA, Bailer AJ, Bolger PM, Borak J, Cai L, Cedergreen N, Cherian MG, Chiueh CC, Clarkson TW, Cook RR, et al. Toxicol. Appl. Pharmacol. 2007;222:122–128. doi: 10.1016/j.taap.2007.02.015. [DOI] [PubMed] [Google Scholar]
  • 82.Xu X, Zhan M, Duan W, Prabhu V, Brenneman R, Wood W, Firman J, Li H, Zhang P, Ibe C, Zonderman AB, Longo DL, Poosala S, Becker KG, Mattson MP. Gene expression atlas of the mouse central nervous system: impact and interactions of age, energy intake and gender. Genome Biol. 2007;8:R234. doi: 10.1186/gb-2007-8-11-r234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Calabrese V, Cornelius C, Dinkova-Kostova AT, Calabrese EJ, Mattson MP. Cellular stress responses, the hormesis paradigm, and vitagenes: novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid. Redox Signal. 2010;13:1763–1811. doi: 10.1089/ars.2009.3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kraft AD, Johnson DA, Johnson JA. Nuclear factor E2-related factor 2-dependent antioxidant response element activation by tert-butylhydroquinone and sulforaphane occurring preferentially in astrocytes conditions neurons against oxidative insult. J. Neurosci. 2004;24:1101–1112. doi: 10.1523/JNEUROSCI.3817-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kim SJ, et al. Curcumin stimulates proliferation of embryonic neural progenitor cells and neurogenesis in the adult hippocampus. J. Biol. Chem. 2008;283:14497–14505. doi: 10.1074/jbc.M708373200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Spencer JP. The impact of fruit flavonoids on memory and cognition. Br. J. Nutr. 2010;104(supplement 3):S40–S47. doi: 10.1017/S0007114510003934. [DOI] [PubMed] [Google Scholar]
  • 87.Roriz-Cruz M, Rosset I, Wada T, Sakagami T, Ishine M, De Sá Roriz-Filho J, Cruz TR, Hosseinkhani M, Rodrigues RP, Sudoh S, Arai H, Wakatsuki Y, Souza AC, Nakagawa M, Kita T, Matsubayashi K. Cognitive impairment and frontal-subcortical geriatric syndrome are associated with metabolic syndrome in a stroke-free population. Neurobiol. Aging. 2007;28:1723–1736. doi: 10.1016/j.neurobiolaging.2006.07.013. [DOI] [PubMed] [Google Scholar]
  • 88.Gunstad J, Paul RH, Cohen RA, Tate DF, Spitznagel MB, Gordon E. Elevated body mass index is associated with executive dysfunction in otherwise healthy adults. Compr. Psychiatry. 2007;48:57–61. doi: 10.1016/j.comppsych.2006.05.001. [DOI] [PubMed] [Google Scholar]
  • 89.van den Berg E, Biessels GJ, de Craen AJ, Gussekloo J, Westendorp RG. The metabolic syndrome is associated with decelerated cognitive decline in the oldest old. Neurology. 2007;69:979–985. doi: 10.1212/01.wnl.0000271381.30143.75. [DOI] [PubMed] [Google Scholar]
  • 90.Sturman MT, de Leon CF, Bienias JL, Morris MC, Wilson RS, Evans DA. Body mass index and cognitive decline in a biracial community population. Neurology. 2008;70:360–367. doi: 10.1212/01.wnl.0000285081.04409.bb. [DOI] [PubMed] [Google Scholar]
  • 91.Li Y, Dai Q, Jackson JC, Zhang J. Overweight is associated with decreased cognitive functioning among school-age children and adolescents. Obesity (Silver Spring) 2008;16:1809–1815. doi: 10.1038/oby.2008.296. [DOI] [PubMed] [Google Scholar]
  • 92.Huizinga MM, Beech BM, Cavanaugh KL, Elasy TA, Rothman RL. Low numeracy skills are associated with higher BMI. Obesity (Silver Spring) 2008;16:1966–1968. doi: 10.1038/oby.2008.294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sabia S, Kivimaki M, Shipley MJ, Marmot MG, Singh-Manoux A. Body mass index over the adult life course and cognition in late midlife: the Whitehall II Cohort Study. Am. J. Clin. Nutr. 2009;89:601–607. doi: 10.3945/ajcn.2008.26482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Volkow ND, Wang GJ, Telang F, Fowler JS, Goldstein RZ, Alia-Klein N, Logan J, Wong C, Thanos PK, Ma Y, Pradhan K. Inverse association between BMI and prefrontal metabolic activity in healthy adults. Obesity (Silver Spring) 2009;17:60–65. doi: 10.1038/oby.2008.469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Fergenbaum JH, Bruce S, Lou W, Hanley AJ, Greenwood C, Young TK. Obesity and lowered cognitive performance in a Canadian First Nations population. Obesity (Silver Spring) 2009;17:1957–1963. doi: 10.1038/oby.2009.161. [DOI] [PubMed] [Google Scholar]
  • 96.Granholm AC, Bimonte-Nelson HA, Moore AB, Nelson ME, Freeman LR, Sambamurti K. Effects of a saturated fat and high cholesterol diet on memory and hippocampal morphology in the middle-aged rat. J. Alzheimers Dis. 2008;14:133–145. doi: 10.3233/jad-2008-14202. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES