You are on page 1of 14

J Neuropathol Exp Neurol Vol. 68, No.

1
Copyright Ó 2008 by the American Association of Neuropathologists, Inc. January 2009
pp. 1Y14

REVIEW ARTICLE

Neuropathology and Cognitive Impairment in Alzheimer Disease:


A Complex but Coherent Relationship
Peter T. Nelson, MD, PhD, Heiko Braak, MD, and William R. Markesbery, MD

there have been many controversies. This review of the lit-


Abstract erature on CP studies in AD is intended to describe the prog-
Amyloid plaques and neurofibrillary tangles (NFTs) are the ress and complexities for a broad scientific audience.
pathological hallmarks of Alzheimer disease (AD). There is con- We interpret CP studies to provide robust support for
troversy regarding the use of current diagnostic criteria for AD and the importance of both amyloid plaques and neurofibrillary
whether amyloid plaques and NFTs contribute to cognitive impair- tangles (NFTs) in the clinical manifestations of AD (Fig. 1;
ment. Because AD is specific to humans, rigorous and compre- Table 1). We review the large body of pertinent literature

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


hensive clinicopathologic studies are necessary to test and refine and describe the medical, technical, and anatomical aspects
hypotheses of AD diagnosis and pathogenesis. Neither the clinical relevant to interpreting these data. Because the pathological
nor the pathological aspects of AD evolve in a linear manner, but features of AD and many other aspects of the aged brain are
the predictable sequence of AD pathology allows for stage-based distinct in Homo sapiens, we focus primarily on studies on
correlations with cognitive deterioration. We discuss subsets of humans.
patients with clinical dementia who lack amyloid plaques and NFTs
and, conversely, whether individuals without antemortem cognitive
impairment can harbor severe AD-type pathological findings at au- CLINICOPATHOLOGIC CORRELATION IN THE
topsy. There are many medical, technical, and anatomical challenges CONTEXT OF AD: THE GOALS AND THE
to clinicopathologic studies in AD. For example, at least two thirds POTENTIAL OBSTACLES
of persons older than 80 years have non-AD brain diseases that can The goal of CP studies is to understand the clinical and
effect on cognitive function. We argue that existing data strongly biologic importance of identified pathological features. In
support the hypothesis that both amyloid plaques and NFTs principle, pathological severity should correlate meaningfully
contribute to cognitive impairment. with the extent of clinical disease, that is, cognitive impair-
Key Words: Acetylcholine, Aging, Cognition, Lewy, Mini-Mental ment in AD. A desirable correlation is depicted in Figure 2A,
State Examination, Stroke, Tau. in which the ideal correlation requires a linear association
between 2 discrete entities: impairment of health and severity
of pathology. Such correlations are, however, seldom attained
in practice because of functional reserve capacity, biologic
INTRODUCTION variation between individuals in both protective and patho-
Clinicopathologic (CP) studies are essential for deter- genetic pathways, and incomplete understanding of disease
mining the pathobiological importance of amyloid plaques mechanisms.
and neurofibrillary pathology in Alzheimer disease (AD). Furthermore, CP studies require assumptions with re-
Research on CP correlation in AD has been active for sev- spect to the pathological substrates. It has been suggested that
eral decades, and meaningful data have accumulated from amyloid plaques and NFTs may not be pathogenetic in AD,
many international centers. These investigations have been and that these abnormalities could instead be a neuroprotec-
aided by a detailed understanding of both clinical and path- tive response to other disease stimuli such as oxidative stress
ological progression of AD. Along with advances, however, and/or inflammation (1). Neither oxidative nor inflammatory
From the Department of Pathology and Division of Neuropathology,
brain insults have, however, been shown to induce NFTs in
University of Kentucky Medical Center, Sanders-Brown Center on any wild-type animal model. Are NFTs Bneuroprotective[
Aging (PTN, WRM); and Alzheimer’s Disease Center, University of exclusively in humans? Interpreting AD-related changes as
Kentucky, Lexington, Kentucky (PTN, WRM); and Institute for Clinical adaptive becomes even more difficult as one seeks explana-
Neuroanatomy, Goethe University Frankfort, Frankfurt am Main, tions for the peculiar amyloid plaque pattern and the selec-
Germany (HB).
Send correspondence and reprint requests to: Peter T. Nelson, MD, PhD, tive vulnerability of only a few types of nerve cells in AD. In
Department of Pathology, Division of Neuropathology, Room 311, principle, all nerve cells and nonneuronal cells can be ex-
Sanders-Brown Center on Aging, University of Kentucky, 800 S. pected to react similarly to oxidative stress and inflammation,
Limestone, Lexington, KY 40536-0230; E-mail: pnels2@email.uky.edu although not necessarily always to the same extent. We in-
This study was supported by Grant No. 5-P30-AG028383 and Grant No.
K08 NS050110 from the National Institutes of Health, Bethesda,
terpret the genetic evidence (discussed later) and qualitative
Maryland, by a grant from the Healy Family Foundation, and funding pathological assessments of AD brains, particularly the neu-
from the Deutsche Forschungsgemeinschaft. rofibrillary pathology that appears to warp and impinge upon

J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009 1

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Nelson et al J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009

Additional questions have been raised regarding the


biologic relevance of amyloid plaques and NFTs (1). Eval-
uating these arguments requires consideration of the specificity
of plaques and NFTs in AD, the importance of concomitant
disease processes, and the challenges in performing cognitive
assessments.

Specificity of AD Lesions: Amyloid Plaques and


NFTs are Seen Outside of AD
It has been suggested that neurofibrillary pathology and
amyloid plaques are not specific to AD. If this is true, does
nonspecificity argue for or against the importance of NFTs
and amyloid plaques in AD?
Neurofibrillary tangles are not specific for AD, partic-
ularly if a broader definition of NFTs includes different tau
isoforms or if one expands the expectations of the morpholo-
FIGURE 1. Histopathologic hallmarks of Alzheimer disease gical characteristics of NFTs (2, 3). In addition to AD, NFTs
demonstrated with Bielschowsky silver impregnation. (A) are also found in some frontotemporal dementias, myotonic
Neuritic plaques are extracellular fibrillary amyloid deposits,

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


surrounded by swollen, degenerating, argyrophilic neurites.
dystrophy, viral panencephalitis, dementia pugilistica, some
(B) Neurofibrillary tangles are composed of intracellular, prion diseases, and other brain diseases (4, 5). For many of
insoluble, and protease-resistant fibrillary polymers of tau these disorders, the severity of NFT pathology is less than
protein. In both panels, there are wispy argyrophilic neuropil that observed in end-stage AD. No condition characterized by
threads. Scale bars = 25 Km. widespread neocortical NFTs lacks extensive neurodegenera-
tion and clinical dementia. On the other hand, there are many
subtypes of chronic brain diseases in which there are ex-
normal cell constituents (Fig. 1), to indicate more of a toxic tensive neurodegeneration and clinical dementia without
than protective role. There may be both harmful and adap- NFTs, such as many subtypes of frontotemporal dementias,
tive aspects of a given phenomenon, of course, and AD le- synucleinopathies, subacute or chronic infarcts, metabolic,
sions presumably both result from, and in turn stimulate, demyelinating, developmental, and trinucleotide repeats dis-
other toxic factors. eases. Nor is there any doubt that tau protein itself can

TABLE 1. Definition of Terms


Pathological terms
Neurofibrillary pathology
Neurofibrillary pathology comprises aberrant, insoluble, and protease-resistant tau aggregates in various cellular compartments. Neuropil threads and the
intracellular components of neuritic plaques are subsets of neurofibrillary pathology that are present within dendrites or neurites.
Neurofibrillary tangle
Neurofibrillary tangle is the term that describes neurofibrillary pathology found in cell bodies.
Amyloid plaques
Amyloid plaques are extracellular, often roughly spherical, proteinaceous deposits stainable with Congo red, silver stains, and thioflavine histologically.
Fibrillary polymers of the AA peptide comprise the structural core of amyloid plaques in Alzheimer disease. Different subsets of amyloid plaques are
defined later.
Neuritic plaques vs diffuse plaques
Neuritic plaques are extracellular amyloid deposits invested by swollen degenerating neurites. The swollen neurites contain filamentous tau protein
aggregates identical structurally to the inclusions within neurofibrillary tangles. Diffuse plaques lack the presumed degenerating and/or aberrant
tau-immunoreactive neuritis.
Braak Stages
Braak stages refer to the relatively predictable progression of neurofibrillary tangle-type pathology in the brain during the course of Alzheimer disease. In
early stages (IYIII) the pathology is mostly isolated to the mesial temporal lobe structures, but later stages (IVYVI) progressively affect the neocortex
(see later).
Anatomical terms
Mesial temporal lobe structures (allocortex)
Mesial temporal lobe structures in the human brain comprise allocortical structures including the entorhinal cortex, amygdala, and the cornu ammonis fields
(CA1-CA4) and subiculum of the hippocampus. Mesial temporal lobe areas functionally play an important role in consolidating short-term memory.
Isocortex or neocortex
Neocortex refers to areas of cerebral cortex, also known as isocortex, that have 6 cellular layers. Neocortical areas subserve higher-order functions
including aspects of judgment, executive function, and so on. The contradistinction between mesial temporal lobe areas and neocortical areas is
important in comprehending the predictable, but nonlinear, progression of pathology in Alzheimer disease.

2 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009 Neuropathology and Cognitive Decline in AD

(11Y13). Notably, without NFTs or amyloid angiopathy, am-


yloid plaques are not associated with neurodegeneration (see
later). In sum, AD involves a specific combination of neu-
ritic amyloid plaques and NFTs; the neuritic plaques are more
specific to the disease, and the NFTs seem more likely to
induce neurodegeneration.
Specificity of AD Lesions: General and
Theoretical Considerations About the Clinical
Context of AD, Neuroanatomy, and Synergy
Between Plaques and NFTs
One challenge of CP studies in AD is to place nonlinear
disease processes onto quasi-linear continua for purposes of
correlation. The pathological changes in AD follow a complex
stereotyped neuroanatomical pattern (14). Prior studies have
produced a general scheme for how the pathology correlates
with clinical outcomes (Figs. 2 and 3), but it is important
to consider the clinical context for this association.
Epidemiological studies indicate that clinical AD prev-

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


alence doubles every half decade after age 65 years so that
approximately 25% of 90-year-olds carry the diagnosis of
AD (15, 16). The time from formal clinical diagnosis to death
is only approximately 8 years (17). Structural and cognitive

FIGURE 2. (A) An idealized clinicopathological correlation may


not be applicable to Alzheimer disease. (B) A conventional
metric for cognition, the Mini-Mental State Examination
(MMSE) and the Braak staging method provide a relatively
strong correlation between pathological severity and antemor-
tem cognitive decline. Data depicted are from the University of
Kentucky Alzheimer’s Disease Center (63). Each patient in this
series lacked concomitant pathological processes, for example,
advanced cerebrovascular disease, and died within a year of
their last MMSE score. Numbers of patients from each group:
Braak Stage 0, n = 8; I, n = 15; II, n = 18; III, n = 13; IV,
n = 12; V, n = 6; and VI, n = 25. Error bars are SD.

directly trigger neurodegeneration: many germ line mutations


in tau produce clinical dementia with NFTs (3, 6). These
tauopathies are distinct from AD, but common pathways may
be involved. The bottom line is that NFTs appear in multiple
brain diseases, and some researchers, including the present
authors, infer that NFTs contribute to neurodegeneration in
more than 1 disease state.
Unlike NFTs, the appearance of amyloid plaques in
AD brains is unique. Dystrophic neurites that contain
amyloid precursor protein are seen in traumatic brain injury
(7, 8), and Bdiffuse plaques[ can be observed in association FIGURE 3. Alzheimer disease (AD) progresses in a nonlinear
with dementia pugilistica (9). BLewy plaques[ have also been manner that renders clinicopathological correlation a chal-
described, with a corona of dystrophic >-synucleinYpositive lenge. The disease duration is long, there is a preclinical phase,
neurites (10). By contrast, the particular appearance of neu- in which pathological findings are present, but there are no
ritic plaques (i.e. AA peptideYcontaining extracellular lesions clinical manifestations. Further, each patient has a unique
surrounded by tau neurofibrillary pathology) is considered to constellation of clinical and pathological features. Nonetheless,
there is a relatively predictable progression of both clinical and
be specific for AD. Amyloid plaques are not a nonspecific
pathological indices. This schematic illustrates that the associ-
reaction to neurofibrillary pathology because non-AD tauo- ation between mesial temporal neurofibrillary tangles (NFTs)
pathies lack amyloid plaques. Further attesting to the spec- and neocortical neuritic plaques with cognitive decline may be
ificity of AD-type amyloid plaques is the fact that mutations weak, whereas the association between neocortical NFTs and
or duplications in the amyloid precursor protein gene pro- cognitive decline is strong. Early mesial temporal NFTs may
duce the specific features of AD, clinically and pathologically play a role in memory dysfunction in early AD.

Ó 2008 American Association of Neuropathologists, Inc. 3

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Nelson et al J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009

disease is present over a longer time span, however, certainly As if factoring in both the clinical course and the
over a decade before death and perhaps considerably longer anatomical aspects of the disease was not difficult enough,
(18Y24). Using these data, we deduce that an average person there is added complexity in the lesions of AD. By definition,
dying at age 78 years will have approximately 5% to 10% AD is characterized by the presence of both amyloid plaques
likelihood of having an antemortem diagnosis of AD, but at and neurofibrillary pathology, and there seems to be some
least approximately 20% to 40% likelihood of having signifi- synergy of the two, most definitely in neuritic plaques, but
cant AD-type pathology. Thus, many individuals with AD- the tracking of 2 separate pathologies renders CP correlations
related pathology and minimal or no detectable antemortem challenging. In the multidecade course of AD progression,
cognitive deficits die of other causes. Theoretical and empiri- amyloid plaques and NFTs may be first observed in different
cally derived data agree on this important point, as shown in parts of the brain: NFTs tend to appear first in allocortical
Figure 4. Figure 4B shows data of Del Tredici and Braak (25) structures, whereas amyloid plaques may first be found in the
based on 3,928 cases. The existence of nondemented persons neocortex (26, 27, 31Y35). Because 2 different types of
with AD-type pathology is hence not a potential confound; it pathology develop initially without apparent direct correla-
is obvious that many persons die in the preclinical phase of tion to each other, a linear continuum is difficult to establish.
AD. Preclinical and subclinical diseases are well-accepted A further layer of complexity is that amyloid plaques and
ideas in cancer, atherosclerosis, and in many other diseases, NFTs are probably not static lesions (see later).
but seem to cause confusion in discussions of AD. These considerations help explain why some prior
In addition to the concept of preclinical AD, there are studies have shown apparent general discrepancies with regard
other potential pitfalls in CP studies. Distinct neuroanatom- to the correlation of extent of pathology and the severity of

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


ical regions are affected at different stages of AD, and this premortem cognitive decline (1). However, apparent incon-
must be taken into account for CP correlations. For example, gruities have been asserted by a number of researchers, and a
portions of the allocortex (Table 1), including the hippo- discussion is provided later regarding specific studies.
campus and entorhinal cortex, may be involved severely
early in AD (14, 26Y28) when neocortical areas are still Specificity of AD Lesions: Particular Cases and
relatively well preserved. The involvement of neocortical Pathological Combinations
areas presumably underlies the later-stage cognitive impair- AD is defined by the presence of neuritic plaques and
ment. Therefore, study of hippocampus may be best for neurofibrillary pathology, the abundance of which is hy-
assessing earlier memory changes, but a study limited to pothesized to correlate with the severity of cognitive de-
hippocampus would be inappropriate for correlating overall terioration. If this is so, then certain corollaries must hold
pathology with overall cognitive decline. An important case true: 1) there should not be individuals with extremely abun-
study is that of Patient H.M., who had bilateral near-complete dant plaques and tangles but no cognitive deterioration; 2)
temporal lobectomies in 1953 (29, 30). Although mostly there should not be clinical dementia cases with abundant
amnestic, Patient H.M. is still quite robust cognitively more plaques and no NFTs; 3) there should not be patients with
than 5 decades later without AD (30) because most cognitive clinical dementia in whose brains there is no detectable
domains impaired in AD pertain to neocortical involvement pathological substrate; and 4) although there exist a number
(i.e. nondeclarative memory, judgment, gnosia, visuospatial of tauopathies (progressive supranuclear palsy, corticobasal
skills, language, executive function, etc.). degeneration, Guamanian parkinsonism, FTDP-17, etc.),

FIGURE 4. (A) The epidemiology of Alzheimer disease (AD) and the duration of the disease process predict that many persons
with appreciable AD pathology die before the onset of dementia. Assuming a 10-year preclinical stage, approximately as many
individuals would have significant AD pathology without the clinical disease as would manifest the disease during life. The
epidemiological data represent an average of 6 large studies, as summarized previously (16). (B) Empirical data confirm the
prediction that many individuals who die of other causes show appreciable AD-type pathology. Empirical data are from 3,928
cases in the indicated age ranges (25). Note that Braak Stages III/IV correspond to National Institute on AgingYReagan Institute
Bintermediate likelihood[ for the diagnosis of AD (46).

4 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009 Neuropathology and Cognitive Decline in AD

there should not be a true Btau-only[ AD. These ideas have


TABLE 2. Autopsy Series on Nondemented Persons With
been tested repeatedly and have fostered controversy. Differ-
False-Positive Pathological Diagnosis of National Institute on
ent specific groups will be discussed in turn. AgingYReagan Institute High Likelihood for Alzheimer Disease
(Braak Stages V and VI)
Amyloid Plaques and Neurofibrillary Pathology
Nondemented, Braak Braak Total Braak
But No Dementia n Stage V Stage VI Stage V/VI Reference
Cases with considerable AD-type pathology but with- 1* 0 1* 1 (47)
out documented cognitive decline have been termed patho- 49 0 0 0† (48)
logical aging and presymptomatic aging (36Y38). Because 142 1 0 1 (33)
mild cognitive impairment is considered to constitute an early 39 1 0 1 (49)
expression of AD (39), we here discuss only nondemented 42 3 0 3 (50)
patients. Such patients with incipient AD-like changes have 18 0 0 0† (56)
also been called preclinical AD based on magnetic resonance 17 0 0 0† (55)
imaging data (19, 40), neuropsychological testing (21, 41), 89 ?‡ ?‡ 3‡ (57)
Pittsburgh Compound B positron emission tomography 31 0 0 0 (51)
imaging (42), and cerebrospinal fluid biomarkers (43). 68 1 1 2 (54)
Pathologically, many nondemented cases meet the older 59§ 3 1§ 4
Khachaturian diagnostic criteria (44) and a significant minority n = 555 n = 15
meet Consortium to Establish A Registry for Alzheimer’s

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


Disease definite criteria for AD (41, 45). These criteria have *Case report (47).
†Braak stages inferred.
been superseded for the stand-alone AD diagnosis since 1997 ‡Cases designated as Braak V/VI.
(46). Moreover, these criteria are not relevant directly to a §From the University of Kentucky Alzheimer’s Disease Center, Braak VI case with
discussion of false-positives because they were established for relatively low neocortical neurofibrillary tangles (Fig. 5).
making the diagnosis of AD in clinically demented patients.
The pathological diagnosis of Bintermediate likelihood[
for AD by the current National Institute on AgingYReagan (62, 63). Of 59 longitudinally followed nondemented sub-
Institute (NIARI) criteria corresponds to Braak Stages III and jects with MMSE scores within a year of death, 9 showed
IV (46), neither of which is by itself a substrate for profound Braak Stage IV, 3 Stage V, and 1 Stage VI (Table 2). The
cognitive impairment (Fig. 2). The NIARI criteria for single Braak Stage VI patient from this series had a final
pathological diagnosis of Bhigh likelihood[ for AD corre- MMSE score of 29 within a year of death. Note that in this
spond to Braak Stages V and VI (46). Note that the average brain, there was a relatively low density of neocortical NFTs
final Mini-Mental State Examination (MMSE) score for (Fig. 5, rightmost data points). This person seems to dem-
Braak Stage V is approximately 20 out of a possible 30 onstrate the lowest-severity Braak Stage VI. Thus, even
(Fig. 2), so may include some relatively normal individuals. among Braak Stage VI cases, individuals with lower neo-
A recent case report described a 92-year-old nondemented cortical NFT densities tend to have better preservation of
patient whose brain, on autopsy, contained Braak Stage VI cognitive faculties. Variability in Braak Stage VI severity is
pathology (47), but neocortical NFTs from this patient did underscored by different studies in which the median final
not appear particularly dense, and the patient did demonstrate MMSE scores range from less than 2 (64) to 14 (53) in this
cognitive deficits. In 3 different cognitive tests, she scored at group. Moreover, in a study of 168 autopsied persons
or below the 20th percentile for her age despite previously (Fig. 6), every case with neocortical NFT density greater
normal test results (47). Because there is as yet no published than a particular threshold (blue arrow) had clinical dementia
report of true end-stage pathology in a cognitively intact (MMSE, G20), just as every case with low or absent
individual, an assessment of larger studies is necessary. neocortical NFTs (red arrow) had final MMSE scores greater
In 2004, Fernando and Ince (48) described a population- than 20. In sum, whereas the correlation between AD-type
based autopsy series in which BIsevere neocortical NFTs were pathology and clinical impairment is not absolutely predict-
not seen in any nondemented individual.[ This result has been able in each patient, there seems to be a threshold of
obtained repeatedly from multiple centers (33, 47Y57), as pathology above which every patient, without exception, is
shown in Table 2 (N = 555 nondemented subjects, of whom profoundly impaired.
15 were Braak Stages V or VI). These data from many research
centers indicate a 2.7% false-positive rate in the NIARI criteria
for high-likelihood AD diagnosis, with mostly Braak Stage V ‘‘Tangle-Only‘‘ Dementia
cases. In addition, a number of other studies have noted that There are reports of autopsied patients with neuro-
nondemented patients have NFTs confined predominantly to fibrillary pathologyYpredominant dementia (5, 54, 65, 66).
the mesial temporal lobe structures (58Y61). These data support These cases are characterized by abundant NFTs in medial
the accuracy of the high likelihood NIARI criteria, but leave temporal lobe structures with no or few amyloid plaques
unanswered the important question: are there nondemented throughout the brain. The individuals are usually older and
persons with truly end-stage AD-type pathology? primarily had memory decline and reduced activities of daily
We addressed this issue using data from the University living. Whether this is a subtype of AD or a different entity
of Kentucky Alzheimer’s Disease Center autopsy series remains to be elucidated.

Ó 2008 American Association of Neuropathologists, Inc. 5

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Nelson et al J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009

FIGURE 5. Variations in clinical and pathological indices in Braak Stage VI cases from the University of Kentucky Alzheimer’s
Disease Center database (63). Cases are arranged left-to-right by increasing Mini-Mental State Examination (MMSE) scores. All

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


patients had undergone MMSE testing within a year before death. Patients with higher MMSE scores tended to have fewer
neocortical neurofibrillary tangles (NFTs). One nondemented patient (MMSE score, 29) had a relatively low number of neocortical
NFTs compared with the other more cognitively impaired patients.

Other diseases are now known to be characterized by


NFT formation and minimal or absent amyloid pathology.
These cases are now presumed to reflect tauopathies, which
are a diverse group of diseases that may be associated with
genetic (tau mutations) or environmental (Guamian or
postencephalitic parkinsonism) factors (3, 5). In most
population-based series, and in older patients, tauopathies
are uncommon (54, 57, 67).

‘‘Plaque-Only’’ Dementia
We have no personal experience of cases with
dementia, abundant neocortical neuritic plaques, or diffuse
plaques but with no other clinical or pathological explanation
for the cognitive decline. The phenomenon of plaque-only
dementia has been described (68, 69) but may partly
correspond to patients with dementia with cortical Lewy
bodies (70). Even in the recent series of plaque-only AD (68),
which described a group of 16 brains of persons with clinical
dementia but with no neocortical NFTs and a low number of
amyloid plaques, it is unclear whether or how other prevalent FIGURE 6. Data from the University of Kentucky Alzheimer’s
brain diseases that are capable of contributing to dementia Disease Center database demonstrate that neocortical neuro-
fibrillary tangle (NFT) counts predict dementia in patients with-
were excluded. Thus, with the apparent exception of a rare
out severe concomitant brain pathologies (n = 168). Neocortical
familial variant of historical significance (71), plaque-only counts comprised total counts from occipital (Brodmann area
dementia has not been demonstrated beyond doubt despite 17 and 18), inferior parietal lobule, superior and midtemporal
many studies addressing this topic. Without fuller clarifica- gyri, and middle frontal gyrus (63). Neocortical amyloid plaque
tion, it is possible that the small numbers of plaque-only counts are the combined amount of neuritic and diffuse amyloid
dementia cases described to date had cognitive decline plaques in the same sections. Above a certain neocortical count
caused by other neurodegenerative conditions. (blue arrow), all patients were demented (blue diamonds =
patients with the Mini-Mental State Examination [MMSE] G20).
Cases With Dementia Lacking Amyloid Plaques, By contrast, patients with mild or no dementia (MMSE, 920;
red circles) tend to have lower neocortical NFTs, and all patients
NFTs, and Other Pathological Substrates below a threshold of neocortical NFT counts (shown by red
The literature does not indicate an appreciable subset of arrow) have MMSE greater than 20. There is only a weak
patients with clinical dementia and histopathologically pris- correlation between the amyloid plaques (green triangles) and
tine brains on autopsy. Studies that show a significant the NFTs, but cases with high neocortical NFTs generally have
proportion of cases with dementia, low neurofibrillary high amyloid plaques.

6 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009 Neuropathology and Cognitive Decline in AD

pathology, and no other known pathological substrate, tend to Subcortical and/or lacunar infarcts alter the detection threshold
be the older studies that lack probes for recently described for dementia (74, 85Y87). The change in clinical course is
diseases. In 2000, Crystal et al (72) described a cohort with mirrored by a different pathological appearance in the brain.
Bdementia of unknown etiology,[ however, cognitive impair- Persons with CVD tend to have lower AD-related pathology
ment in this cohort was not extreme, and these patients with a given degree of cognitive impairment (63, 86). In other
showed specific contributory neuropathological features words, the frequent presence of concomitant CVD severely
including hippocampal sclerosis and vascular disease. dampens the association for other pathologies, such as neuro-
There are many additional causes of cognitive decline, fibrillary pathology and amyloid plaques, to the severity of
and these can be clinically diagnosed in most cases. For this antemortem cognitive decline. For example, Braak Stage IV
reason, in clinical series such as those at the University of pathology may combine with CVD to induce significant cog-
Kentucky Alzheimer’s Disease Center, which include rigor- nitive impairment, whereas Braak Stage V pathology may it-
ous longitudinal clinical assessments followed by brain self have more biologic impact, yet still be clinically silent in
autopsy, extremely rare patients with profound dementia the absence of concomitant pathological abnormalities.
and the premortem diagnosis of AD have lacked histopathol-
ogic substrates at autopsy. We and others have found that Concomitant Pathology: An Expanding List of
persons with concomitant pathologies die with less neuro- Non-AD Brain Diseases in Aged Persons
fibrillary pathology (63, 64). Identifying and taking into Although not as prevalent as CVD, non-AD neuro-
account the presence and magnitude of concomitant pathol- degenerative diseases are important for consideration. Synu-
ogies is probably the biggest challenge to studies of the aging cleinopathies are the second most prevalent neurodegenerative

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


human brain. disease category after AD; these include dementia with Lewy
bodies and Parkinson disease dementia. Depending on the
Concomitant Pathology: The Large Impact of study population, dementia with Lewy bodies and Parkinson
Cerebrovascular Diseases disease dementia constitute approximately 5% to 15% of all
The most prevalent concomitant pathology in the neurodegenerative diseases (57, 88). Cortical Lewy body
brains of aged persons, cerebrovascular disease (CVD), is pathology occurs frequently in association with AD (88, 89).
directly relevant to any discussion of CP studies in dementia Whatever the reason for this clustering, the phenomenon is
or AD. The challenges introduced by this widespread but important from the perspective of CP correlation. When cor-
unpredictable comorbidity in aged persons have been tical Lewy bodies are present, the correlation between AD
discussed previously (48, 64, 73Y79). Yet, the profound pathology and cognitive impairment becomes far less strong
impact of CVD on studies pertinent to cognition in the (63), probably because the Lewy body pathology is account-
elderly seems to be underappreciated, or even ignored, in the ing for additional cognitive impairment. Accordingly, we and
broader field of AD research. others have found that patients with AD and dementia with
Cerebrovascular disease spans a wide spectrum includ- Lewy bodies decline at a faster rate than pure AD (90Y92).
ing small- and large-vessel ischemic disease, embolic, ve- The most reliable diagnostic tool for detection of neocortical
nous, inflammatory, hypertensive, hemorrhagic, aneurysmal, Lewy bodies (i.e. immunohistochemistry for >-synuclein)
hypoglycemia, hypoxia, amyloidogenic, and others. The was not developed until the late 1990s, so earlier studies had
importance of CVD is at least 2-fold. First, some cerebrovas- less ability to detect these structures (93, 94). There are now
cular abnormalities are so common as to be the norm in ad- known non-AD dementias linked to mutations in valosin,
vanced old age. Second, CVD induces an unpredictable progranulin, TDP-43, CHMP2B, and various triplet-repeat
change in cognition and hence presents challenges to correlat- alleles (6, 95). These relatively newly discovered diseases
ing the severity of cognitive decline with other diseases. present an important caveat to interpreting older studies that
Clinical and subclinical CVD is common in elderly tackled the problem of CP correlation in dementia.
subjects. Cerebrovascular disease is detectable in 75% to 90% In addition to neurodegenerative diseases per se, some
of persons older than 90 years (63, 80), and whereas frank other common diseases such as hippocampal sclerosis, sub-
clinical strokes affect approximately 750,000 in America each stance abuse, mood disorders, cancer, hematomas, and hydro-
year, more than 11 million discrete but clinically silent cere- cephalus are directly contributory to cognitive impairment in
brovascular events are thought to occur over the same interval the elderly (96Y98). Together, neurodegenerative diseases,
(81). The high frequency of cerebrovascular changes among CVD, and other aging-associated brain diseases weaken the
elderly patients means that both cognition and other pathology apparent association between AD pathology and antemortem
need to be weighed in the context of an altered milieu. cognitive impairment. An analogy can be made to studying
Furthermore, unlike AD, CVD progresses in an anatomically, heart disease: it would be difficult to study the CP correlation
clinically, and temporally unpredictable fashion, and there is between coronary atheromas and heart function if three quar-
no systematic methodology for correlating pathological and ters of cases had bacterial endocarditis and/or severe arrhyth-
clinical findings (82). These are some reasons why CVD- mias as well. That would be true even if clinical parameters
related pathology is frequent in cases termed dementia of could be correctly and confidently assessed in each case.
unknown origin (72).
Importantly for CP studies, CVD shifts the detection Challenges Pertaining to Cognitive Assessment
threshold for cognitive changes during life. In elderly AD pa- Difficulties in CP correlation may have as much to
tients, CVD leads to more rapid cognitive deterioration (83, 84). do with the clinical as to the pathological component. In

Ó 2008 American Association of Neuropathologists, Inc. 7

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Nelson et al J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009

AD-relevant CP studies, the clinical aspect is commonly the variety of techniques, including silver stains, thioflavine S,
severity of cognitive impairment as quantified by test and immunohistochemistry. Excellent studies have put forth
scores. First, it is problematic to align cognition and its differing views of whether one should use neuritic plaques,
many substituent functions on a linear scale. Second, there is diffuse plaques, total plaque load, amyloid burden, or other
added difficulty in correlating brain disease with test scores. pathology metrics (107Y109). Studies also use differing
In other words, even if cognition could be placed on a valid techniques for counting and/or scoring their density (107).
linear continuum (a test score), it would still be unsafe to Despite the wide variations in study designs, several
assume that regression along that continuum will occur in a points emerge consistently among CP studies about amyloid
linear fashion that correlates with brain disease severity, plaques. First, compared with NFTs, there is a weaker direct
whether a histopathologic substrate exists. correlation between the density of amyloid plaques and the
Although outside the scope of this review, specific cog- severity of cognitive decline. Second, the amyloid plaque
nitive assessments are used for differing applications. More subtype that seems to correlate best with the severity of
challenging, memory-oriented, and sensitive tests such as the
AD Assessment Scale, Cognitive component have been used
frequently in clinical trials and clinical imaging studies when
intraindividual assessments can be correlated as an outcome
measure. By contrast, most CP studies have relied on less
sensitive tests such as MMSE, the Blessed Test, or the even
more semiquantitative Clinical Dementia Rating Scale for

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


CP correlation (26, 36, 52, 61, 63, 90, 99). These latter tests
have been more constantly used over time, assess multiple
cognitive domains, have relatively well-established norma-
tive values, and could be more specific for interpersonal and
cross-study CP correlations. It may be best to use a com-
prehensive battery of cognitive and functional tests such as
those used currently in the Uniform Data Set by Alzheimer’s
Disease Centers (100).
A further complication to CP studies is that many
nonstructural factors alter cognitive assessment in the elderly.
These include cardiovascular, metabolic, infectious, medica-
tion related, and mood disorders and debilities such as
fatigue, arthritis, hearing loss, and visual impairments that
can also change test-taking behavior.
Collectively, many clinical factors render inevitable
some degree of variation in the results of cognitive assess-
ments and hence in correlating test scores with any other
parameter. There are thus many layers of complexity to CP
studies of AD. Yet, by taking into account some of the above
challenges, we can begin to address the existing CP litera-
ture and evaluate the hypothesis that amyloid plaques and
NFTs contribute in a biologic sense to the clinical manifes-
tations of AD.

CLINICOPATHOLOGIC STUDIES IN AD: REVIEW


OF THE LITERATURE
FIGURE 7. Neurofibrillary tangles (NFTs) in ascending reticular
Correlation of Amyloid Plaques and activating system (ARAS) including the acetylcholinergic
Cognitive Decline nucleus basalis of Meynert (NbM) do not map directly onto
More than 30 CP studies have assessed correlations most clinicopathologic studies. (A) Many NFTs are stained
between the density of amyloid plaques in human brains and using an antibody against phosphorylated tau protein in the
the severity of antemortem cognitive decline (26, 34, 36, 56, NbM of a midstage Alzheimer disease (AD). Scale bar =
61, 63, 99, 101Y106). These studies varied in research 50 KM. (B) Other ARAS nuclei, including the tuberomammil-
cohorts, anatomical areas examined, pathological methods, lary nucleus (TM), the locus caeruleus (LC), ventral tegmental
area (VTA), and the brainstem raphe areas also show many
amyloid plaque subcategories, plaque-counting techniques, NFTs and neuronal loss in AD patient. Neurotransmitters that
metrics for cognition and the range of cognitive decline are known to augment memory, arousal, attention, and mood
tested, and the rigor with which concomitant pathologies are indicated alongside the ARAS cell groups that produce
were evaluated and/or factored into the study. Controversy them. The ARAS neurons are the only source of those neuro-
exists about the best way to calculate the extent of amyloid transmitters in cerebral cortex, and thus pathological alter-
plaque pathology. Neuritic plaques can be visualized via a ations in ARAS may contribute to clinical symptoms.

8 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009 Neuropathology and Cognitive Decline in AD

cognitive decline is the neuritic plaque. Third, the patterns deposits that are invested by degenerating or dying back
seen in aged persons’ brain seem to segregate into these 3 nerve cell processes (Table 1; Fig. 1). These abnormal
groups (note that many refers to counted lesions in neo- dendrites and axons contain aberrant tau fibrils identical to
cortex): 1) Few plaques, few NFTs, and no cognitive those seen in NFTs. In a given neuritic plaque, axons from a
impairment; 2) Many plaques, few NFTs, and no cognitive variety of different sources expressing distinct neurotrans-
impairment; and 3) many plaques, many NFTs and cognitive mitter signatures may be present (114, 115). This is therefore
impairment. an important clue to AD pathogenesis because neuritic
Because in part of the scarcity of neocortical NFTs in plaques represent unambiguously a nidus in which extrac-
the absence of plaques, it has been suggested that the path- ellular amyloid plaque pathology induces intracellular neuro-
ogenetic effect of amyloid plaques may be mediated through fibrillary pathology and apparent structural and functional
stimulating neurofibrillary pathology (63, 110). The relative disruption (116, 117). Although the particular toxic sub-
importance of amyloid plaques in terms of correlating with stance(s) within neuritic plaques are still not definitively
cognitive decline may be greatest in the earliest stages of the known (i.e. they may represent specific conformation(s) of
disease, before there are widespread NFTs and neocortical the AA peptide [118]), this apparent Bsmoking gun[ is dif-
neurodegeneration (99). By contrast, there is relatively little ficult to interpret in any other way.
evidence of a direct effect of amyloid plaques in contributing
to the later-stage cognitive decline in AD. Germane to this CORRELATION OF NFTs AND
topic but often neglected is the question of lesion turnover. COGNITIVE DECLINE
The apparent success of AA vaccine trials attests to the ability

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


Dozens of research groups have assessed the associa-
of the immune system to clear amyloid plaques (111), but it tion between NFTs and the severity of cognitive decline
is not known whether a toxic plaque substance is left unaf- (26, 34, 36, 56Y58, 61, 63, 99, 101Y106, 119Y122). As with
fected in these studies. Microglia appear to actively scavenge amyloid plaque research, studies of NFTs have been defined
the fibrillar material in amyloid plaques (112). There is more by different staining techniques, followed many different
direct evidence of amyloid plaque turnover in animal models study designs, and led to differing conclusions. Regardless of
(113). This phenomenon would tend to dampen the correla- the staining or counting method, however, the correlation
tion between pathology and clinical features. between neocortical NFTs and antemortem cognitive decline
To a degree far greater than for NFTs, there is a strong is strong in studies that span the clinical spectrum of AD.
association between amyloid plaques and genetic factors of Several important themes emerge, including the importance
risk for AD. As a rule, environmental and genetic factors that of neuroanatomical considerations and the different subtypes
predict AD risk also appear to directly potentiate amyloid of tau pathology in AD brains.
plaques. These risk factors include head trauma, the ApoE4 In the course of AD, the development of NFTs follows
allele, Down syndrome, amyloid precursor protein mutations a predictable pattern (14, 31, 123). This pattern seems to
and duplications, SORL1 variants, and mutations in PSEN1 correlate on the 1 hand with where neurons die (124Y127)
and PSEN2 genes (13). This strong genetic association is and on the other with the cognitive domains affected in AD.
relevant to the question of the correlation with cognitive For example, in the earliest stages of AD, symptoms tend to
symptoms because all of these alleles are also strong risk relate to memory loss. At this stage of the disease, the
factors for dementia. allocortical substrates of memory are strongly affected by
NFTs, but other areas are not. The cognitive domains
The Neuritic Plaque: A Pathological Entity With affected in midstage and late-stage AD expand to include
Strong Mechanistic Implications areas of executive function, judgment, visuospatial capacities,
Neuritic plaques deserve special consideration. Neuritic and speech. This pattern is paralleled by the development of
plaques comprise roughly spherical extracellular amyloid NFTs in the neocortical areas that subserve those functions.

TABLE 3. Summary Points


Nonlinear clinicopathological correlations are expected in Alzheimer disease (AD)
& There are prevalent (9two thirds of patients) comorbidities in aged brains, including cerebrovascular diseases, synucleinopathies, frontotemporal, and
TDP-43Yrelated diseases that inevitably skew correlations between pathology and cognition
& The relationship between cognitive decline and brain disease is not linear, and both are difficult to quantify
& There are 2 separate important subtypes of AD pathologyVamyloid plaques and neurofibrillary tanglesVthat develop in different patterns in AD brains after a
predictable course
Summary
& Evidence from AD clinicopathological studies strongly supports the existence of a specific prevalent disease as defined by the presence of clinical dementia,
amyloid plaques, and neurofibrillary tangles
& End-stage AD patients have extremely high burden of plaques and tangles, and there is not a subset of cases with very dense AD-type lesions lacking cognitive
decline
& There is not a significant subset of cases with severe cognitive decline lacking a pathological substrate
& Results are compatible with the hypothesis that amyloid plaques and neurofibrillary tangle contribute to clinical decline in AD

Ó 2008 American Association of Neuropathologists, Inc. 9

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Nelson et al J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009

A special anatomical consideration referent to NFTs in neurons may harbor intracellular NFTs over several decades
AD is the impact of NFTs on the ascending reticular (148, 149). This finding has been interpreted to indicate that
activating system (ARAS). Ascending reticular activating NFTs are benign (1), although a pathological process that
system neurons send strong inputs to the cerebral cortex that takes place over such a timeframe could well be toxic
augment memory, arousal, attention, and motivation. These ultimately, as in many other diseases of development or
cells also are severely affected in AD. For example, although aging. Before inducing cell death per se, these conspicuous
the Bcholinergic hypothesis[ of AD (128, 129) is now changes in cell bodies, axons, and dendrites might be
considered somewhat dated, the impact from NFTs on the significantly impairing function.
ARAS cholinergic nucleus basalis of Meynert (NbM) may be
profound. NFTs are numerous in the NbM starting early in
the disease (Fig. 7), and NFT density parallels cognitive LIMITATIONS TO CP STUDIES IN AD
decline severity as does cerebral cortical cholinergic tone
Although many CP studies support the hypothesis that
(130Y132). The ARAS neurons, as a rule, supply neuro-
NFTs and amyloid plaques contribute to the cognitive decline
transmitters that are absent from intrinsic cortical neurons.
in AD, there are important limitations to these studies and
For example, the NbM is a major source of cholinergic
significant unanswered questions. The schema of amyloid
neurotransmission in the cerebral cortex, which itself harbors
plaques and NFTs Bcausing[ AD is oversimplistic and in-
no cholinergic cells (133). The importance of cortical
complete. There are at least 2 relevant ideas that must be
cholinergic tone is underscored by the fact that cholinergic
acknowledged: 1) Correlation does not establish causation.
antagonists such as scopolamine induce transient but com-

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


Although the density of NFTs on autopsy correlates with
plete amnesia in humans (134). NFTs are also thought to
cognitive decline severity, and the experimental results align
affect adversely other ARAS neurotransmitters including
with a plausible hypothesis, this does not prove that NFTs are
histamine, norepinephrine, dopamine, and serotonin (corre-
directly neurotoxic; and 2) Results raise the question of what
sponding to neurodegeneration and NFT-containing cells of
induces the formation of amyloid plaques and NFTs. For
the tuberomammillary nucleus, locus caeruleus, ventral teg-
many, if not most, AD patients, there are no known specific
mental area, and pontine raphe, respectively) (135Y141).
genetic or environmental triggers that would account for the
There may be clinical manifestations associated with this
development of amyloid plaques and NFTs. Yet, acknowl-
subcortical pathology, although this would not be detected in
edging that these lesions probably contribute to cognitive
most CP studies because consensus criteria for AD diagnosis
decline is important because it gives us a target for exper-
currently focus on cortical pathology (46).
imental studies.
As with amyloid plaques, there are subcategories of
neurofibrillary pathology; of these, NFTs are just one
(Table 1). Less attention has been focused on the other
subtypes possibly because neurofibrillary pathology is absent SUMMARY AND CONCLUSIONS
in most animal models and not generally amenable to Neuropathology of aging-related brain disease must
positron emission tomographic scans. There is, however, take into account diverse medical, technical, biochemical,
abundant dendritic and axonal neurofibrillary pathology in and anatomical considerations (Table 3). For example,
AD brains (Fig. 1); these also are known as Bneuropil concurrent pathologies are very common in aging brains.
threads[ and Bcurly fibers[ (142Y145). All the neurofibrillary These diseases and many other factors are, collectively,
lesions, which contain tau polymer fibrils called paired formidable obstacles to aligning pathology and cognition
helical filaments (142), may contribute to cognitive decline along linear scales. An extensive literature on CP correlations
in AD (103). They provide another reason why a simple in AD indicates a sequence that may begin with specific
linear correlation between Bplaques and tangles[ would not genetic and environmental factors that increase risk of
be expected in relation to cognitive decline, although the widespread amyloid plaques. At some point, neuritic plaques
pathways involved in the formation of those lesions may appear that combine extracellular amyloid and intracellular
contribute to the disease. neurofibrillary pathology. Whether independently or not,
Also in common with amyloid plaques, NFTs are NFTs develop first in the allocortex and spread to neocortical
quantified only at death, so it is important to consider areas. When neocortical NFTs are abundant, neurodegenera-
whether some NFTs are removed from the brain in the course tion occurs and, over a certain threshold, cognitive impair-
of AD. This has not been proven either way. Some, but not ment is inevitable. The NIARI consensus Bhigh likelihood for
all, studies have indicated that more neurons disappear in AD AD[ pathological criteria are approximately 97% specific for
brains than can be explained directly by the number of NFTs measurable cognitive impairment, and the false-positives
present at autopsy (126, 146, 147, but see Fukutani et al (nondemented persons with plaques and tangles) are brains
[127]); this might indicate NFT turnover and/or a non-NFT in which the highest pathological burden has not been
pathological mechanism. Extracellular NFTs are invested reached. These results are within realistic expectations for
by microglial cells and astrocytes, but it is unclear whether preclinical AD pathology. Among patients with measurable
these microglial scavengers successfully digest the protease- clinical disease, the extent of cognitive impairment parallels
resistant insoluble neurofibrillary material. Morsch et al (148) the severity of neurofibrillary pathology. In sum, the as-
modeled the correlation between NFT counts and neuron sociation of plaques and tangles with cognitive impairment in
loss in human hippocampi and concluded that individual AD is complex but coherent.

10 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009 Neuropathology and Cognitive Decline in AD

ACKNOWLEDGMENTS 21. Galvin JE, Powlishta KK, Wilkins K, et al. Predictors of preclinical
Alzheimer disease and dementia: A clinicopathologic study. Arch
The authors thank all of the participants in the Neurol 2005;62:758Y65
longitudinal aging study and brain banks. The authors thank 22. Mosconi L, De Santi S, Rusinek H, et al. Magnetic resonance and PET
Richard Kryscio, PhD, and Marta Mendiondo, PhD, for data studies in the early diagnosis of Alzheimer’s disease. Expert Rev
management and statistical support; Ela Patel, Ann Tudor, Neurother 2004;4:831Y49
Paula Thomason, Dr Huaichen Liu, and Sonya Anderson for 23. Rusinek H, De Santi S, Frid D, et al. Regional brain atrophy rate
predicts future cognitive decline: 6-Year longitudinal MR imaging
technical support; Frederick Schmitt, PhD, Gregory Jicha, study of normal aging. Radiology 2003;229:691Y96
MD, PhD, Charles Smith, MD, Gregory Cooper, MD, PhD, 24. Backman L, Jones S, Berger AK, et al. Cognitive impairment in
Nancy Stiles, MD, and Allison Caban-Holt, PhD, for clinical preclinical Alzheimer’s disease: A meta-analysis. Neuropsychology
evaluations; and Daron Davis, MD, and Dianne Wilson, 2005;19:520Y31
25. Del Tredici K, Braak H. Neurofibrillary changes of the Alzheimer type
MD, for pathological assessments. in very elderly individuals: Neither inevitable nor benign: Commentary
on BNo disease in the brain of a 115-year-old woman.[ Neurobiol
Aging 2008;29:1133Y36
26. Berg L, McKeel DW Jr, Miller JP, et al. Clinicopathologic studies in
REFERENCES cognitively healthy aging and Alzheimer’s disease: Relation of
1. Castellani RJ, Lee HG, Zhu X, et al. Alzheimer disease pathology as a histologic markers to dementia severity, age, sex, and apolipoprotein
host response. J Neuropathol Exp Neurol 2008;67:523Y31 E genotype. Arch Neurol 1998;55:326Y35
2. Goedert M. Tau protein and neurodegeneration. Semin Cell Dev Biol 27. Giannakopoulos P, Hof PR, Giannakopoulos AS, et al. Regional
2004;15:45Y49 distribution of neurofibrillary tangles and senile plaques in the cerebral
3. Buee L, Bussiere T, Buee-Scherrer V, et al. Tau protein isoforms, cortex of very old patients. Arch Neurol 1995;52:1150Y59

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


phosphorylation and role in neurodegenerative disorders. Brain Res 28. Mitchell TW, Mufson EJ, Schneider JA, et al. Parahippocampal tau
Brain Res Rev 2000;33:95Y130 pathology in healthy aging, mild cognitive impairment, and early
4. Nelson PT, Keller JN. RNA in brain disease: No longer just Bthe Alzheimer’s disease. Ann Neurol 2002;51:182Y89
messenger in the middle.[ J Neuropathol Exp Neurol 2007;66:461Y68 29. Scoville WB, Milner B. Loss of recent memory after bilateral
5. Feany MB, Dickson DW. Neurodegenerative disorders with extensive hippocampal lesions. J Neurol Neurosurg Psychiatry 1957;20:11Y21
tau pathology: A comparative study and review. Ann Neurol 1996;40: 30. Corkin S. What’s new with the amnesic patient H.M.? Nat Rev
139Y48 Neurosci 2002;3:153Y60
6. Cairns NJ, Bigio EH, Mackenzie IR, et al. Neuropathologic diagnostic 31. Braak H, Braak E. Staging of Alzheimer’s diseaseYrelated neuro-
and nosologic criteria for frontotemporal lobar degeneration: Consensus fibrillary changes. Neurobiol Aging 1995;16:271Y8;[discussion 8-84]
of the Consortium for Frontotemporal Lobar Degeneration. Acta 32. Price JL, Davis PB, Morris JC, et al. The distribution of tangles,
Neuropathol (Berl) 2007;114:5Y22 plaques and related immunohistochemical markers in healthy aging and
7. Graham DI, Gentleman SM, Nicoll JA, et al. Altered beta-APP Alzheimer’s disease. Neurobiol Aging 1991;12:295Y312
metabolism after head injury and its relationship to the aetiology of 33. Bennett DA, Schneider JA, Arvanitakis Z, et al. Neuropathology of
Alzheimer’s disease. Acta Neurochir Suppl 1996;66:96Y102 older persons without cognitive impairment from two community-
8. Uryu K, Chen XH, Martinez D, et al. Multiple proteins implicated in based studies. Neurology 2006;66:1837Y44
neurodegenerative diseases accumulate in axons after brain trauma in 34. Markesbery WR, Schmitt FA, Kryscio RJ, et al. Neuropathologic
humans. Exp Neurol 2007;208:185Y92 substrate of mild cognitive impairment. Arch Neurol 2006;63:38Y46
9. Roberts GW, Allsop D, Bruton C. The occult aftermath of boxing. J 35. Petersen RC, Parisi JE, Dickson DW, et al. Neuropathologic fea-
Neurol Neurosurg Psychiatry 1990;53:373Y78 tures of amnestic mild cognitive impairment. Arch Neurol 2006;63:
10. Braak H, Del Tredici K. Invited article: Nervous system pathology in 665Y72
sporadic Parkinson disease. Neurology 2008;70:1916Y25 36. Green MS, Kaye JA, Ball MJ. The Oregon brain aging study:
11. Yoshioka K, Miki T, Katsuya T, et al. The 717Val-Ile substitution in Neuropathology accompanying healthy aging in the oldest old.
amyloid precursor protein is associated with familial Alzheimer’s Neurology 2000;54:105Y13
disease regardless of ethnic groups. Biochem Biophys Res Commun 37. Crystal HA, Dickson DW, Sliwinski MJ, et al. Pathological markers
1991;178:1141Y46 associated with normal aging and dementia in the elderly. Ann Neurol
12. Goate A, Chartier-Harlin MC, Mullan M, et al. Segregation of a 1993;34:566Y73
missense mutation in the amyloid precursor protein gene with familial 38. Morris JC, Storandt M, McKeel DW Jr, et al. Cerebral amyloid
Alzheimer’s disease. Nature 1991;349:704Y6 deposition and diffuse plaques in Bnormal[ aging: Evidence for
13. Mayeux R, Hyslop PS. Alzheimer’s disease: Advances in trafficking. presymptomatic and very mild Alzheimer’s disease. Neurology 1996;
Lancet Neurol 2008;7:2Y3 46:707Y19
14. Braak H, Braak E. Neuropathological stageing of Alzheimer-related 39. Petersen RC, Negash S. Mild cognitive impairment: An overview. CNS
changes. Acta Neuropathol 1991;82:239Y59 Spectr 2008;13:45Y53
15. Hebert LE, Scherr PA, Bienias JL, et al. Alzheimer disease in the US 40. Skoog I. Detection of preclinical Alzheimer’s disease. N Engl J Med
population: Prevalence estimates using the 2000 census. Arch Neurol 2000;343:502Y3
2003;60:1119Y22 41. Schmitt FA, Davis DG, Wekstein DR, et al. BPreclinical[ AD revisited:
16. Katzman R, Kawas C. The epidemiology of dementia and Alzheimer Neuropathology of cognitively normal older adults. Neurology 2000;
disease. In: Terry RD, ed. Alzheimer Disease. New York, NY: Raven 55:370Y76
Press, LTD, 1994;105Y22 42. Mintun MA, Larossa GN, Sheline YI, et al. [11C]PIB in a nondemented
17. Williams MM, Xiong C, Morris JC, et al. Survival and mortality population: Potential antecedent marker of Alzheimer disease. Neurol-
differences between dementia with Lewy bodies vs Alzheimer disease. ogy 2006;67:446Y52
Neurology 2006;67:1935Y41 43. Chong MS, Sahadevan S. Preclinical Alzheimer’s disease: Diagnosis
18. Scarmeas N, Habeck CG, Hilton J, et al. APOE related alterations in and prediction of progression. Lancet Neurol 2005;4:576Y79
cerebral activation even at college age. J Neurol Neurosurg Psychiatry 44. Khachaturian ZS. Diagnosis of Alzheimer’s disease. Arch Neurol 1985;
2005;76:1440Y44 42:1097Y105
19. Smith CD, Chebrolu H, Wekstein DR, et al. Brain structural 45. Mirra SS. The CERAD neuropathology protocol and consensus
alterations before mild cognitive impairment. Neurology 2007;68: recommendations for the postmortem diagnosis of Alzheimer’s disease:
1268Y73 A commentary. Neurobiol Aging 1997;18:S91YS94
20. Thal DR, Del Tredici K, Braak H. Neurodegeneration in normal brain 46. Consensus recommendations for the postmortem diagnosis of Alzheim-
aging and disease. Sci Aging Knowledge Environ 2004;2004:pe26 er’s disease. The National Institute on Aging, and Reagan Institute

Ó 2008 American Association of Neuropathologists, Inc. 11

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Nelson et al J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009

Working Group on Diagnostic Criteria for the Neuropathological 69. Terry RD, Hansen LA, DeTeresa R, et al. Senile dementia of the
Assessment of Alzheimer’s Disease. Neurobiol Aging 1997;18:S1YS2 Alzheimer type without neocortical neurofibrillary tangles. J Neuro-
47. Berlau DJ, Kahle-Wrobleski K, Head E, et al. Dissociation of pathol Exp Neurol 1987;46:262Y68
neuropathologic findings and cognition: Case report of an apolipopro- 70. Hansen LA, Masliah E, Galasko D, et al. Plaque-only Alzheimer
tein E epsilon2/epsilon2 genotype. Arch Neurol 2007;64:1193Y96 disease is usually the Lewy body variant, and vice versa. J Neuropathol
48. Fernando MS, Ince PG. Vascular pathologies and cognition in a Exp Neurol 1993;52:648Y54
population-based cohort of elderly people. J Neurol Sci 2004;226:13Y17 71. Klunemann HH, Fronhofer W, Werner-Fuchtenbusch D, et al. Char-
49. Knopman DS, Parisi JE, Salviati A, et al. Neuropathology of cog- acterization of the kindred of Alois Alzheimer’s patient with
nitively normal elderly. J Neuropathol Exp Neurol 2003;62:1087Y95 plaque-only dementia. Alzheimer Dis Assoc Disord 2006;20:291Y94
50. Xuereb JH, Brayne C, Dufouil C, et al. Neuropathological findings in 72. Crystal HA, Dickson D, Davies P, et al. The relative frequency of
the very old. Results from the first 101 brains of a population-based Bdementia of unknown etiology[ increases with age and is nearly 50%
longitudinal study of dementing disorders. Ann N Y Acad Sci 2000; in nonagenarians. Arch Neurol 2000;57:713Y19
903:490Y96 73. Bowler JV, Munoz DG, Merskey H, et al. Fallacies in the pathological
51. Hulette CM, Welsh-Bohmer KA, Murray MG, et al. Neuropathological confirmation of the diagnosis of Alzheimer’s disease. J Neurol
and neuropsychological changes in Bnormal[ aging: Evidence for Neurosurg Psychiatry 1998;64:18Y24
preclinical Alzheimer disease in cognitively normal individuals. J 74. Schneider JA, Boyle PA, Arvanitakis Z, et al. Subcortical infarcts,
Neuropathol Exp Neurol 1998;57:1168Y74 Alzheimer’s disease pathology, and memory function in older persons.
52. Wang DS, Bennett DA, Mufson E, et al. Decreases in soluble Ann Neurol 2007;62:59Y66
alpha-synuclein in frontal cortex correlate with cognitive decline in 75. Todorov AB, Go RC, Constantinidis J, et al. Specificity of the clinical
the elderly. Neurosci Lett 2004;359:104Y8 diagnosis of dementia. J Neurol Sci 1975;26:81Y98
53. Whitwell JL, Josephs KA, Murray ME, et al. MRI correlates of 76. Chui HC, Zarow C, Mack WJ, et al. Cognitive impact of subcortical
neurofibrillary tangle pathology at autopsy: A voxel-based morphom- vascular and Alzheimer’s disease pathology. Ann Neurol 2006;60:
etry study. Neurology 2008;71:743Y49 677Y87

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


54. Noda K, Sasaki K, Fujimi K, et al. Quantitative analysis of neuro- 77. Rockwood K. Mixed dementia: Alzheimer’s and cerebrovascular dis-
fibrillary pathology in a general population to reappraise neuropatho- ease. Int Psychogeriatr 2003;15(suppl 1):39Y46
logical criteria for senile dementia of the neurofibrillary tangle type 78. Kovari E, Gold G, Herrmann FR, et al. Cortical microinfarcts and
(tangle-only dementia): The Hisayama Study. Neuropathology 2006; demyelination affect cognition in cases at high risk for dementia.
26:508Y18 Neurology 2007;68:927Y31
55. Newell KL, Hyman BT, Growdon JH, et al. Application of the National 79. Gold G, Giannakopoulos P, Herrmann FR, et al. Identification of
Institute on Aging (NIA)-Reagan Institute criteria for the neuro- Alzheimer and vascular lesion thresholds for mixed dementia. Brain
pathological diagnosis of Alzheimer disease. J Neuropathol Exp Neurol 2007;130:2830Y36
1999;58:1147Y55 80. White L, Small BJ, Petrovitch H, et al. Recent clinical-pathologic re-
56. Haroutunian V, Purohit DP, Perl DP, et al. Neurofibrillary tangles in search on the causes of dementia in late life: Update from the
nondemented elderly subjects and mild Alzheimer disease. Arch Neurol Honolulu-Asia aging study. J Geriatr Psychiatry Neurol 2005;18:224Y27
1999;56:713Y18 81. Centers for Disease Control and Prevention. Prevalence of strokeV
57. Sonnen JA, Larson EB, Crane PK, et al. Pathological correlates of United States, 2005. MMWR Morb Mortal Wkly Rep 2007;56:
dementia in a longitudinal, population-based sample of aging. Ann 469Y74
Neurol 2007;62:406Y13 82. Knopman DS. Dementia and cerebrovascular disease. Mayo Clin Proc
58. Arriagada PV, Marzloff K, Hyman BT. Distribution of Alzheimer-type 2006;81:223Y30
pathologic changes in nondemented elderly individuals matches the 83. Mungas D, Reed BR, Ellis WG, et al. The effects of age on rate of
pattern in Alzheimer’s disease. Neurology 1992;42:1681Y88 progression of Alzheimer disease and dementia with associated
59. Iseki E, Tsunoda S, Suzuki K, et al. Regional quantitative analysis of cerebrovascular disease. Arch Neurol 2001;58:1243Y47
NFT in brains of non-demented elderly persons: Comparisons with 84. Regan C, Katona C, Walker Z, et al. Relationship of vascular risk to the
findings in brains of late-onset Alzheimer’s disease and limbic NFT progression of Alzheimer disease. Neurology 2006;67:1357Y62
dementia. Neuropathology 2002;22:34Y39 85. Snowdon DA, Greiner LH, Mortimer JA, et al. Brain infarction and the
60. Price JL, Morris JC. Tangles and plaques in nondemented aging and clinical expression of Alzheimer disease. The Nun Study. JAMA 1997;
Bpreclinical[ Alzheimer’s disease. Ann Neurol 1999;45:358Y68 277:813Y17
61. Bancher C, Jellinger K, Lassmann H, et al. Correlations between 86. Petrovitch H, Ross GW, He Q, et al. Characterization of Japanese-
mental state and quantitative neuropathology in the Vienna longitudinal American men with a single neocortical AD lesion type. Neurobiol
study on dementia. Eur Arch Psychiatry Clin Neurosci 1996;246: Aging 2008;29:1448Y55
137Y46 87. Gold G, Kovari E, Herrmann FR, et al. Cognitive consequences of
62. Davis DG, Schmitt FA, Wekstein DR, et al. Alzheimer neuropathologic thalamic, basal ganglia, and deep white matter lacunes in brain aging
alterations in aged cognitively normal subjects. J Neuropathol Exp and dementia. Stroke 2005;36:1184Y88
Neurol 1999;58:376Y88 88. Jellinger KA. Influence of Alzheimer pathology on clinical diagnostic
63. Nelson PT, Jicha GA, Schmitt FA, et al. Clinicopathologic correlations accuracy in dementia with Lewy bodies. Neurology 2004;62:160;
in a large Alzheimer disease center autopsy cohort: Neuritic plaques [author reply]
and neurofibrillary tangles Bdo count[ when staging disease severity. J 89. Lee VM, Giasson BI, Trojanowski JQ. More than just two peas in a
Neuropathol Exp Neurol 2007;66:1136Y46 pod: Common amyloidogenic properties of tau and alpha-synuclein in
64. Jellinger KA, Attems J. Neuropathological evaluation of mixed neurodegenerative diseases. Trends Neurosci 2004;27:129Y34
dementia. J Neurol Sci 2007;257:80Y87 90. Kraybill ML, Larson EB, Tsuang DW, et al. Cognitive differences in
65. Ulrich J, Spillantini MG, Goedert M, et al. Abundant neurofibrillary dementia patients with autopsy-verified AD, Lewy body pathology, or
tangles without senile plaques in a subset of patients with senile both. Neurology 2005;64:2069Y73
dementia. Neurodegeneration 1992;1:257Y64 91. Olichney JM, Galasko D, Salmon DP, et al. Cognitive decline is faster
66. Yamada M, Itoh Y, Sodeyama N, et al. Senile dementia of the in Lewy body variant than in Alzheimer’s disease. Neurology 1998;51:
neurofibrillary tangle type: A comparison with Alzheimer’s disease. 351Y57
Dement Geriatr Cogn Disord 2001;12:117Y26 92. Nelson PT, Kryscio RJ, Abner EA, et al. Acetylcholinesterase inhibitor
67. Tschanz JT, Treiber K, Norton MC, et al. A population study of treatment is associated with relatively slow cognitive decline in patients
Alzheimer’s disease: Findings from the Cache County Study on with Alzheimer’s disease and AD+DLB. J Alzheimers Dis 2008
Memory, Health, and Aging. Care Manag J 2005;6:107Y14 (in press)
68. Tiraboschi P, Sabbagh MN, Hansen LA, et al. Alzheimer disease with- 93. Berg L, McKeel DW Jr, Miller JP, et al. Neuropathological indexes of
out neocortical neurofibrillary tangles: BA second look.[ Neurology Alzheimer’s disease in demented and nondemented persons aged
2004;62:1141Y47 80 years and older. Arch Neurol 1993;50:349Y58

12 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009 Neuropathology and Cognitive Decline in AD

94. Tomlinson BE, Blessed G, Roth M. Observations on the brains of 118. Shankar GM, Li S, Mehta TH, et al. Amyloid-beta protein dimers
demented old people. J Neurol Sci 1970;11:205Y42 isolated directly from Alzheimer’s brains impair synaptic plasticity
95. Kosaka K, Iseki E. Recent advances in dementia research in Japan: and memory. Nat Med 2008;14:837Y42
Non-Alzheimer-type degenerative dementias. Psychiatry Clin Neurosci 119. Gold G, Bouras C, Kovari E, et al. Clinical validity of Braak
1998;52:367Y73 neuropathological staging in the oldest-old. Acta Neuropathol (Berl)
96. Amador-Ortiz C, Ahmed Z, Zehr C, et al. Hippocampal sclerosis 2000;99:579Y82;[discussion 83Y84]
dementia differs from hippocampal sclerosis in frontal lobe degener- 120. Duyckaerts C, Bennecib M, Grignon Y, et al. Modeling the relation
ation. Acta Neuropathol (Berl) 2007;113:245Y52 between neurofibrillary tangles and intellectual status. Neurobiol
97. Knopman DS, Petersen RC, Cha RH, et al. Incidence and causes of Aging 1997;18:267Y73
nondegenerative nonvascular dementia: A population-based study. 121. Riley KP, Snowdon DA, Markesbery WR. Alzheimer’s neurofibrillary
Arch Neurol 2006;63:218Y21 pathology and the spectrum of cognitive function: Findings from the
98. Erkinjuntti T, Sulkava R, Kovanen J, et al. Suspected dementia: Eval- Nun Study. Ann Neurol 2002;51:567Y77
uation of 323 consecutive referrals. Acta Neurol Scand 1987;76:359Y64 122. Arriagada PV, Growdon JH, Hedley-Whyte ET, et al. Neurofibrillary
99. Tiraboschi P, Hansen LA, Thal LJ, et al. The importance of neuritic tangles but not senile plaques parallel duration and severity of
plaques and tangles to the development and evolution of AD. Alzheimer’s disease. Neurology 1992;42:631Y39
Neurology 2004;62:1984Y89 123. Braak H, Braak E, Bohl J. Staging of Alzheimer-related cortical
100. Beekly DL, Ramos EM, Lee WW, et al. The National Alzheimer’s destruction. Eur Neurol 1993;33:403Y8
Coordinating Center (NACC) database: The uniform data set. 124. Bobinski M, Wegiel J, Tarnawski M, et al. Relationships between
Alzheimer Dis Assoc Disord 2007;21:249Y58 regional neuronal loss and neurofibrillary changes in the hippocampal
101. Guillozet AL, Weintraub S, Mash DC, et al. Neurofibrillary tangles, formation and duration and severity of Alzheimer disease. J Neuro-
amyloid, and memory in aging and mild cognitive impairment. Arch pathol Exp Neurol 1997;56:414Y20
Neurol 2003;60:729Y36 125. Rossler M, Zarski R, Bohl J, et al. Stage-dependent and sector-specific
102. Imhof A, Kovari E, von Gunten A, et al. Morphological substrates of neuronal loss in hippocampus during Alzheimer’s disease. Acta

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016


cognitive decline in nonagenarians and centenarians: A new paradigm? Neuropathol 2002;103:363Y69
J Neurol Sci 2007;257:72Y79 126. Gomez-Isla T, Hollister R, West H, et al. Neuronal loss correlates with
103. Cummings BJ, Pike CJ, Shankle R, et al. Beta-amyloid deposition and but exceeds neurofibrillary tangles in Alzheimer’s disease. Ann Neurol
other measures of neuropathology predict cognitive status in Alzhei- 1997;41:17Y24
mer’s disease. Neurobiol Aging 1996;17:921Y33 127. Fukutani Y, Kobayashi K, Nakamura I, et al. Neurons, intracellular and
104. Davis PC, Gearing M, Gray L, et al. The CERAD experience, Part extracellular neurofibrillary tangles in subdivisions of the hippocampal
VIII: Neuroimaging-neuropathology correlates of temporal lobe cortex in normal ageing and Alzheimer’s disease. Neurosci Lett 1995;
changes in Alzheimer’s disease. Neurology 1995;45:178Y79 200:57Y60
105. Koepsell TD, Kurland BF, Harel O, et al. Education, cognitive 128. Bartus RT, Dean RL 3rd, Beer B, et al. The cholinergic hypothesis of
function, and severity of neuropathology in Alzheimer disease. geriatric memory dysfunction. Science 1982;217:408Y14
Neurology 2008;70:1732Y39 129. Davies P, Maloney AJ. Selective loss of central cholinergic neurons in
106. Silver MH, Newell K, Brady C, et al. Distinguishing between Alzheimer’s disease. Lancet 1976;2:1403
neurodegenerative disease and disease-free aging: Correlating neuro- 130. Mesulam M, Shaw P, Mash D, et al. Cholinergic nucleus basalis
psychological evaluations and neuropathological studies in centenar- tauopathy emerges early in the aging-MCI-AD continuum. Ann Neurol
ians. Psychosom Med 2002;64:493Y501 2004;55:815Y28
107. Braak H, Braak E, Ohm T, et al. Alzheimer’s disease: Mismatch 131. Rasool CG, Svendsen CN, Selkoe DJ. Neurofibrillary degeneration of
between amyloid plaques and neuritic plaques. Neurosci Lett 1989; cholinergic and noncholinergic neurons of the basal forebrain in
103:24Y28 Alzheimer’s disease. Ann Neurol 1986;20:482Y88
108. Nagy Z, Esiri MM, Jobst KA, et al. Relative roles of plaques and 132. Saper CB, German DC, White CL 3rd. Neuronal pathology in the
tangles in the dementia of Alzheimer’s disease: Correlations using three nucleus basalis and associated cell groups in senile dementia of the
sets of neuropathological criteria. Dementia 1995;6:21Y31 Alzheimer’s type: Possible role in cell loss. Neurology 1985;35:1089Y95
109. Thal DR, Rub U, Schultz C, et al. Sequence of Abeta-protein 133. Saper CB, Chelimsky TC. A cytoarchitectonic and histochemical study
deposition in the human medial temporal lobe. J Neuropathol Exp of nucleus basalis and associated cell groups in the normal human
Neurol 2000;59:733Y48 brain. Neuroscience 1984;13:1023Y37
110. Bennett DA, Schneider JA, Wilson RS, et al. Neurofibrillary tangles 134. Izquierdo I. Mechanism of action of scopolamine as an amnestic.
mediate the association of amyloid load with clinical Alzheimer disease Trends Pharmacol Sci 1989;10:175Y77
and level of cognitive function. Arch Neurol 2004;61:378Y84 135. Airaksinen MS, Paetau A, Paljarvi L, et al. Histamine neurons in
111. Holmes C, Boche D, Wilkinson D, et al. Long-term effects of Abeta42 human hypothalamus: Anatomy in normal and Alzheimer diseased
immunisation in Alzheimer’s disease: Follow-up of a randomised, brains. Neuroscience 1991;44:465Y81
placebo-controlled phase I trial. Lancet 2008;372:216Y23 136. Grudzien A, Shaw P, Weintraub S, et al. Locus coeruleus neuro-
112. Wisniewski HM, Barcikowska M, Kida E. Phagocytosis of beta/A4 fibrillary degeneration in aging, mild cognitive impairment and early
amyloid fibrils of the neuritic neocortical plaques. Acta Neuropathol Alzheimer’s disease. Neurobiol Aging 2007;28:327Y35
1991;81:588Y90 137. German DC, White CL 3rd, Sparkman DR. Alzheimer’s disease:
113. Meyer-Luehmann M, Spires-Jones TL, Prada C, et al. Rapid appear- Neurofibrillary tangles in nuclei that project to the cerebral cortex.
ance and local toxicity of amyloid-beta plaques in a mouse model of Neuroscience 1987;21:305Y12
Alzheimer’s disease. Nature 2008;451:720Y24 138. Rub U, Del Tredici K, Schultz C, et al. The evolution of Alzheimer’s
114. Benzing WC, Mufson EJ, Armstrong DM. Immunocytochemical disease-related cytoskeletal pathology in the human raphe nuclei.
distribution of peptidergic and cholinergic fibers in the human Neuropathol Appl Neurobiol 2000;26:553Y67
amygdala: Their depletion in Alzheimer’s disease and morphologic 139. Zweig RM, Ross CA, Hedreen JC, et al. The neuropathology of
alteration in non-demented elderly with numerous senile plaques. Brain aminergic nuclei in Alzheimer’s disease. Ann Neurol 1988;24:
Res 1993;625:125Y38 233Y42
115. Armstrong DM, Benzing WC, Evans J, et al. Substance P and so- 140. Ishino H, Otsuki S. Frequency of Alzheimer’s neurofibrillary tangles in
matostatin coexist within neuritic plaques: Implications for the patho- the basal ganglia and brain-stem in Alzheimer’s disease, senile dementia
genesis of Alzheimer’s disease. Neuroscience 1989;31:663Y71 and the aged. Folia Psychiatr Neurol Jpn 1975;29:279Y87
116. Wisniewski HM, Ghetti B, Terry RD. Neuritic (senile) plaques and 141. Torack RM, Morris JC. The association of ventral tegmental area
filamentous changes in aged rhesus monkeys. J Neuropathol Exp histopathology with adult dementia. Arch Neurol 1988;45:497Y501
Neurol 1973;32:566Y84 142. Ohtsubo K, Izumiyama N, Kuzuhara S, et al. Curly fibers are
117. Wisniewski HM, Vorbrodt AW, Moretz RC, et al. Pathogenesis of tau-positive strands in the pre- and post-synaptic neurites, consisting
neuritic (senile) and amyloid plaque formation. Exp Brain Res 1982 of paired helical filaments: Observations by the freeze-etch and replica
(suppl 5):3Y9 method. Acta Neuropathol 1990;81:111Y15

Ó 2008 American Association of Neuropathologists, Inc. 13

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.
Nelson et al J Neuropathol Exp Neurol  Volume 68, Number 1, January 2009

143. Braak H, Braak E, Grundke-Iqbal I, et al. Occurrence of neuropil of Alzheimer’s disease exceeds extracellular neurofibrillary tangle
threads in the senile human brain and in Alzheimer’s disease: A third formation. Acta Neuropathol 2002;103:370Y76
location of paired helical filaments outside of neurofibrillary tangles 147. Vogt BA, Vogt LJ, Vrana KE, et al. Multivariate analysis of
and neuritic plaques. Neurosci Lett 1986;65:351Y55 laminar patterns of neurodegeneration in posterior cingulate cortex in
144. Mitchell TW, Nissanov J, Han LY, et al. Novel method to quantify Alzheimer’s disease. Exp Neurol 1998;153:8Y22
neuropil threads in brains from elders with or without cognitive 148. Morsch R, Simon W, Coleman PD. Neurons may live for decades
impairment. J Histochem Cytochem 2000;48:1627Y38 with neurofibrillary tangles. J Neuropathol Exp Neurol 1999;58:
145. Markesbery WR, Wang HZ, Kowall NW, et al. Morphometric image 188Y97
analysis of neuropil threads in Alzheimer’s disease. Neurobiol Aging 149. Lovell MA, Robertson JD, Buchholz BA, et al. Use of bomb pulse
1993;14:303Y7 carbon-14 to age senile plaques and neurofibrillary tangles in
146. Kril JJ, Patel S, Harding AJ, et al. Neuron loss from the hippocampus Alzheimer’s disease. Neurobiol Aging 2002;23:179Y86

Downloaded from http://jnen.oxfordjournals.org/ by guest on May 22, 2016

14 Ó 2008 American Association of Neuropathologists, Inc.

Copyright @ 2008 by the American Association of Neuropathologists, Inc. Unauthorized reproduction of this article is prohibited.

You might also like