2,654
Views
51
CrossRef citations to date
0
Altmetric
Translational Research

Neuropathological approaches to cerebral aging and neuroplasticity

Enfoques neuropatológicos del envejecimiento y la neuroplasticidad cerebral

Approche neuropathologique de la neuroplasticité et du vieillissement cérébral

&

Abstract

Cerebral aging is a complex and heterogenous process related to a large variety of molecular changes involving multiple neuronal networks, due to alterations of neurons (synapses, axons, dendrites, etc), particularly affecting strategically important regions, such as hippocampus and prefrontal areas. A substantial proportion of nondemented, cognitively unimpaired elderly subjects show at least mild to moderate, and rarely even severe, Alzheimer-related lesions, probably representing asymptomatic preclinical Alzheimer's disease, and/or mixed pathologies. While the substrate of resilience to cognitive decline in the presence of abundant pathologies has been unclear, recent research has strengthened the concept of cognitive or brain reserve, based on neuroplasticity or the ability of the brain to manage or counteract age-related changes or pathologies by reorganizing its structure, connections, and functions via complex molecular pathways and mechanisms that are becoming increasingly better understood. Part of neuroplasticity is adult neurogenesis in specific areas of the brain, in particular the hippocampal formation important for memory function, the decline of which is common even in “healthy” aging. To obtain further insights into the mechanisms of brain plasticity and adult neurogenesis, as the basis for prevention and potential therapeutic options, is a major challenge of modern neurosciences.

El envejecimiento cerebral es un proceso complejo y heterogéneo que se relaciona con una gran variedad de cambios moleculares que involucran múltiples redes neuronales, a causa de alteraciones en las neuronas (sinapsis, axones, dendritas, etc.), que particularmente afecta regiones estratégicamente importantes como el hipocampo y las áreas prefrontales. Una proporción significativa de sujetos no dementes y cognitivamente no deteriorados muestran a lo menos lesiones leves a moderadas o rara vez graves que se relacionan con el Alzheimer; lesiones que representan probablemente la Enfermedad de Alzheimer preclínica asintomática ylo patologías mixtas. Si bien el sustrato de la resiliencia para la declinación cognitiva en diversas patologías ha sido poco claro, la investigación retiente ha reforzado el concepto de reserva cognitiva o cerebral, basado en la neuroplasticidad o la capacidad del cerebro de controlar o contrarrestar los cambios o patologías relacionadas con la edad al reorganizar su estructura, conexiones y funciones a través de complejas vías y mecanismos moleculares que están siendo cada vez mejor comprendidos. Parte de la neuroplasticidad es la neurogénesis adulta en áreas específicas del cerebro, en particular en la formación hipocámpica; esta área es importante para la función de memoria y se reduce con frecuencia incluso en el envejecimiento “saludable”. La obtencion de mayores conocimientos sobre los mecanismos de plasticidad cerebral y neurogénesis adulta, como base para la prevención y potentiales opciones terapéuticas, constituye un importante desafío para las modernas neurociencias.

Le vieillissement cérébral est un processus complexe et hétérogène associé à de nombreuses modifications moléculaires impliquant de multiples réseaux neuronaux en raison d'altérations de neurones (synapses, axones, dendrites, etc.) touchant particulièrement des régions stratégiques comme l'hippocampe et le cortex préfrontal. Un pourcentage important de sujets âgés non déments et sans troubles cognitifs présentent des lésions de type Alzheimer de niveau au moins léger à modéré et plus rarement sévère, témoignant probablement d'une maladie d'Alzheimer préclinique asymptomatique et/ou de troubles mixtes. Le substrat de la résilience du déclin cognitif au cours de nombreuses pathologies n'est pas clair mais des recherches récentes ont renforcé le concept de réserve cérébrale ou cognitive sur la base de la neuroplasticité ou de la capacité du cerveau à maîtriser ou à s'opposer aux modifications ou aux pathologies liées à l'âge en réorganisant ses structures, ses connexions et ses fonctions grâce à des mécanismes et des voies moléculaires complexes de mieux en mieux compris. La neurogenèse adulte fait partie de la neuroplasticité dans des aires cérébrales spécifiques comme l'hippocampe, important pour la mémoire, qui décline de façon courante même chez le sujet « sain ». Mieux comprendre les mécanismes de la plasticité cérébrale et de la neurogenèse adulte, comme base de la prévention et des solutions éventuelles de traitement, est un défi majeur pour les sciences modernes.

Introduction

“Healthy” aging is defined as aging without disease. With the current attempts to increase the life span, understanding the molecular interactions and mechanisms involved in normal brain aging continues to be a challenge. Cerebral aging is a complex and heterogenous process that is associated with a high degree of interindividual variability. The last 20 years have witnessed a great increase in our knowledge of its basic mechanisms. Functional analyses have identified signaling pathways acting as master regulators of aging and lifespan that are conserved in many animals, suggesting that the rate of aging is not inevitably fixed, but is plastic and open to modifications. Based on experimental evidence, the evolution of aging is probably the result of determinants of neuronal vulnerability, which include altered protein interaction networks, mitochondria, reactive oxidative species and intracellular calcium homeostasis, autophagy, signal transduction pathways, stem cell proliferation, and stress resistance mechanisms.Citation1,Citation2 Perturbations in the functional state of these processes may lead to a state of decreased homeostatic reserve, where the aged neurons could still maintain adequate function during normal activity, although they become vulnerable. Neurons have significant homeostatic control of essential physiological functions like synaptic excitability, gene expression, and metabolic regulation. Any deviation in these physiological events can have severe consequences, as observed in aging.Citation3 A recent study in a large cohort of >10 000 persons showed that a measurable decline in generalized cortical function is already present by 45 to 49 years of age, with evidence of faster decline in older people.Citation4 Dementia due to Alzheimer's disease (AD) is preceded by about 5 to 6 years of accelerated decline of multiple cognitive functions; by contrast, little decline is evident in persons who do not develop AD. Compromised brain energy metabolism/oxygen delivery to neurons and blood flow differences in the regions most vulnerable to neurodegeneration are possible mechanisms of progression from healthy to “unhealthy” brain aging.Citation5

The human brain is uniquely powerful with respect to cognitive abilities, yet many neuronal networks, in particular the hippocampal and neocortical circuits that mediate such complex functions, are highly vulnerable to aging. Loss of neurons, now recognized to be more modest than previously suggested, mainly involves these specific neuroanatomical areas. Cognition and its decline associated with brain aging also seems to be variable and possibly open to modifications.Citation1 Studies in humans and animal models suggest that age-related cognitive decline is more likely to be associated with alterations in synaptic connectivity than with neuronal loss and white matter changes.Citation6,Citation7 According to recent studies, alterations of intracellular γ-secretase mediated signaling pathways may be involved in synaptic pathogenesis of AD,Citation8 and apolipoprotein E is suggested to enhance the toxic effects of oligomeric amyloid beta (Aβ), causing synapse loss, a major correlate of cognitive decline in AD.Citation9 Although dementia-associated hallmarks of AD pathology (neuritic plaques and neurofibrillary tangles) become less prominent with increasing age, synaptic marker abnormalities in dementia remain constant and may represent an independent substrate of dementia spanning all ages.Citation10 These and other changes induce functional network disruptions in degenerative dementia,Citation11,Citation12 suggesting that disease progress is transmitted by neural pathways.Citation13

Age-related brain changes are widely documented. Postmortem and in vivo magnetic resonance imaging (MRI) studies of healthy brains have reported different location, extent, and severity of these changes with aging, some brain regions with greater activation being linked to better cognitive performance. Besides hemispheric asymmetry reduction they indicated increased activity in (pre)frontal regions, suggesting posterior-anterior shift models of functional brain aging.Citation14 There is a strong relationship between cognitive ability and cortical fine structure in the prefrontal cortex.Citation15 Postmortem studies of human brains revealed more prominent age-related changes in the anterior and posterior white matter, but not in gray matter volumes, histology showing less severe changes than the imaging methods.Citation16 While in previous studies postmortem MRI of white matter lesions (WMLs) was less sensitive than pathology, more recent ones showed that postmortem MRI is a valid tool for the assessment of subcortical pathologies.Citation17 MRI investigations showed widespread age-related changes in prefrontal cortex and white matter, somatosensory cortex, and, to a lesser degree, in motor cortex, the prefrontal white matter being most susceptible to the influence of age.Citation18 In cognitively normal elderly subjects, WMLs were inversely correlated with gray matter volume, with greatest volume loss in the frontal cortex. Both advancing age and hypertension predict higher WML load, which is itself associated with gray matter atrophy.Citation19 Low white matter grade and ventricular grade on MRI are powerful determinants of long-term survival among older individuals.Citation20 Recent functional neuroimaging studies indicated reduced cortical activation in the default-mode network for mild cognitive impairment patients, compared with age-matched healthy elderly persons, mainly in the retrosplenial region/posterior cingulate cortex, left hippocampus, and bilateral inferior and middle frontal areas, while increased activation for patients was observed in the medial prefrontal and bilateral middle temporal/ angular cortex, probably as a compensatory mechanism.Citation21 Resting state networks have been found to be hierarchically organized.Citation22

Age-related atrophy is observed in the hippocampal region.Citation23 This region is of particular interest given its contribution to memory function, working memory decline being a common complaint in healthy agingCitation24 and one of the earliest signs of AD. Impaired hippocampal synaptic function is an early detectable pathologic alteration, well before amyloid plaque accumulation and cell death.Citation25 Positive relationships emerged consistently between the hippocampal formation, global cognition, and memory, and between frontal measures and executive function.Citation26 The hippocampal formation and the Papez circuit are targeted differentially by diseases of late life.Citation27 Volumetric MRI of temporal and parietal brain structures distinguishes AD patients from healthy subjects, volumetry of the left and right hippocampus providing the highest diagnostic accuracy in separating these groups.Citation28 Recent advances in imaging techniques (diffusion tensor imaging [DTI] and magnetization transfer imaging [MTI]) indicate that age-related small-vessel disease is a diffuse process affecting the whole brain and that WMLs are probably only the tip of the iceberg,Citation19,Citation29-Citation31 while decreased gray matter diffusivity might be a potential new biomarker for early AD.Citation32 Aβ-associated cortical thinning has been observed in clinically normal elderly subjects.Citation33

Age-related neuronal dysfunction involves a host of subtle changes such as reduction in the complexity of dendritic arborization and length, decrease in spine numbers and related synaptic densities, changes involving receptors, neurotransmitters, cytology, electric transmission, vascular or Alzheimer-related changes, and myelin dystrophy. Together, these multiple alterations in the brain may lead to age-related cognitive dysfunction.Citation1,Citation2 However, every lesion in the nervous system triggers an endogenous neuroprotective reaction, combining neuroplasticity and neurogenesis, which are initiated and regulated by neurotrophic factors in a multimodal way.Citation34 Extrusion of misfolded and aggregated (toxic) proteins may be a protective strategy of aging neurons.Citation35

Neuropathology findings in cognitively normal aged subjects

It is increasingly recognized that the correlation between neuropathological lesions and cognition is modest and accounts for about a quarter of the variance in cognition of older adults. Concerning factors that modify or mediate the association between neuropathology and cognition, it was hypothesized that the concept of resilient aging can be useful to understand mechanisms that underlie healthy aging amidst disease-related pathology.Citation34 Some individuals maintain normal cognitive function despite significant brain pathology, while others suffer varying degrees of cognitive and neurological deterioration. Many aged people do not exhibit cognitive impairment or other symptoms of disease and live “normal” lives, but nonetheless display pathological changes that are characteristic of AD, Parkinson's disease (PD), cerebrovascular disease (CVD), or other disorders.Citation36-Citation38 Although the best morphologic correlates of cognitive impairment/dementia are; (i) the number of neocortical neurofibrillary tangles (NFTs)Citation39-Citation43; and (ii) loss of synapses,Citation44-Citation47 between 8% and 45% of nondemented, often cognitively stable older adults were found to have AD-related pathologies.Citation38,Citation43,Citation48-Citation58 Many of them showed only minimal to mild neuritic changes corresponding to Braak tau stages 0-IV,Citation59 while 31% to 88% showed National Institute for Aging and Reagan Institute (NIA-RI) criteria of no likelihood for AD criteria.Citation51,Citation53 The frequency of intermediate likelihood of AD criteria ranged from 11.9% to 35.8%, Citation37,Citation53,Citation56 and only 1.5 to 3% were scored as having a high likelihood of AD.Citation53 The presence of AD lesions in nondemented aged individuals may represent AD at a stage prior to clinical expression (presymptomatic or unrecognized early forms) .Citation54,Citation55,Citation58,Citation60,Citation61, This is supported by observations that the mechanisms responsible for these changes in nondemented elderly appear similar if not identical to those found in AD,Citation60,Citation62 and their distribution corresponds to the hierarchical topographical procession associated with symptomatic AD.Citation48,Citation49,Citation61 The concept of “preclinical” ADCitation58,Citation60,Citation61 pathology has been further solidified in biomarker studies using CSF Aβ-42Citation63 and more directly in vivo positron emission tomography (PET) amyloid scanning, demonstrating that 20% to 30% of healthy elderly subjects have elevated PIB signals indicative of extensive amyloid deposition.Citation64 These data suggest a high frequency of preclinical AD pathology in normal elderly similar to that seen in clinico-pathologic cohorts.Citation65,Citation66 They further suggest that preclinical changes are not static, but progress over time.Citation67,Citation68

Among 555 nondemented persons with false-positive pathological NIA-RI high likelihood for AD, only 1.6% corresponded to Braak stage V, 0.5% to stage VI, and 2.6% to stage V-VI,Citation41 while in other studies between 35% and 88% were NIA-RI negativeCitation43,Citation51; 18% to 25% met the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) criteria for AD.Citation51,Citation52,Citation56 Review of the data from National Disease Coordinating Center (NDCC) database and the Nun Study emphasized that there may be no documented example of truly end-stage neurofibrillary pathology with intact cognition.Citation69 Although in the Adult Changes in Thought (ACT) and Nun Studies, nondemented seniors with severe AD pathology (mean age of 89.15±6.9 to 90.80±5.2 years) amounted to 8% and 12%, respectively, most of them showed neuritic Braak stage V, and frontal NFT counts were slightly lower than in a comparable dementia group.Citation49 Moreover, review of clinical data from those studies revealed that most of the seniors classified as nondemented were indeed significantly memory-impaired.Citation49 A recent study of nondemented elderly demonstrated 62% with low and 28% with high NFT levels70; 87 nondemented elderly (mean age 87 ±5.9 years; mean MMSE 28.3) showed mean Braak stage 3.0±0.9, a total NFT score of 4.5±2.5, and mean neuritic density of 1.3±1.1, whereas AD cases showed much higher cortical neuritic and striatal amyloid plaque scores.Citation71 The 90+ study revealed significantly less severe Aβ, α-synuclein, and TPD-43 pathologies, and hippocampal sclerosis in nondemented subjects, while Aβ distribution showed no essential differences; nondemented individuals had limited hippocampal tau and neocortical Aβ pathology.Citation72

A recent clinicopathologic study of 296 persons without cognitive impairment of the Religious Order Study (ROS) and the Memory and Aging Project (MAP) showed a common presence of AD pathology and macroscopic infarctions. Amyloid load was related to global cognition (P<0.05), with only a trend for NFTs (P =0.08), while NFTs and macroscopic infarctions were related to episodic memory (P =0.03 and 0.02, respectively); AD pathology and Aβ load to working memory (P =0.02 and 0.03, respectively).Citation73

Comparing the biochemistry of AD and nondemented nonagenerians revealed the lack of clear amyloid-related pathological/ biochemical determination between both groups.Citation74 A personal retrospective study of 100 nondemented elderly (mean age 81.23±5.47 years, mean Mini Mental State Examination (MMSE) score 29) revealed negative Khachaturian criteria and CERAD stage 0 in 83% and 86%, respectively, only 13% with CERAD stage A and 1% stage B. Braak neuritic stages ranged from 0 to IV with an average score of 2.3±0.8. 12% were scored NIA-RI low, and only 2% intermediate likelihood for AD.Citation37 Thus, mounting evidence from clinicopathologic studies support the view that AD is a continuous spectrum between asymptomatic lesions in cognitively normal elderly and dementia, with mild cognitive impairment (MCI) as a transition phase between them.Citation75 Although correlations between cognitive deficits and the severity and extension of senile plaques (SP) and NFTs (see ref 42) have been found, at least in those brains without other pathologies, the distinction between “physiological” (in nondemented subjects) and “pathological” aging (PA) is difficult. A postmortem classification for individuals reported to be cognitively normal before death, their brains showing plaque pathology similar in extent to AD with only minimal cortical tau pathology, may also be difficult.Citation76 Recent biochemical studies found extensive overlap with only subtle quantitative differencies between Aβ levels, peptide profiles, solubility, and oligomeric assemblies in PA and AD brains, suggesting that PA represents an initial prodromal stage of AD and that these individuals would eventually develop clinical symptoms, if they lived long enough, or an inherent individual resistance to the toxic effects of Aβ.Citation77 Recent studies suggest that two independent processes (synapse-mediated and ApoE-mediated) may contribute to region-specific Aβ accumulation in nondemented individuals, and may influence the mechanisms of the regional vulnerability to Aβ accumulation, which is prevented by ApoE.Citation78 A coding mutation (A673T) in the APP gene that reduces the P-cleavage of APP may protect against AD and also against cognitive decline in the elderly without AD.Citation79

Older persons with overall normal cognitive function and preclinical AD changes by brain autopsy usually have lower scores on cognitive function tests, particularly episodic and working memory.Citation24,Citation54 Aβ biomarker studies also confirmed the relations between preclinical AD and cognition,Citation80,Citation81 and a clinicopathologic study indicated that elders with AD changes but without overt dementia are more likely to have memory complaints.Citation82

The definition of nondemented subjects with AD pathology raises important questions regarding the cognitive profile of these people who are relatively protected from the devastating effects of AD-related lesions. A default hypothesis for AD is that it is a part of a “normal aging process,” such that plaques and tangles are secondary to aging or that the primary aging effect is on synapses and neurons independent of these morphological AD markers. AD is indeed a disease that accompanies human aging, but it is not an inevitable consequence of it.Citation83,Citation84 However, the suggestion that plaques and tangles may “cause” this disorder is oversimplified or even wrong, since accumulating evidence suggests that AD pathology represents effect rather than cause or at least a host response to injury, equaling adaptive or neuroprotective reactions.Citation85

Many studies emphasize multiple additional pathologies in nondemented elders, in particular cerebrovascular lesions (CVLs), eg, small or large cerebral infarctions, lacunes, WMLs, in 22 up to almost 100%. Citation36,Citation49,Citation51-Citation53 Evaluation of 336 cognitively normal (CN) seniors from four studies revealed moderately to frequent neuritic plaque density in 47%; of these 6% also had Braak stages V or VI; medullary, nigral, and cortical Lewy bodies in 15%, 8%, and 4%, respectively; cerebral microinfarcts in 33% and high-level cerebral microinfarcts in 10%. The burden of brain lesions and comorbidities varied widely within each study but was similar across studies.Citation86 Among 418 nondemented participants of the Religious Order study (mean age 88.5±5.3 years), 35% showed macroscopical infarcts, 8% microinfarcts, 14.8% arteriosclerosis, 5.7% both, only 37.5% being free of CVLs.Citation87 Up to 75% of CN seniors had various degrees of cerebral amyloid angiopathy (CAA),Citation53 occasional hippocampal sclerosis,Citation53,Citation56 Lewy body pathologies in up to 18%,Citation36,Citation37,Citation51-Citation53,Citation56,Citation57 argyrophilic grains in up to 23 %,Citation53 and mixed pathologies in 7% to 14.8%.Citation36,Citation57 In a small autopsy series of CN elders, only 16% showed no additional pathology.Citation55 Among 100 nondemented seniors, mild, moderate and severe intracranial atherosclerosis was present in 31%, 17%, and 6%, respectively, lacunar state in basal ganglia and/or white matter in 73%, hippocampal sclerosis in 3%, whereas only 9% were free of CVLs. Lewy bodies were observed in 5% , tau pathology in brain stem in 60%, and mixed cerebral pathologies (CVLs and moderate neuritic Braak stages) in 6%.Citation37 A recent British nondemented sample (n = 53; mean age 81.5±7.4 years; MMSE score 27-30) showed maximum score neuritic plaques in 32% to 49%, NFTs in hippocampus and neocortex in 81% and 30.8%, respectively, white matter changes 55% to 83.7%, small vascular disease 45%, infarcts 13.7%, lacunes 6%, and hemorrhages 10%.Citation88 Thus, clinically silent pathology is widespread in normal aging, and the term ”healthy aging“ is inappropriate at the cellular level, and is manifested by regional heterogeneity in the scenario of general volume loss in the human brain.

Brain aging and neuroplasticity

Aging is associated with progressive loss in function across multiple systems, including sensation, cognition, memory, motor control, and affect. The traditional view has been that functional decline in aging is unavoidable because it is a direct consequence of brain machinery wearing down over time. In recent years, however, an alternative perspective has emerged that, based on extensive experimental work, argues that as people age, brain plasticity processes with negative consequences begin to dominate brain functioning. Four core factors—reduced schedules of brain activity, noisy processing, weakened modulatory control, and negative learning—interact to create a self-reinforcing downward spiral of degraded brain function. These interrelated functions promote plastic changes in the brain that result in substantial improvement in function and/or recovery from functional losses.Citation89 Neuroplasticity can be defined as the ability of the nervous system to respond to intrinsic and extrinsic stimuli by reorganizing its structure, function, and connections. It is both a substrate of learning and memory and a mediator of responses to neuronal attrition and injury (compensatory plasticity). This continuous process in reaction to neuronal activity and injury involves modulation of structural and functional processes of dendrites, axons, and synapses. Plasticity is an intrinsic property of the brain across the lifespan. However, mechanisms of neuroplasticity may vary with age, and occur in many variations and in many contexts, while common areas of plasticity that emerge across diverse CNS conditions include experience dependence and circuit training.Citation70

The concept of “cognitive reserve”

Contrary to assumptions that changes in brain networks are possible only during crucial periods of development, recent research has supported the idea of a permanent plastic brain. Novel experience, altered afferent input due to environmental changes, and learning new skills are now recognized as modulators of brain function and underlying neuroanatomic circuitry.

Results in animal experiments and discovery of increases in gray and white matter in the adult human brain as a result of learning and exercise have reinforced the old concept of “cognitive reserve,” that is, the ability to reinforce brain volume in certain areas and thus provide a greater threshold for age-dependent deficits, or the capacity of the brain to manage pathology or age-related changes, thereby minimizing clinical manifestation.Citation90-Citation94 The concept of “cognitive reserve” and a broader theory of “brain reserve” was originally proposed to help explain epidemiological data indicating that individuals who engaged higher levels of mental and physical activity via education, occupation, and recreation were associated with slower cognitive decline in healthy aging and are at lower risk of developing AD and other forms of dementia.Citation95-Citation98 The aging process that results in loss of synapses and possible neurons may be far more detrimental for those with little brain reserve as compared with those with a high one.Citation99

The construct of “cognitive reserve” is a set of variables including intelligence, education, and mental stimulation which putatively allows the brain to adapt to underlying pathologies by maintaining cognitive function despite underlying neuronal changes. It also indicates a resilience to neuropathological damage, and could be defined as the ability to optimize or maximize performance through effective recruitment of brain networks and/or alternative cognitive strategies. Childhood cognition, educational attainment, and adult occupation all contribute to cognitive reserve independently. Enriched environment and physical activity influence the rate of neurogenesis in adult animal model hippocampi.Citation100 In people with high reserve, deterioration occurs rapidly once the threshold is reached.Citation101

Structural and functional brain imaging studies have revealed selective changes in aging brain that reflect neural decline as well as compensatory neural recruitment, representing possible neural substrates of cognitive reserve, but its neural basis is still a topic of ongoing research.Citation102 While aging is associated with reductions in cortical thickness, white matter integrity, transmitter activity, and functional engagement in the hippocampus and occipital areas, there are compensatory increases in frontal functional engagement that correlate with better behavioral performance in the elderly.Citation103,Citation104 Those cortical regions most consistently shrinking in aging—prefrontal and parietal cortices—are the same regions showing increased regional activation in aging, suggesting that losses in regional brain integrity drive functional reorganization through changes in processing strategy.Citation105 Cognitive reserve allows individuals greater neural efficacy, greater neural capacities, and the ability for compensation via the recruitment of additional brain regions.Citation106 Frontal and supramarginal cortical activity has been suggested to compensate for an age-related decrease in inferior-frontal junction recruitment of verbal fluency processing. Larger brain and hippocampal values, and neuronal hypertrophyCitation107 were associated with preserved cognitive function despite a high burden of AD pathology (asymptomatic AD).Citation108 The structural and functional imaging correlates of cognitive and brain reserve hypothesis have recently been reviewed.Citation109 A complementary hypothesis of “metabolic” reserve is characterized by neuronal circuits that respond adaptively to perturbations in cellular energy metabolism and thereby protect against declining function, mediated by neurotrophic factor signaling, and glucose metabolism.Citation93 Increased basal forebrain metabolism in MCI is an evidence for brain reserve in incipient dementia.Citation110 Neuroprotective effects of noradrenaline both in vivo and in vitro suggest noradrenaline's key role in mediating cognitive reserve—by disease compensation, modification, or a combination of both, a viable hypothesis.Citation111

Structural basis of neuroplasticity

The structural elements that embody plasticity include synaptic efficacy and remodeling, synaptogenesis, neurite extension including axonal sprouting and dendritic remodeling, neurogenesis, and recruitment from neural progenitor cells. Phenomenological processes that manifest plasticity are: synapse, neurite, neuronal cell bodies, anterograde and retrograde transport, cell interactions (neuron-glia), neuronal networks, and related activities.Citation35 They include intraneuronal, interneuronal, and intercellular signaling through glia, and involve extracellular matrix molecules, immunoglobulins, myelin-associated inhibitors, tyrosine kinase receptors, neurotrophic and growth factors, inflammatory cytokines, and neurotransmitters.Citation110 These processes are regulated by cell-autonomous and intercellular programs that mediate responses of neuronal cells to environmental input. By generating energy and regulating subcellular Ca2+ and redox homeostasis, mitochondria may play important roles in controlling fundamental plasticity processes,Citation112 including neuronal and synaptic differentiation, neurite outgrowth, neurotransmitter release, and dendritic remodeling. Receptor protein tyrosine phosphorylase ς (RPTPς) regulates synapse structure, function, and plasticity.Citation113 Emerging data suggest that mitochondria emit molecular signals, eg, reactive oxygen species, proteins, and lipid mediators that can act locally or travel to distant targets. Disorders in mitochondrial functions and signalling may play roles in impaired neuroplasticity and neurodegeneration.Citation114,Citation115

Both aging and Aβ that as a normal product of neuronal metabolism has an essential regulatory function at the synapse, independently decrease neuronal plasticity.Citation116 The major growth of Aβ burden occurs during a preclinical stage of AD, prior to the onset of AD-related symptoms.Citation117 It is associated with lower cognitive performance both in AD patients and normal elderly, but the association is modified by cognitive reserve, suggesting that this may be protective against amyloid-related cognitive impairment.Citation80 On the other hand, endogenous Aβ is necessary for hippocampal plasticity and memory within the normal CNS, due to regulation of transmitter release, activation of nicotinic acetylcholine receptors, and Aβ-42 production. The basis of age-related toxicity partly resides in mitochondrial dysfunction and an oxidative shift in mitochondrial and cytoplasmic redox potential. In turn, signaling through phosphorylated extracellular signal-regulated protein kinases is affected along with an age-independent increase in phosphorylated cyclic adenosine monophosphate (cAMP) response element-binding protein.Citation118 Furthermore, the production of inflammatory mediators (inflammatory cytokines, interleukins, neurotrophins), activation of glia and other immune cells disrupting the delicate balance needed for the physiological action of immune processes produces direct effects on neural plasticity and neurogenesis, facilitating many forms of neuropathology associated with normal aging as well as neurodegenerative diseases.Citation119 Recent evidence shows that key regulations of communication between neuron and microglia disruption in the aged brain may be one of the factors that precedes and initiates the increase in chronic inflammatory states underlying age-related impairments of cognition and hippocampal neurogenesis.Citation120 Effective treatments that dampen inflammatory activity are expected to have beneficial effects on cognitive performance and neural plasticity.Citation121

Functional recovery of synaptic circuitry requires that reactive synaptogenesis not exacerbate dysfunction, since aberrant misconnection by innervating the wrong target may cause misguided synaptogenesis, and inhibition of sprouting may be protective by sequestering dysfunctional neurons. Hippocampal synaptic plasticity in AD has been observed in transgenic models.Citation25 Aberrant, excessive, insufficient, or mistimed plasticity may represent the pathogenic cause of neurodevelopmental and neurodegenerative disorders.Citation122 Neuroplasticity is impaired in patients with AD and PD as a result of diminished growth factor expressionCitation123 and failure of delayed nonsynaptic neural plasticity mechanisms.Citation124

Understanding normative changes in brain structure that occur as a result of environmental changes is pivotal to understanding the ability of the brain to adapt.Citation125 Neuroplastic changes in cerebral gray matter may be induced by training.Citation126 Studies in animals and humans revealed dramatic effects of environmental enrichment, increased physical exercise documenting positive effects of mental and physical exercise, mediating brain and cognitive reserve,Citation127-Citation129 thus showing no compromise in daily life despite higher Aβ plaque load.Citation130 Other studies in animal models showed preventive or therapeutic action of environmental enrichment counteracting Aβ pathology by different molecular mechanismsCitation131 and by mitigating Alzheimer-like pathology, and increasing synaptic immunore activityCitation132,Citation133 due to reduction of cerebral oxidative stress.Citation134

Examination of synaptic physiology revealed that environmental experience significantly enhanced axonal transport in hippocampal and cortical neurons after enrichment, enhanced hippocampus long-term potentiation, without notable alterations in synaptic transmission. These data suggest that environmental modulation can rescue the impaired phenotype of the AD brain and that induction of brain plasticity may represent therapeutic and preventive avenues in AD.Citation135,Citation136

Recent studies demonstrated that the magnitude of the contribution of education is greater than the negative impact of either neuropathological burden of AD or CVLs with standardized regression weights of -0.14 for hyperintensities and -0.20 for hippocampal atrophy.Citation137 However, a large clinicopathologic study at 27 AD centers found no evidence of larger education-related differences in cognitive function when AD pathology was more advanced, suggesting that the advances of cognitive reserve may ultimately be overwhelmed by AD pathology.Citation138

Neurogenesis in the aging brain

Neurogenesis or the birth of new neural cells was thought to occur only in the developing nervous system, but recent studies have demonstrated that it does indeed continue into and throughout adult life. However, the age of olfactory bulb neurones, that are assumed to be derived from neuroblasts via the rostral migratory stream (RMS), has been assessed recently by measuring the levels of nuclear bomb test-derived 14C in genomic DNA. Data from this study suggest that there is very limited, if any, postnatal neurogenesis in the human olfactory bulb.Citation139 Certain areas of the brain may retain pluripotent precursors with the capacity to self-renew and differentiate into new neural lineages in adult mammals, nonhuman primates,Citation140 and humans.Citation141 Physical activity causes a robust increase in neurogenesis in the dentate gyrus of the hippocampus, a process that would implement a form of network plasticity analogous to that at the synaptic level, but occurring at the cellular network level.Citation142,Citation143

Neurogenesis represents a key factor of adult brain to response to environmental stimuli,Citation144 and abnormalities in neurogenesis have been detected in neurodegenerative disorders such as AD.Citation145,Citation146 It occurs in the subventricular zone and the subgranuiar layer of the hippocampus, and follows a multistep process probably in five stages, including proliferation, differentiation, migrating, targeting, and integration phases, respectively.Citation147,Citation148 Stimuli that entail an increase in neuronal activity have been shown to stimulate neurogenesis and enhance survival of new neurons in the adult mammalian hippocampus.Citation149 The incorporation of functional adult-generated neurons into existing neural networks provides higher capacity for plasticity, while they favor the encoding and storage of certain types of memories.Citation150-Citation152 Although neurogenesis continues throughout life, its rate declines with increasing age,Citation153,Citation154 and the proportion of neuronal stem cells that survive to become mature neuronal ceils is reduced.Citation155 This may be due to intrinsic decline in neuronal stem cell responsiveness to stimulating environmental cues, to a decrease in or disappearance of these environmental cues, or to accumulation of inhibitory factors.Citation156 Intrinsic properties of neural progenitor cells such as gene transcription and telomere activity change with age, which may contribute to decline in neurogenesis. While most studies indicated a correlation between decreased hippocampal neurogenesis and impaired performance in hippocampus-dependent cognitive tasks in age mice, few have demonstrated that young and aged mice are equivalent in their cognitive ability. The lack of neuronal ability to divide may be overcome by replacing damaged neurons or by restoring their function. Thus, Kittappa et alCitation157 revisited the molecular mechanisms responsible for neuronal renewal from stem cells, which are present in specific niches within the adult brain. The authors provided the novel notion that even non-terminaliy differentiated neural stem cells play roles in the regeneration of neurons and their synaptic function by mechanisms beyond mere cell replacement. These cells signal specific survival pathways that are worth investigating in search for novel therapeutic strategies against neurodegeneration. According to this notion, noninvasive tools to follow up synaptic function in the living brain are therefore essential for our better understanding of neuronal regeneration.

Although neuronal turnover is reduced in every neurogenic region of the aged brain, neuronal precursor cells clearly survive, remain responsive to growth factors and other physiological stimuli, and can increase their activity in response to damage.Citation157 Exploration of the regulation of neuronal progenitor cells in the aging brain is critical not only for understanding age-related cognitive deficits, but also for progress toward the goal of using the brain's regenerative potential to restore functional loss. Dysregulated or impaired neurogenesis may compromise plasticity and neuronal function in the hippocampus and other neuronal systems, and exacerbate neuronal vulnerability. Interestingly, increasing evidence suggests that molecular players in AD, including presenilin1, amyloid precursor protein, and its metabolites, play a role in adult neurogenesis, while alterations in tau phosporylation may interfere with the potential role of tau proteins in neuronal maturation and differentiation. This indicates a crosstalk between signaling molecules involved in both neurogenesis and neurodegeneration, and the ways by which AD-linked dysfunction of these signaling molecules affect neurogenesis in the adult brain.Citation158,Citation159 In AD, both increased and decreased neurogenesis has been reported and cholinergic activity may be involved in neurogenesis. However, most of these new neurons die, and fibrillar Aβ-42 seems to be involved in generating an inappropriate environment for those neurons to mature. These findings open up prospects for new strategies that can increase neurogenesis in pathologic processes in the aging brain.Citation160 Recent studies confirming the assumption that cholinergic pathology has a detrimental influence on neurogenesisCitation161 suggest an attenuation of stem cells together with compensatory increased proliferation that, however, does not result in an increased number of migratory neuroblasts and differentiated neurons in AD.Citation162

There are indications that neurogenesis is impaired in PD, which might be due to a lack of dopamine in the subventricular zone, but recent studies did not find evidence that dopamine has a direct effect on human stem cell proliferation in vitro. Thus, it was concluded that the number of adult neural stem cells is probably not diminished, and the proliferative capacity of the subventricular zone is maintained in the parkinsonian brain.Citation163 Neural stem cells have been identified also in areas where neurogenesis does not occur under physiological conditions, such as the midbrain and striatum, suggesting that they may have the potential to be used as a non-invasive cell replacement therapy in PD. Recent studies have shown that the deleterious effects of α-synuclein on newly generated neurons, in particular on their dendritic outgrowth and spine development, thus having negative impact on adult neurogenesis and neuronal maturation.Citation164 Further elucidation of the mechanisms regulating the synaptic integration of adult-born neurons is not only crucial for our understanding of the age- and disease-related neuroplasticity/brain plasticity, but also provides a framework for the manipulation and monitoring of endogenous adult neurogenesis as well as grafted cells potential therapeutic applications.Citation165-Citation167

Conclusions and outlook

A major problem in studying aging is how to separate the effects of aging from disease. Cerebral aging is a complex and heterogenous process that is associated with a high variety of molecular interactions, morphological, and functional changes, summarized in Table I. The interrelations between them need further elucidation. Brain aging results in loss of synapses and possible neurons, which is associated with structural changes in cerebral areas and neural neworks that are essential for cognitive and memory function. Many cognitively unimpaired eldery subjects are involved by Alzheimer-related or other pathologies of various severity and extent. Knowing the substrate of the resilience to cognitive decline in the presence of abundant AD and/or mixed pathology might be crucial not only for the understanding of the pathophysiology of nondemented aged people, but also to discover new prophylactic and/or therapeutic targets for aging processes. As expected from the significant clinicopathologic correlations of synaptic and neuronal loss in AD, “high-pathology nondemented” controls have preserved densities of synaptophysinlabeled presynaptic terminals and dendritic spines as compared with AD dementia patients with a similar burden of plaques and tangles.Citation99,Citation168 Greater amounts of specific presynaptic proteins and distinct protein-protein intreactions may be components of cognitive reserve that reduce the risk of dementia with aging.Citation168 They may have no significant neuronal loss, not even in vulnerable regions, such as the entorhinal cortex and hippocampus,Citation54,Citation169 and have lower levels of neuroinflammatory markers than pathology-matched AD patients.Citation170 This resistance to AD pathology has also been related to a nucleolar, nuclear, and cell body hypertrophy of the hippocampal and cortical neurons, suggestive of a compensatory metabolic activation to face the neurotoxic effects of AD lesions.Citation108,Citation171 Resilience to AD is also attributed to genetic factors, particularly apolipoprotein E2 and combinations of other genetic polymorphisms.Citation172 Premorbid brain volume has been found to provide protection against clinical manifestation of dementia despite evidence of AD pathology, supporting the brain reserve hypothesis of resilience to AD.Citation173 Although multiple factors and possible interventions may influence cognitive reserve and susceptibility to dementia, much work is required on the mechanisms of action in order to determine which, if any, may improve the clinical and epidemiological picture.Citation174 On the other hand, the unique observation of a cognitively intact woman aged 115 years with only slight tau pathology corresponding to Braak stage II, almost no plaques or vascular changes, and normal neuron count in the locus ceruleus indicates that the limits of human cognitive function extend far beyond the range that is currently enjoyed by most individuals and that brain disease, even in supercentenarians, is not inevitable.Citation175 The association between “vulnerability” and “protective” factors varies with age, since the effects of these factors on the risk for AD may differ in younger (age <80) versus older (age >80) individuals. The understanding of the dynamic of these factors at different age periods will be essential for the implementation of primary prevention treatments for AD.Citation176

TABLE I. Summary of key points on cerebral aging.

The importance of understanding ageing and the complex interplay of multiple influences on successful cognitive ageing is clear.Citation176,Citation177 Understanding how brain reserve might be influenced to minimize the impact of neuropathologies associated with dementia could have enormous public health implications. This is a crucial prerequisite to meaningful research in dementia and illustrates how life-long intellectual engagement can mitigate the negative impact of brain pathology even on healthy ageing.Citation178 The neuronal underpinning of the dynamic compensatory mechanism opens the possibility for strategic interventions based on environmental approaches.

Future work should measure the contribution of more diverse influences on cognitive reserve that might operate in early and midlife, such as socioeconomic conditions and social relationships, which might be modified through public education in order to have a positive impact on the looming public health disaster that is dementia. Recent studies in a nondemented population have shown that intellectual and physical activity lifestyle factors were not assessed with AD biomarkers, while intellectual lifestyle factors explained the variability in the cognitive performance, providing evidence that lifestyle activities may delay the onset of dementia, but do not significantly influence the expression of AD pathophysiology.Citation179

The neuropathological distinction between nondemented, cognitively intact, and cognitively impaired/demented subjects, elucidation of the relationship of additional pathologies with minor—often clinically latent—AD lesions observed in many but not all elderly persons without cognitive impairment is important, allowing further insights into the mechanisms of brain plasticity and the basic mechanisms of adult neurogenesis warrants further experimental and prospective, well documented clinico-pathological studies of elderly individuals. In this continuously growing field, new acquisitions, derived from basic research and clinical grounds, on cognitive reserve mechanisms, neuroplasticity, and the potential application of novel therapeutic targets in neurodegeneration and aging disorders are necessary.Citation180 As a basis for potential prophylactic and therapeutic options for brain aging, they are major challenges for modern neurosciences.

Acknowledgments: The authors thank many colleagues from clinical departments and the Institute of Pathology, Otto Wagner Hospital, Vienna, for clinical and autopsy data, and Mr E. Mitter-Ferstl, PhD, for secretarial and computer work. The study was supported by the Society for Support of Research in Experimental Neurology, Vienna, Austria.

Conflicts of interest: The authors have nothing to disclose.

REFERENCES

  • BishopNA.LuT.YanknerBA.Neural mechanisms of ageing and cognitive decline.Nature.201046452953520336135
  • MattsonMP.MagnusT.Ageing and neuronal vulnerability.Nat Rev Neurosci.2006727829416552414
  • DicksteinDL.KabasoD.RocherAB.LuebkeJI.WearneSL.HofPR.Changes in the structural complexity of the aged brain.Aging Cell.2007627528417465981
  • Singh-ManouxA.KivimakiM.GlymourMM.et alTiming of onset of cognitive decline: results from Whitehall II prospective cohort study.BMJ.20113447622
  • AanerudJ.BorghammerP.ChakravartyMM.et alBrain energy metabolism and blood flow differences in healthy aging.J Cereb Blood Flow Metab.2012321177118722373642
  • MorrisonJH.BaxterMG.The ageing cortical synapse: hallmarks and implications for cognitive decline.Nat Rev Neurosci.20121324025022395804
  • BaliettiM.TamagniniF.FattorettiP.et alImpairments of synaptic plasticity in aged animals and in animal models of Alzheimer's disease.Rejuvenation Res.20121523523822533439
  • MatsuiC.InoueE.KakitaA.et alInvolvement of the γ-secretase-mediated EphA4 signaling pathway in synaptic pathogenesis of Alzheimer's disease.Brain Pathol.20122277678722404518
  • KoffieRM.HashimotoT.TaiHC.et alApolipoprotein E4 effects in Alzheimer's disease are mediated by synaptotoxic oligomeric amyloid-beta.Brain.20121352155216822637583
  • BeeriMS.HaroutunianV.SchmeidlerJ.et alSynaptic protein deficits are associated with dementia irrespective of extreme old age.Neurobiol Aging.2012331125.e11125.e822206847
  • PievaniM.de HaanW.WuT.SeeleyWW.FrisoniGB.Functional network disruption in the degenerative dementias.Lancet Neurol.20111082984321778116
  • SchroeterML.VogtB.FrischS.et alExecutive deficits are related to the inferior frontal junction in early dementia.Brain.201213520121522184615
  • RajA.KuceyeskiA.WeinerM.A network diffusion model of disease progression in dementia.Neuron.2012731204121522445347
  • EylerLT.SherzaiA.KaupAR.JesteDV.A review of functional brain imaging correlates of successful cognitive aging.Biol Psychiatry.20117011512221316037
  • van VeluwSJ.SawyerEK.CloverL.et alPrefrontal cortex cytoarchitecture in normal aging and Alzheimer's disease: a relationship with IQ.Brain Struct Funct. In press. DOI: 10.1007/s00429-00012-00381-x.
  • PiguetO.DoubleKL.KrilJJ.et alWhite matter loss in healthy ageing: a postmortem analysis.Neurobiol Aging.2009301288129518077060
  • McAleeseKE.FirbankM.HallR.et alMagnetic resonance imaging detects subcortical vascular pathology in post-mortem brains.Neuropathol Appl Neurobiol.201238(suppl)748(O32)22276301
  • MalykhinN.VahidyS.MichielseS.et alStructural organization of the prefrontal white matter pathways in the adult and aging brain measured by diffusion tensor imaging.Brain Struct Funct.201121641743121559982
  • RajiCA.LopezOL.KullerLH.et alWhite matter lesions and brain gray matter volume in cognitively normal elders.Neurobiol Aging.201233834e83781621943959
  • KullerLH.ArnoldAM.LongstrethWT.Jr.et alWhite matter grade and ventricular volume on brain MRI as markers of longevity in the cardiovascular health study.Neurobiol Aging.2007281307131516857296
  • De VogelaereF.SantensP.AchtenE.BoonP.VingerhoetsG.Altered default-mode network activation in mild cognitive impairment compared with healthy aging.Neuroradiology.2012541195120622527687
  • LeeMH.HackerCD.SnyderAZ.et alClustering of resting state networks.PLoS One.20127e4037022792291
  • DuAT.SchuffN.ChaoLL.et alAge effects on atrophy rates of entorhinal cortex and hippocampus.Neurobiol Aging.20062773374015961190
  • WangM.GamoNJ.YangY.et alNeuronal basis of age-related working memory decline.Nature.201147621021321796118
  • MarchettiC.MarieH.Hippocampal synaptic plasticity in Alzheimer's disease: what have we learned so far from transgenic models?Rev Neurosci.20112237340221732714
  • KaupAR.MirzakhanianH.JesteDV.EylerLT.A review of the brain structure correlates of successful cognitive aging.J Neuropsychiatry Clin Neurosci.20112361521304134
  • HornbergerM.WongS.TanR.et alIn vivo and post-mortem memory circuit integrity in frontotemporal dementia and Alzheimer's disease.Brain.20121353015302523012333
  • HanggiJ.StrefferJ.JanckeL.HockC.Volumes of lateral temporal and parietal structures distinguish between healthy aging, mild cognitive impairment, and Alzheimer's disease.J Alzheimers Dis.20112671973421709375
  • GouwAA.SeewannA.van der FlierWM.et alHeterogeneity of small vessel disease: a systematic review of MRI and histopathology correlations.J Neurol Neurosurg Psychiatry.20118212613520935330
  • SchmidtR.SchmidtH.HaybaeckJ.et alHeterogeneity in age-related white matter changes.Acta Neuropathol.201112217118521706175
  • WangL.GoldsteinFC.LeveyAl.et alWhite matter hyperintensities and changes in white matter integrity in patients with Alzheimer's disease.Neuroradiology.20115337338121152911
  • JacobsHI.van BoxtelMP.GronenschildEH.UylingsHB.JollesJ.VerheyFR.Decreased gray matter diffusivity: a potential early Alzheimer's disease biomarker?Alzheimers Dement.20129939722651939
  • BeckerJA.HeddenT.CarmasinJ.et alAmyloid-beta associated cortical thinning in clinically normal elderly.Ann Neurol.2011691032104221437929
  • TeterB.AshfordJW.Neuroplasticity in Alzheimer's disease.J Neurosci Res.20027040243712391603
  • DoehnerJ.GenoudC.ImhofC.KrsticD.KnueselI.Extrusion of misfolded and aggregated proteins - a protective strategy of aging neurons?Eur J Neurosci.2012351938195022708604
  • SchneiderJA.AggarwalNT.BarnesL.BoyleP.BennettDA.The neuropathology of older persons with and without dementia from community versus clinic cohorts.J Alzheimers Dis.20091869170119749406
  • JellingerKA.AttemsJ.Neuropathology and general autopsy findings in nondemented aged subjects.Clin Neuropathol.201231879822385790
  • O'BrienRJ.ResnickSM.ZondermanAB.et alNeuropathologic studies of the Baltimore Longitudinal Study of Aging (BLSA).J Alzheimers Dis.20091866567519661626
  • DuyckaertsC.DelatourB.PotierMC.Classification and basic pathology of Alzheimer disease.Acta Neuropathol.200911853619381658
  • NelsonPT.JichaGA.SchmittFA.et alClinicopathologic correlations in a large Alzheimer disease center autopsy cohort: neuritic plaques and neurofibrillary tangles “do count” when staging disease severity.J Neuropathol Exp Neurol.2007661136114618090922
  • NelsonPT.BraakH.MarkesberyWR.Neuropathology and cognitive impairment in Alzheimer disease: a complex but coherent relationship.J Neuropathol Exp Neurol.20096811419104448
  • NelsonPT.AlafuzoffI.BigioEH.et alCorrelation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature.J Neuropathol Exp Neurol.20127136238122487856
  • Serrano-PozoA.FroschMP.MasliahE.HymanBT.Neuropathological Alterations in Alzheimer Disease.Cold Spring Harb Perspect Med.20111a00618922229116
  • ScheffSW.PriceDA.Synapse loss in the temporal lobe in Alzheimer's disease.Ann Neurol.1993331901998434881
  • ScheffSW.PriceDA.SchmittFA.MufsonEJ.Hippocampal synaptic loss in early Alzheimer's disease and mild cognitive impairment.Neurobiol Aging.2006271372138416289476
  • ScheffSW.PriceDA.SchmittFA.DeKoskyST.MufsonEJ.Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment.Neurology.2007681501150817470753
  • TerryR.HansenL.MasliahE.Structural basis of the cognitive alterations in Alzheimer disease. In: Terry R, Katzman R, eds.Alzheimer Disease. New York, NY: Raven;1994179196
  • ArriagadaPV.MarzloffK.HymanBT.Distribution of Alzheimer-type pathologic changes in nondemented elderly individuals matches the pattern in Alzheimer's disease.Neurology.199242168116881307688
  • SantacruzKS.SonnenJA.PezhouhMK.DesrosiersMF.NelsonPT.TyasSL.Alzheimer disease pathology in subjects without dementia in 2 studies of aging: the nun study and the adult changes in thought study.J Neuropathol Exp Neurol.20117083284021937909
  • MitchellTW.MufsonEJ.SchneiderJA.et alParahippocampal tau pathology in healthy aging, mild cognitive impairment, and early Alzheimer's disease.Ann Neurol.20025118218911835374
  • DavisDG.SchmittFA.WeksteinDR.MarkesberyWR.Alzheimer neuropathologic alterations in aged cognitively normal subjects.J Neuropathol Exp Neurol.19995837638810218633
  • KnopmanDS.ParisiJE.SalviatiA.et alNeuropathology of cognitively normal elderly.J Neuropathol Exp Neurol.2003621087109514656067
  • JentoftM.ParisiJ.DicksonD.et alNeuropathologic findings in 32 non-demented elderly subjects.J Neuropathol Exp Neurol.201170531
  • PriceJL.McKeelDW.Jr.BucklesVD.et alNeuropathology of nondemented aging: presumptive evidence for preclinical Alzheimer disease.Neurobiol Aging.2009301026103619376612
  • MorrisJC.StorandtM.McKeelDW.Jr.et alCerebral amyloid deposition and diffuse plaques in “normal” aging: Evidence for presymptomatic and very mild Alzheimer's disease.Neurology.1996467077198618671
  • SchmittFA.DavisDG.WeksteinDR.SmithCD.AshfordJW.MarkesberyWR.“Preclinical” AD revisited: neuropathology of cognitively normal older adults.Neurology.20005537037610932270
  • WhiteL.Brain lesions at autopsy in older Japanese-American men as related to cognitive impairment and dementia in the final years of life: a summary report from the Honolulu-Asia aging study.J Alzheimers Dis.20091871372519661625
  • GalvinJE.PowlishtaKK.WilkinsK.et alPredictors of preclinical Alzheimer disease and dementia: a clinicopathologic study.Arch Neurol.20056275876515883263
  • BraakH.BraakE.Neuropathological stageing of Alzheimer-related changes.Acta Neuropathol.1991822392591759558
  • TroncosoJC.CataldoAM.NixonRA.et alNeuropathology of preclinical and clinical late-onset Alzheimer's disease.Ann Neurol.1998436736769585365
  • PriceJL.MorrisJC.Tangles and plaques in nondemented aging and “preclinical” Alzheimer's disease.Ann Neurol.19994535836810072051
  • HaroutunianV.PerlDP.PurohitDP.et alRegional distribution of neuritic plaques in the nondemented elderly and subjects with very mild Alzheimer disease.Arch Neurol.199855118511919740112
  • HampelH.ShenY.WalshDM.et alBiological markers of amyloid beta-related mechanisms in Alzheimer's disease.Exp Neurol.201022333434619815015
  • WolkDA.KlunkW.Update on amyloid imaging: from healthy aging to Alzheimer's disease.Curr Neurol Neurosci Rep.2009934535219664363
  • JohnsonKA.FoxNC.SperlingRA.KlunkWE.Brain imaging in Alzheimer disease.Cold Spring Harb Perspect Med.20122a00621322474610
  • VillemagneVL.PikeKE.DarbyD.et alAbeta deposits in older nondemented individuals with cognitive decline are indicative of preclinical Alzheimer's disease.Neuropsychologia.2008461688169718343463
  • SojkovaJ.ZhouY.AnY.et alLongitudinal patterns of beta-amyloid deposition in nondemented older adults.Arch Neurol.20116864464921555640
  • StomrudE.HanssonO.ZetterbergH.BlennowK.MinthonL.LondosE.Correlation of longitudinal cerebrospinal fluid biomarkers with cognitive decline in healthy older adults.Arch Neurol.20106721722320142530
  • AbnerEL.KryscioRJ.SchmittFA.et al“End-stage” neurofibrillary tangle pathology in preclinical Alzheimer's disease: fact or fiction?J Alzheimers Dis.20112544545321471646
  • KramerPL.XuH.WoltjerRL.et alAlzheimer disease pathology in cognitively healthy elderly: a genome-wide study.Neurobiol Aging.2011322113212220452100
  • BeachTG.SueLI.WalkerDG.et alStriatal amyloid plaque density predicts Braak neurofibrillary stage and clinicopathological Alzheimer's disease: implications for amyloid imaging.J Alzheimers Dis.20112886987622112552
  • RobinsonJL.GeserF.CorradaMM.et alNeocortical and hippocampal amyloid-beta and tau measures associate with dementia in the oldest-old.Brain.20111343708371522120149
  • BennettDA.WilsonRS.BoylePA.BuchmanAS.SchneiderJA.Relation of neuropathology to cognition in persons without cognitive impairment.Ann Neurol.20127259960923109154
  • MaaroufCL.DaugsID.KokjohnTA.et alAlzheimer's disease and nondemented high pathology control nonagenarians: comparing and contrasting the biochemistry of cognitively successful aging.PLoS One.20116e2729122087282
  • PetersenRC.RobertsRO.KnopmanDS.et alMild cognitive impairment: ten years later.Arch Neurol.2009661447145520008648
  • JellingerKA.Is there a gold standard for the pathological diagnosis of dementing disorders: routes out of the swamp?Am J Neuroprotec Neuroregen.20102317
  • MooreBD.ChakrabartyP.LevitesY.et alOverlapping profiles of Abeta peptides in the Alzheimer's disease and pathological aging brains.Alzheimers Res Ther.201241822621179
  • ShinoharaM.PetersenRC.DicksonDW.BuG.Brain regional correlation of amyloid-beta with synapses and apolipoprotein E in non-demented individuals: potential mechanisms underlying regional vulnerability to amyloid-beta accumulation.Acta Neuropathol. In press
  • JonssonT.AtwalJK.SteinbergS.et alA mutation in APP protects against Alzheimer's disease and age-related cognitive decline.Nature.2012488969922801501
  • RentzDM.LocascioJJ.BeckerJA.et alCognition, reserve, and amyloid deposition in normal aging.Ann Neurol.20106735336420373347
  • ResnickSM.SojkovaJ.Amyloid imaging and memory change for prediction of cognitive impairment.Alzheimers Res Ther.20113321345176
  • BarnesLL.SchneiderJA.BoylePA.BieniasJL.BennettDA.Memory complaints are related to Alzheimer disease pathology in older persons.Neurology.2006671581158517101887
  • NelsonPT.HeadE.SchmittFA.et alAlzheimer's disease is not “brain aging”: neuropathological, genetic, and epidemiological human studies.Acta Neuropathol.201112157158721516511
  • SwerdlowRH.Is aging part of Alzheimer's disease, or is Alzheimer's disease part of aging?Neurobiol Aging.2007281465148016876913
  • CastellaniRJ.ZhuX.LeeHG.SmithMA.PerryG.Molecular pathogenesis of Alzheimer's disease: Reductionist versus expansionist approaches.Int J Mol Sci.2009101386140619399255
  • SonnenJA.Santa CruzK.HemmyLS.et alEcology of the aging human brain.Arch Neurol.2011681049105621825242
  • BuchmanAS.LeurgansSE.NagS.BennettDA.SchneiderJA.Cerebrovascular disease pathology and parkinsonian signs in old age.Stroke.2011423183318921885844
  • StephanBC.MatthewsFE.MaB.et alAlzheimer and vascular neuropathological changes associated with different cognitive States in a nondemented sample.J Alzheimers Dis.20122930931822233761
  • MahnckeHW.BronstoneA.MerzenichMM.Brain plasticity and functional losses in the aged: scientific bases for a novel intervention.Prog Brain Res.20061578110917046669
  • Bartres-FazD.Arenaza-UrquijoEM.Structural and functional imaging correlates of cognitive and brain reserve hypotheses in healthy and pathological aging.Brain Topogr.20112434035721853422
  • MortimerJA.Important role of brain reserve in lowering the risk of Alzheimer's disease. Editorial.Future Neurol.2009414
  • SternY.Cognitive reserve.Neuropsychologia.2009472015202819467352
  • StranahanAM.MattsonMP.Metabolic reserve as a determinant of cognitive aging.J Alzheimers Dis.201230S5S1322045480
  • SternY.Cognitive reserve in ageing and Alzheimer's disease.Lancet Neurol.2012111006101223079557
  • MengX.D'ArcyC.Education and dementia in the context of the cognitive reserve hypothesis: a systematic review with meta-analyses and qualitative analyses.PLoS One.20127e3826822675535
  • BellevilleS.ClementF.MellahS.GilbertB.FontaineF.GauthierS.Training-related brain plasticity in subjects at risk of developing Alzheimer's disease.Brain.20111341623163421427462
  • GreenwoodPM.ParasuramanR.Neuronal and cognitive plasticity: a neurocognitive framework for ameliorating cognitive aging.Front Aging Neurosci.2010215021151819
  • RoeCM.XiongC.MillerJP.MorrisJC.Education and Alzheimer disease without dementia: support for the cognitive reserve hypothesis.Neurology.20076822322817224578
  • ArnoldSE.LounevaN.CaoK.et alCellular, synaptic, and biochemical features of resilient cognition in Alzheimer's disease.Neurobiol Aging.20133415716822554416
  • BrownJ.Cooper-KuhnCM.KempermannG.et alEnriched environment and physical activity stimulate hippocampal but not olfactory bulb neurogenesis.Eur J Neurosci.2003172042204612786970
  • SternY.AlbertS.TangMX.TsaiWY.Rate of memory decline in AD is related to education and occupation: cognitive reserve?Neurology.1999531942194710599762
  • SteffenerJ.SternY.Exploring the neural basis of cognitive reserve in aging.Biochim Biophys Acta.2012182246747321982946
  • GohJO.ParkDC.Neuroplasticity and cognitive aging: the scaffolding theory of aging and cognition.Restor Neurol Neurosci.20092739140319847066
  • Sole-PadullesC.Bartres-FazD.JunqueC.et alBrain structure and function related to cognitive reserve variables in normal aging, mild cognitive impairment and Alzheimer's disease.Neurobiol Aging.2009301114112418053618
  • GreenwoodPM.Functional plasticity in cognitive aging: review and hypothesis.Neuropsychology.20072165767317983277
  • TuckerAM.SternY.Cognitive reserve in aging.Curr Alzheimer Res.2011835436021222591
  • Erten-LyonsD.WoltjerRL.DodgeH.et alFactors associated with resistance to dementia despite high Alzheimer disease pathology.Neurology.20097235436019171833
  • laconoD.O'BrienR.ResnickSM.et alNeuronal hypertrophy in asymptomatic Alzheimer disease.J Neuropathol Exp Neurol.20086757858918520776
  • Bartrés-FazD.Arenaza-UrquijoEM.Structural and functional imaging correlates of cognitive and brain reserve hypotheses in healthy and pathological aging.Brain Topogr.20112434035721853422
  • KimMJ.LeeKM.SonYD.JeonHA.KimYB.ChoZH.Increased basal forebrain metabolism in mild cognitive impairment: an evidence for brain reserve in incipient dementia.J Alzheimers Dis.20123292793822903128
  • RobertsonIH.A noradrenergic theory of cognitive reserve: implications for Alzheimer's disease.Neurobiol Aging.20133429830822743090
  • PuzzoD.PriviteraL.FaM.et alEndogenous amyloid-beta is necessary for hippocampal synaptic plasticity and memory.Ann Neurol.20116981983021472769
  • HornKE.XuB.GobertD.et alReceptor protein tyrosine phosphatase sigma regulates synapse structure, function and plasticity.J Neurochem.201212214716122519304
  • MattsonMP.GleichmannM.ChengA.Mitochondria in neuroplasticity and neurological disorders.Neuron.20086074876619081372
  • ChengA.HouY.MattsonMP.Mitochondria and neuroplasticity.ASN Neuro.20102e0004520957078
  • MaT.KlannE.Amyloid beta: linking synaptic plasticity failure to memory disruption in Alzheimer's disease.J Neurochem.2012120(suppl 1)14014822122128
  • VlassenkoAG.MintunMA.XiongC.et alAmyloid-beta plaque growth in cognitively normal adults: longitudinal [11C]Pittsburgh compound B data.Ann Neurol.20117085786122162065
  • PariharMS.BrewerGJ.Amyloid-beta as a modulator of synaptic plasticity.J Alzheimers Dis.20102274176320847424
  • YirmiyaR.GoshenI.Immune modulation of learning, memory, neural plasticity and neurogenesis.Brain Behav Immun.20112518121320970492
  • GemmaC.BachstetterAD.BickfordPC.Neuron-microglia dialogue and hippocampal neurogenesis in the aged brain.Aging Dis.2010123224421961084
  • KohmanRA.Aging microglia: relevance to cognition and neural plasticity.Methods Mol Biol.201293419321822933148
  • FoscarinS.RossiF.CarulliD.Influence of the environment on adult CNS plasticity and repair.Cell Tissue Res.201234916116722143260
  • EnciuAM.NicolescuMl.ManoleCG.MuresanuDF.PopescuLM.PopescuBO.Neuroregeneration in neurodegenerative disorders.BMC Neurol.2011117521699711
  • WatsonSN.RislingTE.HermannPM.WilderingWC.Failure of delayed nonsynaptic neuronal plasticity underlies age-associated long-term associative memory impairment.BMC Neurosci.20121310322898271
  • MayA.Experience-dependent structural plasticity in the adult human brain.Trends Cogn Sci.20111547548221906988
  • DraganskiB.GaserC.BuschV.SchuiererG.BogdahnU.MayA.Neuroplasticity: changes in grey matter induced by training.Nature.200442731131214737157
  • FosterPP.RosenblattKP.KuljisRO.Exercise-induced cognitive plasticity, implications for mild cognitive impairment and Alzheimer's disease.Front Neurol.201122821602910
  • HerringA.AmbreeO.TommM.et alEnvironmental enrichment enhances cellular plasticity in transgenic mice with Alzheimer-like pathology.Exp Neurol.200921618419219118549
  • NithianantharajahJ.HannanAJ.The neurobiology of brain and cognitive reserve: mental and physical activity as modulators of brain disorders.Prog Neurobiol.20098936938219819293
  • LewejohannL.ReefmannN.WidmannP.et alTransgenic Alzheimer mice in a semi-naturalistic environment: more plaques, yet not compromised in daily life.Behav Brain Res.20092019910219428622
  • HerringA.LewejohannL.PanzerAL.et alPreventive and therapeutic types of environmental enrichment counteract beta amyloid pathology by different molecular mechanisms.Neurobiol Dis.20114253053821406231
  • CracchioloJR.MoriT.NazianSJ.TanJ.PotterH.ArendashGW.Enhanced cognitive activity — over and above social or physical activity — is required to protect Alzheimer's mice against cognitive impairment, reduce Abeta deposition, and increase synaptic immunoreactivity.Neurobiol Learn Mem.20078827729417714960
  • HerringA.DonathA.YarmolenkoM.et alExercise during pregnancy mitigates Alzheimer-like pathology in mouse offspring.FASEB J.20122611712821948247
  • HerringA.BlomeM.AmbreeO.SachserN.PaulusW.KeyvaniK.Reduction of cerebral oxidative stress following environmental enrichment in mice with Alzheimer-like pathology.Brain Pathol.20102016617519134003
  • HuYS.XuP.PiginoG.BradyST.LarsonJ.LazarovO.Complex environment experience rescues impaired neurogenesis, enhances synaptic plasticity, and attenuates neuropathology in familial Alzheimer's diseaselinked APPswe/PS1DeltaE9 mice.FASEB J.2010241667168120086049
  • StranahanAM.MattsonMP.Recruiting adaptive cellular stress responses for successful brain ageing.Nat Rev Neurosci.20121320921622251954
  • MurrayAD.StaffRT.McNeilCJ.et alThe balance between cognitive reserve and brain imaging biomarkers of cerebrovascular and Alzheimer's diseases.Brain.20111343687369622102649
  • KoepsellTD.KurlandBF.HarelO.JohnsonEA.ZhouXH.KukullWA.Education, cognitive function, and severity of neuropathology in Alzheimer disease.Neurology.2008701732173918160675
  • BergmannO.LieblJ.BernardS.et alThe age of olfactory bulb neurons in humans.Neuron.20127463463922632721
  • MaslovAY.BaroneTA.PlunkettRJ.PruittSC.Neural stem cell detection, characterization, and age-related changes in the subventricular zone of mice.J Neurosci.2004241726173314973255
  • CurtisMA.ErikssonPS.FaullRL.Progenitor cells and adult neurogenesis in neurodegenerative diseases and injuries of the basal ganglia.Clin Exp Pharmacol Physiol.20073452853217439428
  • van PraagH.Neurogenesis and exercise: past and future directions.Neuromolecular Med.20081012814018286389
  • DeisserothK.SinglaS.TodaH.MonjeM.PalmerTD.MalenkaRC.Excitation-neurogenesis coupling in adult neural stem/progenitor cells.Neuron.20044253555215157417
  • GeS.GohEL.SailorKA.KitabatakeY.MingGL.SongH.GABA regulates synaptic integration of newly generated neurons in the adult brain.Nature.200643958959316341203
  • WinnerB.KohlZ.GageFH.Neurodegenerative disease and adult neurogenesis.Eur J Neurosci.2011331139115121395858
  • MarlattMW.LucassenPJ.Neurogenesis and Alzheimer's disease: biology and pathophysiology in mice and men.Curr Alzheimer Res.2010711312519860727
  • von Bohlen und HalbachO.Immunohistological markers for staging neurogenesis in adult hippocampus.Cell Tissue Res.200732940942017541643
  • von Bohlen und HalbachO.Immunohistological markers for proliferative events, gliogenesis, and neurogenesis within the adult hippocampus.Cell Tissue Res.201134511921647561
  • LeunerB.Mendolia-LoffredoS.KozorovitskiyY.SamburgD.GouldE.ShorsTJ.Learning enhances the survival of new neurons beyond the time when the hippocampus is required for memory.J Neurosci.2004247477748115329394
  • LeeSW.ClemensonGD.GageFH.New neurons in an aged brain.Behav Brain Res.201222749750722024433
  • MongiatLA.SchinderAF.Adult neurogenesis and the plasticity of the dentate gyrus network.Eur J Neurosci.2011331055106121395848
  • MuY.GageFH.Adult hippocampal neurogenesis and its role in Alzheimer's disease.Mol Neurodegener.201168522192775
  • KempermannG.Adult hippocampal neurogenesis. In: Kempermann G, ed.Adult Neurogenesis. 2nd ed. New York, NY: Oxford University Press2011185215
  • DemarsM.HuYS.GadadharA.LazarovO.Impaired neurogenesis is an early event in the etiology of familial Alzheimer's disease in transgenic mice.J Neurosci Res.2010882103211720209626
  • TangJ.XuHW.FanXT.et alTargeted migration and differentiation of engrafted neural precursor cells in amyloid beta-treated hippocampus in rats.Neurosci Bull.20072326327017952134
  • GalvanV.JinK.Neurogenesis in the aging brain.Clin Interv Aging.2007260561018225461
  • KittappaR.ChangWW.AwatramaniRB.McKayRD.The foxa2 gene controls the birth and spontaneous degeneration of dopamine neurons in old age.PLoS Biol.20075e32518076286
  • LazarovO.MarrRA.Neurogenesis and Alzheimer's disease: at the crossroads.Exp Neurol.201022326728119699201
  • RodriguezJJ.VerkhratskyA.Neurogenesis in Alzheimer's disease.J Anat.2011219788921323664
  • ShrusterA.MelamedE.OffenD.Neurogenesis in the aged and neurodegenerative brain.Apoptosis.2010151415142120339917
  • SchliebsR.ArendtT.The cholinergic system in aging and neuronal degeneration.Behav Brain Res.201122155556321145918
  • PerryEK.JohnsonM.EkonomouA.PerryRH.BallardC.AttemsJ.Neurogenic abnormalities in Alzheimer's disease differ between stages of neurogenesis and are partly related to cholinergic pathology.Neurobiol Dis.20124715516222504537
  • van den BergeSA.van StrienME.KoreckaJA.et alThe proliferative capacity of the subventricular zone is maintained in the parkinsonian brain.Brain.20111343249326322075520
  • WinnerB.RegensburgerM.SchreglmannS.et alRole of alpha-synuclein in adult neurogenesis and neuronal maturation in the dentate gyrus.J Neurosci.201232169061691623175842
  • ArtegianiB.CalegariF.Age-related cognitive decline: Can neural stem cells help us?Aging (Albany NY).2012417618622466406
  • ToniN.SultanS.Synapse formation on adult-born hippocampal neurons.Eur J Neurosci.2011331062106821395849
  • Abdel-SalamOM.Stem cell therapy for Alzheimer's disease.CNS Neurol Disord Drug Targets.20111045948521495961
  • HonerWG.BarrAM.SawadaK.et alCognitive reserve, presynaptic proteins and dementia in the elderly.Transl Psychiatry.20122e11422832958
  • WestMJ.KawasCH.StewartWF.RudowGL.TroncosoJC.Hippocampal neurons in pre-clinical Alzheimer's disease.Neurobiol Aging.2004251205121215312966
  • LueLF.BrachovaL.CivinWH.RogersJ.Inflammation, A beta deposition, and neurofibrillary tangle formation as correlates of Alzheimer's disease neurodegeneration.J Neuropathol Exp Neurol.199655108310888858005
  • RiudavetsMA.laconoD.ResnickSM.et alResistance to Alzheimer's pathology is associated with nuclear hypertrophy in neurons.Neurobiol Aging.2007281484149217599696
  • BerlauDJ.CorradaMM.HeadE.KawasCH.APOE epsilon2 is associated with intact cognition but increased Alzheimer pathology in the oldest old.Neurology.20097282983419255410
  • NegashS.XieS.DavatzikosC.et alCognitive and functional resilience despite molecular evidence of Alzheimer's disease pathology.Alzheimers Dement. In press
  • EsiriMM.ChanceSA.Cognitive reserve, cortical plasticity and resistance to Alzheimer's disease.Alzheimers Res Ther.20124722380508
  • den DunnenWF.BrouwerWH.BijlardE.et alNo disease in the brain of a 115-year-old woman.Neurobiol Aging.2008291127113218534718
  • DeppCA.HarmellA.VahiaIV.Successful cognitive aging.Curr Top Behav Neurosci.201210355022275006
  • DaffnerKR.Promoting successful cognitive aging: a comprehensive review.J Alzheimers Dis.2010191101112220308777
  • De CarliC.KawasC.MorrisonJH.Reuter-LorenzPA.SperlingRA.WrightCB.Session II: Mechanisms of age-related cognitive change and targets for intervention: neural circuits, networks, and plasticity.J Gerontol A Biol Sci Med Sci.20126774775322570135
  • VemuriP.LesnickTG.PrzybelskiSA.et alEffect of lifestyle activities on Alzheimer disease biomarkers and cognition.Ann Neurol.20127273073823280791
  • BorroniB.PremiE.BozzaliM.PadovaniA.Reserve mechanisms in neurodegenerative diseases: from bench to bedside and back again.Curr Med Chem.2012196112611823072353