Você está na página 1de 7

Journal of Pathology

J Pathol 2007; 211: 181–187


Published online in Wiley InterScience
(www.interscience.wiley.com) DOI: 10.1002/path.2089

Review Article

Ageing and the brain


MM Esiri*
Department of Clinical Neurology, University of Oxford, and Department of Neuropathology, Oxford Radcliffe NHS Trust, Oxford, UK

*Correspondence to: Abstract


MM Esiri, Neuropathology
Department, West Wing, John In this review, the evidence for changes in the human brain with ageing at both the
Radcliffe Hospital, Headington, macroscopic and microscopic levels is summarized. Loss of neurons is now recognized to
Oxford OX3 9DU, UK. be more modest than initial studies suggested and only affects some neuron populations.
E-mail: Accompanying loss of neurons is some reduction in the size of remaining neurons. This
margaret.esiri@clneuro.ox.ac.uk reflects a reduced size of dendritic and axonal arborizations. Some of the likely causes
of these changes, including free radical damage resulting from a high rate of oxidative
No conflicts of interest were
declared. metabolism in neurons, glycation and dysregulation of intracellular calcium homeostasis,
are discussed. The roles of genes and environmental factors in causing and responding to
ageing changes are explored.
Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John
Wiley & Sons, Ltd.
Keywords: human brain; ageing; oxidative damage; advanced glycation end products
(AGEs); calcium homeostasis

Our brains are seventy-year clocks. The Angel of Life the last 100 or so years as a result of improved
winds them up once for all, then closes the case, and gives nutrition during development. Thus, as the twentieth
the key into the hand of the Angel of Resurrection. [Oliver
Wendell Homes (1809–1894), physician and poet]
century progressed, young brains in the developed
world, where most measurements have been made,
became larger and heavier by about 1 g/year [1].
Introduction Second, although weights and volumes of thousands
of brains have been recorded over the years, the
For many people approaching old age, deteriorating number of very elderly brains examined has been
brain function is the aspect of growing old that they relatively low until recently [2,3]. Third, it is known
most fear. The brain seems to contain one’s essence that diseases that cause a reduction in brain weight
and the thought of losing that is hard to contemplate. and volume, particularly Alzheimer’s disease (AD),
But the changes that occur in the brain as it ages we increase in prevalence exponentially with age [4] and
now recognize to be much less tied to chronological many studies of brain weight and volume in old
time than Holmes suggested. Some peoples’ brains age have not taken steps to exclude brains with the
age much better than others. In this review I consider pathology of this disease from their studies. Fourth, it
what is known about why this might be, and how is only recently, with the advent of non-invasive brain
more of us can improve the chances of preserving imaging, that longitudinal measures on the same brains
our brains as they age. I first consider the evidence over time have been possible and studies of this type
about what changes occur in human brains with ageing have followed changes over relatively short periods of
and then consider additional evidence about the likely time [5].
cellular and molecular underpinnings of these, much
of it derived from animal studies.
Macroscopic brain changes with age

Ageing of the human brain Most studies of large numbers of post mortem human
brains indicate that in adults between the ages of
Brain changes with ageing have been studied since approximately 20 and 60 years there is a small loss
the nineteenth century but there is still a surprising of brain weight of about 0.1%/year, but more rapid
degree of uncertainty about what constitutes these loss thereafter [1–3,6] suggested that weights remain
changes. There are several reasons why this is so. stable until about the age of 50 years if the so-
First, measures of weight and volume of the brain called ‘secular’ change in brain weights over time is
as it ages need to take account of changes in these taken into account, and then, as other work suggests,
measures in young brains that have occurred over progressively decrease.

Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
www.pathsoc.org.uk
182 MM Esiri

When non-invasive brain imaging was introduced neuron size. This has been found to decrease mod-
in the 1970s, brain volume was similarly found to estly with age, particularly in cerebral cortex [31–34].
decrease with age [7–9]. This decrease is relatively Neuron size is thought to reflect the extent of the den-
diffuse and uniform in cerebral white matter but shows dritic and axonal arborizations of the cell, as a more
some regional differences in grey matter, with frontal extensive cell requires more energy, more protein syn-
and parietal cortex more affected than temporal and thesis and so on, requiring a larger cell body to furnish
occipital cortex and with the striatum also affected these. It is therefore no surprise to find that studies of
[10–13]. Brain volume reductions increase from about synapses, located mainly, although not exclusively, on
0.1–0.2%/year at age 30–50 years to 0.3–0.5%/year dendritic spines in cerebral cortex, have shown over-
over the age of 70 years, in agreement with brain all decreases with age, although the branching patterns
weight studies. The ventricular system expands to of dendrites suggests that there may be compensatory
fill the space vacated by reduced brain volume. The increases in some dendrites to make up for loss of
leptomeninges tend to thicken slightly with age and others [35]. A 46% reduction in spine number and
the subarachnoid space enlarges. density has been reported in humans over 50 years
of age [36]. Shrinkage of the dendritic trees probably
Microscopic brain changes with age accounts for subtle changes in the parameters of cor-
tical mini-columns in association cortex with human
Views about the microscopic basis for the macro- ageing [37]. Mini-columns are arrays of cortical neu-
scopic brain changes that are observed with ageing rons arranged in columns perpendicular to the pial sur-
have changed over the years. The main controversy face. Mini-columns in normal elderly subjects become
has been over the presence and extent of neuron loss. narrower in auditory association cortex, a region that
Initial studies, commenced in the 1950s, examined retains some plasticity in adult life. In contrast, the
changes in neuron density in two-dimensional space. normally more narrowly spaced mini-columns of the
They concluded that substantial loss of neurons occurs less plastic primary auditory cortex show little change
with age, varying in the range 10–60%, depending on with age [37].
the study and on the neuronal population examined. Some of the other microscopic alterations that occur
However, some populations were recognized to show in the brain with ageing are:
no loss of neurons with age, eg cranial nerve nuclei
[14]. Cerebral cortex [15,16] and hippocampus [17] • An increase in the number of corpora amylacea
were thought to be particularly affected, but also cere- [38]; these are spherical laminated polyglycosan
bellar Purkinje cells [18]. More recent studies employ- bodies, prominent in periodic acid–Schiff (PAS)-
ing stereologically-based sampling [19–22] to derive stained sections. They have a preferential location
estimates of neuronal numbers in three-dimensional around blood vessels or close to pial or ependymal
space have arrived at more conservative conclusions, surfaces. Some are localizable ultrastructurally to
that neuron loss with ageing is either undetectable or astrocyte processes.
relatively mild [23,24]. Estimating the magnitude of • Increase in the amount of detectable iron [39]; iron
neuronal loss in humans is complicated by the fact reaches the brain by a selective uptake mechanism
that most elderly brains from subjects over the age of operating across the blood–brain barrier but little is
80 years are affected by the pathological changes of known of how it is released and why it accumulates
amyloid plaque and neurofibrillary tangle formation, with age. Iron is an essential component of many
the two hallmarks, when present in substantial num- enzymes in the brain but high concentrations of
bers, of AD. They are also affected by cerebrovascular reactive iron can facilitate oxidative stress.
disease [25]. There is, of course, a debate to be had • Increase in the amount of advanced glycation end
over whether these features represent part of ‘normal products [40].
ageing’ or ‘disease’, but for the purposes of this review • Increase in the number and size of astrocytes and
I have considered that they represent ‘disease’. Taking microglia. Microglia are generally thought of as
this view, these features need to be checked for before pathogenic when activated but they may also be
allocating the brain of an elderly subject to the ‘nor- neuroprotective under some circumstances [41].
mal’ category. Relatively few brains of normal elderly • An increase in the lipofuscin content of neurons
subjects checked in this way have had stereologically- [42]. The amount of lipofuscin that accumulates
based estimates of neuronal population numbers. In varies considerably from one type of neuron to
non-human primates, significant loss of neurons in the another, but it tends to accumulate more in large
hippocampus and most of the neocortex is not a fea- cortical and thalamic neurons, inferior olive neurons
ture of ageing. However, a 30% reduction in neurons and motor neurons than in others. Lipofuscin con-
in seen in dorsolateral prefrontal cortex, a change that sists of material derived from lysosomal degrada-
correlates with impairment on a working memory task tion. In neurons of the substantia nigra, the melanin
which is dependent on the function of this region of pigment that accumulates does so as a by-product
the brain [26–30]. of synthesis by the cells of their specific neurotrans-
If studies of neuron numbers have yielded contro- mitter, dopamine. There is barely any detectable
versial findings, there has been more agreement about melanin pigment in these cells at birth but it is

J Pathol 2007; 211: 181–187 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Ageing and the brain 183

readily visible by the time children reach the age radicals, which have the capacity to damage proteins,
of 8 years. Neuromelanin and lipofuscin have some nucleic acids and lipids. Highly reactive superox-
features in common and it is interesting that neu- ide ion radicals are produced that lead to generation
romelanin starts to accumulate long before old age of hydrogen peroxide, hydroxyl radicals, which are
is reached. Lipofuscin accumulation, as a product formed by the reaction of hydrogen peroxide with iron
of lysosomal degradation, is related to autophagy. and copper ions, and peroxynitrite, formed by the reac-
Recently it has been shown that mice engineered tion of superoxide with nitric acid. These molecules
to lack genes enabling autophagy to occur develop oxidize proteins, lipids and DNA, yielding a large
neurodegeneration, illustrating the importance of number of compounds, which can be estimated to give
autophagy in the housekeeping functions in neurons a measure of how much oxidative damage a brain has
[43]. been exposed to. These compounds include oxidized
proteins [47] 4-hydroxynonenal, which results from
It is possibly no coincidence that the two com- peroxidation of ω-6-conjugated fatty acids [48], and
monest neurodegenerative diseases of ageing, AD and 8-hydroxy-2 -deoxyguanosine, a measure of oxida-
Parkinson’s disease (PD), particularly affect cells that tive damage to DNA. Mitochondria themselves suffer
are selectively vulnerable to ageing itself — cortical oxidative damage, which may render them less effi-
and hippocampal pyramidal cells in the case of AD cient at energy generation and liable to generate more
and pigmented brain stem neurons in PD. Common, superoxide ions.
almost universal, functional changes occur in memory These alterations interfere with many aspects of
and motor performance with age. These are functions normal cell metabolism and function. DNA damage
that depend on networks involving these neurons that leads to reduced gene expression, or to the generation
are affected in AD and PD. We need to understand of abnormal proteins that have to be eliminated by
the brain changes that occur with ageing if we are to processes such as proteosomal degradation. A recent
understand the common, sporadic, forms of these dis- study of RNA obtained from human frontal cortex
eases. Although early-onset forms of both diseases are of individuals in the age range 26–106 years found
recognized that are caused by genetic mutations, most that many genes — about 4% of the 11 000 genes
cases of these diseases (ca. 95%) occur in old age and studied — showed reduced expression after the age
the factors that cause late-onset disease are likely to be of 40 years. These genes were involved in synap-
much more heterogeneous, involving both genetic and tic function and plasticity, vesicular transport, mito-
environmental risk factors. To understand the interplay chondrial function and calcium homeostasis. In con-
between these factors, we have to improve understand- trast, some gene expression was increased, includ-
ing of brain ageing. ing genes involved in protein folding and stress
responses, antioxidant defence, metal ion homeosta-
Cellular and molecular changes sis and the inflammatory response [49]. This increased
gene expression is readily interpretable as a response
Cellular and molecular changes in brain ageing have to the oxidative and other stresses referred to above.
been well reviewed recently [44–46]. Here we con- Antioxidant enzymes and growth factors, which might
sider some of the more important factors. be expected to combat the effects of oxidative damage,
have altered effectiveness through alterations in their
High energy demands of neurons render them signalling pathways or lower production with ageing
vulnerable to ageing [45]. Antioxidant defences in the brain are, at best,
A key factor that plays a role in brain ageing is relatively low. Conversely, the brain is rich in pro-
the great demand that neurons have for oxidative oxidant iron and in unsaturated fatty acids that are
metabolism in the generation of energy. The energy particularly vulnerable to peroxidation [50].
needs of neurons throughout a lifetime are exception- Another change to which proteins, lipids and nucleic
ally high, driven by: acids are vulnerable with ageing is the production of
advanced glycation end-products (AGEs), which are
• The enormous size of some neurons, necessitating adducts or cross-links that form non-enzymatically in
maintenance of a very large surface membrane and a reaction between reducing sugars, including glucose,
an energy-hungry system of transport of molecules and the amino groups of proteins and other compounds
and organelles to enable distant parts of the cell to through the Maillard reaction, a process that can occur
be supplied with nutrients and subcellular machin- extra- or intracellularly. The effects of AGEs are
ery. diverse; some are receptor-dependent and some are
• The electrical activity involved in impulse transmis- mediated via other endogenous binding sites, including
sion, which requires ion gradients to be maintained lactoferrin and the macrophage scavenger receptors.
over long stretches of axonal membrane, at great Engagement of AGEs with the receptor (RAGE) can
energetic expense. result in oxidant stress, and by directly affecting
proteins AGEs can alter the shape and interfere with
Mitochondrial activity needed for oxidative meta- the function of proteins. AGEs have been reported to
bolism involves inevitable generation of oxygen free accumulate with age in many tissues, including the

J Pathol 2007; 211: 181–187 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
184 MM Esiri

brain [51,52]. There are enzymes (glyoxalases) that in synaptic plasticity as well as in ageing and neu-
provide protection against glycation. These have been rodegeneration [60]. Some of these do appear to be
found to be up-regulated in the brain in a transgenic related to apoptosis [57]. Whether altered Ca2+ home-
model of AD [53,54]. ostasis plays a role in these forms of degeneration is
not clear, but neurotrophic factors such as bFGF can
exert a direct protective effect on synapses [61,62].
Calcium dysregulation The ubiquitin–proteosome system appears to play an
important role in dendritic, synaptic and axonal degen-
Decreasing mitochondrial efficiency with ageing, and eration [63,64].
its knock-on effects in increasing oxidative stress in
neurons, is intimately coupled to changes in calcium
homeostasis. Aged brain mitochondria are persistently Genes that influence brain ageing
depolarized [55], which is likely to result in an alter- There is a vast array of genes that may potentially
ation in the calcium gradient across mitochondrial influence brain ageing. In principle, any of the genes
membranes. These membranes can normally take up involved in the control of all the factors discussed
calcium from the cytosol when calcium ions accu- above could be involved. In addition, hormonal factors
mulate there, secondary to neuronal membrane ionic may be important [65]. Much more attention has been
fluxes involved in impulse transmission. Impaired abil- given to discovering genes that are important in age-
ity of the mitochondria to store calcium exposes the related neurodegenerative diseases, such as AD and
neuronal cytosol to higher calcium levels, particularly PD, but some consideration has also been given to
after excitatory stimulation. genes that are important in brain ageing [66]. We have
Excitation opens voltage-dependent calcium and N- already seen that many genes influencing the processes
methyl-D-aspartate (NMDA) glutamate receptor chan- described above are either up- or down-regulated in
nels and leads to a transient rise in intracellular cal- human frontal cortex after the age of 40 years [49].
cium levels, even in healthy young neurons. This is The ability to up-regulate genes that repair damage
increased in normal ageing [46]. Ca2+ imaging stud- due to oxidation, glycosylation and so on probably
ies have shown that the resting Ca2+ concentration in has a major influence. This would be predicted by
CA1 hippocampal neurons does not change with age- the attractive ‘disposable soma’ view of ageing as the
ing, but that these cells show a greater rise in Ca2+ in result of a trade-off between somatic maintenance and
response to stimulation [56]. Elevated levels of intra- repair and other functions, particularly reproduction,
cellular Ca2+ are liable to activate calcium-activated in the allocation of an animal’s metabolic resources
proteases (caspases), with potentially damaging effects [67,68].
on cells. At their worst, these effects can include cell Two genes with variants that influence successful
death by apoptosis, but it is possible that they can have brain ageing are the apolipoprotein E (ApoE ) gene
more localized effects in brain ageing, for example, and the prion protein gene, PRNP [69,70]. The ApoE
causing apoptosis limited to dendrites [57,58]. gene has three isoforms, ε2 , ε3 and ε4 . ε3 is the
A likely scenario is that, in the ageing brain, dis- commonest allele, ε4 the next most common and ε2
rupted Ca2+ homeostasis remains compatible with nor- is uncommon. ApoE ε2 is protective against AD and
mal impulse transmission and neurotransmitter release, ε4 is a risk factor for late-onset AD [71]. ApoE ε2
but that it renders the brain very liable to damage if is over-represented in centenarians, suggesting that
there is an imposed stress, such as mild hypoxia. it influences successful brain ageing. It is uncertain
Mild degrees of cerebrovascular disease were found exactly how ApoE exerts this influence. In normal
to be present in over 75% of elderly unselected UK individuals ApoE ε4 is associated with lower cognitive
subjects in the MRC Cognitive Function and Age- performance at 11 years and even more at 80 years of
ing Study [25], and many elderly subjects also suffer age [72].
from heart disease, which is likely to give rise to tran- Prion protein is the protein whose tertiary structure
sient episodes of hypoxia. One of the consequences of is altered in Creutzfeldt–Jakob disease (CJD). The
hypoxia is excess glutamate release, which operates normal function of the protein is not entirely clear
on postsynaptic membranes to open NMDA recep- but it is thought that it may play a role in protecting
tor channels, leading to calcium entry and the risk neurons from the effects of cellular stress. It may play
of calcium overload, with the consequences this may an antioxidant role by influencing uptake of copper
have for the cell. Whether apoptosis is the mode of [73]. The PRNP codon 129 influences susceptibility
death of neurons that are lost with ageing remains to CJD and also to AD [74–77]. Individuals who
uncertain. Non-apoptotic modes of death of neurons or were homozygous for the methionine allele at codon
sub-regions of neurons are also recognized, and may 129 were found to perform cognitively better at age
be important in dendritic spine loss and dying back 79 years than those who were heterozygous, in a study
of axons, for example. Anti-apoptotic interventions do that measured cognitive ability at age 11 years and
not prevent axonal degeneration [59]. Dendrites and again at 79 years in a Scottish cohort [70]. Other genes
synapses also seem to contain compartmentalized self- that were found to exert a mild effect on cognitive
degenerative programmes, which may be important ability at 79 years in this cohort were the genes for

J Pathol 2007; 211: 181–187 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Ageing and the brain 185

nicastrin, Klotho and catechol-O-methyl transferase The key questions with regard to ageing are whether
[78–80]. Other genes thought to play a role in brain the ageing process is in fact clock-driven and what
evolutionary factors might have shaped the design of the
ageing are those involved in insulin signalling [81], clock . . .. Our answer to the question ‘Is it a clock?’ is a
DNA and protein methylation and acetylation [82], definite ‘No’ [67].
DNA repair [83] and lipid metabolism [84].

References
Environmental factors and brain ageing
1. Miller AK, Corsellis JA. Evidence for a secular increase in
A major environmental factor in ageing generally and human brain weight during the past century. Ann Hum Biol
brain ageing in particular is diet. It has been known 1977;4:253–257.
for many decades that restricting daily calorie intake 2. Dekaban AS. Changes in brain weights during the span of human
life: relation of brain weights to body heights and body weights.
in rodents and some other animals increases longevity
Ann Neurol 1978;4:345–356.
substantially [85,86]. It is speculated that this may 3. Ho KC, Roessmann U, Straumfjord JV, Monroe G. Analysis of
be because in times of famine reproductive activity brain weight. I. Adult brain weight in relation to sex, race, and
is reduced, since any offspring would be unlikely to age. Arch Pathol Lab Med 1980;104:635–639.
survive. Accordingly, energy is switched to enhanc- 4. Skoog I, Nilsson L, Palmertz B, Andreasson LA, Svanborg A.
A population-based study of dementia in 85 year-olds. N Engl J
ing maintenance of the soma, which improves the
Med 1993;328:153–158.
repair of cellular damage and reduces the produc- 5. Fox NC, Schott JM. Imaging cerebral atrophy: normal ageing to
tion of reactive oxygen species. Calorie restriction also Alzheimer’s disease. Lancet 2004;363:392–394.
enhances brain-derived neurotrophic factor production 6. Blinkov S, Glezer I. The Human Brain in Figures and Tables: A
and neurogenesis [45]. Conversely, it is likely that Quantitative Handbook. Plenum: New York, 1968.
excess calorie intake has a deleterious effect on brain 7. Coffey CE, Wilkinson WE, Parashos IA, Soady SA, Sullivan RJ,
Patterson LJ, et al. Quantitative cerebral anatomy of the aging
metabolism. Elevated cholesterol and blood pressure, human brain: a cross-sectional study using magnetic resonance
both linked to obesity and excess calorie intake, and imaging. Neurology 1992;42:527–536.
elevated blood homocysteine are risk factors for the 8. Jernigan TL, Press GA, Hesselink JR. Methods for measuring
development both of AD and cerebrovascular disease brain morphologic features on magnetic resonance images.
[87–89]. Other beneficial influences on brain function Validation and normal aging. Arch Neurol 1990;47:27–32.
9. Pfefferbaum A, Mathalon DH, Sullivan EV, Rawles JM,
in old age are physical exercise [90], extended years Zipursky RB, Lim KO. A quantitative magnetic resonance
of education [91,92], cognitive stimulation [93] and imaging study of changes in brain morphology from infancy to
a high intake of polyunsaturated fatty acids [94] and late adulthood. Arch Neurol 1994;51:874–887.
B vitamins, particularly vitamins B6, B12 and folate 10. Raz N. The ageing brain: structural changes and their implications
[95] and statins [96]. The influence of toxins in caus- for cognitive ageing. In New Frontiers in Cognitive Ageing,
Dixon R, Backman L, Nilsson L (eds). Oxford University Press:
ing brain damage in normal brain ageing is not clear Oxford, 2004; 115–134.
but vigilance is called for, as some environmental tox- 11. Resnick SM, Pham DL, Kraut MA, Zonderman AB, Davatzikos C.
ins have been implicated as risk factors in age-related Longitudinal magnetic resonance imaging studies of older adults:
disease, for example, the pesticide rotenone and the a shrinking brain. J Neurosci 2003;23:3295–3301.
herbicide paraquat in PD [97] and a product of cycad 12. Scahill RI, Frost C, Jenkins R, Whitwell JL, Rossor MN, Fox NC.
A longitudinal study of brain volume changes in normal aging
seeds, β-methylamino-L-alanine (BMAA), in Guam using serial registered magnetic resonance imaging. Arch Neurol
disease [98]. 2003;60:989–994.
13. Trollor JN, Valenzuela MJ. Brain ageing in the new millennium.
Aust NZ J Psychiatr 2001;35:788–805.
14. Esiri M. Dementia and normal aging: neuropathology. In Dementia
Conclusions and Normal Aging, Huppert F, Brayne C, O’Connor D (eds).
Cambridge University Press: Cambridge, 1994; 385–436.
The brain undergoes subtle but definite decline in 15. Brody H. Organization of the cerebral cortex. III. A study of aging
in the human cerebral cortex. J Comp Neurol 1955;102:511–516.
structure and function with age. The extent of these
16. Henderson G, Tomlinson BE, Gibson PH. Cell counts in human
changes is variable and subject to numerous influ- cerebral cortex in normal adults throughout life using an image
ences, both genetic and environmental. Progressive, analysing computer. J Neurol Sci 1980;46:113–136.
clinically significant, decline over a lifetime may 17. Ball MJ. Neuronal loss, neurofibrillary tangles and granulovacuo-
not be inevitable and may even, in future, prove lar degeneration in the hippocampus with ageing and dementia.
A quantitative study. Acta Neuropathol (Berl) 1977;37:111–118.
to be reversible, given relatively recent understand-
18. Hall T, Miller K, Corsellis JA. Variations in the human Purkinje
ing that: (a) neurogenesis can persist into adulthood; cell population according to age and sex. Neuropathol Appl
(b) protective neurotrophic factor production can be Neurobiol 1975;1:267–292.
enhanced under certain conditions (exercise, diet); and 19. Benes FM, Lange N. Two-dimensional versus three-dimensional
(c) oxidative damage to the brain can be influenced cell counting: a practical perspective. Trends Neurosci
by diet and other lifestyle factors. To return to the 2001;24:11–17.
20. Howard C, Reed M. Unbiased Stereology: Three-dimensional
quotation at the start of this review, I conclude with Measurement in Microscopy. Bios Scientific: London, 1998.
another quotation, admittedly about ageing in general, 21. West MJ. New stereological methods for counting neurons.
but equally pertinent to brain ageing: Neurobiol Aging 1993;14:275–285.

J Pathol 2007; 211: 181–187 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
186 MM Esiri

22. West MJ. Stereological methods for estimating the total number of 46. Toescu EC. Normal brain ageing: models and mechanisms. Phil
neurons and synapses: issues of precision and bias. Trends Neurosci Trans R Soc Lond B Biol Sci 2005;360:2347–2354.
1999;22:51–61. 47. Stadtman ER. Protein oxidation and aging. Science
23. Hof PR, Morrison JH. The aging brain: morphomolecular 1992;257:1220–1224.
senescence of cortical circuits. Trends Neurosci 2004;27:607–613. 48. Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry
24. West MJ, Coleman PD, Flood DG, Troncoso JC. Differences in of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free
the pattern of hippocampal neuronal loss in normal ageing and Radic Biol Med 1991;11:81–128.
Alzheimer’s disease. Lancet 1994;344:769–772. 49. Lu T, Pan Y, Kao SY, Li C, Kohane I, Chan J, et al. Gene
25. Neuropathology Group of the Medical Research Council Cognitive regulation and DNA damage in the ageing human brain. Nature
Function and Ageing Study (MRC CFAS). Pathological correlates 2004;429:883–891.
of late-onset dementia in a multicentre, community-based 50. Floyd RA, Hensley K. Oxidative stress in brain aging. Implica-
population in England and Wales. Lancet 2001;357:169–175. tions for therapeutics of neurodegenerative diseases. Neurobiol
26. Gazzaley AH, Thakker MM, Hof PR, Morrison JH. Preserved Aging 2002;23:795–807.
number of entorhinal cortex layer II neurons in aged macaque 51. Kimura T, Takamatsu J, Ikeda K, Kondo A, Miyakawa T, Hori-
monkeys. Neurobiol Aging 1997;18:549–553. uchi S. Accumulation of advanced glycation end products of the
27. Keuker JI, Luiten PG, Fuchs E. Preservation of hippocampal Maillard reaction with age in human hippocampal neurons. Neu-
neuron numbers in aged rhesus monkeys. Neurobiol Aging rosci Lett 1996;208:53–56.
2003;24:157–165. 52. Kimura T, Takamatsu J, Miyata T, Miyakawa T, Horiuchi S.
28. Merrill DA, Roberts JA, Tuszynski MH. Conservation of neuron Localization of identified advanced glycation end-product
number and size in entorhinal cortex layers II, III, and V/VI of structures, N-ε (carboxymethyl)lysine and pentosidine, in age-
aged primates. J Comp Neurol 2000;422:396–401. related inclusions in human brains. Pathol Int 1998;48:575–579.
29. Peters A, Leahu D, Moss MB, McNally KJ. The effects of aging 53. Chen F, Wollmer MA, Hoerndli F, Munch G, Kuhla B, Rogaev EI,
on area 46 of the frontal cortex of the rhesus monkey. Cereb Cortex et al. Role for glyoxalase I in Alzheimer’s disease. Proc Natl Acad
1994;4:621–635. Sci USA 2004;101:7687–7692.
30. Smith DE, Rapp PR, McKay HM, Roberts JA, Tuszynski MH. 54. Thornalley PJ. Glyoxalase I — structure, function and a critical
Memory impairment in aged primates is associated with focal role in the enzymatic defence against glycation. Biochem Soc Trans
death of cortical neurons and atrophy of subcortical neurons. 2003;31:1343–1348.
J Neurosci 2004;24:4373–4381. 55. Xiong J, Verkhratsky A, Toescu EC. Changes in mitochondrial
31. Anderson JM, Hubbard BM, Coghill GR, Slidders W. The effect status associated with altered Ca2+ homeostasis in aged cerebellar
of advanced old age on the neurone content of the cerebral cortex.
granule neurons in brain slices. J Neurosci 2002;22:10761–10771.
Observations with an automatic image analyser point counting
56. Thibault O, Hadley R, Landfield PW. Elevated postsynaptic
method. J Neurol Sci 1983;58:235–246.
[Ca2+ ]i and L-type calcium channel activity in aged hippocampal
32. Haug H. Are neurons of the human cerebral cortex really lost
neurons: relationship to impaired synaptic plasticity. J Neurosci
during ageing? A morphometric examination. In Senile Dementia
2001;21:9744–9756.
of the Alzheimer Type, Traber J, Gispen W (eds). Springer: Berlin,
57. Cowan CM, Thai J, Krajewski S, Reed JC, Nicholson DW,
1985; 150–163.
Kaufmann SH, et al. Caspases 3 and 9 send a pro-apoptotic signal
33. Meier-Ruge W, Iwangoff P, Reichlmeier K, Sandoz P. Neuro-
from synapse to cell body in olfactory receptor neurons. J Neurosci
chemical findings in the aging brain. Adv Biochem Psychophar-
2001;21:7099–7109.
macol 1980;23:323–338.
58. Gilman CP, Mattson MP. Do apoptotic mechanisms regulate
34. Terry RD, DeTeresa R, Hansen LA. Neocortical cell counts in
synaptic plasticity and growth-cone motility? Neuromol Med
normal human adult aging. Ann Neurol 1987;21:530–539.
2002;2:197–214.
35. Buell SJ, Coleman PD. Quantitative evidence for selective
dendritic growth in normal human aging but not in senile dementia. 59. Sagot Y, Dubois-Dauphin M, Tan SA, de Bilbao F, Aebischer P,
Brain Res 1981;214:23–41. Martinou JC, et al. Bcl-2 overexpression prevents motoneuron cell
36. Jacobs B, Driscoll L, Schall M. Life-span dendritic and spine body loss but not axonal degeneration in a mouse model of a
changes in areas 10 and 18 of human cortex: a quantitative Golgi neurodegenerative disease. J Neurosci 1995;15:7727–7733.
study. J Comp Neurol 1997;386:661–680. 60. Gillingwater TH, Thomson D, Mack TG, Soffin EM, Mattison RJ,
37. Chance SA, Casanova MF, Switala AE, Crow TJ, Esiri MM. Coleman MP, et al. Age-dependent synapse withdrawal at
Minicolumn thinning in temporal lobe association cortex but not axotomized neuromuscular junctions in Wld(s) mutant and
primary auditory cortex in normal human ageing. Acta Neuropathol Ube4b/Nmnat transgenic mice. J Physiol 2002;543:739–755.
(Berl) 2006;111:459–464. 61. Guo ZH, Mattson MP. Neurotrophic factors protect cortical
38. Mrak RE, Griffin ST, Graham DI. Aging-associated changes in synaptic terminals against amyloid and oxidative stress-induced
human brain. J Neuropathol Exp Neurol 1997;56:1269–1275. impairment of glucose transport, glutamate transport and
39. Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR. Iron, mitochondrial function. Cereb Cortex 2000;10:50–57.
brain ageing and neurodegenerative disorders. Nat Rev Neurosci 62. Mattson MP, Guo ZH, Geiger JD. Secreted form of amyloid
2004;5:863–873. precursor protein enhances basal glucose and glutamate transport
40. Schmitt HP. ε-Glycation, APP and Aβ in ageing and Alzheimer and protects against oxidative impairment of glucose and glutamate
disease: a hypothesis. Med Hypotheses 2006;66:898–906. transport in synaptosomes by a cyclic GMP-mediated mechanism.
41. Esiri MM. The interplay between inflammation and neurodegen- J Neurochem 1999;73:532–537.
eration in CNS disease. J Neuroimmunol 2006;(in press). 63. Coleman MP, Perry VH. Axon pathology in neurological disease:
42. Mann DM, Yates PO, Stamp JE. The relationship between a neglected therapeutic target. Trends Neurosci 2002;25:532–537.
lipofuscin pigment and ageing in the human nervous system. 64. Gillingwater TH, Ribchester RR. The relationship of neuromus-
J Neurol Sci 1978;37:83–93. cular synapse elimination to synaptic degeneration and pathol-
43. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, ogy: insights from WldS and other mutant mice. J Neurocytol
Suzuki-Migishima R, et al. Suppression of basal autophagy in 2003;32:863–881.
neural cells causes neurodegenerative disease in mice. Nature 65. Peters R. Ageing and the brain. Postgrad Med J 2006;82:84–88.
2006;441:885–889. 66. Goyns MH. Genes, telomeres and mammalian ageing. Mech
44. Burke SN, Barnes CA. Neural plasticity in the ageing brain. Nat Ageing Dev 2002;123:791–799.
Rev Neurosci 2006;7:30–40. 67. Kirkwood TB. Time of our lives. What controls the length of life?
45. Mattson MP, Maudsley S, Martin B. BDNF and 5-HT. a dynamic EMBO Rep 2005;6:S4–8.
duo in age-related neuronal plasticity and neurodegenerative 68. Kirkwood TB. Understanding the odd science of aging. Cell
disorders. Trends Neurosci 2004;27:589–594. 2005;120:437–447.

J Pathol 2007; 211: 181–187 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Ageing and the brain 187

69. Deary IJ, Wright AF, Harris SE, Whalley LJ, Starr JM. Searching 84. Yehuda S, Rabinovitz S, Carasso RL, Mostofsky DI. The role
for genetic influences on normal cognitive ageing. Trends Cogn of polyunsaturated fatty acids in restoring the aging neuronal
Sci 2004;8:178–184. membrane. Neurobiol Aging 2002;23:843–853.
70. Kachiwala SJ, Harris SE, Wright AF, Hayward C, Starr JM, 85. Merry BJ. Molecular mechanisms linking calorie restriction and
Whalley LJ, et al. Genetic influences on oxidative stress and longevity. Int J Biochem Cell Biol 2002;34:1340–1354.
their association with normal cognitive ageing. Neurosci Lett 86. Martin B, Mattson MP, Maudsley S. Caloric restriction and
2005;386:116–120. intermittent tasting; two potential diets for successful brain aging.
71. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Ageing Research Reviews 2006;5:332–353.
Gaskell PC, Small GW, et al. Gene dose of apolipoprotein E type 87. Budge M, Johnston C, Hogervorst E, de Jager C, Milwain E,
4 allele and the risk of Alzheimer’s disease in late-onset families. Iversen SD, et al. Plasma total homocysteine and cognitive
Science 1993;261:921–923. performance in a volunteer elderly population. Ann NY Acad Sci
72. Deary IJ, Whiteman MC, Pattie A, Starr JM, Hayward C, 2000;903:407–410.
Wright AF, et al. Apolipoprotein e gene variability and cognitive 88. Cutler RG, Kelly J, Storie K, Pedersen WA, Tammara A, Hatan-
functions at age 79: a follow-up of the Scottish mental survey of paa K, et al. Involvement of oxidative stress-induced abnor-
1932. Psychol Aging 2004;19:367–371. malities in ceramide and cholesterol metabolism in brain
73. Brown DR. Prion protein expression aids cellular uptake aging and Alzheimer’s disease. Proc Natl Acad Sci USA
and veratridine-induced release of copper. J Neurosci Res 2004;101:2070–2075.
1999;58:717–725. 89. Mayeux R. Epidemiology of neurodegeneration. Annu Rev
74. Dermaut B, Croes EA, Rademakers R, Van den Broeck M, Neurosci 2003;26:81–104.
Cruts M, Hofman A, et al. PRNP Val129 homozygosity increases 90. Cotman CW, Berchtold NC. Exercise: a behavioral intervention
risk for early-onset Alzheimer’s disease. Ann Neurol to enhance brain health and plasticity. Trends Neurosci
2003;53:409–412. 2002;25:295–301.
75. Golanska E, Hulas-Bigoszewska K, Rutkiewicz E, Styczynska M, 91. Shenkin SD, Bastin ME, MacGillivray TJ, Deary IJ, Starr JM,
Peplonska B, Barcikowska M, et al. Polymorphisms within the Wardlaw JM. Childhood and current cognitive function in healthy
prion (PrP ) and prion-like protein (Doppel ) genes in AD. 80 year-olds: a DT-MRI study. NeuroReport 2003;14:345–349.
Neurology 2004;62:313–315. 92. Staff RT, Murray AD, Deary IJ, Whalley LJ. What provides
76. Knight RS, Will RG. Prion diseases. J Neurol Neurosurg Psychiat cerebral reserve? Brain 2004;127:1191–1199.
2004;75(Suppl 1):i36–42. 93. Lazarov O, Robinson J, Tang YP, Hairston IS, Korade-Mirnics Z,
77. Riemenschneider M, Klopp N, Xiang W, Wagenpfeil S, Lee VM, et al. Environmental enrichment reduces Aβ levels and
Vollmert C, Muller U, et al. Prion protein codon 129 polymor- amyloid deposition in transgenic mice. Cell 2005;120:701–713.
phism and risk of Alzheimer disease. Neurology 2004;63:364–366. 94. Kyle D. The role of docosahexaenoic acid in the evolution and
78. Deary IJ, Hamilton G, Hayward C, Whalley LJ, Powell J, Starr JM, function of the human brain. In Brain Lipids and Disorders
et al. Nicastrin gene polymorphisms, cognitive ability level and in Biological Psychiatry, Skinner E (ed.). Elsevier Science:
cognitive ageing. Neurosci Lett 2005;373:110–114. Amsterdam, 2002; 1–22.
79. Deary IJ, Harris SE, Fox HC, Hayward C, Wright AF, Starr JM, 95. Elias MF, Robbins MA, Budge MM, Elias PK, Brennan SL,
et al. KLOTHO genotype and cognitive ability in childhood and Johnston C, et al. Homocysteine, folate, and vitamins B6 and
old age in the same individuals. Neurosci Lett 2005;378:22–27. B12 blood levels in relation to cognitive performance: the Maine-
80. Harris SE, Wright AF, Hayward C, Starr JM, Whalley LJ, Syracuse study. Psychosom Med 2006;68:547–554.
Deary IJ. The functional COMT polymorphism, Val 158 Met, 96. DeKosky ST. Statin therapy in the treatment of Alzheimer disease:
is associated with logical memory and the personality trait what is the rationale? Am J Med 2005;118(Suppl 12A):48–53.
intellect/imagination in a cohort of healthy 79 year olds. Neurosci 97. Di Monte DA. The environment and Parkinson’s disease: is the
Lett 2005;385:1–6. nigrostriatal system preferentially targeted by neurotoxins? Lancet
81. Wolkow CA. Life span: getting the signal from the nervous system. Neurol 2003;2:531–538.
Trends Neurosci 2002;25:212–216. 98. Kisby GE, Ellison M, Spencer PS. Content of the neurotoxins
82. Mattson MP. Methylation and acetylation in nervous system cycasin (methylazoxymethanol β-D-glucoside) and BMAA (β-
development and neurodegenerative disorders. Ageing Res Rev N-methylamino-L-alanine) in cycad flour prepared by Guam
2003;2:329–342. Chamorros. Neurology 1992;42:1336–1340.
83. Kyng KJ, Bohr VA. Gene expression and DNA repair in progeroid
syndromes and human aging. Ageing Res Rev 2005;4:579–602.

J Pathol 2007; 211: 181–187 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Você também pode gostar