Vous êtes sur la page 1sur 16

Please cite this article in press as: Bartsch T, Wulff P.

The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084

Neuroscience xxx (2015) xxxxxx

EDITORIAL
THE HIPPOCAMPUS IN AGING AND DISEASE: FROM PLASTICITY
TO VULNERABILITY
T. BARTSCH a* AND P. WULFF b! Key words: hippocampus, vulnerability, neuroplasticity, sub-
a field imaging, Alzheimers disease, aging.
Department of Neurology, Memory Disorders and Plasticity
Group, University Hospital Schleswig-Holstein, Kiel, Germany
b
Institute of Physiology, Neurophysiology, University of
Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
INTRODUCTION

AbstractThe hippocampus has a pivotal role in learning


The hippocampus is widely regarded as being in the
and in the formation and consolidation of memory and is center of a brain network supporting encoding,
critically involved in the regulation of emotion, fear, anxiety, consolidation and retrieval of memory and, being central
and stress. Studies of the hippocampus have been central to to the study of human memory, has been implicated in
the study of memory in humans and in recent years, the episodic and semantic long-term memory, novelty
regional specialization and organization of hippocampal detection, sleep-dependent memory consolidation,
functions have been elucidated in experimental models pattern discrimination, spatial navigation and the binding
and in human neurological and psychiatric diseases. The of temporally and spatially distributed representations
hippocampus has long been considered a classic model (Bartsch, 2012). Beyond these cognitive functions, the
for the study of neuroplasticity as many examples of synap-
hippocampus is also involved in the regulation of emotion,
tic plasticity such as long-term potentiation and -depression
have been identified and demonstrated in hippocampal cir-
fear, anxiety, and stress. The hippocampus has an
cuits. Neuroplasticity is the ability to adapt and reorganize intriguing cyto- and network architecture and it has been
the structure or function to internal or external stimuli and suggested that the particular circuit arrangement in dier-
occurs at the cellular, population, network or behavioral ent subregions of the hippocampus subserves dierential
level and is reflected in the cytological and network architec- mnemonic operations (Kesner and Rolls, 2015). Indeed,
ture as well as in intrinsic properties of hippocampal in recent years, a dierential and complex modular orga-
neurons and circuits. The high degree of hippocampal neu- nization of hippocampal anatomy and function emerged
roplasticity might, however, be also negatively reflected in (Strange et al., 2014). Also, the concept of a regional
the pronounced vulnerability of the hippocampus to delete- specialization and organization of hippocampal functions
rious conditions such as ischemia, epilepsy, chronic stress,
has been increasingly studied in humans using high-
neurodegeneration and aging targeting hippocampal
structure and function and leading to cognitive deficits.
resolution MR subfield imaging indeed showing that
Considering this framework of plasticity and vulnerability, mnemonic operations can be attributed to subnetworks
we here review basic principles of hippocampal anatomy and subregions of the hippocampus (Bakker et al.,
and neuroplasticity on various levels as well as recent find- 2008; Mueller et al., 2011) (Chetelat, this issue).
ings regarding the functional organization of the hippocam- The hippocampus has long been considered as a
pus in light of the regional vulnerability in Alzheimers classic example for the study of functional
disease, ischemia, epilepsy, neuroinflammation and aging. neuroplasticity as many models of synaptic plasticity
This article is part of a Special Issue entitled: such as long-term potentiation (LTP) and -depression
Hippocampus. ! 2015 IBRO. Published by Elsevier Ltd. All (LTD), and spike-timing-dependent plasticity have been
rights reserved.
observed in hippocampal circuits and are thought to be
fundamental to learning and memory (Bliss and
Schoepfer, 2004; Pastalkova et al., 2006). Neuroplasticity
*Corresponding author. Tel: +49-0-431-597-8550; fax: +49-0-431- is considered the ability to adapt and reorganize the
597-8502. structure or function to internal or external stimuli and
E-mail addresses: t.bartsch@neurologie.uni-kiel.de (T. Bartsch), occurs on the cellular, population, network or behavioral
p.wul@physiologie.uni-kiel.de (P. Wul).
!
Tel: +49-431-880-2806; fax:+49-431-880-4580.
level (Cramer et al., 2011). Neuroplastic mechanisms
Abbreviations: Ab, amyloid-beta; AD, Alzheimers disease; DG, are reflected in the cyto- and network architecture and
dentate gyrus; EAE, experimental autoimmune encephalomyelitis; are mirrored in the intrinsic properties of hippocampal
EC, entorhinal cortex; IL-1b, interleukin-1 beta; LTD, long-term neurons and circuits. Structural plasticity in hippocampal
depression; LTP, long-term potentiation; MS, multiple sclerosis; MTL,
medial temporal lobe; NFT, neurofibrillary tangle; ROS, reactive neurons and circuits includes modifications of dendritic
oxygen species; TLE, temporal lobe epilepsy. tree size and spines, synapse number as well as the

http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
0306-4522/! 2015 IBRO. Published by Elsevier Ltd. All rights reserved.

1
2 T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx

formation of new neurons (Leuner and Gould, 2010). Cel- lucidum, stratum radiatum). The structure of this feed-
lular neuroplasticity is not confined to physiology but also forward circuit with its limited redundancy may be critical
present in the context of progressive pathology, such as for learning and memory but may also contribute to its vul-
neurodegeneration in Alzheimers disease (AD) in nerability during insults (Lavenex and Amaral, 2000).
humans and is increasingly studied (Mufson et al., Learning and memory processes within hippocampal
2015). On a network level, neuroplasticity in hippocampal circuits are regulated by synaptic plasticity mechanisms
circuits drives changes in connectivity, structural modifi- that require activation of specific molecular cascades
cations and behavioral outcome (Finke et al., 2013a; (Aksoy-Aksel and Manahan-Vaughan, 2015). For exam-
Ryan et al., 2015). ple, the induction of LTP in the CA1 region involves post-
This high degree of hippocampal plasticity, however, synaptic calcium ion entry via NMDA receptors with
is accompanied by the pronounced vulnerability of the subsequent activation of protein kinases (Aksoy-Aksel
hippocampus to deleterious conditions such as and Manahan-Vaughan, 2015).
ischemia, epilepsy, neuroinflammation, chronic stress, The hippocampus receives large amounts of sensory
neurodegeneration and aging suggesting that the information from neocortical structures, which is
instrinsic properties of hippocampal neurons and circuits integrated in several recurrent subnetworks with distinct
that are critical for neuroplasticity such as glutamatergic computational operations (van Strien et al., 2009). The
excitability may also predispose to metabolic injuries DG with its three layers (molecular, granular, and polymor-
occurring in the process of various neurological and phic layers) consists mainly of granule cells and receives
psychiatric diseases (Bartsch et al., 2015). This view is polymodal input form the EC. The axons of the DG granule
reflected in the suggestion by Bruce McEwen that the cells form the mossy fiber system and project to CA3
plasticity of the hippocampus is the reason for its vulner- (Amaral et al., 2007). Mossy fibers also project back onto
ability (McEwen, 1994). granule cells, thus forming a recurrent network. In addition
In this special issue of Neuroscience: The the DG receives information from the contralateral hip-
hippocampus in aging and disease: from plasticity to pocampus via commissural projections (Amaral et al.,
vulnerability, we will review basic principles of 2007). Axon collaterals of CA3 pyramidal neurons synapse
hippocampal anatomy and neuroplasticity on various onto other CA3 neurons, forming a recurrent autoassocia-
levels as well as recent findings regarding its functional tive network whereas CA3 neurons projecting back to the
organization with respect to regional vulnerability, which dentate network form a heteroassociative network
is critical for the understanding of neurocognitive (Lisman, 1999). CA1 pyramidal neurons receive informa-
diseases (Bartsch, 2012). tion which has been pre-processed in the subnetworks of
the DG and CA3, but also receives direct projections from
the EC suggesting that the function of CA1 neurons
HIPPOCAMPAL ANATOMY includes comparing new information from the EC with
stored information via CA3 in terms of mismatch, error
Encoding, consolidation and retrieval of mnemonic and novelty detection (Lisman and Otmakhova, 2001).
information is critically dependent on a large reciprocal Besides the structural layout of connectivity of these
network of regions that includes neocortical association hippocampal circuits, it is believed that mnemonic
regions, subcortical nuclei, the medial temporal lobe information is represented and processed by spatio-
(MTL), parahippocampal areas and the hippocampal temporal patterns of firing in groups of neurons, also
formation (Fig. 1). The hippocampus is considered the referred to as cell assemblies involving fast oscillatory
central node in this circuit. It receives input from dynamics in various frequency bands (e.g. theta, sharp-
almost all neocortical association areas via perirhinal wave ripples, slow oscillations and gamma oscillations)
and parahippocampal cortices and finally through the that are synchronized and temporally coordinated within
entorhinal cortex (EC) (van Strien et al., 2009). The hip- and across cortical regions (Wul et al., 2009; Battaglia
pocampus is a three-layered allocortical structure that is et al., 2011; Buzsaki and Moser, 2013).
reciprocally connected to other cortical and subcortical
areas (Figs. 1 and 2). The principal neurons of the hip-
NEUROPLASTICITY AND THE HIPPOCAMPUS
pocampus are organized in layers and receive unidirec-
tional polymodal input from the EC, where layer II
neurons project via the perforant path to granule cells in
Neuroplasticity can be broadly defined as the ability of the
the dentate gyrus (DG) (Strange et al., 2014). The trisy- nervous system to respond to intrinsic and extrinsic stimuli
naptic pathway from the DG to CA3 via mossy fibers by reorganizing its structure, function and connections; can
and onward to CA1 via Schaer collaterals is the principal be described at many levels, from molecular to cellular to sys-
feed-forward circuit involved in the processing of informa- tems to behavior; and can occur during development, in
tion through the hippocampus (Fig. 2). Additionally, layer response to the environment, in support of learning, in
III neurons from the EC directly project to CA1 neurons response to disease, or in relation to therapy. Such plasticity
via the temporoammonic path (perforant path to CA1). can be viewed as adaptive when associated with a gain in
CA1 pyramidal cells -the major output relay neurons function or as maladaptive when associated with negative
project via the subicular complex back to deep layers of consequences such as loss of function or increased injury,
the EC and to various subcortical and cortical areas points illustrated by animal models and some human studies.
(Murray et al., 2011). The regions CA1CA3 are sepa-
rated into four layers (pyramidal, stratum oriens, stratum (Cramer et al., 2011).

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx 3

Fig. 1. (A) Principal anatomy of the hippocampal memory systems and the brain regions involved in learning and memory. (B) Connectivity map of
critical memory structures of the temporal lobe system, diencephalic nuclei and neocortical association areas highlighting the reciprocal information
flow. Abbreviations: Ant, anterior thalamic nuclei; CA, cornu ammonis; DG, dentate gyrus; EC, entorhinal cortex; MB, mamillary bodies; Med, medial
thalamic nuclei; MTT, mamillothalamic tract (bundle of Vicq dAzyr); SN, septal nuclei.

reciprocal interaction between neural structure and


function (Kolb and Muhammad, 2014). Neuroplasticity
can occur on various functional and structural levels from
changes in membrane excitability, synaptic plasticity to
changes in dendritic and axonal structure (Kolb and
Gibb, 2014). Neuroplasticity can also occur as structural
and functional adaptations and reorganization of neuronal
populations, which is reflected in modifications of recruit-
ment and strength of connectivity of networks and circuits.
Finally, neuroplasticity can occur on a phenomenological
level mirrored in changes and modifications of cognitive
and behavioral strategies (Cramer et al., 2011) (Fig. 4).
Neuroplasticity can show a time- and age-dependency
and can result in positive adaptive or negative (maladap-
tive) eects (Kolb and Gibb, 2014).
The hippocampus has long been considered a classic
example for the study of neuroplasticity as many
paradigms of synaptic plasticity such as LTP and LTD,
EPSP-spike potentiation and spike-timing-dependent
Fig. 2. (A) Intrinsic anatomy of the hippocampus depicting the
trisynaptic pathway as the principal feed forward neural circuit
plasticity, which are thought to represent cellular
involved in the processing of information through the hippocampus. correlates of learning and memory, have been identified
(B) Flow diagram of the information flow through the hippocampus and demonstrated in hippocampal circuits. Similarly,
highlighting the parallel processing of information as well as the structural plasticity in response to external stimulation
reciprocal auto- and heteroassociative networks of the DG and CA3.
such as modifications of dendritic tree size, number of
Abbreviations: CA, cornu ammonis; DG, dentate gyrus; EC, entorhi-
nal cortex. dendritic spines and number of synapses as well as the
formation of new neurons have been demonstrated in
the hippocampus (Leuner and Gould, 2010). Mechanisms
Neuroplasticity is the ability of the brain or brain of plasticity in the hippocampus are also aected during
structures to adapt in response to intrinsic or extrinsic the early stages of AD during which neurodegenerative
stimuli such as changes in the environment, changes of extracellular amyloid-beta (Ab) deposition
development or lesions and consists in principle of a and intracellular neurofibrillary tangle (NFT) formation

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
4 T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx

accounts for molecular synaptic and intraneuronal remod- Recent findings also indicate that markers of plasticity
eling (Mufson et al., 2015). Axonal sprouting is another such as LTP as well as the formation of new neurons in
example of structural plasticity after damage that may the DG are dierentially organized in the dorsal and ven-
result in maladaptive hippocampal circuit reorganization. tral hippocampus thus (Wu and Hen, 2014; Opendak and
On the molecular level, plasticity can be driven by Gould, 2015). Furthermore, on a cellular level, this
enhanced expression of plasticity-related genes, such dichotomic organization also correlates to a dierential
as BDNF, CaMKII, CREB as well as increased surface expression of NMDA and AMPA receptor subunits in the
expression of glutamatergic AMPA and NMDA receptors. dorsal and ventral hippocampus of the rat (Pandis et al.,
Activity-dependent neuroplastic mechanisms in the hip- 2006).
pocampus that are fundamental to learning and memory This dichotomic view of the functional organization is
such as LTP (Pastalkova et al., 2006) can be modified currently being replaced by a more dierentiated concept
by preceding synaptic activity. This type of regulatory as some functional dierences are organized in a more
mechanism of plasticity itself by prior activity is called gradient-like way along the longitudinal axis. The
metaplasticity and has been implicated not only in physi- distribution of NMDA receptors, the connectivity to
ological regulation but also in aging and disease mecha- cortical and subcortical structures, the size of so called
nisms (Hulme et al., 2013; Jones, 2015). On the place fields suggest a gradual longitudinal axis
network level, plastic changes in the hippocampal mem- distribution whereas genomic expression analyses
ory system to a perturbation in terms of a network reorga- demarcate CA1 and DG into three (ventral, intermediate
nization has been shown in patients with hippocampal and dorsal) and CA3 into nine modules (Thompson
damage where a recruitment of an extended et al., 2008). This arrangement suggests a superimposed
hippocampal-neocortical network compensating for hip- pattern of gradient-like as well as a modular organization
pocampal dysfunction could be observed (Finke et al., along the anterior-posterior axis of the hippocampus
2013a). (Strange et al., 2014). However, it is currently unclear,
In this Special Issue of Neuroscience, the concept of how the anatomical organization, in particular the molecu-
neuroplasticity has been applied to various neurological lar expression patterns, can be reconciled with a functional
and psychiatric diseases as well as aging including AD modular organization along the anterior-posterior axis.
(Mufson et al., 2015; de Flores et al., 2015a), depression
(Malykhin and Coupland, 2015), schizophrenia Functional subfield organization
(Tamminga and Suzanne Zukin, 2015) aging (Gray and
Barnes, 2015), epilepsy (Sierra et al., 2015; Blumcke The hippocampus is organized into dierent cytoar-
and Coras, 2015), neurodevelopmental disorders (van chitectonic subfields that include the cornu ammonis
der Marel et al., 2015), metabolic disorders (Stranahan, fields (CA1CA4), the DG, and the subiculum.
2015), neuroinflammation (Barrientos et al., 2015; Heine Considering the intriguing anatomy of the hippocampal
et al., 2015) including multiple sclerosis (MS) (Novkovic subnetworks with its recurrent associative networks
et al., 2015). embedded into the unidirectional trisynaptic pathway but
with parallel projections from the EC to the CA3 and
CA1 region, it has been suggested that these networks
REGIONAL SPECIALIZATION AND perform dierential computations in dierent cognitive
ORGANIZATION OF HIPPOCAMPAL tasks (Marr, 1971; OReilly and McClelland, 1994;
FUNCTIONS Kesner and Rolls, 2015). Using animal models, an activa-
tion of these hippocampal subnetworks has been corre-
Hippocampal organization along the longitudinal axis
lated with distinct cognitive or mnemonic operations
Earlier experimental models of hippocampal function thereby reflecting distinct electrophysiological- and
regarded the hippocampus as a unitary functional and neuroplastic-properties of subfield neurons (Chen et al.,
structural entity. With a greater refinement of 2009). For example, it has been argued that the DG dur-
hippocampal circuit anatomy in recent years, however, a ing encoding is involved in pattern separation a cogni-
regional specialization of hippocampal functions along tive function whereby a discrimination of similar and
the longitudinal axis emerged (Small, 2002). Animal data overlapping neuronal patterns into more dissimilar pat-
using behavioral lesion models, studies of intrahippocam- terns is performed using minimal dierences in the given
pal connectivity and electrophysiological findings suggest cues (Guzowski et al., 2004; Yassa and Stark, 2011;
a dichotomic organization of hippocampal networks into Neunuebel and Knierim, 2014). The separation of pat-
functional compartments such as the dorsal hippocampus terns on the basis of similarity is thought to play a role
performing cognitive functions, in particular spatial pro- in the non-ambiguous and distinct representation of simi-
cessing, and the ventral hippocampus being involved in lar memories in the context of learning and memory, pre-
the regulation of stress, emotion, and aect (Moser and venting memory interference and allowing novelty
Moser, 1998; Bannerman et al., 2004; Thompson et al., detection (Bakker et al., 2008). It is also thought that pat-
2008; Dong et al., 2009; Fanselow and Dong, 2010). This tern separation is impaired in aging and mild cognitive
distinction was partly derived from findings that the ventral impairment (Yassa et al., 2011). The autoassociative
hippocampus has stronger connectivity with the amyg- CA3 circuitry is thought to be involved in binding of disso-
dala, hypothalamic and endocrine nuclei as well as the ciative representations and pattern completion, a mnemo-
critical role of the ventral hippocampus in the stress nic function which allows the recall of a memory by only
response (Kjelstrup et al., 2002; Strange et al., 2014). partial or fragmented representation of cues that were

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx 5

Fig. 3. High-resolution MR-imaging of the hippocampus with an analysis of hippocampal subfields.

present during initial learning. Both operations are linked dierential patterns of pathological changes (Mueller et al.,
together, as pattern separation in the DG is a prerequisite 2007; Mueller and Weiner, 2009). These imaging patterns
for pattern completion in CA3 (Lisman, 1999). The CA3 are currently evaluated in terms of developing imaging
network has also been implied in spatial rapid one-trial biomarkers for disease and treatment outcomes
learning and spatial sequence learning. The CA1 network (Malykhin and Coupland, 2015; Sierra et al., 2015).
receives input from CA3 and the EC and has been
thought to be engaged in input integration and match-
mismatch detection between predictions from CA3 and REGIONAL VULNERABILITY OF THE
perceptual input from the EC and novelty detection HIPPOCAMPUS
(Rolls and Kesner, 2006; Hunsaker and Kesner, 2013).
Acute damage to the hippocampus
The CA1 region is also involved in the integration of tem-
poral and object representations. Behaviorally, it has For over 100 years it has been known that acute
been shown that CA1 is critical for autobiographical pathological conditions, such as ischemia, hypo-
memory retrieval (Kesner and Hunsaker, 2009; Bartsch glycemia, epileptic seizures and other neurological
et al., 2011). Furthermore, the CA1 subregion is involved conditions can cause damage to the hippocampus
in our ability to learn a map-like representation of an envi- (Michaelis, 2012). This damage of the hippocampus is
ronment which is in accordance with the cognitive-map most evident in CA1 hippocampal neurons and is a reflec-
theory, which suggests that allocentric spatial representa- tion of a selective vulnerability of the hippocampus to
tions of locations are processed in the hippocampus acute conditions impairing the metabolic homeostasis of
(Bartsch et al., 2010). CA1 pyramidal neurons to glutamate-dependent and
Only recently, with the emergence of high-field MR calcium-mediated mechanisms of neuronal cytotoxicity
scanning and new imaging protocols it has been (Schmidt-Kastner and Freund, 1991; Bartsch et al.,
possible to shed light on the longitudinal organization of 2006, 2008). A brief episode of cerebral ischemia results
the hippocampus and to correlate dierential in the selective cell death of hippocampal CA1 neurons
hippocampal subfield activation to dierent mnemonic whereas the DG, CA3, and most cortical neurons appear
functions in humans (Fig. 3) (Small, 2002; Das et al., to be more resistant. The structural changes in CA1 that
2012; Engvig et al., 2012; Poppenk et al., 2013). High- follow the insult are most prominent after a delay of
resolution functional and volumetric imaging studies indi- 34 days and thus have been termed delayed neuronal
cate a dierential involvement of hippocampal subfields death (Pulsinelli et al., 1982; Schmidt-Kastner and
in memory and learning showing that CA3 and the DG Freund, 1991; Kirino, 2000). The phenotype, evolution
are involved in patterns separation, memory encoding and pattern of lesions in the hippocampal CA1 region in
and early retrieval, and an involvement of CA1 in late acute neurological disorders was recently studied using
retrieval, consolidation, spatial encoding, autobiographical high-resolution MR-imaging (Bartsch et al., 2015). In
retrieval and recognition (Bakker et al., 2008; Suthana patients with hippocampal ischemia, neuroinflammation,
et al., 2009; Mueller et al., 2011; Bonnici et al., 2012, after status epilepticus and transient global amnesia, the
2013; Deuker et al., 2014). These high-resolution imaging CA1 region was selectively aected compared to other
methods have been applied to neurological, neurodegen- cornu ammonis (CA-) regions of the hippocampus. The
erative and psychiatric diseases as well as aging showing maximum impairment of CA1 cellular metabolism as

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
6 T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx

Fig. 4. Schematic overview of the various contributing factors, dierent scales and methodological and conceptual levels determining
neuroplasticity in the hippocampus.

measured by apparent diusion coecient (ADC) in MRI numbers, dendritic branching, and dendritic spine density
was seen 48 h-72 h after the insult, irrespective of the nat- are not substantially reduced during the aging process
ure of the insult. Hypoxic-ischemic insults led to a signifi- (West et al., 1994; Lister and Barnes, 2009). Also, bio-
cant stronger reduction of ADC suggesting that the physical properties of hippocampal neurons such as the
ischemic insult results in a stronger impairment of CA1 resting membrane potential, membrane time constant,
metabolism compared to inflammation and epilepsy input resistance, threshold to action potential are largely
(Bartsch et al., 2015). These results show that acute dam- preserved over the lifespan (Burke and Barnes, 2010).
age to the hippocampus selectively aects the CA1 sub- There are, however, observations of synaptic loss in the
region, a region that is highly plastic. In contrast, CA3 layer II entorhinal cortical input to the granule cells in
and the DG are only marginally aected. The cellular the DG (Burke and Barnes, 2010). This change in con-
mechanisms that lead to a regional vulnerability to a nectivity may aect the function of hippocampal subnet-
glutamate- and calcium-mediated excitotoxicity are possi- works for example during pattern separation (Burke and
bly a reflection of genomic-dependent regional dierences Barnes, 2010). Synaptic plasticity such as the induction
in the distribution of glutamatergic NMDA receptors in and maintenance of LTP in line with alterations in
CA1 neurons that express considerably higher densities Ca2+ homeostasis changes with age (Lister and
of NMDA NR2 subunits than do CA3 or DG neurons, as Barnes, 2009). Along these lines, subregion-specific
reported for rodents (Coultrap et al., 2005; Gee et al., changes in the expression of genes subserving the regu-
2006; Quintana et al., 2006; Newrzella et al., 2007; lation of neuroplasticity such as LTP have been observed
Butler et al., 2010). Furthermore, regional dierences in in the aging hippocampus (Burger, 2010). Furthermore,
antioxidant enzymes as well as inflammatory reactions cognitive changes during aging can also be attributed to
may play a role in enhanced vulnerability of the CA1 changes in the neural dynamics of hippocampal circuits,
region (Wang and Michaelis, 2010; Michaelis, 2012). e.g. in place cell maintenance, stability and plasticity
Interestingly, hippocampal interneurons are believed to during remapping and pattern separation processes
be more resistant to the impact of transient ischemia than (Burke and Barnes, 2010). Neurogenesis in the DG also
larger pyramidal neurons (Harry and Lefebvre decreases with age, which may contribute to the changes
dHellencourt, 2003; Avignone et al., 2005). in neuroplasticity over the lifespan (Olariu et al., 2007). In
humans, histo-pathological studies confirm that CA1 does
not show a pronounced cell loss during aging, an obser-
Aging and the hippocampus vation that was confirmed in high-resolution subfield
Neuroplasticity mechanisms in the hippocampus sub- imaging (La Joie et al., 2010) whereas other imaging stud-
serving cognitive functions are also subject to aging ies did show an eect of aging on subfield volume
processes and it has been suggested that neuroplastic (Mueller and Weiner, 2009; Shing et al., 2011; Wisse
mechanisms are particularly vulnerable during aging et al., 2014), probably depending on study cohort and
(Burke and Barnes, 2006). Interestingly, principal cell imaging methodology.

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx 7

CHRONIC INFLUENCES ON THE modifications of hippocampal functions (Bergmann and


HIPPOCAMPUS AND DURING AGING Frisen, 2013). The process of adult neurogenesis is sub-
ject to various internal and external modulatory influences
In chronic diseases such as neurodegenerative diseases, during physiological and pathophysiological states (Voss
chronic epilepsy or neuropsychiatric disorders, the time et al., 2013). It has been suggested that adult neurogene-
course of pathological changes is much slower than with sis contributes to the plastic adaptation of learning and
e.g. acute ischemia and might be more complex as memory in complex environments as an impairment of
multiple pathological pathways overlap. Furthermore, in neurogenesis correlates with reduced hippocampus-
contrast to acute lesions to the hippocampus, the dependent memory performance (Drapeau et al., 2003).
regional vulnerability of the hippocampus may shift to One process that has been suggested to play a role in
other hippocampal structures such as the DG or the adaptive behavior is pattern separation, during which sim-
CA3 area (Small et al., 2011; Falkai et al., 2012). ilar representations are processed into distinct, dissimilar
In AD, extracellular amyloid deposits consisting of Ab non-overlapping representations, a function that is
protein and intra-neuronal NFTs composed of required in discriminating similar experiences (Sahay
hyperphosphorylated tau protein are a hallmark of the et al., 2011; Yassa and Stark, 2011). Pattern separation
cellular pathological process. The deposition of NFTs as well as its complementary function pattern completion
leads to neurodegeneration and the hippocampal CA1 is are thought to be dependent on the neural circuity of the
one of the first structures aected before other subfields DG and the hippocampal CA3 region (Deng et al., 2010). It
such as the subiculum, CA2, CA3 and CA4/DG are has further been suggested that impairments of this pat-
included in the pathological process (West et al., 1994; tern separation process contribute to the development of
Braak and Braak, 1995, 1996; Chetelat et al., 2008; certain neuropsychiatric diseases such as anxiety disor-
Mufson et al., 2015). In some imaging studies, the integ- ders and depression and may be caused by disturbed
rity of CA3/DG (Mueller et al., 2010) or CA2 subregions adult neurogenesis (Sahay and Hen, 2007; Kheirbek
was preserved (Wisse et al., 2014). Deficits in encoding et al., 2012). Impairment of hippocampal neurogenesis
were correlated with CA1 volume whereas deficits in may thus contribute to cognitive deficits seen in depres-
retrieval were correlated with a disruption of a sion. Besides the putative role in pattern separation, adult
hippocampal-parietal-frontal network in aMCI (Fouquet neurogenesis is also thought to play a role in encoding
et al., 2012). Apolipoprotein E4 status has an eect on temporal context representation in the hippocampus and
the volume of CA3/DG in healthy controls and AD in the resolution of memory representations (Aimone
(Mueller and Weiner, 2009). In other studies, it could be et al., 2014). Furthermore, neurogenesis is involved in
shown that the volume of CA3/DG is associated to verbal the formation of a developmental phenotype of individual-
learning and early retrieval whereas CA1 atrophy is corre- ity as a reflection of a neuroplastic interaction between
lated with impaired consolidation/delayed retrieval genetic and environmental variations (Freund et al.,
(Mueller et al., 2007, 2011). 2013). Although the relevance of adult neurogenesis for
Accordingly, imaging studies suggest an association enhancing neuroplasticity in the hippocampus has been
between CA1 and subiculum volume loss and the demonstrated in rodents, the functional relevance in
development of later dementia and emphasize subfield humans is not entirely clear (Lazarov et al., 2010; Ming
imaging as sensitive biomarker tool for the detection and Song, 2011). Adult neurogenesis in the hippocampus
and study of early AD processes (de Flores et al., 2015b). is dynamically regulated and positively modulated by var-
ious physiological influences such as physical exercise,
HIPPOCAMPAL NEUROPLASTICITY AND enriched environment, diet and hippocampal learning,
which facilitate cell proliferation, maturation and new neu-
ADULT NEUROGENESIS
ron survival and modulates mechanisms of synaptic plas-
Plasticity in the central nervous system requires the ticity such as DG LTP (Brown et al., 2003; Voss et al.,
adaptation of brain functions and neural circuits and it 2013; Aimone et al., 2014). On the other side, influences
has long been assumed that these plastic changes take such as aging and stress reduce cell proliferation, survival,
place at hard-wired neural connections. It has been and neuronal dierentiation of the newly formed neurons
shown, however, that new neuronal cells can be created (Opendak and Gould, 2015). Thus, hippocampal neuroge-
in the adult mammalian hippocampus; these new cells nesis was found to be altered in experimental models of
might play an important role in the lifelong plasticity neuropsychiatric diseases such as major depression, anx-
mechanisms of learning and adaptation (Ming and Song, iety disorders, stroke, AD and epilepsy. In these disease
2011). Adult neurogenesis in mammals is restricted to states, either a reduction of neurogenesis by decreased
two brain areas the olfactory bulb and hippocampus, stem cell activity or a reduction of neuronal survival can
where neurons are generated by neural stem cells be observed. Further, abnormal stem cell maturation could
throughout adult life. In humans, adult neurogenesis is lead to aberrant integration into hippocampal circuits. The
thought to be restricted to the subgranular zone (SGZ) in reduction of adult neurogenesis in AD may at least partly
the DG of the hippocampus. The functional relevance be modulated by disease risk genes such as human pre-
of this continuous generation of new cells has been senillin (PS) variants. In epilepsy, seizures lead to
extensively studied in the last years, in particular with increased cell proliferation but also to mis-migration of
regard to the structural integration into preexisting newly formed neurons and altered circuit integration
neural circuits within the hippocampus with subsequent (Ming and Song, 2011). Cerebral ischemia stimulates cell

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
8 T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx

proliferation in the DG, thereby giving rise to newly gener- with the objective of restoring homeostasis through
ated neurons and supporting the activity-dependent role of behavioral and physiological adaptations (de Kloet,
adult neurogenesis in plasticity and compensation 2012). These changes in homeostatic regulation can lead
(Schwamm et al., 1998; Bueters et al., 2008; Miles and from adaptive to maladaptive consequences at multiple
Kernie, 2008). Interestingly, the development of levels and are considered to contribute to various neu-
hippocampal neural stem/progenitor cells can not only ropsychiatric diseases in which deficits of hippocampal
be directed toward the neuronal lineage but can be also function such as cognitive impairment can be observed
directed toward the oligodendrocyte lineage, which could (Dohring et al., 2014; Lucassen et al., 2014). The hip-
result in the generation of human oligodendrocytes, critical pocampal system with its circuitry is functionally and
for remyelination processes in response to demyelinating structurally integrated in the stress responses to behav-
lesions (Czepiel et al., 2015). With regard to the mecha- ioral stressors including fear and anxiety. In the context
nisms of plasticity following hippocampal damage, it has of stress, the hippocampus is subject to an involvement
been shown that at least in the rat global ischemia by the hypothalamicpituitaryadrenal axis, which influ-
results in an almost complete loss of CA1 neurons, which ences the hippocampus by the release of stress hor-
is reflected by robust deficits in spatial memory. The reap- mones (de Kloet et al., 2005; McEwen, 2007; Fanselow
pearance of nearly 40% of the CA1 neurons within 90 days and Dong, 2010). Stress targets hippocampal function
of the insult was associated with a restoration of hip- on dierent levels via glucocorticoid receptor activation
pocampal spatial memory function, which indicates that leading to various cellular and systemic eects on synap-
the newly formed neurons that originate in the lateral tic plasticity, neuronal survival, hippocampal neurogene-
periventricular region have the potential to restore sis, hippocampal connectivity and memory (McEwen
hippocampal function (Bendel et al., 2005). Metabolic and Milner, 2007; Sandi and Pinelo-Nava, 2007; Segal
diseases such diabetic states and high-caloric intake also et al., 2010; de Kloet, 2012). As an example, hippocampal
impair hippocampal neurogenesis, synaptic plasticity and LTP and LTD in CA1 can be aected by acute behavioral
learning possibly via changes in glucocorticoid levels stress (Howland and Wang, 2008). Stress may lead also
(Stranahan et al., 2008). Radiation and chemotherapy- to cytotoxicity and metabolic vulnerability in CA1 neurons
induced damage to neural progenitor populations may due to glucocorticoid-enhanced glutamatergic transmis-
lead to impaired adult hippocampal neurogenesis and sion with an increase of calcium influx (Joels, 2009). On
decreased subcortical white matter integrity contributing a structural level, stress may lead to altered dendritic
to neurocognitive deficits (Monje and Dietrich, 2012). architecture in hippocampal circuits as it may induce a
Only recently, the interconnection between adult retraction of CA3 pyramidal neuron dendrites and reduc-
neural stem cell regulation and the immune system tion of dendritic spine density as well as a loss of
acting on hippocampal function has been studied synapses (de Kloet et al., 2005).
(Carpentier and Palmer, 2009; Williamson and Bilbo,
2013; Barrientos et al., 2015). A plethora of literature
demonstrates the impact of an activated immune NEUROINFLAMMATION IN THE HIPPOCAMPUS
response such as microglia and astrocyte activation, infil-
tration of T cells, cytokines, chemokines, prostaglandins The role of neuroinflammatory mechanisms in
and reactive oxygen species (ROS) on hippocampal func- hippocampal function and dysfunction has only recently
tion (Novkovic et al., 2015). This immune activation in been recognized. Neuroinflammatory states can
inflammatory states such as during insult, injury and in significantly contribute to the vulnerability of the
neurodegenerative diseases such as AD and aging may hippocampus leading to cognitive impairment. This does
lead to an impairment of hippocampal function including not only pertain to pathological inflammatory states such
hippocampal neurogenesis, cellular plasticity and learning as during generalized inflammation or sepsis but also
(Barrientos et al., 2012; Williamson and Bilbo, 2013). during dysregulation in autoimmunity as in MS and
In sum, adult neurogenesis in the hippocampus permits limbic encephalitis (Kayser and Dalmau, 2014; Kostic
long-term neuroplastic adaptation in hippocampal circuits et al., 2015). Peripheral inflammatory responses in the
to complex environmental conditions. Considering the hippocampus result in an interleukin-1 beta (IL-1b) activa-
positive and negative regulation of adult hippocampal tion with subsequent T and B lymphocyte proliferation and
neurogenesis by diverse external stimuli such as stimulation of natural killer cell activity. IL-1b facilitates the
environmental factors, the influence of newly generated production of other cytokines such as TNFa and
cells on hippocampal plasticity highlights the potential of interleukin-6 (IL-6), which acts on neural activity in the hip-
neurogenesis for neuroplastic intervention in aging and pocampus (Baier et al., 2009; Tan et al., 2014). An aggra-
disease states (Lazarov et al., 2010; Ming and Song, vated glutamate excitotoxicity may lead to further
2011). deleterious eects in hippocampal neurons (Semmler
et al., 2013; Tan et al., 2014; Michels et al., 2015).
THE HIPPOCAMPUS AND THE REGULATION Increasing evidence suggests that neuroimmune signal-
ing and neuroinflammation is also present in a wide spec-
OF STRESS
trum of pathophysiological processes such as during
Stress as a concept is defined as a multidimensional neurodegeneration in AD (Heneka et al., 2015). Neuroim-
construct consisting of (i) stress input with perception mune signaling encompasses microglia and astrocyte
and appraisal of the stressor, (ii) the processing of activation, T-cell infiltration and messengers such as
stressful information and (iii) the stress response itself cytokines, ROS and prostaglandins.

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx 9

The influence of the immune system is not restricted to reflecting impairments of large-scale cognitive networks
states of an activated immune system. The idea has been (Roosendaal et al., 2008, 2010). The plasticity in hip-
put forward that a basal level of neuroimmune interaction pocampal memory networks in MS patients with cognitive
in the hippocampus is also functionally relevant and deficits was recently studied (Hulst et al., 2012). In an epi-
present in physiological states and during development sodic memory fMRI task, hypoactivation of hippocampus-
(Williamson and Bilbo, 2013). For example, a role for dependent networks was observed in MS patients with
cytokines has been observed in hippocampal learning, cognitive deficits compared to controls. However,
LTP and neurogenesis (Baier et al., 2009; Braun et al., hyperactivation in the hippocampal-cingulate memory
2013; Williamson and Bilbo, 2013). It has further been sug- system during encoding was seen in MS patients without
gested that hippocampal-neuroimmunological interactions cognitive deficits. This dierent activation pattern was
via neuroimmune signaling molecules including chemoki- interpreted as functional adaptation within the hippocam-
nes and cytokines contribute to the homeostatic balance pal memory system before cognitive deficits become
of vulnerability, plasticity and resilience mechanisms in clinically relevant.
the hippocampus (Williamson and Bilbo, 2013). On a cellular level, hippocampal demyelination and
Alterations of the neuro-immune signature such as inflammation are robust findings in post-mortem
microglial sensitization seems to be a hallmark of a specimens (Geurts et al., 2007). Most demyelination
normal aging process resulting in facilitated and lesions are chronic with subpial or subependymal loca-
exaggerated neuroinflammatory response in the aging tion. Whereas Papadopoulos et al. reported neuronal cell
brain to an immune challenge (Barrientos et al., 2015). loss in the CA1 and CA3-2 regions of approximately 30%
This neuroinflammatory response is further enhanced by (Papadopoulos et al., 2009), Dutta et al., reported only
a dysregulated neuroendocrine response in the aged ani- minor neuronal cell loss (Dutta et al., 2011). However,
mal leading to higher cortisol levels. Notably, these neu- the latter study showed a significant decrease in synaptic
roimmunological responses seem to be most prominent density in the demyelinated hippocampus (Dutta et al.,
in the hippocampal formation (Barrientos et al., 2015). 2011). Synaptophysin-positive pre-synaptic terminals in
These mechanisms may lead to long lasting elevations in CA1 and DG were reduced possibly due to
pro-inflammatory cytokines in the hippocampus resulting demyelination-associated synaptic pruning. In compar-
in impaired plasticity and cognitive deficits (Barrientos ison to motor cortex, levels of proteins involved in axonal
et al., 2015). transport, synaptic plasticity, neuronal survival and
glutamatergic neurotransmission were altered in the
hippocampus. This suggests that the molecular machin-
MULTIPLE SCLEROSIS ery associated with neuroplasticity (such as LTP) may
be impaired in MS (Dutta et al., 2011; Novkovic et al.,
MS is an inflammatory and demyelinating disease 2015). Experimentally, inflammatory changes in MS can
that aects the central nervous system and is be modeled using experimental autoimmune
commonly associated with white-matter damage. encephalomyelitis (EAE), an established animal model
Neuroinflammation is mediated by microglia activation of MS that shows acute or chronically relapsing inflamma-
and acts against myelin, the neuropathological hallmark tory and demyelinating autoimmune processes (Novkovic
of the disease process in MS. However, activation of et al., 2015). EAE leads to microglia-mediated neuroin-
the neuroinflammatory cascade also causes neuronal flammation, demyelination, loss of hippocampal CA1
and synaptic damage leading to gray matter neurons and behavioral deficits in a spatial learning task
demyelination, atrophy and degeneration (Wegner and (Peruga et al., 2011).
Stadelmann, 2009). The majority of MS patients exhibits The inflammatory changes in EAE also aect synaptic
a clinically relevant cognitive dysfunction during the plasticity in the hippocampus as LTP in CA1 is
course of the disease including learning and memory dif- compromised possibly due to enhanced IL-1b release
ficulties suggesting involvement of the hippocampal mem- from infiltrating lymphocytes or activated microglia
ory system (Sicotte et al., 2008; Sacco et al., 2015). (Nistico et al., 2014; Novkovic et al., 2015). Also, a
Indeed, human imaging studies have shown that the certain role of the complement system has been attribu-
cognitive deficits are at least partly dependent on the ted to the synaptic changes in demyelinated hippocampi
distribution of demyelinating lesions in the hippocampus (Michailidou et al., 2015). Notably, remyelination of the
of severely-aected patients (Brainin et al., 1988). In hippocampal neurons reversed these alterations (Dutta
patients with cognitive deficits, selective and progressive et al., 2013).
hippocampal atrophy was observed that was focused on
the CA1 subregion (Sicotte et al., 2008). This hippocam-
pal atrophy in turn was associated with deficits in memory IMMUNE-MEDIATED NEUROINFLAMMATORY
encoding and retrieval (Sicotte et al., 2008; Anderson LIMBIC ENCEPHALITIS AND HIPPOCAMPAL
et al., 2010). MS patients with depressive symptoms have
FUNCTION
smaller volume in CA2, CA3 and DG but show elevated
cortisol levels (Gold et al., 2010, 2014). Besides reduction In the last decade, a new entity of inflammatory diseases
in hippocampal volume, connectivity of the hippocampus was recognized that typically aects the limbic system
as measured using resting-state functional connectivity including the medial temporal lobe system and the
between the hippocampus and anterior cingulate hippocampus (Varley et al., 2015). The limbic encephalitis
gyrus, thalamus and prefrontal cortex was decreased is considered an autoimmune encephalitis as antibodies

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
10 T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx

targeting cell-surface or intracellular antigens can be Key elements include a direct cellular toxicity, alterations
detected. In paraneoplastic limbic encephalitis, onconeu- in cellular metabolism, facilitation of oxidative stress and
ral (paraneoplastic) antibodies, such as anti-Hu, anti-Ma2 induction of pro-inflammatory processes leading to myelin
(anti-Ta), CRMP5/CV2 and ANNA-3 target intracellular toxicity, disruption of neurogenesis (Kaiser et al., 2014).
antigens (Irani et al., 2014). On the other side, antibodies Thus, modern chemotherapeutic substances exert their
against neuronal surface antigens attack voltage-gated neurotoxic eect via several modes of action. Considering
potassium channels, glutamic acid decarboxylase (GAD) the role of the hippocampus in learning and memory, its
and NMDA, AMPA and GABA(B) receptors and thus ability for plasticity and its vulnerability, an impairment of
directly interfere with synaptic transmission and neuronal neurogenesis has been suggested to be a key element
plasticity (Irani et al., 2014). Antibodies are not directly in the deleterious eects of chemotherapy on hippocam-
pathogenic and neuronal damage rather seems to be pal function (Kaiser et al., 2014; Dietrich et al., 2015).
mediated by cytotoxic T cells (Irani et al., 2014). Patients Experimentally, damage to neural progenitor cell
with autoimmune limbic encephalitis present with suba- populations within germinal zones of the adult CNS has
cute memory disturbances that are accompanied by vari- been identified as one of the key concepts in
ous neuropsychiatric symptoms, such as disorientation, chemotherapy-associated impairments of neuroplasticity
confusion, temporal lobe seizures and behavioral abnor- and learning and memory (Seigers et al., 2013). Some
malities, that are suggestive of impaired limbic and hip- chemotherapies show a neurotoxic eect on glial progen-
pocampal structures. Memory impairment is particularly itor cells and post-mitotic oligodendrocytes, the myelin
pronounced in the episodic domain (Bartsch et al., forming cells of the CNS that constitute white matter
2010, 2011; Bettcher et al., 2014). For example, antibod- integrity (Dietrich et al., 2015). Experimentally, it could
ies against NMDARs result in impaired hippocampal be shown that chemotherapy leads to an impairment of
synaptic function and leads to memory disturbances, psy- hippocampus-dependent learning and frontal-temporal
chiatric symptoms and seizures (Hughes et al., 2010; networks. In chemotherapy-treated patients, several
Moscato et al., 2010; Bettcher et al., 2014). In patients human imaging studies suggest a reduction of hippocam-
with NMDA encephalitis, neuroinflammatory processes pal volume accompanied by memory deficits, albeit there
lead to an impairment of hippocampal functional connec- are also studies that did not find volume loss and memory
tivity and white matter integrity that correlated with individ- deficits (Inagaki et al., 2007).
ual memory impairment (Finke et al., 2013b). In mice, the
human NMDAR antibodies cause a decrease in the den-
sity of total and synaptic NMDAR clusters and total HIPPOCAMPAL DYSFUNCTION IN METABOLIC
NMDAR protein concentration in the hippocampus DISEASES
(Planaguma et al., 2015). This suggests that these cell-
surface antibodies impair hippocampal NMDAR functions In the last years, it has been shown that metabolic
and synaptic plasticity thus providing a rationale for func- influences on the cellular and systems level can
tional and neurodegenerative changes leading to cogni- enhance or impair hippocampal plasticity (Fotuhi et al.,
tive deficits. 2012). It became clear that metabolic alterations in obe-
Potassium channel complex antibodyassociated sity and diabetes increase the likelihood of cognitive
encephalitis is the most common form of non- decline and accelerate the conversion of cognitive
paraneoplastic limbic encephalitis. MR imaging shows a impairment to dementia (Stranahan, 2015). A model of
preferential aection of the hippocampus with an neurocognitive impairment in obesity and diabetes has
extension to the parahippocampal or fusiform gyrus and, been suggested emphasizing a bidirectional influence of
in some patients, to remote areas such as the thalamus. the metabolic condition on cellular mechanisms involved
In many patients, the hippocampus is selectively in synaptic plasticity (Stranahan and Mattson, 2011).
aected. The hippocampi show prominent swelling in the For example, a sedentary lifestyle, diabetes and obesity
T2-weighted imaging typically seen within the first three may reduce adaptive cellular stress responses with
months of disease onset. Typically, atrophic temporal- oxidative stress, and inflammatory pathways leading to
mesial or hippocampal structures can be observed in the impaired synaptic plasticity, decreased neurogenesis
later stages (Urbach et al., 2006; Bien et al., 2007). and increased neurodegeneration (Fontan-Lozano et al.,
2007; Stranahan, 2015). These mechanisms may lead
to enhanced vulnerability toward neurocognitive impair-
CHEMOTHERAPY, COGNITIVE IMPAIRMENT ment and AD. On the other hand, exercise and caloric
restriction enhance neuroprotection, neurogenesis, and
AND HIPPOCAMPAL TOXICITY
synaptic plasticity via induction of adaptive cellular stress
Modern therapeutic options targeting cancer cells can responses, e.g. via BDNF (Stranahan and Mattson,
have also eects on the cognitive domain including 2011). Some of the underlying mechanisms have been
learning and memory, attention, executive, processing identified. Diabetic states and high-caloric intake seem
speed as well as mood that are clinically relevant and to, at least partly, exert their eects via increased
add to the morbidity of patients (Seigers et al., 2013). In glucocorticoid receptor activation and impaired insulin
recent years, putative key elements of this CNS toxicity and leptin signaling (Stranahan et al., 2008; Stranahan
of chemotherapy have been identified on a cellular level and Mattson, 2011). A link between diabetes and neu-
and it has been suggested that neurotoxic eects act on rodegenerative diseases such as Alzheimers has been
neural function and plasticity (Monje and Dietrich, 2012). suggested as the insulin-degrading enzyme (IDE) is also

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx 11

involved in the removal of beta-amyloid from the brain. It of epileptic seizures leading to cell loss (Forster et al.,
is thus conceivable that both conditions, diabetes and 2012; Bartsch et al., 2015; Sierra et al., 2015). This itself
amyloid burden enhance each other by competing for may stimulate mechanisms of neural plasticity and further
the same rescue pathway (Yang and Song, 2013). In contribute to epileptogenesis. For example, it has been
addition, obesity-induced neuroinflammation has been shown that during epileptogenesis, aberrant neuronal cir-
linked to impaired hippocampal function (Yang and cuits are formed by adult-generated granule cells in the
Song, 2013; Erion et al., 2014). In sum, metabolic factors DG that itself are capable of inducing seizure activity
do show an important influence on various cellular and (Pun et al., 2012). These results suggest that seizure
systems mechanisms involved in the regulation of hip- induced neurogenesis may play critical role in epileptoge-
pocampal plasticity which may lead to an increased vul- nesis in the hippocampus. In human patients, aspects of
nerability in disease states. Along these lines the microstructural damage and plasticity in the hippocampus
relevance of modifiable environmental factors including in epileptogenesis have begun to be studied using
cognitive activation, learning, life style factors, physical MR-imaging aiming at identifying biomarkers of the
activity and metabolic factors on hippocampal function epileptogenic process, which are critical for the prognosis
has been emphasized (Fotuhi et al., 2012). of epilepsy as well as for the assessment of possible
treatment strategies (Sierra et al., 2015).

THE HIPPOCAMPUS IN EPILEPSY


SLEEP AND HIPPOCAMPAL VULNERABILITY
Epilepsy is a neurological disorder that is characterized by
recurrent seizures. Seizures are correlates of abnormal, The hippocampus plays a key role in sleep-dependent
excessive or synchronous neuronal activity within the memory consolidation in animals and humans
CNS. The human hippocampus in particular is capable (Diekelmann and Born, 2010). Consolidation of newly
in generating epileptic seizures and to facilitate a formed memories refers to the process of stabilization of
chronic increase of cellular excitability which in turn is a memory trace either by strengthening or reducing the
thought to be the correlate of temporal lobe epilepsy susceptibility to interference. In humans, the hippocam-
(TLE). TLE is a focal epilepsy characterized by seizures pus and adjacent cortical structures contribute to
originating in or primarily involving temporal lobe sleep-dependent memory consolidation through interac-
structures including the hippocampus (ODell et al., tions with distributed brain areas. According to a major
2012; Sloviter and Bumanglag, 2013). The disease hypothesis of sleep-dependent memory consolidation of
course of TLE typically correlates with pathomorphologi- episodic memory, neuronal activity associated with the
cal neuronal changes termed hippocampal sclerosis encoding process during wakefulness becomes replayed
(HS; syn. Ammons horn sclerosis) that manifest with seg- (reactivated) during ensuing sleep and thus mediates sys-
mental neuronal cell loss in the pyramidal cell layer of the tem consolidation processes involving the redistribution of
cornu ammonis CA1 and CA4 associated with reactive memories from an intermediate hippocampus-dependent
astrogliosis (Cendes et al., 2014; Blumcke and Coras, to a long-term hippocampus-independent store (Wilson
2015). It is dicult, however, to disentangle causes and and McNaughton, 1994; Diekelmann and Born, 2010).
consequences of epileptogenesis and seizures in hip- This transfer is thought to be mediated by synchronous
pocampal circuits. The current concept of TLE favors network pattern in the hippocampus during slow-wave
complex multifactorial influences and mechanisms on dif- sleep (ONeill et al., 2010). Sleep has also been
ferent temporal scales and suggests a structural and implicated in the regulation of plasticity as it has been
functional re-organization of neural circuits in CA3 and suggested that one of the critical functions of sleep is
DG with aberrant maturation and ectopic migration within the restoration of synaptic homeostasis in terms of renor-
the dentate circuitry. These alterations of synaptic cir- malization of synaptic strength. This homeostatic regula-
cuitry include aberrant mossy fiber sprouting, alterations tion of synaptic strength is thought to increase neuronal
in dendritic branching, spine density and may lead to a signal-to-noise ratios and thus to facilitate the integration
formation of recurrent excitatory collaterals that transform and consolidation of new memories (Tononi and Cirelli,
the CA3 principal cells into an epileptogenic population 2014). As described, many critical cognitive functions of
promoting seizure initiation and/or propagation by the hippocampus require specific brain states such as
increasing the net excitatory drive of dentate granule neu- slow-wave sleep for the processing of mnemonic informa-
rons (Kienzler et al., 2009; ODell et al., 2012). Other tion. Thus, hippocampal function is very sensitive to a dis-
potential components of hippocampal epileptogenesis ruption of these processes by sleep deprivation and sleep
include, among others, molecular rearrangement/plastic- loss leading to impaired formation of hippocampus-
ity of ion channel and neurotransmitter receptor expres- dependent memories (Kreutzmann et al., 2015). Sleep
sion, neural injury, viral infections, as well as genetic deprivation not only interrupts sleep-dependent cognitive
variations, epigenetic modifications, neuroinflammation, processing leading to impaired cognitive functioning, but
blood brain-barrier leakage, angiogenesis and endocrine has widespread eects on synaptic plasticity mechanisms
influences. However, the precise role of these influences in the hippocampus (Prince and Abel, 2013). Short
in the generation of epileptogenic networks remains periods of sleep deprivation impair hippocampal plasticity
unclear (Pitkanen and Sutula, 2002; ODell et al., 2012). by aecting LTP, second-messenger signaling (cAMP-
Hippocampal neurons are in particular vulnerable to protein kinase A (PKA)), adenosine, cellular transcription
metabolic stress conditions and excitotoxicity in the wake factors (CREB), neurotrophic signaling (BDNF), and

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
12 T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx

glutamate receptor expression (Havekes et al., 2012). Bakker A, Kirwan CB, Miller M, Stark CE (2008) Pattern separation in
Long-term sleep loss may lead to reduced hippocampal the human hippocampal CA3 and dentate gyrus. Science
319:16401642.
cell proliferation and impaired neurogenesis thus impair-
Bannerman DM, Rawlins JN, McHugh SB, Deacon RM, Yee BK, Bast
ing hippocampal plasticity and structural integrity on a T, Zhang WN, Pothuizen HH, Feldon J (2004) Regional
longer time scale (Kreutzmann et al., 2015). Sleep depri- dissociations within the hippocampusmemory and anxiety.
vation after learning in humans also alters the connectivity Neurosci Biobehav Rev 28:273283.
between brain regions as a shift from the hippocampo- Barrientos RM, Frank MG, Watkins LR, Maier SF (2012) Aging-
neocortical network to an increased striatal navigation- related changes in neuroimmune-endocrine function: implications
related activity was observed (Orban et al., 2006). for hippocampal-dependent cognition. Horm Behav 62:219227.
Barrientos RM, Kitt MM, Watkins LR, Maier SF (2015)
Neuroinflammation in the normal aging hippocampus.
CONCLUDING REMARKS Neuroscience. http://dx.doi.org/10.1016/
j.neuroscience.2015.03.007.
The research of the last decades has highlighted Bartsch T (2012) The Clinical Neurobiology of the
the multifaceted structure and function of the Hippocampus. Oxford, UK: Oxford University Press.
hippocampus as a pivotal structure involved in cognitive Bartsch T, Alfke K, Stingele R, Rohr A, Freitag-Wolf S, Jansen O,
Deuschl G (2006) Selective aection of hippocampal CA-1
and behavioral regulation. These functions of the
neurons in patients with transient global amnesia without long-
hippocampus are a reflection of the high degree of term sequelae. Brain 129:28742884.
neuroplasticity and tightly linked to the instrinsic Bartsch T, Alfke K, Wol S, Rohr A, Jansen O, Deuschl G (2008)
properties of hippocampal neurons, circuits and neural Focal MR spectroscopy of hippocampal CA-1 lesions in transient
populations. This particular degree of neuroplasticity global amnesia. Neurology 70:10301035.
may also be involved in the particular vulnerability and Bartsch T, Dohring J, Rohr A, Jansen O, Deuschl G (2011) CA1
neurons in the human hippocampus are critical for
susceptibility of the hippocampus to deleterious
autobiographical memory, mental time travel, and autonoetic
conditions such as ischemia, epilepsy, chronic stress consciousness. Proc Natl Acad Sci U S A 108:1756217567.
and neurodegeneration. From a clinical viewpoint, Bartsch T, Dohring J, Reuter S, Finke C, Rohr A, Brauer H, Deuschl
the unique level of neuroplastic mechanisms in the G, Jansen O (2015) Selective neuronal vulnerability of human
hippocampus may be further explored with regard to hippocampal CA1 neurons: lesion evolution, temporal course, and
questions of resilience, regeneration and restoration pattern of hippocampal damage in diusion-weighted MR
to metabolic and behavioral stress that could lead to imaging. J Cereb Blood Flow Metab. http://dx.doi.org/10.1038/
jcbfm.2015.137.
therapeutic interventions on a cellular and systems level
Bartsch T, Schonfeld R, Muller FJ, Alfke K, Leplow B, Aldenho J,
in neurological, neurodegenerative, and psychiatric Deuschl G, Koch JM (2010) Focal lesions of human hippocampal
diseases. CA1 neurons in transient global amnesia impair place memory.
Science 328:14121415.
AcknowledgmentsT.B. has been supported by the German Battaglia FP, Benchenane K, Sirota A, Pennartz CM, Wiener SI
Research Foundation SFB 654, FOR 2093, the German Cluster (2011) The hippocampus: hub of brain network communication for
memory. Trends Cognit Sci 15:310318.
of Excellence Inflammation-at-Interfaces (ExC 306) and by the
Bendel O, Bueters T, von Euler M, Ove Ogren S, Sandin J, von Euler
Faculty of Medicine, University of Kiel, Germany. Peer Wul
G (2005) Reappearance of hippocampal CA1 neurons after
was supported by the Medical Research Council grant
ischemia is associated with recovery of learning and memory. J
G1100546, the German Research Foundation FOR 2143 and
Cereb Blood Flow Metab 25:15861595.
the Faculty of Medicine, University of Kiel, Germany. Bergmann O, Frisen J (2013) Neuroscience. Why adults need new
brain cells. Science 340:695696.
Bettcher BM, Gelfand JM, Irani SR, Neuhaus J, Forner S, Hess CP,
REFERENCES Geschwind MD (2014) More than memory impairment in voltage-
gated potassium channel complex encephalopathy. Eur J Neurol
21:13011310.
Aimone JB, Li Y, Lee SW, Clemenson GD, Deng W, Gage FH (2014)
Bien CG, Urbach H, Schramm J, Soeder BM, Becker AJ, Voltz R,
Regulation and function of adult neurogenesis: from genes to
Vincent A, Elger CE (2007) Limbic encephalitis as a precipitating
cognition. Physiol Rev 94:9911026.
event in adult-onset temporal lobe epilepsy. Neurology
Aksoy-Aksel A, Manahan-Vaughan D (2015) Synaptic strength at the
69:12361244.
temporoammonic input to the hippocampal CA1 region in vivo is
Bliss T, Schoepfer R (2004) Neuroscience. Controlling the ups and
regulated by NMDA receptors, metabotropic glutamate receptors
downs of synaptic strength. Science 304:973974.
and voltage-gated calcium channels. Neuroscience. http://
Blumcke I, Coras R (2015) Clinico-pathological subtypes of
dx.doi.org/10.1016/j.neuroscience.2015.03.014.
hippocampal sclerosis in temporal lobe epilepsy and their
Amaral DG, Scharfman HE, Lavenex P (2007) The dentate gyrus:
dierential impact on memory impairment. Neuroscience, in
fundamental neuroanatomical organization (dentate gyrus for
press. http://dx.doi.org/10.1016/j.neuroscience.2015.08.003.
dummies). Prog Brain Res 163:322.
Bonnici HM, Chadwick MJ, Kumaran D, Hassabis D, Weiskopf N,
Anderson VM, Fisniku LK, Khaleeli Z, Summers MM, Penny SA,
Maguire EA (2012) Multi-voxel pattern analysis in human
Altmann DR, Thompson AJ, Ron MA, Miller DH (2010)
hippocampal subfields. Front Hum Neurosci 6:290.
Hippocampal atrophy in relapsing-remitting and primary
Bonnici HM, Chadwick MJ, Maguire EA (2013) Representations of
progressive MS: a comparative study. Mult Scler 16:10831090.
recent and remote autobiographical memories in hippocampal
Avignone E, Frenguelli BG, Irving AJ (2005) Dierential responses to
subfields. Hippocampus 23:849854.
NMDA receptor activation in rat hippocampal interneurons and
Braak H, Braak E (1995) Staging of Alzheimers disease-related
pyramidal cells may underlie enhanced pyramidal cell
neurofibrillary changes. Neurobiol Aging 16:271278. Discussion
vulnerability. Eur J Neurosci 22:30773090.
278284.
Baier PC, May U, Scheller J, Rose-John S, Schielholz T (2009)
Braak H, Braak E (1996) Evolution of the neuropathology of
Impaired hippocampus-dependent and -independent learning in
Alzheimers disease. Acta Neurol Scand Suppl 165:312.
IL-6 deficient mice. Behav Brain Res 200:192196.

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx 13

Brainin M, Goldenberg G, Ahlers C, Reisner T, Neuhold A, Deecke L of age and Alzheimers disease on hippocampal subfields:
(1988) Structural brain correlates of anterograde memory deficits comparison between manual and FreeSurfer volumetry. Hum
in multiple sclerosis. J Neurol 235:362365. Brain Mapp 36:463474.
Braun O, Dewitz C, Moller-Hackbarth K, Scheller J, Schielholz T, de Kloet ER, Joels M, Holsboer F (2005) Stress and the brain: from
Baier PC, Rose-John S (2013) Eects of blockade of peripheral adaptation to disease. Nat Rev Neurosci 6:463475.
interleukin-6 trans-signaling on hippocampus-dependent and de Kloet R (2012) Stress and the Hippocampus. In: The clinical
independent memory in mice. J Interferon Cytokine Res neurobiology of the hippocampus (Th. Bartsch, ed), pp 77104.
33:254260. Oxford: Oxford University Press.
Brown J, Cooper-Kuhn CM, Kempermann G, Van Praag H, Winkler J, Deng W, Aimone JB, Gage FH (2010) New neurons and new
Gage FH, Kuhn HG (2003) Enriched environment and physical memories: how does adult hippocampal neurogenesis aect
activity stimulate hippocampal but not olfactory bulb learning and memory? Nat Rev Neurosci 11:339350.
neurogenesis. Eur J Neurosci 17:20422046. Deuker L, Doeller CF, Fell J, Axmacher N (2014) Human
Bueters T, von Euler M, Bendel O, von Euler G (2008) Degeneration neuroimaging studies on the hippocampal CA3 region
of newly formed CA1 neurons following global ischemia in the rat. integrating evidence for pattern separation and completion.
Exp Neurol 209:114124. Front Cell Neurosci 8:64.
Burger C (2010) Region-specific genetic alterations in the aging Diekelmann S, Born J (2010) The memory function of sleep. Nat Rev
hippocampus: implications for cognitive aging. Front Aging Neurosci 11:114126.
Neurosci 2:140. Dietrich J, Prust M, Kaiser J (2015) Chemotherapy, cognitive
Burke SN, Barnes CA (2006) Neural plasticity in the ageing brain. Nat impairment and hippocampal toxicity. Neuroscience. http://
Rev Neurosci 7:3040. dx.doi.org/10.1016/j.neuroscience.2015.06.016.
Burke SN, Barnes CA (2010) Senescent synapses and hippocampal Dohring J, Schmuck A, Bartsch T (2014) Stress-related factors in the
circuit dynamics. Trends Neurosci 33:153161. emergence of transient global amnesia with hippocampal lesions.
Butler TR, Self RL, Smith KJ, Sharrett-Field LJ, Berry JN, Littleton Front Behav Neurosci 8:287.
JM, Pauly JR, Mulholland PJ, Prendergast MA (2010) Selective Dong HW, Swanson LW, Chen L, Fanselow MS, Toga AW (2009)
vulnerability of hippocampal cornu ammonis 1 pyramidal cells to Genomic-anatomic evidence for distinct functional domains in
excitotoxic insult is associated with the expression of polyamine- hippocampal field CA1. Proc Natl Acad Sci U S A
sensitive N-methyl-D-asparate-type glutamate receptors. 106:1179411799.
Neuroscience 165:525534. Drapeau E, Mayo W, Aurousseau C, Le Moal M, Piazza PV, Abrous
Buzsaki G, Moser EI (2013) Memory, navigation and theta rhythm in DN (2003) Spatial memory performances of aged rats in the water
the hippocampal-entorhinal system. Nat Neurosci 16:130138. maze predict levels of hippocampal neurogenesis. Proc Natl Acad
Carpentier PA, Palmer TD (2009) Immune influence on adult neural Sci U S A 100:1438514390.
stem cell regulation and function. Neuron 64:7992. Dutta R, Chang A, Doud MK, Kidd GJ, Ribuado MV, Young EA, Fox
Cendes F, Sakamoto AC, Spreafico R, Bingaman W, Becker AJ RJ, Staugaitis SM, Trapp BD (2011) Demyelination causes
(2014) Epilepsies associated with hippocampal sclerosis. Acta synaptic alterations in hippocampi from multiple sclerosis
Neuropathol 128:2137. patients. Ann Neurol 69:445454.
Chen PE, Errington ML, Kneussel M, Chen G, Annala AJ, Rudhard Dutta R, Chomyk AM, Chang A, Ribaudo MV, Deckard SA, Doud MK,
YH, Rast GF, Specht CG, Tigaret CM, Nassar MA, Morris RG, Edberg DD, Bai B, Li M, Baranzini SE, Fox RJ, Staugaitis SM,
Bliss TV, Schoepfer R (2009) Behavioral deficits and subregion- Macklin WB, Trapp BD (2013) Hippocampal demyelination and
specific suppression of LTP in mice expressing a population of memory dysfunction are associated with increased levels of the
mutant NMDA receptors throughout the hippocampus. Learn neuronal microRNA miR-124 and reduced AMPA receptors. Ann
Mem 16:635644. Neurol 73:637645.
Chetelat G, Fouquet M, Kalpouzos G, Denghien I, De la Sayette V, Engvig A, Fjell AM, Westlye LT, Skaane NV, Sundseth O, Walhovd
Viader F, Mezenge F, Landeau B, Baron JC, Eustache F, KB (2012) Hippocampal subfield volumes correlate with memory
Desgranges B (2008) Three-dimensional surface mapping of training benefit in subjective memory impairment. Neuroimage
hippocampal atrophy progression from MCI to AD and over 61:188194.
normal aging as assessed using voxel-based morphometry. Erion JR, Wosiski-Kuhn M, Dey A, Hao S, Davis CL, Pollock NK,
Neuropsychologia 46:17211731. Stranahan AM (2014) Obesity elicits interleukin 1-mediated deficits
Coultrap SJ, Nixon KM, Alvestad RM, Valenzuela CF, Browning MD in hippocampal synaptic plasticity. J Neurosci 34:26182631.
(2005) Dierential expression of NMDA receptor subunits and Falkai P, Gruber O, Schmitt A (2012) Schizophrenia. In: The clinical
splice variants among the CA1, CA3 and dentate gyrus of the neurobiology of the hippocampus (Th. Bartsch, ed). Oxford:
adult rat. Brain Res Mol Brain Res 135:104111. Oxford University Press.
Cramer SC, Sur M, Dobkin BH, OBrien C, Sanger TD, Trojanowski Fanselow MS, Dong HW (2010) Are the dorsal and ventral
JQ, Rumsey JM, Hicks R, Cameron J, Chen D, Chen WG, Cohen hippocampus functionally distinct structures? Neuron 65:719.
LG, deCharms C, Duy CJ, Eden GF, Fetz EE, Filart R, Freund Finke C, Bruehl H, Duzel E, Heekeren HR, Ploner CJ (2013a) Neural
M, Grant SJ, Haber S, Kalivas PW, Kolb B, Kramer AF, Lynch M, correlates of short-term memory reorganization in humans with
Mayberg HS, McQuillen PS, Nitkin R, Pascual-Leone A, Reuter- hippocampal damage. J Neurosci 33:1106111069.
Lorenz P, Schi N, Sharma A, Shekim L, Stryker M, Sullivan EV, Finke C, Kopp UA, Scheel M, Pech LM, Soemmer C, Schlichting J,
Vinogradov S (2011) Harnessing neuroplasticity for clinical Leypoldt F, Brandt AU, Wuerfel J, Probst C, Ploner CJ, Pruss H,
applications. Brain 134:15911609. Paul F (2013b) Functional and structural brain changes in anti-N-
Czepiel M, Boddeke E, Copray S (2015) Human oligodendrocytes in methyl-D-aspartate receptor encephalitis. Ann Neurol
remyelination research. Glia 63:513530. 74:284296.
Das SR, Avants BB, Pluta J, Wang H, Suh JW, Weiner MW, Mueller Fontan-Lozano A, Saez-Cassanelli JL, Inda MC, de los Santos-
SG, Yushkevich PA (2012) Measuring longitudinal change in the Arteaga M, Sierra-Dominguez SA, Lopez-Lluch G, Delgado-
hippocampal formation from in vivo high-resolution T2-weighted Garcia JM, Carrion AM (2007) Caloric restriction increases
MRI. Neuroimage 60:12661279. learning consolidation and facilitates synaptic plasticity through
de Flores R, La Joie R, Chetetat G (2015a) Structural imaging of mechanisms dependent on NR2B subunits of the NMDA receptor.
hippocampal subfields in healthy aging and Alzheimers disease. J Neurosci 27:1018510195.
Neuroscience, in press. http://dx.doi.org/10.1016/j.neuroscience. Forster A, Griebe M, Gass A, Kern R, Hennerici MG, Szabo K (2012)
2015.08.033. Diusion-weighted imaging for the dierential diagnosis of
de Flores R, La Joie R, Landeau B, Perrotin A, Mezenge F, de La disorders aecting the hippocampus. Cerebrovasc Dis
Sayette V, Eustache F, Desgranges B, Chetelat G (2015b) Eects 33:104115.

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
14 T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx

Fotuhi M, Do D, Jack C (2012) Modifiable factors that alter the size of Joels M (2009) Stress, the hippocampus, and epilepsy. Epilepsia
the hippocampus with ageing. Nat Rev Neurol 8:189202. 50:586597.
Fouquet M, Desgranges B, La Joie R, Riviere D, Mangin JF, Landeau Jones O (2015) Astrocyte mediated metaplasticity in the
B, Mezenge F, Pelerin A, de La Sayette V, Viader F, Baron JC, hippocampus: Help or hindrance? Neuroscience, in press. http://
Eustache F, Chetelat G (2012) Role of hippocampal CA1 atrophy dx.doi.org/10.1016/j.neuroscience.2015.08.035.
in memory encoding deficits in amnestic mild cognitive Kaiser J, Bledowski C, Dietrich J (2014) Neural correlates of
impairment. Neuroimage 59:33093315. chemotherapy-related cognitive impairment. Cortex 54:3350.
Freund J, Brandmaier AM, Lewejohann L, Kirste I, Kritzler M, Kruger Kayser MS, Dalmau J (2014) Anti-NMDA receptor encephalitis,
A, Sachser N, Lindenberger U, Kempermann G (2013) autoimmunity, and psychosis. Schizophr Res. http://dx.doi.org/
Emergence of individuality in genetically identical mice. Science 10.1016/j.schres.2014.10.007.
340:756759. Kesner RP, Hunsaker MR (2009) The temporal attributes of episodic
Gee CE, Benquet P, Raineteau O, Rietschin L, Kirbach SW, Gerber memory. Behav Brain Res 215:299309.
U (2006) NMDA receptors and the dierential ischemic Kesner RP, Rolls ET (2015) A computational theory of hippocampal
vulnerability of hippocampal neurons. Eur J Neurosci function, and tests of the theory: new developments. Neurosci
23:25952603. Biobehav Rev 48:92147.
Geurts JJ, Bo L, Roosendaal SD, Hazes T, Daniels R, Barkhof F, Kheirbek MA, Klemenhagen KC, Sahay A, Hen R (2012)
Witter MP, Huitinga I, van der Valk P (2007) Extensive Neurogenesis and generalization: a new approach to stratify
hippocampal demyelination in multiple sclerosis. J Neuropathol and treat anxiety disorders. Nat Neurosci 15:16131620.
Exp Neurol 66:819827. Kienzler F, Norwood BA, Sloviter RS (2009) Hippocampal injury,
Gold SM, Kern KC, OConnor MF, Montag MJ, Kim A, Yoo YS, atrophy, synaptic reorganization, and epileptogenesis after
Giesser BS, Sicotte NL (2010) Smaller cornu ammonis 2 perforant pathway stimulation-induced status epilepticus in the
3/dentate gyrus volumes and elevated cortisol in multiple mouse. J Comp Neurol 515:181196.
sclerosis patients with depressive symptoms. Biol Psychiatry Kirino T (2000) Delayed neuronal death. Neuropathology
68:553559. 20(Suppl):S95S97.
Gold SM, OConnor MF, Gill R, Kern KC, Shi Y, Henry RG, Pelletier Kjelstrup KG, Tuvnes FA, Steenach HA, Murison R, Moser EI,
D, Mohr DC, Sicotte NL (2014) Detection of altered hippocampal Moser MB (2002) Reduced fear expression after lesions of the
morphology in multiple sclerosis-associated depression using ventral hippocampus. Proc Natl Acad Sci U S A 99:1082510830.
automated surface mesh modeling. Hum Brain Mapp 35:3037. Kolb B, Gibb R (2014) Searching for the principles of brain plasticity
Gray DT, Barnes CA (2015) Distinguishing adaptive plasticity from and behavior. Cortex 58:251260.
vulnerability in the aging hippocampus. Neuroscience, in press. Kolb B, Muhammad A (2014) Harnessing the power of neuroplasticity
http://dx.doi.org/10.1016/j.neuroscience.2015.08.001. for intervention. Front Hum Neurosci 8:377.
Guzowski JF, Knierim JJ, Moser EI (2004) Ensemble dynamics of Kostic M, Stojanovic I, Marjanovic G, Zivkovic N, Cvetanovic A (2015)
hippocampal regions CA3 and CA1. Neuron 44:581584. Deleterious versus protective autoimmunity in multiple sclerosis.
Harry GJ, Lefebvre dHellencourt C (2003) Dentate gyrus: alterations Cell Immunol 296:122132.
that occur with hippocampal injury. Neurotoxicology 24:343356. Kreutzmann JC, Havekes R, Abel T, Meerlo P (2015) Sleep
Havekes R, Vecsey CG, Abel T (2012) The impact of sleep deprivation and hippocampal vulnerability: changes in neuronal
deprivation on neuronal and glial signaling pathways important plasticity, neurogenesis and cognitive function. Neuroscience.
for memory and synaptic plasticity. Cell Signal 24:12511260. http://dx.doi.org/10.1016/j.neuroscience.2015.04.053.
Heine J, Pruess H, Bartsch T, Ploner CJ, Paul F, Finke C (2015) La Joie R, Fouquet M, Mezenge F, Landeau B, Villain N, Mevel K,
Imaging of autoimmune encephalitis relevance for clinical Pelerin A, Eustache F, Desgranges B, Chetelat G (2010)
practice and hippocampal function. Neuroscience. http:// Dierential eect of age on hippocampal subfields assessed
dx.doi.org/10.1016/j.neuroscience.2015.05.037. using a new high-resolution 3T MR sequence. Neuroimage
Heneka MT, Golenbock DT, Latz E (2015) Innate immunity in 53:506514.
Alzheimers disease. Nat Immunol 16:229236. Lavenex P, Amaral DG (2000) Hippocampal-neocortical interaction: a
Howland JG, Wang YT (2008) Synaptic plasticity in learning and hierarchy of associativity. Hippocampus 10:420430.
memory: stress eects in the hippocampus. Prog Brain Res Lazarov O, Mattson MP, Peterson DA, Pimplikar SW, van Praag H
169:145158. (2010) When neurogenesis encounters aging and disease.
Hughes EG, Peng X, Gleichman AJ, Lai M, Zhou L, Tsou R, Parsons Trends Neurosci 33:569579.
TD, Lynch DR, Dalmau J, Balice-Gordon RJ (2010) Cellular and Leuner B, Gould E (2010) Structural plasticity and hippocampal
synaptic mechanisms of anti-NMDA receptor encephalitis. J function. Annu Rev Psychol 61(111140):C111C113.
Neurosci 30:58665875. Lisman JE (1999) Relating hippocampal circuitry to function: recall of
Hulme SR, Jones OD, Abraham WC (2013) Emerging roles of memory sequences by reciprocal dentate-CA3 interactions.
metaplasticity in behaviour and disease. Trends Neurosci Neuron 22:233242.
36:353362. Lisman JE, Otmakhova NA (2001) Storage, recall, and novelty
Hulst HE, Schoonheim MM, Roosendaal SD, Popescu V, Schweren detection of sequences by the hippocampus: elaborating on the
LJ, van der Werf YD, Visser LH, Polman CH, Barkhof F, Geurts JJ SOCRATIC model to account for normal and aberrant eects of
(2012) Functional adaptive changes within the hippocampal dopamine. Hippocampus 11:551568.
memory system of patients with multiple sclerosis. Hum Brain Lister JP, Barnes CA (2009) Neurobiological changes in the
Mapp 33:22682280. hippocampus during normative aging. Arch Neurol 66:829833.
Hunsaker MR, Kesner RP (2013) The operation of pattern separation Lucassen PJ, Pruessner J, Sousa N, Almeida OF, Van Dam AM,
and pattern completion processes associated with dierent Rajkowska G, Swaab DF, Czeh B (2014) Neuropathology of
attributes or domains of memory. Neurosci Biobehav Rev stress. Acta Neuropathol 127:109135.
37:3658. Malykhin NV, Coupland NJ (2015) Hippocampal neuroplasticity in
Inagaki M, Yoshikawa E, Matsuoka Y, Sugawara Y, Nakano T, major depressive disorder. Neuroscience. http://dx.doi.org/
Akechi T, Wada N, Imoto S, Murakami K, Uchitomi Y (2007) 10.1016/j.neuroscience.2015.04.047.
Smaller regional volumes of brain gray and white matter Marr D (1971) Simple memory: a theory for archicortex. Philos Trans
demonstrated in breast cancer survivors exposed to adjuvant R Soc Lond B Biol Sci 262:2381.
chemotherapy. Cancer 109:146156. McEwen B (1994) The plasticity of the hippocampus is the reason for
Irani SR, Gelfand JM, Al-Diwani A, Vincent A (2014) Cell-surface its vulnerability. Semin Neurosci 6:239246.
central nervous system autoantibodies: clinical relevance and McEwen BS (2007) Physiology and neurobiology of stress and
emerging paradigms. Ann Neurol 76:168184. adaptation: central role of the brain. Physiol Rev 87:873904.

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx 15

McEwen BS, Milner TA (2007) Hippocampal formation: shedding light multiple sclerosis. Neuroscience. http://dx.doi.org/10.1016/
on the influence of sex and stress on the brain. Brain Res Rev j.neuroscience.2015.03.008.
55:343355. ODell CM, Das A, Gt Wallace, Ray SK, Banik NL (2012)
Michaelis E (2012) Selective Neuronal Vulnerability in the Understanding the basic mechanisms underlying seizures in
Hippocampus: Relationship to Neurological Diseases and mesial temporal lobe epilepsy and possible therapeutic targets:
Mechanisms for Dierential Sensitivity of Neurons to Stress. In: a review. J Neurosci Res 90:913924.
The clinical neurobiology of the hippocampus (Th. Bartsch, ed), ONeill J, Pleydell-Bouverie B, Dupret D, Csicsvari J (2010) Play it
pp 5476 Oxford: Oxford University Press. again: reactivation of waking experience and memory. Trends
Michailidou I, Willems JG, Kooi EJ, van Eden C, Gold SM, Geurts JJ, Neurosci 33:220229.
Baas F, Huitinga I, Ramaglia V (2015) Complement C1q-C3 OReilly RC, McClelland JL (1994) Hippocampal conjunctive
associated synaptic changes in multiple sclerosis hippocampus. encoding, storage, and recall: avoiding a trade-o.
Ann Neurol 77:10071026. Hippocampus 4:661682.
Michels M, Vieira AS, Vuolo F, Zapelini HG, Mendonca B, Mina F, Olariu A, Cleaver KM, Cameron HA (2007) Decreased neurogenesis
Dominguini D, Steckert A, Schuck PF, Quevedo J, Petronilho F, in aged rats results from loss of granule cell precursors without
Dal-Pizzol F (2015) The role of microglia activation in the lengthening of the cell cycle. J Comp Neurol 501:659667.
development of sepsis-induced long-term cognitive impairment. Opendak M, Gould E (2015) Adult neurogenesis: a substrate for
Brain Behav Immun 43:5459. experience-dependent change. Trends Cogn Sci 19:151161.
Miles DK, Kernie SG (2008) Hypoxic-ischemic brain injury activates Orban P, Rauchs G, Balteau E, Degueldre C, Luxen A, Maquet P,
early hippocampal stem/progenitor cells to replace vulnerable Peigneux P (2006) Sleep after spatial learning promotes covert
neuroblasts. Hippocampus 18:793806. reorganization of brain activity. Proc Natl Acad Sci U S A
Ming GL, Song H (2011) Adult neurogenesis in the mammalian brain: 103:71247129.
significant answers and significant questions. Neuron Pandis C, Sotiriou E, Kouvaras E, Asprodini E, Papatheodoropoulos
70:687702. C, Angelatou F (2006) Dierential expression of NMDA and
Monje M, Dietrich J (2012) Cognitive side eects of cancer therapy AMPA receptor subunits in rat dorsal and ventral hippocampus.
demonstrate a functional role for adult neurogenesis. Behav Brain Neuroscience 140:163175.
Res 227:376379. Papadopoulos D, Dukes S, Patel R, Nicholas R, Vora A, Reynolds R
Moscato EH, Jain A, Peng X, Hughes EG, Dalmau J, Balice-Gordon (2009) Substantial archaeocortical atrophy and neuronal loss in
RJ (2010) Mechanisms underlying autoimmune synaptic multiple sclerosis. Brain Pathol 19:238253.
encephalitis leading to disorders of memory, behavior and Pastalkova E, Serrano P, Pinkhasova D, Wallace E, Fenton AA,
cognition: insights from molecular, cellular and synaptic studies. Sacktor TC (2006) Storage of spatial information by the
Eur J Neurosci 32:298309. maintenance mechanism of LTP. Science 313:11411144.
Moser MB, Moser EI (1998) Functional dierentiation in the Peruga I, Hartwig S, Thone J, Hovemann B, Gold R, Juckel G, Linker
hippocampus. Hippocampus 8:608619. RA (2011) Inflammation modulates anxiety in an animal model of
Mueller SG, Chao LL, Berman B, Weiner MW (2011) Evidence for multiple sclerosis. Behav Brain Res 220:2029.
functional specialization of hippocampal subfields detected by MR Pitkanen A, Sutula TP (2002) Is epilepsy a progressive disorder?
subfield volumetry on high resolution images at 4T. Neuroimage Prospects for new therapeutic approaches in temporal-lobe
56:851857. epilepsy. Lancet Neurol 1:173181.
Mueller SG, Schu N, Yae K, Madison C, Miller B, Weiner MW Planaguma J, Leypoldt F, Mannara F, Gutierrez-Cuesta J, Martn-
(2010) Hippocampal atrophy patterns in mild cognitive impairment Garca E, Aguilar E, Titulaer M, Jain A, Balice-Gordon R,
and Alzheimers disease. Hum Brain Mapp 31:13391347. Lakadamyali M, Graus F, Maldonado R, Dalmau J (2015)
Mueller SG, Stables L, Du AT, Schu N, Truran D, Cashdollar N, Human N-Methyl-D-aspartate receptor antibodies alter memory
Weiner MW (2007) Measurement of hippocampal subfields and and behavior in mice. Brain 138:94109.
age-related changes with high resolution MRI at 4T. Neurobiol Poppenk J, Evensmoen HR, Moscovitch M, Nadel L (2013) Long-axis
Aging 28:719726. specialization of the human hippocampus. Trends Cogn Sci
Mueller SG, Weiner MW (2009) Selective eect of age, Apo e4, and 17:230240.
Alzheimers disease on hippocampal subfields. Hippocampus Prince TM, Abel T (2013) The impact of sleep loss on hippocampal
19:558564. function. Learn Mem 20:558569.
Mufson EJ, Mahady L, Waters D, Counts SE, Perez SE, DeKosky ST, Pulsinelli WA, Brierley JB, Plum F (1982) Temporal profile of neuronal
Ginsberg SD, Ikonomovic MD, Sche SW, Binder LI (2015) damage in a model of transient forebrain ischemia. Ann Neurol
Hippocampal plasticity during the progression of Alzheimers 11:491498.
disease. Neuroscience. http://dx.doi.org/10.1016/j.neuroscience. Pun RY, Rolle IJ, Lasarge CL, Hosford BE, Rosen JM, Uhl JD,
2015.03.006. Schmeltzer SN, Faulkner C, Bronson SL, Murphy BL, Richards
Murray AJ, Sauer JF, Riedel G, McClure C, Ansel L, Cheyne L, DA, Holland KD, Danzer SC (2012) Excessive activation of mTOR
Bartos M, Wisden W, Wul P (2011) Parvalbumin-positive CA1 in postnatally generated granule cells is sucient to cause
interneurons are required for spatial working but not for reference epilepsy. Neuron 75:10221034.
memory. Nat Neurosci 14:297299. Quintana P, Stefano A, Hakkoum D, Muller D (2006) Glutamate
Neunuebel JP, Knierim JJ (2014) CA3 retrieves coherent receptor changes associated with transient anoxia/hypoglycaemia
representations from degraded input: direct evidence for CA3 in hippocampal slice cultures. Eur J Neurosci 23:975983.
pattern completion and dentate gyrus pattern separation. Neuron Rolls ET, Kesner RP (2006) A computational theory of hippocampal
81:416427. function, and empirical tests of the theory. Prog Neurobiol
Newrzella D, Pahlavan PS, Kruger C, Boehm C, Sorgenfrei O, Schrock 79:148.
H, Eisenhardt G, Bischo N, Vogt G, Wafzig O, Rossner M, Maurer Roosendaal SD, Hulst HE, Vrenken H, Feenstra HE, Castelijns JA,
MH, Hiemisch H, Bach A, Kuschinsky W, Schneider A (2007) The Pouwels PJ, Barkhof F, Geurts JJ (2010) Structural and functional
functional genome of CA1 and CA3 neurons under native hippocampal changes in multiple sclerosis patients with intact
conditions and in response to ischemia. BMC Genomics 8:370. memory function. Radiology 255:595604.
Nistico R, Mori F, Feligioni M, Nicoletti F, Centonze D (2014) Synaptic Roosendaal SD, Moraal B, Vrenken H, Castelijns JA, Pouwels PJ,
plasticity in multiple sclerosis and in experimental autoimmune Barkhof F, Geurts JJ (2008) In vivo MR imaging of hippocampal
encephalomyelitis. Philos Trans R Soc Lond B Biol Sci lesions in multiple sclerosis. J Magn Reson Imaging 27:726731.
369:20130162. Ryan TJ, Roy DS, Pignatelli M, Arons A, Tonegawa S (2015)
Novkovic T, Shchyglo O, Gold R, Manahan-Vaughan D (2015) Memory. Engram cells retain memory under retrograde amnesia.
Hippocampal function is compromised in an animal model of Science 348:10071013.

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084
16 T. Bartsch, P. Wul / Neuroscience xxx (2015) xxxxxx

Sacco R, Bisecco A, Corbo D, Della Corte M, dAmbrosio A, Docimo allocentric encoding of spatial information. J Neurosci
R, Gallo A, Esposito F, Esposito S, Cirillo M, Lavorgna L, 29:1051210519.
Tedeschi G, Bonavita S (2015) Cognitive impairment and memory Tamminga CA, Suzanne Zukin R (2015) Schizophrenia: evidence
disorders in relapsing-remitting multiple sclerosis: the role of white implicating hippocampal GluN2B protein and REST epigenetics in
matter, gray matter and hippocampus. J Neurol 262:16911697. psychosis pathophysiology. Neuroscience. http://dx.doi.org/
Sahay A, Hen R (2007) Adult hippocampal neurogenesis in 10.1016/j.neuroscience.2015.07.038.
depression. Nat Neurosci 10:11101115. Tan H, Cao J, Zhang J, Zuo Z (2014) Critical role of inflammatory
Sahay A, Wilson DA, Hen R (2011) Pattern separation: a common cytokines in impairing biochemical processes for learning and
function for new neurons in hippocampus and olfactory bulb. memory after surgery in rats. J Neuroinflammation 11:93.
Neuron 70:582588. Thompson CL, Pathak SD, Jeromin A, Ng LL, MacPherson CR,
Sandi C, Pinelo-Nava MT (2007) Stress and memory: behavioral Mortrud MT, Cusick A, Riley ZL, Sunkin SM, Bernard A, Puchalski
eects and neurobiological mechanisms. Neural Plast RB, Gage FH, Jones AR, Bajic VB, Hawrylycz MJ, Lein ES (2008)
2007:78970. Genomic anatomy of the hippocampus. Neuron 60:10101021.
Schmidt-Kastner R, Freund TF (1991) Selective vulnerability of the Tononi G, Cirelli C (2014) Sleep and the price of plasticity: from
hippocampus in brain ischemia. Neuroscience 40:599636. synaptic and cellular homeostasis to memory consolidation and
Schwamm LH, Koroshetz WJ, Sorensen AG, Wang B, Copen WA, integration. Neuron 81:1234.
Budzik R, Rordorf G, Buonanno FS, Schaefer PW, Gonzalez RG Urbach H, Soeder BM, Jeub M, Klockgether T, Meyer B, Bien CG
(1998) Time course of lesion development in patients with acute (2006) Serial MRI of limbic encephalitis. Neuroradiology
stroke: serial diusion- and hemodynamic-weighted magnetic 48:380386.
resonance imaging. Stroke 29:22682276. van der Marel K, Bouet V, Meerho GF, Freret T, Boulouard M,
Segal M, Richter-Levin G, Maggio N (2010) Stress-induced dynamic Dauphin F, Klomp A, Lucassen PJ, Homberg JR, Dijkhuizen RM,
routing of hippocampal connectivity: a hypothesis. Hippocampus Reneman L (2015) Eects of long-term methylphenidate
20:13321338. treatment in adolescent and adult rats on hippocampal shape,
Seigers R, Schagen SB, Van Tellingen O, Dietrich J (2013) functional connectivity and adult neurogenesis. Neuroscience.
Chemotherapy-related cognitive dysfunction: current animal http://dx.doi.org/10.1016/j.neuroscience.2015.04.044.
studies and future directions. Brain Imaging Behav 7:453459. van Strien NM, Cappaert NL, Witter MP (2009) The anatomy of
Semmler A, Widmann CN, Okulla T, Urbach H, Kaiser M, Widman G, memory: an interactive overview of the parahippocampal-
Mormann F, Weide J, Fliessbach K, Hoeft A, Jessen F, Putensen hippocampal network. Nat Rev Neurosci 10:272282.
C, Heneka MT (2013) Persistent cognitive impairment, Varley J, Vincent A, Irani SR (2015) Clinical and experimental studies
hippocampal atrophy and EEG changes in sepsis survivors. J of potentially pathogenic brain-directed autoantibodies: current
Neurol Neurosurg Psychiatry 84:6269. knowledge and future directions. J Neurol 262:10811095.
Shing YL, Rodrigue KM, Kennedy KM, Fandakova Y, Bodammer N, Voss MW, Vivar C, Kramer AF, van Praag H (2013) Bridging animal
Werkle-Bergner M, Lindenberger U, Raz N (2011) Hippocampal and human models of exercise-induced brain plasticity. Trends
subfield volumes: age, vascular risk, and correlation with Cogn Sci 17:525544.
associative memory. Front Aging Neurosci 3:2. Wang X, Michaelis EK (2010) Selective neuronal vulnerability to
Sicotte NL, Kern KC, Giesser BS, Arshanapalli A, Schultz A, Montag oxidative stress in the brain. Front Aging Neurosci 2:12.
M, Wang H, Bookheimer SY (2008) Regional hippocampal Wegner C, Stadelmann C (2009) Gray matter pathology and multiple
atrophy in multiple sclerosis. Brain 131:11341141. sclerosis. Curr Neurol Neurosci Rep 9:399404.
Sierra A, Grohn O, Pitkanen A (2015) Imaging microstructural damage West MJ, Coleman PD, Flood DG, Troncoso JC (1994) Dierences in
and plasticity in the hippocampus during epileptogenesis. the pattern of hippocampal neuronal loss in normal ageing and
Neuroscience. http://dx.doi.org/10.1016/j.neuroscience.2015. Alzheimers disease. Lancet 344:769772.
04.054. Williamson LL, Bilbo SD (2013) Chemokines and the hippocampus: a
Sloviter RS, Bumanglag AV (2013) Defining epileptogenesis and new perspective on hippocampal plasticity and vulnerability. Brain
identifying antiepileptogenic targets in animal models of Behav Immun 30:186194.
acquired temporal lobe epilepsy is not as simple as it might Wilson MA, McNaughton BL (1994) Reactivation of hippocampal
seem. Neuropharmacology 69:315. ensemble memories during sleep. Science 265:676679.
Small SA (2002) The longitudinal axis of the hippocampal formation: Wisse LE, Biessels GJ, Heringa SM, Kuijf HJ, Koek DH, Luijten PR,
its anatomy, circuitry, and role in cognitive function. Rev Neurosci Geerlings MI (2014) Hippocampal subfield volumes at 7T in early
13:183194. Alzheimers disease and normal aging. Neurobiol Aging
Small SA, Schobel SA, Buxton RB, Witter MP, Barnes CA (2011) A 35:20392045.
pathophysiological framework of hippocampal dysfunction in Wu MV, Hen R (2014) Functional dissociation of adult-born neurons
ageing and disease. Nat Rev Neurosci 12:585601. along the dorsoventral axis of the dentate gyrus. Hippocampus
Stranahan AM (2015) Models and mechanisms for hippocampal 24:751761.
dysfunction in obesity and diabetes. Neuroscience. http:// Wul P, Ponomarenko AA, Bartos M, Korotkova TM, Fuchs EC,
dx.doi.org/10.1016/j.neuroscience.2015.04.045. Bahner F, Both M, Tort AB, Kopell NJ, Wisden W, Monyer H
Stranahan AM, Arumugam TV, Cutler RG, Lee K, Egan JM, Mattson (2009) Hippocampal theta rhythm and its coupling with gamma
MP (2008) Diabetes impairs hippocampal function through oscillations require fast inhibition onto parvalbumin-positive
glucocorticoid-mediated eects on new and mature neurons. interneurons. Proc Natl Acad Sci U S A 106:35613566.
Nat Neurosci 11:309317. Yang Y, Song W (2013) Molecular links between Alzheimers disease
Stranahan AM, Mattson MP (2011) Bidirectional metabolic regulation and diabetes mellitus. Neuroscience 250:140150.
of neurocognitive function. Neurobiol Learn Mem 96:507516. Yassa MA, Mattfeld AT, Stark SM, Stark CE (2011) Age-related
Strange BA, Witter MP, Lein ES, Moser EI (2014) Functional memory deficits linked to circuit-specific disruptions in the
organization of the hippocampal longitudinal axis. Nat Rev hippocampus. Proc Natl Acad Sci U S A 108:88738878.
Neurosci 15:655669. Yassa MA, Stark CE (2011) Pattern separation in the hippocampus.
Suthana NA, Ekstrom AD, Moshirvaziri S, Knowlton B, Bookheimer Trends Neurosci 34:515525.
SY (2009) Human hippocampal CA1 involvement during

Please cite this article in press as: Bartsch T, Wulff P. The hippocampus in aging and disease: From plasticity to vulnerability. Neuroscience (2015),
http://dx.doi.org/10.1016/j.neuroscience.2015.07.084

Vous aimerez peut-être aussi