ArticlePDF Available

The Emerging Role of Mechanics in Synapse Formation and Plasticity

Authors:

Abstract

The regulation of synaptic strength forms the basis of learning and memory, and is a key factor in understanding neuropathological processes that lead to cognitive decline and dementia. While the mechanical aspects of neuronal development, particularly during axon growth and guidance, have been extensively studied, relatively little is known about the mechanical aspects of synapse formation and plasticity. It is established that a filamentous actin network with complex spatiotemporal behavior controls the dendritic spine shape and size, which is thought to be crucial for activity-dependent synapse plasticity. Accordingly, a number of actin binding proteins have been identified as regulators of synapse plasticity. On the other hand, a number of cell adhesion molecules (CAMs) are found in synapses, some of which form transsynaptic bonds to align the presynaptic active zone (PAZ) with the postsynaptic density (PSD). Considering that these CAMs are key components of cellular mechanotransduction, two critical questions emerge: (i) are synapses mechanically regulated? and (ii) does disrupting the transsynaptic force balance lead to (or exacerbate) synaptic failure? In this mini review article, I will highlight the mechanical aspects of synaptic structures—focusing mainly on cytoskeletal dynamics and CAMs—and discuss potential mechanoregulation of synapses and its relevance to neurodegenerative diseases.
MINI REVIEW
published: 06 December 2018
doi: 10.3389/fncel.2018.00483
The Emerging Role of Mechanics in
Synapse Formation and Plasticity
Devrim Kilinc*
INSERM U1167, Institut Pasteur de Lille, Lille, France
Edited by:
Kyle Miller,
Michigan State University,
United States
Reviewed by:
Olivier Thoumine,
Centre National de la Recherche
Scientifique (CNRS), France
Brenton Hoffman,
Duke University, United States
*Correspondence:
Devrim Kilinc
devrim.kilinc@pasteur-lille.fr
Received: 01 September 2018
Accepted: 27 November 2018
Published: 06 December 2018
Citation:
Kilinc D (2018) The Emerging Role of
Mechanics in Synapse Formation
and Plasticity.
Front. Cell. Neurosci. 12:483.
doi: 10.3389/fncel.2018.00483
The regulation of synaptic strength forms the basis of learning and memory, and is a key
factor in understanding neuropathological processes that lead to cognitive decline and
dementia. While the mechanical aspects of neuronal development, particularly during
axon growth and guidance, have been extensively studied, relatively little is known
about the mechanical aspects of synapse formation and plasticity. It is established
that a filamentous actin network with complex spatiotemporal behavior controls the
dendritic spine shape and size, which is thought to be crucial for activity-dependent
synapse plasticity. Accordingly, a number of actin binding proteins have been identified
as regulators of synapse plasticity. On the other hand, a number of cell adhesion
molecules (CAMs) are found in synapses, some of which form transsynaptic bonds to
align the presynaptic active zone (PAZ) with the postsynaptic density (PSD). Considering
that these CAMs are key components of cellular mechanotransduction, two critical
questions emerge: (i) are synapses mechanically regulated? and (ii) does disrupting
the transsynaptic force balance lead to (or exacerbate) synaptic failure? In this mini
review article, I will highlight the mechanical aspects of synaptic structures—focusing
mainly on cytoskeletal dynamics and CAMs—and discuss potential mechanoregulation
of synapses and its relevance to neurodegenerative diseases.
Keywords: dendritic spine, cytoskeleton, cell adhesion molecules, motor proteins, mechanotransduction, synaptic
scaffold proteins
INTRODUCTION
Chemical synapses of the central nervous system (CNS) mediate the directional information flow
between neurons and form the basis of learning and memory. A precisely-defined synaptic cleft
separates the opposing pre- and postsynaptic terminals that are held in place via transsynaptic
cell adhesion molecules (CAMs). While presynaptic terminals are specialized in neurotransmitter
release, postsynaptic terminals house neurotransmitter receptors and various signaling and
scaffolding proteins. Functional diversity of synapses is reflected in structural diversity: most
inhibitory synapses form directly on the dendrite shaft, while most excitatory synapses form
on dendritic spines—morphologically diverse membrane protrusions from the dendrite shaft.
Dendritic spines may form enlarged heads with relatively narrow necks, resulting in signaling hubs
with restricted electrical and chemical connection to the dendrite shaft. The postsynaptic density
(PSD), a dense protein matrix beneath the postsynaptic membrane, forms at the tip of the spine
Abbreviations: ABP, actin-binding protein; AD, Alzheimer’s disease; AMPAR, α-amino-3-hydroxy-5-methyl-4-
isoxazolepropionic acid receptor; APP, amyloid precursor protein; CAM, cell adhesion molecule; CaMKII, Ca2+/calmodulin-
dependent protein kinase; CNS, central nervous system; D1R, dopamine D1 receptor; ECM, extracellular matrix; F-actin,
filamentous actin; IgCAM, immunoglobulin superfamily cell adhesion molecule; LDP, long-term depression; LTP, long-term
potentiation; MMP, matrix metalloproteinase; NCAM, neural cell adhesion molecule; NMDAR, N-methyl-D-aspartate
receptor; PAZ, presynaptic active zone; PSD, postsynaptic density.
Frontiers in Cellular Neuroscience | www.frontiersin.org 1December 2018 | Volume 12 | Article 483
Kilinc Mechanical Regulation of Synapses
head and orchestrates synaptic functions (Sheng and
Kim, 2011). The size and shape of individual spines are
regulated in an activity-dependent fashion, supported by
a specialized protein synthesis and degradation system
(Alvarez-Castelao and Schuman, 2015). Synapses may last
from seconds to decades; thus, highly sophisticated regulatory
mechanisms are required to effectively control their dynamics,
during development and adulthood.
The role of mechanics in neurodevelopment is best
characterized at the growth cones (Franze, 2013), highly
motile tips of growing axons that integrate mechanical and
chemical cues during axon pathfinding (Kerstein et al.,
2015). Much less is known about the mechanical aspects of
synapse formation and plasticity. This is—partly—due to
the increased complexity of mature neurons compared to
developing axons. Nevertheless, recent studies established that
synapse formation and plasticity require unique mechanisms
involving the cytoskeleton, molecular motors, CAMs and the
extracellular matrix (ECM; Figure 1). Considering that these
components are either force-generating or force-bearing, two
critical questions emerge: (i) are synapses mechan(ochem)ically
regulated? and (ii) does disrupting the transsynaptic force
balance lead to (or exacerbate) synaptic failure? In this mini
review article, I will highlight the mechanical aspects of synaptic
structures—focusing mainly on cytoskeletal dynamics and
CAMs—and discuss potential mechanoregulation of synapses
and its relevance to neurodegenerative diseases.
DYNAMIC CYTOSKELETAL
INTERACTIONS SHAPE SYNAPSES
The cytoskeleton is an interconnected network of dynamic
filaments and regulatory proteins mediating not only the
mechanical processes such as shape change and cell motility,
but also the global intracellular organization. This is achieved
through combining stable, long-range interactions and highly
dynamic, short range interactions (Fletcher and Mullins, 2010).
Synapses rely on cytoskeletal processes to accomplish specific
tasks, from activity-dependent structural change to long-term
maintenance of established connections. I will discuss actin,
microtubule and neurofilament networks separately, despite the
tight coupling between them (Coles and Bradke, 2015).
Dynamic Actin Networks Control Dendritic
Spine Shape and Size
Dendritic spines are structurally supported by a filamentous
actin (F-actin) framework, which controls the spine shape and
organizes the signaling machinery (reviewed in Hotulainen
and Hoogenraad, 2010). The F-actin retrograde flow in spines,
i.e., from tip to base, is reminiscent of the same in developing
axons (Nichol et al., 2016), albeit with shorter filaments and
lower flow rates (50 nm/s; Frost et al., 2010). However,
this resemblance is disputed by reports showing that the
F-actin flow slows down (from 35 nm/s to 20 nm/s) as
dendritic filopodia turn into spines (Chazeau et al., 2014),
and that the polarization is lost (Tatavarty et al., 2012).
Long-term potentiation (LTP) and depression (LTD) induce
FIGURE 1 | Mechanically-relevant components of an excitatory synapse.
Presynaptic vesicle fusion machinery and postsynaptic receptors are held in
place by their respective scaffold proteins, which are physically linked to cell
adhesion molecules (CAMs) and the cytoskeleton. Direct interactions are
indicated with thin, continuous arrows. Translational movements are indicated
with thin, broken arrows. Only postsynaptic cytoskeleton is depicted for
simplicity. Major cytoskeletal filaments (actin, neurofilaments, microtubules)
and a select set of associated molecules (microtubule end-binding protein
EB3, actin severing/stabilizing protein cofilin, actin branch-inducing
Arp2/3 complex) are depicted. Spine base and center are occupied by stable,
dense F-actin, whereas the periphery is occupied by dynamic, branched
F-actin. F-actin rings line the spine shaft. Microtubules occasionally invade
spines and interact with the postsynaptic density (PSD), but the role of
neurofilaments is not clear. Actin and microtubule polymerization creates
tensile forces (green arrows) favoring the expansion of the spine head. Myosin
motors pull on actin filaments to generate actomyosin contractile forces (red
arrows) favoring the shrinkage of the spine head. Kinesin and dynein motors
transport cargo on microtubules anterograde and retrograde, respectively. The
latter also interacts with the PSD. A select set of CAMs are depicted, most of
which form transsynaptic homophilic bonds. N-cadherin and SynCAM bonds
encircle the presynaptic active zone (PAZ) and the PSD. β-catenin links
N-cadherin to the F-actin cytoskeleton directly or via α-catenin and vinculin.
This linkage pulls on transsynaptic CAM bonds, which potentially induce
signaling in pre- and postsynaptic compartments, i.e., mechanotransduction.
Powered by actomyosin contractile forces, integrins pull on the extracellular
matrix (ECM), where force is transmitted via the focal adhesion complex (of
which vinculin is a member). The force balance between CAMs and the actin
cytoskeleton results in actin retrograde flow and allows for the rapid
expansion/shrinkage of the spine structure. Not drawn to scale.
actin polymerization and depolymerization in spines causing
them to enlarge or shrink, respectively (Bosch and Hayashi,
2012). Three distinct pools of F-actin occupy the dendritic spines:
a stable core, forming the center and the base, a dynamic shell,
Frontiers in Cellular Neuroscience | www.frontiersin.org 2December 2018 | Volume 12 | Article 483
Kilinc Mechanical Regulation of Synapses
extending towards the periphery, and an apical pool associated
with the head enlargement during LTP (Honkura et al., 2008).
In addition, there are periodic F-actin rings that shape the
spine neck (Bär et al., 2016). The dynamic and stable pools
have drastically different turnover rates, i.e., polymer half-life of
tens of seconds vs. tens of minutes (Stefen et al., 2016). These
differences in F-actin dynamics likely arise from the distinct
spatiotemporal organization of various actin-binding proteins
(ABPs) in dendritic spines.
Multiple ABPs regulate F-actin in dendritic spines: cortactin
and drebrin localize to the stable core, whereas cofilin localizes to
the dynamic shell (Rácz and Weinberg, 2013). Cortactin directly
interacts with N-methyl-D-aspartate receptors (NMDARs) and
Shank scaffold in the PSD, and regulates the branch-inducing
Arp2/3 complex (Hering and Sheng, 2003), which is required
for spine maturation (Spence et al., 2016). During LTP, the
actin severing protein cofilin is rapidly recruited into the spine
and forms stable complexes with F-actin (as cofilin’s effect
paradoxically shifts from severing to stabilizing with increasing
stoichiometric ratio), which occupy the base of spines and
consolidate their expansion (Bosch et al., 2014). Importantly,
cofilin regulates NMDA-dependent synapse remodeling in LTP
and LTD (Pontrello et al., 2012). Altogether, these data suggest
that actin is the primary structural element in postsynapses.
Microtubules Transiently Invade Dendritic
Spines (Not Only) for Cargo Delivery
Microtubules transiently invade mature spines (residing there
for a few minutes) and the occurrence and duration of these
invasions correlate with neuronal activity (Hu et al., 2008).
Transient nature and activity-dependence of microtubule spine
invasions suggest that they drive cargo in and out; e.g., kinesin-3
delivers synaptotagmin-IV, an LTP regulator, to spine heads
(McVicker et al., 2016). Mitochondria, however, undergo an
actomyosin handoff, i.e., switch from microtubule-based to
actin-based motor transport, to reach the spine head. Similarly,
recycling endosomes containing α-amino-3-hydroxy-5-methyl-
4-isoxazolepropionic acid receptors (AMPARs) are transported
into spines via myosins Va (Correia et al., 2008) and Vb (Wang
et al., 2008). During LTP they undergo syntaxin-4-mediated
exocytosis such that AMPAR are inserted into the plasma
membrane adjacent to the PSD (Kennedy et al., 2010). However,
to what extent AMPAR are actively transported and/or diffuse
laterally into the spine head is debated (Penn et al., 2017). Apart
from retrogradely transporting neurotrophic factors, dendritic
kinesin-4 also regulates microtubule dynamics (Ghiretti et al.,
2016), and is required for learning and memory (Muhia et al.,
2016).
The microtubule plus end-binding protein EB3 directly
interacts with the postsynaptic scaffold protein PSD-95, an
event that decreases EB3-microtubule interaction (Sweet et al.,
2011), suggesting a functional role for dendritic microtubules
in synaptic plasticity. Indeed, spine invasion by EB3-capped
microtubules constrains the ABP p140CAP to the PSD and
maintains the spine size (Jaworski et al., 2009). Accordingly,
during LTD, NMDAR-mediated Ca2+influx removes EB3 from
growing microtubule tips, which causes EB3 accumulation in the
dendrite shaft and suppresses microtubule entry into the spine
(Kapitein et al., 2011). Interestingly, microtubule spine invasions
require a cortactin-dependent increase in the F-actin remodeling
at the base of the synapse, but do not require EB3- and drebrin-
mediated F-actin-microtubule linkages (Schätzle et al., 2018).
Altogether, these studies suggest that microtubules indirectly
regulate synapses via transporting cargo and regulating the actin
cytoskeleton.
Neurofilaments—Additional Structural
Support for Postsynaptic Density?
Neurofilaments are neuron-specific intermediate filaments
associated with axon caliber regulation (Lee and Cleveland,
1996). All four neurofilament subunits found in the CNS—NF-L,
NF-M, NF-H and α-internexin—localize to the synapses
(enriched in postsynapses), are distinct from their axonal
counterparts, and have no known functions (Yuan et al., 2015).
Neurofilaments—but not F-actin or microtubules—directly
interact with SAPAP (Hirao et al., 2000), a member of PSD-
95/SAPAP/Shank core complex, the major scaffold of the PSD
(Zhu et al., 2017). Mice lacking neurofilament subunits have
structurally normal brains, but exhibit synapse plasticity and
memory deficits, indicating a functional role. In support of this,
dopamine D1 receptor (D1R)-induced LTP was modulated by
NF-M, which anchors D1R-containing endosomes to build up
a reservoir of D1Rs for their rapid recycling to the postsynaptic
membrane (Yuan et al., 2015). Curiously, the cytoplasmic tail of
NR1 subunit of NMDAR binds neurofilaments and inhibits their
assembly (Ehlers et al., 1998), consistent with the findings that
synaptic vesicles and endosomes dock onto neurofilament-based
scaffolds, and that vesicle recycling requires neurofilaments to
interact with microtubule motors (Yuan et al., 2017). While
these data suggest that neurofilaments mainly act as postsynaptic
scaffolds, further effort is required to decipher the specific
synaptic function(s) of their subunits.
CELL ADHESION MOLECULES INITIATE,
SPECIFY AND REGULATE SYNAPSES
CAMs are cell surface molecules that link cells to the ECM and to
other cells via homophilic and heterophilic interactions. Synaptic
CAMs are defined as CAMs with potential to induce synapses
via trans interactions (Frei and Stoeckli, 2014). However, other
CAMs also localize to synapses and contribute to synapse
formation and plasticity, through transsynaptic recognition and
signaling processes, respectively (Dalva et al., 2007). In fact, the
subtype of the CAM(s) recruited can determine the synapse
type: for example, alternative splicing of postsynaptic neuroligins
leads to either inhibitory or excitatory synapses (Chih et al.,
2005). CAMs are hierarchically expressed; some are required
for core synaptic functions and others for specialized functions.
Moreover, ‘‘early’’ and ‘‘late’’ synaptic genes are co-expressed and
co-regulated in the same neuron, suggesting that their differential
localization and combinatorial use define synapse specificity and
network connectivity rules (Földy et al., 2016). Here, I will
describe the major CAM families involved; for a complete list of
Frontiers in Cellular Neuroscience | www.frontiersin.org 3December 2018 | Volume 12 | Article 483
Kilinc Mechanical Regulation of Synapses
CAMs in synapses, see recent reviews (Jang et al., 2017; Chamma
and Thoumine, 2018).
Integrins Regulate Functional and
Structural Plasticity of Dendritic Spines
Integrins are heterodimeric transmembrane receptors that link
ECM components to the actin cytoskeleton via adaptor proteins,
e.g., talin and vinculin, forming a ‘‘molecular clutch’’ that
transmits actomyosin contractile forces to the ECM (Sun
et al., 2016). Integrins regulate the spine functional plasticity
through controlling receptor trafficking in a subunit-specific
manner: α3β1-integrins regulate LTP through modulating
NMDAR activity (Chan et al., 2007), whereas αVβ3-integrins
regulate synaptic strength through stabilizing AMPAR in the
membrane (Cingolani et al., 2008). LTP induces long-lasting
(30 min) Rho activity in individual spines that is thought
to relay the transient activation of Ca2+/calmodulin-dependent
protein kinase II (CaMKII; 10 s to structural plasticity
(Murakoshi et al., 2011). Rho and Rac act at different
phases of LTP, mediating the spine neck formation and
driving the spine head expansion, respectively (Rex et al.,
2009). In fact, while ROCK1 regulates the early phase by
forming stable actomyosin bundles that create spine polarity,
ROCK2 regulates the late phase by controlling Rac activity and
by deactivating cofilin (Newell-Litwa et al., 2015). Rac1 activity,
in turn, maintains the globular shape of the spine through
regulating the localization and dynamics of the branched
F-actin network (Chazeau et al., 2014). Separately, integrins
regulate the spine structural plasticity through controlling
actin remodeling: the integrin/focal adhesion pathway regulates
multiple ABPs, including Arp2/3 complex (Serrels et al.,
2007), cortactin (Hotulainen and Hoogenraad, 2010) and
cofilin (Heredia et al., 2006). Together, these studies suggest
that integrins regulate synapses through multiple, intertwined
mechanisms. Moreover, recent proteomic analyses identified
new, non-canonical adhesion components with emerging
functions in mechanotransduction and receptor trafficking
(Humphries et al., 2015). Their potential roles in synapse
formation and plasticity are yet to be explored.
Cadherin/Catenin Complexes Are
Important Regulators of Synapse Plasticity
Cadherins are Ca2+-dependent transmembrane proteins found
at intercellular adherent junctions. Their ectodomains form
homo- and occasional heterophilic bonds and their intracellular
tails interact with various partners—notably catenins and
vinculins—to induce downstream signaling. Cadherins can form
transsynaptic bonds with different adhesiveness and kinetics,
thanks to their large repertoire of homophilic interactions: trans
dimers (slip bond), X-dimers (catch bond) and clusters, which
combine cis and trans bonds (Leckband and de Rooij, 2014).
Cadherin mechanotransduction is highly complex (reviewed
in Hoffman and Yap, 2015): on one hand, cadherin-catenin
complex directly interacts with F-actin via a two state catch
bond (that strengthens under force, as opposed to slip bonds
that weaken under force), reinforcing intercellular adhesion
(Buckley et al., 2014). On the other hand, force induces a
conformational change in α-catenin, revealing cryptic sites
for vinculin binding. Activated vinculin not only directly
binds to F-actin, but also recruits Ena/VASP family proteins
to promote actin assembly, further reinforcing the cadherin-
cytoskeleton coupling (le Duc et al., 2010). In the CNS, classical
cadherins (e.g., N-cadherin) localize to pre- and postsynapses,
and border the presynaptic active zone (PAZ; Uchida et al., 1996).
During development, neuron-neuron interactions regulate the
activity-dependent dendrite arborization, which is mediated by
cadherin/catenin surface levels (Tan et al., 2010). It should
be noted that, due to structural differences, N-cadherin and
E-cadherin dimers have different disassembly kinetics, where
the former depend strongly on Ca2+binding and cannot
form X-dimers (Vunnam and Pedigo, 2012). It is therefore
likely that N-cadherin and E-cadherin have distinct mechanical
behavior. Similarly, αN-catenin differs from αE-catenin in terms
of β-catenin binding kinetics (Pokutta et al., 2014), further
suggesting that mechanotransduction mechanisms identified for
E-cadherin/αE-catenin may not be applicable to synapses.
Cadherins take part in spine and synapse formation,
particularly in excitatory neurons (Seong et al., 2015). In
postnatal, excitatory synapses, N-cadherin is required for
LTP and spine enlargement—but not LTD or spine density
and morphology, suggesting that cadherins selectively regulate
synapse plasticity (Bozdagi et al., 2010). Indeed, cadherin
accumulation on synaptic membranes is required for stabilizing
postsynaptic receptors, e.g., kainate receptors (Fièvre et al., 2016)
and AMPAR (Mills et al., 2017). Catenins also regulate synapses:
upon NMDAR activation, β-catenin is redistributed from the
dendrite shaft to the spines (where it binds N-cadherin), leading
to synapse enlargement, consistent with its role in learning and
memory (Murase et al., 2002). Furthermore, spines compete for
surface-bound N-cadherin/β-catenin complexes, which appear
to be the key drivers for activity-dependent spine pruning,
where β-catenin redistribution determines the fate of individual
spines, i.e., stabilizing one while eliminating its neighbors (Bian
et al., 2015). Cumulatively, these data suggest that cadherin and
its intracellular binding partners are important regulators of
synapse plasticity.
Several Other CAM Families Localize to
Synapses
Ectodomains of the immunoglobulin superfamily CAM
(IgCAM) contain tandem repeats of Ig-like domains, which
permit the formation of a variety of trans and cis bonds,
leading to molecular zippers (Aricescu and Jones, 2007). Such
flexibility is ideal for mechanosignaling, where force-induced
conformational change regulates intracellular signaling (Johnson
et al., 2007). For example, neural CAM (NCAM)—critical
for neurodevelopment (Maness and Schachner, 2007)—forms
at least two types of homophilic bonds differing in force
sensitivity and intercellular distance (Wieland et al., 2005).
NCAM regulates synapses by crosslinking to NMDAR and
CaMKII via a spectrin-based postsynaptic scaffold (Sytnyk et al.,
2006). Additionally, NCAM interacts with dynein to tether
microtubule plus-ends, an event that enhances synapse stability
Frontiers in Cellular Neuroscience | www.frontiersin.org 4December 2018 | Volume 12 | Article 483
Kilinc Mechanical Regulation of Synapses
(Perlson et al., 2013). SynCAM, originally identified as an
IgCAM promoting synaptogenesis (Biederer et al., 2002), forms
transsynaptic homophilic bonds at spine heads encircling the
synaptic cleft (Perez de Arce et al., 2015). Importantly, SynCAM
complexes enlarge during LTD, suggesting that they control the
cleft diameter.
Presynaptic neurexins interact with postsynaptic neuroligins,
which directly bind to and recruit PSD-95—a function regulated
through phosphorylation (Giannone et al., 2013; Bemben et al.,
2014). The neurexin-neuroligin bond stabilizes the dendritic
filopodia during synaptogenesis (Chen et al., 2010), and regulates
the synapse specificity in an isoform-dependent manner (Graf
et al., 2004; Boucard et al., 2005). In fact, neurexins are
heparan sulfate proteoglycans and neuroligin binding to heparan
sulfate chains on neurexin is necessary for synapse development
(Zhang et al., 2018). In mature synapses, NMDAR activation
leads to juxtamembrane cleavage of neuroligin by matrix
metalloproteinase 9 (MMP-9) or by ADAM10, which destabilizes
neurexins and decreases synaptic strength by altering presynaptic
release (Peixoto et al., 2012; Suzuki et al., 2012).
Another CAM type required for synapse specificity is the
clustered protocadherin (Kostadinov and Sanes, 2015). Clustered
protocadherins form homodimers and antiparallel homophilic
trans interactions to regulate dendritic self-avoidance (Nicoludis
et al., 2015). Unfortunately, whether CAMs other than integrins
and cadherins participate in the mechanotransduction is largely
unknown. One exception to this is the ephrin-Eph receptor
pair, which bidirectionally regulates synapse formation and
maturation in the adult, i.e., ephrins can signal into the
Eph-receptor-expressing cell (forward) or into their host cell
(reverse; reviewed in Klein, 2009). For example, the dynamics
of EphB2 kinase activity at the tip of a dendritic filopodium
upon binding to axonal ephrin-B1 determines whether the
filopodium retracts or establishes a synapse (Mao et al., 2018).
On the other hand, dendritic ephrin-B3 directly interacts with
PSD-95 to control its localization and stability, via activity-
dependent phosphoregulation (Hruska et al., 2015). Importantly,
physically restraining the ephrin-A1 modulates cytoskeletal
dynamics by blocking the EphA2 receptor clustering, indicating
that ephrin-Eph pair is mechanosensitive (Salaita et al., 2010).
Together, these data suggest that mechanotransduction via
CAMs may be a general mechanism in synapse regulation, and
not unique to integrins and cadherins.
OTHER POTENTIAL
MECHANOREGULATORS OF SYNAPSE
PLASTICITY: SCAFFOLD PROTEINS,
MECHANOSENSITIVE ION CHANNELS,
AND THE EXTRACELLULAR MATRIX
Apart from major mechanical actors (cytoskeleton, CAMs),
synapses contain other mechanically-relevant components.
Synaptic scaffolds support the dynamic components of the
PAZ and the PSD (Ziv and Fisher-Lavie, 2014) and, since
they physically couple CAMs to the underlying cytoskeleton,
they potentially bear tensile forces. While non-specifically
pulling on a neurite is sufficient to recruit Bassoon presynaptic
scaffold into a potential presynapse (Suarez et al., 2013), the
mechanosensitivity of scaffold proteins remains unknown.
Mechanosensitive ion channels, however, are expressed in
neurons (Hu et al., 2015), although most do not localize to
synapses. An important exception to this is NMDAR, which
may be activated through increased membrane tension (Paoletti
and Ascher, 1994) or by cytoskeletal forces acting on its
intracellular domain (Singh et al., 2012). The ECM is another
potential mechanoregulator of synapses: the composition,
structure and stiffness of the ECM (reciprocally) regulate
the cellular mechanotransduction (reviewed in Humphrey
et al., 2014). Components of the ECM form a perineuronal
net that surrounds dendritic spines and extends into the
synaptic cleft (Dansie and Ethell, 2011). In the adult brain,
chondroitin sulfate proteoglycans stabilize dendritic spine
structure and movement, whereas, other glycoproteins
(notably, reelin, agrin and tenascins) are important regulators
of synapse plasticity (reviewed in Levy et al., 2014). For
example, cleavage of agrin (an integrin αvβ1ligand) by
neurotrypsin promotes LTP by facilitating new dendritic
filopodia (Matsumoto-Miyai et al., 2009). Similarly, cleavage
of the hyaluronan receptor CD44 by MMP-9, results in its
detachment from the ECM and leads to dendritic spine
elongation (Bijata et al., 2017). These data suggest that ECM
proteolysis regulates synapses, potentially by modifying their
force balance.
IS SYNAPSE PLASTICITY MECHANICALLY
REGULATED?
As illustrated above, mechanotransduction takes place during
synapse plasticity. It is clear that mechanical processes convey
biochemical signals into spine remodeling; however, whether
they also convey purely mechanical signals (e.g., forces with a
certain magnitude, rate, duration and frequency; Hoffman et al.,
2011) to invoke structural change is unknown. In fact, changes
in plasma membrane curvature and tension (due to spine
remodeling) may be sufficient to induce mechanotransduction
(Diz-Muñoz et al., 2013). Identifying such mechanisms remains
a challenge due to the high level of complexity, i.e., synapses
involve numerous CAMs—some of which crosstalk via common
effectors (Mui et al., 2016)—and intertwined cytoskeletal
networks (Figure 1). Since pulling on a transsynaptic CAM
(e.g., cadherin) bond modulates not only its unbinding rate,
but also its downstream signaling by revealing cryptic domains
(e.g., α-catenin-vinculin interaction), the acting force needs to be
tightly controlled, such that intracellular signaling precedes bond
breakage.
Notably, cadherin transsynaptic bonds can be synapse-
specific: synapses between hippocampal CA3 and CA1 neurons
with high-magnitude LTP require cis dimers of postsynaptic
cadherins-6 and -10 to form trans bonds with presynaptic
cadherin-9 (Basu et al., 2017). This level of complexity
suggests mechanosignaling to take place, considering the
differences in the force-dependency of these bonds. Indeed,
N-cadherins stabilize filopodial F-actin through counteracting
Frontiers in Cellular Neuroscience | www.frontiersin.org 5December 2018 | Volume 12 | Article 483
Kilinc Mechanical Regulation of Synapses
the actomyosin pulling force—a process associated with the
transition of the dendritic filopodia into spines (Chazeau
et al., 2015). Remarkably, pulling on N-cadherins at the tips of
dendritic filopodia—via optical tweezers—resulted in rapid actin
accumulation and mushroom-like spine morphology (Chazeau
et al., 2015), suggesting that cadherin mechanotransduction
alone can trigger synapse remodeling. Nevertheless, to
what degree mechanotransduction and signal transduction
mechanisms overlap, whether their activation is synchronized,
and whether they operate synergistically (e.g., to strengthen
synaptic signaling) remain open questions.
IS SYNAPSE MECHANICS RELEVANT TO
NEURODEGENERATIVE DISEASES?
Whether synapse mechanics is relevant to neurodegenerative
diseases is another open question. Synaptic failure is a key event
in most neurodegenerative diseases, particularly in Alzheimer’s
disease (AD). Accordingly, cytoskeletal proteins, including
cofilin (Rahman et al., 2014), drebrin (Gordon-Weeks, 2016)
and NF-L (Bacioglu et al., 2016) were implicated in AD
pathophysiology. Separately, multiple CAMs, including integrin
(Caltagarone et al., 2007), N-cadherin (Andreyeva et al., 2012),
NCAM (Leshchyns’ka et al., 2015) and neurexin-neuroligin
(Brito-Moreira et al., 2017) are involved in amyloid-β-induced
synaptotoxicity, a major event in AD. In fact, amyloid precursor
protein (APP), whose cleavage products include amyloid-β, is a
transsynaptic CAM (Ludewig and Korte, 2016). APP regulates
the PAZ organization (Laßek et al., 2016) and the dendritic spine
shape (Weyer et al., 2014), and its dimerization is regulated
by N-cadherin (Asada-Utsugi et al., 2011). These observations
suggest a link between synapse mechanics and synaptic failure
in AD, but direct evidence is missing.
CONCLUSIONS AND OUTLOOK
In this mini review article, I attempted to highlight the
mechanically-relevant mechanisms that take part in synapse
formation and plasticity. Growing evidence suggests that
synapses employ mechanosensitive molecules and mechanical
processes; however, direct evidence for the mechanoregulation
of synapse behavior is currently lacking. Implementing new
technologies for force application or measurement (Kilinc et al.,
2015), combined with novel single-molecule or super-resolution
approaches (Jin et al., 2018) may help discover such mechanisms.
To this end, magnetic tweezers would be an excellent tool
to specifically-target CAMs on dendritic filopodia (or axon
shafts) and to deliver well-defined forces to identify stretch
paradigms leading to spine (or PAZ) formation. Downstream
effects of the mechanical input may be monitored live in
terms of: (i) cytoskeletal dynamics; (ii) activity of cytoskeleton-
associated (e.g., ABPs) or signaling proteins (e.g., CaMKII); and
(iii) secondary messengers, such as Ca2+or cyclic nucleotides
(Blasiak et al., 2017). Notably, the frequency of local Ca2+
transients in dendritic filopodia upon initial contact with an
excitatory axon determines whether the connection will be lost
or stabilized (Lohmann and Bonhoeffer, 2008), suggesting that
Ca2+may be an intermediary to mechanical signaling. Finally,
single-molecule tension sensors (Cost et al., 2015) may be used
to identify load-bearing proteins in synapses during activity-
dependent remodeling, as well as to measure forces acting on
these proteins as a function of spine size and shape. A better
understanding of synapse mechanoregulation could pave the way
for mechanically modulated therapies against synaptic failure.
AUTHOR CONTRIBUTIONS
DK wrote the manuscript.
FUNDING
This work was funded by the 2017 Pilot Research Grant from
Fondation Vaincre Alzheimer and by the EU Joint Programme—
Neurodegenerative Diseases Research (JPND; 3DMiniBrain).
ACKNOWLEDGMENTS
I would like to thank Esther Stoeckli, Beatriz Rico, Marina
Mikhaylova and Agata Blasiak for critical reading of the
manuscript.
REFERENCES
Alvarez-Castelao, B., and Schuman, E. M. (2015). The regulation of synaptic
protein turnover. J. Biol. Chem. 290, 28623–28630. doi: 10.1074/jbc.R115.
657130
Andreyeva, A., Nieweg, K., Horstmann, K., Klapper, S., Müller-Schiffmann, A.,
Korth, C., et al. (2012). C-terminal fragment of N-cadherin accelerates synapse
destabilization by amyloid-β.Brain 135, 2140–2154. doi: 10.1093/brain/
aws120
Aricescu, A. R., and Jones, E. Y. (2007). Immunoglobulin superfamily cell
adhesion molecules: zippers and signals. Curr. Opin. Cell Biol. 19, 543–550.
doi: 10.1016/j.ceb.2007.09.010
Asada-Utsugi, M., Uemura, K., Noda, Y., Kuzuya, A., Maesako, M., Ando, K., et al.
(2011). N-cadherin enhances APP dimerization at the extracellular domain and
modulates Aβproduction. J. Neurochem. 119, 354–363. doi: 10.1111/j.1471-
4159.2011.07364.x
Bacioglu, M., Maia, L. F., Preische, O., Schelle, J., Apel, A., Kaeser, S. A., et al.
(2016). Neurofilament light chain in blood and CSF as marker of disease
progression in mouse models and in neurodegenerative diseases. Neuron 91,
56–66. doi: 10.1016/j.neuron.2016.05.018
Bär, J., Kobler, O., van Bommel, B., and Mikhaylova, M. (2016). Periodic
F-actin structures shape the neck of dendritic spines. Sci. Rep. 6:37136.
doi: 10.1038/srep37136
Basu, R., Duan, X., Taylor, M. R., Martin, E. A., Muralidhar, S., Wang, Y., et al.
(2017). Heterophilic type II cadherins are required for high-magnitude synaptic
potentiation in the hippocampus. Neuron 96, 160.e8–176.e8. doi: 10.1016/j.
neuron.2017.09.009
Bemben, M. A., Shipman, S. L., Hirai, T., Herring, B. E., Li, Y., Badger, J. D., et al.
(2014). CaMKII phosphorylation of neuroligin-1 regulates excitatory synapses.
Nat. Neurosci. 17, 56–64. doi: 10.1038/nn.3601
Bian, W.-J., Miao, W.-Y., He, S.-J., Qiu, Z., and Yu, X. (2015). Coordinated
spine pruning and maturation mediated by inter-spine competition for
cadherin/catenin complexes. Cell 162, 808–822. doi: 10.1016/j.cell.2015.07.018
Biederer, T., Sara, Y., Mozhayeva, M., Atasoy, D., Liu, X., Kavalali, E. T., et al.
(2002). SynCAM, a synaptic adhesion molecule that drives synapse assembly.
Science 297, 1525–1531. doi: 10.1126/science.1072356
Frontiers in Cellular Neuroscience | www.frontiersin.org 6December 2018 | Volume 12 | Article 483
Kilinc Mechanical Regulation of Synapses
Bijata, M., Labus, J., Guseva, D., Stawarski, M., Butzlaff, M., Dzwonek, J., et al.
(2017). Synaptic remodeling depends on signaling between serotonin receptors
and the extracellular matrix. Cell Rep. 19, 1767–1782. doi: 10.1016/j.celrep.
2017.05.023
Blasiak, A., Kilinc, D., and Lee, G. U. (2017). Neuronal cell bodies remotely
regulate axonal growth response to localized Netrin-1 treatment via second
messenger and DCC dynamics. Front. Cell. Neurosci. 10:298. doi: 10.3389/fncel.
2016.00298
Bosch, M., Castro, J., Saneyoshi, T., Matsuno, H., Sur, M., and Hayashi, Y. (2014).
Structural and molecular remodeling of dendritic spine substructures during
long-term potentiation. Neuron 82, 444–459. doi: 10.1016/j.neuron.2014.
03.021
Bosch, M., and Hayashi, Y. (2012). Structural plasticity of dendritic spines. Curr.
Opin. Neurobiol. 22, 383–388. doi: 10.1016/j.conb.2011.09.002
Boucard, A. A., Chubykin, A. A., Comoletti, D., Taylor, P., and Südhof, T. C.
(2005). A splice code for trans-synaptic cell adhesion mediated by binding of
neuroligin 1 to α- and β-neurexins. Neuron 48, 229–236. doi: 10.1016/j.neuron.
2005.08.026
Bozdagi, O., Wang, X. B., Nikitczuk, J. S., Anderson, T. R., Bloss, E. B.,
Radice, G. L., et al. (2010). Persistence of coordinated long-term potentiation
and dendritic spine enlargement at mature hippocampal CA1 synapses requires
N-cadherin. J. Neurosci. 30, 9984–9989. doi: 10.1523/jneurosci.1223-10.2010
Brito-Moreira, J., Lourenco, M. V., Oliveira, M. M., Ribeiro, F. C., Ledo, J. H.,
Diniz, L. P., et al. (2017). Interaction of amyloid-β(Aβ) oligomers with
neurexin 2αand neuroligin 1 mediates synapse damage and memory loss in
mice. J. Biol. Chem. 292, 7327–7337. doi: 10.1074/jbc.M116.761189
Buckley, C. D., Tan, J., Anderson, K. L., Hanein, D., Volkmann, N., Weis, W. I.,
et al. (2014). Cell adhesion. The minimal cadherin-catenin complex binds to
actin filaments under force. Science 346:1254211. doi: 10.1126/science.1254211
Caltagarone, J., Jing, Z., and Bowser, R. (2007). Focal adhesions regulate Aβ
signaling and cell death in Alzheimer’s disease. Biochim. Biophys. Acta 1772,
438–445. doi: 10.1016/j.bbadis.2006.11.007
Chamma, I., and Thoumine, O. (2018). Dynamics, nanoscale organization, and
function of synaptic adhesion molecules. Mol. Cell. Neurosci. 91, 95–107.
doi: 10.1016/j.mcn.2018.04.007
Chan, C. S., Levenson, J. M., Mukhopadhyay, P. S., Zong, L., Bradley, A.,
Sweatt, J. D., et al. (2007). α3-integrins are required for hippocampal long-term
potentiation and working memory. Learn. Mem. 14, 606–615. doi: 10.1101/lm.
648607
Chazeau, A., Garcia, M., Czöndör, K., Perrais, D., Tessier, B., Giannone, G.,
et al. (2015). Mechanical coupling between transsynaptic N-cadherin adhesions
and actin flow stabilizes dendritic spines. Mol. Biol. Cell 26, 859–873.
doi: 10.1091/mbc.e14-06-1086
Chazeau, A., Mehidi, A., Nair, D., Gautier, J. J., Leduc, C., Chamma, I., et al. (2014).
Nanoscale segregation of actin nucleation and elongation factors determines
dendritic spine protrusion. EMBO J. 33, 2745–2764. doi: 10.15252/embj.
201488837
Chen, S. X., Tari, P. K., She, K., and Haas, K. (2010). Neurexin-neuroligin cell
adhesion complexes contribute to synaptotropic dendritogenesis via growth
stabilization mechanisms in vivo.Neuron 67, 967–983. doi: 10.1016/j.neuron.
2010.08.016
Chih, B., Engelman, H., and Scheiffele, P. (2005). Control of excitatory
and inhibitory synapse formation by neuroligins. Science 307, 1324–1328.
doi: 10.1126/science.1107470
Cingolani, L. A., Thalhammer, A., Yu, L. M., Catalano, M., Ramos, T.,
Colicos, M. A., et al. (2008). Activity-dependent regulation of synaptic AMPA
receptor composition and abundance by β3 integrins. Neuron 58, 749–762.
doi: 10.1016/j.neuron.2008.04.011
Coles, C. H., and Bradke, F. (2015). Coordinating neuronal actin-microtubule
dynamics. Curr. Biol. 25, R677–R691. doi: 10.1016/j.cub.2015.06.020
Correia, S. S., Bassani, S., Brown, T. C., Lisé, M. F., Backos, D. S., El-
Husseini, A., et al. (2008). Motor protein-dependent transport of AMPA
receptors into spines during long-term potentiation. Nat. Neurosci. 11,
457–466. doi: 10.1038/nn2063
Cost, A. L., Ringer, P., Chrostek-Grashoff, A., and Grashoff, C. (2015). How to
measure molecular forces in cells: a guide to evaluating genetically-encoded
FRET-based tension sensors. Cell. Mol. Bioeng. 8, 96–105. doi: 10.1007/s12195-
014-0368-1
Dalva, M. B., Mcclelland, A. C., and Kayser, M. S. (2007). Cell adhesion
molecules: signaling functions at the synapse. Nat. Rev. Neurosci. 8, 206–220.
doi: 10.1038/nrn2075
Dansie, L. E., and Ethell, I. M. (2011). Casting a net on dendritic spines:
the extracellular matrix and its receptors. Dev. Neurobiol. 71, 956–981.
doi: 10.1002/dneu.20963
Diz-Muñoz, A., Fletcher, D. A., and Weiner, O. D. (2013). Use the force:
membrane tension as an organizer of cell shape and motility. Trends Cell Biol.
23, 47–53. doi: 10.1016/j.tcb.2012.09.006
Ehlers, M. D., Fung, E. T., O’Brien, R. J., and Huganir, R. L. (1998). Splice
variant-specific interaction of the NMDA receptor subunit NR1 with neuronal
intermediate filaments. J. Neurosci. 18, 720–730. doi: 10.1523/jneurosci.18-02-
00720.1998
Fièvre, S., Carta, M., Chamma, I., Labrousse, V., Thoumine, O., and Mulle, C.
(2016). Molecular determinants for the strictly compartmentalized expression
of kainate receptors in CA3 pyramidal cells. Nat. Commun. 7:12738.
doi: 10.1038/ncomms12738
Fletcher, D. A., and Mullins, R. D. (2010). Cell mechanics and the cytoskeleton.
Nature 463, 485–492. doi: 10.1038/nature08908
Földy, C., Darmanis, S., Aoto, J., Malenka, R. C., Quake, S. R., and Südhof, T. C.
(2016). Single-cell RNAseq reveals cell adhesion molecule profiles in
electrophysiologically defined neurons. Proc. Natl. Acad. Sci. U S A 113,
E5222–E5231. doi: 10.1073/pnas.1610155113
Franze, K. (2013). The mechanical control of nervous system development.
Development 140, 3069–3077. doi: 10.1242/dev.079145
Frei, J. A., and Stoeckli, E. T. (2014). SynCAMs extend their functions beyond the
synapse. Eur. J. Neurosci. 39, 1752–1760. doi: 10.1111/ejn.12544
Frost, N. A., Shroff, H., Kong, H., Betzig, E., and Blanpied, T. A. (2010). Single-
molecule discrimination of discrete perisynaptic and distributed sites of actin
filament assembly within dendritic spines. Neuron 67, 86–99. doi: 10.1016/j.
neuron.2010.05.026
Ghiretti, A. E., Thies, E., Tokito, M. K., Lin, T., Ostap, E. M., Kneussel, M., et al.
(2016). Activity-dependent regulation of distinct transport and cytoskeletal
remodeling functions of the dendritic kinesin KIF21B. Neuron 92, 857–872.
doi: 10.1016/j.neuron.2016.10.003
Giannone, G., Mondin, M., Grillo-Bosch, D., Tessier, B., Saint-Michel, E.,
Czöndör, K., et al. (2013). Neurexin-1βbinding to neuroligin-1 triggers
the preferential recruitment of PSD-95 versus gephyrin through tyrosine
phosphorylation of neuroligin-1. Cell Rep. 3, 1996–2007. doi: 10.1016/j.celrep.
2013.05.013
Gordon-Weeks, P. R. (2016). The role of the drebrin/EB3/Cdk5 pathway in
dendritic spine plasticity, implications for Alzheimer’s disease. Brain Res. Bull.
126, 293–299. doi: 10.1016/j.brainresbull.2016.06.015
Graf, E. R., Zhang, X., Jin, S. X., Linhoff, M. W., and Craig, A. M. (2004).
Neurexins induce differentiation of GABA and glutamate postsynaptic
specializations via neuroligins. Cell 119, 1013–1026. doi: 10.1016/j.cell.2004.
11.035
Heredia, L., Helguera, P., de Olmos, S., Kedikian, G., Solá Vigo, F., Laferla, F.,
et al. (2006). Phosphorylation of actin-depolymerizing factor/cofilin by
LIM-kinase mediates amyloid β-induced degeneration: a potential mechanism
of neuronal dystrophy in Alzheimer’s disease. J. Neurosci. 26, 6533–6542.
doi: 10.1523/jneurosci.5567-05.2006
Hering, H., and Sheng, M. (2003). Activity-dependent redistribution and
essential role of cortactin in dendritic spine morphogenesis. J. Neurosci. 23,
11759–11769. doi: 10.1523/jneurosci.23-37-11759.2003
Hirao, K., Hata, Y., Deguchi, M., Yao, I., Ogura, M., Rokukawa, C., et al.
(2000). Association of synapse-associated protein 90/postsynaptic density-
95-associated protein (SAPAP) with neurofilaments. Genes Cells 5, 203–210.
doi: 10.1046/j.1365-2443.2000.00318.x
Hoffman, B. D., Grashoff, C., and Schwartz, M. A. (2011). Dynamic molecular
processes mediate cellular mechanotransduction. Nature 475, 316–323.
doi: 10.1038/nature10316
Hoffman, B. D., and Yap, A. S. (2015). Towards a dynamic understanding of
cadherin-based mechanobiology. Trends Cell Biol. 25, 803–814. doi: 10.1016/j.
tcb.2015.09.008
Honkura, N., Matsuzaki, M., Noguchi, J., Ellis-Davies, G. C., and Kasai, H. (2008).
The subspine organization of actin fibers regulates the structure and plasticity
of dendritic spines. Neuron 57, 719–729. doi: 10.1016/j.neuron.2008.01.013
Frontiers in Cellular Neuroscience | www.frontiersin.org 7December 2018 | Volume 12 | Article 483
Kilinc Mechanical Regulation of Synapses
Hotulainen, P., and Hoogenraad, C. C. (2010). Actin in dendritic spines:
connecting dynamics to function. J. Cell Biol. 189, 619–629. doi: 10.1083/jcb.
201003008
Hruska, M., Henderson, N. T., Xia, N. L., Le Marchand, S. J., and Dalva, M. B.
(2015). Anchoring and synaptic stability of PSD-95 is driven by ephrin-B3. Nat.
Neurosci. 18, 1594–1605. doi: 10.1038/nn.4140
Hu, W., An, C., and Chen, W. J. (2015). Molecular mechanoneurobiology:
an emerging angle to explore neural synaptic functions. Biomed Res. Int.
2015:486827. doi: 10.1155/2015/486827
Hu, X., Viesselmann, C., Nam, S., Merriam, E., and Dent, E. W. (2008). Activity-
dependent dynamic microtubule invasion of dendritic spines. J. Neurosci. 28,
13094–13105. doi: 10.1523/JNEUROSCI.3074-08.2008
Humphrey, J. D., Dufresne, E. R., and Schwartz, M. A. (2014).
Mechanotransduction and extracellular matrix homeostasis. Nat. Rev.
Mol. Cell Biol. 15, 802–812. doi: 10.1038/nrm3896
Humphries, J. D., Paul, N. R., Humphries, M. J., and Morgan, M. R.
(2015). Emerging properties of adhesion complexes: what are they and
what do they do? Trends Cell Biol. 25, 388–397. doi: 10.1016/j.tcb.2015.
02.008
Jang, S., Lee, H., and Kim, E. (2017). Synaptic adhesion molecules and excitatory
synaptic transmission. Curr. Opin. Neurobiol. 45, 45–50. doi: 10.1016/j.conb.
2017.03.005
Jaworski, J., Kapitein, L. C., Gouveia, S. M., Dortland, B. R., Wulf, P. S.,
Grigoriev, I., et al. (2009). Dynamic microtubules regulate dendritic spine
morphology and synaptic plasticity. Neuron 61, 85–100. doi: 10.1016/j.neuron.
2008.11.013
Jin, D., Xi, P., Wang, B., Zhang, L., Enderlein, J., and van Oijen, A. M. (2018).
Nanoparticles for super-resolution microscopy and single-molecule tracking.
Nat. Methods 15, 415–423. doi: 10.1038/s41592-018-0012-4
Johnson, C. P., Tang, H. Y., Carag, C., Speicher, D. W., and Discher, D. E.
(2007). Forced unfolding of proteins within cells. Science 317, 663–666.
doi: 10.1126/science.1139857
Kapitein, L. C., Yau, K. W., Gouveia, S. M., van der Zwan, W. A., Wulf, P. S.,
Keijzer, N., et al. (2011). NMDA receptor activation suppresses microtubule
growth and spine entry. J. Neurosci. 31, 8194–8209. doi: 10.1523/JNEUROSCI.
6215-10.2011
Kennedy, M. J., Davison, I. G., Robinson, C. G., and Ehlers, M. D. (2010). Syntaxin-
4 defines a domain for activity-dependent exocytosis in dendritic spines. Cell
141, 524–535. doi: 10.1016/j.cell.2010.02.042
Kerstein, P. C., Nichol, R. H. IV., and Gomez, T. M. (2015). Mechanochemical
regulation of growth cone motility. Front. Cell. Neurosci. 9:244.
doi: 10.3389/fncel.2015.00244
Kilinc, D., Blasiak, A., and Lee, G. U. (2015). Microtechnologies for studying the
role of mechanics in axon growth and guidance. Front. Cell. Neurosci. 9:282.
doi: 10.3389/fncel.2015.00282
Klein, R. (2009). Bidirectional modulation of synaptic functions by Eph/ephrin
signaling. Nat. Neurosci. 12, 15–20. doi: 10.1038/nn.2231
Kostadinov, D., and Sanes, J. R. (2015). Protocadherin-dependent dendritic
self-avoidance regulates neural connectivity and circuit function. Elife
4:e08964. doi: 10.7554/eLife.08964
Laßek, M., Weingarten, J., Wegner, M., Mueller, B. F., Rohmer, M.,
Baeumlisberger, D., et al. (2016). APP is a context-sensitive regulator of
the hippocampal presynaptic active zone. PLoS Comput. Biol. 12:e1004832.
doi: 10.1371/journal.pcbi.1004832
le Duc, Q., Shi, Q., Blonk, I., Sonnenberg, A., Wang, N., Leckband, D., et al. (2010).
Vinculin potentiates E-cadherin mechanosensing and is recruited to actin-
anchored sites within adherens junctions in a myosin II-dependent manner.
J. Cell Biol. 189, 1107–1115. doi: 10.1083/jcb.201001149
Leckband, D. E., and de Rooij, J. (2014). Cadherin adhesion and
mechanotransduction. Annu. Rev. Cell Dev. Biol. 30, 291–315. doi: 10.1146/
annurev-cellbio-100913-013212
Lee, M. K., and Cleveland, D. W. (1996). Neuronal intermediate filaments.
Annu. Rev. Neurosci. 19, 187–217. doi: 10.1146/annurev.ne.19.030196.
001155
Leshchyns’ka, I., Liew, H. T., Shepherd, C., Halliday, G. M., Stevens, C. H.,
Ke, Y. D., et al. (2015). Aβ-dependent reduction of NCAM2-mediated synaptic
adhesion contributes to synapse loss in Alzheimer’s disease. Nat. Commun.
6:8836. doi: 10.1038/ncomms9836
Levy, A. D., Omar, M. H., and Koleske, A. J. (2014). Extracellular matrix control
of dendritic spine and synapse structure and plasticity in adulthood. Front.
Neuroanat. 8:116. doi: 10.3389/fnana.2014.00116
Lohmann, C., and Bonhoeffer, T. (2008). A role for local calcium signaling in
rapid synaptic partner selection by dendritic filopodia. Neuron 59, 253–260.
doi: 10.1016/j.neuron.2008.05.025
Ludewig, S., and Korte, M. (2016). Novel insights into the physiological function
of the APP (gene) family and its proteolytic fragments in synaptic plasticity.
Front. Mol. Neurosci. 9:161. doi: 10.3389/fnmol.2016.00161
Maness, P. F., and Schachner, M. (2007). Neural recognition molecules of the
immunoglobulin superfamily: signaling transducers of axon guidance and
neuronal migration. Nat. Neurosci. 10, 19–26. doi: 10.1038/nn0207-263b
Mao, Y. T., Zhu, J. X., Hanamura, K., Iurilli, G., Datta, S. R., and Dalva, M. B.
(2018). Filopodia conduct target selection in cortical neurons using differences
in signal kinetics of a sngle kinase. Neuron 98, 767.e8–782.e8. doi: 10.1016/j.
neuron.2018.04.011
Matsumoto-Miyai, K., Sokolowska, E., Zurlinden, A., Gee, C. E., Lüscher, D.,
Hettwer, S., et al. (2009). Coincident pre- and postsynaptic activation induces
dendritic filopodia via neurotrypsin-dependent agrin cleavage. Cell 136,
1161–1171. doi: 10.1016/j.cell.2009.02.034
McVicker, D. P., Awe, A. M., Richters, K. E., Wilson, R. L., Cowdrey, D. A.,
Hu, X., et al. (2016). Transport of a kinesin-cargo pair along microtubules
into dendritic spines undergoing synaptic plasticity. Nat. Commun. 7:12741.
doi: 10.1038/ncomms12741
Mills, F., Globa, A. K., Liu, S., Cowan, C. M., Mobasser, M., Phillips, A. G., et al.
(2017). Cadherins mediate cocaine-induced synaptic plasticity and behavioral
conditioning. Nat. Neurosci. 20, 540–549. doi: 10.1038/nn.4503
Muhia, M., Thies, E., Labonté, D., Ghiretti, A. E., Gromova, K. V., Xompero, F.,
et al. (2016). The kinesin KIF21B regulates microtubule dynamics and
is essential for neuronal morphology, synapse function, and learning and
memory. Cell Rep. 15, 968–977. doi: 10.1016/j.celrep.2016.03.086
Mui, K. L., Chen, C. S., and Assoian, R. K. (2016). The mechanical regulation of
integrin-cadherin crosstalk organizes cells, signaling and forces. J. Cell Sci. 129,
1093–1100. doi: 10.1242/jcs.183699
Murakoshi, H., Wang, H., and Yasuda, R. (2011). Local, persistent activation of
Rho GTPases during plasticity of single dendritic spines. Nature 472, 100–104.
doi: 10.1038/nature09823
Murase, S., Mosser, E., and Schuman, E. M. (2002). Depolarization drives β-
Catenin into neuronal spines promoting changes in synaptic structure and
function. Neuron 35, 91–105. doi: 10.1016/s0896-6273(02)00764-x
Newell-Litwa, K. A., Badoual, M., Asmussen, H., Patel, H., Whitmore, L., and
Horwitz, A. R. (2015). ROCK1 and 2 differentially regulate actomyosin
organization to drive cell and synaptic polarity. J. Cell Biol. 210, 225–242.
doi: 10.1083/jcb.201504046
Nichol, R. H. IV., Hagen, K. M., Lumbard, D. C., Dent, E. W., and Gómez, T. M.
(2016). Guidance of axons by local coupling of retrograde flow to point
contact adhesions. J. Neurosci. 36, 2267–2282. doi: 10.1523/JNEUROSCI.2645-
15.2016
Nicoludis, J. M., Lau, S. Y., Schärfe, C. P. I., Marks, D. S., Weihofen, W. A., and
Gaudet, R. (2015). Structure and sequence analyses of clustered protocadherins
reveal antiparallel interactions that mediate homophilic specificity. Structure
23, 2087–2098. doi: 10.1016/j.str.2015.09.005
Paoletti, P., and Ascher, P. (1994). Mechanosensitivity of NMDA receptors in
cultured mouse central neurons. Neuron 13, 645–655. doi: 10.1016/0896-
6273(94)90032-9
Peixoto, R. T., Kunz, P. A., Kwon, H., Mabb, A. M., Sabatini, B. L., Philpot, B. D.,
et al. (2012). Transsynaptic signaling by activity-dependent cleavage of
neuroligin-1. Neuron 76, 396–409. doi: 10.1016/j.neuron.2012.07.006
Penn, A. C., Zhang, C. L., Georges, F., Royer, L., Breillat, C., Hosy, E., et al. (2017).
Hippocampal LTP and contextual learning require surface diffusion of AMPA
receptors. Nature 549, 384–388. doi: 10.1038/nature23658
Perez de Arce, K., Schrod, N., Metzbower, S. W. R., Allgeyer, E., Kong, G. K.,
Tang, A. H., et al. (2015). Topographic mapping of the synaptic cleft into
adhesive nanodomains. Neuron 88, 1165–1172. doi: 10.1016/j.neuron.2015.
11.011
Perlson, E., Hendricks, A. G., Lazarus, J. E., Ben-Yaakov, K., Gradus, T.,
Tokito, M., et al. (2013). Dynein interacts with the neural cell adhesion
molecule (NCAM180) to tether dynamic microtubules and maintain synaptic
Frontiers in Cellular Neuroscience | www.frontiersin.org 8December 2018 | Volume 12 | Article 483
Kilinc Mechanical Regulation of Synapses
density in cortical neurons. J. Biol. Chem. 288, 27812–27824. doi: 10.1074/jbc.
M113.465088
Pokutta, S., Choi, H.-J., Ahlsen, G., Hansen, S. D., and Weis, W. I. (2014).
Structural and thermodynamic characterization of cadherin.β-catenin.α-
catenin complex formation. J. Biol. Chem. 289, 13589–13601. doi: 10.1074/jbc.
M114.554709
Pontrello, C. G., Sun, M. Y., Lin, A., Fiacco, T. A., Defea, K. A., and Ethell, I. M.
(2012). Cofilin under control of β-arrestin-2 in NMDA-dependent dendritic
spine plasticity, long-term depression (LTD), and learning. Proc. Natl. Acad.
Sci. U S A 109, E442–E451. doi: 10.1073/pnas.1118803109
Rácz, B., and Weinberg, R. J. (2013). Microdomains in forebrain spines: an
ultrastructural perspective. Mol. Neurobiol. 47, 77–89. doi: 10.1007/s12035-
012-8345-y
Rahman, T., Davies, D. S., Tannenberg, R. K., Fok, S., Shepherd, C., Dodd, P. R.,
et al. (2014). Cofilin rods and aggregates concur with tau pathology and
the development of Alzheimer’s disease. J. Alzheimers Dis. 42, 1443–1460.
doi: 10.3233/jad-140393
Rex, C. S., Chen, L. Y., Sharma, A., Liu, J., Babayan, A. H., Gall, C. M., et al.
(2009). Different Rho GTPase-dependent signaling pathways initiate sequential
steps in the consolidation of long-term potentiation. J. Cell Biol. 186, 85–97.
doi: 10.1083/jcb.200901084
Salaita, K., Nair, P. M., Petit, R. S., Neve, R. M., Das, D., Gray, J. W., et al.
(2010). Restriction of receptor movement alters cellular response: physical force
sensing by EphA2. Science 327, 1380–1385. doi: 10.1126/science.1181729
Schätzle, P., Esteves da Silva, M., Tas, R. P., Katrukha, E. A., Hu, H. Y.,
Wierenga, C. J., et al. (2018). Activity-dependent actin remodeling at the base of
dendritic spines promotes microtubule entry. Curr. Biol. 28, 2081.e6–2093.e6.
doi: 10.1016/j.cub.2018.05.004
Seong, E., Yuan, L., and Arikkath, J. (2015). Cadherins and catenins
in dendrite and synapse morphogenesis. Cell Adh. Migr. 9, 202–213.
doi: 10.4161/19336918.2014.994919
Serrels, B., Serrels, A., Brunton, V. G., Holt, M., McLean, G. W., Gray, C. H., et al.
(2007). Focal adhesion kinase controls actin assembly via a FERM-mediated
interaction with the Arp2/3 complex. Nat. Cell Biol. 9, 1046–1056.
doi: 10.1038/ncb1626
Sheng, M., and Kim, E. (2011). The postsynaptic organization of synapses. Cold
Spring Harb. Perspect. Biol. 3:a005678. doi: 10.1101/cshperspect.a005678
Singh, P., Doshi, S., Spaethling, J. M., Hockenberry, A. J., Patel, T. P., Geddes-
Klein, D. M., et al. (2012). N-methyl-D-aspartate receptor mechanosensitivity
is governed by C terminus of NR2B subunit. J. Biol. Chem. 287, 4348–4359.
doi: 10.1074/jbc.M111.253740
Spence, E. F., Kanak, D. J., Carlson, B. R., and Soderling, S. H. (2016).
The Arp2/3 complex is essential for distinct stages of spine synapse
maturation, including synapse unsilencing. J. Neurosci. 36, 9696–9709.
doi: 10.1523/JNEUROSCI.0876-16.2016
Stefen, H., Chaichim, C., Power, J., and Fath, T. (2016). Regulation of the
postsynaptic compartment of excitatory synapses by the actin cytoskeleton
in health and its disruption in disease. Neural Plast. 2016:2371970.
doi: 10.1155/2016/2371970
Suarez, F., Thostrup, P., Colman, D., and Grutter, P. (2013). Dynamics of
presynaptic protein recruitment induced by local presentation of artificial
adhesive contacts. Dev. Neurobiol. 73, 98–106. doi: 10.1002/dneu.22037
Sun, Z., Guo, S. S., and Fassler, R. (2016). Integrin-mediated
mechanotransduction. J. Cell Biol. 215, 445–456. doi: 10.1083/jcb.201609037
Suzuki, K., Hayashi, Y., Nakahara, S., Kumazaki, H., Prox, J., Horiuchi, K., et al.
(2012). Activity-dependent proteolytic cleavage of neuroligin-1. Neuron 76,
410–422. doi: 10.1016/j.neuron.2012.10.003
Sweet, E. S., Tseng, C. Y., and Firestein, B. L. (2011). To branch or not to
branch: how PSD-95 regulates dendrites and spines. Bioarchitecture 1, 69–73.
doi: 10.4161/bioa.1.2.15469
Sytnyk, V., Leshchyns’ka, I., Nikonenko, A. G., and Schachner, M. (2006). NCAM
promotes assembly and activity-dependent remodeling of the postsynaptic
signaling complex. J. Cell Biol. 174, 1071–1085. doi: 10.1083/jcb.2006
04145
Tan, Z. J., Peng, Y., Song, H. L., Zheng, J. J., and Yu, X. (2010). N-cadherin-
dependent neuron-neuron interaction is required for the maintenance of
activity-induced dendrite growth. Proc. Natl. Acad. Sci. U S A 107, 9873–9878.
doi: 10.1073/pnas.1003480107
Tatavarty, V., Das, S., and Yu, J. (2012). Polarization of actin cytoskeleton
is reduced in dendritic protrusions during early spine development in
hippocampal neuron. Mol. Biol. Cell 23, 3167–3177. doi: 10.1091/mbc.E12-
02-0165
Uchida, N., Honjo, Y., Johnson, K. R., Wheelock, M. J., and Takeichi, M.
(1996). The catenin/cadherin adhesion system is localized in synaptic junctions
bordering transmitter release zones. J. Cell Biol. 135, 767–779. doi: 10.1083/jcb.
135.3.767
Vunnam, N., and Pedigo, S. (2012). X-interface is not the explanation for the slow
disassembly of N-cadherin dimers in the apo state. Protein Sci. 21, 1006–1014.
doi: 10.1002/pro.2083
Wang, Z., Edwards, J. G., Riley, N., Provance, D. W. Jr., Karcher, R.,
Li, X. D., et al. (2008). Myosin Vb mobilizes recycling endosomes and AMPA
receptors for postsynaptic plasticity. Cell 135, 535–548. doi: 10.1016/j.cell.2008.
09.057
Weyer, S. W., Zagrebelsky, M., Herrmann, U., Hick, M., Ganss, L., Gobbert, J.,
et al. (2014). Comparative analysis of single and combined APP/APLP
knockouts reveals reduced spine density in APP-KO mice that is prevented
by APPsαexpression. Acta Neuropathol. Commun. 2:36. doi: 10.1186/2051-59
60-2-36
Wieland, J. A., Gewirth, A. A., and Leckband, D. E. (2005). Single molecule
adhesion measurements reveal two homophilic neural cell adhesion molecule
bonds with mechanically distinct properties. J. Biol. Chem. 280, 41037–41046.
doi: 10.1074/jbc.M503975200
Yuan, A., Rao, M. V., Veeranna, and Nixon, R. A. (2017). Neurofilaments and
neurofilament proteins in health and disease. Cold Spring Harb. Perspect. Biol.
9:a018309. doi: 10.1101/cshperspect.a018309
Yuan, A., Sershen, H., Veeranna, Basavarajappa, B. S., Kumar, A., Hashim, A.,
et al. (2015). Neurofilament subunits are integral components of synapses and
modulate neurotransmission and behavior in vivo.Mol. Psychiatry 20, 986–994.
doi: 10.1038/mp.2015.45
Zhang, P., Lu, H., Peixoto, R. T., Pines, M. K., Ge, Y., Oku, S., et al. (2018). Heparan
sulfate organizes neuronal synapses through neurexin partnerships. Cell 174,
1450.e23–1464.e23. doi: 10.1016/j.cell.2018.07.002
Zhu, J., Zhou, Q., Shang, Y., Li, H., Peng, M., Ke, X., et al. (2017). Synaptic targeting
and function of SAPAPs mediated by phosphorylation-dependent binding
to PSD-95 MAGUKs. Cell Rep. 21, 3781–3793. doi: 10.1016/j.celrep.2017.
11.107
Ziv, N. E., and Fisher-Lavie, A. (2014). Presynaptic and postsynaptic
scaffolds: dynamics fast and slow. Neuroscientist 20, 439–452.
doi: 10.1177/1073858414523321
Conflict of Interest Statement: The author declares that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2018 Kilinc. This is an open-access article distributed under the terms
of the Creative Commons Attribution License (CC BY). The use, distribution or
reproduction in other forums is permitted, provided the original author(s) and the
copyright owner(s) are credited and that the original publication in this journal
is cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
Frontiers in Cellular Neuroscience | www.frontiersin.org 9December 2018 | Volume 12 | Article 483
... Traditionally, NFs have been considered as structural components in the axon, albeit also present in dendrites and synaptosomes [6][7][8][9]. Although the role of NFs in signal transduction was proposed decades ago [10][11][12], a link between NF function in synapses and synaptic disorders has started to emerge only in the last years [13][14][15][16]. Indeed, NFs were identified at pre-and postsynaptic sites of excitatory and inhibitory synapses in the form of short 9-10 nm long filaments corresponding to oligomeric structures [17,18]. ...
... NFs belong to the most abundant proteins in neurons and are found in both axonal and dendritic regions. Although several studies suggest other NF functions beyond axonal structural support, and connections with psychiatric disorders are known, their role at the synapse is only beginning to emerge [13][14][15]. In the last decade, the presence of NF oligomers has been reported at postsynaptic sites, where they influence receptor function, LTP induction, and spine morphology [3,13]. ...
... The presence of longer filaments with an internexin-rich content cannot be excluded. Together with the demonstration of isoform-specific interaction with receptors at synapses (e.g., NFL and NMDA receptors, NFM, and D1 dopamine receptors [14,16,24]), this data points at isoform-specific functions of NFs within the context of the postsynapses. ...
Article
Full-text available
Neurofilaments are one of the main cytoskeletal components in neurons; they can be found in the form of oligomers at pre- and postsynapses. How their presence is regulated at the postsynapse remains largely unclear. Here we systematically quantified, by immunolabeling, the occurrence of the neurofilament isoform triplet neurofilament light (NFL), medium (NFM), and heavy (NFH) at the postsynapse using STED nanoscopy together with markers of synaptic strength and activity. Our data show that, within dendritic spines, neurofilament isoforms rarely colocalize with each other and that they are present to different extents, with NFL being the most abundant isoform. The amount of the three isoforms correlates with markers of postsynaptic strength and presynaptic activity to varying degrees: NFL shows the highest correlation to both synaptic traits, suggesting its involvement in synaptic response, while NFM exhibits the lowest correlations. By quantifying the presence of neurofilaments at the postsynapse within the context of the synaptic status, this work sheds new light on the regulation of synaptic neurofilaments and their possible contribution to synaptopathies.
... Adhesion is associated with cancer and the mechanism of cellular infiltration; however, CAMs have a much broader role in the body and are a fundamental functional construct of the brain [12,[24][25][26][27][28][29][30][31]; hence this chapter will outline the diseases and cellular processes in which the involvement of CAMs has been documented. ...
... Learning and memory-involve a vast array of CAMs [12,22,29]; therefore, we will present only selected examples here. Following Wu and Reedy [22], integrins are involved in learning and memory through a few mechanisms. ...
... Similar results were documented in mice [22]. Integrins may regulate LTP via modulating N-methyl-D-aspartic acid receptors (NMDARs) or α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) activity [29]. Furthermore, any functional changes associated with integrins result in morphological changes in a synapse structure and function [22]. ...
Article
Full-text available
The brain functions through neuronal circuits and networks that are synaptically connected. This type of connection can exist due to physical forces that interact to stabilize local contacts in the brain. Adhesion is a fundamental physical phenomenon that allows different layers, phases, and tissues to connect. Similarly, synaptic connections are stabilized by specialized adhesion proteins. This review discusses the basic physical and chemical properties of adhesion. Cell adhesion molecules (CAMs) such as cadherins, integrins, selectins, and immunoglobulin family of cell adhesion molecules (IgSF) will be discussed, and their role in physiological and pathological brain function. Finally, the role of CAMs at the synapse will be described. In addition, methods for studying adhesion in the brain will be presented.
... Key distinctions in structural features of individual glutamatergic synapses including spine size, volume (Matsuzaki et al., 2004;Arellano et al., 2007;Holtmaat and Svoboda, 2009;Bopp et al., 2017;Holler et al., 2021), and PSD area (Konur et al., 2003;Gray et al., 2006;Statman et al., 2014;Kilinc, 2018;Borczyk et al., 2019) show correlation with physiological transmission strength. Whether or not proteomic complexity at synapses including that of GluN2 synaptic heterogeneity may be causative, correlative (Antal et al., 2008;Hruska et al., 2022), or independent of PSD or synapse size (Racca et al., 2000;Shinohara and Hirase, 2009;Hruska et al., 2022), we suggest that synaptic distinctiveness, rooted in GluN2 subtype diversity, is a critical determinant in grading subthreshold glutamatergic synaptic strength. ...
Article
Full-text available
The unitary postsynaptic response to presynaptic quantal glutamate release is the fundamental basis of excitatory information transfer between neurons. The view, however, of individual glutamatergic synaptic connections in a population as homogenous, fixed-strength units of neural communication is becoming increasingly scrutinized. Here, we used minimal stimulation of individual glutamatergic afferent axons to evoke single synapse resolution postsynaptic responses from central sensory lamina I neurons in an ex vivo adult rat spinal slice preparation. We detected unitary events exhibiting a NMDA receptor component with distinct kinetic properties across synapses conferred by specific GluN2 subunit composition, indicative of GluN2 subtype-based postsynaptic heterogeneity. GluN2A, 2A and 2B, or 2B and 2D synaptic predominance functioned on distinct lamina I neuron types to narrowly, intermediately, or widely tune, respectively, the duration of evoked unitary depolarization events from resting membrane potential, which enabled individual synapses to grade differentially depolarizing steps during temporally patterned afferent input. Our results lead to a model wherein a core locus of proteomic complexity prevails at this central glutamatergic sensory synapse that involves distinct GluN2 subtype configurations. These findings have major implications for subthreshold integrative capacity and transmission strength in spinal lamina I and other CNS regions.
... Also, throughout their lifetime neurons are exposed to heterogeneous stiffness and several mechanical cues, which, in addition to modulating neuronal differentiation and development, likely influence the plasticity of neuronal circuits. Indeed, mechanical force is emerging as a key factor for the regulation of axon guidance, growth, synapse formation, and plasticity [27][28][29][30] . Several studies have suggested that synaptic changes driven by activity involve dynamic cell interactions, which are mediated by molecules coupled to the cytoskeleton and governed by trans-synaptic forces 25,[31][32][33][34][35] . ...
Article
Full-text available
Wired neurons form new presynaptic boutons in response to increased synaptic activity, however the mechanism(s) by which this occurs remains uncertain. Drosophila motor neurons (MNs) have clearly discernible boutons that display robust structural plasticity, being therefore an ideal system in which to study activity-dependent bouton genesis. Here, we show that in response to depolarization and in resting conditions, MNs form new boutons by membrane blebbing, a pressure-driven mechanism that occurs in 3-D cell migration, but to our knowledge not previously described to occur in neurons. Accordingly, F-actin is decreased in boutons during outgrowth, and non-muscle myosin-II is dynamically recruited to newly formed boutons. Furthermore, muscle contraction plays a mechanical role, which we hypothesize promotes bouton addition by increasing MN confinement. Overall, we identified a mechanism by which established circuits form new boutons allowing their structural expansion and plasticity, using trans-synaptic physical forces as the main driving force.
... Key distinctions in structural features of individual glutamatergic synapses including spine size, volume [66][67][68][69][70] , and PSD area [71][72][73][74][75] show correlation with physiological transmission strength. Whether or not proteomic complexity at synapses including that of GluN2 synaptic heterogeneity may be causative, correlative, or independent of PSD or synapse size, we suggest that synaptic distinctiveness, rooted in GluN2 subtype diversity, is a critical determinant in grading subthreshold glutamatergic synaptic strength. ...
Preprint
The unitary postsynaptic response to presynaptic quantal glutamate release is the fundamental basis of excitatory information transfer between neurons. The view, however, of individual glutamatergic synaptic connections in a population as homogenous, fixed-strength units of neural communication is becoming increasingly scrutinized. Here, we used minimal stimulation of individual glutamatergic afferent axons to evoke single synapse resolution postsynaptic responses from central sensory lamina I neurons in an ex vivo adult rat spinal slice preparation. We detected unitary events exhibiting a NMDA receptor component with distinct kinetic properties across synapses conferred by specific GluN2 subunit composition, indicative of GluN2 subtype-based postsynaptic heterogeneity. GluN2A, 2A and 2B, or 2B and 2D synaptic predominance functioned on distinct lamina I neuron types to narrowly, intermediately, or widely tune, respectively, the duration of evoked unitary depolarization events from resting membrane potential, which enabled individual synapses to grade differentially depolarizing steps during temporally-patterned afferent input. Our results lead to a model wherein a core locus of proteomic complexity prevails at this central glutamatergic sensory synapse that involves distinct GluN2 subtype configurations. These findings have major implications for subthreshold integrative capacity and transmission strength in spinal lamina I and other CNS regions.
Article
The ability of neurons to rapidly remodel their synaptic structure and strength in response to neuronal activity is highly conserved across species and crucial for complex brain functions. However, mechanisms required to elicit and coordinate the acute, activity-dependent structural changes across synapses are not well understood, as neurodevelopment and structural plasticity are tightly linked. Here, using an RNAi screen in Drosophila against genes affecting nervous system functions in humans, we uncouple cellular processes important for synaptic plasticity and synapse development. We find mutations associated with neurodegenerative and mental health disorders are 2-times more likely to affect activity-induced synaptic remodeling than synapse development. We report that while both synapse development and activity-induced synaptic remodeling at the fly NMJ require macroautophagy (hereafter referred to as autophagy), bifurcation in the autophagy pathway differentially impacts development and synaptic plasticity. We demonstrate that neuronal activity enhances autophagy activation but diminishes degradative autophagy, thereby driving the pathway towards autophagy-based secretion. Presynaptic knockdown of Snap29, Sec22, or Rab8, proteins implicated in the secretory autophagy pathway, is sufficient to abolish activity-induced synaptic remodeling. This study uncovers secretory autophagy as a transsynaptic signaling mechanism modulating synaptic plasticity.
Article
Strumpellin/Wiskott-Aldrich syndrome protein and SCAR homologue (WASH) complex subunit 5 (WASHC5) is a core component of the WASH complex, and its mutations confer pathogenicity for hereditary spastic paraplegia (HSP) type SPG8, a rare neurodegenerative gait disorder. WASH complex activates actin-related protein-2/3-mediated actin polymerization and plays a pivotal role in intracellular membrane trafficking in endosomes. In this study, we examined the role of strumpellin in the regulation of structural plasticity of cortical neurons involved in gait coordination. Administration of a lentivirus containing a strumpellin-targeting short hairpin RNA (shRNA) to cortical motor neurons lead to abnormal motor coordination in mice. Strumpellin knockdown using shRNA attenuated dendritic arborization and synapse formation in cultured cortical neurons, and this effect was rescued by wild-type strumpellin expression. Compared with the wild-type, strumpellin mutants N471D or V626F identified in patients with SPG8 exhibited no differences in rescuing the defects. Moreover, the number of F-actin clusters in neuronal dendrites was decreased by strumpellin knockdown and rescued by strumpellin expression. In conclusion, our results indicate that strumpellin regulates the structural plasticity of cortical neurons via actin polymerization.
Article
Do dendritic spines, which comprise the postsynaptic component of most excitatory synapses, exist only for their structural dynamics, receptor trafficking, and chemical and electrical compartmentation? The answer is no. Simultaneous investigation of both spine and presynaptic terminals has recently revealed a novel feature of spine synapses. Spine enlargement pushes the presynaptic terminals with muscle-like force and augments the evoked glutamate release for up to 20 min. We now summarize the evidence that such mechanical transmission shares critical features in common with short-term potentiation (STP) and may represent the cellular basis of short-term and working memory. Thus, spine synapses produce the force of learning to leave structural traces for both short and long-term memories.
Article
Full-text available
Synapses are fundamental units of communication in the brain. The prototypical synapse-organizing complex neurexin-neuroligin mediates synapse development and function and is central to a shared genetic risk pathway in autism and schizophrenia. Neurexin's role in synapse development is thought to be mediated purely by its protein domains, but we reveal a requirement for a rare glycan modification. Mice lacking heparan sulfate (HS) on neurexin-1 show reduced survival, as well as structural and functional deficits at central synapses. HS directly binds postsynaptic partners neuroligins and LRRTMs, revealing a dual binding mode involving intrinsic glycan and protein domains for canonical synapse-organizing complexes. Neurexin HS chains also bind novel ligands, potentially expanding the neurexin interactome to hundreds of HS-binding proteins. Because HS structure is heterogeneous, our findings indicate an additional dimension to neurexin diversity, provide a molecular basis for fine-tuning synaptic function, and open therapeutic directions targeting glycan-binding motifs critical for brain development.
Article
Full-text available
Synaptic adhesion molecules not only provide a physical link between pre- and post-synaptic membranes, but also contribute to synaptic differentiation and plasticity by organizing functional elements, in particular neurotransmitter receptors. The wealth of existing adhesive protein families including many isoforms and splice variants, calls for systematic identification of the levels and exchange rates of each of those protein members at specific synapse types. Complementary to electron microscopy to identify individual synaptic contacts and biochemistry to reveal protein-protein interactions, recent super-resolution light microscopy methods combined with appropriate fluorescent labeling provide a way to measure the dynamics and sub-micron organization of selective molecular components, and their inter-relations at the synapse. In this review, we summarize current knowledge on the dynamics, nanoscale localization, and function of key synaptic adhesion complexes.
Article
Full-text available
The PSD-95/SAPAP/Shank complex functions as the major scaffold in orchestrating the formation and plasticity of the post-synaptic densities (PSDs). We previously demonstrated that the exquisitely specific SAPAP/Shank interaction is critical for Shank synaptic targeting and Shank-mediated synaptogenesis. Here, we show that the PSD-95/SAPAP interaction, SAPAP synaptic targeting, and SAPAP-mediated synaptogenesis require phosphorylation of the N-terminal repeat sequences of SAPAPs. The atomic structure of the PSD-95 guanylate kinase (GK) in complex with a phosphor-SAPAP repeat peptide, together with biochemical studies, reveals the molecular mechanism underlying the phosphorylation-dependent PSD-95/SAPAP interaction, and it also provides an explanation of a PSD-95 mutation found in patients with intellectual disabilities. Guided by the structural data, we developed potent non-phosphorylated GK inhibitory peptides capable of blocking the PSD-95/SAPAP interaction and interfering with PSD-95/SAPAP-mediated synaptic maturation and strength. These peptides are genetically encodable for investigating the functions of the PSD-95/SAPAP interaction in vivo.
Article
Full-text available
Rewiring of synaptic circuitry pertinent to memory formation has been associated with morphological changes in dendritic spines and with extracellular matrix (ECM) remodeling. Here, we mechanistically link these processes by uncovering a signaling pathway involving the serotonin 5-HT7 receptor (5-HT7R), matrix metalloproteinase 9 (MMP-9), the hyaluronan receptor CD44, and the small GTPase Cdc42. We highlight a physical interaction between 5-HT7R and CD44 (identified as an MMP-9 substrate in neurons) and find that 5-HT7R stimulation increases local MMP-9 activity, triggering dendritic spine remodeling, synaptic pruning, and impairment of long-term potentiation (LTP). The underlying molecular machinery involves 5-HT7R-mediated activation of MMP-9, which leads to CD44 cleavage followed by Cdc42 activation. One important physiological consequence of this interaction includes an increase in neuronal outgrowth and elongation of dendritic spines, which might have a positive effect on complex neuronal processes (e.g., reversal learning and neuronal regeneration).
Article
In neurons, microtubules form dense bundles and run along the length of axons and dendrites. Occasionally, dendritic microtubules can grow from the shaft directly into dendritic spines. Microtubules target dendritic spines that are undergoing activity-dependent changes, but the mechanism by which microtubules enter spines has remained poorly understood. Using live-cell imaging, high-resolution microscopy, and local glutamate uncaging, we show that local actin remodeling at the base of a spine promotes microtubule spine targeting. Microtubule spine entry is triggered by activation of N-Methyl-D-aspartic acid (NMDA) receptors and calcium influx and requires dynamic actin remodeling. Activity-dependent translocation of the actin remodeling protein cortactin out of the spine correlates with increased microtubule targeting at a single spine level. Our data show that the structural changes in the actin cytoskeleton at the base of the spine are directly involved in microtubule entry and emphasize the importance of actin-microtubule crosstalk in orchestrating synapse function and plasticity.
Article
We review the use of luminescent nanoparticles in super-resolution imaging and single-molecule tracking, and showcase novel approaches to super-resolution imaging that leverage the brightness, stability, and unique optical-switching properties of these nanoparticles. We also discuss the challenges associated with their use in biological systems, including intracellular delivery and molecular targeting. In doing so, we hope to provide practical guidance for biologists and continue to bridge the fields of super-resolution imaging and nanoparticle engineering to support their mutual advancement.
Article
Dendritic filopodia select synaptic partner axons by interviewing the cell surface of potential targets, but how filopodia decipher the complex pattern of adhesive and repulsive molecular cues to find appropriate contacts is unknown. Here, we demonstrate in cortical neurons that a single cue is sufficient for dendritic filopodia to reject or select specific axonal contacts for elaboration as synaptic sites. Super-resolution and live-cell imaging reveals that EphB2 is located in the tips of filopodia and at nascent synaptic sites. Surprisingly, a genetically encoded indicator of EphB kinase activity, unbiased classification, and a photoactivatable EphB2 reveal that simple differences in the kinetics of EphB kinase signaling at the tips of filopodia mediate the choice between retraction and synaptogenesis. This may enable individual filopodia to choose targets based on differences in the activation rate of a single tyrosine kinase, greatly simplifying the process of partner selection and suggesting a general principle.
Article
Hippocampal CA3 neurons form synapses with CA1 neurons in two layers, stratum oriens (SO) and stratum radiatum (SR). Each layer develops unique synaptic properties but molecular mechanisms that mediate these differences are unknown. Here, we show that SO synapses normally have significantly more mushroom spines and higher-magnitude long-term potentiation (LTP) than SR synapses. Further, we discovered that these differences require the Type II classic cadherins, cadherins-6, -9, and -10. Though cadherins typically function via trans-cellular homophilic interactions, our results suggest presynaptic cadherin-9 binds postsynaptic cadherins-6 and -10 to regulate mushroom spine density and high-magnitude LTP in the SO layer. Loss of these cadherins has no effect on the lower-magnitude LTP typically observed in the SR layer, demonstrating that cadherins-6, -9, and -10 are gatekeepers for high-magnitude LTP. Thus, Type II cadherins may uniquely contribute to the specificity and strength of synaptic changes associated with learning and memory.
Article
Long-term potentiation (LTP) of excitatory synaptic transmission has long been considered a cellular correlate for learning and memory. Early LTP (less than 1 h) had initially been explained either by presynaptic increases in glutamate release or by direct modification of postsynaptic AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor function. Compelling models have more recently proposed that synaptic potentiation can occur by the recruitment of additional postsynaptic AMPA receptors (AMPARs), sourced either from an intracellular reserve pool by exocytosis or from nearby extra-synaptic receptors pre-existing on the neuronal surface. However, the exact mechanism through which synapses can rapidly recruit new AMPARs during early LTP remains unknown. In particular, direct evidence for a pivotal role of AMPAR surface diffusion as a trafficking mechanism in synaptic plasticity is still lacking. Here, using AMPAR immobilization approaches, we show that interfering with AMPAR surface diffusion markedly impairs synaptic potentiation of Schaffer collaterals and commissural inputs to the CA1 area of the mouse hippocampus in cultured slices, acute slices and in vivo. Our data also identify distinct contributions of various AMPAR trafficking routes to the temporal profile of synaptic potentiation. In addition, AMPAR immobilization in vivo in the dorsal hippocampus inhibited fear conditioning, indicating that AMPAR diffusion is important for the early phase of contextual learning. Therefore, our results provide a direct demonstration that the recruitment of new receptors to synapses by surface diffusion is a critical mechanism for the expression of LTP and hippocampal learning. Since AMPAR surface diffusion is dictated by weak Brownian forces that are readily perturbed by protein-protein interactions, we anticipate that this fundamental trafficking mechanism will be a key target for modulating synaptic potentiation and learning.
Article
Neurofilaments (NFs) are unique among tissue-specific classes of intermediate filaments (IFs) in being heteropolymers composed of four subunits (NF-L [neurofilament light]; NF-M [neuro-filament middle]; NF-H [neurofilament heavy]; and a-internexin or peripherin), each having different domain structures and functions. Here, we review how NFs provide structural support for the highly asymmetric geometries of neurons and, especially, for the marked radial expansion of myelinated axons crucial for effective nerve conduction velocity. NFs in axons extensively cross-bridge and interconnect with other non-IF components of the cytoskeleton, including microtubules, actin filaments, and other fibrous cytoskeletal elements, to establish a regionally specialized network that undergoes exceptionally slow local turnover and serves as a docking platform to organize other organelles and proteins. We also discuss how a small pool of oligomeric and short filamentous precursors in the slow phase of axonal transport maintains this network. A complex pattern of phosphorylation and dephosphorylation events on each subunit modulates filament assembly, turnover, and organization within the axonal cytoskeleton. Multiple factors, and especially turnover rate, determine the size of the network, which can vary substantially along the axon. NF gene mutations cause several neuroaxonal disorders characterized by disrupted subunit assembly and NF aggregation. Additional NF alterations are associated with varied neuropsychiatric disorders. New evidence that subunits of NFs exist within postsynaptic terminal boutons and influence neurotransmission suggests how NF proteins might contribute to normal synaptic function and neuropsychiatric disease states. © 2017 Cold Spring Harbor Laboratory Press; all rights reserved.