Você está na página 1de 47

Physiol Rev 96: 647– 693, 2016

Published March 9, 2016; doi:10.1152/physrev.00010.2015

CELLULAR AND SYSTEM BIOLOGY OF MEMORY:


TIMING, MOLECULES, AND BEYOND
Martin Korte and Dietmar Schmitz

Zoological Institute, Division of Cellular Neurobiology, Braunschweig, Germany; Helmholtz Centre for Infection
Research, AG NIND, Braunschweig, Germany; and Neuroscience Research Centre, Charité Universitätsmedizin
Berlin, Berlin, Germany

Korte M, Schmitz D. Cellular and System Biology of Memory: Timing, Molecules,

L
and Beyond. Physiol Rev 96: 647– 693, 2016. Published March 9, 2016;
doi:10.1152/physrev.00010.2015.—The storage of information in the mammalian
nervous systems is dependent on a delicate balance between change and stability of
neuronal networks. The induction and maintenance of processes that lead to changes
in synaptic strength to a multistep process which can lead to long-lasting changes, which starts and
ends with a highly choreographed and perfectly timed dance of molecules in different cell types of
the central nervous system. This is accompanied by synchronization of specific networks, resulting
in the generation of characteristic “macroscopic” rhythmic electrical fields, whose characteristic
frequencies correspond to certain activity and information-processing states of the brain. Molec-
ular events and macroscopic fields influence each other reciprocally. We review here cellular
processes of synaptic plasticity, particularly functional and structural changes, and focus on timing
events that are important for the initial memory acquisition, as well as mechanisms of short- and
long-term memory storage. Then, we cover the importance of epigenetic events on the long-time
range. Furthermore, we consider how brain rhythms at the network level participate in processes
of information storage and by what means they participating in it. Finally, we examine memory
consolidation at the system level during processes of sleep.

I. INTRODUCTION 647 rons in neuronal networks, which is so important for ani-


II. CELLULAR BIOLOGY OF MEMORY 649 mals in general and for humans specifically?
III. SYSTEMS BIOLOGY OF MEMORY 673
IV. PERSPECTIVES 681 It became evident in the last decades that neurons in memory
relevant areas of the mammalian brain are highly plastic. This
means that the activity patterns generated by experience can
I. INTRODUCTION modify neuronal function and structure. Especially prominent
to learning and memory events are processes of synaptic plas-
“There is nothing like future and past (ь ь ь). There is only ticity, which include activity-dependent alterations of the effi-
the presence of the past, the presence of the presence, and cacy of synaptic transmission (functional plasticity) and
the presence of the future. These three I see in the soul, but changes in the structure and number of synaptic connections
I cannot see them independent of it: present is the memory
(structural plasticity). But in addition to processes of synaptic
of the past, present is the perception of the presence, and
plasticity, it also became evident that the storage of informa-
present is the expectation of the future” (Augustine, Con-
tion is highly influenced by activity patterns of neuronal
fessions, Chapter 20 in Book 11).
networks and any approach which tries to explain the cel-
Life is in many respects a remembrance of things of the past. lular foundation of memory storage needs to take the net-
In this context, information storage works on different time work level into account. In addition, the timing of network
scales in the genome (evolution and epigenetic), on behav- activities is as important as the history of prior events,
ior, and in the nervous system. One of the most intriguing which influences the propensity of neurons to be plastic and
questions is how does the brain capture and store informa- by this means to change its input-output characteristic.
tion in such an efficient and long-lasting way? To do so, the Therefore, in this review, we will summarize the knowledge
brain has to perform a tremendous task: it has to process a about the interplay of different types of neurons and the
continuous input from our sensory organs and at the same rhythm of their activity to prepare a network of neurons to
time it must be able to store memories, sometimes even for receive and to store information. In addition, we would like
a lifetime (and sensory processing does not stop when we to report about these network events and follow them all
store or retrieve information). What is the cellular founda- the way to the molecular level, which have to finally imple-
tion of this long-term information storage capacity of neu- ment the necessary plastic changes that are needed for the

0031-9333/16 Copyright © the American Physiological Society 647


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

processes of memory and recall. In particular, we will focus hemispheres (FIGURE 1). In particular, the hippocampus is
on neuron-related timing events that are important for the essential for the formation and also for recall of spatial,
initial steps of memory acquisition. We will then consider contextual, and episodic memory (99, 327, 412) by inte-
cellular processes modulating the input and output charac- grating sensory and spatial information from the entorhinal
teristics of a neuron due to changes in neuronal activity cortex (EC) to create context-specific representations (225,
(synaptic plasticity). Next, we will examine the importance 279, 423). In humans, the hippocampus plays a major role
of epigenetic events on the long-range time scale of months in processes related to declarative memory, the type of
and years, and finally, we will focus on hypothesis to ex- knowledge that comprises facts about the self and the sur-
plain how brain rhythms are organized at the network level rounding world. Famous cases of hippocampal lesions, as
to assist in, or being central, to information storage before the case of the patient H.M., impressively show the impor-
examining memory consolidation at the systems level. In tance of the hippocampus in memory-related functions.
this context, the timing of single events is important, since After a bilateral surgical removal of the hippocampus (hip-
the encoding of a memory trace on the behavioral level pocampectomy), H.M. suffered from permanent antero-
requires time scales that differ substantially from those re- grade amnesia and partial retrograde amnesia (387). Very
quired for processes of synaptic plasticity and the review old memories could be retrieved, demonstrating the fact
will address the question of which cellular or network that the hippocampus is not the storage site for long-term
mechanisms allow bridging these different time scales. memories but serves for the processing of memories by con-
necting pieces of information and preparing them for inde-
To give the reader a sense of what we consider the major issues
pendent long-term storage in higher cortical brain regions.
emerging in the research about learning and memory mecha-
nisms, we have been highly selective in the topics we cover and
in the number of papers we can cite. This selective approach is
bound to exclude a large body of work on memory processes.
We can only focus on several major questions (related to our
own research fields) that we consider particularly challenging
and that have recently evolved into a broader focus. This is a
huge field to cover, in time (from nanoseconds to years) and
space (from nanometers within the synaptic cleft to centime-
ters of neuronal networks), and therefore, we had to limit
ourselves to some aspects of the events in neuronal networks,
neurons, synapses, and signaling pathways.

Throughout this review we will stress that learning and


Rec.
memory processes are the result of the interplay between Stim.
synaptic plasticity and orchestrated network activity that
finally culminates in the long-term storage of information.
Overall, information storage is a dynamic and interactive CA1
process that starts with the encoding of new information
and progresses to the rather fragile short-term/intermediate
memory, before a memory trace (engram) is stored in dif- CA2
ferent forms of long-term memory (191). At this stage the DG
engram might be either consolidated for a lifetime, destabi-
lized, or even restabilized in the course of memory retrieval. CA3
These neuronal dynamics start and end with synaptic and
cellular plasticity and extend to more distributed larger neu- FIGURE 1. Anatomy of the hippocampus. Anatomically, the mouse
ronal networks and can be observed at the behavioral level. hippocampus is formed by the dentate gyrus (DG) and the ammon’s
horn (cornu ammonis, CA). The CA region can be subdivided into
Although synaptic plasticity has been found in most regions CA1, CA2, and CA3. Information is delivered from the starting point
at the entorhinal cortex (EC) via the perforant path (green axonal
of the vertebrate brain (and for that matter in all other
projection) into the DG and the CA3 region or (pink) to the CA1
neuronal networks of all other animal species), most of the pyramidal cells. From the DG, signals are transferred via mossy
research on activity-dependent synaptic plasticity in mam- fibers (violet) to the CA3 region and after that via the Schaffer
mals has been performed on a brain region called the hip- collaterals (yellow) to CA1 pyramidal cells before they are delivered
pocampus. back to the EC (blue). For the analysis of synaptic plasticity, stimulus
electrodes (Stim.) are employed for activating axon fiber bundles
from Schaffer collaterals and the signals are recorded from apical or
The hippocampus is an evolutionarily old part of the cere- basal dendrites of CA1 pyramidal cells using a recording electrode
bral cortex, lying bilaterally in the depth of the cerebral (Rec.).

648 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

Its highly organized network structure renders it to an ideal (SPM) hypothesis formulated by Morris and colleagues and
model for investigation of specific synaptic contacts be- which is used as a road map for this part of the review:
tween groups of neurons. The hippocampal formation is “Activity-dependent synaptic plasticity is induced at appro-
organized in transverse lamellae. Herein, axons are project- priate synapses during memory formation, and is both nec-
ing in line with pyramidal cells so that slices can keep the essary and sufficient for the information storage underlying
neurons and most of their projections alive. It comprises the the type of memory mediated by the brain area in which
dentate gyrus (DG) and the ammon’s horn (CA, lat. cornu that plasticity is observed” (284).
ammonis). The hippocampal formation includes three sub-
regions with distinct populations of excitatory neurons,
forming a trisynaptic circuit between excitatory neurons A. Processes of Timing on the Cellular Level:
(see FIGURE 1). Information enters the hippocampus from Storage of Information
the EC, terminating on granular cells of the DG. These
neurons send axons, the mossy fibers to pyramidal neurons On the cellular and molecular level, timing and the orches-
of the area CA3, and these in turn connect by the Schaffer tration of memory events is crucial. With respect to timing,
collaterals to CA1 pyramidal neurons. In addition to excit- “Hebb’s rule” is by far the best-known example for the role
atory neurons, there are highly diverse populations of in- of timing events as a central mechanism for information
hibitory interneurons in all subregions mediating feedback storage. The key concept postulated by D. O. Hebb tempo-
and feed-forward inhibition within an area and shaping rally links neural activity representing aspects of informa-
rhythms of activity by grouped discharge predisposing the tion processing and storage to neuronal assemblies (165).
network for alterations. In all subregions, modification and Hebb postulated that not a single cell but networks of neu-
interconnection take place, and at each of the stations, con-
rons, which change the strength of their connections and
nections can be altered. The hippocampal CA3-CA1 syn-
therefore their input/output characteristics, can be used to
apse is the best-specified synapse in the mammalian brain in
store information. Hebb also suggested that the timing of
terms of synaptic plasticity. Interestingly, the morphologi-
events is crucial for the learning and memory processes in
cal and physiological characteristics of the CA3 region are
the brain (165): “When an axon of cell A is near enough to
essential in the encoding of novel information involving
excite a cell B and repeatedly or persistently takes part in
associations between cues and spatial locations, and the
firing it, some growth process [structural changes] or met-
CA3-CA1 connection is mandatory for processes of recall
abolic change [functional changes] takes place in one or
and pattern completion (for review, see Ref. 209). But re-
both cells such that A’s efficiency, as one of the cells firing B,
cently, also the CA2 region received some attention: it is a
is increased.” In the Hebbian rule, timing (associativity) and
fairly small region between the CA3 and CA1 subregion.
input specificity are of upmost importance. In other words,
CA2 neurons get their input from DG cells and project to
memories are stored not in single cells but in networks of
CA1 pyramidal neurons (228). In addition, it might also get
input from the EC. The CA2 region might have its own neurons where the information is stored by altering the
specific function, since it could be shown that mice, in contact sites between nerve cells. These changes take place
which the CA2 subregion was specifically inactivated, at synapses and are therefore called activity-dependent syn-
showed deficits in social learning (168). aptic plasticity. Hebb’s postulate also includes an important
rule, predicting that synaptic weights change when the pre-
and postsynaptic neurons show coincident activity (associa-
II. CELLULAR BIOLOGY OF MEMORY tivity), thus changing the input/output characteristic of neu-
ronal assemblies. In 1973, such an enhancement of synaptic
Many different molecular players have to be orchestrated to strength was finally observed for the first time by Timothy
play in order to store and retrieve memory events. In this Bliss and Terje Lømo. After they applied a short train of
section, we will discuss the cellular level, including pre- and high-frequency pulses under in vivo conditions to a hip-
postsynaptic processes, as well as the role of extracellular pocampal pathway (see FIGURE 1 and sect. I), they could
matrix, glia cells, and modulatory inputs that are essential observe a long-lasting strengthening in the electrophysio-
for memory formation. It can be assumed that the brain of logical response properties of the recorded neurons (36), a
animals and humans alike encodes internal as well as exter- phenomenon that is now termed long-term potentiation
nal events as spatiotemporal activity patterns that are pro- (LTP). LTP is defined as an increase in synaptic strength
cessed in a neuronal circuit. The output of such a neuronal that lasts for at least 1 h (36). It consists of an induction
circuit (sometimes also called a neuronal ensemble) is de- phase, including processes that trigger the alterations lead-
fined by the weight of synaptic connections between neu- ing to the changes in synaptic efficacy followed by the ex-
rons in such a functional entity. An experimentally well- pression or maintenance phase of LTP. LTP can be divided
supported hypothesis is therefore that information storage in different types: at 1-3 h, it is independent of transcription
is defined as a change in the pattern of synaptic strength in and translation (early or E-LTP); if it lasts longer than 3 h,
a specific neuronal circuit involved in the learned behavior. it is generally dependent on altered gene expression and
This is described in the synaptic plasticity and memory referred to as late LTP (L-LTP) (35, 196).

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 649


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

Hebb’s postulate for the requirements of synaptic strength- no-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)
ening asks for coincident neuronal activity at the pre- and receptors, which allows Na⫹ influx in response to glutamate
postsynapse and for a subcellular mechanism of detection in a binding (FIGURE 2). The NMDA receptor is a ligand-coupled
narrow time window. In 1986, a cellular implementation of ion channel, permeable for Na⫹ and Ca2⫹. Under resting
this requirement was found. Wigström and Gustafsson (462) membrane condition, the ion channel pore is occluded by
showed a strong potentiation of synaptic responses in a hip- Mg2⫹ ions; therefore, binding of glutamate is not sufficient
pocampal slice preparation by simultaneously activating an to open the channel pore for Ca2⫹ or Na⫹. Only the depo-
axon bundle of CA3 neurons and a postsynaptic neuron in the larization of the postsynaptic terminal releases the Mg2⫹
CA1 region (see also sect. I). In many excitatory synapses, a block in addition to glutamate binding allowing influx of
specific role for the induction of plastic alterations is exerted Na⫹ and most importantly of Ca2⫹ into the cell (167) (FIG-
2⫹
by the N-methyl-D-aspartate (NMDA) receptor, an iono- URE 2A). Elevation of the Ca concentration within the
tropic glutamate receptor. This was shown by the inhibition postsynaptic neuron then triggers several intracellular reac-
of LTP induction in area CA1 by application of a receptor tions via the activation of kinases. The requirement for
antagonist for the NMDA receptor. Regular (basal) synaptic postsynaptic depolarization during glutamate binding de-
transmission is mediated predominantly by ionotropic ␣-ami- mands precise timing and coordinated activity of the pre-

A
FIGURE 2. Basic cellular processes re-
sulting in long-term potentiation (LTP) or
long-term depression (LTD). A: under basic
synaptic conditions, AMPA receptors (AM-
PAR) regulate the Na⫹ influx in the postsyn-
aptic dendritic spine depending on the
amount of released glutamate. Ion chan-
nels of NMDA receptors (NMDARs) are
AMPAR NMDAR Mg2+ blocked by Mg2⫹ ions (left). If the postsyn-
Mg2+
aptic membrane is depolarized, the Mg2⫹
block of the NMDA receptor channel is re-
moved and Ca2⫹ and Na⫹ ions enter the
Na+ Na+ Ca2+ spine (right). B: under baseline synaptic
Na+
conditions (top), AMPA receptors cycle be-
tween intracellular compartments and the
Depolarization outer membrane of the postsynapse. This
is accomplished through lateral mobility of
the AMPA receptors out of the postsyn-
apse into endocytose-active zones. Here
AMPA receptors are included into early en-
B AMPAR NMDAR dosomes, which then are transferred to
recycling endosomes, then they return to
the cell membrane via exocytosis, followed
by lateral movement into the synapse
LTP LTD where they are retained. After LTP induc-
Exo- Endo- tion by tetanic stimulation (bottom left), en-
cytosis cytosis
hanced AMPA receptor exocytosis occurs
and these receptors are then stabilized at
the synapse through a Ca2⫹-mediated pro-
Recycling cess that includes protein kinases (e.g.,
CaMKII) and the fusion of recycling endo-
somes mediated by Rab11a. After the in-
NMDAR AMPAR duction of LTD by low-frequency stimulation
NMDAR AMPAR
(bottom right), endocytosis of AMPA recep-
tors is increased, and it occurs at extrasyn-
aptic sites in a process that is Ca2⫹ depen-
CaMKII
dent and included the activation of protein
Endo- Ca2+ phosphatases [e.g., protein phosphatases
cytosis Calcineurin
Ca2+ 1 (PP1) or calcineurin]. After the induction
PP1
of LTD, AMPA receptors are retained
Exocytose within the cell and are stored in endosomes
Endocytose or degraded.
Rab11a

650 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

and postsynaptic side. By this means, the NMDA receptor strengthen it. And indeed, synaptic depression (termed
acts as a molecular switch, a real “molecular coincidence long-term depression, LTD) constitutes a necessary coun-
detector.” Only when the postsynaptic cell or a stretch of terpart of LTP. By reducing synaptic efficacy, LTD estab-
dendrite is already (or still) active and the Mg2⫹ block is lishes the necessary differences in strength between syn-
expelled from the NMDAR channel while presynaptic glu- apses, and at the same time, it helps to prevent excessive
tamate is released, an intracellular Ca2⫹ signal is generated. synaptic activity. An earlier notion was that LTD is a cor-
The requirement for presynaptic activity, i.e., glutamate relate of forgetting by weakening unused connections, but it
release, underlines a key feature of LTP: input specificity. is now undisputed that also activity-dependent reduction of
This means that LTP is restricted to synapses, which have synaptic strength is directly involved in learning. A common
been activated (or are very close to activated synapses, see theory is that LTP establishes connectivity within a net-
Ref. 104) rather than to all of the synapses on a given work, whereas LTD accomplishes the fine-tuning of these
neuron. This was experimentally proven by the induction of connections (272). LTD was first described by Dudek and
LTP using intracellular depolarization of a neuron while Bear (95) and Mulkey and Malenka (310). These groups
applying single low-frequency pulses to axons converging observed that prolonged low-frequency stimulation can in-
onto that neuron, which without this postsynaptic depolar- duce a long-lasting reduction of synaptic responses, at the
ization did not show LTP (463; see also Ref. 104). Bliss and same synapses that are capable to show LTP. Even more
Lomo (36) could achieve the same effect by high-frequency surprisingly, it was found that the same molecular players
stimulation of axons. Here, the postsynaptic depolariza- that mediate LTP were also involved in LTD. However,
tion, caused by repeated transmitter release, summed up to they act in opposite directions. While AMPA receptors are
exceed the voltage threshold for the activation of the internalized during the induction of LTD, they are in-
NMDA receptor. This mechanism ensures the second im- serted into the membrane which contribute to the induc-
portant characteristic of LTP: cooperativity. To confine tion of LTP (FIGURE 2B). Some forms of LTD are initiated
plastic alterations to stimuli representing either simultane- by activation of NMDA receptors, by prolonged low-
ous heterosynaptic information or strong (homosynaptic) frequency stimulation (LFS; 1 Hz for 15 min is a typical
inputs, these have to pass a certain intensity threshold due protocol) resulting in a modest increase of cytosolic Ca2⫹
to effects of cooperativity (37; for a review, see Ref. 146). levels (whereas Ca2⫹ levels after tetanic stimulation in-
crease strongly). Instead of resulting in the activation of
With these means, cooperativity and specificity, changes in kinases, a modest increase in Ca2⫹ levels leads predomi-
synaptic strength are largely confined to active connection. nantly to activation of phosphatases, due to their higher
On glutamatergic synapses, the postsynaptic elements are affinity for Ca2⫹ (250; for a review, see Ref. 409). Espe-
located on small membrane protrusions with bottle-neck cially the phosphatase calcineurin (also alternatively ab-
connections to the dendrite, so-called dendritic spines. The breviated as PP2B) is involved here, as well as PP1 (FIG-
geometry of these spine-necks, together with the passive URE 2B). NMDA receptor-dependent LTD is easier to
signal propagation in dendrites, ensures that electric and induce in the hippocampus during development of the
ionic alterations, especially local alterations of cytosolic nervous system, and the likelihood of induction decreases
Ca2⫹, stay largely confined to the active part of the dendrite with age, concomitantly with a switch between NMDA
(for a review, see Ref. 484). Here, the fast kinetics of the receptor subunits with different opening kinetics. During
AMPA receptors and the local extrusion by Na⫹/Ca2⫹ development, the expression of the NMDA receptor sub-
pumps ensure restriction of synaptic alterations. The spec- unit 2B (NR2B) decreases (which helps to mediate LTD),
ificity, however, is not absolute; part of the depolarization whereas the expression of the NR2A subunit rises in-
spreads to nearby portions of dendrite (see Ref. 104). This creasing the likelihood for LTP expression (18, 22, 63).
contributes to the phenomenon of cooperativity, permit-
ting that inputs converging in close spatial vicinity can be In addition, a form of NMDAR-independent LTD was dis-
potentiated if they are active together, but it also explains covered, which relied on the activation of metabotropic
the third feature of LTP: associativity. A weak synaptic glutamate receptors (mGluR) (19, 176). This third type of
connection can be potentiated if it coincides with a stron- glutamate receptors is not linked to an ion channel but
ger one, because the depolarization from nearby active instead to G proteins. The mechanism of mGluR-dependent
synapses helps to surpass the threshold of the NMDA LTD is much less clear. As it is blocked by the postsynaptic
receptor or of voltage-dependent Ca2⫹ channels (219). injection of Ca2⫹ chelators, its induction seems to rely on
So, new connections between previously unrelated inputs postsynaptic mechanisms. However, it results in a de-
can be formed, if one of them surpasses the induction creased frequency but not amplitude of miniature excit-
threshold within a narrow time window (374; but see also atory postsynaptic potentials (mEPSPs), indicating a pre-
the clustered plasticity model in Ref. 146). synaptic phenomenon that can be observed with the patch-
clamp techniques. This suggests a presynaptic locus of
In fine-tuned electrical circuits, you also expect to find expression (for a review, see Ref. 12). Occlusion studies
mechanisms that weaken synaptic input and not only indeed indicate that LFS-induced LTD (NMDAR depen-

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 651


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

dent) can still be induced after mGluR-dependent LTD is recording of precisely timed spikes in pre- and postsyn-
saturated. This confirms that the two forms of LTD rely on aptic neurons (280). In the presynaptic neuron, the elec-
independent signaling mechanisms and that the activation trical stimulation causes well-timed neurotransmitter re-
of the two receptor types occurs independently. NMDA lease; in the postsynaptic neuron, electrical stimulation
receptor-dependent LTD is readily induced in juvenile, but generates a back-propagating action potential (bAP) run-
not in adult mice, suggesting that its main task is to refine ning from the soma into the dendritic compartment. If
the young neuronal network by targeted pruning of less presynaptic activity precedes the postsynaptic spike by
significant contacts during late phases of development. 5–15 ms (representing a situation in which a presynaptic
mGluR-dependent LTD is still inducible in the adult system neuron contributes to firing the postsynaptic neuron), the
(176) and thus seems to be one of the main mechanism strength of the active synapses will be enhanced (LTP).
needed to keep the balance of synaptic weights in the adult On the contrary, if the postsynaptic spike occurs shortly
hippocampus. before transmitter release and binding, uncorrelated ac-
tivity is mimicked and synaptic strength is reduced (in-
ducing LTD). As a consequence, according to this model,
B. Spike-Timing Dependent Plasticity
inputs on a given neuron compete for grouped activity to
form stronger connections, leaving less precisely timed
LTP and LTD can be induced via alterations of stimulus
frequency of the presynaptic neuron alone or via pre- and inputs to be weakened. STDP was observed so far on
postsynaptic stimulation. As outlined above, Hebb postu- more than 20 neuronal connections in different brain
lated that coincident activation of the pre- and postsynapse areas (109) and may represent an important instructor of
leads to a strengthening of these specific synapses, the developing brain (79) as well as a crucial mechanism
whereas noncoincident synaptic activity will lead to the for memory storage in the mature brain (109).
weakening of the connectivity. In this context of timing,
spike-timing dependent plasticity (STDP) is an interest- Functionally, Hebbian STDP strengthens synaptic connec-
ing concept, since it confirms Hebb’s rule and offers an tions whose activity is causal for the postsynaptic spike and
explanation for the time dependency of storage events in does weaken noncausal synaptic activity (1). Additionally,
the brain taking into account the opening time of the the opposite phenomenon called Anti-Hebbian-STDP has
NMDA receptor (FIGURE 3; reviewed in Ref. 79). Exper- been observed. In this case of Anti-Hebbian-STDP, synaptic
imentally, STDP can be induced by paired triggering and neurotransmitter release (presynaptic spike) precedes the
synaptic weight

tpre – tpost [ms]

FIGURE 3. Spike-timing-dependent synaptic plasticity


l
tia

(STDP). Coordinated timing of presynaptic activity and post-


sy

n
n

te

synaptic back-propagating action potential (bAP) cause de-


ap

po
tic

polarization and activation of spines, subsequent Ca2⫹ in-


n
in

tio
pu

flux, and finally, can lead to the induction of LTP or LTD.


ac
t

spine
depolari-
sation

Ca2+ influx

LTP/LTD

652 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

postsynaptic spike and will lead to LTD (whereas the oc- coupled receptors, and it is significantly boosted by the
currence of the postsynaptic spike before presynaptic trans- coincidence of depolarization and receptor activation. At
mitter release will lead to LTP). This phenomenon has been these synapses, the PLC serves as the coincidence detector of
detected at excitatory synapses on medium spiny neurons presynaptic activity (which activates mGluR and conse-
(which are inhibitory themselves), on cholinergic interneu- quently the release of Ca2⫹ from intercellular stores) and
rons (118), as well as on GABAergic interneurons (241, postsynaptic activation of VSCC via the bAP (for further
242). In most cases it is associated with LTD (for details, see details, see Ref. 163).
Ref. 109).
Taken together, precise timing develops on the one hand
An attractive candidate for mechanisms to regulate the tim- from molecular coincidence detectors (NMDA receptor,
ing of pre- and postsynaptic spikes is phase-phase coupling, PLC) and on the other hand from the dynamics of electrical
or phase synchronization (429). Here, spike times are de- signaling in dendrites and spines (this includes the AMPA
termined by the oscillation frequencies of the neuronal as- receptor-mediated signals and the bAP).
semblies of which the pre- and postsynaptic neurons are a
part (further details will be outlined in sect. IIIB). By this Different induction mechanisms for changing synaptic
mechanism, an oscillatory input that impinges on a cluster properties (and consequently the characteristics of neuronal
of rhythmic discharging neurons might meet either the cri- networks) are not restricted to synapses between different
teria of Hebbian STDP (“in-phase” rhythmic activity of neuronal types or even brain regions in the CNS. Even
input and target assembly) or those of anti-Hebbian STDP within the same neuron, synapses may be ruled by different
(“out of phase” rhythm of input and target assembly; for a regimes: bAPs are short lasting and propagate with decre-
review, see Ref. 110). ment. The bAP loses already 50% of its magnitude in the
first 100 ␮m from the soma and disappears completely in
C. Cellular and Molecular Mechanisms of the more distal parts of dendrites of pyramidal neurons
STDP (411). In layer 5 pyramidal neurons of the cortex, Hebbian
STDP can induce LTP only in proximal synapses, whereas
distal synapses show no LTP under these conditions (for
Normal Hebbian STDP is most likely implemented via
further details, see Ref. 400). This mechanism is also pres-
NMDA receptor function, as described above in the context
ent in phylogenetically older palaeocortical structures like
of LTP at the CA3-CA1 hippocampal synapses. The same
the piriform cortex (188). De facto, this implies that the
would hold up for LTD; here the NMDA receptor activa-
rules for synaptic plasticity change between different zones
tion leads to a moderate Ca2⫹ influx (in contrast to the high
Ca2⫹ entry via the NMDA receptor during strong stimula- within a single pyramidal neuron in the neocortex, piriform
tion). The induction of LTP or LTD leads to the removal or cortex, or hippocampus. In other words, the molecular
to the insertion of AMPA receptors into the postsynaptic mechanisms and the timing requirements for modifying the
membrane, respectively (177, 276). At other synapses, dif- connectivity of neuronal networks might be defined by dif-
ferent mechanisms of LTP/LTD are realized, e.g., at the ferent synaptic plasticity thresholds (146, 411). This means,
hippocampal mossy fiber-CA3 as well as at the parallel fiber in simple terms, that proximal synapses will be affected by
synapses in the cerebellum, as the induction and expression the bAP and will most likely implement STDP-LTP. Conse-
mechanisms are presynaptic (for a review, see Ref. 321). quently, this LTP will not or to a lesser extent depend on the
firing rate and on synaptic cooperativity. In contrast, syn-
For STDP it is mechanistically important that Ca2⫹ entry apses at the distal part of large dendritic trees will be prone
into the cell via the NMDA receptor or voltage-sensitive to show STDP-LTD (129) or even anti-Hebbian-LTD
Ca2⫹ channels (VSCC) is timed precisely. VSCC are acti- (399). Furthermore, the proximal synapses will depend to a
vated via the bAP leading to a nonlinear increase in the larger extent on firing rate and cooperativity. The most
cytosolic Ca2⫹ when paired with a synaptic response. Al- distal synapses may be completely outside the STDP regime
ternatively, activation of mGluR resulting in the release of depending on cooperativity and firing rates largely by
Ca2⫹ from intracellular stores plus Ca2⫹ entry via VSCC, neighboring inputs (an example is given in Ref. 143).
again activated via the bAP, fulfill the same purpose (227).
For the implementation of this supralinear Ca2⫹ entry, tim- Taken together, depending on the localization at the den-
ing is important. The bAP needs to activate VSCC and by dritic tree, different plasticity thresholds and diverse timing
this means depolarizes the membrane in spines, which leads rules are implemented to increase or decrease the influence
to the removal of the Mg2⫹ block inside the channel of of STDP on different forms of synaptic plasticity.
NMDA receptors resulting in the maximal Ca2⫹ entry. At
some hippocampal synapses, phospholipase C (PLC) is used Recent studies have questioned the necessity of bAP for the
as a coincidence detector instead of the NMDAR. Here induction of plastic changes under all circumstances (158,
release of endocannabinoids (neuromodulatory lipids) are 257, 396). One has especially to consider that synaptic
induced by either depolarization or activation of G protein- strength changes are not exclusively determined by den-

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 653


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

drites; they are also implemented by molecular mechanisms These findings support the hypothesis that STDP-like plas-
and biophysical properties of synapses themselves. As out- ticity indeed occurs in the human CNS and is instrumental
lined above, the relevance of STDP seems to vary between for sensory perception. Overall, at least for simple visual
different types of synapses and their distance from the perceptions, stimulus-timing dependent plasticity changes
soma. Additionally, one of the most important parameters the orientation and also spatial position perception with a
for the induction of synaptic plasticity is the strength of time regime compatible with STPD rules. Also, data ob-
postsynaptic depolarization. Here, STDP is only one out of tained in humans using transcranial magnetic stimulation
several variables influencing the physical properties of den- (TMS) are in line with timing requirements predicted from
dritic membranes (which include AMPA receptor-mediated STDP rules (259, 469). Finally, for hippocampal place cells,
depolarization, spike timing, spike frequency, and postsyn- it could be shown that the sequence learning of spatial po-
aptic depolarization) within a multifactorial plasticity pa- sitions is compatible with Hebbian-STPD-LTP on the cel-
rameters (for a review, see Ref. 158). A limitation of STDP lular level (293). Moreover, the rate and timing dependent
has been so far the fact that under experimental conditions STDP model fits to results obtained by Bush et al. (49) to
bAPs are usually generated by somatic current injection, explain spatial sequence learning of place cells with over-
whereas in vivo, action potentials are most likely the result lapping place fields.
of an strong excitatory input (reviewed in Ref. 257). “To
‘naturalize’ STDP, it will be important to focus on the mul- Taken together, timing between pre- and postsynaptic neu-
tiple natural sources of synaptic depolarization rather than rons is certainly an important feature of plasticity events in
on artificially induced spikes” (158). For instance, the de- neuronal networks. The relatively new concept of STDP has
polarization achieved via the AMPA receptor channel al- significantly broadened the understanding on how this tim-
ready has a major impact on Ca2⫹ influx. In addition, the ing can be achieved and directed the focus on the functional
strength of synapses is influenced via Ca2⫹ spikes in spines, involvement of the postsynaptic neuron in general and bAPs
which are active before, during, and after the dendritic spike in particular. But despite the conceptional strength of
(bAP). Here, an intriguing idea is that form and function STDP, several important questions remain: how important
depend on each other; the morphology of spines could have is the absolute level of postsynaptic depolarization and
Ca2⫹ entry, despite the relatively moderate effects of bAPs?
a significant effect on synaptic function, e.g., changing the
And in which way do trans-synaptic signaling and other cell
diameter of the spine neck could affect Ca2⫹ diffusion out
types contribute to plasticity and how can this be related to
of the spine into dendrites and by this means the effectivity
STDP? In this respect, STDP has improved the tools and the
of a bAP into spines might be influenced (271, 323).
theoretical analysis of learning-related plasticity in neuro-
nal networks. But for future explorations it seems to be
It is speculated that STDP is not only important as a cellular
more important to focus on the weight of postsynaptic de-
learning rule, but it is also of utmost importance for the
polarization (74, 258) and an increase in cytosolic Ca2⫹
implementation of topographic maps and receptive fields
concentration (396). For this, bAPs are important, espe-
during the development of the sensory systems of the brain
cially considering that bAP evoked Ca2⫹ transients can be
(410). Additionally, it might assist in sharpening the com-
enhanced by neuronal activity (189). However, they are not
petition between convergent inputs (1). In the adult nervous
the only current source to reach a high degree of depolariza-
system, Hebbian STDP supports learning of temporal se- tion or to mobilize Ca2⫹. STDP focuses heavily on the impor-
quences (356). The logic goes as follows: sequential, recur- tant contribution of the postsynaptic neuron as an instructor
rent activation of a neuronal network would drive LTP in for the early events in inducing synaptic plasticity. But in ad-
the so-called forward direction, and LTD would be driven dition to postsynaptic events, a more general view also has to
in the reverse route. It would also be mandatory for the include presynaptic changes in function and structure, trans-
prediction of future events from stimuli experienced in the synaptic signaling at the synaptic cleft, and the role of astro-
past. And indeed, with the use of the tadpole visual system, cytes as members of the so-called tripartite synapse (for re-
experimental evidence has been provided for an STDP views, see Refs. 169, 329). A detailed description of the mo-
mechanism in vivo (105, 114). In addition, it should be lecular players taking place in the different cellular
noted that some psychophysical experiments with human compartments of neurons and glia cells will be given in the
visual perception do support the timing rules of STDP following sections.
(480). STDP was also observed via the variation of the
stimulus timing for the perception of retinotopic position
(130). This could also be observed for higher-order visual D. Well-Timed Molecular Events Underlie
processing: in an experiment concerning face perception, in Processes of Synaptic Plasticity
which a rapid series of two faces (100 pairings over 2 min)
are presented to test persons, a bias for the perception of the The initiation of many synaptic plasticity events in the hip-
second presented face was observed. Following the STDP pocampus seems to be orchestrated by the postsynaptic
prediction, strict timing requirements have to be fulfilled, compartment, which in excitatory neurons is the spine. As
since it only works for pairing delays up to 60 ms (291). outlined above, a spine constitutes a distinct subcompart-

654 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

ment (almost like an organelle) at dendrites that detects spines of excitatory neurons and its inhibition prevents LTP
timing coincidence. The spine utilizes the electrical asym- induction (273). One of the first events after the rise of Ca2⫹
metry (“easy in, hard to get out”) implemented by the ge- levels in the postsynaptic compartment is the activation of
ometry of the spine neck to act as a processing machinery calmodulin upon binding of Ca2⫹, and the resulting com-
for incoming signals. The geometry of the narrow spine plex activates CaMKII (FIGURE 4). As a consequence, CaM-
neck isolates the small volume protrusion, and this has two KII autophosphorylates and is now constitutively active,
important functional consequences for processes of synap- independent of Ca2⫹ or calmodulin. CaMKII translocates
tic plasticity: 1) bAPs invade the spine without decreasing in to the postsynaptic density (PSD), a meshwork of densely
amplitude, and 2) minimal amounts of ion flux over the cell packed postsynaptic proteins juxtaposed to the presynaptic
membrane result in significant depolarization (5, 171). The active zone (134, 277). At the PSD, CaMKII phosphorylates
asymmetry in electric signal propagation becomes clear by amino acid residues on the AMPA receptor subunit GluR1,
considering the direction of current flow from the spine thus increasing its opening probability. It also activates fur-
(high impedance) to the dendrite (low impedance): a spine-
ther signaling molecules, which mediate the insertion of
generated EPSP is highly attenuated when it flows into the
more AMPA receptors into the postsynaptic membrane
branch of a dendrite (10, 37, 271). Therefore, the spine can
(265; for a review, see Ref. 177).
be studied as a separated functional unit (for a detailed
discussion, see Ref. 158). This supports the fact that the
A possible site at which CaMKII (and also for PKA) phos-
spine is not just a little protrusion of the dendrite, but
instead a densely packed compartment itself filled with phorylates the AMPA receptor subunit GluR1 is a serine at
actin, actin binding proteins (70, 119, 174, 492), and position 831 (Ser831). If Ser831 is phosphorylated, the
postsynaptic molecules (214, 393). These molecules need opening probability of the AMPA receptor increases (27,
to interact in a precise spatial-temporal pattern to induce 406), thus resulting in an increase in the EPSP size at this
and to maintain changes in the strength of neuronal con- particular synapse. But the phosphorylation of AMPAR
nectivity (FIGURE 4 – 6). In this spatial-temporal interplay subunits has an even more important effect on the receptor
at glutamatergic synapses, the depolarization achieved insertion into the postsynaptic membrane. It induces the
by AMPA receptors is the first sensitizing event which delivery of GluR1 subunit to the PSD at a higher rate (FIG-
depolarizes the postsynaptic membrane and thus allows URE 4) (48, 87, 274, 409; for a review, see Ref. 177). AMPA
coincidence detection of the activity of the pre- and post- receptors are tetramers (GluR1-GluR4) and are delivered in
synapse via the NMDA receptors. This AMPA receptor- pairs of subunits (367; for a review, see Ref. 177). Mostly,
mediated depolarization can also be important for the GluR1/R2 and GluR2/R3 AMPA receptor compositions/
additive activation of VDCC, when a bAP enters the assemblies are inserted into the postsynaptic membrane
spine (compare sect. IIB) (131, 132, 170). (460). Under baseline synaptic conditions, GluR1 subunits
undergo a cycle of membrane insertion and removal with a
slow rate, whereas GluR2/R3 subunits cycle at a constantly
E. From the Pre- to the Postsynapse: higher rate (164, 340, 395; for a review, see Refs. 71, 177).
Events in Time During elevation of intracellular Ca2⫹, GluR1/R2 insertion
increases, probably caused by exocytosis of endosomes lo-
An integrated view at the paradigmatic CA3-CA1 hip- cated in the spine. This event involves the small G protein
pocampal synapse (see sect. I) could be envisioned like this. Rab11a inserting GluR1/R2 into the spine membrane, most
Activity-dependent increase in intracellular Ca2⫹ levels in
likely at extrasynaptic sites. Therefore, the synaptic strength
the postsynapse, the common crucial step for converting
increases only if lateral diffusion delivers these AMPA recep-
electrical signals into biochemical activity, is achieved by
tors to the PSD (for a recent review, see Ref. 177). Incorpora-
three sources: opening of NMDA receptor channels, VGCC
tion into the PSD is mediated by a specific TARP, the accessory
activity, and/or release from internal stores (388). The in-
subunit stargazin, which mediates the interaction of AMPA
crease in Ca2⫹ levels induces a cascade of events involving
the activation of kinases (372) and ultimately leading to receptors with PSD95 (one of the most prominent scaffolding
the amplification of synaptic transmission (FIGURE 4). proteins in the PSD) (382). Phosphorylation of stargazin leads
The concentration of cytosolic Ca2⫹ is a good predictor to a decrease in AMPA receptor mobility trapping the recep-
of the magnitude of synaptic potentiation (319). High tors at the PSD. By this plethora of highly regulated mecha-
Ca2⫹ levels activate different effector kinases, like PKA, nisms, the composition and total number of postsynaptic
Ca2⫹/calmodulin-dependent protein kinase II (CaMKII), AMPA receptors change after LTP induction. In addition to
Fyn, Scr, and protein kinase Mzeta (PKM␨, an atypical the direct posttranslational modifications of AMPAR, it is cur-
PKC isoform). These kinases initiate processes to main- rently debated if another crucial step for LTP induction is the
tain an increase in synaptic strength acting in various par- phosphorylation of transmembrane AMPA receptor regula-
allel and serial signal transduction pathways (for a review, tory proteins (TARPs), which would then alter AMPAR inser-
see Refs. 197, 372). One of the first kinases discovered in tion, membrane clustering, and traffic (434, 435; for a discus-
this respect is CaMKII, which is highly concentrated in sion of this issue, see Ref. 177).

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 655


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

A B
PRESYNAPTIC PRESYNAPTIC
Neurotransmitter
NEURON molecules AXON
TERMINAL
Synaptic
Depolarization vesicle

Neurotransmitter
Synaptic Vesicle uptake
vesicle 1 mobilization
SYNAPTIC Endocytosys
2 B Neu- PSA-
CLEFT Glutamate
Activ A C rexin NCAM NCAM
Na+ Mg2+ e zone EphrinB
Ca2+ Ca2+ release Docking and SYNAPTIC
Na+ Fusion and CLEFT
Na+ priming
VSCC axocytosis
Kainate 6
AMPA NMDA AMPAR Retrograde
receptor receptor receptor P NMDAR messenger
P (BDNF, NO, endo-
PSA- cannabinoids)
NCAM
EphB Neuro- NCAM
P login
Calcium PSD
Depolarization AMPA influx
receptor Lateral
PKA diffusion Exocytosis
trafficking POSTSYNAPTIC
Adenyl DENDRIC
cyclase AMPAR
Endocytosis SPINE
cAMP
PKC Degradation
P
Intracellular instead of
MAPK Ca2+ Calmodulin International

CaMKII
CREB

POSTSYNAPTIC
Protein synthesis NEURON

FIGURE 4. Events and signaling pathways mediating functional synaptic plasticity. Different events partici-
pate in the strengthening of a synapse and in the manifestation of LTP. A: signaling from the pre- to the
postsynapse: action potentials arriving at the presynaptic terminal initiate Ca2⫹ influx through VSCCs resulting
in the release of glutamate at the active zone. After activation by binding of their ligand glutamate, postsynaptic
AMPA and kainate receptors conduct Na⫹ initiating the depolarization of the postsynaptic membrane. Upon
depolarization, Mg2⫹ ions are removed from NMDA receptors channels allowing the Ca2⫹ influx into the
postsynaptic cytosol resulting in the activation of Ca2⫹-dependent enzymes (e.g., CaMKII, PKC, adenyl cyclase).
Activation of the CaMKII cascade facilitates AMPA receptor trafficking by the phosphorylation of specific
AMPAR subunits. B: retrograde signaling from the post- to the presynapse: diffusible retrograde signaling
messengers like BDNF, NO, and endocannabinoids are released by the postsynapse. Retrograde messengers
bind to their receptors in the presynaptic membrane modulating the neurotransmitter release facility by, e.g.,
modulating the Ca2⫹ influx into the presynapse. Increased Ca2⫹ concentrations are followed by an extensive
vesicle mobilization resulting in the augmented vesicle docking and priming and finally in vesicle fusion and
neurotransmitter release. Vesicles are recycled by endocytosis and neurotransmitters are regained by specific
transporters. Retrograde messengers cause changes in the release probabilities of neurotransmitters at the
presynaptic side, by this means the strengthening of the synapse would be ensured via presynaptic mecha-
nism, including a higher mobilization of vesicles (indicated as 1), and the release probably could be increase
(indicated as 2). Trans-synaptic signaling (6): presynaptic neurexins (NX) and postsynaptic neuroligins (NL)
cross the synaptic cleft transducing signals to the respective compartment. Cadherins transmit signals in a
Ca2⫹-dependent manner and immunoglobulins like NCAM and PSA-NCAM contribute to the adhesion between
pre- and postsynapse. Extracellular matrix (ECM): the netlike ECM contains proteoglycans and glycoproteins
and is considered to maintain stabilized structures. It might control the lateral diffusion of various receptors in
their cell membrane and for this contribute to processes of synaptic plasticity.

The functional effects of AMPA receptor insertion into the AMPA receptors into the postsynaptic membrane of silent
spine membrane are gradual for most synapses, but for synapses which so far only contained NMDA receptors.
some synapses they are extreme. Prior to LTP induction, the This insertion implies that EPSPs appear after LTP induc-
so-called silent synapses have no transmission at all (307). tion when no depolarization was detectable before (138,
Their functional significance and characteristics are still 307). Furthermore, the terminology is imprecise and con-
controversial, overlapping with the debate of the pre- or fusing, because synapses lacking AMPA receptors are not
postsynaptic locus for LTP induction in the hippocampus. really silent, but “mute” as opposed to a truly silent syn-
But some experimental evidence points to the fact that apse, which would not undergo any transmitter release and
strong postsynaptic depolarization leads to the insertion of thus not “speak.” Speaking of “mute” synapses would in-

656 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

A
LTP
Presynaptic Presynaptic

LTD
AMPAR

NMDAR

Synaptic
vesicles
Spine head Spine head

docked synaptic vesicles


AMPAR content

Number of
PSD size

Spine volume PSD size

B polymerization
G-actin F-actin G-actin F-actin

barbed polymerization
G-actin F-actin end polymerization

F-actin
pointed depolymerization ↓↓
end
depolymerization ↓↓ F-actin stability ↑↑
depolymerization ↑

F-actin stability G-actin depolymerization F-actin stability ↑↑

C
N-cadherin
Vezatin

syndecan-2

5-HT2AR
integrin α5

integrin α3
TSPAN7
GluA2

EphB2

TrkB

PAK
Par-1 ka
p190RhoGAP

n
a t eni PICK1 GIT1 CASK ka
lir ERK1/2 Rac1 lir
in
β-c αPIX
FAK in Tiam1 PAK
PAK3

spine enlargement

FIGURE 5. Structural plasticity and actin dynamics. A: structural plasticity induced via activity-dependent
synaptic plasticity can lead to a growth in the postsynaptic density (PSD) and the whole spine volume (after LTP),
or to a shrinkage (LTD) accompanied by the change in the number of AMPA receptors in the PSD and
presynaptically the number of docked presynaptic vesicles. B: actin filament plays a major role in the regulation
of the spine dynamics. Upon LTP induction, actin polymerization increases close to the PSD and the G-actin to
F-actin ratio shifts towards the filamentous F-actin resulting in spine head enlargement. LTD induction leads to
the shrinkage of the spine head and increased actin depolymerization. C: depicted is a set of molecules that
interacts with the actin cytoskeleton. This graph highlights the main PSD-95-associated membrane or scaf-
folding molecules, which are involved in the regulation of spine formation and changes of in spine shape.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 657


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

Folded
m7G protein LOCAL
5 Ubiquitin DEGRADATION
40S
eEF2 Ub Ub
60S Ub Ub

Nascent
polypeptide Proteasome

40S
elF4E m7G

elF4G 40S 3 Met-i


elF2
PABP Degraded
GTP
protein
2

elF4E PABP

4EBP

1 RBPS
RBPS

LOCAL
TRANSLATION

RNA trafficking Synapse to nucleus signal

FIGURE 6. Local protein synthesis and degradation in dendrites and spines. mRNAs for spine-localized
proteins are transcribed in the nucleus and transported into spines by specific transport granules (RNA
trafficking). Here, the 3’UTR of the mRNA is bound by CPEB until the translation is initiated by synaptic
activation. Now, CPEB is phosphorylated and recruits the poly(A) increasing machinery. After PABP binding,
the translatory factors are captured and the local translation is performed. Proteins supposed to be specifically
degraded in proteasomes of spines after ubiquitin-dependent labeling in an activity-dependent manner. CPEB,
cytoplasmic polyadenylation element binding protein; eIF4G, eukaryotic translation initiation factor 4 gamma;
eIF4E, eukaryotic translation initiation factor 4E; PABP, poly(A) binding protein; Ub, ubiquitin; 3’UTR, 3’
untranslated region; arrows indicate where neuronal activity changes and alter the dynamic the system.

658 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

dicate that transmitter release is not initially absent, but the status, and history of neuronal activity at a particular syn-
amount of postsynaptic AMPA receptors is not sufficient to aptic side. GPCR regulatory signaling primarily works to
induce postsynaptic responses (452, 453). control a network of interacting protein kinases that cata-
lyze the transfer of phosphoryl groups to definite serine and
In the context of AMPA receptor cycling in and out of the threonine residues in regulatory target proteins. This sets
cell membrane of spine, it is noteworthy that evidence is the scene for future events quite powerfully; it has been
accumulating showing also NMDA receptors cycle in and estimated that a third of all cytoplasmic proteins are subject
also out of the postsynaptic membrane (for a detailed dis- to kinase regulation (421). However, reviewing the actions
cussion, see Ref. 202). of these important molecules is beyond the scope of this
review; see the following excellent and recent reviews: Ref-
Finally, it should be considered that the so far described erences 12, 23, 139, 263, 272, 309, 415.
early LTP (E-LTP), lasting for 1–3 h, does not necessarily
lead to long-term changes in synaptic strength (376). It is
F. From the Post- to the Presynapse:
generally believed that distinct changes at the synapse lead
Retrograde Signaling
either to LTP, which lasts 1–3 h (E-LTP), or to L-LTP,
which lasts for 3–10 h and probably even longer. Distinc-
So far we have described the prototypic CA3-CA1 LTP
tive inducing stimuli trigger very different but partially
induction mechanism as being implemented in the postsyn-
overlapping biochemical pathways depending on how long
apse, but the mechanisms underlying synaptic plasticity are
the plasticity changes at synapses are maintained. Notably,
pleiotropic and include different cell types and cell compart-
induction of L-LTP leads to activation of signaling path-
ments. Directly following the postsynaptic actions which
ways including PKA and MAPK (also known as ERK) (317,
initiate LTP induction at CA3-CA1 hippocampal synapses,
318). In addition, several transcription factors [e.g., cAMP/
presynaptic changes due to retrograde signaling have to be
Ca2⫹-responsive-element binding protein (CREB), Elk-1]
taken into account (169, 196). Retrograde trans-synaptic
which are expressed constitutively are phosphorylated and
signaling occurs via diffusible retrograde messengers [can-
by this means activated to strengthen transcription of didates are endocannabinoids, BDNF, nitric oxide (NO),
downstream elements that most likely mediate structural and arachidonic acid]. Depending on the activation pattern,
and functional changes of the synapses (196, 486). In recent as well as on the type of neuronal circuit in different brain
years, two prominent candidates, the brain-derived neu- areas, different signaling systems might be activated. In ad-
rotrophic factor (BDNF) and the brain-specific, atypical dition, or alternatively, to the NMDA receptor also
PKC isoform PKM␨ have emerged as key molecular players metabotropic glutamate receptors (mGluR9) or endocan-
for maintaining L-LTP and processes of long-term memory nabinoid type 1 receptor (CB1) signaling mediate different
(333, 341, 373). The overall excitability of a neuron on a forms of plasticity (447).
larger time scale also depends on neuromodulatory neu-
rotransmitters acting via different metabotropic receptors 1. Endocannabinoids
to activate signaling cascades, which boost or maintain the
initial NMDA receptor-mediated plasticity (30). These As a paradigmatic example for a retrograde signaling sys-
modulatory inputs have to be taken into account, because tem involved in processes of synaptic plasticity, endocan-
they give input to excitatory as well as inhibitory neurons in nabinoids (endogenous cannabinoids, eCB) and their recep-
addition to glutamatergic or GABAergic transmission. tors will be presented here (66, 89, 126, 237, 348, 465).
Therefore, the picture of molecular timing events would not Endocannabinoid signaling influences the propensity of the
be complete without considering metabotropic G protein- synaptic network for plasticity alterations by modifying
coupled receptors (GPCRs). GPCRs exist for many ligands GABAergic inhibitory contributions or by regulating re-
with a variety of subtypes for each ligand coupled to differ- lease probabilities of excitatory synapses (494), thereby re-
ent signaling pathways (30). GPCRs are highly clustered in alizing a form of metaplasticity (4). Metaplasticity means
the vicinity of synaptic membranes and provide a sort of that the previous history of synapses changes the rules of
modulation attuned to a wide range of different input sce- synaptic plasticity (see sect. IIO).
narios. Overall, one of the most important functions of the
metabotropic receptors is the modulation of synaptic From the molecular viewpoint, eCBs, the ligands of CB1
strength in response to previous synaptic activity. Just to receptors, are hydrophobic lipids and do not need to be
give an example, there are at least three different metabo- packed into vesicles. They are produced out of glycerophos-
tropic dopamine receptors that interact with heterotrimeric pholipids and are directly released from the cell membrane
G proteins generally promoting LTP (57), and there are at after the activation of the appropriate enzymes (205). It is
least two other dopamine receptors that activate other G now well recognized that postsynaptic neurons in the hip-
proteins that generally promote LTD (57). The ensemble of pocampus and several other brain regions release eCBs in
GPCRs, G proteins, and cellular signal transduction com- response to a rather strong depolarization. This might also
ponents at a particular synapse varies with age, epigenetic occur after the activation of G protein-coupled receptors

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 659


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

(e.g., mGluR or muscarinic receptors). This results in the include “turning on” mute synapses, as has also been
activation of presynaptic CB1 receptors inhibiting neurotrans- shown in hippocampal granule cell autapses (chemical syn-
mitter release at either excitatory or inhibitory synapses (FIG- apse formed by the axon on its dendrites of the same neu-
URE 4). CB1 receptors are one of the most abundant G pro- ron) (436). Recordings of Ca2⫹ transients in the postsyn-
tein-coupled receptors in the CNS and are predominantly lo- apse implied that presynaptic release sites, which have been
calized on axon terminals (205). In the hippocampus, eCBs are silent previously, can be switched on following LTP induc-
for example mandatory for a certain form of LTD at inhibi- tion (360).
tory as well as excitatory synapses, thereby restricting LTP
(65, 403). They also regulate the heights of LTP directly (305). Taken together, even if NMDA receptor-dependent forms
of LTP have been in the spotlight of experimental research
2. Presynaptic release machinery for LTP induction, also presynaptic components have to be
considered and could further increase the cellular instru-
An important presynaptic parameter to change/affect syn- ments to implement fine-tuned changes in synaptic strength.
aptic strength is the release probability. The process of neu- In addition to the induction mechanisms that are imple-
rotransmitter release is highly regulated, and in principle, mented directly at excitatory or inhibitory synapses, also
each of the highly controlled events leading to neurotrans- other cell types (astrocytes or microglia cells) and neuro-
mitter release would be suitable for change due to altered modulators (such as dopamine, acetylcholine, and sero-
neuronal activity, namely, vesicle recycling, storage and tonin) may be major components of the transition from
mobilization, docking at the presynaptic membrane, prim- short- to long-term memory processes (96, 141, 142, 426).
ing, and fusion (184, 398). Good candidates for presynaptic
regulatory elements are synapsins, which can be phosphor-
ylated via kinases regulated by neuronal activity. Their G. Trans-synaptic Signaling at the Synaptic
phosphorylation alters the tethering of the actin cytoskele- Cleft Contributing to Synaptic Plasticity
ton with synaptic vesicles and regulates vesicles in the read-
ily releasable pool (61). And indeed, synapsin-deficient/ There is also accumulating evidence indicating that the
knockout mice do have a reduced number of vesicles in the space surrounding neurons (peri-neuronal net, PNN) and
reserve pool and show behavioral abnormalities in learning the extracellular space between synapses (synaptic cleft)
tasks (365). Further on the regulation of the different vesi- must be considered as players in processes of activity-de-
cles pools, also the docking and fusion of synaptic vesicles is pendent neuronal plasticity. The tiny space between the pre-
highly regulated. Here, vesicles undergo a maturation pro- and the postsynapse (20 nm) is densely packed with cell
cess to become fusion competent. In this process, SNARE adhesion molecules (CAMs), and it is not an empty space
proteins (soluble NSF attachment protein receptor) come through which neurotransmitters and neuromodulators dif-
into close contact with the plasma membrane and with fuse. The most important CAMs are neurexins, neuroligins,
Ca2⫹ channels. The active zone of the presynaptic mem- cadherins, and CAMs with immunoglobulin domains
brane where the vesicles release their neurotransmitters is (478).
organized by the cytomatrix at the active zone (CAZ),
which is a likely a candidate for activity-dependent changes 1. Neuroligin/neurexins
in the release machinery (150). A particularly good example
is the study by Matz et al. (288) performed in hippocampal The importance of cell adhesion became evident for the
cultures. This study provides good evidence that the num- heterotypic interaction between the presynaptically ex-
ber of CAZ-related proteins present at individual active pressed cell adhesion protein neurexin (NX) and its post-
zones changes fast in response to alterations in neuronal synaptically expressed counterpart neuroligin (NL). Due to
activity (150, 288, 398). At the level of individual signaling a large variety of splice variants, these molecules were orig-
pathways, pharmacological as well as genetic analyses inally thought to provide the potential for a highly specific
show that an increase in presynaptic cAMP is mandatory trans-synaptic combinatorial code during the development
for mossy fiber synaptic plasticity. The same holds true for of the nervous system (239). It became evident that they
cerebellar parallel fibers (reviewed in Ref. 398). Indeed, a also act as modulators of synaptic plasticity. The two NL
couple of independent studies implicated that adenylate cycla- isoforms, NL1 and NL2, are targeted specifically to excit-
ses and subsequent PKA activation result in the phosphoryla- atory (NL1) and inhibitory (NL2) synapses, respectively
tion of CAZ-related proteins leading to the presynaptic induc- (44). NL1 has the ability to modulate synaptic transmission
tion of LTP at hippocampal mossy fiber terminals (reviewed in via NMDA receptor activation (81) and by this means con-
Ref. 321). In line with this is the finding that voltage-depen- trols synaptic plasticity. NL1 is in particular mandatory for
dent Ca2⫹ entry into the presynaptic terminal triggered by the maintenance of synaptic plasticity in the amygdala at
high frequencies of action potentials might directly activate the synapses that express NMDA receptors (194, 215). Here,
adenylate cyclase/PKA pathway. This results in the phos- NL1 knockdown in rats show impaired STDP in thalamic
phorylation of proteins like synapsins (reviewed in Ref. inputs to the amygdala, whereas cortical inputs were not
321). Presynaptic mechanisms for the induction of LTP also affected. These defects in synaptic plasticity result in im-

660 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

pairments in contextual and cued fear conditioning. Dele- 192), and this ensures a direct signaling mechanism to struc-
tion of NL1 also impairs hippocampal CA1 LTP (39, 78). tural changes at synapses. Tang et al. (428) found that LTP
These alterations in synaptic properties after NL1 knock- is compromised if N- and E-cadherin function is disturbed
down in rats (194) are accompanied by impaired spatial via antibodies or specific peptides against the extracellular
memory in a Morris water maze task (215). Interestingly, domains of cadherins.
impaired learning in two water maze tasks following NL1
overexpression in mice and also impaired hippocampal Taken together, cadherins are well suited to act as trans-
CA1 LTP were observed too (78). Changes in synaptic ac- synaptically active sensors of neuronal activity for the pre-
tivity might induce the transport of NX as well as NL to and and postsynaptic neuron to enhance or restrict synaptic
from individual synapses. Evidence for this comes from the plasticity, depending on the precise code of expression and
observation that induction of LTP can be achieved via the the precise type of neuronal activity.
application of forskolin (an activator of adenylate cyclases,
leading to PKA activation), and this results in trafficking of 3. Immunoglobulin superfamily
NL1 to the cell membrane, whereas the opposite, induction
of chemical LTD by stimulation with 3,5-dihydroxyphenyl-
In addition to the Ca2⫹-dependent regulation of cadherins,
glycine (DHPG, a mGluR agonist), causes the internaliza-
also Ca2⫹-independent cell-cell adhesion molecules are im-
tion of NL1 (380).
portant determinants of synaptic plasticity. Here, members
of the immunoglobulin superfamily are important. The
It can be concluded that the expression of the right amounts
most abundant is the neural cell adhesion molecule (NCAM)
of NL1 is crucial for the proper synaptic function and for
and a molecule named L1. Function blocking antibodies
the fine tuning of synaptic plasticity.
against NCAM and also for L1 were able to prevent LTP in the
hippocampus if applied prior to the inducing stimulus (267).
2. Cadherins
NCAM can be modified posttranslationally through the addi-
tion of polysialic acid residues (PSA). PSA-NCAM to NCAM
The cadherin family of adhesion molecules participates in
ratio changes due to neuronal activity, and this alters homo-
processes of synaptic development and plasticity (324, 413;
philic adhesion, a higher PSA-NCAM content increases the
for a review, see Ref. 424). During the early development,
distance between synapses and therefore allows remodeling
mostly E-cadherins (epithelial cadherin) are expressed,
and growth of the synapse (24, 474, 478). PSA-NCAM is
whereas in the development of the nervous system N-cad-
less adhesive compared with NCAM alone. And indeed,
herins (neural cadherin) are expressed at higher levels (for a
removal of PSA from NCAM, which is naturally occurring
review, see Ref. 424). Cadherins form, and this is a very
via the enzyme EndoN (an endoneuraminidase), interferes
interesting feature with respect to synaptic plasticity, Ca2⫹-
with the induction of LTP as well as LTD (390).
dependent homophilic bonds in the synaptic cleft (50, 181,
425, 450); indeed, cadherins are named with reference to
Taken together, the interesting point of regulation is here,
“calcium-dependent adhesion.” The main question is how
that the strength of adhesion can be calibrated via the re-
cadherins sense changes in neuronal activity and if they
moval or addition of PSA, regulated via an activity-depen-
might be able to transmit signals from the extracellular
dent enzyme.
space to the cytosol. With respect to these questions, it is
noteworthy that Kim et al. (217) could visualize via genet-
ically encoded fluorescence resonance energy transfer 4. Extracellular matrix
(FRET) the spatiotemporal dynamics of the interaction of
N-cadherins across the synaptic cleft. The results revealed a In addition to CAMs, the extracellular matrix (ECM) is
fast and partial loss of homophilic interactions upon chela- another factor influencing synaptic plasticity. Most syn-
tion with an increased level of extracellular Ca2⫹. The usage apses are surrounded by a dense meshwork of ECM. The
of a version of the cadherin molecule with a deteriorated ECM contains proteoglycans and glycoproteins of glial or
adhesive activity showed a faster loss of homophilic inter- neuronal origin. A remarkable feature of the ECM in CNS
actions. This approach could also tackle the question of is its netlike appearance (PNN) (60), which is constructed
how the signal might be transmitted to the cell interior, by postnatally in the first 5 wk of mouse development (229).
showing that the Ca2⫹-sensitive modulation of cadherin The ECM is considered to be a stabilizer and is most likely
interactions is transmitted via intracellular ␤-catenin. These important for the maintenance of a memory engram in the
cadherin-␤-catenin complex by this means link synaptic ac- adult CNS possessing inhibitory effects on structural rear-
tivity with processes of synaptic plasticity. Cadherins are rangements and axonal outgrowth (128, 140). Depending
therefore molecules that are responsible for transmitting on its precise composition, ECM might control the diffu-
dynamic information about the extracellular environment sion of receptors in a lateral manner inside the cell mem-
to both cells that form a synapse. This can occur on a fast brane and therefore regulates the proportion of synaptic
time scale in minutes. The highly conserved cytoplasmic and/or extrasynaptic receptors (92, 151). With this means
localized cadherins are associated with the cytoskeleton (3, the netlike structure of the ECM might represent an addi-

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 661


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

tional determinant for synaptic plasticity by restricting pro- What is a possible mechanism for BDNF to increase the
cesses of plasticity, which could in the end also support the likelihood of synapses to undergo activity-dependent
expression of stable engrams. strengthening? The signaling cascade seems to involve
the receptor for BDNF, that TrkB receptor. And indeed,
if the interaction of BDNF with its TrkB receptor is
H. BDNF: It Is the Modulator That Mediates blocked by TrkB-receptor bodies (116) or via anti-BDNF
Important Aspects of Long-Term Storage antibodies (62), hippocampal LTP is strongly impaired.
Using a Cre-deleter mouse line to delete the TrkB recep-
In addition to classical neurotransmitters, also neurotro- tor from the genome only postnatally and only in the
phins might be important players in promoting the likeli- forebrain region, CA3-CA1 LTP was impaired (297,
hood of plastic changes in neuronal networks. Most of all, 298). Furthermore, it was asked if BDNF is an acute
BDNF is implicated in processes of synaptic plasticity. The mediator of synaptic changes or if it is a permissive mod-
transcription of BDNF and its release have been shown to ulator (337, 350)? BDNF is not needed under all circum-
be activity dependent (252, 262; for reviews, see Refs. 431, stances and stimulus protocols (see, e.g., Ref. 473). With
432). BDNF is also a major player in changing functional respect to the discussion about the nature and function of
synaptic properties during the development of the CNS as “mediators” and “modulators” of synaptic plasticity,
well as in the adult nervous system (for reviews, see Refs. BDNF is an instructive example. In our view, depending
144, 337). There is also evidence that BDNF affects neurite on the brain area, the type of neuron, the local connec-
outgrowth and the differentiation of certain neuronal sub- tivity, and the required learning task, BDNF most likely
types. Taken together, BDNF was hypothesized to be a plays a dual role: it acts both as a mediator and as a
major player for the transformation of functional into
modulator of plasticity. It is noteworthy that quite in
structural changes (231, 289, 427). Genetic approaches elu-
contrast to the findings of the TrkB receptor signaling,
cidated that BDNF knockout mice exhibit a significant re-
which mediates LTP, the pan-neurotrophin receptor
duction in synaptic plasticity, most of all in the induction
p75NTR, which also binds BDNF, supports negative syn-
and maintenance of LTP (232, 343, 351). Remarkably, this
aptic plasticity like LTD (366, 470).
impairment in hippocampal LTP could be rescued by the
virus-mediated local expression of BDNF (233) or via the
There is no “exclusive memory molecule,” and also BDNF
application of recombinant BDNF to hippocampal slices
is working in concert with other molecular signaling path-
from BDNF knockout mice (343). In a gain of function
way. In addition, it is a relevant question if BDNF indeed
experiment, the application of BDNF to wild-type slices
belongs to a group of molecules which are major players
increased synaptic strength and by this means increased the
(instructive mediators) or if BDNF is more like a permissive
probability of LTP induction (145, 199, 201). Likewise,
weak electrical stimulation (which alone would not lead to modulator with a more or less supporting role. By this
LTP) together with the local application of BDNF, con- means it would only be expressed in neurons to support
verted short-term plasticity into LTP (235). Also synap- indirectly processes of synaptic plasticity. If this would be
totagmin-IV knockout mice, which release higher amounts the case, BDNF would not be directly incorporated in a
of BDNF, show enhanced LTP (82). In addition to these rather linear signaling line, leading to functional or struc-
slice experiments, there is also evidence that BDNF is me- tural activity-dependent changes. With respect to these
diating learning behavior in vivo as it is upregulated in brain thoughts, experiments in which endogenous BDNF is
areas that are involved in a specific learning task (433). blocked during the induction period of LTP are telling.
Furthermore, BDNF⫹/⫺ mice and conditional tropomyo- Two studies have been published which used a TrkB-IgG
sin-related kinase receptor B (TrkB, which binds BDNF) fusion protein (TrkB-Fc), which scavenges (and by this
knockout mice show deficits in hippocampus-dependent means neutralizes) BDNF with high affinity, and function
learning (298). Notably also studies on humans indicate blocking TrkB receptor antibodies have been used to
that BDNF is involved in processes of learning and infor- block binding of BDNF to the receptor. In both cases,
mation storage. Here the genetic evidence comes from a LTP was impaired in acute slices (116, 201). The inter-
common single-nucleotide polymorphism in the human pretation was complicated by the fact that other neu-
BDNF gene which leads to the substitution of a methionine rotrophins NT3 and NT4/5 can also bind to the TrkB.
(Met) for the valine (Val) at codon 66 (Val66Met). This has This was resolved by Chen et al. (62), who used specific,
the consequence that BDNF secretion is impaired (97, 161, monoclonal antibodies against BDNF or alternatively
468). This is most likely due to misfolding of the BDNF against NT3 at the time point of LTP induction. Under
protein and leading to a less efficient sorting of the these circumstances, only anti-BDNF antibodies were
proBDNF protein into the Golgi apparatus, impairing espe- able to impair LTP. This is supported by experiments in
cially the activity-mediated release of BDNF. The Val-Met which endogenous BDNF was inactivated by “uncaging”
genotype leads to a compromised performance in hip- a BDNF-function-blocking antibody (monoclonal) with
pocampus-dependent learning tasks in humans (97, 161, a light stimulus (which released the antibody-inactiva-
468). tion compound from the antibody) right at the time of

662 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

LTP induction. Only when the BDNF-function blocking an elegant way to solve the problem of how a neuron with
antibody was uncaged by a light stimulus was LTP im- 10,000 synapses can maintain changes in a few specific/
paired (62). These observations showed that indeed selected synapses without affecting others (417, 418).
BDNF directly affects synaptic plasticity, and it strength- But how does a neuron know which synapse shows
ened the view that indeed it might be an instructive me- changes in synaptic strength and has been activated? One
diator, and not just a permissive modulator. solution would be that a tag is set to specifically mark a
certain synapse. Alternatively but not exclusively, the
In addition, it could be shown that BDNF does not only proteome of dendrites and spines might be changed lo-
increase the likelihood of LTP induction but supports also cally at the site of synaptic plasticity alteration via local
L-LTP maintenance and by this means long-term memory. protein translation in dendrites. The presence of protein
Experiments here include studies with BDNF knockout translation in dendrites was first proposed after polyri-
mice (234) as well as results from experiments in which bosomes had been observed in dendrites (416). In addi-
BDNF was applied directly (201), experiments involving tion, several forms of LTP are protein synthesis depen-
TrkB-Fc (116) and conditional TrkB knockout mice (298). dent and could still be maintained, when the soma was
The long-lasting effects of BDNF might be mediated via its cut off from dendrites, but not if translation was phar-
effect on local dendritic translation, and this might produce macologically blocked (176, 200). In addition, it was
proteins that promote cellular processes of memory consol- reported that mRNAs in the soma can bind to proteins,
idation (46). which shuttle them into dendrites (211, 212). These pro-
teins include Staufen, ZBP1 (Zipcode binding protein 1),
However, BDNF action might not be restricted to glutama- and hnRNPA2 (heterogeneous nuclear ribonucleopro-
tergic synapses; it might also be involved in changing the tein A2). They form together with the mRNA a kinesin-
excitation-inhibition balance (for a thorough review, see dependent transport granule that is transported along
Refs. 144, 338). This might be achieved due to the fact that microtubules into the dendrites (211). How the final des-
the GABAergic influence on excitatory neurons is weak- tination is determined is an open question. It might also
ened. be possible that the transport is not directed to specific
spines, but that the transport granule is stopped and sta-
Taken together, BDNF seems to be not part of the core bilized at activated postsynaptic sites (211).
mechanism of LTP induction (even complete BDNF knock-
out mice show residual LTP), but to maintain LTP (L-LTP) The mRNAs are not only transported (or captured) close to
and to fully potentiate a synapse (heights of LTP) BDNF active spines, they also need to be translationally regulated,
seems to be an important modulator of synaptic plasticity adding another layer of complexity and regulation to local
(491; for reviews, see Refs. 144, 338, 488). The functions of protein synthesis in dendrites. One possible scenario is de-
BDNF in orchestrating molecular events leading to memory picted in FIGURE 6. mRNA is transported into dendrites,
storage are depending on the developmental stage, the spe- where cytoplasmic polyadenylation element binding pro-
cific cell type, and also the brain region (488). Overall, tein (CPEB) binds to its 3’ untranslated region (3’UTR),
BDNF is most likely an instructive player in the process of thus inhibiting its translation (FIGURE 6). After the synaptic
functional strengthening of synapses. activation of specific signaling cascades, CPEB can be phos-
phorylated and now recruits the protein machinery neces-
It should also be noted that modulators of synaptic plastic- sary to increase the length of the poly(A) tail of the mRNA
ity are as important as mediators. Examples for this state- (7). This enables PABP [poly(A) binding protein] to bind to
ment are modulatory neurotransmitter systems like acetyl- the mRNA, and this recruits a new group of proteins to
choline, serotonin (5-HT), dopamine, and epinephrine foster local translation (FIGURE 6) (7). Among these trans-
(noradrenaline), which are involved in attention and moti- lation promoting molecules are eukaryotic translation ini-
vational aspects of behavior and have a great influence on tiation factor 4 gamma (eIF4G) and eukaryotic translation
the capacity of long-term memory storage. initiation factor 4E (eIF4E), which initiate the translation of
proteins in a combined effort right at the side of increased
synaptic activity (363, 418).
I. Local Protein Synthesis and Degradation
In the following we present a small list of molecules that
In addition to posttranslational modifications like phos- have been instrumental to understand how protein transla-
phorylation and dephosphorylation of plasticity associ- tion is shaping processes of functional and structural plas-
ated proteins occurring very fast, long-lasting memory ticity.
storage also needs the synthesis or the degradation of a
specific set of proteins. New synthesis can occur either via 1. Arc/Arg3.1
transcription (signaling to the soma, synthesized proteins
are transported back to the activated synapses) or via One of the first discovered examples of activity-regulated
local translation in dendrites. Local mRNA translation is gene transcription and translation is Arc/Arg3.1 (419, 420).

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 663


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

It could be shown that indeed Arc/Arg3.1 likely contributes delicate balance of protein degradation and synthesis is
to L-LTP and to processes of memory consolidation (349, mandatory to transform E-LTP into L-LTP (123).
394). Its translation is robustly induced within the spine by
plasticity-promoting stimulation, and the protein is specif- In summary, net production of new proteins via local trans-
ically targeted to stimulated synaptic sides. Arc/Arg3.1 gene lation in dendrites, possibly right at the side of activity-
targeted mice fail to form long-term memories. This holds dependent changes at synapses, is as important for informa-
true for implicit as well as for explicit learning tasks. This is tion storage as is the regulated degradation of certain regu-
in contrast to short-term memory which stays intact (349). latory components of signaling cascaded in synapses and
This mechanism to support long-lasting plasticity is medi- dendrites.
ated by local translation of plasticity-relevant proteins, like
Arc or CaMKII protein (459, 472). Interestingly, the J. Synaptic Tagging and Capture Hypothesis
mRNAs encoding GluR1 and GluR2 (AMPA receptor sub-
units) have both been found in dendrites of pyramidal neu- In addition to processes of local protein synthesis (transla-
rons of the hippocampus, and they are indeed regulated by tion), transcription in the nucleus has also been shown to be
synaptic activity (148, 193). An interesting twist here is that mandatory for long-term memory storage (196). But this
local eEF2-dependent translation of cytoskeletal regulator leads to the following puzzle: on the one hand, long-term
Arc and its mRNA trigger endocytosis of AMPA receptors memory storage needs transcription in the nucleus (196),
during mGluR-mediated hippocampal LTD (67, 458). and on the other hand, it requires a local specificity for
stimulated synapses (320). But how do the gene products
2. miRNAs generated in the nucleus find the appropriate synapse? A
pyramidal neuron in the hippocampus might have up to
In addition to the inactivation of mRNA interacting pro- 10,000 synapses, and during a certain learning event, plas-
teins that usually inhibit translation, also microRNAs ticity is induced only in a subset of these synapses. One
(miRNAs) seem to play an elegant role in the regulation of solution to this problem is provided by the synaptic tagging
local protein synthesis. Each miRNA, only 22 nucleotides and capture (STC) hypothesis (127, 283). This hypothesis
long, can in principle regulate literally many hundred tran- was formulated as a consequence of the results of experi-
scripts and might therefore orchestrate the expression of ments recording from two hippocampal pathways at the
many genes. The mechanism of miRNAs action is rather same time (127). For this, two CA3 synaptic inputs, shown
simple: via base-pairing with complementary sequences to be independent from each other and to converge on the
within coding mRNA they inhibit translation at ribosomes same population of CA1 hippocampal neurons are em-
of particular proteins (383). The relatively higher abun- ployed. A weak tetanus (a stimulation normally leading to
dance of miRNAs in the brain, some are even restricted to E-LTP) is converted into L-LTP, if shortly after its applica-
the brain, points to their important role in the CNS (for a tion a second, independent pathway is stimulated with a
review, see Ref. 383). miRNAs are particularly well suited strong tetanus. These observations are the core of the STC
to control distally localized mRNAs and their translation. hypothesis and can be explained as follows. The first syn-
Components of the RNA-induced silencing complex (RISC) aptic pathway by itself is not able to activate local protein
machinery have been found in dendrites and synapses and synthesis in the respective dendritic spine as it received te-
can be altered by synaptic activity (14). tanic stimulation that induced E-LTP. But the induction of
E-LTP is able to form a “tag” marking the activated synapse
in a specific manner. For a distinct time period, this tag can
3. Degradation
“capture” the newly synthesized plasticity-related proteins
(PRPs). The activation or synthesis of PRPs is stimulated
In addition to transcription and translation, which lead to when LTP is induced in a neighboring synapse. By this
an increase in the amount of certain proteins in synapses, means, certain amounts of the PRPs can enter the tagged
protein degradation can reduce the copy numbers of specific synapse so that E-LTP is converted into L-LTP (378). In a
proteins. This has been shown for the proteasome-depen- nutshell, a synaptic “mark” or “tag” induced via a transient
dent alterations in the composition of proteins in the post- event, sequesters PRPs from a nearby strong synapse to
synaptic density (PSD) of principal neurons in the hip- consolidate changes in synaptic weight at stimulated syn-
pocampus after LTP induction (98). Like dendritic transla- apses (and by this means maintains synaptic input specific-
tion, protein degradation can also be regulated by changing ity) (127). This concept is so far to our knowledge the most
the local proteome. The ubiquitin proteasome system may comprehensive model to explain late associativity in pro-
be the key regulator of protein degradation, and its compo- cesses of activity-dependent synaptic plasticity (15).
nents exist in dendrites and also at synapses (FIGURE 6). In
addition, proteasomes seem to relocate from dendritic 1. Molecules involved in synaptic tagging
shafts into spines within minutes possibly altering AMPA
receptor trafficking and degradation within spines (for a It is noteworthy that in hippocampal neurons BDNF was
review, see Ref. 269). In 2004 it was discovered that a shown to regulate the local translation of PRPs, including

664 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

Arc/Arg3.1, which is in part involved in controlling the spatial memory (248, 451). It could even be shown that
turnover of the actin cytoskeleton (294, 481; see also for once the PKM␨ inhibitor has been removed, the memory is
a review, Refs. 46, 175). These results suggest that BDNF still erased, but new spatial memories can be formed and
is indeed a major PRP, which also stimulates the produc- stored (341). Thus PKM␨ is the first identified molecular
tion of other PRPs. This notion is supported by experi- component of the storage mechanism of associative long-
ments from Barco et al. (16). This group performed a term memory in the mammalian brain (373). Besides
STC-type two-pathway experiment in hippocampal slices PKM␨, several kinases act in concert to activate new protein
of BDNF heterozygous knockout mice having only a lim- synthesis for L-LTP and the formation of long-term mem-
ited amount of BDNF (16). They stimulated two indepen- ory. For example, selective inhibitors of protein kinases like
dent inputs to the same part of the CA1 region and found CaMKII, MAP kinases, PKA, and mammalian target of
evidence that synaptic tagging and capture was signifi- rapamycin (mTOR) block the synthesis of PKM␨ in LTP
cantly impaired in the BDNF heterozygous knockout (364).
mice (16). Following up on these results it could be
shown that BDNF is only mandatory in specific forms of
3. Sensorin
long-term memory formation and that its role is highly
specific and indeed instructive for synaptic plasticity
(375). The authors used a version of L-LTP, which is Overall, there is indeed good evidence for a mechanism in
induced locally (175) and by this means allows the inves- which generally available proteins enter the activated syn-
tigation of a synapse-specific L-LTP achieved by a weak apses only after synaptic stimulation (330). Synaptic stim-
tetanus that does not activate transcription in the nu- ulation could induce protein synthesis restricted exclusively
cleus. This form of locally induced L-LTP is exclusively to the activated synapse, thus forming the “tag” (146, 418).
achieved via BDNF-mediated local protein synthesis But this scenario requires that specific mRNAs undergo
(175). Under these circumstances the newly synthesized locally restricted translation at stimulated synapses only
PRPs can only be used within small dendritic portion and during processes of long-lasting synaptic plasticity. To ad-
will not be shared with more distant compartments dress this issue, Wang et al. (457) have used the well-estab-
(375). This indicates that PRPs can be spatially restricted lished system of Aplysia sensory neurons co-cultivated with
to a cluster of synapses, e.g., on a certain dendritic motor neurons (196). This monosynaptic connection is a
branch. Surprisingly, theta-burst stimulation (TBS)-in- central part of the gill-withdrawal reflex in Aplysia. Here,
duced LTP is prone to cross-capture, a process that shows the repeated application of 5-HT can lead to a long-term
a positive associative interaction of LTP with LTD, in facilitation (LTF), the Aplysia equivalent to LTP, whereas
which PRPs are shared between these different mecha- the application of the neuropeptide FMRFamide leads to
nisms of synaptic plasticity (317). In addition, the tag synaptic depression (LTD). To monitor local translation
that is set during E-LTD induction exploits BDNF as a during the conversion of short- to long-lasting synaptic
PRP for LTD maintenance, whereas other signaling path- plasticity, the researchers generated an elegant translational
ways involved in synaptic plasticity relied on the activity reporter utilizing the mRNA from sensorin, a sensory neu-
of PKM␨, but not on BDNF, as has been shown in Ref- ron-specific peptide neurotransmitter the mRNA of which
erence 317. localizes to distal neuronal processes. Sensorin is enriched
at synapses, and its translation is necessary for 5-HT-in-
In this context, experiments are needed for addressing the
duced LTF. Indeed, the authors only used the 5’ and
question of how different dendritic compartments with a
3’UTRs of sensorin mRNA that were fused to the coding
specific plasticity threshold act as functional units for stor-
region of the photoconvertible fluorescent protein dendra2
ing long-term memory in the neuronal networks. The term
(152). By this means, the authors could show that in neu-
plasticity threshold refers to the capacity of a synaptic unit
rites, disconnected from the soma, green dendra2 became
to process incoming information. It was shown that hip-
pocampal neurons have different synaptic compartments visible only at stimulated synapses that underwent LTF af-
and that independent “synaptic units” or “clusters” exist ter local 5-HT superfusion, but not at unstimulated syn-
within these compartments (146, 375). Now, it becomes apses and not when LTD was induced. One central finding
clear that future experiments should not exclusively con- of this study was that regulation of local translation re-
centrate on a certain synapse but rather on synaptic com- quires chemical synaptic transmission. Furthermore, this
partments. study also shows an example in which Ca2⫹ changes in the
postsynaptic compartment are correlated with local protein
2. PKM␨ translation in the presynapse. Therefore, a trans-synaptic
signal is required for translational regulation in the presyn-
In addition to BDNF, the atypical protein kinase PKM␨ is aptic side. It will be interesting to see what the hunt after the
discussed to act as a synaptic tag for stimulated synapses. In retrograde signaling will bring to light and if such mecha-
particular, it could be shown that PKM␨ inhibition in the nisms are used in hippocampal and other CNS structures of
hippocampus hours after learning prevents the retention of mammals as well.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 665


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

Local and precisely targeted protein synthesis is one solu- changes are maintained for a long period of time (consoli-
tion to the riddle of synapse specificity of learning events. dation). This memory barrier is of special importance, since
Another, not mutually exclusive, alternative explanation it is a hallmark of animal and human cognitive abilities to
would be to target ubiquitous available proteins into the select salient information to separate this from a noisy back-
stimulated synapse only. This latter idea was tackled by ground. Moreover, it might be even more important to
Okada et al. (330). Under resting conditions, Vesl-1S consolidate the most important facts and features and pro-
(Homer 1a), one of the PRPs required for long-term fear tect them from change, like it is indeed happening during
memory, was absent from the postsynaptic side of a stimu- processes of reconsolidation (314).
lated synapse (in primary cultures of hippocampal neu-
rons). Okada et al. (330) used a fluorescence marker tagged 1. Nogo-A
to Vesl-1S to detect the movement of the corresponding
protein. Only after NMDA receptor activation, Vesl-1S Among the molecules known to restrict neuronal plastic-
could enter spines in an input specific manner. Also Wang et ity are the pan-neurotrophin receptor p75NTR (366, 470),
al. (457) could show that the entry of proteins into spines is adhesion molecules (392), endocannabinoids (64, 305),
tightly controlled by molecules that function as synaptic part of the ephrin family (224), semaphorins (342), and
tags. This finding opens the possibility of a synaptic tag as a the myelin-associated neurite growth inhibitor family
“molecular gatekeeper” and how it can “sense” the spatial and their receptors (385). As an example we would like
and temporal strength of the incoming information. It will to focus on the myelin-associated neurite growth inhibi-
be exciting to see if molecules located at the spine neck, like tor Nogo-A and its receptors NgR1 (Nogo-66 receptor 1;
synaptopodin (84), could fulfill this function (see, e.g., Ref. Ref. 124), S1PR2 (sphingosine 1-phosphate receptor 2;
230). Ref. 208), or PirB (reviewed in Ref. 385). They are ex-
pressed pre- and postsynaptically (207, 251, 261). Re-
Overall, these studies could demonstrate that translation cently, they have been shown to restrict neuronal archi-
and transportation can indeed be restricted to specific syn- tecture (346, 490) and to control synaptic plasticity in
apses. However, data from acute slices of mice support the the developing and also in the mature CNS (83, 208, 249,
view that, under a more complex network situation, there is 299, 353). Moreover, NgR1 has been shown to limit
additional cross-talk and interaction between synapses like experience-driven plasticity in the visual cortex (290).
cross-tagging (377) or competition for PRPs (122, 376). Furthermore, there is accumulating evidence that it also
Experiments presented here (330, 457) are so far the most regulates experience-dependent dynamics of spines as
convincing studies how synapse specificity can be main- well as the axonal varicosities in the somatosensory cor-
tained during the implementation of associative memory tex (8). Indeed, Nogo-A might acutely regulate structural
storage along the line of the synaptic-tagging and capture plasticity due to its influence on the actin cytoskeleton
hypothesis. dynamics via Rho/Rock signaling to restrict plasticity at
individual dendritic spines of hippocampal pyramidal
neurons within minutes (386).
K. Stabilization
However, it is not enough to concentrate on single mol-
Change of synaptic function and structure is one side of ecules promoting stabilization, but of course one has to
the coin, but how can change last and how can an engram take into account that there might be an extensive cross-
be stabilized in an ever-changing neuronal network? talk between signaling pathways promoting plasticity
Long-term memory storage requires on the one hand the and those that promote stability (or restrict possible plas-
activation of positive regulators, which favor informa- tic changes). Indeed, there are experiments suggesting an
tion storage, and on the other hand the removal of re- opposing interaction between inhibitors of plasticity and
straining, negative-acting factors preventing it (2). Inhib- neurotrophic factor signaling pathways. Pretreatment of
itory pathways act at different levels all the way from dissociated hippocampal neurons with BDNF signifi-
synapses to the nucleus of a neuron. Following synaptic cantly reduces the inhibitory effect of myelin, and this is
receptor activation, downstream signal amplifiers, such phospho-CREB dependent (136). On the other hand,
as PKA, are controlled by an inhibitory subunit. Also the Nogo⌬20 (one domain of the Nogo-A molecule) reduces
transactivation function of some transcription factors can CREB activation (190) and decreases pAKT (86). In line
be reduced by inhibitory regulators. These “memory sup- with this, the other Nogo-A domain, Nogo66, reduces
pressor genes” (2) are good checkpoints for long-lasting the BDNF-mediated activation of pAKT (353). In an-
memory storage. These molecules might function as hubs other study it has been shown that the BDNF/TrkB acti-
determining the rate, the occurrence, and the specificity at vated Erk1/2 signaling pathway is increased in NgR1
which changes in neuronal connectivity are either imple- knockout hippocampal neurons (353), and NgR1 knock-
mented, maintained, or prevented. Thus memory control- out neurons are also more sensitive to another growth
ling proteins ensure that only relevant features of an event factor pathway, namely, to FGF2-FGFR signaling (249).
are stored (filter function) and that the synaptic weight It is also noteworthy that growth factor and Nogo-A

666 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

signaling pathways affect the mTOR complex 1 1. Astrocytes


(TORC1) in an antagonistic fashion (for details, see Refs.
344, 353). Specifically mTOR-dependent protein trans- Astrocytes got their name indeed from their star-shaped
lation is regulated in an antagonist fashion via phospho- morphology. The interest in their involvement in processes
CREB, promoting or inhibiting transcription (344, 353). of synaptic plasticity comes from the fact that processes of
Finally, Nogo-A in concert with its receptors (NgR1, protoplasmic astrocytes completely enwrap synapses in the
S1P2R, PirB) limits activity-dependent plasticity and grey matter of the CNS. In the hippocampus, astrocytes
growth-dependent processes, thereby leading to the sta- enwrap ⬃57% of all synapses (467). As far as it is known,
bilization of neuronal assemblies (either developmentally usually a single astrocyte enwraps literally thousands of
or during processes of memory consolation in the mature synapses (up to 6,000 synapses; Ref. 210). Astrocytes signal
nervous system) (for recent reviews, see Refs. 386, 489). to neurons via glioneurotransmitters, like D-serine, a coago-
Taking away the molecular synaptic plasticity brakes al- nist of the NMDA receptor. And indeed, it could be shown
lows the induction of extensive structural and functional that the release of D-serine is a necessary ingredient to pro-
rearrangements and promotes compensatory growth mote hippocampal LTP induction (166, 479). In addition,
processes after an injury of the CNS. However, it is im- astrocytes express a large array of neurotransmitter recep-
portant to keep in mind that this could be a dangerous tors, like glutamatergic, purinergic, GABAergic, cholin-
endeavor, since it might facilitate unwanted and unnec- ergic, and adrenergic receptors (for a review, see Ref. 106).
essary (and probably even maladaptive) neuronal con- Specifically, astrocytes express AMPA and NMDA recep-
nections. tors at their surface, and the removal of astrocytic AMPA
receptors leads to the retraction of astrocyte processes from
Also of note, at some glutamatergic connections, synaptic synapses and to pronounced deficits in motor coordination
plasticity is rather difficult to induce, and it is suggestive to in the adult brain (371). Also, in astrocytes the activation of
speculate that this might be linked to expression of plastic- typical neurotransmitter receptors can lead to Ca2⫹ waves
ity-limiting molecules at the end of CNS development. For within and between astrocytes. An increase in the intracel-
example, within the entorhinal cortex, many different pro- lular Ca2⫹ concentration boosts the release of D-serine and
tocols failed to induce LTP, whereas other studies showed coactivates NMDA receptors (179, 244). As a side note, this
LTP like effects (76, 361, 483). has the potential to explain why there is a correlation be-
tween glial coverage of synapses and LTP in the supraoptic
Molecules that limit synaptic plasticity may therefore play a nucleus of mice (29, 332).
central part in processes of memory formation, especially
when it comes to regulating the association between events However, increases in Ca2⫹ in the cytosol of astrocytes
or facts. The inhibitory processes might be activated in sit- following neuronal activity leads to the release of a whole
uations of CNS injury (e.g., infection, stroke) to prevent plethora of signaling molecules, most prominently ATP,
that the ongoing remodeling processes might endanger es- glutamate, and also tumor necrosis factor (TNF)-␣ (31,
tablished structures and functions of adjacent neuronal net- 111, 345, 414). In return, neurons can supply D-serine
works (385). They are in a way reminiscent of tumor sup- (204, 300). The signaling between neurons and astro-
pressor genes that integrate a variety of molecular signals cytes can also influence the morphology of astrocytes. In
into a single cellular response and help to control cell divi- this context, it has been shown recently that astrocytes
sion. also undergo structural rearrangements after LTP induc-
tion, especially at the perisynaptic astrocytic processes
L. The Tripartite Synapse (29).

So far we have considered the storage of information as a In addition, subtypes of astrocytes and microglia cells are
solely neuronal endeavor. But indeed, the orchestration of involved in structural changes of synapses, e.g., in synapse
memory manifestation is even more complex than “just” elimination and pruning (69, 336). On the signaling level it
the CA3-CA1 excitatory synaptic connection. In addition could be shown that the tyrosine kinase EphA4 on the side
to the pre- and postsynaptic side of a synapse, the inhibitory of excitatory pyramidal cells of the hippocampus interacts
and modulatory inputs to the pre- and postsynaptic neurons directly with the ligand ephrin-A3, which is located directly
and the myriads of molecules therein, also glia cells (astrocytes on processes at synapses from astrocytes (311; see also Ref.
and microglia cells) have to be considered as regulatory ele- 117).
ments in changing synaptic strength and during the mainte-
nance of functional and structural changes (FIGURE 7) (for a 2. Microglia
review, see Ref. 335).
In addition, there is increasing evidence that also microglia
Here we would like to consider glia cells and their contri- cells, the immune cells of the brain, are involved in the
bution to information storage. First of all astrocytes should elimination of synapses (339). Also oligodendrocytes have
be considered. been implicated in processes of plasticity, since the myeli-

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 667


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

nation of axons can change due to changes in neuronal M. Function Follows Form? Structural
activity even in the mature CNS and might therefore further Plasticity and Memory Consolidation
increase the cellular arsenal of plasticity mediating cells in
the CNS (113, 405). One essential arena for future experi- Until this point we have mainly considered the singular
ments is to determine how specific glia cell-secreted mole- contributions of molecular, cellular, as well as network
cules can cross-talk with the neuronal signaling to regulate, events that are correlated to functional Hebbian plasticity
promote, or restrict activity-dependent plasticity. In addi- by which synaptic strength is modulated during and after
tion, it would be important to elucidate how glia cells re- the learning events. However, synaptic networks are not
spond to changes in neuronal activity to secrete molecules only changed and rearranged due to functional modifica-
that are important for functional or structural changes at tions, they are also modified at the structural level, and
synapses. therefore, neuronal connectivity can be remodeled through-

A B
Nucleus
PRESYNAPTIC CRE CRE
TERMINAL
ASTROCYTE
PROCESS
P
Substance SYNAPTIC cAMP MAP
release
CLEFT kinase kinase
Glutamate
transporter
ECM PSA-
cAMP
Calcium PSA- NCAM
rise NCAM

NCAM Adenylyl
PRESYNAPTIC NCAM cyclase
Neuro-
Neuro- POSTSYNAPTIC
AXON login
login
DENDRIC
POSTSYNAPTIC TERMINAL EphBx
TERMINAL
EphB
Endo-
SPINE
cytosis
Neurotransmitter
uptake
Degradation PRP
Neuro- Fusion and (plasticity-
transmitter related
exocytosis proteins)
molecules
Intracellular
NMDAR PSD AMPAR
Regulators
VSCC
zone

Vesicle Exo- Endo-


mobilization Docking cytosis cytosis
and priming
C Synaptic
e

vesicle P
Activ

AMPAR
Neurotransmitter PRESYNAPTIC AMPAR
molecules AXON
Synaptic TERMINAL
vesicle
Lateral
diffusion
Neurotransmitter
Vesicle uptake
mobilization Substance Calcium
release rise
Endocytosis
B PSA-
Neuro- NCAM
A
Activ C EphB login NCAM
e
zone
ASTROCYTE
Docking and Fusion and
priming exocytosis SYNAPTIC
VSCC CLEFT ASTROCYTE
AMPAR
PROCESS
NMDAR

PSA-
P EphB Neuro-NCAM NCAM
login
Lateral PSD
diffusion
Exocytosis POSTSYNAPTIC Modulatory
DENDRIC input
AMPAR SPINE
Endocytosis (Dopamin, ACH, 5'TH)

Degradation

Intracellular
pool
Inhibitory
modulation
(e.g at the soma)
Local Trancription/
translation Translation
(Soma)

668 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

out life. These structural changes include not only the for- structural changes, it is noteworthy that LTP induction
mation of new synapses and their stabilization but also their and maintenance (133, 236, 238) indeed depend on the
elimination. Structure and function of neurons have a re- specific modulation of actin dynamics within dendritic
ciprocal relationship, and spines on dendrites are a partic- spines (331). And the induction of LTD resulted in spine
ularly good example of this fact. Inside the spine, actin retractions (301), whereas LTP induction evoked growth
filaments (F-actin) are highly enriched providing the struc- of spines (301) (FIGURE 5). The highly concentrated actin
tural base for stability as well as for changes in morphology in dendritic spines provides a skeleton for dynamic re-
(115, 286, 287). In turn, the actin cytoskeleton is linked via modeling of spine size and shape (114; reviewed in Ref.
a complex network of proteins to extracellular signals to 367). Actin is highly enriched in the head of spines and
tightly control spine morphology including the coupling of shows a high degree of dynamics (114, 165; reviewed in
functional synaptic changes to structural changes at den- Ref. 41). In line with this, inhibition of LTP also pre-
dritic spine, presynaptic release side (boutons), dendrites vented changes in the dynamics of actin polymerization
themselves, and axons (reviewed in Refs. 59, 379, 487). in spines (255). Moreover, processes of activity-depen-
Changes in the morphology initiated by activity-dependent dent synaptic plasticity leading to LTP induces a shifts
synaptic plasticity events are termed structural plasticity.
from G-actin (globular actin, monomers) to F-actin (fil-
These changes are of functional and behavioral conse-
amentous, linear polymer microfilament). This change in
quence, and they remodel the fine-grained architecture of
the G/F-actin ratio goes hand in hand with an enlarge-
neurons and therefore also the connectivity of networks or
ment of the spine head, whereas processes that lead to
subsets of neuronal nets throughout life.
LTD shift the ratio to G-actin resulting in destabilization
1. Structural spine plasticity of the actin cytoskeleton and spine shrinkage (133, 255,
331, 493, 286, 427). Under in vivo conditions it was
We will focus here on structural changes of spines (for shown in the murine motor cortex that intensive motor
extended reviews, see Refs. 43, 379). Activity-dependent learning leads to structural changes in dendritic spines as
synaptic plasticity includes changes in the size and shape well (477). The extent of these changes can even be highly
of dendritic spines as well in the overall number of spines, correlated with the behavioral improvement in the spe-
and these structural changes are highly correlated to cific task, suggesting a crucial role for structural plastic-
learning processes (103, 275, 437; for reviews, see Refs. ity during learning processes (477). In this context, it is
59, 172). Moreover, increasing evidence indicates that an interesting notion that BDNF supports LTP expres-
functional changes at synapses such as LTP and LTD are sion by increasing the dynamics of cytoskeletal changes
associated with spine shrinkage or spine growth (427) or of spines (348). In addition, the BDNF-TrkB signaling
even the growth of new spines (103, 275, 437) possibly pathway induces the local dendritic synthesis of several
containing functional synapses (243, 315, 495) (FIGURE proteins known to regulate the cytoskeleton, like Arc,
5). With respect to the interdependence of functional and Homer, and LIMK1 (384, 481).

FIGURE 7. The tripartite synapse. A: role of the extracellular matrix (ECM) and of astrocytes in processes of synaptic plasticity. In addition to
pre- and postsynaptic effects, glia cells contribute to the activity of various synapses by signaling via glioneurotransmitters like D-serine (coagonist
of NMDA receptors). Astrocytes themselves express various neurotransmitter receptors on their surface like AMPA or NMDA receptors
monitoring the synaptic activity and responding to changes by influencing Ca2⫹ level-dependent astrocytic processes like signaling molecule
release (D-serine, ATP, glutamate, TNF-␣) and NMDA receptor activity contributing to plasticity processes of the synapse. B: in addition to core
events, translation and transcription must be controlled as well via synaptic plasticity. In addition, astrocytes have to be taken into account, and
the influence of astrocytes on processes of synaptic plasticity are depicted. Changes in neurotransmitter vesicle cycling are prominent features
of presynaptically mediated neuronal plasticity. In general, the release of neurotransmitter starts with the filling of synaptic vesicles with
neurotransmitters. These vesicles are then docked to the plasma membrane and undergo a priming process at the active zone. When the action
potential arrives at the presynaptic side, it induces an influx of calcium ions through voltage-sensitive calcium channels (VSCCs). This triggers
vesicle fusion with the plasma membrane and exocytosis. The neurotransmitter vesicles are then recycled by the means of local reuse (a; “kiss
and stay”), or via fast recycling (b; “kiss and run”), or clathrin-mediated endocytosis (c). Vesicle fusion can be precisely controlled when neuronal
activity changes as exemplified by the regulation of synapsin phosphorylation (1) or the control of RIM protein phosphorylation (2). On the other
side of the synapse, AMPA receptor trafficking can be altered postsynaptically. AMPA receptors, synthesized in the nucleus or via local dendritic
protein translation, can enter a pool of endosomes which are involved in constitutive and regulated membrane trafficking. During processes of
synaptic strengthening, membrane receptor insertion increases the number of AMPA receptors at the synapse (3), where they are anchored
by interactions at the PSD. During processes of synaptic weakening, AMPA receptors have a high likelihood to be endocytosed (3). The
preferential site of exocytosis and endocytosis is most likely extrasynaptic. Within the plasma membrane, lateral diffusion between the synapse
and the point of insertion or removal controls AMPA receptors. Extrasynaptic diffusion of AMPA receptors increases with higher neuronal activity
(4). AMPA receptor trafficking is controlled by phosphorylation of receptor subunits (5), which determines the interaction with intracellular
scaffolding proteins. C: in addition to glia cells, events happening at the prototypic CA3-CA1 synapses and the role of the ECM, also modulatory
and inhibitory inputs from other neurons play a mayor in the modulation of neuronal plasticity. The neuromodulatory neurotransmitters (e.g.,
acetylcholine, serotonin, dopamine) influence the synaptic activity and signaling events of a specific neuron. NMDA, N-methyl-D-aspartate
receptor; TNF-␣, tumor necrosis factor-alpha.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 669


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

2. Spine neck plasticity al. (42) monitored the remodeling of spine structures during
LTP in single spines and observed that proteins translocate
Structural changes do not only affect the spine head, but into the spine through three sequential phases in four dis-
also the spine neck, which connects the spine head with the tinguishable patterns. The initial phase is characterized by
dendrite, restricts the diffusion of ions, and influences the rapid actin remodeling and followed by immense trafficking
electric signal propagation due to its geometric characteris- of the ABP cofilin into the spine. After that, cofilin forms a
tics (485). There is evidence that the resistance between the stable complex with F-actin and contributes to long-term
spine neck and the underlying dendritic branch is changed spine stabilization.
after the induction LTP and that this is due to changes in the
dynamics of the cytoskeleton in spines (38, 438). These 4. CaMKII
studies added the novel concept that spine neck resistance is
not constant and that structural alterations in the cytoskel- On the molecular side, an interesting connecting partner
eton change the diffusion constant of ions and probably between function and structural plasticity is the kinase
also electric signal propagation from spines to dendrites. CaMKII, which is needed after LTP induction due to an
Here, Tønnesen et al. (438) used high-resolution stimulated increase in intracellular Ca2⫹ concentration (see sect. IIE
emission depletion (STED) imaging to provide direct evi- and FIGURE 4). In addition to its role in functional syn-
dence for morphological changes of the spine neck upon aptic plasticity, CaMKII in its inactive form can also bind
LTP induction. This study could show that the spine neck is to F-actin, thereby limiting the access of APBs to F-actin
a highly dynamic structure, which becomes shorter and thus stabilizing the shape and size of spines (216). Ca2⫹-
wider upon activity, possibly facilitating diffusion of second mediated activation of CaMKII via autophosphorylation
messengers from the dendrite into the spine. The biophysi- leads to its dissociation from F-actin. This now allows
cal effects of spine neck changes are quite different from the F-actin remodeling via the now possible interaction with
biochemical effects. Whereas diffusion from dendrites into ABPs. The dissociation of CaMKII from F-actin is fol-
spines will most likely be boosted, the biophysical proper- lowed by a fast inactivation of CaMKII leading again to
ties indicate that these morphological changes lead to a the binding to F-actin thus restabilizing the newly formed
substantial drop in spine head EPSP. Therefore, on the bio- spine structure.
physical side, the resistance of a spine neck is increasing
after LTP induction and is decreased if synaptic activity is In summary, it is commonly accepted that the immense
blocked for a longer period of time (438). capacity of the central nervous system to store information
depends on the precise refinement of synapse structure. Syn-
3. Actin-binding proteins apses are considered today as the single, most fundamental
processing units within neuronal networks, the majority of
To enable structural plasticity, the underlying actin fila- which are located on dendritic spines, which can change
ments (F-actin) have to be assembled or disassembled on a their structure due to changes in the actin cytoskeleton
fast time scale (seconds to minutes) in a process tightly (379).
regulated via numerous actin-binding proteins (ABPs). The
main task is the transformation of receptor-mediated sig-
nals into the cytosol via the action of ABPs. This view de- N. Changes in Gene Expression and
mands that the most dynamic pool of F-actin is located Epigenetic
close to the postsynaptic membrane at the tip of the spine to
promote or restrict growth processes (173, 174; for a re- In addition to events that occur at the site of communica-
view, see Ref. 70) (FIGURE 5B). Since LTP occurs within 1 tion between neurons, when it comes to long-term storage
min after high-frequency stimulation and needs to be stabi- of information also the communication between the syn-
lized in the following 10 –30 min, rapid changes in the actin apses and the nucleus of a particular neuron has to be con-
organization are necessary (268). Increased polymerization sidered (as already mentioned in sect. IIJ). Whereas synaptic
of G-actin to F-actin is mandatory for the stabilization of events that lead to memory storage focus on protein
LTP and pharmacologically induced depolymerization of switches, more enduring processes of long-lasting memory
F-actin caused a decrease in spine head volume and elevated storage also have to include RNAs (translation) and also
internalization of glutamate receptors (133, 268). Overall, events at the DNA level (transcription). Therefore, the
it can be summarized that inhibiting spine head enlarge- sometimes long distance between a particular set of syn-
ment, LTP induction was not possible (133, 213, 238, 354, apses and the nucleus must be overcome by signaling cas-
362). Two recent studies used time-lapse two-photon imag- cades. The fastest routes use Ca2⫹ signaling, either via li-
ing of fluorescently labeled synaptic marker proteins com- gand or voltage-gated Ca2⫹ channels, or Ca2⫹ release from
bined with electron microscopy to analyze the spatiotem- internal stores. By this means Ca2⫹ signals could lead to
poral changes in synaptic morphology upon LTP. Meyer et transcriptional changes of specific genes involved in plastic-
al. (295) demonstrated that in persistently enlarged spines, ity processes genes (160; for a review, see Ref. 13), like the
the sizes of spine, PSD and bouton are correlated. Bosch et expression of immediate early genes after the induction of

670 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

LTP (419, 456), which have been acknowledged to be im- This means that by posttranslational changes, e.g., acetyla-
portant prerequisites of long-term memory storage (for a tion or phosphorylation of certain histone domains or
review, see Ref. 197). In this context hundreds of activity- methylation of DNA elements, the long-lasting transcrip-
regulated changes have been discovered, including MEF2, tion of specific genes involved in processes of long-term
CREB, and NPAS4 Greer (147). information storage can be up- or downregulated.

In addition to immediate changes in gene transcription via Conceptually this means that after a learning event, the
the activation/repression of transcription factors, the over- molecular modifications (up- and downregulation of cer-
all long-lasting (months to years) expression of genes can tain genes) necessary to implement the long-lasting change
also be changed via epigenetic regulation of chromatin in the expression of plasticity-related proteins is mediated
structure. The term epigenetic is used for the heritable and via alterations at the level of the chromatin structure (with-
self-perpetuating modifications to DNA and DNA-associ- out changes in the order of DNA base pairs). By this means,
ated proteins (e.g., histones), without changes in the DNA long-lasting changes in response properties of particular
base pairs sequence itself (FIGURE 8). The epigenetic foot- neurons can be conserved to store an engram for months or
print on the DNA or histones can lead to a specific pattern even years. This is supported by the fact that chromatin
of gene expression and can be considered as a persistent acetylation is of importance for high-level gene expression
form of cellular memory, which can even be transferred precisely at a chromosomal region of active transcription of
from mothers to their offspring. Epigenetic mechanisms are plasticity-related genes (253, 266). Acetylation is catalyzed
used to define the expression pattern of cells in a specific via the histone acetyltransferase (HAT) and de-acetylation
organ of the body, and the adult nervous system is using this is mediated via the histone deacetylase (HDAC). The effect
program for processes of long-term memory storage (253). of acetylation on gene expression is explained by the fact

A Gene expression

Acetylated chromatin
Ac Ac
CREB-1
P P
CBP
TBP

CRE TATA Ac C/EBP

POL II FIGURE 8. Epigenetic mechanisms. Epi-


mRNA genetic mechanisms mediate long-term
storage of memories (cellular event of
memory consolidation). The most promi-
nent molecular events of epigenetic regu-
lation are depicted: acetylated chromatin
B Gene repression (A) and methylated DNA and processes of
deacetylation (B). Ac, acetyl residue; CBP,
Methylated DNA CREB binding protein; C/EBP, CCAAT-en-
hancer-binding proteins; CRE, cAMP re-
CREB-1 Ac Ac sponse element; CREB, CRE binding pro-
P P tein; DMNT, DNA methyltransferase;
CBP DNMT HDAC, histone deacetylase; Me, methyl
Me
TBP residue; Me-CpG-BP, methyl-CpG binding
protein; P, phosphate residue; Pol II, RNA
CRE TATA Ac C/EBP polymerase II; TATA, TATA box; TBP, TATA
box binding protein.
POL II
mRNA
De-acetylated chromatin

Me-CpG-
CREB-
HDAC

TA

Me
TA

CRE C/EBP

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 671


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

that it removes positive charges on histone proteins, and by age. It is noteworthy that a NMDA receptor blocker can
this means it decreases the contact of histones with nega- prevent the synaptic plasticity-associated alterations in
tively charged phosphate groups of DNA. This results in a bdnf DNA methylation (203, 266).
less condensed “relaxed” chromatin and increases the ac-
cess of transcription factors to DNA sections close to the Taken together, activity-dependent changes can lead via
acetylated sides and increases transcription (149). In partic- epigenetic mechanisms to a changed expression of the bdnf
ular, DNA methylation has emerged as an important mech- gene, and this might be utilized for long-lasting memory
anism for epigenetic changes involved in long-term memory storage. In general, evolutionary ancient mechanism of epi-
storage. Here DNA methyltransferases (DNMT) detect se- genetic switches changing chromatin structure and by this
quences of CpG island sites within the DNA. “CpG” means means the long-lasting expression pattern of a series of
“-C-phosphate-G-” (cytosine and guanine linked by a phos- genes can be utilized by neurons to change their long-term
phate), as it occurs in the structure of the DNA or RNA behavior. Therefore, to store memories for a long period of
nucleotide “backbone” of a single nucleotide strand. The time, signaling molecules have to be activated (e.g., cAMP,
methylation of these CpG sites can directly control the rate Ca2⫹) as well as kinases (protein switches), and in the end,
of transcription of certain genes via the alteration of the translational and transcriptional switches in addition to cy-
local chromatin structure (253, 266). CpG islands can be toskeletal switches are needed to transform short-lasting to
found in 40% of the promoters for mammalian genes (108). long-lasting events. Therefore, a different set of “molecular
It is currently believed that DNA methylation is a major switches” is needed (245), some of them transient, some of
epigenetic mechanism that potentially contributes to long- them in the end long-lasting, and these could well include
lasting alterations in the expression pattern of a whole series changes in gene expression via activity-dependent epige-
of activity-regulated genes (32, 203, 245). At least two sub- netic changes in chromatin structure. This makes epigenetic
types of DNMT enzymes (DNMT 1, DNMT 3) are differ- changes part of the molecular toolbox to consolidate infor-
entially expressed in neurons in an activity-dependent man- mation storage for month and years (and some of them for
ner. a life time).

How can this be studied? Here, an example for the epige- O. Homeostasis and Metaplasticity
netic control of the bdnf gene is instrumental. BDNF has
been described earlier in this review as being a mediator of So far we have considered the timing of cellular events from
activity-dependent synaptic plasticity and is discussed as an changes of neuronal activity under leaning conditions to
important mediator especially of long-term memory stor- synaptic plasticity events to maintain these changes for
age. It has been shown that the expression of BDNF de- long-term storage. This includes changes in the postsynap-
pends on neuronal activity (266). And not only that, there is tic spine, retrograde signaling to the presynaptic side, and
also behavioral evidence which implies that bdnf gene ex- changes that happen there, signaling into the dendrites and
pression is activated in the hippocampus after contextual to the nucleus of a neuron, which leads sometimes to long-
and spatial learning (see, e.g., Ref. 157). In addition, the lasting changes in gene expression. But also neuronal activ-
bdnf-gene activation is vital for learning and memory events ity that happens before a particular learning event has an
(17, 157, 301). The bdnf gene itself is therefore rather com- influence on the probability of a specific synapses of a neu-
plex, to fulfill all the regulatory needs. It consists of nine 5’ ron to undergo plasticity changes. The amount of informa-
noncoding exons each linked to individual promoter re- tion a neuron “perceives” is changing all the time as it might
gions, and a 3’ coding exon encoding the pre-protein amino be involved in different neuronal assemblies that store dif-
acid sequence. Some of the BDNF-promoter regions are ferent information. The ability of a neuron to modify its
activity dependent determining the temporal and spatial ability to change over time has indeed been described and
expression pattern of specific isoforms of bdnf transcripts named metaplasticity (4, 178) or synaptic scaling as a ho-
(for a review, see Ref. 47). After contextual fear condition- meostatic mechanism to calibrate the overall excitability of
ing, it could be shown that bdnf exons I, II, and VI are a neuron (444).
methylated and the related mRNA transcripts reduced in
their expression, whereas exon IV is demethylated and the 1. Metaplasticity
corresponding mRNA expression increased. In this context,
it could be shown that methylation of bdnf DNA in the The principal idea behind the concept of metaplasticity is
hippocampus is changed by infusions of zebularine (a DNA that the previous history of a nerve cell determines its future
methyltransferase inhibitor). Overall, these changes lead to ability to undergo synaptic changes; a neuron might be less
different levels of exon-specific bdnf mRNAs and indeed to prepared to change its synapses or it may be more prone to
a different performance in learning tasks related to long- changes in response to a stimulus. For example, depending
term memory storage (203). This indicates that changes in on the previous input to a synapse or to a group of synapses,
DNA methylation are sufficient to trigger a differential bdnf LTP or LTD might have a higher (or lower) induction rate.
transcript regulation affecting long-term information stor- These mechanisms depend on the current “state” of a group

672 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

of functionally connected synapses (a synaptic unit), which counteracted if BDNF, but not the other neurotrophins,
is influenced via current extrinsic factors, like the activity of was applied during the activity blockade (356). Overall,
synaptic inhibition and the actual level of neuromodulators these results indicate that the synaptic activity level controls
acting on a subset of synapses. There is also some evidence cortical inhibition and that BDNF plays a major role in the
that synapses during processes of functional plasticity are regulation of cortical excitability. In a followup study, it
not changed gradually, but rather that they could be in could be shown that the effects of neuron activity blockade
discrete states (306). These different states could be silent, are mediated by changes in the quantal amplitude of EPSPs
active, potentiated, recently silent, or depressed. Therefore, and that this amplitude is regulated via BDNF (370). These
the coming and the future states will be influenced by the data demonstrate that synaptic scaling is of major impor-
status gained during the previous activity level. Not much is tance for the homeostatic control of excitatory as well as of
known about the molecular nature of metaplasticity, but it inhibitory circuits and in the regulation of the balance of
is of great theoretical importance in brain and cognitive
cortical inhibition and excitation (443).
science and it adds another layer of complexity to the mod-
ulation of synapses (306).
Taken together, the induction of processes that lead to
changes in synaptic weight is a long-lasting sequence, which
2. Excitation-inhibition balance and synaptic scaling
starts and ends with a highly choreographed and perfectly
In relation to a remark made by the late Walter Cannon, timed movement of molecules in different cell types, which
one can say: “Somehow the unstable stuff of which neurons operates over a long range of temporal and spatial scales.
are composed of has learned the trick of maintaining stabil- But for sure the journey into molecular mechanisms of long-
ity.” Homeostatic synaptic scaling is a form of synaptic lasting memory storage will go on “ѧ as it happens to the
plasticity that adjusts the strength of all of a neuron’s excit- coastwise voyager, who finds no end to his journey, for
atory synapses up or down to stabilize its firing. It was first behind each headland of clayey dune he conquers, fresh
observed in dissociated cortical cultures (446), which headlands and new distances lure him on” (Thomas Mann,
showed compensatory adjustments in their synaptic Joseph and His Brothers).
strength upon perturbations of the activity in the neuronal
network. The adjustments returned in the end firing rates
back to baseline values. This suggests that nerve cells detect III. SYSTEMS BIOLOGY OF MEMORY
changes in their own firing rates and regulate, e.g., receptor
trafficking accordingly so that synaptic strength is either Although synaptic plasticity processes contribute to mem-
increased, if the overall firing rate of a neuron is very low, or ory formation, they have to be integrated into neuronal
decreased if the overall firing rate is very high (for a review, networks within different brain areas as well as different
see Ref. 445). brain states. In this regard, synchronicity of circuits seems
to be of particular relevance. In this section, we review both
On the molecular level, in addition to Ca2⫹-dependent sen- the encoding of new information during awake states as
sors (for a review, see Ref. 445), BDNF might orchestrate well as the consolidation of fresh memories during sleep
the plasticity threshold of an entire cluster of synapses and from the prospective of the network level of information
might therefore be involved in processes of metaplasticity processing.
and homeostasis. Indeed, BDNF contributes to homeostatic
processes, especially to synaptic scaling of inhibitory neu-
rons, as was shown for cortical circuits in culture, which are A. Encoding: Role of Temporal Compression
also modulated by GABA-releasing interneurons (356). In
the rodent as well as in the primate visual cortex, activity
As described above, the mammalian hippocampus was very
blockade results in a decrease in the expression of GABA.
early on identified as critical for explicit memory (296).
This indicates that neuronal activity is in principle able to
Memory trace encoding on the behavioral level requires
adjust the strength of cortical GABA-mediated inhibition.
In this context it is noteworthy that neuronal activity regu- time scales on the order of hundreds of milliseconds or even
lates the expression of BDNF as well. In addition, BDNF seconds. Thus timing of learning differs substantially from
influences the differentiation and the phenotype of those required for synaptic plasticity rules, which is on the
GABAergic interneurons. Blocking spontaneous activity in order of only a few milliseconds (see above). Which cellular
neuronal cultures leads to a reversible decrease in the num- and/or network mechanisms allow to bridge these different
ber of GABA-expressing neurons without affecting neuro- time scales? In the following sections, we discuss a potential
nal survival (356). Patch-clamp analysis of inhibitory cur- network mechanism, the so-called “phase precession.” We
rents indicates that blockade of neuronal activity also leads will describe some of the features of place cells, present
to a decrease in inhibition mediated by the release of GABA findings on oscillatory activity, and outline the phenome-
onto pyramidal neurons, which results in an increase of the non of phase precession and its hypothetical function in
firing rates of these neurons (356). These effects could be encoding via temporal compression.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 673


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

B. Place Cells rat has been suggested to be encoded by the firing rate (100,
180, 328, 391). However, the correlation between action po-
The activity of many hippocampal pyramidal neurons is tential firing rate and running speed, though higher than be-
closely related to the animal’s position in the environment tween firing rate and other parameters, is low and varies be-
during spatial navigation. These “place cells” show a rise in tween cells. In addition, place cell activity correlated to previ-
their firing rate if the animal navigates through a specific ous spatial behavior has been shown to be recapitulated in
region of the environment, the so-called place field, and different types of sleep (264, 247).
become virtually silent when the animal is completely out-
side the respective place field (FIGURE 9). When an animal is
placed into a completely new environment, place fields are C. Network Oscillations: Theta Rhythm
established within a few minutes and stay stable for long
periods of time if the environment does not change. Never- The activity of the place cell is accompanied by hippocampal
theless, the place fields of different cells may overlap, which theta oscillations (4 –12 Hz) during exploratory behavior and
is an important condition for learning of spatial sequences certain phases of sleep (REM) (325, 328). Summative evi-
(100, 328, 391). dences indicate that theta rhythm is crucial for learning, e.g.,
spatial learning and navigation.
The topographical relationship of the place fields is inde-
pendent of their anatomical representation (359). Fur- Exploratory behavior can be observed, e.g., when an animal
thermore, the relation between place fields varies be- is placed in a rather novel environment and does whisking
tween environments, i.e., cells with neighboring place and sniffing. The characteristic sniffing pattern of a rat is
fields in a certain environment might have place fields also associated with theta activity (327).
apart from each other in another one (358). Not all hip-
pocampal pyramidal neurons show place field activity, 1. Hippocampal theta rhythm
e.g., only ⬃10 –25% of the CA1 cells show this firing
pattern (326, 327). Also, the fraction of active place cells In principle, the hippocampal theta electroencephalogra-
in a specific environment is even smaller and independent phy (EEG) is very prominent and regular in frequency,
of other environments (326, 327). but it shows important differences depending on the re-
cording site. The amplitude as well as the phase of the
The underlying circuits for place fields very likely have theta rhythm change between different hippocampal lay-
important implications for information storage, as they ers. The phase of the theta shifts backward with increas-
provide the spatial context for memories and past expe- ing depth: from Stratum oriens, which is the layer above
riences. Place cells show a large variety of properties, but the CA1 pyramidal layer, to the hilus, beneath the DG
only a few of those most important in the given context granule cell layer. The amplitude is maximal at the hip-
are mentioned here (see Ref. 358 for more details). On pocampal fissure between Stratum lacunosum-molecu-
repeated paths (e.g., linear or rectangular tracks), place lare, the lowest layer in CA1, and Stratum moleculare,
fields are direction specific, i.e., the same cell that is firing the uppermost layer in DG (see also Ref. 52). The under-
when the rat runs in one direction is silent when it runs lying mechanism of the hippocampal theta rhythm is un-
the opposite way (292). On the other hand, in an open known. The “classic” model of theta generation is de-
field environment where the rat can move freely, neuro- scribed as follows (52). The medial septum and the diag-
nal activity does not completely depend on the direction onal band of Broca (MS-DBB) have been assumed to yield
in which the rat is moving through a specific region (Fig- the theta pacemaker input to the hippocampus by di-
ure 1.3B in Ref. 281). rectly innervating CA1 pyramidal cells and interneurons
using acetylcholine as a transmitter. Additionally, the
Later experiments showed that place fields of CA1 pyramidal basket cells (a type of inhibitory hippocampal neurons)
neurons extend opposite to the running direction when the receive rhythmic GABAergic input, which they transmit
animal is frequently running on a linear track. The field may to the pyramidal cells. The pyramidal cells receive further
become asymmetric thereby, i.e., the relationship between po- rhythmic input from the EC (layer III). This “classic”
sition and firing rate gets negatively skewed over time (293). model does not seem to reflect the reality very well for
Such asymmetric place fields are hypothesized to emerge by several reasons. It has been suggested that two indepen-
NMDA receptor-dependent long-term plasticity of synapses dent theta generators are present in the hippocampus
from CA3 to CA1 (100, 293, 391). Familiar environments are (226). This suggestion arises from the finding that two
also made up of symmetric and positively skewed place fields types of theta activity can be distinguished: atropine re-
(180). It has been reported that place cells exhibit a high vari- sistant and atropine sensitive (52). The atropine-resistant
ability of activity within the place field (112). This might indi- theta component is expected to be mediated by the EC to
cate that place cells encode other information than the ani- the hippocampus, and the atropine-sensitive theta origi-
mal’s location in the environment, e.g., texture underfoot, nates intrinsically in the hippocampus in the area of CA3
odor, or stage of task. Furthermore, the running speed of the neurons. The latter theta oscillator requires cholinergic

674 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

activation, which is expected to be mediated by the sep- timing of hippocampal pyramidal cell spiking in relation to
tum (121). In the model presented by Kocsis et al. (226), the theta rhythm while the rat crosses the neurons’ place
rhythmic input from EC layers II/III is transferred to both field. They recorded CA3 and CA1 pyramidal cells extra-
hippocampal interneurons (basket and chandelier cells) cellularly and found that during locomotor behavior, place
and directly to pyramidal and granule cells, while the cells do not fire spikes randomly distributed over the whole
intrinsic CA3 theta generator is assumed to be suppressed theta cycle, but rather discharge spikes that are pooled into
in the intact hippocampus (226). several bursts. O’Keefe and Recce (328) investigated the
temporal relationship between the theta rhythm and those
Recent findings suggest that an intrahippocampal theta os- bursts and uncovered that such spike bursts do not occur at
cillator is located in the network of CA2/CA3 pyramidal a constant phase but shift to lower phases as the rat crosses
cells (120). In this model, the hippocampal subfield does not the cell’s place field. A graphical illustration of this phenom-
obtain extrinsic oscillating input, and the septum is expected enon is given in FIGURE 9 (see also Figure 5 in Ref. 328). The
to provide simply constant cholinergic input. The remaining earliest action potentials, when the rat goes into the place
hippocampal network is assumed to adopt the oscillatory field, occur at constant phases, but the subsequent spikes
pacemaker activity of area CA2/CA3. In fact, the mechanism advance to lower phases. The phase precession continues
underlying the theta oscillations in the hippocampus remains throughout the whole place field traverse so that the phase
still unknown, despite the alternative ideas (for a review on has shifted through roughly an entire theta cycle when the
hippocampal theta activity and alternative solutions, see also animal leaves the field.
Refs. 52, 75, 308).
A phase recession, i.e., shift to higher phases, has never been
Corresponding to the extracellular theta, intracellular observed. The linear correlation of the firing phase with the
membrane potential oscillations at theta frequency have position was stronger than with time. Though focusing on a
been shown to emerge in pyramidal cells (195, 482). The linear relation between phase and position, they mentioned
activity of pyramidal cells, or more precisely place cells, that occasionally position versus phase curves had sigmoi-
during spatial behavior is highly related to the theta rhythm dal shapes indicating nonlinear dependencies (328). Later
as will be outlined in the next section in more detail.
on, it was described by recording only CA1 pyramidal cells
that phase precession shows more complex dynamics (402).
Riding on top of the theta rhythm, a neuronal activity with
The spikes do not shift to lower phases linearly with posi-
the characteristic frequency of the gamma band (40 –100
tion; instead, the shift may accelerate at the end of the place
Hz) is observed in the EEG (45). Theta and gamma rhythms
field, yielding curved phase versus position plots with ver-
are recordable throughout the whole entorhinal-hippocam-
tically expanded clusters for low phases (Figure 7 in Ref.
pal system, including the subiculum, and are tightly cou-
402). Yamaguchi et al. (475) analyzed these features more
pled. The phase of theta scales the amplitude of gamma
thoroughly and described the data by two Gaussian func-
oscillations. This phase-amplitude (or cross-frequency)
tions. They saw that the first Gaussian component belongs
coupling of theta and gamma oscillations has been first
to the early part of the place field, thus to high theta phases
described (for a review, see Ref. 52) and later confirmed for
the hippocampus (407, 439) and other brain areas (58, 85, (⫽180°) and reflects a strong correlation between position
440). More recently, it was reported (2012) that also an- and phase. The second component, which is in the middle of
other theta-gamma coupling phenomenon is observed in the late part of the place field, shows no or less correlation
the hippocampus (26): phase-phase coupling (429). Here, between phase and position (475).
the phases of both rhythms are correlated (for a review, see
Ref. 110). Furthermore, Skaggs et al. (402) extended their experi-
ments to two-dimensional environments, where the rat is
In summary, theta-oscillations are very robust synchronous freely exploring in a rectangular box. Phase precession is
activities observed during exploratory behavior and are also present there but less obvious. It is an interesting
thought to be crucial during encoding. notion that two-dimensional environments contain the
possibility of partial place field crossings (402). Due to its
dependence on the place fields, one would expect a
D. Phase Precession
smaller amount of phase precession. This might explain
The firing of single place cells in hippocampus is specifically the reduced distinctiveness. Skaggs et al. (402) addition-
related to the theta rhythm of hippocampal neurons. This ally reported examples of phase precession in DG granule
spiking takes place at distinct points of the phase of the cells, but the total phase shift was only about half a theta
theta cycle and can show variations in spiking-phase rela- cycle and less obvious in general compared with the hip-
tionship. pocampal pyramidal cells. Furthermore, only very few
examples were reported. Thus whether phase precession
In 1993, O’Keefe and Recce (328) observed the phenome- is already present in earlier levels of processing of the
non of hippocampal phase precession by investigating the hippocampus than CA3 is less clear.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 675


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

In contrast to O’Keefe and Recce (328), Skaggs et al. (402) Additionally, phase precession has been shown to be unaf-
specify absolute measured phase not in correlation to the fected by aging (391) and NMDA receptor blockade (100).
EEG theta rhythm, but instead to the CA1 population ac- On the one hand, both studies reported a very similar loss of
tivity peak, which they previously found to occur at a par- experience-dependent expansion of place fields, as men-
ticular phase of the EEG theta. They did not report the tioned above, suggesting some possible connection between
phase lag between the CA1 population peak and the EEG aging and deficits in NMDA receptor dependent LTP. On
theta explicitly. The CA1 population peak, however, seems the other hand, the overall amount of phase precession as
to precede the maximum of the EEG theta rhythm in the well as the initial firing phase and the firing phase at place
figures found in their publication by at least 45° (see Figures field exit remained unaffected. Due to the smaller field size,
2 and 3 in Ref. 402). If a large number of the principal however, the rate of phase precession, i.e., the change of
neurons are silent during recordings, the population activity phase with position, was higher in both studies. Moreover,
will largely be shaped by the activity of a limited number of both Shen et al. (391) and Ekstrom et al. (100) observed
phase precession throughout the first laps of a specific re-
cells. Spikes of active cells may precede in the phase at the
cording session in a familiar environment, contrary to the
moment of recording, resulting in a nonstationary reference
findings of Mehta et al. (293) (see also Ref. 75).
point. Nevertheless, the first spikes emitted by a CA1 pyra-
midal cells, when the animal enters the place field, were
Taken together, theta-oscillations of the hippocampal EEG
determined to be at 90 –120° after the phase corresponding rhythm and place cell activity are correlated with each other
to the peak CA1 theta activity. The onset of firing of DG with a specific spiking-phase relationship. Is phase-preces-
units was 90° earlier than that of CA1 cells, and the peak of sion an epiphenomenon or of functional relevance? Within
the DG population theta activity preceded the CA1 peak by the next section, we are going to lay down a hypothetical
about 90°. Meanwhile, further studies have been published, function for phase precession.
which investigated different aspects of phase precession,
e.g., on phase precession inheritance from the hippocampal
region CA3 to area CA1 (185). The point of zero phase is E. Temporal Compression as a Hypothetical
defined in different ways among these studies; therefore, the Function of Phase Precession
comparability of absolute phase specifications remains un-
certain. Most focus has been given to CA1 pyramidal neu- Due to its relation to the theta rhythm, which depends on
rons, but some studies present data on CA3 (e.g., Ref. 162). the rat’s location inside the place field, the phase apparently
The latter investigated the relationship between pyramidal encodes the precise position in the place field (186). Tem-
cell firing rate and phase, finding a correlation between poral compression is considered to be a function of phase
both. The phase of firing is observed to be high and constant precession, and it is presented here as a link between the
during low spiking activity and phase advance occurs at cellular and network mechanisms for the encoding of new
times of intense activity. This result even holds true during information. This is an example for temporal coding, i.e.,
nonspatial learning behaviors like wheel running and sleep. the precise timing of the spike(s) in the theta rhythm pos-
The observed correlation supports the idea that phase pre- sesses spatial information. Another way to represent infor-
cession depends on the strength of dendritic depolarization mation is firing rate coding where the number of spikes
of the pyramidal cell, as discussed above. On the other determines the information coded, as in the case of the rate
of action potential firing of a place cell. If phase precession
hand, it could be shown that the observed correlation be-
is only a product of learning, then its function would be
tween the rate of action potential firing and phase might
restricted to the coding or additional coding of position.
result from a combination of position-phase and rate-phase
However, it is unclear whether phase precession truly is a
relationships (180). They support the idea that phase and
product of learning; for instance, phase precession is pres-
rate encode different parameters, e.g., phase for the ani-
ent already during the first exposure to an environment
mal’s position in the place field and the firing rate for the (358, 368), suggesting another functional role. Skaggs et al.
in-field velocity. Thus the absolute place cell activity during (402) proposed an alternative view, how phase precession
various place field traverses does not have to be the same. might facilitate the learning of sequences. In principle, a
discrepancy exists between the time scales of synaptic plas-
Further experiments indicated that a connection between ticity, such as NMDA receptor-dependent LTP, and the
firing rate and phase (293) exists, similar to findings in behaviorally driven changes in neural firing patterns. This
Reference 162. Furthermore, according to their model hy- discrepancy could be removed by means of phase precession
pothesis, they found phase precession only within nega- that provides a temporal code, e.g., for the position, so that
tively skewed place fields, which in their opinion emerge information of different traversed locations can be tempo-
due to NMDA receptor mediated LTP, as described below. rally related to each other. Within this framework, a se-
In contrast, Huxter et al. (180) observed that all possible quence of overlapping, successively traversed place fields of
kinds of place field skewness (negative, positive, symmetric) neurons would be reactivated or “replayed” in a com-
exhibit phase precession (see also Figure 2. 1B in Ref. 180). pressed form during each theta cycle. This arises due to the

676 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

phase precession of the neuronal spikes, while the animal tic one, results in an asymmetric reinforcement of the syn-
moves through a certain sequence of place fields. As illus- aptic connections according to the sequence, the more so,
trated in FIGURE 9, along the path of a running rat neurons since a portion of the whole sequence is replayed in several
with overlapping place fields successively start to fire and theta cycles. This redundancy supports a reliable associa-
process in the phase thereafter, which means their firing tion of the parts of the sequence. As a high degree of con-
advances to the succeeding phase. With the two assump- nectivity in a network is necessary to perform this kind of
tions that 1) firing starts at a fixed phase of the “back- associations, the CA3 region is the prior candidate for the
ground” theta rhythm as a common temporal reference, described mechanism to occur. As a consequence, apart
and that 2) the phase change with position or time (i.e., from the supporting experimental findings (328, 402, 162),
phase precession) is the same for all cells, the temporal phase precession should be expected to emerge already in
order of traversed place fields is conserved in the temporal the CA3 region (185, 302). In sum, temporal compression
order of the place cell firing during a theta cycle. That is the of behavioral time scales could be a function of phase pre-
case, because the firing of a cell, whose place field has been cession: it might link synaptic, cellular, and network mech-
entered before that of a second cell, already occurs at an anisms for the encoding of new memories.
earlier phase inside the theta cycle than the firing of the
second cell. Thus a sequence of overlapping place fields is
represented in each theta cycle. The sequence, however, is F. Sleep: a Long-Lasting Timing Rhythm of
temporally compressed, because the time scale of the replay Memory Consolidation
is in the order of 10 ms.
Memory consolidations on a system level refer to the pos-
The compressed time scale of sequence replay meets the tencoding reorganization of long-term memory over dis-
criteria necessary for NMDA receptor-dependent synaptic tributed neuronal circuits within different brain areas. Sev-
plasticity. Additionally, synaptic modifications depend on eral models of system consolidation have been established,
the timing of pre- and postsynaptic spikes (STDP), i.e., the from the standard consolidation theory, to the multiple
preference of presynaptic activity preceding the postsynap- trace theory, up to the schema assimilation model. The

location

a b c d

a b c d
event
FIGURE 9. Encoding of spatial trajectories
within the hippocampus via temporal com-
pression. Place cells exhibit place cell over-
lap, while at the same time the hippocampal
neuronal network shows robust theta oscilla-
cell a
tions. Overlapping place cell activity and sub-
b sequent temporal compression establish the
time windows for spike-timing dependent syn-
c aptic plasticity (STDP).

c
a

d
b

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 677


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

different hypotheses underlying these models have been to the order of 100 ms, which is strong evidence for the
nicely reviewed (for example, see Ref. 94). In our review we existence of a temporal compression mechanism.
would like to concentrate on the role of sleep, with a par-
ticular focus on high-frequency oscillations, and its func- Different from the standard consolidation theory is that
tion in memory consolidation. both hippocampus and extrahippocampal regions store rel-
evant information. The orchestrating central role of the
Sleep has long been thought to be a rather passive and hippocampus is the integration of different information as-
recreational event. In recent years this view has been chal- pects into one spatiotemporal context, thus binding the dif-
lenged and very recent studies could even link structural ferent components together to form an episodic memory.
plasticity with sleep (476). In mammals as well as in birds, Replay of this information in each SWS cycle and informa-
the sleep behavior is generally divided into two phases: tion transmitted from the hippocampus to extrahippocam-
rapid eye movement (REM sleep) and non-rapid eye move- pal regions might induce plasticity changes in these regions,
ment (NREM, non-REM sleep). Each phase of sleep has a representing the transformation steps necessary to extract
rather distinct set of neurophysiological features associated the semantic content of the episodic memory and store them
with it. REM sleep is linked to dreaming. NREM is divided as semantic engrams (41).
into three distinct stages: N1, N2, and N3. N3 is commonly
also called delta sleep or slow-wave sleep (SWS). 2. Information processing during SWS

Among the different sleep stages, especially the SWS seems A key electrophysiological feature enabling this informa-
to be important with respect to the consolidation of mem- tion processing during SWS is the interaction of neocortical
ory. During SWS, memory sequences are repeatedly re- slow oscillations (SO) with hippocampal sharp wave ripples
played, which has a pivotal role in transferring labile mem- (SWR) and thalamic spindles. SO are characterized by a
ory into stable engrams (for reviews, see Refs. 41, 357). At highly excitable “up” state, setting the framework for exci-
the same time, episodic and decontextualized content is tation-mediated Ca2⫹ influx, which might lead to appropri-
separated and processed in the appropriate networks, the ate synaptic changes (389). The cyclic change between the
hippocampus being mainly responsible for orchestrating low-frequency and high-amplitude EEG in SWS and the
the storage of episodic memory and extrahippocampal re- low-amplitude EEG in the theta range as it occurs during
gions such as the neocortex and the striatum being respon- REM sleep might be a complementary mechanism to enable
sible for semantic and procedural aspects of memory (357). memory consolidation. Consistent with this theory, sleep
primarily seems to be beneficial for hippocampus-depen-
dent memories (101, 448). Animal studies support this hy-
1. Function of distinct stages of sleep
pothesis. Although lesions to the hippocampus did not im-
pair the recall of “what,” “when,” or “where” as separated
Several hypotheses seek to explain the function of distinct information, the recall of contextual memory, which char-
sleep states for memory consolidation (for more details, see acterizes the relation between these aspects, was severely
Ref. 357). An earlier hypothesis proposed that sleep evolved impaired (254). Additionally, sleep was a prerequisite for
to maintain homeostasis (77, 198, 40). In this context, it is the integration of these aspects into a contextual memory
thought that plasticity processes during the awake state (182). Interestingly, these observations are also linked to
result in a net widespread enhancement in the efficacy of the occurrence of SWR and spindles during sleep intervals
synapses; sleep is then to downscale synaptic strength to (33). Moreover, blocking CA3 output by a genetic ap-
baseline that is energetically sustainable and potentially proach and thus eliminating the SWR drive to extrahip-
also more useful for the acquisition of new memories the pocampal regions resulted in a reduced retrieval perfor-
next day (198). The “active system consolidation” theory mance (316). The importance of sleep in hippocampus-de-
(41, 91) is a modification of the standard consolidation pendent memory consolidation is further supported by
theory and incorporates an active and integral role of sleep several behavioral observations. In earlier studies, sleep de-
in long-term memory consolidation. At the heart of this privation only affects the hidden platform version of the
theory lies the observation that hippocampal place cell as- Morris watermaze, which depends on hippocampal func-
semblies (compare sect. IIIB) are replayed in the same order tion (404). Furthermore, it induces a shift from hippocam-
during SWS as experienced during the initial exploration pus-dependent navigation strategies towards striatum-de-
and encoding (247, 401, 464). More specifically, Lee and pendent ones (153–155). Similarly, context-dependent fear
Wilson (247) report on CA1 pyramidal neurons in rats that conditioning is reduced by sleep deprivation, whereas cued
are sequentially activated during spatial behavior, repeat fear conditioning is not (56). Memories not involving the
the same order of firing during sleep, which indicates the hippocampus seem to profit less well from sleep (55). Thus
replay of memories during sleep and emphasizes the impor- the hippocampal system seems to provide a temporary
tance of the hippocampus in sequence learning. The replay memory buffer, in which labile (explicit) information is
during SWS occurs 20 times faster compared with the stored and transferred to other brain regions later on, where
awake exploration. As a result, sequences are compressed the memory becomes more stable. In accordance with this,

678 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

hippocampal lesions conducted 3 h after a learning para- as a result of the repeated replaying of information by the
digm interfered with the establishment of stable memory, hippocampus, probably during sleep, as suggested here.
whereas lesions conducted after 48 h had no effect (442).
Notably, SWS sleep decreases naturally with age, which is
Expanding these findings to a more global level leads to the correlated with a reduction in declarative memory consoli-
viewpoint that sleep is the state of the brain optimized for dation in humans and rodents (159, 278). Furthermore,
memory consolidation, whereas vigilance is the brain state when sleep was possible after implicit training on a motor
optimized for memory encoding (FIGURE 10). Sleep en- sequence, children showed bigger improvements in explicit
dorses the branch-specific formation of dendritic spines af- sequence knowledge after sleep than grownups did. This
ter learning (6, 476). Indeed, the need for sleep as a state of superior explicit knowledge in children was connected to
diminished sensory awareness, immobility, and loss of con- their higher amount of SWS activity as well as to stronger
sciousness might explain that encoding of information and activation of the hippocampus during explicit knowledge
consolidation are mutually exclusive events, since they re- retrieval (352).
cruit overlapping neuronal resources and physiological pro-
cesses (357). Bridging this further to the cellular level, it also
means that the cortex might have a different synaptic plas- G. Neuronal Network Oscillations in
ticity machinery than the hippocampus, e.g., it could indeed Memory Consolidation
be shown that the crucial function of the kinase alpha-
CaMKII during induction of LTP is a different one in the Within the two-stage model of memory consolidation, theta
cortex compared with its role in the hippocampus (125, oscillations and high-frequency oscillations (ripples) fulfill
256). Elegant experiments proved this point in alpha-CaM- complementary functions: theta during encoding and rip-
KII heterozygous mice, which have only half the normal ples during consolidation. A prerequisite of the aforemen-
amount of alpha-CaMKII and these mice showed normal tioned memory consolidation during sleep is that hip-
LTP in the hippocampus, but only short lasting LTP in the pocampal and extrahippocampal regions need to commu-
cortex (125). And indeed mice learned normally a new task, nicate in a carefully orchestrated manner. Prominent
but because LTP in the cortex is only short-lasting in these electrophysiological hallmark of this orchestration are the
mice, long-term memory could not be stabilized (consolida- up and down states of the SWS phases (0.75–5 Hz), sleep
tion was impaired). Also, on the behavioral level, these mice spindles (12–15 Hz), and hippocampal SWR complexes
could perform well initially in “hippocampus-dependent” (100 –200 Hz for ripples). SWS might represent the sleep
learning tasks, but these memories could not be maintained. state most important for the consolidation of declarative
According to Lisman and Morris (256), the cortex can be memory. Memory consolidation is thought to be the result
viewed to be a slow learner, capable of consolidation only of repeated reactivation of the neuronal representation of a
memory. Alongside consolidation, transformation of mem-
ory takes place at several levels: transformation of labile to
consolidation stable memory, but also transformation of abstract content
into semantic or procedural memory (466).

Slow and fast oscillation phenomena seem to set the overall


experience stable memories
“time reference” for the information processing during
sleep in a “top-down” manner. The information carrying
“bottom-up” events like SWR and spindles (see below) are
inhibited during the down state of the slow oscillations (20,
72, 73). Interestingly, slow spindles are delayed by ⬃200 –
500 ms with respect to fast spindles, thus occurring during
the transition from up to down states. SWR and spindles
appear to be driven by the depolarizing transition of down
to up states (20, 183, 246). Successful learning is paralleled
by an increase in density of ripples and spindles (73, 107,
135) and facilitates SO (304). In turn, the boosting of SO in
humans and animals has been shown to improve learning
FIGURE 10. Neuronal network oscillations in memory consolida- (282).
tion. Memory consolidation critically depends on high-frequency net-
work oscillations named ripples. Ripples enable the strengthening of 1. Spindles
newly acquired information on episodic events into stable memories.
The two types of pyramidal neurons within the subicular area at the
hippocampal output (B, burst firing pyramidal cell; R, regular firing Spindles are oscillations in the range of 7–15 Hz, with a
pyramidal neuron) might be differentially embedded during ripple duration of 0.5–3 s generated in the thalamus and spreading
activity and thereby separate information flow into the cortex. into many cortical areas. They are often further subdivided

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 679


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

into fast (12–15 Hz) and slow (10 –12 Hz) spindles, the be the presence of phasic excitation during ripples that is
latter occurring prominently during SWS (9, 80, 303, 430). coherent across the neuronal network and apparent as ex-
GABAergic neurons of the nucleus reticularis are the prime citatory postsynaptic currents and potentials at the single-
pacemaker in spindle generation, which depend to a large cell level (270).
extent on the activation of T-type Ca2⫹ channels (11, 21).
In cortical pyramidal cells, spindle occurrence might lead to The main output of the hippocampus, the subicular area,
a massive Ca2⫹ influx with consequences for the excitability also exhibits SWRs activity (25, 102, 285). Within the
of the cell (34, 389). Likely, one of the targets of this activity subicular area two types of pyramidal cells have been
on the genetic and molecular level is the protein Arg3.1 (88, described based on their specific firing properties: regular
389), which has been introduced in section III. Further- or burst firing pyramids (for a review, see Ref. 25). In
more, similar to SWR, plasticity inducing protocols also contrast to CA1 pyramidal cells, subicular pyramidal
facilitate spindle occurrence (28). Spindle activity seems to cells exhibit a low degree of axonal collateralization and
be positively correlated with successful learning in several project mostly to only one target region (313). Interest-
paradigms; noteworthy, in some paradigms spindle activity
ingly, the two pyramids project to different target areas,
increases selectively in the cortical areas associated with
e.g., the lateral and medial entorhinal cortex, but also
that particular paradigm, e.g., motor cortex (322, 454,
other brain areas (313). Recently, Böhm et al. could show
455). Furthermore, fast spindle activity is in synchrony with
by whole cell patch-clamp recordings in awake mice that
hippocampal and extrahippocampal oscillations starting
shortly before the occurrence of SWR and also outlasting bursting cells are activated during sharp wave-ripples
them (422). In the resulting spindle-ripple complex, ripples whereas regular firing cells are inhibited (39a). Similar
are temporally locked to the trough of a spindle and also observations were made in in vitro models of sharp-wave
synchronize with neocortical gamma activity (73, 397, 461, ripples (102, 285). Böhm and colleagues also identified
422). Slow spindles on the other hand might coincide with the principles that underlie the differential recruitment
the silencing of hippocampal input to the cortex (73, 347). during sharp wave-ripples by using multiple patch clamp
recordings and circuit analysis. Regular firing cells re-
2. Hippocampal SWRs ceive more and stronger inhibition while the pyramidal
cell network favors excitatory input onto burst firing
Hippocampal SWRs are 40 –150 ms periods of massive ac- cells. Thus principal cells that project to the same target
tivation of the entire hippocampal-subicular network (51, regions are connected among themselves, and in addi-
54, 482), in which characteristically slow sharp waves are tion, there is exclusive unidirectional connectivity from
overlaid with 120- to 250-Hz ripple oscillations. SWRs are one group of principal cells, regular firing cells, to the
observed during awake immobility and SWS (51, 54) and other, burst firing cells, but not vice versa. Considering
have been suggested to support the consolidation of re- the cell-type specific extrahippocampal target areas of
cently acquired memories (53, 90, 240, 247, 464). Accord- burst and regular firing cells (218), mainly target regions
ing to a current hypothesis, SWRs represent population of the burst firing cells would receive excitatory input
discharges of basically the entire hippocampal network from the subiculum during SWR. Indeed, neuronal activ-
(53). The basic mechanisms of how SWRs are initiated, ity that is temporally locked to hippocampal sharp waves
maintained for ⬃50 –100 ms, and finally terminated are could be shown in both the medial entorhinal cortex (68,
largely unknown. 471) and the presubiculum (68), which are target areas of
burst firing cells (218). It is tempting to speculate that
Recently, different experimental approaches, including jux-
there is no or less SWR-associated input to the lateral
tacellular recordings in vivo and investigations in hip-
entorhinal cortex, a target area mostly innervated by
pocampal slices, have identified several classes of inhibitory
regular firing cells. This would imply a pivotal role of the
interneurons that are activated during SWRs (187, 206,
220, 221, 222, 156, 334, 408, 449). With these techniques, subiculum in separating information streams and distrib-
it has been shown that ripple network activity is paralleled uting this information to cortical targets.
by an increase or decrease in specific classes of interneurons
(for a review, see Ref. 223). Based on these data, it has been In summary, memory consolidation requires not only cel-
concluded that interneurons induce ripple oscillations, al- lular events at activated synapses, but also fast network
though a direct and causal link has not been established yet. oscillations, and it involves the transfer of information from
Alternative mechanisms of ripple generation have been sug- the hippocampus to various cortical targets. Experimental-
gested where electrical coupling (93, 381) or combined ists in close interaction with theoreticians will further de-
mechanisms involving synaptic and electrical coupling velop the differing models over the near future. In addition,
(441) contribute to the oscillogenesis. In these models, the modern techniques such as cell-type specific optogenetics
phasic discharge of assemblies of pyramidal neurons is the will not only contribute to an improved understanding of
immediate source of ripples. A straightforward prediction the connectivity of neuronal networks, but will also provide
of such a synchronous activation of pyramidal cells would further insights in memory formation on a system level.

680 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

IV. PERSPECTIVE (e.g., Ref. 369). Although this convincingly shows the
necessity of these neuronal populations, the equally im-
In this review we covered a huge research field, not only in portant question of sufficiency remains: is it possible to
terms of papers and results, but also in space: vesicle size in elicit a specific (learning) behavior by directly stimulating
the nanometer scale, phosphorylation of proteins ever precisely the specific population of cells that was active
smaller, up to large centimeter wide neuronal networks, during the learning period? Here an amazing experiment
and the crosstalk of different brain areas during sleep and highlights how future experiments can be conducted to
memory consolidation. But also the time frame covered prove what is necessary and what is sufficient for infor-
shows the complexity of information storage and retrieval mation storage, as was outlined in the review by Morris
in the brain: some events are extremely fast, like the phos- and colleagues (284). In mice, it was recently shown that
phorylation of glutamate receptors due to changes in syn- optogenetic reactivation of hippocampal cells, which
aptic activity, the change in synaptic release machinery and were also active during a learning paradigm which in-
also Ca2⫹ signals inside neuronal compartment, which hap- cluded fear conditioning, is enough to induce freezing
pen in the millisecond to second range. Signaling to the behavior (260). During this experiment a certain popu-
nucleus, expression of early change and the transport of lation of granule cells in the hippocampus were labeled
their products can happen in 30 min up to a few hours, and after they have been activated during fear conditioning
this happens in parallel to structural changes at synaptic with the optogenetic protein channelrhodopsin-2
sides. But these activity-dependent changes are also modu- (ChR2). These neurons have been reactivated optically in
lated and shaped via rhythmic, sometimes oscillatory, brain a different context during the latter part of the experi-
activity, in the time range of seconds and minutes. Long- ment. And indeed, these animals showed a higher freez-
lasting changes in gene expression can last hours, days, ing rate only upon optogenetic stimulation. This out-
weeks, and most likely in concert with epigenetic genes even come indicates a recall of a light-induced fear memory. In
for months and years. It will be an exciting question to a control situation, no freezing behavior could be de-
explore how prior experience minutes, hours, or days ear- tected in mice, which were non-fear-conditioned but ex-
lier can increase or decrease the gain of synaptic plasticity pressed ChR2s (260). All control experiments indicated
and how by what means network activity and synaptic ac- that optogenetically induced fear recall is context spe-
tivity are timed to store, or encode specific memories. It is cific.
also an open question why certain forms of network activ-
ity, theta (5–7 Hz) during encoding and ripples 100 –300 In summary, these experiments show that activating a
Hz during consolidation, foster certain components of sparse but highly specific assembly of hippocampal neu-
memory storage and retrieval. rons, which were involved in the initial learning paradigm,
can be adequate for the recall of that specific memory.
What became evident in the last decade is that a specific Moreover, we think that this experimental paradigm used
memory can be encoded by an astonishingly sparse pop- by Tonegawa and colleagues offers a general paradigm to
ulation of principal neurons. These cells can be tagged map neuronal populations bearing an engram (memory
during the learning events for the subsequent identifica- trace). It also paves the way for not only clarifying the role
tion and manipulation. Moreover, their destruction or of single neurons in a neuronal network storing a specific
their inactivation can result in reduced memory expres- memory content, it also offers the possibility to study the
sion, suggesting their necessity in mnemonic processes molecular nature subserving these events.

TIMING OF EVENTS

Waves and STDP: Implementation Consolidation Epigenetic:


rhythms: Timing on of change at when we sleep the “before”
coupling the network the synapse and the “after”
level of learning

FIGURE 11. Timing of events in the biology of memory formation and consolidation. There is mutual
interdependence at every process for events of memory storage that need precise timing and might influence
current events (like the implementation of changes at the synapse) or future events (like epigenetic factors).

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 681


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

Consolidation is a continuous and long-lasting process on GRANTS


several molecular, network and systems levels, as Dudai
phrased it, “Taken together, the picture that emerges is of We are grateful to the DFG (to M. Korte and D. S. Schmitz)
dynamic engrams that are formed, modified, and re-modi- and N-RENNT, Ministry of Science and Culture of Lower
fied over time at the systems level by using synaptic consol- Saxony (to M. Korte), for funding our work.
idation mechanisms as subroutines. This implies that, con-
trary to interpretations that have dominated neuroscience
DISCLOSURES
for a while, but similar to long-standing cognitive concepts,
consolidation of at least some items in long-term memory
No conflicts of interest, financial or otherwise, are declared
may never really come to an end” (94). In recent years, it
by the authors.
became clear that synaptic plasticity, in particular LTP, is of
crucial importance for memory processes in vivo, and it
became also evident how neuronal assemblies can store in- REFERENCES
formation and represent a memory (see, e.g., Refs. 137,
260, 312, 355). 1. Abbott LF, Nelson SB. Synaptic plasticity: taming the beast. Nat Neurosci 3 Suppl:
1178 –1183, 2000.

For the past 40 years, the expedition into the neural basis of 2. Abel T, Martin KC, Bartsch D, Kandel ER. Memory suppressor genes: inhibitory
constraints on the storage of long- term memory. Science 279: 338 –341, 1998.
learning, memory, and retrieval focused mainly on the mo-
lecular and cellular basis of activity-dependent plasticity. 3. Aberle H, Butz S, Stappert J, Weissig H, Kemler R, Hoschuetzky H. Assembly of the
cadherin-catenin complex in vitro with recombinant proteins. J Cell Sci 107: 3655–
This approach has brought tremendous insight into the mo- 3663, 1994.
lecular machinery and the timing events of learning and
4. Abraham WC. Metaplasticity: tuning synapses and networks for plasticity. Nat Rev
memory so far (FIGURE 11). Nevertheless, here we provide Neurosci 9: 387, 2008.
evidence that a full understanding of the different processes
5. Acker CD, Yan P, Loew LM. Single-voxel recording of voltage transients in dendritic
leading to complex learning and memory behavior will re- spines. Biophys J 101: L11–13, 2011.
quire incorporating the properties of individual cells and
6. Ackermann S, Rasch B. Differential effects of non-REM and REM sleep on memory
the functional dynamics of large-scale networks. Further consolidation? Curr Neurol Neurosci Rep 14: 430, 2014.
progress in examining the conceptional foundation of mem-
7. Afroz T, Skrisovska L, Belloc E, Guillen-Boixet J, Mendez R, Allain FH. A fly trap
ory will require an approach that takes into consideration
mechanism provides sequence-specific RNA recognition by CPEB proteins. Genes
the importance of timing events in the CNS on every level of Dev 28: 1498 –1514, 2014.
complexity.
8. Akbik FV, Bhagat SM, Patel PR, Cafferty WB, Strittmatter SM. Anatomical plasticity of
adult brain is titrated by Nogo Receptor 1. Neuron 77: 859 – 866, 2013.
Therefore, we would like to conclude: “If any one faculty
9. Anderer P, Klosch G, Gruber G, Trenker E, Pascual-Marqui RD, Zeitlhofer J, Barbanoj
of our nature may be called more wonderful than the rest, MJ, Rappelsberger P, Saletu B. Low-resolution brain electromagnetic tomography
I do think it is memory. There seems something more revealed simultaneously active frontal and parietal sleep spindle sources in the human
cortex. Neuroscience 103: 581–592, 2001.
speakingly incomprehensible in the powers, the failures,
the inequalities of memory, than in any other of our 10. Araya R, Jiang J, Eisenthal KB, Yuste R. The spine neck filters membrane potentials.
Proc Natl Acad Sci USA 103: 17961–17966, 2006.
intelligences. The memory is sometimes so retentive, so
serviceable, so obedient–at others, so bewildered and so 11. Astori S, Wimmer RD, Prosser HM, Corti C, Corsi M, Liaudet N, Volterra A, Franken
P, Adelman JP, Luthi A. The Ca(V)3.3 calcium channel is the major sleep spindle
weak– but at others again, so tyrannic, so beyond con- pacemaker in thalamus. Proc Natl Acad Sci USA 108: 13823–13828, 2011.
trol! We are to be sure a miracle every way– but our
12. Atwood BK, Lovinger DM, Mathur BN. Presynaptic long-term depression mediated
powers of recollecting and of forgetting, do seem pecu-
by G-coupled receptors. Trends Neurosci 37: 663– 673, 2014.
liarly past finding out” (Jane Austen in Mansfield Park).
13. Bading H. Nuclear calcium signalling in the regulation of brain function. Nature Rev
Neurosci 14: 593– 608, 2013.
ACKNOWLEDGMENTS
14. Banerjee S, Neveu P, Kosik KS. A coordinated local translational control point at the
synapse involving relief from silencing and MOV10 degradation. Neuron 64: 871– 884,
We thank Wiebke Arendt, Anita Remus, Marta Zagreb- 2009.
elsky, and Benedikt Salmen for critical comments to an 15. Barco A, Lopez de Armentia M, Alarcon JM. Synapse-specific stabilization of plasticity
earlier version of the manuscript. We are especially processes: the synaptic tagging and capture hypothesis revisited 10 years later. Neu-
rosci Biobehav Rev 32: 831– 851, 2008.
thankful to Claudia Brose for the excellent help with the
figures. 16. Barco A, Patterson SL, Alarcon JM, Gromova P, Mata-Roig M, Morozov A, Kandel ER.
Gene expression profiling of facilitated L-LTP in VP16-CREB mice reveals that BDNF
is critical for the maintenance of LTP and its synaptic capture. Neuron 48: 123–137,
Address for reprint requests and other correspondence: M. 2005.
Korte, TU Braunschweig, Zoological Institute, Div. Cellu-
17. Barrientos RM, Sprunger DB, Campeau S, Watkins LR, Rudy JW, Maier SF. BDNF
lar Neurobiology, Spielmannstr. 7, 38106 Braunschweig, mRNA expression in rat hippocampus following contextual learning is blocked by
Germany (e-mail: m.korte@tu-bs.de). intrahippocampal IL-1beta administration. J Neuroimmunol 155: 119 –126, 2004.

682 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

18. Barth AL, Malenka RC. NMDAR EPSC kinetics do not regulate the critical period for 39a.Böhm C, Peng Y, Maier N, Winterer J, Poulet JF, Geiger JR, Schmitz D. Functional
LTP at thalamocortical synapses. Nat Neurosci 4: 235–236, 2001. diversity of subicular principal cells during hippocampal ripples. J Neurosci 35: 13608 –
13618, 2015.
19. Bashir ZI, Bortolotto ZA, Davies CH, Berretta N, Irving AJ, Seal AJ, Henley JM,
Jane DE, Watkins JC, Collingridge GL. Induction of LTP in the hippocampus needs 40. Borbely AA, Achermann P. Sleep homeostasis and models of sleep regulation. J Biol
synaptic activation of glutamate metabotropic receptors. Nature 363: 347–350, Rhythms 14: 557–568, 1999.
1993.
41. Born J, Wilhelm I. System consolidation of memory during sleep. Psychol Res 76:
20. Battaglia FP, Sutherland GR, McNaughton BL. Hippocampal sharp wave bursts coin- 192–203, 2012.
cide with neocortical “up-state” transitions. Learn Mem 11: 697–704, 2004.
42. Bosch M, Castro J, Saneyoshi T, Matsuno H, Sur M, Hayashi Y. Structural and molec-
21. Bazhenov M, Timofeev I, Steriade M, Sejnowski TJ. Self-sustained rhythmic activity in ular remodeling of dendritic spine substructures during long-term potentiation. Neu-
the thalamic reticular nucleus mediated by depolarizing GABAA receptor potentials. ron 82: 444 – 459, 2014.
Nat Neurosci 2: 168 –174, 1999.
43. Bosch M, Hayashi Y. Structural plasticity of dendritic spines. Curr Opin Neurobiol 22:
383–388, 2012.
22. Bear MF. Bidirectional synaptic plasticity: from theory to reality. Philos Trans R Soc
Lond B Biol Sci 358: 649 – 655, 2003. 44. Bottos A, Rissone A, Bussolino F, Arese M. Neurexins and neuroligins: synapses look
out of the nervous system. Cell Mol Life Sci 68: 2655–2666, 2011.
23. Bear MF, Malenka RC. Synaptic plasticity: LTP and LTD. Curr Opin Neurobiol 4: 389 –
399, 1994. 45. Bragin A, Jando G, Nadasdy Z, Hetke J, Wise K, Buzsaki G. Gamma (40 –100 Hz)
oscillation in the hippocampus of the behaving rat. J Neurosci 15: 47– 60, 1995.
24. Beesley PW, Herrera-Molina R, Smalla KH, Seidenbecher C. The Neuroplastin adhe-
sion molecules: key regulators of neuronal plasticity and synaptic function. J Neuro- 46. Bramham CR. Local protein synthesis, actin dynamics, and LTP consolidation. Curr
chem 131: 268 –283, 2014. Opin Neurobiol 18: 524 –531, 2008.

25. Behr J, Wozny C, Fidzinski P, Schmitz D. Synaptic plasticity in the subiculum. Prog 47. Bramham CR, Messaoudi E. BDNF function in adult synaptic plasticity: the synaptic
Neurobiol 89: 334 –342, 2009. consolidation hypothesis. Prog Neurobiol 76: 99 –125, 2005.

26. Belluscio MA, Mizuseki K, Schmidt R, Kempter R, Buzsaki G. Cross-frequency phase- 48. Bredt DS, Nicoll RA. AMPA receptor trafficking at excitatory synapses. Neuron 40:
phase coupling between theta and gamma oscillations in the hippocampus. J Neurosci 361–379, 2003.
32: 423– 435, 2012.
49. Bush D, Philippides A, Husbands P, O’Shea M. Spike-timing dependent plasticity and
27. Benke TA, Luthi A, Isaac JT, Collingridge GL. Modulation of AMPA receptor unitary the cognitive map. Front Computat Neurosci 4: 142, 2010.
conductance by synaptic activity. Nature 393: 793–797, 1998.
50. Buxton RS, Magee AI. Structure and interactions of desmosomal and other cadherins.
28. Bergmann TO, Molle M, Diedrichs J, Born J, Siebner HR. Sleep spindle-related reac- Semin Cell Biol 3: 157–167, 1992.
tivation of category-specific cortical regions after learning face-scene associations.
51. Buzsaki G. Hippocampal sharp waves: their origin and significance. Brain Res 398:
NeuroImage 59: 2733–2742, 2012.
242–252, 1986.
29. Bernardinelli Y, Randall J, Janett E, Nikonenko I, Konig S, Jones EV, Flores CE, Murai
52. Buzsaki G. Theta oscillations in the hippocampus. Neuron 33: 325–340, 2002.
KK, Bochet CG, Holtmaat A, Muller D. Activity-dependent structural plasticity of
perisynaptic astrocytic domains promotes excitatory synapse stability. Curr Biol 24: 53. Buzsaki G. Two-stage model of memory trace formation: a role for “noisy” brain
1679 –1688, 2014. states. Neuroscience 31: 551–570, 1989.

30. Betke KM, Wells CA, Hamm HE. GPCR mediated regulation of synaptic transmission. 54. Buzsaki G, Horvath Z, Urioste R, Hetke J, Wise K. High-frequency network oscillation
Prog Neurobiol 96: 304 –321, 2012. in the hippocampus. Science 256: 1025–1027, 1992.

31. Bezzi P, Carmignoto G, Pasti L, Vesce S, Rossi D, Rizzini BL, Pozzan T, Volterra A. 55. Cai DJ, Shuman T, Gorman MR, Sage JR, Anagnostaras SG. Sleep selectively enhances
Prostaglandins stimulate calcium-dependent glutamate release in astrocytes. Nature hippocampus-dependent memory in mice. Behav Neurosci 123: 713–719, 2009.
391: 281–285, 1998.
56. Cai DJ, Shuman T, Harrison EM, Sage JR, Anagnostaras SG. Sleep deprivation and
32. Bheda P, Schneider R. Epigenetics reloaded: the single-cell revolution. Trends Cell Biol Pavlovian fear conditioning. Learn Mem 16: 595–599, 2009.
24: 712–723, 2014.
57. Calabresi P, Picconi B, Tozzi A, Di Filippo M. Dopamine-mediated regulation of
33. Binder S, Baier PC, Molle M, Inostroza M, Born J, Marshall L. Sleep enhances memory corticostriatal synaptic plasticity. Trends Neurosci 30: 211–219, 2007.
consolidation in the hippocampus-dependent object-place recognition task in rats.
58. Canolty RT, Edwards E, Dalal SS, Soltani M, Nagarajan SS, Kirsch HE, Berger MS,
Neurobiol Learn Mem 97: 213–219, 2012.
Barbaro NM, Knight RT. High gamma power is phase-locked to theta oscillations in
34. Blanco W, Pereira CM, Cota VR, Souza AC, Renno-Costa C, Santos S, Dias G, Guer- human neocortex. Science 313: 1626 –1628, 2006.
reiro AM, Tort AB, Neto AD, Ribeiro S. Synaptic Homeostasis and Restructuring
59. Caroni P, Donato F, Muller D. Structural plasticity upon learning: regulation and
across the Sleep-Wake Cycle. PLoS Comput Biol 11: e1004241, 2015.
functions. Nature Rev Neurosci 13: 478 – 490, 2012.
35. Bliss TV, Collingridge GL. A synaptic model of memory: long-term potentiation in the
60. Celio MR, Spreafico R, De Biasi S, Vitellaro-Zuccarello L. Perineuronal nets: past and
hippocampus. Nature 361: 31–39, 1993. present. Trends Neurosci 21: 510 –515, 1998.
36. Bliss TV, Lomo T. Long-lasting potentiation of synaptic transmission in the dentate 61. Cesca F, Baldelli P, Valtorta F, Benfenati F. The synapsins: key actors of synapse
area of the anaesthetized rabbit following stimulation of the perforant path. J Physiol function and plasticity. Prog Neurobiol 91: 313–348, 2010.
232: 331–356, 1973.
62. Chen G, Kolbeck R, Barde YA, Bonhoeffer T, Kossel AH. Relative contribution of
37. Bloodgood BL, Giessel AJ, Sabatini BL. Biphasic synaptic Ca influx arising from com- endogenous neurotrophins in hippocampal long-term potentiation. J Neurosci 19:
partmentalized electrical signals in dendritic spines. PLoS Biol 7: e1000190, 2009. 7983–7990, 1999.

38. Bloodgood BL, Sabatini BL. Neuronal activity regulates diffusion across the neck of 63. Chen L, Cooper NG, Mower GD. Developmental changes in the expression of
dendritic spines. Science 310: 866 – 869, 2005. NMDA receptor subunits (NR1, NR2A, NR2B) in the cat visual cortex and the effects
of dark rearing. Brain Res Mol Brain Res 78: 196 –200, 2000.
39. Blundell J, Blaiss CA, Etherton MR, Espinosa F, Tabuchi K, Walz C, Bolliger MF, Sudhof
TC, Powell CM. Neuroligin-1 deletion results in impaired spatial memory and in- 64. Chevaleyre V, Castillo PE. Endocannabinoid-mediated metaplasticity in the hip-
creased repetitive behavior. J Neurosci 30: 2115–2129, 2010. pocampus. Neuron 43: 871– 881, 2004.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 683


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

65. Chevaleyre V, Castillo PE. Heterosynaptic LTD of hippocampal GABAergic synapses: 88. Destexhe A, Sejnowski TJ. Interactions between membrane conductances underlying
a novel role of endocannabinoids in regulating excitability. Neuron 38: 461– 472, 2003. thalamocortical slow-wave oscillations. Physiol Rev 83: 1401–1453, 2003.

66. Chevaleyre V, Takahashi KA, Castillo PE. Endocannabinoid-mediated synaptic plas- 89. Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, Gibson D,
ticity in the CNS. Annu Rev Neurosci 29: 37–76, 2006. Mandelbaum A, Etinger A, Mechoulam R. Isolation and structure of a brain constituent
that binds to the cannabinoid receptor. Science 258: 1946 –1949, 1992.
67. Chowdhury S, Shepherd JD, Okuno H, Lyford G, Petralia RS, Plath N, Kuhl D, Huganir
RL, Worley PF. Arc/Arg3.1 interacts with the endocytic machinery to regulate AMPA 90. Diba K, Buzsaki G. Forward and reverse hippocampal place-cell sequences during
receptor trafficking. Neuron 52: 445– 459, 2006. ripples. Nat Neurosci 10: 1241–1242, 2007.

68. Chrobak JJ, Buzsaki G. Selective activation of deep layer (V-VI) retrohippocampal 91. Diekelmann S, Born J. The memory function of sleep. Nat Rev Neurosci 11: 114 –126,
cortical neurons during hippocampal sharp waves in the behaving rat. J Neurosci 14: 2010.
6160 – 6170, 1994.
92. Dityatev A, Schachner M, Sonderegger P. The dual role of the extracellular matrix in
69. Chung WS, Barres BA. The role of glial cells in synapse elimination. Curr Opin Neurobiol synaptic plasticity and homeostasis. Nature Rev Neurosci 11: 735–746, 2010.
22: 438 – 445, 2012.
93. Draguhn A, Traub RD, Schmitz D, Jefferys JG. Electrical coupling underlies high-
70. Cingolani LA, Goda Y. Actin in action: the interplay between the actin cytoskeleton frequency oscillations in the hippocampus in vitro. Nature 394: 189 –192, 1998.
and synaptic efficacy. Nature Rev Neurosci 9: 344 –356, 2008.
94. Dudai Y. The restless engram: consolidations never end. Annu Rev Neurosci 35: 227–
71. Citri A, Malenka RC. Synaptic plasticity: multiple forms, functions, mechanisms. Neu- 247, 2012.
ropsychopharmacology 33: 18 – 41, 2008.
95. Dudek SM, Bear MF. Homosynaptic long-term depression in area CA1 of hippocam-
72. Clemens Z, Molle M, Eross L, Barsi P, Halasz P, Born J. Temporal coupling of para- pus and effects of N-methyl-D-aspartate receptor blockade. Proc Natl Acad Sci USA 89:
hippocampal ripples, sleep spindles and slow oscillations in humans. Brain 130: 2868 – 4363– 4367, 1992.
2878, 2007.
96. Easton A, Douchamps V, Eacott M, Lever C. A specific role for septohippocampal
73. Clemens Z, Molle M, Eross L, Jakus R, Rasonyi G, Halasz P, Born J. Fine-tuned coupling acetylcholine in memory? Neuropsychologia 50: 3156 –3168, 2012.
between human parahippocampal ripples and sleep spindles. Eur J Neurosci 33: 511–
97. Egan MF, Kojima M, Callicott JH, Goldberg TE, Kolachana BS, Bertolino A, Zaitsev E,
520, 2011.
Gold B, Goldman D, Dean M, Lu B, Weinberger DR. The BDNF val66met polymor-
74. Clopath C, Busing L, Vasilaki E, Gerstner W. Connectivity reflects coding: a model of phism affects activity-dependent secretion of BDNF and human memory and hip-
voltage-based STDP with homeostasis. Nat Neurosci 13: 344 –352, 2010. pocampal function. Cell 112: 257–269, 2003.

75. Colgin LL. Mechanisms and functions of theta rhythms. Annu Rev Neurosci 36: 295– 98. Ehlers MD. Activity level controls postsynaptic composition and signaling via the
312, 2013. ubiquitin-proteasome system. Nat Neurosci 6: 231–242, 2003.

76. Craig S, Commins S. Plastic and metaplastic changes in the CA1 and subicular projec- 99. Eichenbaum H. Hippocampus: cognitive processes and neural representations that
tions to the entorhinal cortex. Brain Res 1147: 124 –139, 2007. underlie declarative memory. Neuron 44: 109 –120, 2004.

77. Crick F, Mitchinson G. The function of dream sleep. Nature 304: 111–114, 1983. 100. Ekstrom AD, Meltzer J, McNaughton BL, Barnes CA. NMDA receptor antagonism
blocks experience-dependent expansion of hippocampal “place fields.” Neuron 31:
78. Dahlhaus R, Hines RM, Eadie BD, Kannangara TS, Hines DJ, Brown CE, Christie BR, 631– 638, 2001.
El-Husseini A. Overexpression of the cell adhesion protein neuroligin-1 induces learn-
ing deficits and impairs synaptic plasticity by altering the ratio of excitation to inhibition 101. Ellenbogen JM, Payne JD, Stickgold R. The role of sleep in declarative memory con-
in the hippocampus. Hippocampus 20: 305–322, 2010. solidation: passive, permissive, active or none? Curr Opin Neurobiol 16: 716 –722,
2006.
79. Dan Y, Poo MM. Spike timing-dependent plasticity: from synapse to perception.
Physiol Rev 86: 1033–1048, 2006. 102. Eller J, Zarnadze S, Bauerle P, Dugladze T, Gloveli T. Cell type-specific separation of
subicular principal neurons during network activities. PloS One 10: e0123636, 2015.
80. De Gennaro L, Ferrara M. Sleep spindles: an overview. Sleep Med Rev 7: 423– 440,
2003. 103. Engert F, Bonhoeffer T. Dendritic spine changes associated with hippocampal long-
term synaptic plasticity. Nature 399: 66 –70, 1999.
81. Dean C, Dresbach T. Neuroligins and neurexins: linking cell adhesion, synapse for-
mation and cognitive function. Trends Neurosci 29: 21–29, 2006. 104. Engert F, Bonhoeffer T. Synapse specificity of long-term potentiation breaks down at
short distances. Nature 388: 279 –284, 1997.
82. Dean C, Liu H, Mark DF, Chang PY, Jackson MB, Chapman ER. Synaptotagmin-IV
modulates synaptic function and long-term potentiation by regulating BDNF release. 105. Engert F, Tao HW, Zhang LI, Poo MM. Moving visual stimuli rapidly induce direction
sensitivity of developing tectal neurons. Nature 419: 470 – 475, 2002.
Nat Neurosci 12: 767–776, 2009.
106. Eroglu C, Barres BA. Regulation of synaptic connectivity by glia. Nature 468: 223–231,
83. Delekate A, Zagrebelsky M, Kramer S, Schwab ME, Korte M. NogoA restricts synap-
2010.
tic plasticity in the adult hippocampus on a fast time scale. Proc Natl Acad Sci USA 108:
2569 –2574, 2011. 107. Eschenko O, Molle M, Born J, Sara SJ. Elevated sleep spindle density after learning or
after retrieval in rats. J Neurosci 26: 12914 –12920, 2006.
84. Deller T, Korte M, Chabanis S, Drakew A, Schwegler H, Stefani GG, Zuniga A,
Schwarz K, Bonhoeffer T, Zeller R, Frotscher M, Mundel P. Synaptopodin-deficient 108. Fatemi M, Pao MM, Jeong S, Gal-Yam EN, Egger G, Weisenberger DJ, Jones PA.
mice lack a spine apparatus and show deficits in synaptic plasticity. Proc Natl Acad Sci Footprinting of mammalian promoters: use of a CpG DNA methyltransferase reveal-
USA 1000: 10494 –10499, 2003. ing nucleosome positions at a single molecule level. Nucleic Acids Res 33: e176, 2005.
85. Demiralp T, Bayraktaroglu Z, Lenz D, Junge S, Busch NA, Maess B, Ergen M, Herr- 109. Feldman DE. The spike-timing dependence of plasticity. Neuron 75: 556 –571, 2012.
mann CS. Gamma amplitudes are coupled to theta phase in human EEG during visual
perception. Int J Psychophysiol 64: 24 –30, 2007. 110. Fell J, Axmacher N. The role of phase synchronization in memory processes. Nature
Rev Neurosci 12: 105–118, 2011.
86. Deng K, Gao Y, Cao Z, Graziani EI, Wood A, Doherty P, Walsh FS. Overcoming
amino-Nogo-induced inhibition of cell spreading and neurite outgrowth by 12-O- 111. Fellin T, Pascual O, Gobbo S, Pozzan T, Haydon PG, Carmignoto G. Neuronal syn-
tetradecanoylphorbol-13-acetate-type tumor promoters. J Biol Chem 285: 6425– chrony mediated by astrocytic glutamate through activation of extrasynaptic NMDA
6433, 2010. receptors. Neuron 43: 729 –743, 2004.

87. Derkach VA, Oh MC, Guire ES, Soderling TR. Regulatory mechanisms of AMPA 112. Fenton AA, Muller RU. Place cell discharge is extremely variable during individual
receptors in synaptic plasticity. Nature Rev Neurosci 8: 101–113, 2007. passes of the rat through the firing field. Proc Natl Acad Sci USA 95: 3182–3187, 1998.

684 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

113. Fields RD. Myelin formation and remodeling. Cell 156: 15–17, 2014. 136. Gao Y, Deng K, Hou J, Bryson JB, Barco A, Nikulina E, Spencer T, Mellado W, Kandel
ER, Filbin MT. Activated CREB is sufficient to overcome inhibitors in myelin and
114. Fiete IR, Senn W, Wang CZ, Hahnloser RH. Spike-time-dependent plasticity and promote spinal axon regeneration in vivo. Neuron 44: 609 – 621, 2004.
heterosynaptic competition organize networks to produce long scale-free sequences
of neural activity. Neuron 65: 563–576, 2010. 137. Garner AR, Rowland DC, Hwang SY, Baumgaertel K, Roth BL, Kentros C, Mayford M.
Generation of a synthetic memory trace. Science 335: 1513–1516, 2012.
115. Fifkova E, Delay RJ. Cytoplasmic actin in neuronal processes as a possible mediator of
synaptic plasticity. J Cell Biol 95: 345–350, 1982. 138. Gasparini S, Saviane C, Voronin LL, Cherubini E. Silent synapses in the developing
hippocampus: lack of functional AMPA receptors or low probability of glutamate
116. Figurov A, Pozzo-Miller LD, Olafsson P, Wang T, Lu B. Regulation of synaptic re- release? Proc Natl Acad Sci USA 97: 9741–9746, 2000.
sponses to high-frequency stimulation and LTP by neurotrophins in the hippocampus.
Nature 381: 706 –709, 1996. 139. Gladding CM, Fitzjohn SM, Molnar E. Metabotropic glutamate receptor-mediated
long-term depression: molecular mechanisms. Pharmacol Rev 61: 395– 412, 2009.
117. Filosa A, Paixao S, Honsek SD, Carmona MA, Becker L, Feddersen B, Gaitanos L,
Rudhard Y, Schoepfer R, Klopstock T, Kullander K, Rose CR, Pasquale EB, Klein R. 140. Gogolla N, Caroni P, Luthi A, Herry C. Perineuronal nets protect fear memories from
Neuron-glia communication via EphA4/ephrin-A3 modulates LTP through glial gluta- erasure. Science 325: 1258 –1261, 2009.
mate transport. Nat Neurosci 12: 1285–1292, 2009.
141. Gold PE, Newman LA, Scavuzzo CJ, Korol DL. Modulation of multiple memory
118. Fino E, Deniau JM, Venance L. Cell-specific spike-timing-dependent plasticity in systems: from neurotransmitters to metabolic substrates. Hippocampus 23: 1053–
GABAergic and cholinergic interneurons in corticostriatal rat brain slices. J Physiol 586: 1065, 2013.
265–282, 2008.
142. Gonzalez-Burgos I, Feria-Velasco A. Serotonin/dopamine interaction in memory for-
119. Fischer M, Kaech S, Knutti D, Matus A. Rapid actin-based plasticity in dendritic spines. mation. Prog Brain Res 172: 603– 623, 2008.
Neuron 20: 847– 854, 1998.
143. Gordon U, Polsky A, Schiller J. Plasticity compartments in basal dendrites of neocor-
120. Fischer Y. The hippocampal intrinsic network oscillator. J Physiol 554: 156 –174, 2004. tical pyramidal neurons. J Neurosci 26: 12717–12726, 2006.

121. Fischer Y, Gahwiler BH, Thompson SM. Activation of intrinsic hippocampal theta 144. Gottmann K, Mittmann T, Lessmann V. BDNF signaling in the formation, maturation
oscillations by acetylcholine in rat septo-hippocampal cocultures. J Physiol 519: 405– and plasticity of glutamatergic and GABAergic synapses. Exp Brain Res 199: 203–234,
413, 1999. 2009.

122. Fonseca R, Nagerl UV, Morris RG, Bonhoeffer T. Competing for memory: hippocam- 145. Gottschalk W, Pozzo-Miller LD, Figurov A, Lu B. Presynaptic modulation of synaptic
pal LTP under regimes of reduced protein synthesis. Neuron 44: 1011–1020, 2004. transmission and plasticity by brain-derived neurotrophic factor in the developing
hippocampus. J Neurosci 18: 6830 – 6839, 1998.
123. Fonseca R, Vabulas RM, Hartl FU, Bonhoeffer T, Nagerl UV. A balance of protein
synthesis and proteasome-dependent degradation determines the maintenance of 146. Govindarajan A, Kelleher RJ, Tonegawa S. A clustered plasticity model of long-term
LTP. Neuron 52: 239 –245, 2006. memory engrams. Nat Rev Neurosci 7: 575–583, 2006.

124. Fournier AE, GrandPre T, Strittmatter SM. Identification of a receptor mediating 147. Greer PL, Greenberg ME. From synapse to nucleus: calcium-dependent gene tran-
Nogo-66 inhibition of axonal regeneration. Nature 409: 341–346, 2001. scription in the control of synapse development and function. Neuron 59: 846 – 860,
2008.
125. Frankland PW, O’Brien C, Ohno M, Kirkwood A, Silva AJ. Alpha-CaMKII-dependent
plasticity in the cortex is required for permanent memory. Nature 411: 309 –313, 148. Grooms SY, Noh KM, Regis R, Bassell GJ, Bryan MK, Carroll RC, Zukin RS. Activity
2001. bidirectionally regulates AMPA receptor mRNA abundance in dendrites of hippocam-
pal neurons. J Neurosci 26: 8339 – 8351, 2006.
126. Freund TF, Katona I, Piomelli D. Role of endogenous cannabinoids in synaptic signal-
ing. Physiol Rev 83: 1017–1066, 2003. 149. Grunstein M. Histone acetylation in chromatin structure and transcription. Nature
389: 349 –352, 1997.
127. Frey U, Morris RG. Synaptic tagging and long-term potentiation. Nature 385: 533–
536, 1997. 150. Gundelfinger ED, Fejtova A. Molecular organization and plasticity of the cytomatrix at
the active zone. Curr Opin Neurobiol 22: 423– 430, 2012.
128. Frischknecht R, Heine M, Perrais D, Seidenbecher CI, Choquet D, Gundelfinger ED.
Brain extracellular matrix affects AMPA receptor lateral mobility and short-term 151. Gundelfinger ED, Frischknecht R, Choquet D, Heine M. Converting juvenile into adult
synaptic plasticity. Nat Neurosci 12: 897–904, 2009. plasticity: a role for the brain’s extracellular matrix. Eur J Neurosci 31: 2156 –2165,
2010.
129. Froemke RC, Poo MM, Dan Y. Spike-timing-dependent synaptic plasticity depends on
dendritic location. Nature 434: 221–225, 2005. 152. Gurskaya NG, Verkhusha VV, Shcheglov AS, Staroverov DB, Chepurnykh TV, Frad-
kov AF, Lukyanov S, Lukyanov KA. Engineering of a monomeric green-to-red photo-
130. Fu YX, Djupsund K, Gao H, Hayden B, Shen K, Dan Y. Temporal specificity in the activatable fluorescent protein induced by blue light. Nat Biotechnol 24: 461– 465,
cortical plasticity of visual space representation. Science 296: 1999 –2003, 2002. 2006.

131. Fuenzalida M, Fernandez de Sevilla D, Buno W. Changes of the EPSP waveform 153. Hagewoud R, Bultsma LJ, Barf RP, Koolhaas JM, Meerlo P. Sleep deprivation impairs
regulate the temporal window for spike-timing-dependent plasticity. J Neurosci 27: contextual fear conditioning and attenuates subsequent behavioural, endocrine and
11940 –11948, 2007. neuronal responses. J Sleep Res 20: 259 –266, 2011.

132. Fuenzalida M, Fernandez de Sevilla D, Couve A, Buno W. Role of AMPA and NMDA 154. Hagewoud R, Havekes R, Tiba PA, Novati A, Hogenelst K, Weinreder P, Van der Zee
receptors and back-propagating action potentials in spike timing-dependent plasticity. EA, Meerlo P. Coping with sleep deprivation: shifts in regional brain activity and
J Neurophysiol 103: 47–54, 2010. learning strategy. Sleep 33: 1465–1473, 2010.

133. Fukazawa Y, Saitoh Y, Ozawa F, Ohta Y, Mizuno K, Inokuchi K. Hippocampal LTP is 155. Hagewoud R, Whitcomb SN, Heeringa AN, Havekes R, Koolhaas JM, Meerlo P. A
accompanied by enhanced F-actin content within the dendritic spine that is essential time for learning and a time for sleep: the effect of sleep deprivation on contextual fear
for late LTP maintenance in vivo. Neuron 38: 447– 460, 2003. conditioning at different times of the day. Sleep 33: 1315–1322, 2010.

134. Fukunaga K, Muller D, Miyamoto E. Increased phosphorylation of Ca2⫹/calmodulin- 156. Hajos N, Karlocai MR, Nemeth B, Ulbert I, Monyer H, Szabo G, Erdelyi F, Freund TF,
dependent protein kinase II and its endogenous substrates in the induction of long- Gulyas AI. Input-output features of anatomically identified CA3 neurons during hip-
term potentiation. J Biol Chem 270: 6119 – 6124, 1995. pocampal sharp wave/ripple oscillation in vitro. J Neurosci 33: 11677–11691, 2013.

135. Gais S, Molle M, Helms K, Born J. Learning-dependent increases in sleep spindle 157. Hall J, Thomas KL, Everitt BJ. Rapid and selective induction of BDNF expression in the
density. J Neurosci 22: 6830 – 6834, 2002. hippocampus during contextual learning. Nat Neurosci 3: 533–535, 2000.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 685


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

158. Hao J, Oertner TG. Depolarization gates spine calcium transients and spike-timing- 181. Hyafil F, Babinet C, Jacob F. Cell-cell interactions in early embryogenesis: a molecular
dependent potentiation. Curr Opin Neurobiol 22: 509 –515, 2012. approach to the role of calcium. Cell 26: 447– 454, 1981.

159. Harand C, Bertran F, Doidy F, Guenole F, Desgranges B, Eustache F, Rauchs G. How 182. Inostroza M, Brotons-Mas JR, Laurent F, Cid E, de la Prida LM. Specific impairment of
aging affects sleep-dependent memory consolidation? Front Neurol 3: 8, 2012. “what-where-when” episodic-like memory in experimental models of temporal lobe
epilepsy. J Neurosci 33: 17749 –17762, 2013.
160. Hardingham GE, Arnold FJ, Bading H. Nuclear calcium signaling controls CREB-
mediated gene expression triggered by synaptic activity. Nat Neurosci 4: 261–267, 183. Isomura Y, Sirota A, Ozen S, Montgomery S, Mizuseki K, Henze DA, Buzsaki G.
2001. Integration and segregation of activity in entorhinal-hippocampal subregions by neo-
cortical slow oscillations. Neuron 52: 871– 882, 2006.
161. Hariri AR, Goldberg TE, Mattay VS, Kolachana BS, Callicott JH, Egan MF, Weinberger
DR. Brain-derived neurotrophic factor val66met polymorphism affects human mem- 184. Jahn R, Fasshauer D. Molecular machines governing exocytosis of synaptic vesicles.
ory-related hippocampal activity and predicts memory performance. J Neurosci 23: Nature 490: 201–207, 2012.
6690 – 6694, 2003.
185. Jaramillo J, Schmidt R, Kempter R. Modeling inheritance of phase precession in the
162. Harris KD, Henze DA, Hirase H, Leinekugel X, Dragoi G, Czurko A, Buzsaki G. Spike hippocampal formation. J Neurosci 34: 7715–7731, 2014.
train dynamics predicts theta-related phase precession in hippocampal pyramidal
cells. Nature 417: 738 –741, 2002. 186. Jensen O, Lisman JE. Position reconstruction from an ensemble of hippocampal place
cells: contribution of theta phase coding. J Neurophysiol 83: 2602–2609, 2000.
163. Hashimotodani Y, Ohno-Shosaku T, Tsubokawa H, Ogata H, Emoto K, Maejima T,
Araishi K, Shin HS, Kano M. Phospholipase Cbeta serves as a coincidence detector 187. Jinno S, Klausberger T, Marton LF, Dalezios Y, Roberts JD, Fuentealba P, Bushong EA,
through its Ca2⫹ dependency for triggering retrograde endocannabinoid signal. Neu- Henze D, Buzsaki G, Somogyi P. Neuronal diversity in GABAergic long-range projec-
ron 45: 257–268, 2005. tions from the hippocampus. J Neurosci 27: 8790 – 8804, 2007.

164. Hayashi Y, Shi SH, Esteban JA, Piccini A, Poncer JC, Malinow R. Driving AMPA 188. Johenning FW, Beed PS, Trimbuch T, Bendels MH, Winterer J, Schmitz D. Dendritic
receptors into synapses by LTP and CaMKII: requirement for GluR1 and PDZ domain compartment and neuronal output mode determine pathway-specific long-term po-
interaction. Science 287: 2262–2267, 2000. tentiation in the piriform cortex. J Neurosci 29: 13649 –13661, 2009.

165. Hebb DO. The Organization of Behavior. A Neuropsychological Theory. New York: 189. Johenning FW, Theis AK, Pannasch U, Ruckl M, Rudiger S, Schmitz D. Ryanodine
Wiley, 1949. receptor activation induces long-term plasticity of spine calcium dynamics. PLoS Biol
13: e1002181, 2015.
166. Henneberger C, Papouin T, Oliet SH, Rusakov DA. Long-term potentiation depends
on release of D-serine from astrocytes. Nature 463: 232–236, 2010. 190. Joset A, Dodd DA, Halegoua S, Schwab ME. Pincher-generated Nogo-A endo-
somes mediate growth cone collapse and retrograde signaling. J Cell Biol 188:
167. Herron CE, Lester RA, Coan EJ, Collingridge GL. Frequency-dependent involvement 271–285, 2010.
of NMDA receptors in the hippocampus: a novel synaptic mechanism. Nature 322:
265–268, 1986. 191. Josselyn SA, Kohler S, Frankland PW. Finding the engram. Nature Rev Neurosci 16:
521–534, 2015.
168. Hitti FL, Siegelbaum SA. The hippocampal CA2 region is essential for social memory.
Nature 508: 88 –92, 2014. 192. Jou TS, Stewart DB, Stappert J, Nelson WJ, Marrs JA. Genetic and biochemical dis-
section of protein linkages in the cadherin-catenin complex. Proc Natl Acad Sci USA 92:
169. Ho VM, Lee JA, Martin KC. The cell biology of synaptic plasticity. Science 334: 623–
5067–5071, 1995.
628, 2011.
193. Ju W, Morishita W, Tsui J, Gaietta G, Deerinck TJ, Adams SR, Garner CC, Tsien RY,
170. Holbro N, Grunditz A, Wiegert JS, Oertner TG. AMPA receptors gate spine Ca2⫹
Ellisman MH, Malenka RC. Activity-dependent regulation of dendritic synthesis and
transients and spike-timing-dependent potentiation. Proc Natl Acad Sci USA 107:
trafficking of AMPA receptors. Nat Neurosci 7: 244 –253, 2004.
15975–15980, 2010.
194. Jung SY, Kim J, Kwon OB, Jung JH, An K, Jeong AY, Lee CJ, Choi YB, Bailey CH, Kandel
171. Holthoff K, Zecevic D, Konnerth A. Rapid time course of action potentials in spines
ER, Kim JH. Input-specific synaptic plasticity in the amygdala is regulated by neuroli-
and remote dendrites of mouse visual cortex neurons. J Physiol 588: 1085–1096, 2010.
gin-1 via postsynaptic NMDA receptors. Proc Natl Acad Sci USA 107: 4710 – 4715,
172. Holtmaat A, Svoboda K. Experience-dependent structural synaptic plasticity in the 2010.
mammalian brain. Nat Rev Neurosci 10: 647– 658, 2009.
195. Kamondi A, Acsady L, Wang XJ, Buzsaki G. Theta oscillations in somata and dendrites
173. Honkura N, Matsuzaki M, Noguchi J, Ellis-Davies GC, Kasai H. The subspine organi- of hippocampal pyramidal cells in vivo: activity-dependent phase-precession of action
zation of actin fibers regulates the structure and plasticity of dendritic spines. Neuron potentials. Hippocampus 8: 244 –261, 1998.
57: 719 –729, 2008.
196. Kandel ER. The molecular biology of memory storage: a dialogue between genes and
174. Hotulainen P, Llano O, Smirnov S, Tanhuanpaa K, Faix J, Rivera C, Lappalainen P. synapses. Science 294: 1030 –1038, 2001.
Defining mechanisms of actin polymerization and depolymerization during dendritic
197. Kandel ER. The molecular biology of memory: cAMP, PKA, CRE, CREB-1, CREB-2,
spine morphogenesis. J Cell Biol 185: 323–339, 2009.
and CPEB. Mol Brain 5: 14, 2012.
175. Huang YY, Kandel ER. Theta frequency stimulation induces a local form of late phase
198. Kandel ER, Dudai Y, Mayford MR. The molecular and systems biology of memory. Cell
LTP in the CA1 region of the hippocampus. Learn Mem 12: 587–593, 2005.
157: 163–186, 2014.
176. Huber KM, Kayser MS, Bear MF. Role for rapid dendritic protein synthesis in hip-
pocampal mGluR- dependent long-term depression. Science 288: 1254 –1257, 2000. 199. Kang H, Schuman EM. Long-lasting neutrotrophin-induced enhancement of synaptic
transmisson in the adult hippocampus. Science 267: 1658 –1662, 1995.
177. Huganir RL, Nicoll RA. AMPARs and synaptic plasticity: the last 25 years. Neuron 80:
704 –717, 2013. 200. Kang H, Schuman EM. A requirement for local protein synthesis in neurotrophin-
induced hippocampal synaptic plasticity. Science 273: 1402–1406, 1996.
178. Hulme SR, Jones OD, Abraham WC. Emerging roles of metaplasticity in behaviour
and disease. Trends Neurosci 36: 353–362, 2013. 201. Kang H, Welcher AA, Shelton D, Schuman EM. Neurotrophins and time: different
roles for TrkB signaling in hippocampal long-term potentiation. Neuron 19: 653– 664,
179. Hur YS, Kim KD, Paek SH, Yoo SH. Evidence for the existence of secretory granule 1997.
(dense-core vesicle)-based inositol 1,4,5-trisphosphate-dependent Ca2⫹ signaling
system in astrocytes. PloS One 5: e11973, 2010. 202. Karpova A, Mikhaylova M, Bera S, Bar J, Reddy PP, Behnisch T, Rankovic V, Spilker C,
Bethge P, Sahin J, Kaushik R, Zuschratter W, Kahne T, Naumann M, Gundelfinger ED,
180. Huxter J, Burgess N, O’Keefe J. Independent rate and temporal coding in hippocampal Kreutz MR. Encoding and transducing the synaptic or extrasynaptic origin of NMDA
pyramidal cells. Nature 425: 828 – 832, 2003. receptor signals to the nucleus. Cell 152: 1119 –1133, 2013.

686 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

203. Karpova NN. Role of BDNF epigenetics in activity-dependent neuronal plasticity. 225. Knierim JJ, Lee I, Hargreaves EL. Hippocampal place cells: parallel input streams,
Neuropharmacology 76: 709 –718, 2014. subregional processing, and implications for episodic memory. Hippocampus 16: 755–
764, 2006.
204. Kartvelishvily E, Shleper M, Balan L, Dumin E, Wolosker H. Neuron-derived D-serine
release provides a novel means to activate N-methyl-D-aspartate receptors. J Biol 226. Kocsis B, Bragin A, Buzsaki G. Interdependence of multiple theta generators in the
Chem 281: 14151–14162, 2006. hippocampus: a partial coherence analysis. J Neurosci 19: 6200 – 6212, 1999.

205. Katona I, Freund TF. Endocannabinoid signaling as a synaptic circuit breaker in neu- 227. Koester HJ, Sakmann B. Calcium dynamics in single spines during coincident pre- and
rological disease. Nat Med 14: 923–930, 2008. postsynaptic activity depend on relative timing of back-propagating action potentials
and subthreshold excitatory postsynaptic potentials. Proc Natl Acad Sci USA 95: 9596 –
206. Katona L, Lapray D, Viney TJ, Oulhaj A, Borhegyi Z, Micklem BR, Klausberger T, 9601, 1998.
Somogyi P. Sleep and movement differentiates actions of two types of somatostatin-
expressing GABAergic interneuron in rat hippocampus. Neuron 82: 872– 886, 2014. 228. Kohara K, Pignatelli M, Rivest AJ, Jung HY, Kitamura T, Suh J, Frank D, Kajikawa K,
Mise N, Obata Y, Wickersham IR, Tonegawa S. Cell type-specific genetic and opto-
207. Kempf A, Schwab ME. Nogo-A represses anatomical and synaptic plasticity in the genetic tools reveal hippocampal CA2 circuits. Nat Neurosci 17: 269 –279, 2014.
central nervous system. Physiology 28: 151–163, 2013.
229. Koppe G, Bruckner G, Brauer K, Hartig W, Bigl V. Developmental patterns of pro-
208. Kempf A, Tews B, Arzt ME, Weinmann O, Obermair FJ, Pernet V, Zagrebelsky M, teoglycan-containing extracellular matrix in perineuronal nets and neuropil of the
Delekate A, Iobbi C, Zemmar A, Ristic Z, Gullo M, Spies P, Dodd D, Gygax D, Korte postnatal rat brain. Cell Tissue Res 288: 33– 41, 1997.
M, Schwab ME. The sphingolipid receptor S1PR2 is a receptor for Nogo-A repressing
synaptic plasticity. PLoS Biol 12: e1001763, 2014. 230. Korkotian E, Frotscher M, Segal M. Synaptopodin regulates spine plasticity: mediation
by calcium stores. J Neurosci 34: 11641–11651, 2014.
209. Kesner RP. Behavioral functions of the CA3 subregion of the hippocampus. Learn Mem
14: 771–781, 2007. 231. Korte M. Neuroscience. A protoplasmic kiss to remember. Science 319: 1627–1628,
2008.
210. Kettenmann H, Ransom BR. Neuroglia. New York: Oxford Univ. Press, 2013.
232. Korte M, Carroll P, Wolf E, Brem G, Thoenen H, Bonhoeffer T. Hippocampal long-
211. Kiebler MA, Bassell GJ. Neuronal RNA granules: movers and makers. Neuron 51: term potentiation is impaired in mice lacking brain-derived neurotrophic factor. Proc
685– 690, 2006. Natl Acad Sci USA 92: 8856 – 8860, 1995.

212. Kiebler MA, DesGroseillers L. Molecular insights into mRNA transport and local 233. Korte M, Griesbeck O, Gravel C, Carroll P, Staiger V, Thoenen H, Bonhoeffer T.
translation in the mammalian nervous system. Neuron 25: 19 –28, 2000. Virus-mediated gene transfer into hippocampal CA1 region restores long-term po-
tentiation in brain-derived neurotrophic factor mutant mice. Proc Natl Acad Sci USA
213. Kim CH, Lisman JE. A role of actin filament in synaptic transmission and long-term 93: 12547–12552, 1996.
potentiation. J Neurosci 19: 4314 – 4324, 1999.
234. Korte M, Kang H, Bonhoeffer T, Schuman EM. A role for BDNF in the late-phase of
214. Kim E, Sheng M. PDZ domain proteins of synapses. Nature Rev Neurosci 5: 771–781, hippocampal long-term potentiation. Neuropharmacology 37: 553–559, 1998.
2004.
235. Kovalchuk Y, Hanse E, Kafitz KW, Konnerth A. Postsynaptic induction of BDNF-
215. Kim J, Jung SY, Lee YK, Park S, Choi JS, Lee CJ, Kim HS, Choi YB, Scheiffele P, Bailey mediated long-term potentiation. Science 295: 1729 –1734, 2002.
CH, Kandel ER, Kim JH. Neuroligin-1 is required for normal expression of LTP and
associative fear memory in the amygdala of adult animals. Proc Natl Acad Sci USA 105: 236. Kramar EA, Lin B, Rex CS, Gall CM, Lynch G. Integrin-driven actin polymerization
9087–9092, 2008. consolidates long-term potentiation. Proc Natl Acad Sci USA 103: 5579 –5584, 2006.

216. Kim K, Lakhanpal G, Lu HE, Khan M, Suzuki A, Kato-Hayashi M, Narayanan R, Luyben 237. Kreitzer AC, Regehr WG. Retrograde signaling by endocannabinoids. Curr Opin Neu-
TT, Matsuda T, Nagai T, Blanpied TA, Hayashi Y, Okamoto K. A temporary gating of robiol 12: 324 –330, 2002.
actin remodeling during synaptic plasticity consists of the interplay between the kinase
238. Krucker T, Siggins GR, Halpain S. Dynamic actin filaments are required for stable
and structural functions of CaMKII. Neuron 87: 813– 826, 2015.
long-term potentiation (LTP) in area CA1 of the hippocampus. Proc Natl Acad Sci USA
217. Kim SA, Tai CY, Mok LP, Mosser EA, Schuman EM. Calcium-dependent dynamics of 97: 6856 – 6861, 2000.
cadherin interactions at cell-cell junctions. Proc Natl Acad Sci USA 108: 9857–9862,
239. Krueger DD, Tuffy LP, Papadopoulos T, Brose N. The role of neurexins and neuroli-
2011.
gins in the formation, maturation, and function of vertebrate synapses. Curr Opin
218. Kim Y, Spruston N. Target-specific output patterns are predicted by the distribution Neurobiol 22: 412– 422, 2012.
of regular-spiking and bursting pyramidal neurons in the subiculum. Hippocampus 22:
240. Kudrimoti HS, Barnes CA, McNaughton BL. Reactivation of hippocampal cell assem-
693–706, 2012.
blies: effects of behavioral state, experience, and EEG dynamics. J Neurosci 19: 4090 –
219. Kitajima T, Hara K. A model of the mechanism of cooperativity and associativity of 4101, 1999.
long-term potentiation in the hippocampus: a fundamental mechanism of associative
241. Kullmann DM. The Mother of All Battles 20 years on: is LTP expressed pre- or
memory and learning. Biol Cybern 64: 365–371, 1991.
postsynaptically? J Physiol 590: 2213–2216, 2012.
220. Klausberger T, Magill PJ, Marton LF, Roberts JD, Cobden PM, Buzsaki G, Somogyi P.
242. Kullmann DM, Lamsa KP. LTP and LTD in cortical GABAergic interneurons: emerging
Brain-state- and cell-type-specific firing of hippocampal interneurons in vivo. Nature
rules and roles. Neuropharmacology 60: 712–719, 2011.
421: 844 – 848, 2003.
243. Kwon HB, Sabatini BL. Glutamate induces de novo growth of functional spines in
221. Klausberger T, Marton LF, Baude A, Roberts JD, Magill PJ, Somogyi P. Spike timing of developing cortex. Nature 474: 100 –104, 2011.
dendrite-targeting bistratified cells during hippocampal network oscillations in vivo.
Nat Neurosci 7: 41– 47, 2004. 244. Lalo U, Andrew J, Palygin O, Pankratov Y. Ca2⫹-dependent modulation of GABAA
and NMDA receptors by extracellular ATP: implication for function of tripartite
222. Klausberger T, Marton LF, O’Neill J, Huck JH, Dalezios Y, Fuentealba P, Suen WY, synapse. Biochem Soc Trans 37: 1407–1411, 2009.
Papp E, Kaneko T, Watanabe M, Csicsvari J, Somogyi P. Complementary roles of
cholecystokinin- and parvalbumin-expressing GABAergic neurons in hippocampal 245. Landry CD, Kandel ER, Rajasethupathy P. New mechanisms in memory storage:
network oscillations. J Neurosci 25: 9782–9793, 2005. piRNAs and epigenetics. Trends Neurosci 36: 535–542, 2013.

223. Klausberger T, Somogyi P. Neuronal diversity and temporal dynamics: the unity of 246. Leconte P, Hennevin E. [Temporal characteristics of the augmentation of paradoxal
hippocampal circuit operations. Science 321: 53–57, 2008. sleep following learning in the rat]. Physiol Behav 11: 677– 686, 1973.

224. Klein R. Bidirectional modulation of synaptic functions by Eph/ephrin signaling. Nat 247. Lee AK, Wilson MA. Memory of sequential experience in the hippocampus during
Neurosci 12: 15–20, 2009. slow wave sleep. Neuron 36: 1183–1194, 2002.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 687


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

248. Lee AM, Kanter BR, Wang D, Lim JP, Zou ME, Qiu C, McMahon T, Dadgar J, Fisch- 271. Majewska A, Brown E, Ross J, Yuste R. Mechanisms of calcium decay kinetics in
bach-Weiss SC, Messing RO. Prkcz null mice show normal learning and memory. hippocampal spines: role of spine calcium pumps and calcium diffusion through the
Nature 493: 416 – 419, 2013. spine neck in biochemical compartmentalization. J Neurosci 20: 1722–1734, 2000.

249. Lee H, Raiker SJ, Venkatesh K, Geary R, Robak LA, Zhang Y, Yeh HH, Shrager P, Giger 272. Malenka RC, Bear MF. LTP and LTD: an embarrassment of riches. Neuron 44: 5–21,
RJ. Synaptic function for the Nogo-66 receptor NgR1: regulation of dendritic spine 2004.
morphology and activity-dependent synaptic strength. J Neurosci 28: 2753–2765,
2008. 273. Malenka RC, Kauer JA, Perkel DJ, Mauk MD, Kelly PT, Nicoll RA, Waxham MN. An
essential role for postsynaptic calmodulin and protein kinase activity in long-term
250. Lee HK, Barbarosie M, Kameyama K, Bear MF, Huganir RL. Regulation of distinct potentiation. Nature 340: 554 –557, 1989.
AMPA receptor phosphorylation sites during bidirectional synaptic plasticity. Nature
405: 955–959, 2000. 274. Malenka RC, Nicoll RA. Long-term potentiation–a decade of progress? Science 285:
1870 –1874, 1999.
251. Lee YS, Silva AJ. The molecular and cellular biology of enhanced cognition. Nat Rev
Neurosci 10: 126 –140, 2009. 275. Maletic-Savatic M, Malinow R, Svoboda K. Rapid dendritic morphogenesis in CA1
hippocampal dendrites induced by synaptic activity. Science 283: 1923–1927, 1999.
252. Lessmann V, Gottmann K, Heumann R. BDNF and NT-4/5 enhance glutamatergic
synaptic transmission in cultured hippocampal neurones. NeuroReport 6: 21–25, 1994. 276. Malinow R, Malenka RC. AMPA receptor trafficking and synaptic plasticity. Annu Rev
Neurosci 25: 103–126, 2002.
253. Levenson JM, Sweatt JD. Epigenetic mechanisms in memory formation. Nat Rev Neu-
rosci 6: 108 –118, 2005. 277. Malinow R, Schulman H, Tsien RW. Inhibition of postsynaptic PKC or CaMKII blocks
induction but not expression of LTP. Science 245: 862– 866, 1989.
254. Li JS, Chao YS. Electrolytic lesions of dorsal CA3 impair episodic-like memory in rats.
Neurobiol Learn Mem 89: 192–198, 2008. 278. Mander BA, Rao V, Lu B, Saletin JM, Lindquist JR, Ancoli-Israel S, Jagust W, Walker MP.
Prefrontal atrophy, disrupted NREM slow waves and impaired hippocampal-depen-
255. Lin B, Kramar EA, Bi X, Brucher FA, Gall CM, Lynch G. Theta stimulation polymerizes dent memory in aging. Nat Neurosci 16: 357–364, 2013.
actin in dendritic spines of hippocampus. J Neurosci 25: 2062–2069, 2005.
279. Manns JR, Eichenbaum H. Evolution of declarative memory. Hippocampus 16: 795–
256. Lisman J, Morris RG. Memory. Why is the cortex a slow learner? Nature 411: 248 – 808, 2006.
249, 2001.
280. Markram H, Lubke J, Frotscher M, Sakmann B. Regulation of synaptic efficacy by
257. Lisman J, Spruston N. Postsynaptic depolarization requirements for LTP and LTD: a
coincidence of postsynaptic APs and EPSPs. Science 275: 213–215, 1997.
critique of spike timing-dependent plasticity. Nat Neurosci 8: 839 – 841, 2005.
281. Markus EJ, Qin YL, Leonard B, Skaggs WE, McNaughton BL, Barnes CA. Interactions
258. Lisman J, Spruston N. Questions about STDP as a General Model of Synaptic Plastic-
between location and task affect the spatial and directional firing of hippocampal
ity. Front Synaptic Neurosci 2: 140, 2010.
neurons. J Neurosci 15: 7079 –7094, 1995.
259. Litvak V, Zeller D, Oostenveld R, Maris E, Cohen A, Schramm A, Gentner R, Zaaroor
282. Marshall L, Helgadottir H, Molle M, Born J. Boosting slow oscillations during sleep
M, Pratt H, Classen J. LTP-like changes induced by paired associative stimulation of
potentiates memory. Nature 444: 610 – 613, 2006.
the primary somatosensory cortex in humans: source analysis and associated changes
in behaviour. Eur J Neurosci 25: 2862–2874, 2007. 283. Martin KC, Casadio A, Zhu H, YE, Rose JC, Chen M, Bailey CH, Kandel ER. Synapse-
specific, long-term facilitation of Aplysia sensory to motor synapses: a function for local
260. Liu X, Ramirez S, Pang PT, Puryear CB, Govindarajan A, Deisseroth K, Tonegawa S.
protein synthesis in memory storage. Cell 91: 927–938, 1997.
Optogenetic stimulation of a hippocampal engram activates fear memory recall. Na-
ture 484: 381–385, 2012. 284. Martin SJ, Grimwood PD, Morris RG. Synaptic plasticity and memory: an evaluation of
the hypothesis. Annu Rev Neurosci 23: 649 –711, 2000.
261. Liu YY, Jin WL, Liu HL, Ju G. Electron microscopic localization of Nogo-A at the
postsynaptic active zone of the rat. Neurosci Lett 346: 153–156, 2003. 285. Maslarova A, Lippmann K, Salar S, Rosler A, Heinemann U. Differential participation
of pyramidal cells in generation of spontaneous sharp wave-ripples in the mouse
262. Lohof AM, Ip NY, Poo MM. Potentiation of developing neuromuscular synapses by
the neurotrophins NT-3 and BDNF. Nature 363: 350 –353, 1993. subiculum in vitro. Neurobiol Learn Mem 125: 113–119, 2015.

263. Lopez de Maturana R, Sanchez-Pernaute R. Regulation of corticostriatal synaptic 286. Matsuzaki M, Honkura N, Ellis-Davies GC, Kasai H. Structural basis of long-term
plasticity by G protein-coupled receptors. CNS Neurol Disorders Drug Targets 9: 601– potentiation in single dendritic spines. Nature 429: 761–766, 2004.
615, 2010.
287. Matus A, Ackermann M, Pehling G, Byers HR, Fujiwara K. High actin concentrations
264. Louie K, Wilson MA. Temporally structured replay of awake hippocampal ensemble in brain dendritic spines and postsynaptic densities. Proc Natl Acad Sci USA 79: 7590 –
activity during rapid eye movement sleep. Neuron 29: 145–156, 2001. 7594, 1982.

265. Lu W, Man H, Ju W, Trimble WS, MacDonald JF, Wang YT. Activation of synaptic 288. Matz J, Gilyan A, Kolar A, McCarvill T, Krueger SR. Rapid structural alterations of the
NMDA receptors induces membrane insertion of new AMPA receptors and LTP in active zone lead to sustained changes in neurotransmitter release. Proc Natl Acad Sci
cultured hippocampal neurons. Neuron 29: 243–254, 2001. USA 107: 8836 – 8841, 2010.

266. Lubin FD, Roth TL, Sweatt JD. Epigenetic regulation of BDNF gene transcription in 289. McAllister AK, Katz LC, Lo DC. Opposing roles for endogenous BDNF and NT-3 in
the consolidation of fear memory. J Neurosci 28: 10576 –10586, 2008. regulating cortical dendritic growth. Neuron 18: 767–778, 1997.

267. Lüthi A, Laurent JP, Figurov A, Muller D, Schachner M. Hippocampal long-term 290. McGee AW, Yang Y, Fischer QS, Daw NW, Strittmatter SM. Experience-driven
potentiation and neural cell adhesion molecules L1 and NCAM. Nature 372: 777–779, plasticity of visual cortex limited by myelin and Nogo receptor. Science 309: 2222–
1994. 2226, 2005.

268. Lynch G, Rex CS, Gall CM. LTP consolidation: substrates, explanatory power, and 291. McMahon DB, Leopold DA. Stimulus timing-dependent plasticity in high-level vision.
functional significance. Neuropharmacology 52: 12–23, 2007. Curr Biol 22: 332–337, 2012.

269. Mabb AM, Ehlers MD. Ubiquitination in postsynaptic function and plasticity. Annu Rev 292. McNaughton BL, Barnes CA, O’Keefe J. The contributions of position, direction, and
Cell Dev Biol 26: 179 –210, 2010. velocity to single unit activity in the hippocampus of freely-moving rats. Exp Brain Res
52: 41– 49, 1983.
270. Maier N, Tejero-Cantero A, Dorrn AL, Winterer J, Beed PS, Morris G, Kempter R,
Poulet JF, Leibold C, Schmitz D. Coherent phasic excitation during hippocampal 293. Mehta MR, Quirk MC, Wilson MA. Experience-dependent asymmetric shape of hip-
ripples. Neuron 72: 137–152, 2011. pocampal receptive fields. Neuron 25: 707–715, 2000.

688 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

294. Messaoudi E, Kanhema T, Soule J, Tiron A, Dagyte G, da SB, Bramham CR. Sustained 316. Nakashiba T, Buhl DL, McHugh TJ, Tonegawa S. Hippocampal CA3 output is crucial
Arc/Arg3.1 synthesis controls long-term potentiation consolidation through regula- for ripple-associated reactivation and consolidation of memory. Neuron 62: 781–787,
tion of local actin polymerization in the dentate gyrus in vivo. J Neurosci 27: 10445– 2009.
10455, 2007.
317. Navakkode S, Sajikumar S, Frey JU. Mitogen-activated protein kinase-mediated rein-
295. Meyer D, Bonhoeffer T, Scheuss V. Balance and stability of synaptic structures during forcement of hippocampal early long-term depression by the type IV-specific phos-
synaptic plasticity. Neuron 82: 430 – 443, 2014. phodiesterase inhibitor rolipram and its effect on synaptic tagging. J Neurosci 25:
10664 –10670, 2005.
296. Milner B, Squire LR, Kandel ER. Cognitive neuroscience and the study of memory.
Neuron 20: 445– 468, 1998. 318. Navakkode S, Sajikumar S, Frey JU. The type IV-specific phosphodiesterase inhibitor
rolipram and its effect on hippocampal long-term potentiation and synaptic tagging. J
297. Minichiello L, Calella A, Medina D, Bonhoeffer T, Klein R, Korte M. Mechanism of Neurosci 24: 7740 –7744, 2004.
TrkB-mediated hippocampal long-term potentiation. Neuron 36: 121, 2002.
319. Nevian T, Larkum ME, Polsky A, Schiller J. Properties of basal dendrites of layer 5
298. Minichiello L, Korte M, Wolfer D, Kuhn R, Unsicker K, Cestari V, Rossi-Arnaud C, pyramidal neurons: a direct patch-clamp recording study. Nat Neurosci 10: 206 –214,
Lipp HP, Bonhoeffer T, Klein R. Essential role for TrkB receptors in hippocampus- 2007.
mediated learning. Neuron 24: 401– 414, 1999.
320. Nguyen PV, Abel T, Kandel ER. Requirement of a critical period of transcription for
299. Mironova YA, Giger RJ. Where no synapses go: gatekeepers of circuit remodeling and induction of a late phase of LTP. Science 265: 1104 –1107, 1994.
synaptic strength. Trends Neurosci 36: 363–373, 2013.
321. Nicoll RA, Schmitz D. Synaptic plasticity at hippocampal mossy fibre synapses. Nat Rev
300. Miya K, Inoue R, Takata Y, Abe M, Natsume R, Sakimura K, Hongou K, Miyawaki T, Neurosci 6: 863– 876, 2005.
Mori H. Serine racemase is predominantly localized in neurons in mouse brain. J Comp
322. Nishida M, Walker MP. Daytime naps, motor memory consolidation and regionally
Neurol 510: 641– 654, 2008.
specific sleep spindles. PloS One 2: e341, 2007.
301. Mizuno K, Dempster E, Mill J, Giese KP. Long-lasting regulation of hippocampal Bdnf
323. Noguchi J, Matsuzaki M, Ellis-Davies GC, Kasai H. Spine-neck geometry determines
gene transcription after contextual fear conditioning. Genes Brain Behav 11: 651– 659,
NMDA receptor-dependent Ca2⫹ signaling in dendrites. Neuron 46: 609 – 622, 2005.
2012.
324. Nosten-Bertrand M, Errington ML, Murphy KPSJ, Tokugawa Y, Barboni E, Kozlova E,
302. Mizuseki K, Royer S, Diba K, Buzsaki G. Activity dynamics and behavioral correlates of Michalovich D, Morris RGM, Silver J, Stewart CL, Bliss TVP, Morris RJ. Normal spatial
CA3 and CA1 hippocampal pyramidal neurons. Hippocampus 22: 1659 –1680, 2012. learning despite regional inhibition of LTP in mice lacking Thy-1. Nature 379: 826 –
829, 1996.
303. Molle M, Bergmann TO, Marshall L, Born J. Fast and slow spindles during the sleep
slow oscillation: disparate coalescence and engagement in memory processing. Sleep 325. O’Keefe J. Place units in the hippocampus of the freely moving rat. Exp Neurol 51:
34: 1411–1421, 2011. 78 –109, 1976.

304. Molle M, Eschenko O, Gais S, Sara SJ, Born J. The influence of learning on sleep slow 326. O’Keefe J, Dostrovsky J. The hippocampus as a spatial map. Preliminary evidence
oscillations and associated spindles and ripples in humans and rats. Eur J Neurosci 29: from unit activity in the freely-moving rat. Brain Res 34: 171–175, 1971.
1071–1081, 2009.
327. O’Keefe J, Nadel L. The Hippocampus as a Cognitive Map. Oxford: Clarendon, 1978.
305. Monory K, Polack M, Remus A, Lutz B, Korte M. Cannabinoid CB1 receptor calibrates
excitatory synaptic balance in the mouse hippocampus. J Neurosci 35: 3842–3850, 328. O’Keefe J, Recce ML. Phase relationship between hippocampal place units and the
2015. EEG theta rhythm. Hippocampus 3: 317–330, 1993.

306. Montgomery JM, Madison DV. Discrete synaptic states define a major mechanism of 329. O’Rourke NA, Weiler NC, Micheva KD, Smith SJ. Deep molecular diversity of mam-
synapse plasticity. Trends Neurosci 27: 744 –750, 2004. malian synapses: why it matters and how to measure it. Nature Rev Neurosci 13:
365–379, 2012.
307. Montgomery JM, Pavlidis P, Madison DV. Pair recordings reveal all-silent synaptic
connections and the postsynaptic expression of long-term potentiation. Neuron 29: 330. Okada D, Ozawa F, Inokuchi K. Input-specific spine entry of soma-derived Vesl-1S
691–701, 2001. protein conforms to synaptic tagging. Science 324: 904 –909, 2009.

331. Okamoto K, Nagai T, Miyawaki A, Hayashi Y. Rapid and persistent modulation of actin
308. Montgomery SM, Betancur MI, Buzsaki G. Behavior-dependent coordination of mul-
dynamics regulates postsynaptic reorganization underlying bidirectional plasticity. Nat
tiple theta dipoles in the hippocampus. J Neurosci 29: 1381–1394, 2009.
Neurosci 7: 1104 –1112, 2004.
309. Mukherjee S, Manahan-Vaughan D. Role of metabotropic glutamate receptors in
332. Panatier A, Theodosis DT, Mothet JP, Touquet B, Pollegioni L, Poulain DA, Oliet SH.
persistent forms of hippocampal plasticity and learning. Neuropharmacology 66: 65–
Glia-derived D-serine controls NMDA receptor activity and synaptic memory. Cell
81, 2013.
125: 775–784, 2006.
310. Mulkey RM, Malenka RC. Mechanisms underlying induction of homosynaptic long-
333. Pang PT, Teng HK, Zaitsev E, Woo NT, Sakata K, Zhen S, Teng KK, Yung WH,
term depression in area CA1 of the hippocampus. Neuron 9: 967–975, 1992.
Hempstead BL, Lu B. Cleavage of proBDNF by tPA/plasmin is essential for long-term
hippocampal plasticity. Science 306: 487– 491, 2004.
311. Murai KK, Nguyen LN, Irie F, Yamaguchi Y, Pasquale EB. Control of hippocampal
dendritic spine morphology through ephrin-A3/EphA4 signaling. Nat Neurosci 6: 153– 334. Pangalos M, Donoso JR, Winterer J, Zivkovic AR, Kempter R, Maier N, Schmitz D.
160, 2003. Recruitment of oriens-lacunosum-moleculare interneurons during hippocampal rip-
ples. Proc Natl Acad Sci USA 110: 4398 – 4403, 2013.
312. Nabavi S, Fox R, Proulx CD, Lin JY, Tsien RY, Malinow R. Engineering a memory with
LTD and LTP. Nature 511: 348 –352, 2014. 335. Pannasch U, Rouach N. Emerging role for astroglial networks in information process-
ing: from synapse to behavior. Trends Neurosci 36: 405– 417, 2013.
313. Naber PA, Witter MP. Subicular efferents are organized mostly as parallel projections:
a double-labeling, retrograde-tracing study in the rat. J Comp Neurol 393: 284 –297, 336. Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Giustetto M,
1998. Ferreira TA, Guiducci E, Dumas L, Ragozzino D, Gross CT. Synaptic pruning by
microglia is necessary for normal brain development. Science 333: 1456 –1458, 2011.
314. Nader K, Schafe GE, Le Doux JE. Fear memories require protein synthesis in the
amygdala for reconsolidation after retrieval. Nature 406: 722–726, 2000. 337. Park H, Poo MM. Neurotrophin regulation of neural circuit development and func-
tion. Nat Rev Neurosci 14: 7–23, 2012.
315. Nagerl UV, Kostinger G, Anderson JC, Martin KA, Bonhoeffer T. Protracted synap-
togenesis after activity-dependent spinogenesis in hippocampal neurons. J Neurosci 338. Park H, Poo MM. Neurotrophin regulation of neural circuit development and func-
27: 8149 – 8156, 2007. tion. Nature Rev Neurosci 14: 7–23, 2013.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 689


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

339. Parkhurst CN, Yang G, Ninan I, Savas JN, Yates JR 3rd, Lafaille JJ, Hempstead BL, through increased release probability, recruitment of new release sites, and activation
Littman DR, Gan WB. Microglia promote learning-dependent synapse formation of silent synapses. J Neurosci 24: 3618 –3626, 2004.
through brain-derived neurotrophic factor. Cell 155: 1596 –1609, 2013.
361. Remondes M, Schuman EM. Molecular mechanisms contributing to long-lasting syn-
340. Passafaro M, Piech V, Sheng M. Subunit-specific temporal and spatial patterns of aptic plasticity at the temporoammonic-CA1 synapse. Learn Mem 10: 247–252, 2003.
AMPA receptor exocytosis in hippocampal neurons. Nat Neurosci 4: 917–926, 2001.
362. Rex CS, Lin CY, Kramar EA, Chen LY, Gall CM, Lynch G. Brain-derived neurotrophic
341. Pastalkova E, Serrano P, Pinkhasova D, Wallace E, Fenton AA, Sacktor TC. Storage of factor promotes long-term potentiation-related cytoskeletal changes in adult hip-
spatial information by the maintenance mechanism of LTP. Science 313: 1141–1144, pocampus. J Neurosci 27: 3017–3029, 2007.
2006.
363. Richter JD. CPEB: a life in translation. Trends Biochem Sci 32: 279 –285, 2007.
342. Pasterkamp RJ, Giger RJ. Semaphorin function in neural plasticity and disease. Curr
Opin Neurobiol 19: 263–274, 2009. 364. Roberson ED, English JD, Adams JP, Selcher JC, Kondratick C, Sweatt JD. The mito-
gen-activated protein kinase cascade couples PKA and PKC to cAMP response ele-
343. Patterson SL, Abel T, Deuel TA, Martin KC, Rose JC, Kandel ER. Recombinant BDNF ment binding protein phosphorylation in area CA1 of hippocampus. J Neurosci 19:
rescues deficits in basal synaptic transmission and hippocampal LTP in BDNF knock- 4337– 4348, 1999.
out mice. Neuron 16: 1137–1145, 1996.
365. Rosahl TW, Spillane D, Missler M, Herz J, Selig DK, Wolff JR, Hammer RE, Malenka
344. Peng X, Kim J, Zhou Z, Fink DJ, Mata M. Neuronal Nogo-A regulates glutamate
RC, Südhof TC. Essential functions of synapsins I and II in synaptic vesicle regulation.
receptor subunit expression in hippocampal neurons. J Neurochem 119: 1183–1193,
Nature 375: 488 – 493, 1995.
2011.
366. Rösch H, Schweigreiter R, Barde YA, Bonhoeffer T, Korte M. The neurotrophin
345. Perea G, Araque A. Astrocytes potentiate transmitter release at single hippocampal
receptor p75NTR modulates long-term depression and regulates the expression of
synapses. Science 317: 1083–1086, 2007.
AMPA receptor subunits in the hippocampus. Proc Natl Acad Sci USA 102: 7362–7367,
346. Petrinovic MM, Hourez R, Aloy EM, Dewarrat G, Gall D, Weinmann O, Gaudias J, 2005.
Bachmann LC, Schiffmann SN, Vogt KE, Schwab ME. Neuronal Nogo-A negatively
367. Rosenmund C, Stern-Bach Y, Stevens CF. The tetrameric structure of a glutamate
regulates dendritic morphology and synaptic transmission in the cerebellum. Proc Natl
Acad Sci USA 110: 1083–1088, 2013. receptor channel. Science 280: 1596 –1599, 1998.

347. Peyrache A, Battaglia FP, Destexhe A. Inhibition recruitment in prefrontal cortex 368. Rosenzweig E, Ekstrom A, Redish A, McNaughton B, Barnes C.Phase precession as an
during sleep spindles and gating of hippocampal inputs. Proc Natl Acad Sci USA 108: experience-independent process: hippocampal pyramidal cell phase precession in a
17207–17212, 2011. novel environment and under NMDA-receptor blockade. Soc Neurosci Abstr 26: 982,
2000.
348. Piomelli D. The molecular logic of endocannabinoid signalling. Nat Rev Neurosci 4:
873– 884, 2003. 369. Rumpel S, LeDoux J, Zador A, Malinow R. Postsynaptic receptor trafficking underlying
a form of associative learning. Science 308: 83– 88, 2005.
349. Plath N, Ohana O, Dammermann B, Errington ML, Schmitz D, Gross C, Mao X,
Engelsberg A, Mahlke C, Welzl H, Kobalz U, Stawrakakis A, Fernandez E, Waltereit R, 370. Rutherford LC, Nelson SB, Turrigiano GG. BDNF has opposite effects on the quantal
Bick-Sander A, Therstappen E, Cooke SF, Blanquet V, Wurst W, Salmen B, Bosl MR, amplitude of pyramidal neuron and interneuron excitatory synapses. Neuron 21: 521–
Lipp HP, Grant SG, Bliss TV, Wolfer DP, Kuhl D. Arc/Arg3.1 is essential for the 530, 1998.
consolidation of synaptic plasticity and memories. Neuron 52: 437– 444, 2006.
371. Saab AS, Neumeyer A, Jahn HM, Cupido A, Simek AA, Boele HJ, Scheller A, Le Meur
350. Poo MM. Neurotrophins as synaptic modulators. Nat Rev Neurosci 2: 24 –32, 2001. K, Gotz M, Monyer H, Sprengel R, Rubio ME, Deitmer JW, De Zeeuw CI, Kirchhoff F.
Bergmann glial AMPA receptors are required for fine motor coordination. Science
351. Pozzo-Miller LD, Gottschalk W, Zhang L, McDermott K, Du J, Gopalakrishnan R, Oho 337: 749 –753, 2012.
C, Sheng ZH, Lu B. Impairments in high-frequency transmission, synaptic vesicle
docking, and synaptic protein distribution in the hippocampus of BDNF knockout 372. Sacktor TC. How does PKMzeta maintain long-term memory? Nat Rev Neurosci 12:
mice. J Neurosci 19: 4972– 4983, 1999. 9 –15, 2011.

352. Prehn-Kristensen A, Goder R, Chirobeja S, Bressmann I, Ferstl R, Baving L. Sleep in 373. Sacktor TC. PKMzeta, LTP maintenance, and the dynamic molecular biology of mem-
children enhances preferentially emotional declarative but not procedural memories. ory storage. Prog Brain Res 169: 27– 40, 2008.
J Exp Child Psychol 104: 132–139, 2009.
374. Sajikumar S, Korte M. Different compartments of apical CA1 dendrites have different
353. Raiker SJ, Lee H, Baldwin KT, Duan Y, Shrager P, Giger RJ. Oligodendrocyte-myelin plasticity thresholds for expressing synaptic tagging and capture. Learn Mem 18: 327–
glycoprotein and Nogo negatively regulate activity-dependent synaptic plasticity. J 331, 2011.
Neurosci 30: 12432–12445, 2010.
375. Sajikumar S, Korte M. Metaplasticity governs compartmentalization of synaptic tag-
354. Ramachandran B, Frey JU. Interfering with the actin network and its effect on long- ging and capture through brain-derived neurotrophic factor (BDNF) and protein
term potentiation and synaptic tagging in hippocampal CA1 neurons in slices in vitro. kinase M␨ (PKM␨). Proc Natl Acad Sci USA 108: 2551–2556, 2011.
J Neurosci 29: 12167–12173, 2009.
376. Sajikumar S, Morris RG, Korte M. Competition between recently potentiated synaptic
355. Ramirez S, Liu X, Lin PA, Suh J, Pignatelli M, Redondo RL, Ryan TJ, Tonegawa S.
inputs reveals a winner-take-all phase of synaptic tagging and capture. Proc Natl Acad
Creating a false memory in the hippocampus. Science 341: 387–391, 2013.
Sci USA 111: 12217–12221, 2014.
356. Rao RP, Sejnowski TJ. Spike-timing-dependent Hebbian plasticity as temporal differ-
377. Sajikumar S, Navakkode S, Sacktor TC, Frey JU. Synaptic tagging and cross-tagging:
ence learning. Neural Comput 13: 2221–2237, 2001.
the role of protein kinase Mzeta in maintaining long-term potentiation but not long-
357. Rasch B, Born J. About sleep’s role in memory. Physiol Rev 93: 681–766, 2013. term depression. J Neurosci 25: 5750 –5756, 2005.

358. Redish AD. Beyond the Cognitive Map: From Place Cells to Episodic Memory. Cambridge, 378. Sajikumar SE. Synaptic Tagging and Capture: From Synapses to Behavior. New York:
MA: MIT Press, 1999. Springer, 2015.

359. Redish AD, Battaglia FP, Chawla MK, Ekstrom AD, Gerrard JL, Lipa P, Rosenzweig ES, 379. Sala C, Segal M. Dendritic spines: the locus of structural and functional plasticity.
Worley PF, Guzowski JF, McNaughton BL, Barnes CA. Independence of firing corre- Physiol Rev 94: 141–188, 2014.
lates of anatomically proximate hippocampal pyramidal cells. J Neurosci 21: RC134,
2001. 380. Schapitz IU, Behrend B, Pechmann Y, Lappe-Siefke C, Kneussel SJ, Wallace KE,
Stempel AV, Buck F, Grant SG, Schweizer M, Schmitz D, Schwarz JR, Holzbaur EL,
360. Reid CA, Dixon DB, Takahashi M, Bliss TV, Fine A. Optical quantal analysis indicates Kneussel M. Neuroligin 1 is dynamically exchanged at postsynaptic sites. J Neurosci 30:
that long-term potentiation at single hippocampal mossy fiber synapses is expressed 12733–12744, 2010.

690 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

381. Schmitz D, Schuchmann S, Fisahn A, Draguhn A, Buhl EH, Petrasch-Parwez E, Der- 405. Snaidero N, Mobius W, Czopka T, Hekking LH, Mathisen C, Verkleij D, Goebbels S,
mietzel R, Heinemann U, Traub RD. Axo-axonal coupling, a novel mechanism for Edgar J, Merkler D, Lyons DA, Nave KA, Simons M. Myelin membrane wrapping of
ultrafast neuronal communication. Neuron 31: 831– 840, 2001. CNS axons by PI(3,4,5)P3-dependent polarized growth at the inner tongue. Cell 156:
277–290, 2014.
382. Schnell E, Sizemore M, Karimzadegan S, Chen L, Bredt DS, Nicoll RA. Direct inter-
actions between PSD-95 and stargazin control synaptic AMPA receptor number. Proc 406. Soderling TR, Derkach VA. Postsynaptic protein phosphorylation and LTP. Trends
Natl Acad Sci USA 99: 13902–13907, 2002. Neurosci 23: 75– 80, 2000.

383. Schratt G. microRNAs at the synapse. Nature Rev Neurosci 10: 842– 849, 2009. 407. Soltesz I, Deschenes M. Low- and high-frequency membrane potential oscillations
during theta activity in CA1 and CA3 pyramidal neurons of the rat hippocampus under
384. Schratt GM, Nigh EA, Chen WG, Hu L, Greenberg ME. BDNF regulates the transla- ketamine-xylazine anesthesia. J Neurophysiol 70: 97–116, 1993.
tion of a select group of mRNAs by a mammalian target of rapamycin-phosphatidyl-
inositol 3-kinase-dependent pathway during neuronal development. J Neurosci 24: 408. Somogyi P, Klausberger T. Defined types of cortical interneurone structure space and
7366 –7377, 2004. spike timing in the hippocampus. J Physiol 562: 9 –26, 2005.

385. Schwab ME. Functions of Nogo proteins and their receptors in the nervous system. 409. Song I, Huganir RL. Regulation of AMPA receptors during synaptic plasticity. Trends
Nat Rev Neurosci 11: 799 – 811, 2010. Neurosci 25: 578 –588, 2002.

386. Schwab ME, Strittmatter SM. Nogo limits neural plasticity and recovery from injury. 410. Song S, Abbott LF. Cortical development and remapping through spike timing-depen-
Curr Opin Neurobiol 27C: 53– 60, 2014. dent plasticity. Neuron 32: 339 –350, 2001.

387. Scoville WB, Milner B. Loss of recent memory after bilateral hippocampal lesions. J 411. Spruston N. Pyramidal neurons: dendritic structure and synaptic integration. Nature
Neurol Neurosurg Psychiatry 20: 11–21, 1957. Rev Neurosci 9: 206 –221, 2008.

388. Segal M, Korkotian E. Endoplasmic reticulum calcium stores in dendritic spines. Front 412. Squire LR. Memory systems of the brain: a brief history and current perspective.
Neuroanat 8: 64, 2014. Neurobiol Learn Mem 82: 171–177, 2004.

389. Sejnowski TJ, Destexhe A. Why do we sleep? Brain Res 886: 208 –223, 2000. 413. Staubli U, Vanderklish P, Lynch G. An inhibitor of integrin receptors blocks long-term
potentiation. Behav Neural Biol 53: 1–5, 1990.
390. Senkov O, Sun M, Weinhold B, Gerardy-Schahn R, Schachner M, Dityatev A. Polysia-
lylated neural cell adhesion molecule is involved in induction of long-term potentiation 414. Stellwagen D, Malenka RC. Synaptic scaling mediated by glial TNF-alpha. Nature 440:
and memory acquisition and consolidation in a fear-conditioning paradigm. J Neurosci 1054 –1059, 2006.
26: 10888 –109898, 2006.
415. Stephens GJ. G-protein-coupled-receptor-mediated presynaptic inhibition in the cer-
391. Shen J, Barnes CA, McNaughton BL, Skaggs WE, Weaver KL. The effect of aging on ebellum. Trends Pharmacol Sci 30: 421– 430, 2009.
experience-dependent plasticity of hippocampal place cells. J Neurosci 17: 6769 –
416. Steward O, Levy WB. Preferential localization of polyribosomes under the base of
6782, 1997.
dendritic spines in granule cells of the dentate gyrus. J Neurosci 2: 284 –291, 1982.
392. Shen K, Cowan CW. Guidance molecules in synapse formation and plasticity. Cold
417. Steward O, Schuman EM. Compartmentalized synthesis and degradation of proteins
Spring Harbor Perspect Biol 2: a001842, 2010. in neurons. Neuron 40: 347–359, 2003.
393. Sheng M, Kim E. The postsynaptic organization of synapses. Cold Spring Harb Perspect 418. Steward O, Schuman EM. Protein synthesis at synaptic sites on dendrites. Annu Rev
Biol 3: 1–20, 2011. Neurosci 24: 299 –325, 2001.
394. Shepherd JD, Rumbaugh G, Wu J, Chowdhury S, Plath N, Kuhl D, Huganir RL, Worley 419. Steward O, Wallace CS, Lyford GL, Worley PF. Synaptic activation causes the mRNA
PF. Arc/Arg3.1 mediates homeostatic synaptic scaling of AMPA receptors. Neuron 52: for the IEG Arc to localize selectively near activated postsynaptic sites on dendrites.
475– 484, 2006. Neuron 21: 741–751, 1998.
395. Shi S, Hayashi Y, Esteban JA, Malinow R. Subunit-specific rules governing AMPA 420. Steward O, Worley PF. Selective targeting of newly synthesized Arc mRNA to active
receptor trafficking to synapses in hippocampal pyramidal neurons. Cell 105: 331– synapses requires NMDA receptor activation. Neuron 30: 227–240, 2001.
343, 2001.
421. Stock JB, Zhang S. The biochemistry of memory. Curr Biol 23: R741–745, 2013.
396. Shouval HZ, Wang SS, Wittenberg GM. Spike timing dependent plasticity: a conse-
quence of more fundamental learning rules. Front Computat Neurosci 4: 2010. 422. Sullivan D, Mizuseki K, Sorgi A, Buzsaki G. Comparison of sleep spindles and theta
oscillations in the hippocampus. J Neurosci 34: 662– 674, 2014.
397. Siapas AG, Wilson MA. Coordinated interactions between hippocampal ripples and
cortical spindles during slow-wave sleep. Neuron 21: 1123–1128, 1998. 423. Suzuki WA, Miller EK, Desimone R. Object and place memory in the macaque ento-
rhinal cortex. J Neurophysiol 78: 1062–1081, 1997.
398. Sigrist SJ, Schmitz D. Structural and functional plasticity of the cytoplasmic active zone.
Curr Opin Neurobiol 21: 144 –150, 2011. 424. Tai CY, Kim SA, Schuman EM. Cadherins and synaptic plasticity. Curr Opin Cell Biol 20:
567–575, 2008.
399. Sjostrom PJ, Hausser M. A cooperative switch determines the sign of synaptic plas-
ticity in distal dendrites of neocortical pyramidal neurons. Neuron 51: 227–238, 2006. 425. Takeichi M. Morphogenetic roles of classic cadherins. Curr Opin Cell Biol 7: 619 – 627,
1995.
400. Sjostrom PJ, Rancz EA, Roth A, Hausser M. Dendritic excitability and synaptic plas-
ticity. Physiol Rev 88: 769 – 840, 2008. 426. Takeuchi T, Duszkiewicz AJ, Morris RG. The synaptic plasticity and memory hypoth-
esis: encoding, storage and persistence. Philos Trans R Soc Lond B Biol Sci 369:
401. Skaggs WE, McNaughton BL. Replay of neuronal firing sequences in rat hippocampus 20130288, 2014.
during sleep following spatial experience. Science 271: 1870 –1873, 1996.
427. Tanaka J, Horiike Y, Matsuzaki M, Miyazaki T, Ellis-Davies GC, Kasai H. Protein
402. Skaggs WE, McNaughton BL, Wilson MA, Barnes CA. Theta phase precession in synthesis and neurotrophin-dependent structural plasticity of single dendritic spines.
hippocampal neuronal populations and the compression of temporal sequences. Hip- Science 319: 1683–1687, 2008.
pocampus 6: 149 –172, 1996.
428. Tang L, Hung CP, Schuman EM. A role for the cadherin family of cell adhesion
403. Slanina KA, Roberto M, Schweitzer P. Endocannabinoids restrict hippocampal long- molecules in hippocampal long-term potentiation. Neuron 20: 1165–1175, 1998.
term potentiation via CB1. Neuropharmacology 49: 660 – 668, 2005.
429. Tass P, Rosenblum M, Weule J, Kurths J, Pikovsky A, Volkmann J, Schnitzler A, Freund
404. Smith C, Rose GM. Evidence for a paradoxical sleep window for place learning in the HJ. Detection of n: m phase locking from noisy data: application to magnetoencepha-
Morris water maze. Physiol Behav 59: 93–97, 1996. lography. Phys Rev Lett 81: 3291, 1998.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 691


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
BIOLOGY OF MEMORY EVENTS

430. Terrier G, Gottesmann CL. Study of cortical spindles during sleep in the rat. Brain Res 453. Voronin LL, Cherubini E. “Deaf, mute and whispering” silent synapses: their role in
Bull 3: 701–706, 1978. synaptic plasticity. J Physiol 557: 3–12, 2004.

431. Thoenen H. The changing scene of neurotrophic factors. Trends Neurosci 14: 165– 454. Walker MP, Brakefield T, Morgan A, Hobson JA, Stickgold R. Practice with sleep
170, 1991. makes perfect: sleep-dependent motor skill learning. Neuron 35: 205–211, 2002.

432. Thoenen H. Neurotrophins and neuronal plasticity. Science 270: 593–598, 1995. 455. Walker MP, Brakefield T, Seidman J, Morgan A, Hobson JA, Stickgold R. Sleep and the
time course of motor skill learning. Learn Mem 10: 275–284, 2003.
433. Tokuyama W, Okuno H, Hashimoto T, Xin LY, Miyashita Y. BDNF upregulation
during declarative memory formation in monkey inferior temporal cortex. Nat Neu- 456. Wallace CS, Lyford GL, Worley PF, Steward O. Differential intracellular sorting of
rosci 3: 1134 –1142, 2000. immediate early gene mRNAs depends on signals in the mRNA sequence. J Neurosci
18: 26 –35, 1998.
434. Tomita S, Chen L, Kawasaki Y, Petralia RS, Wenthold RJ, Nicoll RA, Bredt DS. Func-
tional studies and distribution define a family of transmembrane AMPA receptor 457. Wang DO, Kim SM, Zhao Y, Hwang H, Miura SK, Sossin WS, Martin KC. Synapse- and
regulatory proteins. J Cell Biol 161: 805– 816, 2003. stimulus-specific local translation during long-term neuronal plasticity. Science 324:
1536 –1540, 2009.
435. Tomita S, Stein V, Stocker TJ, Nicoll RA, Bredt DS. Bidirectional synaptic plasticity
regulated by phosphorylation of stargazin-like TARPs. Neuron 45: 269 –277, 2005. 458. Waung MW, Pfeiffer BE, Nosyreva ED, Ronesi JA, Huber KM. Rapid translation of
Arc/Arg3.1 selectively mediates mGluR-dependent LTD through persistent increases
436. Tong G, Malenka RC, Nicoll RA. Long-term potentiation in cultures of single hip- in AMPAR endocytosis rate. Neuron 59: 84 –97, 2008.
pocampal granule cells: a presynaptic form of plasticity. Neuron 16: 1147–1157, 1996.
459. Wells DG, Dong X, Quinlan EM, Huang YS, Bear MF, Richter JD, Fallon JR. A role for
437. Toni N, Buchs PA, Nikonenko I, Bron CR, Muller D. LTP promotes formation of the cytoplasmic polyadenylation element in NMDA receptor-regulated mRNA trans-
multiple spine synapses between a single axon terminal and a dendrite. Nature 402: lation in neurons. J Neurosci 21: 9541–9548, 2001.
421– 425, 1999.
460. Wenthold RJ, Petralia RS, Blahos J, II, Niedzielski AS. Evidence for multiple AMPA
438. Tonnesen J, Katona G, Rozsa B, Nagerl UV. Spine neck plasticity regulates compart- receptor complexes in hippocampal CA1/CA2 neurons. J Neurosci 16: 1982–1989,
mentalization of synapses. Nat Neurosci 17: 678 – 685, 2014. 1996.

439. Tort AB, Komorowski RW, Manns JR, Kopell NJ, Eichenbaum H. Theta-gamma cou- 461. Wierzynski CM, Lubenov EV, Gu M, Siapas AG. State-dependent spike-timing rela-
pling increases during the learning of item-context associations. Proc Natl Acad Sci USA tionships between hippocampal and prefrontal circuits during sleep. Neuron 61: 587–
106: 20942–20947, 2009. 596, 2009.

440. Tort AB, Kramer MA, Thorn C, Gibson DJ, Kubota Y, Graybiel AM, Kopell NJ. 462. Wigström H, Gustafsson B. Facilitated induction of hippocampal long-lasting potenti-
Dynamic cross-frequency couplings of local field potential oscillations in rat striatum ation during blockade of inhibition. Nature 301: 603– 604, 1983.
and hippocampus during performance of a T-maze task. Proc Natl Acad Sci USA 105:
20517–20522, 2008. 463. Wigström H, Gustafsson B, Huang YY, Abraham WC. Hippocampal long-term poten-
tiation is induced by pairing single afferent volleys with intracellularly injected depo-
441. Traub RD, Bibbig A. A model of high-frequency ripples in the hippocampus based on larizing current pulses. Acta Physiol Scand 126: 317–319, 1986.
synaptic coupling plus axon-axon gap junctions between pyramidal neurons. J Neurosci
20: 2086 –2093, 2000. 464. Wilson MA, McNaughton BL. Reactivation of hippocampal ensemble memories dur-
ing sleep. Science 265: 676 – 679, 1994.
442. Tse D, Langston RF, Kakeyama M, Bethus I, Spooner PA, Wood ER, Witter MP,
Morris RG. Schemas and memory consolidation. Science 316: 76 – 82, 2007. 465. Wilson RI, Nicoll RA. Endocannabinoid signaling in the brain. Science 296: 678 – 682,
2002.
443. Turrigiano G. Homeostatic synaptic plasticity: local and global mechanisms for stabi-
lizing neuronal function. Cold Spring Harbor Perspect Biol 4: a005736, 2012. 466. Winocur G, Moscovitch M, Bontempi B. Memory formation and long-term retention
in humans and animals: convergence towards a transformation account of hippocam-
444. Turrigiano G. Too many cooks? Intrinsic and synaptic homeostatic mechanisms in pal-neocortical interactions. Neuropsychologia 48: 2339 –2356, 2010.
cortical circuit refinement. Annu Rev Neurosci 34: 89 –103, 2011.
467. Witcher MR, Kirov SA, Harris KM. Plasticity of perisynaptic astroglia during synapto-
445. Turrigiano GG. The self-tuning neuron: synaptic scaling of excitatory synapses. Cell genesis in the mature rat hippocampus. Glia 55: 13–23, 2007.
135: 422– 435, 2008.
468. Witte AV, Kurten J, Jansen S, Schirmacher A, Brand E, Sommer J, Floel A. Interaction
446. Turrigiano GG, Leslie KR, Desai NS, Rutherford LC, Nelson SB. Activity-dependent of BDNF and COMT polymorphisms on paired-associative stimulation-induced cor-
scaling of quantal amplitude in neocortical neurons. Nature 391: 892– 896, 1998. tical plasticity. J Neurosci 32: 4553– 4561, 2012.

447. Tzounopoulos T, Rubio ME, Keen JE, Trussell LO. Coactivation of pre- and postsyn- 469. Wolters A, Schmidt A, Schramm A, Zeller D, Naumann M, Kunesch E, Benecke R,
aptic signaling mechanisms determines cell-specific spike-timing-dependent plasticity. Reiners K, Classen J. Timing-dependent plasticity in human primary somatosensory
Neuron 54: 291–301, 2007. cortex. J Physiol 565: 1039 –1052, 2005.

448. Van der Helm E, Yao J, Dutt S, Rao V, Saletin JM, Walker MP. REM sleep depotentiates 470. Woo NH, Teng HK, Siao CJ, Chiaruttini C, Pang PT, Milner TA, Hempstead BL, Lu B.
amygdala activity to previous emotional experiences. Curr Biol 21: 2029 –2032, 2011. Activation of p75(NTR) by proBDNF facilitates hippocampal long-term depression.
Nat Neurosci 8: 1069 –1077, 2005.
449. Varga C, Golshani P, Soltesz I. Frequency-invariant temporal ordering of interneuro-
nal discharges during hippocampal oscillations in awake mice. Proc Natl Acad Sci USA 471. Wu CP, Huang HL, Asl MN, He JW, Gillis J, Skinner FK, Zhang L. Spontaneous
109: E2726 –2734, 2012. rhythmic field potentials of isolated mouse hippocampal-subicular-entorhinal cortices
in vitro. J Physiol 576: 457– 476, 2006.
450. Vaughn DE, Bjorkman PJ. The (Greek) key to structures of neural adhesion molecules.
Neuron 16: 261–273, 1996. 472. Wu L, Wells D, Tay J, Mendis D, Abbott MA, Barnitt A, Quinlan E, Heynen A, Fallon
JR, Richter JD. CPEB-mediated cytoplasmic polyadenylation and the regulation of
451. Volk LJ, Bachman JL, Johnson R, Yu Y, Huganir RL. PKM-zeta is not required for experience-dependent translation of alpha-CaMKII mRNA at synapses. Neuron 21:
hippocampal synaptic plasticity, learning and memory. Nature 493: 420 – 423, 2013. 1129 –1139, 1998.

452. Voronin LL, Altinbaev RS, Bayazitov IT, Gasparini S, Kasyanov AV, Saviane C, Savtch- 473. Xu B, Gottschalk W, Chow A, Wilson RI, Schnell E, Zang K, Wang D, Nicoll RA, Lu B,
enko L, Cherubini E. Postsynaptic depolarisation enhances transmitter release and Reichardt LF. The role of brain-derived neurotrophic factor receptors in the mature
causes the appearance of responses at “silent” synapses in rat hippocampus. Neuro- hippocampus: modulation of long-term potentiation through a presynaptic mecha-
science 126: 45–59, 2004. nism involving TrkB. J Neurosci 20: 6888 – 6897, 2000.

692 Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.
MARTIN KORTE AND DIETMAR SCHMITZ

474. Yamagata M, Sanes JR, Weiner JA. Synaptic adhesion molecules. Curr Opin Cell Biol 15: 485. Yuste R. Electrical compartmentalization in dendritic spines. Annu Rev Neurosci 36:
621– 632, 2003. 429 – 449, 2013.

475. Yamaguchi Y, Aota Y, McNaughton BL, Lipa P. Bimodality of theta phase precession in 486. Yuste R, Bonhoeffer T. Genesis of dendritic spines: insights from ultrastructural and
hippocampal place cells in freely running rats. J Neurophysiol 87: 2629 –2642, 2002. imaging studies. Nat Rev Neurosci 5: 24 –34, 2004.

476. Yang G, Lai CS, Cichon J, Ma L, Li W, Gan WB. Sleep promotes branch-specific 487. Yuste R, Bonhoeffer T. Morphological changes in dendritic spines associated with
formation of dendritic spines after learning. Science 344: 1173–1178, 2014. long-term synaptic plasticity. Annu Rev Neurosci 24: 1071–1089, 2001.

477. Yang G, Pan F, Gan WB. Stably maintained dendritic spines are associated with lifelong 488. Zagrebelsky M, Korte M. Form follows function: BDNF and its involvement in
memories. Nature 462: 920 –924, 2009. sculpting the function and structure of synapses. Neuropharmacology 76: 628 –
638, 2014.
478. Yang X, Hou D, Jiang W, Zhang C. Intercellular protein-protein interactions at syn-
apses. Protein Cell 5: 420 – 444, 2014. 489. Zagrebelsky M, Korte M. Maintaining stable memory engrams: new roles for
Nogo-A in the CNS. Neuroscience 283: 17–25, 2014.
479. Yang Y, Ge W, Chen Y, Zhang Z, Shen W, Wu C, Poo M, Duan S. Contribution of
astrocytes to hippocampal long-term potentiation through release of D-serine. Proc 490. Zagrebelsky M, Schweigreiter R, Bandtlow CE, Schwab ME, Korte M. Nogo-A
Natl Acad Sci USA 100: 15194 –15199, 2003. stabilizes the architecture of hippocampal neurons. J Neurosci 30: 13220 –13234,
2010.
480. Yao H, Dan Y. Stimulus timing-dependent plasticity in cortical processing of orienta-
tion. Neuron 32: 315–323, 2001. 491. Zakharenko SS, Patterson SL, Dragatsis I, Zeitlin SO, Siegelbaum SA, Kandel ER,
Morozov A. Presynaptic BDNF required for a presynaptic but not postsynaptic com-
481. Ying SW, Futter M, Rosenblum K, Webber MJ, Hunt SP, Bliss TV, Bramham CR. ponent of LTP at hippocampal CA1-CA3 synapses. Neuron 39: 975–990, 2003.
Brain-derived neurotrophic factor induces long-term potentiation in intact adult hip-
pocampus: requirement for ERK activation coupled to CREB and upregulation of Arc 492. Zhao H, Pykalainen A, Lappalainen P. I-BAR domain proteins: linking actin and plasma
synthesis. J Neurosci 22: 1532–1540, 2002. membrane dynamics. Curr Opin Cell Biol 23: 14 –21, 2011.

482. Ylinen A, Soltesz I, Bragin A, Penttonen M, Sik A, Buzsaki G. Intracellular correlates of 493. Zhou Q, Homma KJ, Poo MM. Shrinkage of dendritic spines associated with long-
hippocampal theta rhythm in identified pyramidal cells, granule cells, and basket cells. term depression of hippocampal synapses. Neuron 44: 749 –757, 2004.
Hippocampus 5: 78 –90, 1995.
494. Zhu PJ, Lovinger DM. Persistent synaptic activity produces long-lasting enhancement
483. Yun SH, Mook-Jung I, Jung MW. Variation in effective stimulus patterns for induction of endocannabinoid modulation and alters long-term synaptic plasticity. J Neurophysiol
of long-term potentiation across different layers of rat entorhinal cortex. J Neurosci 97: 4386 – 4389, 2007.
22: RC214, 2002.
495. Zito K, Scheuss V, Knott G, Hill T, Svoboda K. Rapid functional maturation of nascent
484. Yuste R. Dendritic spines and distributed circuits. Neuron 71: 772–781, 2011. dendritic spines. Neuron 61: 247–258, 2009.

Physiol Rev • VOL 96 • APRIL 2016 • www.prv.org 693


Downloaded from www.physiology.org/journal/physrev (179.006.223.112) on February 19, 2019.

Você também pode gostar