You are on page 1of 14

Progress in Neurobiology 97 (2012) 3851

Contents lists available at SciVerse ScienceDirect

Progress in Neurobiology
journal homepage: www.elsevier.com/locate/pneurobio

Dening Alzheimer as a common age-related neurodegenerative process not


inevitably leading to dementia
Isidro Ferrer *
Institute of Neuropathology, University Hospital Bellvitge, University of Barcelona, Idibell, Ciberned, Spain

A R T I C L E I N F O A B S T R A C T

Article history: Since the description by Alois Alzheimer, more than 50 years have passed during which senile dementia
Received 15 January 2012 and pre-senile dementia have been considered Alzheimer disease (AD) on the basis of their common
Received in revised form 10 March 2012 neuropathological and clinical manifestations. AD now covers pre-senile dementia, senile dementia,
Accepted 13 March 2012
mild cognitive impairment and pre-clinical AD, all of them within the context of AD-related pathology.
Available online 21 March 2012
However, there is still a gray area between normal aging with AD-related pathology and AD. Here it is
proposed that Alzheimer (or alzheimer) is an age-related neurodegenerative process distinguished from
Keywords:
normal aging by the presence of senile plaques and neurobrillary tangles. Alzheimer affects about 80%
Alzheimer
of individuals aged 65 years but dementia only occurs in a small percentage of individuals at this age;
Senile plaques
Neurobrillary tangles prevalence of dementia in Alzheimer increases to 25% in individuals aged 80 years. The concepts derived
Amyloid from the b-amyloid hypothesis support b-amyloid as a conductor in the pathogenesis of familial AD and
Tau as a prodding factor in sporadic AD. Moreover, seeding of b-amyloid and truncated tau explains
Mitochondria incorporation, enhancement and perpetuation of AD-related changes. Therefore, the earliest Alzheimer
Oxidative stress changes conned to selected regions are the rst grounds and the main risk factor for developing
Atherosclerosis dementia. The term Alzheimer embraces this assumption and likens its meaning to other degenerative
Biomarkers
biological processes, such as atherosclerosis, that may eventually progress to disease. In this context, the
rst stages of Alzheimer should be considered as primary targets of therapeutic intervention in order to
prevent progression to diseased states.
2012 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
2. Historical evolution of the concepts of AD and senile dementia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
3. Genetic and sporadic AD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
4. Neuropathological hallmarks and molecular pathology of tau b-amyloid in AD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
5. Multi-functional subcellular failure in AD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
6. Involvement of synapses, multiple neuronal types and systems in AD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
7. Categorization of changes in the nervous system attributable to aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
8. Discrimination of normal brain aging from AD-related pathology and AD. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
9. AD, mild cognitive impairment, pre-clinical AD and biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42

Abbreviations: AD, Alzheimers disease; ADAD, autosomal dominant Alzheimers disease; ApoE4, apolipoprotein allele e4; APP, amyloid precursor protein; BACE, b-secretase;
CERAD, Consortium to Establish a Registry of Alzheimers Disease; CSF, cerebrospinal uid; EOAD, early onset Alzheimers disease; FAD, Familial Alzheimers disease; LOAD,
late-onset Alzheimers disease; MAPT, gene encoding microtubule associated protein tau; NINCDS-ADRDA, National Institute of Neurological and Communicative Disorders
Association; NFTs, neurobrillary tangles; PDGF, platelet-derived growth factor; PET, positron emission tomography; PICALM, phosphatidylinositol binding clathrin
assembly protein; PSEN1, presenilin-1; PSEN2, presenilin-2; PSN1, gene encoding presenilin-1; PSN2, gene encoding presenilin-2; sAD, sporadic Alzheimers disease; sMRI,
structural magnetic resonance; SPs, senile plaques.
* Correspondence address: Institute of Neuropathology, Service of Pathology, University Hospital Bellvitge, carrer Feixa LLarga sn, 08907 Hospitalet de LLobregat, Spain.
Tel.: +34 93 2607452.
E-mail address: 8082ifa@gmail.com.

0301-0082/$ see front matter 2012 Elsevier Ltd. All rights reserved.
doi:10.1016/j.pneurobio.2012.03.005
I. Ferrer / Progress in Neurobiology 97 (2012) 3851 39

10. The b-amyloid cascade in the pathogenesis of FAD and sAD, compelling or prodding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
11. Seeding b-amyloid and abnormal tau . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
12. Concluding comments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45

1. Introduction on chromosome 21, the presenilin-1 (PSEN1) gene on chromosome


14, and the presenilin-2 (PSEN2) gene on chromosome 1 (Levy
Alzheimer disease (AD) is a human age-related biological et al., 1990; van Broeckhoven et al., 1990; Chartier-Harlin et al.,
process which causes progressive degeneration of the brain and 1991; Sherrington et al., 1995; Hardy, 1997; Tanzi, 1999; Bateman
which is characterized clinically by cognitive impairment and et al., 2011).
dementia, and neuropathologically by the variable combination of Sporadic AD (sAD) is favored by: (a) individual or combined low-
two hallmarks, neurobrillary tangles (NFTs) and senile plaques penetrating genetic factors linked to apolipoprotein allele e4
(SPs) (Duyckaerts and Dickson, 2003; Lowe et al., 2008). The (ApoE4), a2-macroglobulin, low density lipoprotein protein recep-
meaning of the term AD, rst described as a pre-senile dementia, tor 1 (both in chromosome 12), very low density lipoprotein R
has changed over the years, now covering pre-senile dementia, (chromosome 9), interleukin 1a (chromosome 2), clusterin (chro-
senile dementia, mild cognitive impairment and pre-clinical AD, all mosome 8), phosphatidylinositol binding clathrin assembly protein
of them within the context of AD-related pathology. However, (PICALM, chromosome 11) and complement component (3b/4b)
there is still heated controversy regarding normal aging with AD- receptor 1 (CR1, chromosome 1), among others (Corder et al., 1993;
related pathology and AD. Farrer et al., 1997; Marzolo and Bu, 2009; Bu, 2009; Harold et al.,
2009; Lambert et al., 2009; Seshadri et al., 2010; Jun et al., 2010;
2. Historical evolution of the concepts of AD and senile Hollingworth et al., 2011); (b) environmental factors such as
dementia repeated brain trauma (i.e. dementia pugilistica) (Constanza et al.,
2011); and (c) combined metabolic systemic factors such as
The concept of AD has changed over the years mainly due to atherosclerosis and cardiovascular disease, elevated serum homo-
social aspects as well as to scientic developments. The average cysteine and hypercholesterolemia at midlife, and family history of
lifespan of human beings in Western countries rarely exceeded the dementia (Qiu et al., 2005; Kivipelto and Solomon, 2006; Anstey
sixth decade at the end of the nineteenth century and during the et al., 2008; Solomon et al., 2009; Reitz et al., 2011; Ballard et al.,
rst half of the twentieth. Individuals over 65 were considered old. 2011). Interestingly, genes involved in sAD are not linked to b-
Mental disorders before this age were classied with terms such as amyloid but rather to cholesterol and lipid metabolism, the immune
madness, neurasthenia, presbyophrenia and hysteria, and the system, and synaptic membranes (Jones et al., 2010; Morgan, 2011).
principal cause of dementia was progressive general paresis due to Moreover, epigenetic factors (differences in DNA methylation) may
tertiary syphilis. Mental impairment and dementia in older mediate AD in monozygotic twins (Mastroeni et al., 2009). In
persons was considered a phenomenon of senile dementia closely consequence, AD is a multifactorial, genetically complex and
associated with the process of aging and related to vascular heterogeneous disorder modulated by variegated non-genetic
disease. The study of the brains of many of these old people showed factors (Bertram and Tanzi, 2003; Iqbal and Grundke-Iqbal, 2010).
the presence of SPs and the term pure senile dementia was Approximately 16% of cases are early-onset (under 65 years of
therefore introduced (Klippel and Lhermitte, 1905). The descrip- age: EOAD) and nearly 70% of these are associated with mutations
tion by Alois Alzheimer of a relatively young woman with in APP, PSN1 or PSN2 these mutations being the major cause of
dementia and numerous SPs and new lesions in her brain familial AD (FAD) (Bird, 2008). In contrast, late-onset AD (after 65
described as NFTs, as shown with silver stains, was the starting years of age: LOAD) is largely sporadic although 40% of LOAD
point of pre-senile dementia of Alzheimers type or AD (Alzheimer, patients have at least one ApoE4 allele (Bu, 2009). These gures
1907, 19091910). Although neuropathological studies promptly represent AD cases with clinical manifestations of cognitive
revealed that (pre-senile) AD and pure senile dementia shared the impairment and dementia.
same brain lesions, NFTs and SPs, the distinction between the two
conditions was based on the social assumption of what was 4. Neuropathological hallmarks and molecular pathology of
considered, at that time, to be old. Even in the 1960s, pre-senile tau b-amyloid in AD
dementia or AD and pure senile dementia were considered as
separate entities (Delay and Brion, 1962). Not until the end of that The rst electron microscopic studies revealed the structure of
decade was a relationship established between NFTs, SPs and the b-amyloid deposition and the presence of surrounding dystrophic
degree of dementia (Blessed et al., 1968). neurites lled with altered mitochondria, residual bodies and
Several years of debate and vast amounts of clinical, neuro- abnormal laments in SPs, together with disorganization of the
pathological and genetic evidence were needed to bring about the normal cytoskeleton and accumulation of paired helical laments
abandonment of this dualism in favor of the view that pre-senile such as the subcellular substrates of NFTs (Kidd, 1964; Luse and
dementia of Alzheimers type and senile dementia with neuro- Smith, 1964; Terry et al., 1964; Nakano and Hirano, 1999).
pathological changes consisting of accumulations of NFTs and SPs Biochemical characterizations of the main alterations of SPs and
are the same disorder appearing at different ages. NFTs appeared in the 1980s, rmly demonstrating b-amyloid as
the main component of SPs and hyper-phosphorylated tau as the
3. Genetic and sporadic AD main component of NFTs (Glenner and Wong, 1984a,b; Masters
et al., 1985; Grundke-Iqbal et al., 1986; Iqbal et al., 1986; Kang
Genetically determined AD is triggered by dominant genetic et al., 1987; Goedert et al., 1988, 1989; Delacourte et al., 1999).
factors as in Down syndrome (chromosome 21 trisomy) and in Hyper-phosphorylated 3R and 4R tau isoforms, resulting from
familial cases with autosomal dominant inheritance (ADAD) the imbalance between hyper-activation of kinases and reduced
caused by mutations in the amyloid precursor protein (APP) gene activation of phosphatases, together with tau truncation resulting
40 I. Ferrer / Progress in Neurobiology 97 (2012) 3851

from the activity of different proteases, and interactions of tau with 2008; Sultana and Buttereld, 2009, 2010; Terni et al., 2010).
sulphated glycosaminoglycans, are the main alteration in NFTs Neuronal exhaustion is the term used for the delicate metabolic
(Buee and Delacourte, 2001; Ferrer et al., 2005; Goedert et al., condition resulting from the convergence of detrimental energy
2006; Avila, 2006; Wang et al., 2007, 2010; Hanger et al., 2009; production, increased oxidative damage, impaired glycolysis and
Kovacech and Novak, 2010). Modications of different subunits of increased energy demands in AD (Ferrer, 2009). It is worth
the ubiquitinproteasome system and expression of mutant emphasizing that altered mitochondrial function appears early in
ubiquitin are additional contributory factors to impaired NFT time in AD (Terni et al., 2010), and that it precedes Alzheimer
degradation (van Leeuwen et al., 1998; Keller et al., 2000; Cecarini pathology in mouse models of AD (Yao et al., 2009; Du et al., 2010).
et al., 2007; Oddo, 2008; McMillan et al., 2011). In addition to energetic failure, other molecular pathways fail in
SPs are composed of variable species of b-amyloid forming: (a) AD. Lipid composition of cell membranes and most particularly of
diffuse plaques accompanied with punctate alterations of neuronal lipid rafts is modied when compared with aged-matched controls
processes, and (b) neuritic plaques with a central core of b-amyloid (Pamplona et al., 2005; Martn et al., 2010; Chan et al., 2012);
and a peripheral crown of dystrophic neurites lled with hyper- alteration of lipid raft composition implies increased viscosity and
phosphorylated tau, altered mitochondria and other cellular greater difculty in molecular trafc (Martn et al., 2010).
debris. In addition, b-amyloid angiopathgy (i.e. b-amyloid Endoplasmic reticulum stress and impairment of responses to
deposition in the blood vessel walls) is a common accompanying protein misfolding has also been demonstrated in AD (Hoozemans
alteration in AD (Goedert, 1996; Duyckaerts and Dickson, 2003; et al., 2009; Salminen et al., 2009; Lee et al., 2010; Kaneko et al.,
Masters and Beyreuther, 2003; Lowe et al., 2008; Shepherd et al., 2010). Mechanisms geared to getting rid of altered proteins and
2009; Markesbery, 2010; Serrano-Pozo et al., 2011). organelles, including autophagy, are impaired in AD (Cataldo et al.,
b-amyloid deposits result from the proteolytic cleavage of the 1991; Yu et al., 2005; Nixon et al., 2005; Barrachina et al., 2006;
APP precursor by b-secretases (BACEs) and g-secretases which Cecarini et al., 2007). Impaired removal of altered proteins is
consist of a complex in which presenilins are one of their already manifested in the rst NFT, as hyper-phospohorylated,
components. b-amyloid is composed of several cleaved products, abnormally conformed, and truncated, ubiquitinated tau species
mainly b-amyloid140 and b-amyloid142, which are the principal are resistant to degradation by the ubiquitinproteasome system.
elements of brillar b-amyloid in SPs (Hardy, 2006). Additionally,
nepresylin and insulin-degrading enzyme do not withstand 6. Involvement of synapses, multiple neuronal types and
degradation of b-amyloid; and transport of b-amyloid bound to systems in AD
low-density protein receptor and receptor for advanced glycation
end-products required to pass across the blood brain barrier is Synapses are damaged in AD, resulting in a major cause of
partially blocked in AD, thus favoring accumulation of soluble and cognitive impairment (Gonatas et al., 1967; Terry et al., 1991;
insoluble b-amyloid species in brain (Deane and Zlokovic, 2007; Selkoe, 2002; Arendt, 2003; Bloss et al., 2011). The mechanisms of
Jaeger and Pietrzik, 2008; Weller et al., 2008; Tseng et al., 2008; Utter synaptic alteration are not fully understood but several observa-
et al., 2008; Schmidt et al., 2009; Alvira-Botero and Carro, 2010; tions have provided important pieces of information. On the one
Hawkes et al., 2011; Chalbot et al., 2011). hand, b-amyloid proto-brils are synaptotoxic (ONuallain et al.,
However, the links between b-amyloid and tau hyper- 2010), b-amyloid impairs anterograde transport of mitochondria
phosphorylation are not completely understood (Small and Duff, and degenerates synapses in vitro (Calkins and Reddy, 2011), and
2008; Oddo et al., 2008; Takashima, 2009). b-amyloid immunoreactivity has been detected in mitochondria in
In addition to these basic mechanisms involved in NFTs and b- AD (Lustbader et al., 2004; Caspersen et al., 2005; Gouras et al.,
amyloid deposition, mounting evidence points toward soluble b- 2010). This is further supported by functional studies showing that
amyloid oligomers rather than brillar b-amyloid in plaques as b-amyloid deposition is associated with default activity (Buckner
being causative of neurodegeneration (McLean et al., 1999; Glabe et al., 2005; Sperling et al., 2009). On the other hand, hyper-
and Kayed, 2006; Deshpande et al., 2006; Shankar et al., 2008; van phosphorylated tau and hyper-phosphorylated a-synuclein accu-
Helmond et al., 2010; Ono and Yamada, 2011). Similarly, truncated mulate at the synapses (Muntane et al., 2008) involving more
tau appears to be toxic to neurons (Fasulo et al., 2000; Chung et al., sophisticated alterations in combination with b-amyloid and
2001; Gomez-Ramos et al., 2006; Filipcik et al., 2009; Lasagna- mitochondrial deviations connected with synaptic dysfunction.
Reeves et al., 2010; Zilkova et al., 2011). Several cell types are vulnerable to AD including nuclei of the
brain stem such as raphe nuclei, reticular formation, locus
5. Multi-functional subcellular failure in AD ceruleus, substantia nigra; amygdala; nucleus basalis of Meynert;
striatum; thalamus; hypothalamus; and claustrum, in addition to
Mitochondria are abnormal and dysfunctional in AD (Kidd, 1964; the selective regional and laminar vulnerability of the cerebral
Luse and Smith, 1964; Terry et al., 1964; Kish et al., 1992; Mutisya cortex. Different neuronal types are affected in the various regions
et al., 1994; Hirai et al., 2001; Tsuji et al., 2005; Hauptmann et al., as revealed by several neurotransmitter, neuromodulator, neuro-
2006; Kim et al., 2000; Perez-Gracia et al., 2008; Moreira et al., 2007; peptide, and calcium-binding protein markers. Even the cerebellar
Coskun et al., 2011). This may result in increased oxidative stress and cortex is compromised at advanced stages of the disease. Neuron
damage to different molecules including nucleic acids, proteins and loss and cerebral atrophy are the aftermath of the degenerative
lipids (Pamplona and Barja, 2007; Starkov, 2008). process (Hof, 2001; Vogt et al., 2001; Duyckaerts and Dickson,
b-amyloid deposition is also causative of oxidative stress (Boyd- 2003; Lowe et al., 2008). In short, AD may be categorized as a
Kimball et al., 2005a,b; De Felice et al., 2007; Wan et al., 2011), and b- multi-systemic degenerative process.
amyloid inhibits integrated mitochondrial respiration and key Not only neurons, but also astrocytes, oligodendroglia, microglia,
enzyme activities (Casley et al., 2002 Crouch et al., 2005). Resulting blood vessels and choroid plexus are involved in disease progression.
from the cumulative effect of diverse events, increased oxidative
stress is key in the pathogenesis of AD and is the principal cause of 7. Categorization of changes in the nervous system attributable
damage to quite a variety of substrates including mitochondrial and to aging
energy metabolism-related proteins (Buttereld et al., 2002;
Castegna et al., 2002a,b; Choi et al., 2004, 2005; Pamplona et al., Common characteristics of the central nervous system with
2005; Korolainen et al., 2006; Sultana et al., 2006, 2009; Reed et al., aging in distinct mammalian species, including humans, involve
I. Ferrer / Progress in Neurobiology 97 (2012) 3851 41

the progressive accumulation of lipofuscin granules in the (Becker et al., 2011). However, the implication of these changes,
cytoplasm of neurons and glial cells; mitochondrial alterations and the frontier between normal aging and abnormal aging
(covering alterations in size and deletions in mitochondrial DNA); (mostly in the case of AD) have been and still are the subject of an
increased oxidative stress and oxidative and nitrosative damage of on-going debate that has lasted decades.
lipids, proteins, DNA and RNA; loss of neurons in specic regions of Another important point is the increased prevalence and
the nervous system; reduced dendritic arbours and reduced post- incidence of other pathologies of the human central nervous
synaptic sites particularly in the cerebral cortex; and axonal system with aging, most of them potentially causative of cognitive
degeneration with axon loss (Morrison and Hof, 1997; Peters, impairment. The old term mixed dementia, indicating a
1999; Sandell and Peters, 2001, 2003; Hof and Morrison, 2004; combination of AD and vascular pathology leading to dementia
Peters et al., 2008, 2010; Kabaso et al., 2009; Bowley et al., 2010; (Delay and Brion, 1962), is still valid (Vinters et al., 2000; Grinberg
Dumitriu et al., 2010; Pannesse, 2011). Some of these alterations and Thal, 2010), although the attribution of clinical manifestations
occur in parallel with cognitive decline in rhesus monkey (Moss to each of the two pathologies is a matter for frequent discussion.
et al., 1999). In addition, other degenerative diseases are common in old age,
In addition, b-amyloid deposits come up in old individuals in such as Lewy body diseases, argyrophilic grain disease, mesial
several (but not all) species including dolphins and other cetacean, sclerosis and pathology of TAR DNA binding protein (TDP)-43 (Ince
non-human primates such as Microcebus murinus, Macaca mulatta et al., 1998; Ince, 2001; Uchikado et al., 2006; Amador-Ortiz et al.,
and Macaca arctioides, Chlorocebus aethiops, dogs and bears, among 2007; Ferrer et al., 2008; Zarow et al., 2008; Wilson et al., 2011).
others (Head, 2011; Mutsuga et al., 2012; Finch and Austad, 2012; Studies of large neuropathologically examined series in general
Languille et al., 2012). Phospho-tau deposits surrounding b- hospitals have demonstrated that combined neurodegenerative/
amyloid plaques are common but NFTs are rare in species other vascular pathologies, rather than single entities, are prevalent in
than humans (Price et al., 1991; Morelli et al., 1996; Toledano et al., old age (Kovacs et al., 2008; White, 2009). A fascinating aspect is
2011). Yet protein tau accumulates in the cerebral cortex, but that these combinations are not the result of mere accumulation
lesser in the hippocampus, in a few vulnerable aged mouse lemur, related to age, but rather they have a promoting or facilitating
M. murinus (Giannakopoulos et al., 1997). Importantly, transcrip- effect upon each other. Although it would be worth writing a bit
tome differences can be distinguished during brain aging from AD- more on this subject, a detailed description of this scenario is
like pathology in this species (Abdel Rassoul et al., 2010). These beyond the scope of the present review.
observations indicate that AD pathology is associated with aging
but AD pathology is not brain aging in M. murinus as only a small 8. Discrimination of normal brain aging from AD-related
percentage of old animals have AD-like pathology. Intracellular pathology and AD
phospho-tau lamentous inclusions occur in nerve cells, astrocytes
and oligodendrocytes of aged baboons (Schultz et al., 2000b) and Several classications are based on the assumption that only a
tau pathology increases with age in these primates (Schultz et al., certain number of plaques at a particular age and determinate
2000a). Whether these changes in non-human primates are related regions indicate AD, whereas the same number of lesions in older
to normal aging or to AD is difcult to ascertain. Old C. aethiops have individuals does not. Khachaturian and CERAD (Consortium to
abundant AD-related pathology in their brains although these Establish a Registry of Alzheimers Disease) codications are based
changes are not clearly accompanied by impaired cognitive on the quantication of plaques by using silver or amyloid stains
function, at least as may be determined by our current study (Khachaturian) or dystrophic neurites using Bielchowsky stains
methods, thus suggesting that certain non-human primates may (CERAD) in selected cortical regions (Kachaturian, 1985; Mirra
have extensive AD-related pathology without dementia of et al., 1991, 1994). The number of lesions depending on the age of
Alzheimer type. However, cognitive decline in old dogs correlates the patient serves to make the diagnosis of AD. Beyond the
with increased b-amyloid deposition and tau phosphorylation at difculty in staining and interpreting Bielchowsky stains (Alafuzoff
Ser396 in the parietal cortex and dorsal part of the hippocampus et al., 2008), Khachaturian and CERAD classications are not
(Yu et al., 2011). In short, although AD-related pathology occurs in instrumental approaches to classifying AD but rather tenets to
some aged mammals, it is far from clear the impact of such lesions discriminate AD from normal aging with SPs. Moreover, these
in cognitive performances and behavior. Moreover, lack of a categorizations are sustained on a problematic basis as the number
denitive categorization of dementia in non-human species of SPs barely correlates with neurological decits whereas
ignores the possibility of AD and has prejudged the presence of dementia is more closely associated with NFTs, synaptic loss
SPs and phospho-tau deposits in neurons in these species as and nerve cell loss in the cerebral cortex (Bierer et al., 1995;
normal aging. Masliah and Salmon, 1999; Hof et al., 1999). These classications
Neuropsychological studies have demonstrated that loss of have for years permeated the notion that AD is a different
recent memory, explicit and implicit secondary memories, condition from normal aging with SPs.
language, and sustained attention occurs with age in humans; Other studies have stressed that, albeit a heterogeneous pattern
these changes are currently interpreted as a result of normal aging of SPs, NFTs in non-demented old people are consistently present
(Coubard et al., 2011). Clinical studies have assumed that cognitive among individuals in the entorhinal cortex, CA1 region of the
decline does not occur before the age of 60 (Hedden and Gabrieli, hippocampus, and, less often, the temporal and frontal cortex
2004). However, recent work carried out on the Whitehall II cohort (Braak and Braak, 1991; Bussiere and Hof, 2001). These changes in
of British civil servants of both sexes followed during 10 years has individuals with no concurrent major cognitive disability have
demonstrated a 3.6% cognitive decline in men aged 4549 and been largely categorized as being related to normal aging (Hof
9.6% in those aged 6570, whereas the corresponding decline was et al., 1999).
3.6% in women aged 4549 and 7.4% in women aged 6570, Regardless of the clinical threshold between normal aging and
probably increasing to 11.4% when considering cross sectional AD, it is clear that the type of lesionsNFTs and SPsand the
effects (Singh-Manoux et al., 2011). regional vulnerability are similar in the group of individuals who
Some papers have highlighted a link between normal aging and present SPs and NFTs with aging and cases with dementia with AD-
AD-related changes particularly in relation with b-amyloid related pathology, apart from the order of magnitude and extent of
deposition in temporal cortex (Mormino et al., 2009; Rentz such lesions, which are markedly greater in cases with dementia
et al., 2010) and with cortical thinning in clinically normal elderly (Arriagada et al., 1992; Morris et al., 1996; Hof et al., 1999).
42 I. Ferrer / Progress in Neurobiology 97 (2012) 3851

A signicant advance was brought about after the study of post- AD-related changes does not always match with severe neurological
mortem aged brains by using silver methods (Braak and Braak, 1991, decits (Erten-Lyons et al., 2009; Nelson et al., 2009; Abner et al.,
1999). Systematic anatomical study of cases with AD-related 2011; SantaCruz et al., 2011). Further studies are also needed to
pathology has prompted a staging classication of sAD. Stages I elucidate the neuropathological and molecular correlates of
and II are dened by the presence of NFTs in the entorhinal cortex different types of dementia in AD cases (Murray et al., 2011).
(stage I) and progression to the transentorhinal cortex and mild The present distinction between AD (once accepted as a unique
involvement of the CA1 region (stage II). Limbic stages III and IV disease) and normal aging accompanied by variable numbers of
implicate the presence of NFTs in the upper and inner layers of the SPs and NFTs (AD-related pathology) is circular and dependent on
entorhinal cortex, transentorhinal cortex, CA1 region of the the arbitrary number and extent of NFTs and SPs based on
hippocampus, subiculum, anterodorsal thalamic nucleus, amygdala, statistical measures of what is considered normal mental
magnocellular nuclei of the basal forebrain (including Meynert performance according to typical forms and frequencies of a
nucleus), tubero-mammillary nucleus (stage III), plus associated socially established behavior.
areas of the temporal cortex, striatal neurons, raphe nucleus and Recent proposals aimed at facilitating the neuropathological
locus ceruleus (stage IV). Neocortical stages V and VI require, in assessment of AD recommend the selection of certain areas and
addition, NFTs in cortical association areas, claustrum, reticular staining of sections to cover AD-related changes and concomitant
nucleus of the thalamus and substantia nigra (stage V), plus primary pathologies such as vascular disease, Lewy body diseases, TDP-43
sensory areas (stage VI). Similarly, SP burden and distribution allows inclusions, and mesial sclerosis as well (Montine et al., 2012). Yet
the classication of SP pathology into stages A (basal neocortex the ABC score suggested for AD neuropathological classication
including orbitary and temporal cortices), B (isocortical involvement has added little of value to previous instrumental classications
covering associated cortices) and C (involving in addition the intended to substantially increase our understanding of AD.
primary cortical areas). Further renements have been made with
immunohistochemistry, thus permitting a harmonization of stain- 9. AD, mild cognitive impairment, pre-clinical AD and
ing in different laboratories (Braak et al., 2006a,b). Neuropathologi- biomarkers
cal inquiries have also given rise to more sophisticated
classications of b-amyloid pathology in relation to SPs and b- The National Institute of Neurological and Communicative
amyloid angiopathy (Thal et al., 2002). Phases of b-amyloid Disorders Association (NINCDS-ADRDA) criteria of 1984 (McKhann
deposition are classied as phase I: neocortex; phase 2: plus et al., 1984) were revised in 2007 by the International Working
allocortical brain regions; phase 3: plus diencephalic nuclei, Group for New Research Criteria for the Diagnosis of Alzheimers
striatum, cholinergic nuclei of the basal forebrain; phase 4: plus disease (Dubois et al., 2007). Subsequently, improvements
nuclei of the brain stem; phase 5: plus cerebellar deposition. appeared as a result of annual meetings and workshops. In
Stages of AD-related pathology have supported a hypothesis of contrast to NINCDS-ADRDA criteria which determined the
AD progression based on the putative sequence of NFT pathology diagnosis of AD as probable, possible and denitive, this last
from entorhinal and transentorhinal cortex, to the hippocampus category requiring neuropathological examination of the brain, the
and limbic brain, and eventually to the entire cerebral cortex. new criteria are mostly based on clinical, laboratory, and
Similarly, amyloid plaques are suggested as spreading from the neuroimaging methods (Dubois et al., 2010).
orbitary and temporal cortices to the entire brain, and nally to the Currently, the term biomarker is used to designate variegated
cerebellum. These seminal observations are the primordium of the signs derived from clinical probes, biochemical data and neurora-
concept that AD develops from the rst tangle or amyloid plaque in diological techniques, formerly considered as complementary or
particular regions of the cerebral cortex (Ohm et al., 1995). ancillary examinations to characterize a particular disease. Several
No similar studies have been carried out in individuals carrying biomarkers are currently available for the clinical diagnosis of
APP, PSN1 and PSN2 mutations, and we therefore have very little dementia of Alzheimer type covering skill decrement and
information with which to formulate a tenable staging of AD- degraded responses in several neuropsychological tests, reduced
related pathology in ADAD (Klunk et al., 2007). However, AD- levels of b-amyloid and increased ratio of phospho-tau/tau in the
related pathology in Down syndrome is rst manifested with CSF, increasing atrophy of certain cerebral regions such as the
widespread distribution of b-amyloid deposits, followed by NFTs hippocampus and amygdala revealed by structural magnetic
(Mann and Esiri, 1989; Leverenz and Raskind, 1998). In the same resonance imaging (sMRI) or computerized tomography, and
line, b-amyloid plaques precede the appearance of NFTs in some increased positron emission tomography (PET) signals of abnor-
transgenic mice bearing mutations in APP-related proteins and mally accumulated proteins in the brain such as b-amyloid and
mutations in MAPT, the gene encoding protein tau (Oldo et al., tau, as well as reduced glucose consumption (Jack et al., 2008;
2003; Perez et al., 2005; Ribe et al., 2005). Although limited, Morris et al., 2009; Mueller et al., 2010; Vemuri et al., 2010; Dubois
available data suggest that chronology of SPs and NFTs may differ et al., 2010; Jack et al., 2011a,b).
in sAD and, at least some genetic AD and certain AD-related Unveiling the ordered appearance of the different biomarkers,
models. Yet bigenic mice resulting from the crossing of PDAPP combining the appropriate use of several known biomarkers, and
transgenic mice carrying the APP V717F mutation driven by the discovering and applying new biomarkers may help at identifying
PDGF promoter with PS19 transgenic mice harbouring the P301S early stages of AD even before the appearance of clinical symptoms
mutation in MAPT routed by the prion protein promoter rst (Jack et al., 2010; Hu et al., 2010).
manifest tau pathology following a pattern similar to that seen in Rened recommendations to diagnose dementia and mild
single PS19 transgenic mice, reminiscent of Braak staging in cognitive impairment due to AD and pre-clinical stages of AD have
humans (Hurtado et al., 2010). recently been the objective of several guidelines published under
Post-mortem neuropathological studies of neurologically exam- the patronage of the National Institute on AgingAlzheimers
ined cases have shown that manifestations of neurological Association (Jack et al., 2011a,b; Albert et al., 2011; McKhann et al.,
impairment as early memory decits and mild cognitive im- 2011; Sperling et al., 2011). Identication of pre-clinical stages of
pairment occur at stages III and IV of Braak, whereas dementia does AD is relevant, although there is no full agreement among different
not appear until stage V and, more precisely, stage VI, at the time proposals. Staging categories of pre-clinical AD proposed by the
when the brain is lled with NFTs and SPs (Braak et al., 2006a,b; Price National Institute on Aging and the Alzheimers Association
et al., 2009; Markesbery, 2010). However, the presence of abundant Workgroup cover stage 1, described as asymptomatic cerebral
I. Ferrer / Progress in Neurobiology 97 (2012) 3851 43

amyloidosis (altered b-amyloid by PET or in the CSF); stage 2, has been estimated after the sequential study of a series of cases of
characterized as asymptomatic amyloidosis plus markers of AD-related pathology; it takes about 14 years from stage II to stage
neurodegeneration (as revealed by sMRI, 18-uorodeoxyglucose III, 13 years from stages III to IV but 5 years from stages IV to V
PET and altered tau/phospho-tau ratio in the CSF); and stage 3, (Ohm et al., 1995). This means that the biological process starts at
combining markers of amyloidosis, markers of neurodegeneration middle age, develops slowly during the rst stages and then
plus subtle cognitive and behavioral decline (Sperling et al., 2011). progresses more rapidly at advanced stages. Individual differences
The new lexicon proposed by the International Working Group in the progression and severity of the process are not commensu-
for New Research Criteria for the Diagnosis of Alzheimers disease rate to maintain the pervasive opinion that the origin of AD-related
incorporates AD (typical and atypical); pre-dementia stage of AD pathology at stages I, II and III differs from the origin of AD-related
(prodromal AD); and pre-clinical states of AD, grouping asymp- pathology stages IV, V and VI.
tomatic at-risk state for AD (similar to pre-clinical stage 2 of the Moreover, it has been claimed that the multifaceted nature of
previous proposal) and pre-symptomatic AD (carriers of mono- the altered neuronal function and neuronal connections in mild
genic forms of ADAD) (Dubois et al., 2010). cognitive impairment due to AD suggests that there is no single
These clinical proposals are innovative tools that clearly event which precipitates this prodromal stage of AD (Mufson et al.,
identify AD as a continuum of alterations that may lead from 2012).
mild or moderate cognitive impairment as a prodromal stage to
dementia of Alzheimer type. Moreover, these criteria also advance 10. The b-amyloid cascade in the pathogenesis of FAD and sAD,
the possibility of considering AD even in the absence of apparent compelling or prodding
clinical symptoms but in the presence of positive biomarkers. In
short, the focus of biomarker research at present is not only the The discovery that FAD is linked to mutations in APP and in other
identication of AD the disease but also the identication of genes encoding proteins involved in the cleavage of APP and the
individuals at risk of suffering dementia of Alzheimer type. production of b-amyloid, and to duplications in the genetic loading
Unfortunately, available biomarkers are still too uneven and not of APP (St George-Hyslop et al., 1987; Hardy, 1997; Tanzi, 1999;
precise enough to detect subtle alterations. Available biomarkers, Sleegers et al., 2006; Cabrejo et al., 2006; Brouwers et al., 2006;
other than genetic tests related to ADAD, are capable of visualizing Bateman et al., 2011), prompted the hypothesis of the b-amyloid
large accumulations of certain altered proteins such as b-amyloid, cascade as the origin of AD (Hardy and Higgins, 1992; Hardy and
focal atrophy involving severe loss of neurons and connections, Selkoe, 2002, Tanzi and Bertram, 2005; Hardy, 2009). This was
and disturbed proportions of altered proteins discharged in the supported by the fact that transgenic mice expressing mutations in
CSF. Yet altered composition of proteins in CSF follows massive APP present b-amyloid plaques with hyper-phosphorylated tau
neuronal damage as for tau or retained in the brain as a deposition in surrounding dystrophic neurites but not accompanied
consequence of severe impairment of b-amyloid clearance in by NFTs in their brains (Games et al., 1995; Hsiao et al., 1996;
the blood vessel wall and CSF barriers. The available biomarkers Sturchler-Pierrat et al., 1997; Borchelt et al., 1997; Puig et al., 2004;
unfortunately do not detect the process at early stages in McGowan et al., 2006; Gotz and Ilttner, 2008; Woodruff-Pak, 2008;
neuropathological terms. Aso et al., 2011). In contrast, individuals with mutations in MAPT
The application of biomarkers to the diagnosis of AD (excepting develop another type of dementia accompanied by neuronal and
gene study in ADAD) is, at present, a procedure giving witness to glial hyper-phosphorylated 4R tau inclusions but not accompanied
the pace of progression of the degenerative process rather than a by SPs (Munoz and Ferrer, 2008; Dickson et al., 2011). Moreover,
system allowing the detection of the rst stages of such process in a transgenic mice bearing tau mutations do not reproduce the
particular individual. Moreover, the ordering pattern or sequence presence of b-amyloid plaques but accumulate hyper-phosphory-
of positive biomarkers does not reect the real sequence of events lated tau in neurons (Zhang et al., 2004; Gotz and Ilttner, 2008).
in sAD as revealed in post-mortem neuropathological studies. The Therefore, mutations in APP-related genes give rise to incomplete
rst biomarker of AD identies accumulation of b-amyloid plaques AD lacking NFTs while mutations in MAPT give rise to a tauopathy.
and abnormal clearance of b-amyloid in the CSF, and this is Generation of mice bearing both SPs and NFTs is only accomplished
followed by altered ratio of tau/phospho-tau in the CSF (Dubois following the co-expression of mutant APP together with mutant or
et al., 2010; Jack et al., 2010; Sperling et al., 2011; Ossenkoppele altered tau (Lewis et al., 2001; Oldo et al., 2003; Perez et al., 2005;
et al., 2011). However, the rst pathological changes in cases with Ribe et al., 2005; Hurtado et al., 2010).
sAD-related pathology are characterized by NFTs while b-amyloid Although valid in FAD, the strict interpretation of the b-amyloid
plaques appear later. The hierarchical ordering of current cascade hypothesis does not completely t with the morphological
biomarkers further reinforces the misconception that plaque neuropathology of early stages in sAD. AD-related pathology in
formation precedes NFT formation in sAD. sAD is rst manifested in the form of NFTs whereas it is important
The major criticism of biomarker research is that present to recall that no SPs are normally encountered at the rst stages of
clinical approaches bias the recognition of AD as a biological sAD-related pathology (Braak and Braak, 1991, 1997, 1999; Braak
process in which the majority of persons will not develop an illness et al., 2011; Schonheit et al., 2004). Moreover, the distribution of
in the sense that this process may have no major impact on the SPs at early stages of AD-related pathology does not t with the
current life of the individual. localization of NFTs (Braak et al., 2011).
AD-related pathology, at least limited to the entorrhinal and Whether b-amyloid oligomers are present years before the
transentorrhinal cortices (stages III), is found in about 7080% of appearance of tau pathology in sAD and trigger the rst alterations
neuropathologically assessed post-mortem individuals aged 65 in tau is not known at present. This does not rule out the possibility
years belonging to a non-biased general population and dying as a that b-amyloid might accelerate tau pathology in different animal
result of various non-neurologically related conditions. These models, and in sAD. Indeed, NFT pathology is enhanced in
values increase to about 90% in individuals aged 80 years but then transgenic mice expressing mutant tau and APP (Lewis et al.,
remain steady in centenarians (Braak and Braak, 1997; Braak et al., 2001). Similarly, tau pathology is accelerated in bigenic mice
2011). It is worth recalling that the prevalence of dementia in AD is crossing PDAPP and PS19, but tau pathology does not alter b-
approximately 1 per 100 persons between 65 and 69 years of age, amyloid plaques when comparing bigenic mice with single
and between 25 and 50 per 100 individuals over the age of 85. The transgenic counterparts (Hurtado et al., 2010). These results are
speed of progression of AD-related pathology in relation with NFTs in line with studies showing that b-amyloid oligomers can induce
44 I. Ferrer / Progress in Neurobiology 97 (2012) 3851

tau phosphorylation in a mouse model (Tomiyama et al., 2010). It has been largely defended that NFTs, as revealed with silver
Consequently, these observations support the concept that b- stains, can be encountered in different conditions, but 3R/4R-tau
amyloid fuels tauopathy in sAD (Masters and Beyreuther, 2006; NFTs are practically exclusive of AD, excepting rare familial
Delacourte, 2006). prionopathies and a limited number of familial tauopathies due to
MAPT mutations (Munoz and Ferrer, 2008; Alzualde et al., 2010).
11. Seeding b-amyloid and abnormal tau Moreover, 3R/4R-tau NFTs not accompanied by glial tau inclusions
and not accompanied by PrP amyloid deposition are restricted to
Recent studies have shown that b-amyloid deposition can be AD-related pathology.
stimulated by b-amyloid under certain conditions in vivo. Thus The occurrence of certain numbers of NFTs in particular regions
plaque formation is accelerated in transgenic mice bearing FAD in most people means that this process is common but not
mutations following the intracerebral injection of diluted extracts necessarily normal. Moreover, normal aging does not exclude the
from AD brains or from old AD transgenic mice (Walker et al., 2002; circumstance of a concomitant putative age-dependent degenera-
Meyer-Luehmann et al., 2006; Walker et al., 2006). Similar results tive process. The fact that we cannot at present identify the clinical
are found following peripheral inoculation of these b-amyloid consequences of NFTs in the inner temporal lobe, corresponding to
seeds (Eisele et al., 2010). early stages of AD-related pathology (Knopman et al., 2003; Price
Transgenic rats over-expressing human APP (APP21 line) do not and Morris, 1999; Jack et al., 2010), does not mean that we should
develop b-amyloid plaques at any point in their lifespan, but SPs and overlook NFT lesions in these regions as a part of the AD spectrum.
b-amyloid angiopathy are seen in the same rats at nine months Furthermore, we do not have fair criteria to consider the well-
following intra-hippocampal injection of diluted AD extracts known memory and other cognitive decits in most individuals at
containing aggregated b-amyloid (Rosen et al., 2012). Similar pre-senile ages not resulting from AD-related alterations in the
observations have been reported in transgenic mice over-expressing inner temporal cortex.
APP, which also do not develop b-amyloid plaques during their life Recent observations have shown the appearance of NFTs in the
span, following intra-hipocampal injection of diluted AD extracts dorsal raphe nucleus and locus coeruleus in the absence of cortical
(Morales et al., 2011). Plaques not only develop locally but extend as tangles in young individuals (Rub et al., 2000; Grinberg et al., 2009;
well to the cerebral cortex at a distance from the injection. These Simic et al., 2009; Braak et al., 2011), suggesting that early AD-
ndings support the possibility that once the rst b-amyloid related lesions start in the brain stem. Whether these lesions may
deposits appear they may accelerate the accumulation of additional explain pre-cognitive decits related to AD-related pathology such
seeding in other parts of the brain, thus contributing to the as disturbances in mood, emotion and sleep, and confusion,
exponential deposition of b-amyloid. Interestingly, soluble forms of depression and others already reported years before the diagnosis
b-amyloid are particularly effective at inducing plaque formation of AD deserves attention (Jost and Grossberg, 1996).
(Langer et al., 2011). Additional studies have established that One of the major setbacks to acceptance of the spectrum of AD-
presence of b-amyloid seeds, and not the age of the host per se, is related pathology as AD is the term disease which implies some
critical to the initiation of b-amyloid aggregation in brain, while b- disability or disturbing health problem according to the WHO. It is
amyloid deposition, actuated in a brain area, eventually spreads clear that cases with AD-related pathology restricted to the
throughout the brain (Hamaguchi et al., 2012). entorhinal cortex, transentorhinal cortex and limited parts of the
These observations may apply not only to genetic-prone cases CA1 region may not suffer AD as a disease per se. Moreover, most of
with over-expression of APP, such as patients with Down these individuals probably would have never developed signicant
syndrome and with duplications in the APP locus or altered cognitive impairment or dementia. Nevertheless, they are indeed
regulatory sequences in the APP promoter region (Sleegers et al., experiencing a degenerative process within the spectrum of AD.
2006; Cabrejo et al., 2006; Brouwers et al., 2006), but may also be Similarly, the spectrum or atherosclerosis covers the presence of
considered contributory factors in sAD. isolated plaques of lipid deposition in the walls of arteries and
In addition, injection of brain extract from mutant P301S tau- ischemic complications linked with occlusion of arterial blood
expressing mice into the brain of transgenic wild-type tau- vessels. It would hardly be acceptable to speak of normal aging in
expressing animals induces assembly of wild-type human tau into the presence of a certain number of arterial plaques whilst
laments and the spread of the pathology from the site of injection reserving the term atherosclerosis exclusively for the moment
to neighbouring brain regions (Clavaguera et al., 2009). In the same when the arteries are clogged with fatty deposits and the process is
line, in vitro models have also shown that introduction of small manifested as infarction.
quantities of misfolded preformed tau brils into tau-expressing One possibility for circumventing the implications of the use
cells recruits soluble tau into lamentous inclusions reminiscent of disease when dealing with AD-related pathology would be to use
NFTs (Guo and Lee, 2011). Moreover, seeding differs in 3R and 4R the common term Alzheimer (or alzheimer) at the same level that
tau (Dinkel et al., 2011). we use the term atherosclerosis.
These observations have important consequences as they imply Under this thinking, Alzheimer is then considered an age-
that seeding and spreading of b-amyloid plaques and altered tau related biological process in certain mammals, mainly in non-
species may occur, under appropriate conditions, at a cellular level human primates and particularly in humans, which causes
in a similar way as occurs in prion diseases under appropriate progressive degeneration of the brain neuropathologically charac-
conditions (Frost and Diamond, 2010; Aguzzi and OConnor, 2010; terized by the variable combination of the neuropathological
Goedert et al., 2010; Jucker and Walker, 2011). hallmarks NFTs and SPS, and which may cause neurological
symptoms principally manifested as cognitive impairment and
12. Concluding comments dementia. This term covers early stages of AD-related pathology
with presently unavailable clinical and biomarker data, cases with
The presence of NFTs in the inner regions of the temporal cortex altered biomarkers without clinical signs, and cases with early or
in the absence of clinical symptoms in aged individuals has been advanced stages of mental deterioration and cognitive im-
largely considered as normal aging. pairment.
Yet these rst lesions are present in the majority of people over It may be argued that Alois Alzheimer described a disease
65 years of age and 100% of AD cases have entorhinal and whereas Alzheimer may be or may not be manifested as disease. It
transentorhinal pathology. is worth recalling that, in a pure sense, Alois Alzheimer described
I. Ferrer / Progress in Neurobiology 97 (2012) 3851 45

the terminal stage of a pre-senile dementia that was considered for Aguzzi, A., OConnor, T., 2010. Protein aggregation diseases: pathogenicity and
therapeutic perspectives. Nat. Rev. Drug Discov. 9, 237248.
years to be different from senile dementia with AD-related Alafuzoff, I., Pikkarainen, M., Arzberger, T., Thal, D.R., Al-Sarraj, S., Bell, J., Bodi, I.,
pathology. Moreover, cognitive impairment with AD-related Budka, H., Capetillo-Zarate, E., Ferrer, I., Gelpi, E., Gentleman, S., Giaccone, G.,
pathology was never envisaged by Alois Alzheimer as a prodromal Kavantzas, N., King, A., Korkolopoulou, P., Kovacs, G.G., Meyronet, D., Monoranu,
C., Parchi, P., Patsouris, E., Roggendorf, W., Stadelmann, C., Streichenberger, N.,
stage of pre-senile or senile dementia. Tagliavini, F., Kretzschmar, H., 2008. Inter-laboratory comparison of neuropath-
In addition to clarifying the concept of the full spectrum of AD- ological assessments of beta-amyloid protein: a study of the BrainNet Europe
related pathology, the term Alzheimer will have two important consortium. Acta Neuropathol. 115, 533546.
Albert, M.S., DeKosky, S.T., Dickson, D., Dubois, B., Feldman, H.H., Fox, N.C., Gamst,
implications. One of them is to center attention on the origin of A., Holtzman, D.M., Jagust, W.J., Petersen, R.C., Snyder, P.J., Carrillo, M.C., Thies,
Alzheimer in the very early stages of AD-related pathology because B., Phelps, C.H., 2011. The diagnosis of mild cognitive impairment due to
tau pathology and later b-amyloid pathology are already present Alzheimers disease: recommendations from the National Institute on Ag-
ingAlzheimers Association workgroups on diagnostic guidelines for Alzhei-
on a small scale during these rst stages. In addition, the
mers disease. Alzheimers Dement. 7, 270279.
hypothesis of seeding, if proved to be true in humans, implies a Alvira-Botero, X., Carro, E.M., 2010. Clearance of amyloid-b peptide across the
progression of the disease once the rst extracellular deposition of choroid plexus in Alzheimers disease. Curr. Aging Sci. 3, 219229.
b-amyloid brils or the intracellular accumulation of truncated tau Alzheimer, A., 1907. Ueber eine eigenartige Erkrankung der Hirnrinde. Centralblatt
f. Nervenh. und Psychiat. 18, 177.
is initiated. The b-amyloid cascade hypothesis further supports Alzheimer, A., 19091910. Ueber eine eigenartige Krankheitsfalle des spateren
self-recruitment of lesions. Based on these observations, thera- Alters. Zeltschr. F. D. Ges. Neurol. U. Psychiat. 4, 365385.
peutic strategies should be shifted, in part, to halting early changes Alzualde, A., Indakoetxea, B., Ferrer, I., Moreno, F., Barandiaran, M., Gorostidi, A.,
Estanga, A., Ruiz, I., Calero, M., van Leeuwen, F.W., Atares, B., Juste, R., Rodriguez-
in addition to focusing on the middle and late stages of the process. Martnez, A.B., Lopez de Munain, A., 2010. A novel PRNP Y218N mutation in
It must be stressed that the proposed term Alzheimer is not Gerstmann-Straussler-Scheinker disease with neurobrillary degeneration. J.
synonymous with Alzheimer disease or Alzheimer dementia, Neuropathol. Exp. Neurol. 69, 789800.
Amador-Ortiz, C., Lin, W.L., Ahmed, Z., Personett, D., Davies, P., Duara, R., Graff-
which must be contemplated in a clinical setting. Rather, Radford, N.R., Hutton, M.L., Dickson, D.W., 2007. TDP-43 immunoreactivity in
Alzheimer comprises molecular, morphological and accompanying hippocampal sclerosis and Alzheimers disease. Ann. Neurol. 61, 435445.
neuronal dysfunctions that can lead, or not, to the progression of Anstey, K.L., Lipnicki, D.M., Low, L.F., 2008. Cholesterol as a risk factor for dementia
and cognitive decline: a systematic review of prospective studies with meta-
neurological decits. It is true, at present, that the main hallmarks analysis. Am. J. Geriatr. Psychiatry 16, 343354.
of Alzheimer are the morphological alterations NFTs and SPs, but it Arendt, T., 2003. Synaptic plasticity and cell cycle activation i are alternative effector
may be advanced that these alterations occur in a very complex pathways: the Dr. Jekyll and Mr. Hyde concept of Alzheimers disease or the yin
and yang of neuroplasticity. Prog. Neurobiol. 71, 83248.
and still poorly understood metabolic and molecular background,
Arriagada, P.V., Marzloff, K., Hyman, B.T., 1992. Distribution of Alzheimer-type
including mitochondrial dysfunction and altered lipids in cellular pathologic changes in nondemented elderly individuals matches the pattern in
membranes. More precisely, NFTs and SPS can be considered as by- Alzheimers disease. Neurology 42, 16811688.
products or residual products resulting from intricate processes Aso, E., Lomoio, S., Lopez-Gonzalez, I., Joda, L., Carmona, M., Fernandez, N., Moreno,
J., Juves, S., Pujol, A., Pamplona, R., Portero-Otin, M., Martn, V., Daz, M., Ferrer, I.,
covering altered formation and inefcient removal of different 2011. Amyloid generation and dysfunctional immunoproteasome activation
proteins in particular molecular and metabolic settings. Therefore, with disease progression in animal model of familial Alzheimer disease. Brain
it can be envisaged that combinations of various at risk genome/ Pathol. (epub ahead of print).
Avila, J., 2006. Tau phosphorylation and aggregation in Alzheimers disease pathol-
transcriptome/proteome/lipidome/metabolomes, which make up ogy. FEBS Lett. 580, 29222927.
such predisposing molecular and metabolic backgrounds, will be Ballard, C., Gauthier, S., Corbett, A., Brayne, C., Aarsland, C., Jones, E., 2011. Alzhei-
identied in the near future. Considering this rationale, the term mers disease. Lancet 377, 10191031.
Barrachina, M., Maes, T., Buesa, C., Ferrer, I., 2006. Up-regulation of lysosome
AD-related pathology seems confusing as it implies a relationship associated-membrane protein 1 (Lamp-1) in Alzheimers disease. Neuropathol.
but not identication with the process of Alzheimer. The Appl. Neurobiol. 32, 505516.
expression Alzheimer pathology seems appropriate to describe Bateman, R.J., Aisen, P.S., De Strooper, B., Fox, N.C., Lemere, C.A., Ringman, J.M.,
Salloway, S., Sperling, R.A., Windisch, M., Xiong, C., 2011. Autosomal-dominant
the neuropathology of key alterations, even at the rst stages.
Alzheimers disease: a review and proposal for the prevention of Alzheimers
No less important are the social implications of the term disease. Alzheimers Res. Ther. 3, 1.
Alzheimer (or alzheimer) as an alternative to AD. The term AD is Becker, J.A., Hedden, T., Carmasin, J., Maye, J., Rentz, D.M., Putcha, D., Fischl, B.,
Greve, D.N., Marshall, G.A., Salloway, S., Marks, D., Buckner, R.L., Sperling, R.A.,
associated with the idea of a terrible untreatable condition that
Johnson, K.A., 2011. Amyloid-b associated cortical thinning in clinically normal
disrupts our minds and undermines our status as humans. In elderly. Ann. Neurol. 69, 10321042.
contrast, alzheimer denotes a very common condition in old age Bertram, L., Tanzi, R., 2003. Genetics of Alzheimers disease. In: Dickson, D. (Ed.),
that only in a percentage of cases progresses to dementia. Suffering Neurodegeneration: The Molecular Pathology of Dementia and Movement
Disorders. ISN Neuropath Press, Basel, pp. 4046.
from alzheimer does not imply a verdict of dementia, but rather Bierer, L.M., Hof, P.R., Purohit, D.P., Carlin, L., Schmeidler, J., Davis, K.L., Perl, D.P.,
encompasses understanding that most humans at a certain age will 1995. Neocortical neurobrillary tangles correlate with dementia severity in
face a degenerative disease of the brain with similar possibilities Alzheimers disease. Arch. Neurol. 52, 8188.
Bird, T.D., 2008. Genetic aspects of Alzheimer disease. Genet. Med. 10, 231
for treatment as atherosclerosis. The present rationale also permits 239.
an assertive conciliation to break the hierarchical status between Blessed, G., Tomlinson, B.E., Roth, M., 1968. The association between quantitative
normal and disease regarding degenerative processes associated measures and senile change in the cerebral grey matter of elderly subjects. Br. J.
Psychyatry 114, 797811.
with aging. Bloss, E.B., Morrison, J.H., Hof, P.R., Dickstein, D.L., 2011. Inuence of aging and
neurodegeneration on dendritic spine morphology. Transl. Neurosci. 2, 4960.
Acknowledgment Borchelt, D.R., Ratovitski, T., van Lare, J., Lee, M.K., Gonzales, V., Jenkins, N.A.,
Copeland, N.G., Price, D.L., Sisodia, S.S., 1997. Accelerated amyloid deposition
in the brains of transgenic mice coexpressing mutant presenilin 1 and amyloid
I wish to thank T. Yohannan for editorial assistance. precursor proteins. Neuron 19, 939945.
Bowley, M.P., Cabral, H., Rosene, D.L., Peters, A., 2010. Age changes in myelinated
References nerve bers of the cingulate bundle and corpus callosum in the rhesus monkey.
J. Comp. Neurol. 518, 30463064.
Boyd-Kimball, D., Castegna, A., Sultana, R., Poon, H.F., Petroze, R., Lynn, B.C., Klein,
Abdel Rassoul, R., Alves, S., Pantesco, V., De Vos, J., Michel, B., Perret, M., Mestre- J.B., Buttereld, D.A., 2005a. Proteomic identication of proteins oxidized by
Frances, N., Verdier, J.M., Devau, G., 2010. Distinct transcriptome expression of Ab(142) in synaptosomes: implications for Alzheimers disease. Brain Res.
the temporal cortex of the primate Microcebus murinus during brain aging 1044, 206215.
versus Alzheimers disease like pathology. PLoS ONE 5, e12770. Boyd-Kimball, D., Castegna, A., Sultana, R., Poon, H.F., Petroze, R., Lynn, B.C., Klein,
Abner, E.L., Kryscio, R.J., Schmitt, F.A., Santacruz, K.S., Jicha, G.A., Lin, Y., Neltner, J.M., J.B., Buttereld, D.A., 2005b. Proteomic identication of proteins specically
Smith, C.D., Van Eldik, L.J., Nelson, P.T., 2011. End-stage neurobrillary tangle oxidized by intracerebral injection of amyloid b-peptide (142) into rat brain:
pathology in preclinical Alzheimers disease: fact or ction? J. Alzheimers Dis. implications for Alzheimers disease. Neuroscience 132, 313324.
25, 445453.
46 I. Ferrer / Progress in Neurobiology 97 (2012) 3851

Braak, H., Alafuzoff, I., Arzberger, T., Kretzschmar, H., Del Tredici, K., 2006a. Staging Clavaguera, F., Bolmont, T., Crowther, R.A., Abramowski, D., Frank, S., Probst, A.,
of Alzheimer disease-associated neurobrillary pathology using parafn sec- Fraser, G., Stalder, A.K., Beibel, M., Staufenbiel, M., Jucker, M., Goedert, M.,
tions and immunocytochemistry. Acta Neuropathol. 112, 389404. Tolnay, M., 2009. Transmission and spreading of tauopathy in transgenic mouse
Braak, H., Rub, U., Del Tredici, K., 2006b. Cognitive decline correlates with neuro- brain. Nat. Cell Biol. 11, 909913.
pathological stage in Parkinsons disease. J. Neurol. Sci. 248, 255258. Constanza, A., Weber, K., Gandy, S., Bouras, C., Hof, P.R., Giannakopoulos, P., Canuto,
Braak, H., Braak, E., 1991. Neuropathological staging of Alzheimer-related changes. A., 2011. Review: contact sport-related chronic traumatic encephalopathy in
Acta Neuropathol 82, 239259. the elderly: clinical expression and structural substrates. Neuropathol. Appl.
Braak, H., Braak, E., 1997. Frequency of stages of Alzheimer-related lesions in Neurobiol. 37, 570584.
different age categories. Neurobiol. Aging 18, 351357. Corder, E.H., Saunders, A.M., Strittmatter, W.J., Schmechel, D.E., Gaskell, P.C., Small,
Braak, H., Braak, E., 1999. Temporal sequence of Alzheimers disease-related pa- G.W., Roses, A.D., Haines, J.L., Pericak-Vance, M.A., 1993. Gene dose of apolipo-
thology. In: Peters, A., Morrison, J.H. (Eds.), Cerebral Cortex: Vol. 14, Neurode- protein E type 4 allele and the risk of Alzheimers disease in late onset families.
generative and Age-Related Changes in Structure and Function of Cerebral Science 261, 921923.
Cortex. Kluwer Academic/Plenum, New York/Boston/Dordrecht/London/ Coskun, P., Wyrembak, J., Eschriner, S.E., Chen, H.W., Marciniack, C., LaFerla, F.,
Moscow, pp. 475512. Wallace, D.C., 2011. A mitochondrial etiology of Alzheimer and Parkinson
Braak, H., Thal, D.R., Ghebremedhin, E., del Tredici, K., 2011. Stages of the pathologic disease. Biochem. Biophys. Acta (epub ahead of print).
process in Alzheimer disease: age categories from 1 to 100 years. J. Neuropathol. Coubard, O.A., Ferruno, L., Boura, M., Gripon, A., Renaud, M., Bherer, L., 2011.
Exp. Neurol. 70, 960969. Attentional control in normal aging and Alzheimers disease. Neuropsychology
Brouwers, N., Sleegers, K., Engelborghs, S., Bogaerts, V., Serneels, S., Kamali, K., 25, 353367.
Corsmit, E., De Leenheir, E., Martin, J.J., De Deyn, P.P., Van Broeckhoven, C., Crouch, P.J., Blake, R., Duce, J.A., Ciccotosto, G.D., Li, Q.X., Barnham, K.J., Curtain, C.C.,
Theuns, J., 2006. Genetic risk and transcriptional variability of amyloid precur- Cherny, R.A., Cappai, R., Dyrks, T., Masters, C.L., Trounce, I.A., 2005. Copper-
sor protein in Alzheimers disease. Brain 129, 29842991. dependent inhibition of human cytochrome c oxidase by a dimeric conformer of
Bu, G., 2009. Apolipoprotein E and its receptors in Alzheimers disease: pathways, amyloid-b142. J. Neurosci. 25, 672679.
pathogenesis and therapy. Nat. Rev. Neurosci. 10, 333344. Deane, R., Zlokovic, B.V., 2007. Role of the bloodbrain barrier in the pathogenesis of
Buckner, R.L., Snyder, A.Z., Shannon, B.J., LaRossa, G., Sachs, R., Fotenos, A.F., Sheline, Alzheimers disease. Curr. Alzheimer Res. 4, 191197.
Y.I., Klunk, W.E., Mathis, C.A., Morris, J.C., Mintun, M.A., 2005. Molecular, De Felice, F.G., Velasco, P.T., Lambert, M.P., Viola, K., Fernandez, S.J., Ferreira, S.T.,
structural, and functional characterization of Alzheimers disease: evidence Klein, W.L., 2007. Abeta oligomers induce neuronal oxidative stress through an
for a relationship between default activity, amyloid, and memory. J. Neurosci. N-methyl-D-aspartate receptor-dependent mechanism that is blocked by the
25, 77097717. Alzheimer drug memantine. J. Biol. Chem. 282, 1159011601.
Buee, L., Delacourte, A., 2001. Tau phosphorylation. In: Hof, P.R., Mobbs, C.V. Delacourte, A., 2006. The natural and molecular history of Alzheimers disease. J.
(Eds.), Functional Neurobiology of Aging. Academic Pres, San Diego/San Alzheimers Dis. 9, 187194.
Francisco/New York/Boston/London/Sydney/Tokyo, pp. 315332. Delacourte, A., David, J.P., Sergeant, N., Buee, L., Wattez, A., Vermersch, P., Ghozali, F.,
Bussiere, T., Hof, P.R., 2001. Morphological changes in human cerebral cortex during Fallet-Bianco, C., Pasquier, F., Lebert, F., Petit, H., Di Menza, C., 1999. The
aging. In: Hof, P.R., Mobbs, C.H. (Eds.), Functional Neurobiology of Aging. biochemical pathway of neurobrillary degeneration in aging and Alzheimers
Academic Press, San Diego/San Francisco/New York/Boston/London/Sydney/ disease. Neurology 52, 11581162.
Tokyo, pp. 7784. Delay, J., Brion, S., 1962. Les demences tardives. Mason and Cie, Paris.
Buttereld, D.A., Castegna, A., Lauderback, C.M., Drake, J., 2002. Evidence that Deshpande, A., Mina, E., Glabe, C., Busciglio, J., 2006. Different conformation of
amyloid beta-peptide-induced lipid peroxidation and its sequelae in Alzhei- amyloid beta induce neurotoxicity by distinct mechanisms in human cortical
mers disease brain contribute to neuronal death. Neurobiol. Aging 23, 264 neurons. J. Neurosci. 26, 60116018.
655. Dickson, D.W., Kouri, N., Murray, M.E., Josephs, K.A., 2011. Neuropathology of
Cabrejo, L0., Guyant-Marechal, L., Laquerriere, A., Vercelletto, M., De la Fourniere, F., frontotemporal lobar degeneration-tau (FTLD-Tau). J. Mol. Neurosci. 45, 384
Thomas-Anterion, C., Verny, C., Letournel, F., Pasquier, F., Vital, A., Checler, F., 389.
Frebourg, T., Campion, D., Hannequin, D., 2006. Phenotype associated with APP Dinkel, P.D., Siddiqua, A., Huynh, H., Shah, M., Margittai, M., 2011. Variations in
duplication in ve families. Brain 129, 29662976. lament conformation dictate seeding barrier between three- and four-repeat
Calkins, M.J., Reddy, P.H., 2011. Amyloid beta impairs mitochondrial anterograde tau. Biochemistry 50, 43304336.
transport and degenerates synapses in Alzheimers disease neurons. Biochem. Du, H., Guo, L., Yan, S., Sosunov, A., McKhann, G.M., Yan, S.S., 2010. Early decits in
Biophys. Acta 1812, 507513. synaptic mitochondria in an Alzheimers disease mouse model. Proc. Natl. Acad.
Casley, C.S., Canevari, L., Land, J.M., Clark, J.B., Sharpe, M.A., 2002. b-Amyloid inhibits Sci. U.S.A. 107, 1867018675.
integrated mitochondrial respiration and key enzyme activities. J. Neurochem. Dubois, B., Feldman, H.H., Jacova, C., Cummings, J.L., Dekosky, S.T., Barberger-
80, 91100. Gateau, P., Delacourte, A., Frisoni, G., Fox, N.C., Galasko, D., Gauthier, S., Hampel,
Caspersen, C., Wang, N., Yao, J., Sosunov, A., Chen, X., Lustbader, J.W., Xu, H.W., Stern, H., Jicha, G.A., Meguro, K., OBrien, J., Pasquier, F., Robert, P., Rossor, M., Salloway,
D., McKhann, G., Yan, S.D., 2005. Mitochondrial Ab: a potential focal point for S., Sarazin, M., de Souza, L.C., Stern, Y., Visser, P.J., Scheltens, P., 2010. Revising
neuronal metabolic dysfunction in Alzheimers disease. FASEB J. 19, 20402041. the denition of Alzheimers disease: a new lexicon. Lancet Neurol. 9, 1118
Castegna, A., Aksenov, M., Aksenova, M., Thongboonkerd, V., Klein, J.B., Pierce, W.M., 1127.
Booze, R., Markesbery, W.R., Buttereld, D.A., 2002a. Proteomic identication of Dubois, B., Feldman, H.H., Jacova, C., Dekosky, S.T., Barberger-Gateau, P., Cummings,
oxidatively modied proteins in Alzheimers disease brain. Part I: creatine J., Delacourte, A., Galasko, D., Gauthier, S., Jicha, G., Meguro, K., Obrien, J.,
kinase BB, glutamine synthase, and ubiquitin carboxy-terminal hydrolase L- Pasquier, F., Robert, P., Rossor, M., Salloway, S., Stern, Y., Visser, P.J., Scheltens, P.,
1. Free Radic. Biol. Med. 33, 562571. 2007. Research criteria for the diagnosis of Alzheimers disease: revising the
Castegna, A., Aksenov, M., Thongboonkerd, V., Klein, J.B., Pierce, W.M., Booze, R., NINCDS-ADRDA criteria. Lancet Neurol. 6, 734746.
Markesbery, W.R., Buttereld, D.A., 2002b. Proteomic identication of oxida- Dumitriu, D., Hao, J., Hara, Y., Kaufmann, J., Janssen, W.G.M., Lou, W., Rapp, P.R.,
tively modied proteins in Alzheimers disease brain. Part II: dihydropyrimi- Morrison, J.H., 2010. Selective changes in thin spine density and morphology in
dinase-related protein 2, alpha-enolase and heat shock cognate 71. J. monkey prefrontal cortex correlate with aging-related cognitive impairment. J.
Neurochem. 82, 15241532. Neurosci. 30, 75077515.
Cataldo, A.M., Paskevich, P.A., Kominami, E., Nixon, R.A., 1991. Lysosomal hydro- Duyckaerts, C., Dickson, D.W., 2003. Neuropathology of Alzheimers disease. In:
lases of different classes are abnormally distributed in brains of patients with Dickson, D.W. (Ed.), Neurodegeneration: The Molecular Pathology of Dementia
Alzheimer disease. Proc. Natl. Acad. Sci. U.S.A. 88, 10998111002. and Movement Disorders. ISN Neuropath Press, Basel, pp. 4765.
Cecarini, V., Ding, Q., Keller, J.N., 2007. Oxidative inactivation of the proteasome in Eisele, Y.S., Obermuller, U., Heilbronner, G., Baumann, F., Kaeser, S.A., Wolburg, H.,
Alzheimers disease. Free Radic. Res. 41, 673680. Walker, L.C., Staufenbiel, M., Heikenwalder, M., Jucker, M., 2010. Peripherally
Chalbot, S., Zetterberg, H., Blennow, K., Fladby, T., Andreasen, N., Grundke-Iqbal, I., applied Ab-containing inoculates induce cerebral b-amyloidogenesis. Science
Iqbal, K., 2011. Blood-cerebrospinal uid barrier permeability in Alzheimers 330, 980982.
disease. J. Alzheimers Dis. 25, 505515. Erten-Lyons, D., Woltjer, R.L., Dodge, H., Nixon, R., Vorobik, R., Calvert, J.F., Leahy, M.,
Chan, R.B., Oliveira, T.G., Cortes, E.P., Honig, L.S., Duff, K.E., Smal l, S.A., Wenk, M.R., Montine, T., Kaye, J., 2009. Factors associated with resistance to dementia
Shui, G., Di Paolo, G., 2012. Comparative lipidomic analysis of mouse and human despite high Alzheimer disease pathology. Neurology 72, 354360.
brain with Alzheimer disease. J. Biol. Chem. 287, 26782688. Farrer, L.A., Cupples, L.A., Haines, J.L., Hyman, B., Kukull, W.A., Mayeux, R., Myers,
Chartier-Harlin, M.C., Crawford, F., Houlden, H., Warren, A., Hughes, D., Fidani, L., R.H., Pericak-Vance, M.A., Risch, N., van Duijn, C.M., 1997. Effects of age, sex, and
1991. Early-onset Alzheimers disease caused by mutations at codon 717 of the ethnicity on the association between apolipoprotein E genotype and Alzheimer
beta-amyloid precursor protein gene. Nature 353, 844846. disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consor-
Choi, J., Levey, A.I., Weintraub, S.T., Rees, H.D., Gearing, M., Chin, L.S., Li, L., 2004. tium. JAMA 27, 13491356.
Oxidative modications and down-regulation of ubiquitin carboxyl-terminal Fasulo, L., Ugolini, G., Visintin, M., Bradbury, A., Brancolini, C., Verzillo, V., Novak, M.,
hydrolase L1 associated with idiopathic Parkinsons and Alzheimers diseases. J. Cattaneo, A., 2000. The neuronal microtubule-associated protein tau is a
Biol. Chem. 279, 1325613264. substrate for caspase-3 and an effector of apoptosis. J. Neurochem. 75, 624633.
Choi, J., Rees, H.D., Weintraub, S.T., Levey, A.I., Chin, L.S., Li, L., 2005. Oxidative Ferrer, I., 2009. Altered mitochondria, energy metabolism, voltage-dependent anion
modications and aggregation of Cu,Zn-superoxide dismutase associated with channel, and lipid rafts converge to exhaust neurons in Alzheimers disease. J.
Alzheimer and Parkinson diseases. J. Biol. Chem. 280, 1164811655. Bioenerg. Biomembr. 841, 425431.
Chung, C.W., Song, Y.H., Kim, I.K., Yoon, W.J., Ryu, B.R., Jo, D.G., Woo, H.N., Kwon, Y.K., Ferrer, I., Gomez-Isla, T., Puig, B., Freixes, M., Ribe, E., Dalfo, E., Avila, J., 2005. Current
Kim, H.H., Gwag, B.J., Mook-Jung, I.H., Jung, Y.K., 2001. Pro-apoptotic effects of advances on different kinases involved in tau phosphorylation, and implications
tau cleavage product generated by caspase-3. Neurobiol. Dis. 8, 162172. in Alzheimers disease and tauopathies. Curr. Alzheimer Res. 2, 318.
I. Ferrer / Progress in Neurobiology 97 (2012) 3851 47

Ferrer, I., Santpere, G., van Leeuwen, F.W., 2008. Argyrophilic grain disease. Brain Livingston, G., Bass, N.J., Gurling, H., McQuillin, A., Gwilliam, R., Deloukas, P., Al-
131, 14161432. Chalabi, A., Shaw, C.E., Tsolaki, M., Singleton, A.B., Guerreiro, R., Muhleisen, T.W.,
Finch, C.E., Austad, S.N., 2012. Primate aging in the mammalian scheme: the puzzle Nothen, M.M., Moebus, S., Jockel, K.H., Klopp, N., Wichmann, H.E., Carrasquillo,
of extreme variation in brain aging. Age (Dordr) (epub ahead of print). M.M., Pankratz, V.S., Younkin, S.G., Holmans, P.A., ODonovan, M., Owen, M.J.,
Filipcik, P., Cente, M., Krajciova, G., Vanicky, I., Novak, M., 2009. Cortical and Williams, J., 2009. Genome-wide association study identies variants at GLU
hippocampal neurons from truncated tau transgenic rat express multiple and CRI associated with Alzheimers disease. Nat. Genet 41, 10881093.
markers of neurodegeneration. Cell Mol. Neurobiol. 29, 895900. Hauptmann, S., Keil, U., Scherping, I., Bonert, A., Eckert, A., Muller, W.E., 2006.
Frost, B., Diamond, M.I., 2010. Prion-like mechanisms in neurodegenerative dis- Mitochondrial dysfunction in sporadic and genetic Alzheimers disease. FASEB J.
eases. Nat. Rev. Neurosci. 11, 155159. 19, 20402041.
Games, D., Adams, D., Alessandrini, R., Barbour, R., Berthelette, P., Blackwell, C., Carr, Hawkes, C.A., Hartig, W., Kacza, J., Schliebs, R., Weller, R.O., Nicoll, J.A., Carare, R.O.,
T., Clemens, J., Donaldson, T., Gillespie, F., et al., 1995. Alzheimer-type neuro- 2011. Perivascular drainage of solutes is impaired in the ageing mouse brain
pathology in transgenic mice overexpressing V717F beta-amyloid precursor and in the presence of cerebral amyloid angiopathy. Acta Neuropathol. 121,
protein. Nature 373, 523527. 431443.
Giannakopoulos, P., Silhol, S., Jallageas, V., Mallet, J., Bons, N., Bouras, C., Delaere, P., Head, E., 2011. Neurobiology of the aging dog. Age (Dordr) 33, 485496.
1997. Quantitative analysis of tau protein-immunoreactive accumulations and Hedden, T., Gabrieli, J.D., 2004. Insights into the ageing mind: a view from cognitive
beta amyloid protein deposits in the cerebral cortex of the mouse lemur, neuroscience. Nat. Rev. Neurosci. 5, 8796.
Microcebus murinus. Acta Neuropathol. 94, 131139. Hirai, K., Aliev, G., Nunomura, A., Fujioka, H., Russell, R.L., Atwood, C.S., Johnson, A.B.,
Glabe, C.G., Kayed, R., 2006. Common structure and toxic function of amyloid Kress, Y., Vinters, H.V., Tabaton, M., Shimohama, S., Cash, A.D., Siedlak, S.L.,
oligomers implies a common mechanism of pathogenesis. Neurology 66, S74 Harris, P.L., Jones, P.K., Petersen, R.B., Perry, G., Smith, M.A., 2001. Mitochondrial
S78. abnormalities in Alzheimers disease. J. Neurosci. 21, 30173023.
Glenner, G.G., Wong, C.W., 1984a. Alzheimers disease: initial report of the puri- Hollingworth, P., Harold, D., Sims, R., Gerrish, A., Lambert, J.C., Carrasquillo, M.M.,
cation and characterization of a novel cerebrovascular amyloid protein. Bio- Abraham, R., Hamshere, M.L., Pahwa, J.S., Moskvina, V., Dowzell, K., Jones, N.,
chem. Biophys. Res. Commun. 120, 885890. Stretton, A., Thomas, C., Richards, A., Ivanov, D., Widdowson, C., Chapman, J.,
Glenner, G.G., Wong, C.W., 1984b. Alzheimers disease and Downs syndrome: Lovestone, S., Powell, J., Proitsi, P., Lupton, M.K., Brayne, C., Rubinsztein, D.C., Gill,
sharing of a unique cerebrovascular amyloid bril protein. Biochem. Biophys. M., Lawlor, B., Lynch, A., Brown, K.S., Passmore, P.A., Craig, D., McGuinness, B.,
Res. Commun. 122, 11311135. Todd, S., Holmes, C., Mann, D., Smith, A.D., Beaumont, H., Warden, D., Wilcock, G.,
Goedert, M., 1996. Tau protein and the neurobrillary pathology of Alzheimers Love, S., Kehoe, P.G., Hooper, N.M., Vardy, E.R., Hardy, J., Mead, S., Fox, N.C., Rossor,
disease. Ann. N. Y. Acad. Sci. 777, 121131. M., Collinge, J., Maier, W., Jessen, F., Ruther, E., Schurmann, B., Heun, R., Kolsch, H.,
Goedert, M., Clavaguera, F., Tolnay, M., 2010. The propagation of prion-like protein van den Bussche, H., Heuser, I., Kornhuber, J., Wiltfang, J., Dichgans, M., Frolich, L.,
inclusions in neurodegenerative diseases. Trends Neurosci. 33, 317325. Hampel, H., Gallacher, J., Hull, M., Rujescu, D., Giegling, I., Goate, A.M., Kauwe, J.S.,
Goedert, M., Klug, A., Crowther, R.A., 2006. Tau protein, the paired helical lament Cruchaga, C., Nowotny, P., Morris, J.C., Mayo, K., Sleegers, K., Bettens, K., Engel-
and Alzheimers disease. J. Alzheimers Dis. 9, 195207. borghs, S., De Deyn, P.P., Van Broeckhoven, C., Livingston, G., Bass, N.J., Gurling, H.,
Goedert, M., Spillantini, M.G., Jakes, R., Rutherford, D., Crowther, R.A., 1989. Multiple McQuillin, A., Gwilliam, R., Deloukas, P., Al-Chalabi, A., Shaw, C.E., Tsolaki, M.,
isoforms of human microtubule-associated protein tau: sequences and locali- Singleton, A.B., Guerreiro, R., Muhleisen, T.W., Nothen, M.M., Moebus, S., Jockel,
zation in neurobrillary tangles of Alzheimers disease. Neuron 3, 519526. K.H., Klopp, N., Wichmann, H.E., Pankratz, V.S., Sando, S.B., Aasly, J.O., Barci-
Goedert, M., Wischik, C.M., Crowther, R.A., Walker, J.E., Klug, A., 1988. Cloning and kowska, M., Wszolek, Z.K., Dickson, D.W., Graff-Radford, N.R., Petersen, R.C.,
sequencing of a core protein of the paired helical lament of Alzheimer disease: Alzheimers Disease Neuroimaging Initiative, van Duijn, C.M., Breteler, M.M.,
Identication as the microtubule-associated protein tau. Proc. Natl. Acad. Sci. Ikram, M.A., DeStefano, A.L., Fitzpatrick, A.L., Lopez, O., Launer, L.J., Seshadri, S.,
U.S.A. 85, 40514055. consortium, CHARGE., Berr, C., Campion, D., Epelbaum, J., Dartigues, J.F., Tzourio,
Gomez-Ramos, A., Daz-Hernandez, M., Cuadros, R., Hernandez, F., Avila, J., 2006. C., Alperovitch, A., Lathrop, M., EADI1 consortium, Feulner, T.M., Friedrich, P.,
Extracellular tau is toxic to neuronal cells. FEBS Lett. 580, 48424850. Riehle, C., Krawczak, M., Schreiber, S., Mayhaus, M., Nicolhaus, S., Wagenpfeil, S.,
Gonatas, N.K., Anderson, W., Evangelista, I., 1967. The contribution of altered Steinberg, S., Stefansson, H., Stefansson, K., Snaedal, J., Bjornsson, S., Jonsson, P.V.,
synapses in the senile plaques: an electron microscopic study in Alzheimers Chouraki, V., Genier-Boley, B., Hiltunen, M., Soininen, H., Combarros, O., Zelenika,
disease. J. Neuropathol. Exp. Neurol. 26, 2539. D., Delepine, M., Bullido, M.J., Pasquier, F., Mateo, I., Frank-Garcia, A., Porcellini, E.,
Gotz, J., Ilttner, L.M., 2008. Animal models of Alzheimers disease and frontotem- Hanon, O., Coto, E., Alvarez, V., Bosco, P., Siciliano, G., Mancuso, M., Panza, F.,
poral dementia. Nat. Rev. Neurosci. 9, 532544. Solfrizzi, V., Nacmias, B., Sorbi, S., Bossu, P., Piccardi, P., Arosio, B., Annoni, G.,
Gouras, G.K., Tampellini, D., Takahashi, R.H., Capetillo-Zarate, E., 2010. Intraneur- Seripa, D., Pilotto, A., Scarpini, E., Galimberti, D., Brice, A., Hannequin, D., Licastro,
onal amyloid accumulation and synapse pathology in Alzheimers disease. Acta F., Jones, L., Holmans, P.A., Jonsson, T., Riemenschneider, M., Morgan, K., Younkin,
Neuropathol. 119, 523541. S.G., Owen, M.J., ODonovan, M., Amouyel, P., Williams, J., 2011. Common variants
Grinberg, L.T., Rub, U., Ferretti, R.E., Nitrini, R., Farfel, J.M., Polichiso, L., et al., at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with
Brazilian Brain Bank Study Group, 2009. The dorsal raphe nucleus shows Alzheimers disease. Nat. Genet. 43, 429435.
phospho-tau neurobrillary changes before the transentorhinal region in Alz- Hof, P.R., 2001. Regional and laminar patterns of selective vulnerabilit. In: Alzhei-
heimers disease. A precocious onset? Neuropathol. Appl. Neurobiol. 35, 406 mers, disease., In: Hof, P.R., Mobbs, C.V. (Eds.), Functional Neurobiology of
416. Aging. Academic Press, San Diego/San Francisco/New York/Boston/London/
Grinberg, L.T., Thal, D.R., 2010. Vascular pathology in the aged human brain. Acta Sydney/Tokyo, pp. 95110.
Neuropathol. 119, 277290. Hof, P.R., Bouras, C., Morrison, J.H., 1999. Cortical neuropathology in aging and
Grundke-Iqbal, I., Iqbal, K., Quinlan, M., Tung, Y.C., Zaidi, M.S., Wisniewski, H.M., dementing disorders: neuronal typology, connectivity, and selective vulnera-
1986. Microtubule-associated protein tau. A component of Alzheimer paired bility. In: Peters, A., Morrison, J.H. (Eds.), Cerebral Cortex: Vol. 14: Neurodegen-
helical laments. J. Biol. Chem. 261, 60846089. erative and Age-Related Changes in Structure and Function of Cerebral Cortex.
Guo, J.L., Lee, V.M., 2011. Seeding of normal tau by pathological tau conformers Kluwer Academic/Plenum, New York/Boston/Dordrecht/London/Moscow, pp.
drives pathogenesis of Alzheimer-like tangles. J. Biol. Chem. 286, 1531715331. 175312.
Hamaguchi, T., Eisele, Y., Varvel, N., Lamb, B.T., Walker, L.C., Jucker, M., 2012. The Hof, P.R., Morrison, J.H., 2004. The aging brain: morphomolecular senescence of
presence of Ab seeds, and not age per se, is critical to the initiation of Ab cortical circuits. Trends Neurosci. 27, 607613.
deposition in the brain. Acta Neuropathol. 123, 3137. Hoozemans, J.J., van Haastert, E.S., Nijholt, D.A., Rozemuller, A.J., Eikelenboom, P.,
Hanger, D.P., Seereeram, A., Noble, W., 2009. Mediators of tau phosphorylation in Scheper, W., 2009. The unfolded protein response is activated in pretangle
the pathogenesis of Alzheimers disease. Expert Rev. Neurother. 9, 16471666. neurons in Alzheimers disease hippocampus. Am. J. Pathol. 174, 12411251.
Hardy, J., 1997. Amyloid, the presenilins and Alzheimers disease. Trends Neurosci. Hsiao, K., Chapman, P., Nilsen, S., Eckman, C., Harigaya, Y., Younkin, S., Yang, F., Cole,
20, 154159. G., 1996. Correlative memory decits, Abeta elevation, and amyloid plaques in
Hardy, J., 2006. Has the amyloid cascade hypothesis for Alzheimers disease been transgenic mice. Science 274, 99102.
proved? Curr. Alzheimer Res. 3, 7173. Hu, W.T., Chen-Plotkin, A., Arnold, S.E., Grossman, M., Clark, C.M., Shaw, L.M.,
Hardy, J., 2009. The amyloid hypothesis for Alzheimers disease: a critical reap- Pickering, E., Kuhn, M., Chen, Y., McCluskey, L., Elman, L., Karlawish, J., Hurtig,
praisal. J. Neurochem. 110, 11291134. H.I., Siderowf, A., Lee, V.M., Soares, H., Trojanowski, J.Q., 2010. Novel CSF
Hardy, J.A., Higgins, G.A., 1992. Alzheimers disease: the amyloid cascade hypothe- biomarkers for Alzheimers disease and mild cognitive impairment. Acta Neu-
sis. Science 256, 184185. ropathol. 119, 669678.
Hardy, J., Selkoe, D.J., 2002. The amyloid hypothesis of Alzheimers disease. Progress Hurtado, D.E., Molina-Porcel, L., Iba, M., Aboagye, A.K., Paul, S.M., Trojanowski, J.Q.,
and problems on the road to therapeutics. Science 297, 353356. Lee, V.M., 2010. Ab accelerates the spatiotemporal progression of tau pathology
Harold, D., Abraham, R., Hollingworth, P., Sims, R., Gerrish, A., Hamshere, M.L., and augments tau amyloidosis in an Alzheimer mouse model. Am. J. Pathol. 177,
Pahwa, J.S., Moskvina, V., Dowzell, K., Williams, A., Jones, N., Thomas, C., 19771988.
Stretton, A., Morgan, A.R., Lovestone, S., Powell, J., Proitsi, P., Lupton, M.K., Iqbal, K., Grundke-Iqbal, I., 2010. Alzheimers disease, a multifactorial disorder
Brayne, C., Rubinsztein, D.C., Gill, M., Lawlor, B., Lynch, A., Morgan, K., Brown, seeking multitherapies. Alzheimers Dement. 6, 420424.
K.S., Passmore, P.A., Craig, D., McGuinness, B., Todd, S., Holmes, C., Mann, D., Iqbal, K., Grundke-Iqbal, I., Zaidi, T., Merz, P.A., Wen, G.Y., Shaikh, S.S., Wisniewski,
Smith, A.D., Love, S., Kehoe, P.G., Hardy, J., Mead, S., Fox, N., Rossor, M., Collinge, H.M., Alafuzoff, I., Winblad, B., 1986. Defective brain microtubule assembly in
J., Maier, W., Jessen, F., Schurmann, B., van den Bussche, H., Heuser, I., Kornhu- Alzheimers disease. Lancet 2, 421426.
ber, J., Wiltfang, J., Dichgans, M., Frolich, L., Hampel, H., Hull, M., Rujescu, D., Ince, P., 2001. Dementia with Lewy bodies. Adv. Exp. Med. Biol. 487, 135145.
Goate, A.M., Kauwe, J.S., Cruchaga, C., Nowotny, P., Morris, J.C., Mayo, K., Ince, P.G., Perry, E.K., Morris, C.M., 1998. Dementia with Lewy bodies. A distinct non-
Sleegers, K., Bettens, K., Engelborghs, S., De Deyn, P.P., Van Broeckhoven, C., Alzheimer dementia syndrome? Brain Pathol. 8, 299324.
48 I. Ferrer / Progress in Neurobiology 97 (2012) 3851

Jack, C.R., Albert, M.S., Knopman, D.S., McKhann, G.M., Sperling, R.A., Carrillo, M.C., Korolainen, M.A., Goldsteins, G., Nyman, T.A., Alafuzoff, I., Koistinaho, J., Pirttila, T.,
Thies, B., Phelps, C.H., 2011a. Introduction to the recommendations from the 2006. Oxidative modication of proteins in the frontal cortex of Alzheimers
National Institute on AgingAlzheimers Association workgroups on diagnostic disease brain. Neurobiol. Aging 27, 4253.
guidelines for Alzheimers disease. Alzheimers Dement. 7, 257262. Kovacech, B., Novak, M., 2010. Tau truncation is a productive posttranslational
Jack, C.R., Vemuri, P., Wiste, H.J., Weigand, S.D., Aisen, P.S., Trojanowski, J.Q., Shaw, modication of neurobrillary degeneration in Alzheimers disease. Curr. Alz-
L.M., Bernstein, M.A., Petersen, R.C., Weiner, M.W., Knopman, D.S., Alzheimers heimer Res. 7, 708716.
Disease Neuroimaging Initiative, 2011b. Evidence for ordering of Alzheimer Kovacs, G.G., Alafuzoff, I., Al-Sarraj, S., Arzberger, T., Bogdanovic, N., Capellari, S.,
disease biomarkers. Arch. Neurol. 68, 15261535. Ferrer, I., Gelpi, E., Kovari, V., Kretzschmar, H., Nagy, Z., Parchi, P., Seilhean, D.,
Jack, C.R., Lowe, V.J., Senjem, M.L., Weigand, S.D., Kemp, B.J., Shiung, M.M., Knopman, Soininen, H., Troakes, C., Budka, H., 2008. Mixed brain pathologies in dementia:
D.S., Boeve, B.F., Klunk, W.E., Mathis, C.A., Petersen, R.C., 2008. 11C PiB and the BrainNet Europe consortium experience. Dement. Geriatr. Cogn. Disord. 26,
structural MRI provide complementary information in imaging of Alzheimers 343350.
disease and amnestic mild cognitive impairment. Brain 131, 665680. Lambert, J.C., Heath, S., Even, G., Campion, D., Sleegers, K., Hiltunen, M., Combarros,
Jack, C.R., Wiste, H.J., Vemuri, P., Weigand, S.D., Senjem, M.L., Zeng, G., Bernstein, O., Zelenika, D., Bullido, M.J., Tavernier, B., Letenneur, L., Bettens, K., Berr, C.,
M.A., Gunter, J.L., Pankratz, V.S., Aisen, P.S., Weiner, M.W., Petersen, R.C., Shaw, Pasquier, F., Fievet, N., Barberger-Gateau, P., Engelborghs, S., De Deyn, P., Mateo,
L.M., Trojanowski, J.Q., Knopman, D.S., Alzheimers Disease Neuroimaging I., Franck, A., Helisalmi, S., Porcellini, E., Hanon, O., European Alzheimers
Initiative, 2010. Brain beta-amyloid measures and magnetic resonance imaging Disease Initiative Investigators, de Pancorbo, M.M., Lendon, C., Dufouil, C.,
atrophy both predict time-to-progression from mild cognitive impairment to Jaillard, C., Leveillard, T., Alvarez, V., Bosco, P., Mancuso, M., Panza, F., Nacmias,
Alzheimers disease. Brain 133, 33363348. B., Bossu, P., Piccardi, P., Annoni, G., Seripa, D., Galimberti, D., Hannequin, D.,
Jaeger, S., Pietrzik, C.U., 2008. Functional role of lipoprotein receptors in Alzheimers Licastro, F., Soininen, H., Ritchie, K., Blanche, H., Dartigues, J.F., Tzourio, C., Gut, I.,
disease. Curr. Alzheimer Res. 5, 1525. Van Broeckhoven, C., Alperovitch, A., Lathrop, M., Amouyel, P., 2009. Genome-
Jones, L., Holmans, P.A., Hamshere, M.L., Harold, D., Moskvina, V., Ivanov, D., wide association study identies variants at CLU and CR1 associated with
Pocklington, A., Abraham, R., Hollingworth, P., Sims, R., Gerrish, A., Pahwa, Alzheimers disease. Nat. Genet. 41, 10491094.
J.S., Jones, N., Stretton, A., Morgan, A.R., Lovestone, S., Powell, J., Proitsi, P., Langer, F., Eisele, Y.S., Fritschi, S.K., Staufenbeil, M., Walker, L.C., Jucker, M., 2011.
Lupton, M.K., Brayne, C., Rubinsztein, D.C., Gill, M., Lawlor, B., Lynch, A., Morgan, Soluble Ab seeds are potent inducers of cerebral b-amyloid deposition. J.
K., Brown, K.S., Passmore, P.A., Craig, D., McGuinness, B., Todd, S., Holmes, C., Neurosci. 31, 1448814495.
Mann, D., Smith, A.D., Love, S., Kehoe, P.G., Mead, S., Fox, N., Rossor, M., Collinge, Languille, S., Blanc, S., Blin, O., Canale, C.I., Dal-Pan, A., Devau, G., Dhenain, M.,
J., Maier, W., Jessen, F., Schurmann, B., Heun, R., Kolsch, H., van den Bussche, H., Dorieux, O., Epelbaum, J., Gomez, D., Hardy, I., Henry, P.Y., Irving, E.A., Marchal,
Heuser, I., Peters, O., Kornhuber, J., Wiltfang, J., Dichgans, M., Frolich, L., Hampel, J., Mestre-Frances, N., Perret, M., Picq, J.L., Pifferi, F., Rahman, A., Schenker, E.,
H., Hull, M., Rujescu, D., Goate, A.M., Kauwe, J.S., Cruchaga, C., Nowotny, P., Terrien, J., Thery, M., Verdier, J.M., Aujard, F., 2012. The grey mouse lemur: a
Morris, J.C., Mayo, K., Livingston, G., Bass, N.J., Gurling, H., McQuillin, A., non-human primate model for ageing studies. Ageing Res. Rev. 11, 150162.
Gwilliam, R., Deloukas, P., Al-Chalabi, A., Shaw, C.E., Singleton, A.B., Guerreiro, Lasagna-Reeves, C.A., Castillo-Carranza, D.L., Guerrero-Munoz, M.J., Jackson, G.R.,
R., Muhleisen, T.W., Nothen, M.M., Moebus, S., Jockel, K.H., Klopp, N., Wich- Kayed, R., 2010. Preparation and characterization of neurotoxic tau oligomers.
mann, H.E., Ruther, E., Carrasquillo, M.M., Pankratz, V.S., Younkin, S.G., Hardy, J., Biochemistry 49, 1003910041.
ODonovan, M.C., Owen, M.J., Williams, J., 2010. Genetic evidence implicates the Lee, J.H., Won, S.M., Suh, J., Son, S.J., Moon, G.J., Park, U.J., Gwaq, B.J., 2010. Induction
immune system and cholesterol metabolism in the aetiology of Alzheimers of the unfolded protein response and cell death pathway in Alzheimers disease,
disease. PLoS ONE 5, e13950. but not in aged Tg2576 mice. Exp. Mol. Med. 42, 386394.
Jost, B.C., Grossberg, G.T., 1996. The evolution of psychiatric symptoms in Alzhei- Leverenz, J.B., Raskind, M.A., 1998. Early amyloid deposition in the medial temporal
mers disease: a natural history study. J. Am. Geriatr. Soc. 44, 10781081. lobe of young Down syndrome patients: a regional quantitative analysis. Exp.
Jucker, M., Walker, L.C., 2011. Pathogenic protein seeding in Alzheimer disease and Neurol. 150, 296304.
other neurodegenerative disorders. Ann. Neurol. 70, 532540. Levy, E., Carman, M.D., Fernandez-Madrid, I.J., Power, M.D., Lieberburg, I., van
Jun, G., Naj, A.C., Beecham, G.W., Wang, L.S., Buros, J., Gallins, P.J., Buxbaum, J.D., Duinen, S.G., Bots, G.T., Luyendijk, W., Frangione, B., 1990. Mutation of the
Ertekin-Taner, N., Fallin, M.D., Friedland, R., Inzelberg, R., Kramer, P., Rogaeva, E., Alzheimers disease amyloid gene in hereditary cerebral hemorrhage, Dutch
St George-Hyslop, P., Alzheimers Disease Genetics Consortium, Cantwell, L.B., type. Science 248, 11241126.
Dombroski, B.A., Saykin, A.J., Reiman, E.M., Bennett, D.A., Morris, J.C., Lunetta, Lewis, J., Dickson, D.W., Lin, W.L., Chisholm, L., Corral, A., Jones, G., Yen, S.H., Sahara,
K.L., Martin, E.R., Montine, T.J., Goate, A.M., Blacker, D., Tsuang, D.W., Beekly, D., N., Skipper, L., Yager, D., Eckman, C., Hardy, J., Hutton, M., McGowan, E., 2001.
Cupples, L.A., Hakonarson, H., Kukull, W., Foroud, T.M., Haines, J., Mayeux, R., Enhanced neurobrillary degeneration in transgenic mice expressing mutant
Farrer, L.A., Pericak-Vance, M.A., Schellenberg, G.D., 2010. Meta-analysis con- tau and APP. Science 293, 14871491.
rms CR1, CLU, and PICALM as Alzheimer disease risk loci and reveals inter- Lowe, J., Mirra, S.S., Hyman, B., Dickson, D.W., 2008. Ageing and Dementia. In: Love,
actions with APOE genotypes. Arch. Neurol. 67, 14731484. S., Louis, D.N., Ellison, D.W. (Eds.), Greenelds Neuropathology, vol. 1. Hodder
Kabaso, D., Coskren, P.J., Henry, B.I., Hof, P.R., Wearne, S.L., 2009. The electronic Arnold, London, pp. 10311152.
structure of pyramidal neurons contributing to prefrontal cortical circuits in Luse, S.A., Smith Jr., K.R., 1964. The ultrastructure of senile plaques. Am. J. Pathol. 44,
macaque monkeys is signicantly altered in aging. Cereb. Cortex 19, 2248 553563.
2268. Lustbader, J.W., Cirilli, M., Lin, C., Xu, H.W., Takuma, K., Wang, N., Caspersen, C.,
Kachaturian, Z.S., 1985. Diagnosis of Alzheimers disease. Arch. Neurol. 42, 1097 Chen, X., Pollak, S., Chaney, M., Trinchese, F., Liu, S., Gunn-Moore, F., Lue, L.F.,
1105. Walker, D.G., Kuppusamy, P., Zewier, Z.L., Arancio, O., Stern, D., Yan, S.S., Wu, H.,
Kaneko, M., Koike, H., Saito, R., Kitamura, Y., Okuma, Y., Nomura, Y., 2010. Loss of 2004. ABAD directly links Ab to mitochondrial toxicity in Alzheimers disease.
HRD1-mediated protein degradation causes amyloid precursor protein accu- Science 304, 448452.
mulation and amyloid-beta generation. J. Neurosci. 30, 39243932. Mann, D.M., Esiri, M.M., 1989. The pattern of acquisition of plaques and tangles in
Kang, J., Lemaire, H.G., Unterbeck, A., Salbaum, J.M., Masters, C.L., Grzeschik, K.H., the brains of patients under 50 years of age with Downs syndrome. J. Neurol.
Multhaup, G., Beyreuther, K., Muller-Hill, B., 1987. The precursor of Alzheimers Sci. 89, 169179.
disease amyloid A4 protein resembles a cell-surface receptor. Nature 325, 733 Markesbery, W.R., 2010. Neuropathologic alterations in mild cognitive impairment:
736. a review. J. Alzheimers Dis. 19, 221228.
Keller, J.N., Hanni, K.B., Markesbery, W.R., 2000. Impaired proteasome function in Martn, V., Fabelo, N., Santpere, G., Puig, B., Marn, R., Ferrer, I., Diaz, M., 2010. Lipid
Alzheimers disease. J. Neurochem. 75, 436439. alterations in lipid rafts from Alzheimers disease human brain cortex. J.
Kidd, M., 1964. Alzheimers disease: an electron microscopical study. Brain 87, 307 Alzheimers Dis. 19, 489502.
320. Marzolo, M.P., Bu, G., 2009. Lipoprotein receptors and cholesterol in APP trafcking
Kim, S.H., Vlonsky, R., Cairos, N., Lubec, G., 2000. Decreased levels of complex III core and proteolytic processing, implications for Alzheimers disease. Semin. Cell.
protein and complex 1 and complex V beta in brains from patients with Dev. Biol. 20, 191200.
Alzheimers disease and Down syndrome. Cell Mol. Life Sci. 57, 18101816. Masliah, E., Salmon, D.P., 1999. Neuropathological correlations of dementia in
Kish, S.J., Bergeron, C., Rajput, A., Dozic, S., Mastrogiacomo, F., Chang, L.J., Wilsonm, Alzheimers disease. In: Peters, A., Morrison, J.H. (Eds.), Cerebral Cortex: Vol.
J.M., DiStefanom, L.M., Nobregam, J.N., 1992. Brain cytochrome oxidase in 14: Neurodegenerative and Age-Related Changes in Structure and Function of
Alzheimers disease. J. Neurochem. 59, 776779. Cerebral Cortex. Kluwer Academic/Plenum, New York/Boston/Dordrecht/Lon-
Kivipelto, M., Solomon, A., 2006. Cholesterol as a risk factor for Alzheimers don/Moscow, pp. 513552.
diseaseepidemiological evidence. Acta Neurol. Scand. 114, 5057. Masters, C.L., Beyreuther, K., 2003. Molecular pathogenesis of Alzheimers dis-
Klippel, M., Lhermitte, J., 1905. Les demences. Anatomie pathologique et patho- ease. In: Dickson, D.W. (Ed.), Neurodegeneration: The Molecular Pathology
genie. Rev. Psychiat. 9, 485514. of Dementia and Movement Disorders. ISN Neuropath Press, Basel, pp. 69
Klunk, W.E., Price, J.C., Mathis, C.A., Tsopelas, N.D., Lopresti, B.J., Ziolko, S.K., Bi, W., 73.
Hoge, J.A., Cohen, A.D., Ikonomovic, M.D., Saxton, J.A., Snitz, B.E., Pollen, D.A., Masters, C.L., Beyreuther, K., 2006. Alzheimers centennial legacy: prospects for
Moonis, M., Lippa, C.F., Swearer, J.M., Johnson, K.A., Rentz, D.M., Fischman, A.J., rational therapeutic intervention targeting the Ab amyloid pathway. Brain 129,
Aizenstein, H.J., DeKosky, S.T., 2007. Amyloid deposition begins in the striatum 28232839.
of presenilin-1 mutation carriers from two unrelated pedigrees. J. Neurosci. 27, Masters, C.L., Simms, G., Weinman, N.A., Multhaup, G., McDonald, B.L., Beyreuther,
61746184. K., 1985. Amyloid plaque core protein in Alzheimer disease and Down syn-
Knopman, D.S., Parisi, J.E., Salviati, A., Floriach-Robert, M., Boeve, B.F., Ivnik, R.J., drome. Proc. Natl. Acad. Sci. U.S.A. 82, 42454249.
Smith, G.E., Dickson, D.W., Johnson, K.A., Petersen, L.E., McDonald, W.C., Braak, Mastroeni, D., McKee, A., Grover, A., Rogers, J., Coleman, P.D., 2009. Epigenetic
H., Petersen, R.C., 2003. Neuropathology of cognitively normal elderly. J. Neu- differences in cortical neurons from a pair of monozygotic twins discordant for
ropathol. Exp. Neurol. 62, 10871095. Alzheimers disease. PLoS ONE 4, e6617.
I. Ferrer / Progress in Neurobiology 97 (2012) 3851 49

McGowan, E., Riksen, J., Hutton, M., 2006. A decade of modelling Alzheimers disease Mutsuga, M., Chambers, J.K., Uchida, K., Tei, M., Makibuchi, T., Mizorogi, T., Taka-
in transgenic mice. Trends Genet. 22, 281289. shima, A., Nakayama, H., 2012. Binding of curcumin to senile plaques and
McKhann, G., Drachman, D., Folstein, M., Katzman, R., Price, D., Stadlan, E.M., 1984. cerebral amyloid angiopathy in the aged brain of various animals and to
Clinical diagnosis of Alzheimers disease: report of the NINCDS-ADRDA Work neurobrillary tangles in Alzheimers brain. J. Vet. Med. Sci. 74, 5157.
Group under the auspices of Department of Health and Human Services Task Mutisya, E.M., Bowling, A.C., Beal, M.F., 1994. Cortical cytochrome oxidase activity is
Force on Alzheimers disease. Neurology 34, 939944. reduced in Alzheimers disease. J. Neurochem. 63, 21792184.
McKhann, G.M., Knopman, D.S., Chertkow, H., Hyman, B.T., Jack Jr., C.R., Kawas, Nakano, I., Hirano, A., 1999. Ultrastructural changes in dementing illness. In: Peters,
C.H., Klunk, W.E., Koroshetz, W.J., Manly, J.J., Mayeux, R., Mohs, R.C., Morris, A., Morrison, J.H. (Eds.), Cerebral Cortex: Vol. 14, Neurodegenerative and Age-
J.C., Rossor, M.N., Scheltens, P., Carrillo, M.C., Thies, B., Weintraub, S., Phelps, Related Changes in Structure and Function of Cerebral Cortex. Kluwer Academ-
C.H., 2011. The diagnosis of dementia due to Alzheimers disease. Recom- ic/Plenum, New York/Boston/Dordrecht/London/Moscow, pp. 399432.
mendations from the National Institute on AgingAlzheimers Association Nelson, P.T., Braak, H., Markesbery, W.R., 2009. Neuropathology and cognitive
workgroups on diagnostic guidelines for Alzheimers disease. Alzheimers impairment in Alzheimer disease: a complex but coherent relationship. J.
Dement. 7, 263269. Neuropathol. Exp. Neurol. 68, 114.
McLean, C.A., Cherny, R.A., Fraser, F.W., Fuller, S.J., Smith, M.J., Beyreuther, K., Bush, Nixon, R.A., Wegiel, J., Kumar, A., Yu, W.H., Peterhoff, C., Cataldo, A., Cuervo, A.M.,
A.I., Masters, C.L., 1999. Soluble pool of Abeta amyloid as a determinant of 2005. Extensive involvement of autophagy in Alzheimer disease: an immuno-
severity of neurodegeneration in Alzheimers disease. Ann. Neurol. 46, 466 electron microscopy study. J. Neuropathol. Exp. Neurol. 64, 113122.
860. Oddo, S., 2008. The ubiquitinproteasome system in Alzheimers disease. J. Cell.
McMillan, P.J., Kraemer, B.C., Robinson, L., Leverenz, J.B., Raskind, M., Schellenberg, Mol. Med. 12, 363373.
G., 2011. Truncation of tau at E391 promotes early pathologic changes in Oddo, S., Caccamo, A., Tseng, B., Cheng, D., Vasilevko, V., Cribbs, D.H., LaFerla, F.M.,
transgenic mice. J. Neuropathol. Exp. Neurol. 70, 10061019. 2008. Blocking Abeta42 accumulation delays the onset and progression of tau
Meyer-Luehmann, M., Coomaraswamy, J., Bolmont, T., Kaeser, S., Schaefer, C., Kilger, pathology via the C terminus of heat shock protein70-interacting protein: a
E., Neuenschwander, A., Abramowski, D., Frey, P., Jaton, A.L., Vigouret, J.M., mechanistic link between Abeta and tau pathology. J. Neurosci. 28, 12163
Paganetti, P., Walsh, D.M., Mathews, P.M., Ghiso, J., Staufenbiel, M., Walker, L.C., 12175.
Jucker, M., 2006. Exogenous induction of cerebral b-amyloidogenesis is gov- Ohm, T.G., Muller, H., Braak, H., Bohl, J., 1995. Close-meshed prevalence rates of
erned by agent and host. Science 313, 17811784. different stages as a tool to uncover the rate of Alzheimers disease-related
Mirra, S.S., Gearing, M., McKeel Jr., D.W., Crain, B.J., Hughes, J.P., van Belle, G., neurobrillary changes. Neuroscience 64, 209217.
Heyman, A., 1994. Inter-laboratory comparison of neuropathology assessments Oldo, S., Caccamo, A., Shepherd, J.D., Murphy, M.P., Golde, T.E., Kayed, R., Metherate,
in Alzheimers disease: a study of the Consortium to Establish a Registry for R., Mattson, M.P., Akbari, Y., LaFerla, F.M., 2003. Triple-transgenic model of
Alzheimers Disease (CERAD). J. Neuropathol. Exp. Neurol. 53, 303315. Alzheimers disease with plaques and tangles: intracellular Ab and synaptic
Mirra, S.S., Heyman, A., McKeel, D., Sumi, S.M., Crain, B.J., Brownlee, L.M., Vogel, F.S., dysfunction. Neuron 39, 409421.
Hughes, J.P., van Belle, G., Berg, L., 1991. The Consortium to Establish a Registry Ono, K., Yamada, M., 2011. Low-n oligomers as therapeutic targets of Alzheimers
for Alzheimers Disease (CERAD). Part II. Standardization of the neuropathologic disease. J. Neurochem. 117, 1928.
assessment of Alzheimers disease. Neurology 41, 479486. ONuallain, B., Freir, D.B., Nicoll, A.J., Risse, E., Ferguson, N., Herron, C.E., Collinge, J.,
Montine, T.J., Phelps, C.H., Beach, T.G., Bigio, E.H., Cairns, N.J., Dickson, D.W., Walsh, D.M., 2010. Amyloid b-protein dimers rapidly form stable synaptotoxic
Duyckaerts, C., Frosch, M.P., Masliah, E., Mirra, S.S., Nelson, P.T., Schneider, protobrils. J. Neurosci. 30, 1441114419.
J.A., Thal, D.R., Trojanowski, J.Q., Vinters, H.V., Hyman, B.T., 2012. National Ossenkoppele, R., van Berckel, B.N., Prins, N.D., 2011. Amyloid imaging in prodromal
Institute on aging-Alzheimers Association guidelines for the neuropathologic Alzheimers disease. Alzheimers Res. Ther. 3, 26.
assessment of Alzheimers disease: a practical approach. Acta Neuropathol. 123, Pamplona, R., Barja, G., 2007. Highly resistant macromolecular components and low
111. rate of generation of endogenous damage: two key traits of longevity. Ageing
Morales, R., Duran-Aniotz, C., Castilla, J., Estrada, L.D., Soto, C., 2011. De novo Res. Rev. 6, 189210.
induction of amyloid-b deposition in vivo. Mol. Psychiatry (epub ahead of Pamplona, R., Dalfo, E., Ayala, V., Bellmunt, M.J., Prat, J., Ferrer, I., Portero-Otin, M.,
print). 2005. Proteins in human brain cortex are modied by oxidation, glycoxidation,
Moreira, P.I., Santos, M.S., Oliveira, C.R., 2007. Alzheimers disease: a lesson from and lipoxidation: effects of Alzheimer disease and identication of lipoxidation
mitochondrial dysfunction. Antioxid. Redox Signal. 9, 16211630. targets. Mol. J. Biol. Chem. 280, 2152221530.
Morelli, L., Wei, L., Amorim, A., McDermid, J., Abee, C.R., Frangione, B., Walker, L.C., Pannesse, E., 2011. Morphological changes in nerve cells during normal aging. Brain
Levy, E., 1996. Cerebrovascular amyloidosis in squirrel monkeys and rhesus Struct. Funct. 216, 8589.
monkeys: apolipoprotein E genotype. FEBS Lett. 379, 132134. Perez, M., Ribe, E., Rubio, A., Lim, F., Moran, M.A., Ramos, P.G., Ferrer, I., Isla, M.T.,
Morgan, K., 2011. The three new pathways leading to Alzheimers disease. Neuro- Avila, J., 2005. Characterization of a double (amyloid precursor protein-tau)
pathol. Appl. Neurobiol. 37, 353357. transgenic: tau phosphorylation and aggregation. Neuroscience 130, 339
Mormino, Kluth, E.C., Madison, J.T., Rabinovici, C.M., Baker, G.D., Miller, S.L., Koeppe, 347.
B.L., Mathis, R.A., Weiner, C.A., Jagust, M.W., Alzheimers Disease Neuroimaging Perez-Gracia, E., Torrejon-Escribano, B., Ferrer, I., 2008. Dystrophic neurites of senile
Initiative, W.J., 2009. Episodic memory loss is related to hippocampal-mediated plaques in Alzheimers disease are decient in cytochrome c oxidase. Acta
beta-amyloid deposition in elderly subjects. Brain 132, 13101323. Neuropathol. 116, 261268.
Morris, J.C., Roe, C.M., Grant, E.A., Head, D., Storandt, M., Goate, A.M., Fagan, A.M., Peters, A., 1999. Normal aging in the cerebral cortex of primates. In: Peters, A.,
Holtzman, D.M., Mintun, M.A., 2009. Pittsburgh compound B imaging and Morrison, J.H. (Eds.), Cerebral Cortex: Vol. 14: Neurodegenerative and Age-
prediction of progression from cognitive normality to symptomatic Alzheimer Related Changes in Structure and Function of Cerebral Cortex. Kluwer Academ-
disease. Arch. Neurol. 66, 14691475. ic/Plenum, New York/Boston/Dordrecht/London/Moscow, pp. 4980.
Morris, J.C., Storandt, M., McKeel Jr., D.W., Rubin, E.H., Price, J.L., Grant, E.A., Berg, L., Peters, A., Sethares, C., Luebke, J.I., 2008. Synapses are lost during aging in the
1996. Cerebral amyloid deposition and diffuse plaques in normal aging: Evi- primate prefrontal cortex. Neuroscience 152, 970981.
dence for presymptomatic and very mild Alzheimers disease. Neurology 46, Peters, A., Sethares, C., Moss, M.B., 2010. How the primate fornix is affected by age. J.
707719. Comp. Neurol. 518, 39623980.
Morrison, J.H., Hof, P.R., 1997. Life and death of neurons in the aging brain. Science Price, D.L., Martin, L.J., Sisodia, S.S., Wagster, M.V., Koo, E.H., Walker, L.C., Koliatsos,
278, 412419. V.E., Cork, L.C., 1991. Aged non-human primates: an animal model of age-
Moss, M.B., Killiany, R.J., Herndon, J.G., 1999. Age-related cognitive decline in the associated neurodegenerative disease. Brain Pathol. 1, 287296.
rhesus monkey. In: Peters, A., Morrison, J.H. (Eds.), Cerebral Cortex: Vol. 14: Price, J.L., McKeel Jr., D.W., Buckles, V.D., Roe, C.M., Xiong, C., Grundman, M., Hansen,
Neurodegenerative and Age-Related Changes in Structure and Function of L.A., Petersen, R.C., Parisi, J.E., Dickson, D.W., Smith, C.D., Davis, D.G., Schmitt,
Cerebral Cortex. Kluwer Academic/Plenum, New York/Boston/Dordrecht/Lon- F.A., Markesbery, W.R., Kaye, J., Kurlan, R., Hulette, C., Kurland, B.F., Higdon, R.,
don/Moscow, pp. 2148. Kukull, W., Morris, J.C., 2009. Neuropathology of non-demented aging: pre-
Mueller, S.G., Schuff, N., Yaffe, K., Madison, C., Miller, B., Weiner, M.W., 2010. sumptive evidence for preclinical Alzheimer disease. Neurobiol. Aging 30,
Hippocampal atrophy patterns in mild cognitive impairment and Alzheimers 10261036.
disease. Hum. Brain Mapp. 31, 13391347. Price, J.L., Morris, J.C., 1999. Tangles and plaques in nondemented aging and
Mufson, E.J., Binder, L., Counts, S.E., DeKosky, S.T., deToledo-Morrel, L., Ginsberg, preclinical Alzheimers disease. Ann. Neurol. 45, 358368.
S.D., Ikonomovic, M.D., Perez, S.E., Scheff, S.W., 2012. Mild cognitive im- Puig, B., Gomez-Isla, T., Ribe, E., Cuadrado, M., Torrejon-Escribano, B., Dalfo, E.,
pairment: pathology and mechanisms. Acta Neuropathol. 123, 1330. Ferrer, I., 2004. Expression of stress-activated kinases c-Jun N-terminal kinase
Muntane, G., Dalfo, E., Martinez, A., Ferrer, I., 2008. Phosphorylation of tau and (SAPK/JNK-P) and p38 kinase (p38-P), and tau hyperphosphorylation in neur-
alpha-synuclein in synaptic-enriched fractions of the frontal cortex in Alzhei- ites surrounding betaA plaques in APP Tg2576 mice. Neuropathol. Appl. Neu-
mers disease, and in Parkinsons disease and related a-synucleinopathies. robiol. 30, 491502.
Neuroscience 152, 913923. Qiu, C., Winblad, B., Fratiglioni, L., 2005. The age-dependent relation of blood
Munoz, D.G., Ferrer, I., 2008. Neuropathology of hereditary forms of frontotemporal pressure to cognitive function and dementia. Lancet Neurol. 4, 487499.
dementia and parkinsonism. In: Duyckaerts, C., Litvan, I. (Eds.), Handbook of Reed, T., Perluigi, M., Sultana, R., Pierce, W.M., Klein, J.B., Turner, D.M., Coccia, R.,
Clinical Neurology: Dementias, Edinburgh/London/New York/Oxford/Philadel- Markesbery, W.R., Buttereld, D.A., 2008. Proteomic identication of 4-hy-
phia/St. Louis/Sydney/Toronto. Elsevier, pp. 393414. droxy-2-nonenal-modied brain proteins in amnestic mild cognitive im-
Murray, M.E., Graff-Radford, N.R., Ross, O.A., Petersen, R.C., Duara, R.C., Dickson, pairment: insight into the role of lipid peroxidation in the progression and
D.W., 2011. Neuropathologically dened subtypes of Alzheimers disease with pathogenesis of Alzheimers disease. Neurobiol. Dis. 30, 107120.
distinct clinical characteristics: a retrospective study. Lancet Neurol. 10, 785 Reitz, C., Brayne, C., Mayeux, R., 2011. Epidemiology of Alzheimer disease. Nat. Rev.
796. Neurol. 7, 137152.
50 I. Ferrer / Progress in Neurobiology 97 (2012) 3851

Rentz, D.M., Locascio, J.J., Becker, J.A., Moran, E.K., Eng, E., Buckner, R.L., Sperling, Sperling, R.A., Laviolette, P.S., OKeefe, K., OBrien, J., Rentz, D.M., Pihlajamaki, M.,
R.A., Johnson, K.A., 2010. Cognition, reserve, and amyloid deposition in normal Marshall, G., Hyman, B.T., Selkoe, D.J., Hedden, T., Buckner, R.L., Becker, J.A.,
aging. Ann. Neurol. 67, 353364. Johnson, K.A., 2009. Amyloid deposition is associated with impaired default
Ribe, E.M., Perez, M., Puig, B., Gich, I., Lim, F., Cuadrado, M., Sesma, T., Catena, S., network function in older persons without dementia. Neuron 63, 178188.
Sanchez, B., Nieto, M., Gomez-Ramos, P., Moran, M.A., Cabodevilla, F., Samar- Solomon, A., Kivipelto, M., Wolozin, B., Zhou, J., Whitmer, R.A., 2009. Midlife serum
anch, L., Ortiz, L., Perez, A., Ferrer, I., Avila, J., Gomez-Isla, T., 2005. Accelerated cholesterol and increased risk of Alzheimers and vascular dementia three
amyloid deposition, neurobrillary degeneration and neuronal loss in double decades later. Dement. Geriatr. Cogn. Disord. 28, 7580.
mutant APP/tau transgenic mice. Neurobiol. Dis. 20, 814822. St George-Hyslop, P.H., Tanzi, R.E., Polinsky, R.J., Haines, J.L., Nee, L., Watkins, P.C.,
Rosen, R.F., Fritz, J.J., Dooyema, J., Cintron, A.F., Hamaguchi, T., Lah, J.J., Levine 3rd, H., Myers, R.H., Feldman, R.G., Pollen, D., Drachman, D., et al., 1987. The genetic
Jucker, M., Walker, L.C., 2012. Exogenous seeding of cerebral b-amyloid depo- defect causing familial Alzheimers disease maps on chromosome 21. Science
sition in bAPP-transgenic rats. J. Neurochem. 120, 660666. 235, 885890.
Rub, U., Del Tredici, K., Schultz, C., Thal, D.R., Braak, E., Braak, H., 2000. The evolution Starkov, A.A., 2008. The role of mitochondria in reactive oxygen species metabolism
of Alzheimers disease-related cytoskeletal pathology in the human raphe and signaling. Ann. N. Y. Acad. Sci. 1147, 3752.
nuclei. Neuropathol. Appl. Neurobiol. 26, 553567. Sturchler-Pierrat, C., Abramowski, D., Duke, M., Wiederhold, K.H., Mistl, C., Rotha-
Salminen, A., Kauppinen, A., Suuronen, T., Kaarniranta, K., Ojala, J., 2009. ER stress in cher, S., Ledermann, B., Burki, K., Frey, P., Paganetti, P.A., Waridel, C., Calhoun,
Alzheimers disease: a novel neuronal trigger for inammation and Alzheimers M.E., Jucker, M., Probst, A., Staufenbiel, M., Sommer, B., 1997. Two amyloid
pathology. J. Neuroinammation 6, 4.1. precursor protein transgenic mouse models with Alzheimer disease-like pa-
Sandell, J.H., Peters, A., 2001. Effects of age on nerve bres in the rhesus monkey thology. Proc. Natl. Acad. Sci. U.S.A. 94, 1328713292.
optic nerve. J. Comp. Neurol. 466, 1430. Sultana, R., Buttereld, D.A., 2009. Oxidatively modied, mitochondria-relevant
Sandell, J.H., Peters, A., 2003. Disrupted myelin and axon loss in the anterior brain proteins in subjects with Alzheimer disease and mild cognitive im-
commissure of the aged rhesus monkey. J. Comp. Neurol. 466, 1430. pairment. J. Bioenerg. Biomembr. 41, 441446.
SantaCruz, K.S., Sonnen, J.A., Pezhouh, M.K., Desrosiers, M.F., Nelson, P.T., Tyas, S.L., Sultana, R., Buttereld, D.A., 2010. Role of oxidative stress in the progression of
2011. Alzheimer disease pathology in subjects without dementia in 2 studies of Alzheimers disease. J. Alzheimers Dis. 19, 341353.
aging: the Nun Study and the Adult Changes in Thought Study. J. Neuropathol. Sultana, R., Perluigi, M., Buttereld, D.A., 2009. Oxidatively modied proteins in
Exp. Neurol. 70, 832840. Alzheimers disease (AD), mild cognitive impairment and animal models of AD:
Schmidt, A.M., Sahagan, B., Nelson, R.B., Selmer, J., Rothlein, R., Bell, J.M., 2009. The role of Abeta in pathogenesis. Acta Neuropathol. 118, 131150.
role of RAGE in amyloid-beta peptide-mediated pathology in Alzheimers Sultana, R., Poon, H.F., Cai, J., Pierce, W.M., Merchant, M., Klein, J.B., Markesbery,
disease. Curr. Opin. Investig. Drugs 10, 672680. W.R., Buttereld, D.A., 2006. Identication of nitrated proteins in Alzheimers
Schonheit, B., Zarski, R., Ohm, T.G., 2004. Spatial and temporal relationships disease brain using a redox proteomics approach. Neurobiol. Dis. 22, 7687.
between plaques and tangles in Alzheimer-pathology. Neurobiol. Aging 25, Takashima, A., 2009. Amyloid-b, tau, and dementia. J. Alzheimers Dis. 17, 729
697711. 736.
Schultz, C., Dehghani, F., Hubbard, G.B., Thal, D.R., Struckhoff, G., Braak, E., Braak, H., Tanzi, R.E., 1999. A genetic dichotomy model for the inheritance of Alzheimers
2000a. Filamentous tau pathology in nerve cells, astrocytes, and oligodendro- disease and common age-related disorders. J. Clin. Invest. 104, 11751179.
cytes of aged baboons. J. Neuropathol. Exp. Neurol. 59, 3952. Tanzi, R.E., Bertram, L., 2005. Twenty years of the Alzheimers disease amyloid
Schultz, C., Hubbard, G.B., Rub, U., Braak, E., Braak, H., 2000b. Age-related progres- hypothesis: a genetic perspective. Cell 120, 545555.
sion of tau pathology in brains of baboons. Neurobiol. Aging 21, 905912. Terni, B., Boada, J., Portero-Otn, M., Pamplona, R., Ferrer, I., 2010. Mitochondrial
Selkoe, D.J., 2002. Alzheimers disease is a synaptic failure. Science 298, 789 ATP-synthase in the entorhinal cortex is a target of oxidative stress at stages I/II
791. of Alzheimers disease pathology. Brain Pathol. 20, 222233.
Serrano-Pozo, A., Frosch, M.P., Masliah, E., Hyman, B.T., 2011. Neuropathological Terry, R.D., Gonatas, N.K., Weiss, M., 1964. Ultrastructural studies in Alzheimers
alterations in Alzheimer disease. Cold Spring Harb. Perspest. Biol. 3, a006189. presenile dementia. Am. J. Pathol. 44, 269297.
Seshadri, S., Fitzpatrick, A.L., Ikram, M.A., DeStefano, A.L., Gudnason, V., Boada, M., Terry, R.D., Masliah, E., Salmon, D.P., Butters, N., DeTeresa, R., Hill, R., Hansen, L.A.,
Bis, J.C., Smith, A.V., Carassquillo, M.M., Lambert, J.C., Harold, D., Schrijvers, E.M., Katzman, R., 1991. Physical basis of cognitive alterations in Alzheimers disease:
Ramirez-Lorca, R., Debette, S., Longstreth Jr., W.T., Janssens, A.C., Pankratz, V.S., synapse loss is the major correlate of cognitive impairment. Ann. Neurol. 30,
Dartigues, J.F., Hollingworth, P., Aspelund, T., Hernandez, I., Beiser, A., Kuller, 572580.
L.H., Koudstaal, P.J., Dickson, D.W., Tzourio, C., Abraham, R., Antunez, C., Du, Y., Thal, D.R., Rub, U., Orantes, M., Braak, H., 2002. Phases of A beta-deposition in the
Rotter, J.I., Aulchenko, Y.S., Harris, T.B., Petersen, R.C., Berr, C., Owen, M.J., Lopez- human brain and its relevance for the development of AD. Neurology 58, 1791
Arrieta, J., Varadarajan, B.N., Becker, J.T., Rivadeneira, F., Nalls, M.A., Graff- 1800.
Radford, N.R., Campion, D., Auerbach, S., Rice, K., Hofman, A., Jonsson, P.V., Toledano, A., Alvarez, M.I., Lopez-Rodrguez, A.B., Toledano-Daz, A., Fernandez-
Schmidt, H., Lathrop, M., Mosley, T.H., Au, R., Psaty, B.M., Uitterlinden, A.G., Verdecia, C.I., 2011. Does Alzheimers disease exist in all primates? Alzheimer
Farrer, L.A., Lumley, T., Ruiz, A., Williams, J., Amouyel, P., Younkin, S.G., Wolf, pathology in non-human primates and its pathophysiological implications (II).
P.A., Launer, L.J., Lopez, O.L., van Duijn, C.M., Breteler, M.M., CHARGE Consor- Neurologia (epub ahead of print).
tium, GERAD1 Consortium, EADI1 Consortium, 2010. Genome-wide analysis of Tomiyama, T., Matsuyama, S., Iso, H., Umeda, T., Takuma, H., Ohnishi, K., Ishibashi,
genetic loci associated with Alzheimer disease. JAMA 303, 18321840. K., Teraoka, R., Sakama, N., Yamashita, T., Nishitsuji, K., Ito, K., Shimada, H.,
Shankar, G.M., Li, S., Metha, T.H., Garcia-Munoz, A., Shepardson, N.E., Smith, I., Brett, Lambert, M.P., Klein, W.L., Mori, H., 2010. A mouse model of amyloid beta
F.M., Farrell, M.A., Rowan, M.J., Lemere, C.A., Regan, C.M., Walsh, D.M., Sabatini, oligomers: their contribution to synaptic alteration, abnormal tau phosphor-
B.L., Selkoe, D.J., 2008. Amyloid-beta protein dimers isolated directly from ylation, glial activation, and neuronal loss in vivo. J. Neurosci. 30, 4845
Alzheimers brains impair synaptic plasticity and memory. Nat. Med. 14, 4856.
837842. Tseng, B.P., Green, K.N., Chan, J.L., Blurton-Jones, M., LaFerla, F.M., 2008. Ab inhibits
Shepherd, C., McCann, H., Halliday, G.M., 2009. Variations in the neuropathology of the proteasome and enhances amyloid and tau accumulation. Neurobiol. Aging
familial Alzheimers disease. Acta Neuropathol. 118, 3752. 29, 16071618.
Sherrington, R., Rogaev, E.I., Liang, Y., Rogaeva, E.A., Levesque, G., Ikeda, M., Chi, H., Tsuji, T., Shiozaki, A., Shimohama, S., 2005. The application of proteomics in
Lin, C., Li, G., Holman, K., Tsuda, T., Mar, L., Foncin, J.F., Bruni, A.C., Montesi, M.P., neurology. Curr. Proteomics 2, 4153.
Sorbi, S., Rainero, I., Pinessi, L., Nee, L., Chumakov, I., Pollen, D., Brookes, A., Uchikado, H., Lin, W.L., DeLucia, M.W., Dickson, D.W., 2006. Alzheimer disease with
Sanseau, P., Polinsky, R.J., Wasco, W., Da Silva, H.A., Haines, J.L., Perkicak-Vance, amygdala Lewy bodies: a distinct form of alpha-synucleinopathy. J. Neuro-
M.A., Tanzi, R.E., Roses, A.D., Fraser, P.E., Rommens, J.M., St George-Hyslop, P.H., pathol. Exp. Neurol. 65, 685697.
1995. Cloning of a gene bearing missense mutations in early-onset familial Utter, S., Tamboli, I.Y., Walter, J., Upadhaya, A.R., Birkenmeier, G., Pietrzik, C.U.,
Alzheimers disease. Nature 375, 754760. Ghebremedhin, E., Thal, D.R., 2008. Cerebral small vessel disease-induced
Simic, G., Stanic, G., Mladinov, M., Jovanov-Milosevic, N., Kostovic, I., Hof, P.R., 2009. apolipoprotein E leakage is associated with Alzheimer disease and the accu-
Does Alzheimers disease begin in the brainstem? Neuropathol. Appl. Neuro- mulation of amyloid beta-protein in perivascular astrocytes. J. Neuropathol.
biol. 35, 532554. Exp. Neurol. 67, 842856.
Singh-Manoux, A., Kivimaki, M., Glymour, M.M., Elbaz, A., Berr, C., Ebmeier, K.P., van Broeckhoven, C., Haan, J., Bakker, E., Hardy, J.A., Van Hul, W., Wehnert, A.,
Ferrie, J.E., Dugravot, A., 2011. Timing of onset of cognitive decline: results from Vegter-Van der Vlis, M., Roos, R.A., 1990. Amyloid beta protein precursor gene
Whitehall II prospective cohort study. Br. Med. J. 344, d7622. and hereditary cerebral hemorrhage with amyloidosis (Dutch). Science 248,
Sleegers, K., Brouwers, N., Gijselinck, I., Theuns, J., Goossens, D., Wauters, J., Del- 11201122.
Favero, J., Cruts, M., van Duijn, C.M., Van Broeckhoven, C., 2006. APP duplication van Helmond, Z., Miners, J.S., Kehoe, P.G., Love, S., 2010. Oligomeric Ab in Alzhei-
is sufcient to cause early onset Alzheimers dementia with cerebral amyloid mers disease: relationship to plaque and tangle pathology, APOE genotype and
angiopathy. Brain 129, 29772983. cerebral amyloid angiopathy. Brain Pathol. 20, 468480.
Small, S.A., Duff, K., 2008. Linking Ab and tau in late-onset Alzheimers disease: a van Leeuwen, F.W., de Kleijn, D.P., van den Hurk, H.H., Neubauer, A., Sonnemans,
dual hypothesis. Neuron 60, 534542. M.A., Sluijs, J.A., Koycu, S., Ramdjielal, R.D., Salehi, A., Martens, G.J., Grosveld,
Sperling, R.A., Aisen, P.S., Beckett, L.A., Bennett, D.A., Craft, S., Fagan, A.M., Iwatsubo, F.G., Peter, J., Burbach, H., Hol, E.M., 1998. Frameshift mutants of b-amyloid
T., Jack Jr., C.R., Kaye, J., Montine, T.J., Park, D.C., Reiman, E.M., Rowe, C.C., precursor protein and ubiquitin-B in Alzheimers and Down patients. Science
Siemers, E., Stern, Y., Yaffe, K., Carrillo, M.C., Thies, B., Morrison-Bogorad, M., 279, 242247.
Wagster, M.V., Phelps, C.H., 2011. Toward dening the preclinical stages of Vemuri, P., Wiste, H.J., Weigand, S.D., Knopman, D.S., Trojanowski, J.Q., Shaw, L.M.,
Alzheimers disease: recommendations from the National Institute on Aging Bernstein, M.A., Aisen, P.S., Weiner, M., Petersen, R.C., Jack Jr., C.R., Alzheimers
Alzheimers Association workgroups on diagnostic guidelines for Alzheimers Disease Neuroimaging Initiative, 2010. Serial MRI and CSF biomarkers in normal
disease. Alzheimers Dement. 7, 280292. aging, MCI, and AD. Neurology 75, 143151.
I. Ferrer / Progress in Neurobiology 97 (2012) 3851 51

Vinters, H.V., Ellis, W.G., Zarow, C., Zaias, B.W., Jagust, W.J., Mack, W.J., Chui, H.C., Wilson, A.C., Dugger, B.N., Dickson, D.W., Wang, D.S., 2011. TDP-43 in aging and
2000. Neuropathologic substrates of ischemic vascular dementia. J. Neuro- Alzheimers diseasea review. Int. J. Clin. Exp. Pathol. 4, 147155.
pathol. Exp. Neurol. 59, 931945. Woodruff-Pak, D.S., 2008. Animal models of Alzheimers disease: therapeutic
Vogt, B.A., Vogt, L.J., Hof, P.R., 2001. Patterns of cortical neurodegeneration in Alzhei- implications. J. Alzheimers. Dis. 15, 507521.
mers disease: subgroups, subtypes, and implications for staging strategies. In: Hof, Yao, J., Irwin, R.W., Zhao, L., Nilsen, J., Hasmilton, R.T., Diaz Brinton, R., 2009.
P.R., Mobbs, C.V. (Eds.), Functional Neurobiology of Aging. Academic Press, San Mitochondrial bioenergic decit precedes alzhiemrs pathology in female
Diego/San Francisco/New York/Boston/London/Sydney/Tokyo, pp. 111130. mouse model of Alzheimers disease. Proc. Natl. Acad. Sci. U.S.A. 106, 14670
Walker, L.C., Callahan, M.J., Bian, F., Durham, R.A., Roher, A.E., Lipinski, W.J., 2002. 14675.
Exogenous induction of cerebral beta-amyloidosis in betaAPP-transgenic mice. Yu, W.H., Cuervo, A.M., Kumar, A., Peterhoff, C.M., Schmidt, S.D., Lee, J.H., Mohan,
Peptides 23, 12411247. P.S., Mercken, M., Farmery, M.R., Tjernberg, L.O., Jiang, Y., Duff, K., Uchiyama, Y.,
Walker, L.C., Levine, H., Mattson, M.P., Jucker, M., 2006. Inducible proteopathies. Naslund, J., Mathews, P.M., Cataldo, A.M., Nixon, R.A., 2005. Macroautophagya
Trends Neurosci. 29, 438443. novel b-amyloid peptide-generating pathway activated in Alzheimers disease.
Wan, L., Nie, G., Zhang, J., Luo, Y., Zhang, P., Zhang, Z., Zhao, B., 2011. b-Amyloid J. Cell. Biol. 171, 8798.
peptide increases levels of iron content and oxidative stress in human cell and Yu, C.H., Song, G.S., Yhee, J.Y., Kim, J.H., Im, K.S., Nho, W.G., Lee, J.H., Sur, J.H., 2011.
Caenorhabditis elegans models of Alzheimer disease. Free Radic. Biol. Med. 50, Histopathological and immunohistochemical comparison of the brain of human
122129. patients with Alzheimers disease and the brain of aged dogs with cognitive
Wang, Y.P., Biernat, J., Pickhardt, M., Mandelkow, E., Mandelkow, E.M., 2007. dysfunction. J. Comp. Pathol. 145, 4558.
Stepwise proteolysis liberates tau fragments that nucleate the Alzheimer-like Zarow, C., Sitzer, T.E., Chui, H.C., 2008. Understanding hippocampal sclerosis in the
aggregation of full-length tau in a neuronal cell model. Proc. Natl. Acad. Sci. elderly: epidemiology, characterization, and diagnostic issues. Curr. Neurol.
U.S.A. 104, 1025210257. Neurosci. Rep. 8, 363370.
Wang, Y., Garg, S., Mandelkow, E.M., Mandelkow, E., 2010. Proteolytic processing of Zhang, B., Higuchi, M., Yoshiyama, Y., Ishihara, T., Forman, M.S., Martinez, D., Joyce,
tau. Biochem. Soc. Trans. 38, 955961. S., Trojanowski, J.Q., Lee, V.M., 2004. Retarded axonal transport of R406W
Weller, R.O., Subash, M., Preston, S.D., Mazanti, I., Carare, R.O., 2008. Perivascular mutant tau in transgenic mice with a neurodegenerative tauopathy. J. Neurosci.
drainage of amyloid-beta peptides from the brain and its failure in cerebral 24, 46574667.
amyloid angiopathy and Alzheimers disease. Brain Pathol. 18, 253266. Zilkova, M., Zilka, N., Kovac, A., Kovacech, B., Skrabana, R., Skrabanova, M., Novak, M.,
White, L., 2009. Brain lesions at autopsy in older Japanese-American men as related 2011. Hyperphosphorylated truncated protein tau induces caspase-3 indepen-
to cognitive impairment and dementia in the nal years of life: a summary dent apoptosis-like pathway in the Alzheimers disease cellular model. J.
report from the Honolulu-Asia aging study. J. Alzheimers Dis. 18, 713725. Alzheimers Dis. 23, 161169.

You might also like