Dynamin Superfamily at Pre - and Postsynapses

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

974313

review-article2020
NROXXX10.1177/1073858420974313The NeuroscientistArriagada-Diaz et al.

Review
The Neuroscientist

Dynamin Superfamily at Pre- and


1­–18
© The Author(s) 2020
Article reuse guidelines:
Postsynapses: Master Regulators of sagepub.com/journals-permissions
DOI: 10.1177/1073858420974313
https://doi.org/10.1177/1073858420974313

Synaptic Transmission and Plasticity journals.sagepub.com/home/nro

in Health and Disease

Jorge Arriagada-Diaz1,2*, Lorena Prado-Vega1,2*, Ana M. Cárdenas Díaz1,


Alvaro O. Ardiles1,3,4, and Arlek M. Gonzalez-Jamett1

Abstract
Dynamin superfamily proteins (DSPs) comprise a large group of GTP-ases that orchestrate membrane fusion and
fission, and cytoskeleton remodeling in different cell-types. At the central nervous system, they regulate synaptic vesicle
recycling and signaling-receptor turnover, allowing the maintenance of synaptic transmission. In the presynapses, these
GTP-ases control the recycling of synaptic vesicles influencing the size of the ready-releasable pool and the release of
neurotransmitters from nerve terminals, whereas in the postsynapses, they are involved in AMPA-receptor trafficking
to and from postsynaptic densities, supporting excitatory synaptic plasticity, and consequently learning and memory
formation. In agreement with these relevant roles, an important number of neurological disorders are associated with
mutations and/or dysfunction of these GTP-ases. Along the present review we discuss the importance of DSPs at
synapses and their implication in different neuropathological contexts.

Keywords
endocytic synaptic vesicle recycling, neurosecretion, AMPAR trafficking, synaptic plasticity, excitatory synapses,
dendritic spines, synaptopathies, dynamin superfamily, dynamin GTP-ase activity

Introduction 2020) and microtubule-instability (Hamao and others


2009; Ishida and others 2011; Maeda and others 1992).
Dynamin superfamily proteins (DSPs) are mechano- In mammals, classical dynamins are encoded by
enzymes with GTP-ase activity that catalyze membrane three genes: DNM1, DNM2, and DNM3 located in
remodeling (Antonny and others 2016; Ferguson and De chromosomes 9 (Newman-Smith and others 1997), 19
Camilli 2012; Gonzalez-Jamett and others 2013b; (Zuchner and others 2005), and 1 (Noakes and others
Gonzalez-Jamett and others 2014; Praefcke and McMahon 1999), respectively. These genes can be subjected to
2004; Singh and others 2018) during different cellular alternative splicing, producing each one from four to
processes including clathrin-mediated endocytosis (CME) thirteen spliced variants (Cao and others 1998). These
(Damke and others 1994; Loerke and others 2009;
Rappoport and Simon 2003; van der Bliek and others
1993; Warnock and Schmid 1996), caveolae internaliza- 1
Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad
tion (Henley and others 1998), exocytosis (Gonzalez- de Valparaíso, Valparaíso, Chile
2
Jamett and others 2010; Gonzalez-Jamett and others Programa de Magister en Ciencias, mención Neurociencia,
2013a; Wong and others 2015), endosomal recycling Universidad de Valparaíso, Valparaíso, Chile
3
Centro de Neurología Traslacional, Facultad de Medicina,
(Nicoziani and others 2000), post-trans-Golgi network Universidad de Valparaíso, Valparaíso, Chile
transportation of secretory vesicles to the plasma mem- 4
Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina,
brane (Jones and others 1998; Kreitzer and others 2000) Universidad de Valparaíso, Viña del Mar, Chile
and mitochondria fusion and fission (Mears and others *These authors contributed equally to this work.
2011). In addition, DSPs are involved in membrane-inde-
pendent processes such as actin cytoskeleton dynamics Corresponding Author:
Arlek M. Gonzalez-Jamett, Centro Interdisciplinario de Neurociencia
(Gonzalez-Jamett and others 2013a; Gonzalez-Jamett de Valparaíso, Universidad de Valparaíso, Gran Bretaña 1111 Playa
and others 20117; Gu and others 2010; Mooren and oth- Ancha, Valparaiso, 2340000, Chile.
ers 2009; Yamada and others 2013; Zhang and others Email: arlek.gonzalez@cinv.cl
2 The Neuroscientist 00(0)

Figure 1.  Structure and assembly of dynamin superfamily proteins (DSPs). (A) Domains of classical dynamins and dynamin-
related proteins are schematized. Classical dynamins are composed by a GTP-ase domain, a middle domain, a pleckstrin
homology domain (PHD), a GTP-ase effector domain (GED), and a proline-rich domain (PRD). Dynamin-related proteins lack
PHD and PRD but include an “insert” variable sequence. The “bundle signaling element” (BSE) formed by helices from GTP-ase
and GED, together with the “stalk” region formed by helices from MD and GED mediate GTP-dependent dynamin self-assembly.
(B) Schematic representations of dimers and tetramers showing the interfaces required for dynamin’s oligomerization.

three dynamin isoforms exhibit different expression pat- and others 2014; Praefcke and McMahon 2004; Singh
terns. While dynamin-1 is exclusively expressed in neu- and others 2018). Three helices located on the N- and
ronal tissues, dynamin-2 is ubiquitous, and dynamin-3 C-terminus of GD together with a C-terminus helix
is present in brain, heart, testis, and lungs (Ferguson and from GED form the “bundle signaling element” (BSE)
De Camilli 2012; Gonzalez-Jamett and others 2014; (Chappie and others 2009). Helices from the MD and
Singh and others 2018). As outlined in Figure 1A, clas- the amino-terminal portion of GED constitute the
sic dynamins are composed by five conserved domains: “stalk” region (Antonny and others 2016; Chappie and
an amino-terminal catalytic GTP-ase domain (GD), a others 2009; Ford and others 2011) (Fig. 1) The BSE
middle domain (MD) required for their self-assembly, a seems to sense and convey GTP-dependent conforma-
phosphoinositides-binding pleckstrin homology domain tional changes from the GD to the stalk region, control-
(PHD), a GTP-ase-effector domain (GED), and a car- ling dynamin’s activity (Chappie and others 2009). In
boxy-terminal proline- and arginine-rich domain (PRD) turn, depending on the information received from the
that binds SH3 domain-containing partners (Antonny BSE, the stalk region mediates dynamin oligomeriza-
and others 2016; Ferguson and De Camilli 2012; tion (Wenger and others 2013). In this regard, stalks
Gonzalez-Jamett and others 2013a; Gonzalez-Jamett from neighboring monomers interact in an extensive
Arriagada-Diaz et al. 3

interface (interface 2) forming a cross-shaped dimer dementia (Mulugeta and others 2014), and sporadic forms
with their GDs linked to one side of the cross, and their of Alzheimer’s disease (AD) (Aidaralieva and others
PHDs linked to the other side (Chappie and others 2011; 2008; Baek and others 2017; Kamagata and others 2009;
Faelber and others 2011; Kong and others 2018) (Fig. Kandimalla and others 2017).
1B) In the absence of lipids, dynamins exist predomi- In the current review, we discuss the importance of
nantly as tetramers, which are also stabilized by stalk- DSPs at central synapses, with special attention on their
localized interfaces (1 and 3). While interface 1 is at the roles at the pre- and postsynaptic compartments and their
tips of interacting stalks, proximal to GD and BSE, implication in neuropathological contexts.
interface 3 is at the distal membrane–facing ends of the
interacting stalks (Chappie and others 2011; Faelber and Presynaptic Dynamins and Synaptic
others 2011; Ford and others 2011; Kong and others
2018) (Fig. 1B). Vesicle Recycling
In the presence of lipids dynamins form higher order The endocytic recycling of SV that compensates
helical and ring-like structures (Hinshaw and Schmid SV-exocytosis is critical for keeping constant the surface
1995) that have up to 100-fold higher GTP-ase activity membrane area and ensuring neurotransmission efficiency
(Warnock and Schmid 1996). In a GTP hydrolysis (Cardenas and Marengo 2010). Dynamins’ participation
dependent manner, dynamin-oligomers constrict, favor- in SV-recycling was first demonstrated in the neuromus-
ing membrane tubulation and scission (Antonny and cular junction of the Drosophila shibire mutant (Chen and
others 2016). Factors such as low ionic strength others 1991), which expresses a mutation in a dynamin
(Hinshaw and Schmid 1995) or the presence of negative orthologue that affects its catalytic activity (Chen and oth-
tubular templates such as lipid membranes (Lin and oth- ers 1991; Kawasaki and others 2000). Shibire is a temper-
ers 1997; Salim and others 1996), microtubules (Maeda ature-sensitive phenotype in Drosophila that results in
and others 1992) and actin bundles (Gu and others 2010; locomotion alterations and paralysis at nonpermissive
Mooren and others 2009) have shown to potentiate temperatures (>30°C) (Poodry and Edgar 1979). The lat-
dynamin oligomerization promoting its catalytic activ- ter is consequence of the inability of endocytic pits to
ity and membrane fission property. Besides classical achieve the fission process, leading to aberrant accumula-
dynamins, DSPs also include dynamin-related proteins tion of omega-shaped structures at the presynaptic zone
(DRPs) which lack PHD and/or PRD (Williams and and depletion of SV at nerve terminals (Koenig and Ikeda
Kim 2014) but exhibit important structural (Fig. 1A) 1989). Such defect is more evident on high-frequency
and functional similarities. In fact, DRPs are predicted stimulation, when a continuous recycling of SV is critical
to carry out membrane tubulation and scission in a simi- for maintaining the synaptic transmission. Indeed, in teta-
lar manner to the classical dynamins, exerting important nized shibire-neuromuscular junctions, synaptic currents
roles in intracellular trafficking (Bonekamp and others exhibit a constant decline until completely cease due to
2010), endocytosis and mitochondria fusion and seg- depletion of the SV ready-releasable pool (RRP) (Delgado
mentation (Mears and others 2011). and others 2000). These observations support the impor-
In the nervous system, different types of DSPs are tance of the dynamins’ catalytic activity in maintaining
expressed at the pre- and postsynaptic levels and partici- SV availability and have been confirmed by a consider-
pate in several processes modulating synaptic transmis- able number of reports in the two last decades in both
sion. In presynaptic compartments, classical dynamins peripheral and central synapses (Cheung and Cousin
play a key function supporting synaptic vesicle (SV) 2019; Clark and others 1997; Ferguson and others 2007;
recycling (Ferguson and others 2007; Raimondi and oth- Hayashi and others 2008; Linares-Clemente and others
ers 2011; Tanifuji and others 2013) and they appear to 2015; Lu and others 2009; Newton and others 2006;
exert differential roles in response to different patterns of Raimondi and others 2011; Singh and others 2018;
neuronal activity (Tanifuji and others 2013). At the post- Tanifuji and others 2013; Yamashita and others 2005).
synaptic level, they regulate the surface availability of However, it is not entirely clear whether different dyna-
neurotransmitter receptors by tuning receptor turnover min isoforms equally participate in SV endocytic recy-
(Carroll and others 1999; Kabbani and others 2004). cling mechanisms or they have differential roles.
Consistently, dysfunctions of DSPs associate with a con- Electron microscopy studies from De Camilli’s labo-
siderable number of peripheral (PNS) and central nervous ratory demonstrated that the knock-out (KO) of dyna-
system (CNS) disorders, including Charcot-Marie-Tooth min-1 produces accumulation of large clathrin-coated
neuropathy (Zuchner and others 2005), centronuclear structures at the nerve terminals of mouse primary cul-
myopathy (Bitoun and others 2005; Bitoun and others tured cortical neurons, which associate with impairment
2009), encephalopathy and epilepsy (Li and others 2015; of the inhibitory synaptic transmission(Ferguson and oth-
von Spiczak and others 2017), stroke and vascular ers 2007). A later report showed that the structural
4 The Neuroscientist 00(0)

changes produced by the absence of dynamin-1 are more


evident at the nerve terminals of γ-aminobutyric acid
(GABA)-ergic synapses (Hayashi and others 2008), sug-
gesting that CME is a predominant SV-recycling mecha-
nism in inhibitory neurons upon tonic activity. Since
inhibitory synapses are importantly modulated by tonic
activity, they require an efficient endocytic machinery
and likely are affected by the absence of dynamin-1 on
basal synaptic transmission (Evergren and others 2006).
Unlike that observed in GABA-ergic synapses, excitatory
transmission seems to efficiently operate on spontaneous
neuronal activity. In fact as shown by Ferguson and col-
laborators, although SV-endocytosis is severely impaired
during strong stimulation it is restored once stimulation
ceases, suggesting that dynamin 1–independent mecha-
nisms support SV recycling on tonic activity in excitatory
synapses (Ferguson and others 2007). One possibility is
that other dynamin isoforms compensate the absence of
dynamin-1. In agreement with this idea, Ferguson and
coworkers showed that the overexpression of dynamin-2
and dynamin-3 partially or completely rescue, respec- Figure 2.  Schematic diagram of the role of dynamin
tively, impairments in stimulus-induced SV-recycling superfamily proteins (DSPs) in the presynaptic mechanisms
(Ferguson and others 2007). The generation of a dyna- supporting neurotransmission. In response to different
min-1/dynamin-3 double KO mouse (dKO) provided action potential (AP) firing rates, classical dynamin isoforms
are differentially recruited to mediate SV recycling. On
additional evidence in favor of this idea, as dKO mice are
high-frequency stimulation, dynamin-1 (DYN1) acts with a
still able to develop efficient synapses and to recycle SV fast kinetics to resupply SV to the RRP (brown arrow) in a
(Raimondi and others 2011). However, some aberrant large window time, greater than 50 ms. Independent of the
mechanisms seem to operate in such condition. In this stimulation pattern, dynamin-3 (DYN3) works with slow
sense, Lou and collaborators demonstrated that hippo- kinetics (orange arrow), but in a faster time window on the
campal neurons from dynamin 1/3 dKO mice exhibit AP arrival (20 ms). Dynamin-2 (DYN2) acts in an intermediate
defective CME of SV, accumulation of arrested clathrin- way, responding quickly after the AP-triggering during high
coated pits, reduction in the RRP-size and impairments in frequency neuronal activity (Tanifuji and others 2013).
Mitochondria segmentation mediated by DRP1 modulates the
excitatory postsynaptic currents (Lou and others 2012). SV recycling by regulating mobilization from RP to RRP. GTP-
Moreover, dKO neurons express a strong facilitation of ase activity of DSPs also regulates quantal size and kinetics of
the post-synaptic response, which is accompanied by neurotransmitter release.
lower release probability in the absence of dynamin-1
and dynamin-3 (Lou and others 2012). Thus, the absence
of both dynamin-1 and dynamin-3 significantly compro- mediates SV-recycling in response to high frequency
mises the SV maintenance, even though they can be neuronal stimulation, acting quickly after AP-triggering
replaceable to some extent by dynamin-2. and allowing a fast RRP resupply (Fig. 2) (Tanifuji and
In 2013, experiments from Mochida’s laboratory others 2013). These data suggest that the three classical
demonstrated that the role of different dynamin isoforms dynamins are differentially required on specific patterns
in SV recycling depends on neuronal activity. Specific of neuronal activity, being able to participate in different
knock-down of each classical dynamin isoform in rat endocytic mechanisms that refill the SV population at
superior cervical ganglion neurons showed that the the nerve terminals. In this regard, RRP and RP (the
replenishment of RRP is mediated by dynamin-1, dyna- reserve pool mobilized during intense neuronal activity)
min-2, or dynamin-3 with different kinetics, as a func- (Richards and others 2003) have shown to be maintained
tion of the action potential (AP)–firing pattern. On by different endocytosis modes. This is how, during
high-frequency stimulation, dynamin-1 mediates SV sparse activity, an “ultrafast endocytosis” (UFE) is trig-
recycling with a fast kinetics in a slow window time gered to rapidly restore the surface area (Watanabe and
greater than 50 ms (Fig. 2), while dynamin-3 operates others 2013; Watanabe and others 2014). The most
independent of the AP-firing pattern with slow kinetics understood CME appears to be triggered during mild
but in a faster time window within 20 ms of the incoming periods of neuronal activity (Granseth and others 2006).
AP (Fig. 2) (Tanifuji and others 2013). Dynamin-2 Remarkably, both UFE and CME have shown to rapidly
Arriagada-Diaz et al. 5

saturate on high frequency of AP-firings (Clayton and three dynamin-isoforms have a differential participation
Cousin 2008; Granseth and others 2006; Soykan and oth- in the presynaptic homeostatic mechanisms that operate
ers 2017) making “bulk-endocytosis” relevant (Clayton in response to neuronal silencing (Calabrese and Halpain
and Cousin 2008; Hayashi and others 2008; Wenzel and 2015).
others 2012; Wu and others 2014). This mechanism An important process that has been shown to be rele-
relies on the formation of large endocytic structures vant in the presynaptic mechanisms of synaptic transmis-
named “bulk endosomes,” from which SV are subse- sion is mitochondrial dynamics (Ma and others 2009).
quently generated (Kokotos and Cousin 2015). It has Mitochondria are the main regulators of the cellular
been suggested that both, UFE and CME, directly refill metabolism, which becomes critical in neurons to support
the RRP (Granseth and others 2006; Watanabe and oth- the high energy demand during synaptic transmission and
ers 2014), whereas bulk endocytosis contributes to the plasticity (Mattson 2007; Todorova and Blokland 2017).
SV maintenance by replenishing the RP (Cheung and Mitochondrial dynamics includes fission, fusion, and
Cousin 2019). As demonstrated by Watanabe and col- motility and, together with Ca2+ signaling, finely modu-
laborators, UFE depends on dynamin GTP-ase activity lates mitochondrial functions (Mishra and Chan 2016). A
and actin-cytoskeleton polymerization, and unlike CME, set of DRPs regulate mitochondrial dynamics. Mitofusins
it occurs adjacent to the active zones at a rate 200 times (Mfn1/2) and optic atrophy-1 (Opa1) are required for
faster than CME in hippocampal synapses (Watanabe fusion of the outer and inner mitochondrial membrane,
and others 2013). Wu and collaborators in 2014 reported respectively, whereas dynamin-related protein 1 (DRP1)
that in dynamin-1/3 dKO neurons, in which CME is sig- and the adaptors mitochondrial fission protein 1 (Fis1),
nificantly impaired (Lou and others 2012), the formation and mitochondrial fission factor (MFF) regulate mito-
of bulk endosomes is unaffected (Wu and others 2014). chondrial segmentation (Fig. 2) (Tilokani and others
The latter suggests that bulk endocytosis requires neither 2018). Fission/fusion imbalance severely compromises
dynamin-1 nor dynamin-3, opening up the possibility mitochondrial function, affecting neuronal activity and
that dynamin-2 mediates this mechanism. In fact, previ- synaptic transmission (Wu and others 2019). In this
ous reports confirmed the occurrence of dynamin-1-in- regard, DRP1 plays a pivotal role. In 2014, Berthet and
dependent forms of bulk endocytosis in cultured colleagues demonstrated that the deletion of DRP1 in
hippocampal neurons in response to intense stimulation midbrain dopaminergic neurons leads to axonal mito-
(Hayashi and others 2008). Since dynamin-2 quickly chondria depletion, axonal degeneration and neuronal
orchestrates SV-recycling on high-frequency neuronal death (Berthet and others 2014) highlighting the impor-
stimulation (Tanifuji and others 2013), it is feasible that tance of mitochondria segmentation for neuronal mor-
this isoform mediates the formation of bulk endosomes. phology and survival. Furthermore, DRP1 knock-out
Interestingly, it was recently demonstrated that dyna- (DRP1KO) neurons exhibit aberrant large mitochondria
min-1 is also necessary for bulk endocytosis but, rather and decreased levels of mitochondrial-derived ATP on
than participate in the formation of bulk endosomes, it enhanced neuronal activity (Shields and others 2015).
appears to be important for SV generation from bulk- Such defects are evident at nerve terminals but not at
endosomes (Cheung and Cousin 2019). This latter mech- soma and correlate with impaired SV-recycling and trans-
anism is compatible with a role of both, dynamin-2 mission (Shields and others 2015). How the absence of
mediating the formation of bulk endosomes, and dyna- DRP1 could affect the resupply of SV? As demonstrated
min-1 mediating the SV formation from bulk endosomes in Drosophila neuromuscular junctions, SV-mobilization
during high frequency stimulation patterns. However, from RP to RRP on intense neuronal activity depends on
the participation of these and other dynamin-isoforms in synaptic DRP1 and on mitochondrial ATP-production
SV bulk endocytosis needs to be further investigated. (Verstreken and others 2005). In fact, DRP1 depletion in
The truth is that DSPs seem to be differentially hippocampal neurons delays the recovery of the RRP on
recruited, participating in specific presynaptic mecha- high-frequency stimulation (Li and others 2013) pointing
nisms that depend on neuronal activity. In this regard, its involvement in the presynaptic mechanisms that sup-
Calabrese and Halpain (2015) demonstrated that changes port synaptic transmission. Along the same line, Singh
in the AP-firing pattern affect the presynaptic clustering and collaborators, reported that the presynaptic removal
of dynamin-isoforms. Both dynamin-1 and dynamin-3, of DRP1 in calyx of Held synapses, besides to impair
but not dynamin-2, shown to be sensitive to the chronic mitochondrial morphology, leads to a reduction in the
silencing of neuronal activity induced by the Na+- RRP size negatively impacting on synaptic transmission
voltage-dependent-channel inhibitor tetrodotoxin (TTX). (Singh and others 2018).
However, dynamin-1 was excluded from nerve terminals In addition to their role in the endocytic-SV recycling,
in response to TTX, whereas dynamin-3 was accumu- classical dynamins could have other important functions
lated at the presynapses. This strongly suggests that these in the regulation of neurotransmission. As reported by
6 The Neuroscientist 00(0)

Mahapatra and coworkers, the conditional knockout of that transiently connects the vesicular lumen with the
dynamin-1 (cKO) increases transmission-failures during extracellular space allowing transmitter-output to the
high-frequency stimulation in calyx of Held synapses. synaptic cleft (Jena and others 2003). These mechanisms
However, and contrary to the expected consequence of appear to rely on the dynamin-actin interplay (Gonzalez-
reducing SV recycling, cKO enhances the steady-state Jamett and others 2013a; Shin and others 2018) and could
neurotransmission and reduces short-term depression explain defects in neurotransmitter release on dynamin
during brief periods of high frequency stimulation disruption and sustained neuronal activity (Fa and others
(Mahapatra and others 2016). As reduction in short-term- 2014; Mahapatra and others 2016), independent of the
depression was not due to modifications in the SV recycling.
SV-resupply rate nor to significant changes in the RRP In summary, different DSPs are implicated in the pre-
size (Mahapatra and others 2016) these authors suggested synaptic mechanisms that support transmission and plas-
the possibility that an enhanced clearance of the release ticity of the synaptic response and in particular classical
sites operate at the active zones (AZs) in the absence of dynamins seem to be relevant during high frequency stim-
dynamin-1. Since a rapid local membrane retrieval is ulations. Since intense and sustained neuronal activity is
important during high rates of transmitter release, fast thought to underlie plastic phenomena leading to memory
and massive endocytosis such as bulk endocytosis could formation, dynamin’s activity is likely required for this
become predominant during brief intense stimulation process (Fa and others 2014).
(Hayashi and others 2008; Watanabe and others 2013)
supporting the efficient clearance of the SV components Postsynaptic Dynamins and
and consequently promoting transmitter release and Signaling-Receptor Trafficking:
reducing depression (Mahapatra and others 2016). In this
Implications in Postsynaptic
regard, dynamin-1 action could be a limiting factor con-
trolling transmitter release and transmission. Indeed, oth- Mechanisms of Plasticity
ers reports also support the involvement of classical In addition to presynaptic mechanisms, synaptic trans-
dynamins in the mechanisms that control neurotransmit- mission has a very important post-synaptic component
ter release on repetitive neuronal activity. This is how that determines how efficiently cells respond to neu-
inhibition of dynamin’s GTP-ase activity with dynasore rotransmitters. Besides modifications in the efficiency
has been shown to accelerate the synaptic fatigue (SF) with which transmitters are released from nerve termi-
evoked during high frequency stimulation in mouse hip- nals, plasticity of the synaptic response relies on density,
pocampal slices (Fa and others 2014) and rat superior cer- type and properties of the postsynaptic receptors. Both,
vical ganglion neurons (Lu and others 2009). As SF is a excitatory (Bear and Malenka 1994; Malenka and Bear
form of short-term synaptic plasticity that reflects presyn- 2004) and inhibitory synapses (Gaiarsa and others 2002),
aptic depletion of the RRP (Zucker and Regehr 2002) exhibit plastic mechanisms that weaken or strengthen
these results are in agreement with a dynamin-dependent their efficacy. However, excitatory synaptic plasticity has
tuning of the release of transmitters on sustained stimula- received much more attention and the postsynaptic mech-
tion. Similarly post-tetanic stimulation, another form of anisms controlling its induction and expression are better
short-term synaptic plasticity dependent on sustained understood. Of these, long-term potentiation (LTP) and
neurotransmitter release, is also affected by dynasore (Fa long-term-depression (LTD) of the excitatory synaptic
and others 2014). Clearly, disruption of dynamins’ activ- transmission at the hippocampal Shaffer Collateral-
ity not only affects SV recycling at the presynapses but to-CA1 synapses are the most studied forms of synaptic
also impacts on the efficiency of neurosecretion. In this plasticity. Both LTP and LTD have widely shown to be
regard, the dynamin GTP-ase activity has been widely relevant for learning acquisition and memory formation
implicated in the mechanisms controlling kinetics and (Lee and Kirkwood 2011). Glutamate α-amino-3-
amount of transmitter released from endocrine cells (Min hydroxy-5-methyl-4-isoxazolepropionic acid recep-
and others 2007), neuroendocrine cells (Anantharam and tors (AMPARs) mediate the majority of fast excitatory
others 2011; Gonzalez-Jamett and others 2010; Gonzalez- synaptic transmission in the mammalian brain (Anggono
Jamett and others 2013a; Graham and others 2002; Tsuboi and Huganir 2012). Therefore, AMPAR trafficking into
and others 2004; Yang and others 2001) and neurons and out of synapses is a major regulatory mechanism
(Wong and others 2015). The dynamins’ activity appears underlying modifications in the efficacy of excitatory
to further modulate the exocytotic release of transmitters synaptic transmission (Anggono and Huganir 2012). In
by favoring expansion (Anantharam and others 2011; this regard, an increase in AMPAR density at the syn-
Gonzalez-Jamett and others 2013a; Samasilp and others apses results in LTP (Lu and others 2001; Park and others
2012) and reclosure (Lasic and others 2017; Tsuboi and 2004), whereas the endocytic AMPAR removal from syn-
others 2004) of the fusion pore, a nanometric “channel” apses leads to LTD (Carroll and others 1999; Man and
Arriagada-Diaz et al. 7

others 2000). Ca2+-dependent mechanisms determine


intracellular signaling cascades that modulate the direc-
tionality of the synaptic plasticity, for instance, by defining
the state of AMPAR phosphorylation/dephosphorylation,
which affects AMPAR trafficking, synaptic-targeting, and
function (Lee and others 2003). Remarkably, all these
mechanisms have shown to be regulated in some way by
the activity of DSPs. Among classical dynamins, isoforms
2 and 3 are most relevant in postsynapses (Okamoto and
others 2001) and seem to play major functions in AMPAR
trafficking. In 1999, Carroll and colleagues demonstrated
that overexpression of dynamin-2-K44A, a mutant unable
to bind and hydrolyze GTP, perturbs CME of AMPAR in
cultured hippocampal neurons (Carroll and others 1999).
This process occurs at specific endocytic zones (EZ)
adjacent to the postsynaptic-densities (PSDs), an elec-
tron-dense specialization of the postsynaptic membrane
that is located in close apposition to the presynaptic active
zone (Lu and others 2007a). Dynamin-2 appears to direct
AMPAR internalization in a way dependent on the activ-
ity-regulated-cytoskeleton-associated protein Arc/Ag3.1 Figure 3.  Schematic representation of the participation of
dynamin superfamily proteins (DSPs) in AMPAR-trafficking
(Chowdhury and others 2006) which in turns enhances
to and from postsynaptic densities. Dynamin-2 (DYN2)
the dynamin-GTP-ase activity (Byers and others 2015). directs lateral diffusion of AMPAR from dendritic shaft to
From EZs, AMPAR undergo endosomal sorting processes spines in response to synaptic activity. Dynamin-3 (DYN3)
that can take them to a lysosomal degradation pathway associates with Homer-1 coupling the endocytic zones
via late endosomes or back to the plasma membrane via (EZs) to postsynaptic densities (PSDs) and allowing efficient
recycling endosomes (REs) (Ehlers 2000; Park and others AMPAR recycling. DRP1 and DYN2 mediate mitochondrial
2004). On high frequencies of synaptic activity, REs segmentation allowing structural and functional synaptic
plasticity.
move into dendritic spines allowing a local delivery of
AMPAR (Park and others 2006). In this regard, although
some reports suggest a direct exocytosis of AMPAR into others 2007b). As reported by these authors, experimental
PSDs during LTP (Gerges and others 2006; Kopec and approaches that disrupt the Homer-1/dynamin-3 interac-
others 2006) most evidences suggest that postsynaptic tion induce the uncoupling of EZs from PSDs, negatively
AMPAR insertion occurs first extrasynaptically in the impacting on AMPAR PSD-targeting (Fig. 3) Congruently,
dendritic shaft, followed by a lateral diffusion from den- EZ/PSD uncoupling impairs AMPAR-mediated spontane-
dritic shaft to spines (Ashby and others 2004; Groc and ous excitatory postsynaptic currents (Lu and others 2007b)
others 2004; Tardin and others 2003; Yudowski and oth- and stimulus-induced synaptic potentiation (Petrini and
ers 2007). In 2009, Jaskolski and collaborators reported others 2009) further supporting an important role of dyna-
that the disruption of dynamin-2 GTP-ase activity with min-3 in excitatory synaptic transmission and plasticity.
the pharmacological agent dynasore or with the K44A As occurs during presynaptic mechanisms, dynamins’
mutant inhibits the NMDAR-induced lateral diffusion of participation in AMPAR trafficking seems to depends on
AMPAR from dendritic shaft to spines (Jaskolski and neuronal activity. In this sense, constitutive AMPAR
others 2009).The latter strongly suggests a key role of internalization and recycling in hippocampal neurons
dynamin-2 in driving a membrane drift that overrides the does not require CME or dynamin GTP-ase activity
topologic barrier imposed by dendritic spines and pro- (Fujii and others 2017; Glebov and others 2015).
motes AMPAR delivery into PSDs (Fig. 3) As the dyna- Homeostatic downscaling of the synaptic response,
min GTP-ase activity is required for endosomal-recycling induced by chronic enhancement of the excitatory neuro-
pathways in different cell types (Nicoziani and others nal activity, relies on the removal of AMPAR from the
2000; van Dam and Stoorvogel 2002), it could also regu- plasma membrane. This mechanism neither seems to
late PSD-targeting of AMPAR by modulating their recy- involve the dynamin activity, but it does require actin-
cling from REs. In this regard, Lu and collaborators cytoskeleton remodeling (Glebov and others 2015).
showed that dynamin-3, in association with the scaffold- Notwithstanding the apparent irrelevance of dynamin’s
ing protein Homer-1, links EZs to PSDs and that such GTP-ase activity in constitutive AMPAR recycling,
association is critical for AMPAR-recycling (Lu and induction of LTD by using low-frequency stimulation
8 The Neuroscientist 00(0)

Figure 4.  Pharmacological inhibition of dynamin’s GTPase activity decreases long-term potentiation (LTP). (A) Representative
diagram of the experimental setup used to obtain postsynaptic excitatory field records in hippocampal slices from 6-month-old
wild type mice. The stimulation electrode was placed on the Schaffer collaterals, while the recording electrode was placed on
stratum radiatum. Stimulation was elicited by a theta-burst (TBS) protocol to induce LTP. Field excitatory postsynaptic potentials
(fEPSP) were recorded in the Shaffer collateral-CA1 synapse during 60 minutes after TBS. Treatment with dynole 34-2 (Dyn 34-
2) and its inactive analogue dynole 31-2 (Dyn 31-2) was added 10 minutes before, during and after the LTP protocol application.
(B) Representative Dyn 31-2 (light brown) and Dyn34-2 (purple) traces of fEPSPs obtained before and after application of the
LTP protocol. (C) Note that the presence of Dyn 34-2 decreases the potentiation of the synaptic responses compared to Dyn
31-2. Dyn 31-2 (n = 19 slices from 5 mice), Dyn 34-2 (n = 11 slices from 3 mice). All data are plotted as mean ± SEM. Statistical
differences were calculated using t test,*P < 0.0001. Experimental protocols with animals were approved by the Ethics and
Animal Care Committee of Universidad de Valparaíso (BEA131-18).

pulses is significantly affected by dynamin’s inhibition impairs LTP in wild type hippocampal slices compared
(Fujii and others 2017; Glebov and others 2015; Man to that observed in slices treated with its inactive ana-
and others 2000) thus confirming dynamin’s requirement logue dynole 31-2 (Fig. 4; unpublished observations by
for AMPAR internalization on specific patterns of syn- Arriagada-Díaz). These data further support the rele-
aptic activity. Along the same line, Zheng and collabora- vance of the dynamin GTP-ase activity in AMPAR traf-
tors demonstrated that AMPARs recycle in a ficking and in the synaptic mechanisms depending on it.
dynamin-independent way in the absence of synaptic Besides intrinsic dynamin’s properties allowing mem-
stimulation but in a CME and dynamin GTP-ase- brane remodeling, another important regulation point
dependent way on chemical induction of LTP (Zheng through which the dynamin GTP-ase activity could mod-
and others 2015). In agreement with these findings we ulate AMPAR trafficking is the actin-cytoskeleton
have confirmed in our laboratory the importance of the dynamics. In fact, the dynamin activity orchestrates actin
dynamin’s GTP-ase activity for LTP expression in excit- remodeling in different experimental models by promot-
atory hippocampal synapses (Fig. 4). In our hands, treat- ing actin polymerization (Gonzalez-Jamett and others
ment with the dynamin inhibitor dynole 34-2 significantly 2017; Gu and others 2010; Mooren and others 2009) and
Arriagada-Diaz et al. 9

by modulating actin filament stabilization (Yamada and the dynamin’s activity. For instance, dopamine-induced
others 2013). Furthermore, actin dynamics constitutes an D2 receptor internalization in primary-cultured striatal
important regulator of the intracellular trafficking neurons depends on the dynamin-2 function (Kabbani
(Lanzetti 2007). At the dendritic spines, a rich actin net- and others 2004) suggesting its involvement in dopami-
work regulates exocytosis, endocytosis and endosomal nergic neurotransmission. Similarly, agonist- and antag-
recycling of AMPARs (Hanley 2014). Congruently, dis- onist-mediated internalization of serotonin 5-HT2A
ruption of actin remodeling suppresses potentiation of receptors, a major target of antipsychotic medications,
the synaptic transmission (Fukazawa and others 2003; requires dynamin GTP-ase activity (Bhatnagar and oth-
Kim and Lisman 1999). In addition to a functional role in ers 2001). Moreover, endocytosis of GABAA receptors, a
AMPAR trafficking, spine-actin has an important struc- major mechanism modulating experience-induced LTP
tural role supporting modifications in spine architecture and extinction of aversive memories, also depends on the
during induction of synaptic plasticity (Bosch and others function of dynamins (Wang and others 2017).
2014; Hanley 2014). This is how actin polymerization Thus, DSPs are critical components of the postsynap-
associates with spine enlargement on LTP (Lin and oth- tic machinery, modulating most of the mechanisms that
ers 2005), while actin depolymerization associates with govern synaptic transmission and plasticity.
spine shrinkage on LTD (Fortin and others 2012;
Okamoto and others 2009; Zhou and others 2004). Given
that classical dynamins are important regulators of the Dynamin Implication in Synaptic
actin dynamics (Gonzalez-Jamett and others 2017; Gu Dysfunction and Neurological
and others 2010; Mooren and others 2009; Yamada and Diseases
others 2013) they could also affect other actin-dependent
mechanisms during structural synaptic plasticity. Consistent with the role played by DSPs in the mecha-
Regarding structural plasticity, mitochondrial dynam- nisms that support synaptic transmission and plasticity,
ics is an important, although still understudied, check- dysfunction of these proteins has been associated with an
point. Mitochondria are enriched in dendrites where important number of neuropathologies. Table 1 summa-
“flashes” of excitable-mitochondrial events mediate rizes some of the most relevant synaptic pathologies
structural LTP (Fu and others 2017). Remarkably, LTP implicating DSP dysfunction.
induction prompts a burst of DRP1-mediated mitochon- In 2007, Waterham and collaborators reported a neo-
drial-segmentation, which in turns is necessary for synap- nathal lethal case characterized by encephalopathy and
togenesis, spine-remodeling and potentiation of the cerebral dysgenesis. This severe disorder was associated
synaptic strength (Divakaruni and others 2018). In this with defects in mitochondrial and peroxisomal fission
regard, DRP1 overexpression, along with enhance mito- and caused by a point mutation in the middle domain of
chondrial fragmentation, increases dendritic spine den- DRP1 (Waterham and others 2007). Other cases of
sity and promotes synapse formation in cultured encephalopathy linked to mutations in the DRP1-
hippocampal neurons (Li and others 2004). Interestingly, encoding gene DNM1L have been later reported (Vanstone
in addition to DRP1 activation, dynamin-2 is also required and others 2016; Yoon and others 2016). Some of them
for mitochondrial segmentation. Whilst DRP1 initiates also associate with seizures and refractory epilepsy
mitochondrial constriction facilitating dynamin-2 assem- (Vanstone and others 2016). Moreover, de novo missense
bly, dynamin-2 appears to mediate the last step of mito- mutations in the GTP-ase effector domain of DRP1 that
chondrial fission to complete division (Divakaruni and lead to static encephalopathy and seizures also were
others 2018; Lee and others 2016). This latter supports recently reported (Assia Batzir and others 2019).
the involvement of different DSPs in the mitochondrial- Classical dynamins have also been implicated in
dependent postsynaptic mechanisms that regulate excit- molecular mechanisms causing epileptogenic activity.
atory synaptic plasticity (Fig. 4). Patients expressing de novo mutations in the DNM1 gene,
Finally, and besides AMPAR trafficking, dynamin-2 located in the middle and GTP-ase domains of dynamin-1,
also participates in the endocytic recycling of N-methyl- suffer from seizures that start in childhood and frequently
d-aspartate receptors (NMDARs) in excitatory glutama- progress to the Lennox-Gastaut syndrome, a rare and
tergic synapses. As demonstrated by Montgomery and severe kind of refractory epilepsy, accompanied with
colleagues, NMDARs are down-regulated on low-fre- intellectual disabilities and hypotonia (von Spiczak and
quency stimulation patterns in a way that is dependent on others 2017). Interestingly, seizure-like activity is sup-
dynamin-mediated endocytosis (Montgomery and others pressed in shibire fly mutants (Kroll and others 2015) sug-
2005). Furthermore, the endocytic recycling of other gesting that dynamin-dependent SV recycling could be a
postsynaptic receptors has also shown to be mediated by potential therapeutic target in refractory epilepsy.
10
Table 1.  Synaptic Pathologies Involving Dysfunction of Dynamin Superfamily Proteins (DSPs): A Nonexhaustive List of Synaptic Disorders Linked to Deregulations in DSPs’
Activity.

DSP Synaptic Pathology Mechanism References


DRP1 Encephalopathy, microcephaly, Mutations in DNM1L gene (middle domain)/aberrant (Vanstone and others 2016; Waterham and
cerebral dysgenesis, seizures, mitochondria fragmentation others 2007; Yoon and others 2016)
refractory epilepsy
  Mutations in DNM1L gene (GTP-ase effector domain)/ (Assia Batzir and others 2019)
aberrant mitochondria fragmentation
  Alzheimer’s disease Exacerbated Aβ-induced GTP-ase activation of DRP1/ (Baek and others 2017; Kandimalla and
exacerbated mitochondrial segmentation others 2017; Yan and others 2015)
Mitofusin-2 Charcot-Marie-Tooth disease Mutations in the MFN2 gene/deregulated mitochondrial fusion (Kijima and others 2005)
Dynamin-1 Lennox-Gastaut syndrome, Mutations in DNM1 (middle and GTP-ase domains) (von Spiczak and others 2017)
refractory epilepsy, intellectual
disabilities, hypotonia
  Vascular dementia Decreased levels of dynamin-1 (Mulugeta and others 2014)
  Alzheimer’s disease Decreased levels of dynamin-1 (Kelly and others 2005; Watanabe and
others 2010)
Dynamin-2 Alzheimer’s disease Decreased levels of dynamin-2/Accumulation of membrane- (Kamagata and others 2009)
surface APP/ enhanced Aβ secretion
  Single nucleotide polymorphisms (Aidaralieva and others 2008)
  Charcot-Marie-Tooth disease Mutations in DNM2 (PH domain) (Zuchner and others 2005)
  Reduced capability to bind phospholipids (Kenniston and Lemmon 2010)
  Impaired CME/impaired microtubules instability (Claeys and others 2009; Sidiropoulos and
others 2012; Tanabe and Takei 2009)
  Impaired clathrin-independent endocytosis and membrane (Liu and others 2011)
trafficking
  Centronuclear myopathy Mutations in DNM2 (middle and PH domains) (Jeannet and others 2004)
  Cognitive impairments/learning disabilities (Bohm and others 2012; Echaniz-Laguna and
others 2007; Fischer and others 2006)
  Impaired centrosomal localization (Bitoun and others 2005)
  Impaired CME (Ali and others 2019; Bitoun and others
2009; Koutsopoulos and others 2011)
  Increased dynamin oligomerization and basal GTP-ase activity (Kenniston and Lemmon 2010; Wang and
in in vitro others 2010)
  Impaired clathrin-independent endocytosis and membrane (Liu and others 2011)
trafficking
  Impaired cytoskeletal actin dynamics and membrane (Gonzalez-Jamett and others 2017)
trafficking
Arriagada-Diaz et al. 11

A study analyzing postmortem brain tissue from Accumulation of Aβ in the AD context also modifies
patients who had suffered from cerebrovascular insults the expression levels of classical dynamins. As demon-
and developed vascular dementia (VaD) reported a cor- strated by Kelly and colleagues in 2005, Aβ significantly
relation between cognitive dysfunctions and dynamin-1 decreases dynamin-1 in hippocampal neurons, in a time
expression in affected tissues (Mulugeta and others and dose-dependent way. The mechanisms include both
2014). The study included samples from patients with increased calpain-mediated degradation and down-regu-
VaD, VaD linked to Alzheimer’s disease (VD-AD), and lation of dynamins’ gene expression (Kelly and others
stroke without dementia (SND) who in life were sub- 2005) and suggest involvement of these GTPases in the
jected to cognitive tests. Interestingly, bad punctuation in Aβ-induced neurodegeneration. Furthermore, rats
these tests were highly correlated with significant induced to transient cerebral ischemia in combination
decrease in dynamin-1 levels in white matter of VaD and with synthetic Aβ peptide exhibit significant memory
VaD-AD patients, but not in SND brain patients in which defects and reduction in dynamin-1 expression levels
dynamin-1 levels were even increased poststroke (Watanabe and others 2010). These changes were not
(Mulugeta and others 2014). These observations suggest observed on induction of cerebral ischemia alone
that dynamin-1 constitutes a protecting factor that reduces (Watanabe and others 2010), supporting an Aβ-triggered
the risk of develop dementia associated to cerebrovascu- modification in dynamin-1 expression/degradation.
lar insults. Regarding dynamin-2, its gene expression appears to
One of the most common forms of dementia in the be also affected in the AD context as a significant diminu-
elderly is AD. It is characterized by progressive neuronal tion in its mRNA levels was reported in cortical tissue of
deterioration and cognitive decline (Selkoe 2001). A AD brains (Kamagata and others 2009). In the same work
majority of the AD cases are sporadic forms of the disease the expression of the GTP-ase defective K44A dynamin-2
with late-onset (LOAD) and unknown cause (Selkoe mutant led to increase Aβ secretion from SH-SY5Y
2001). An earlier event in LOAD is a synaptic dysfunction human neuroblastoma cells (Kamagata and others 2009).
caused by accumulation of soluble Aβ oligomers (Selkoe Since most of the amyloid precursor protein (APP) in
2002). Although Aβ has multiple targets to exert its neuro- these cells was observed in the surface membrane, the
toxicity it has been shown to accumulate in mitochondria authors raised the possibility that the inhibition of dyna-
in AD brains (Maurer and others 2000) causing structural min-2 GTP-ase activity impairs APP internalization
and functional abnormalities such as oxidative stress, cal- resulting in its accumulation at the plasma membrane and
cium dyshomeostasis, impaired bioenergetics and apopto- enhanced Aβ secretion (Kamagata and others 2009). In
sis all mechanisms implicated in the Aβ-mediated fact, something similar could happen in the AD brains in
neuronal degeneration (Cha and others 2012; Lustbader which dynamin-2 expression is reduced. Interestingly,
and others 2004). A considerable number of reports sug- single nucleotide polymorphisms in the dynamin-2-en-
gest that an excessive mitochondrial segmentation linked coding DNM2 gene have been identified in AD patients
to DRP1 deregulation contributes to the LOAD pathogen- (Aidaralieva and others 2008). This latter study suggests
esis. In this regard, Yan and collaborators showed that Aβ that, like other locus in the human chromosome 19, that
triggers cytotoxic cascades that enhance the GSK3β is, the allele ε4 of the apolipoprotein E (Borgaonkar and
kinase activity promoting phosphorylation and activation others 1993), DNM2 polymorphisms could also consti-
of DRP1 (Yan and others 2015). Cultured neurons express- tute a genetic risk factor in LOAD.
ing DRP1-phosphomimetics exhibit increased mitochon- Mutations in DSPs have also been linked to peripheral
drial fragmentation and become more susceptible to synapses dysfunction in the context of neuromuscular
Aβ-induced apoptosis (Yan and others 2015) suggesting diseases. Charcot-Marie-Tooth disease (CMT) a periph-
that DRP1 inhibition could be a potential protecting factor eral neuropathy featured by neuropathic pain, muscular
to ameliorate Aβ-toxicity. In fact, inhibition of DRP1 with weakness, and atrophy (Claeys and others 2009) is caused
the pharmacological drug mdivi-1 manages to reduce mito- among others by mutations in genes encoding DSPs.
chondrial dysfunctions and spatial memory defects in a Mutations in the GTP-ase domain of the mitochondrial
mouse model of AD (Baek and others 2017). In the same fusion protein mitofusin-2 cause an axonal form of CMT
line, a partial reduction of DRP1 in a mouse model of AD (Kijima and others 2005) in which degeneration of neuro-
that expresses hyperphosphorylated forms of the microtu- nal axons causes muscle weakness and distal sensory loss
bule-associated protein Tau reduces mitochondrial dysfunc- (Kijima and others 2005). Intermediate forms of CMT,
tion and decreases the production of hyperphosphorylated characterized by axonal demyelination and axonal degen-
Tau (Kandimalla and others 2017). Certainly, these evi- eration are caused by mutations in the dynamin-2-encod-
dences could support a potential role of DRP1 as a thera- ing gene DNM2 (Zuchner and others 2005). Such
peutic target to intervene some of the pathological mutations mainly localize in the PH domain of dyna-
mechanisms in AD. min-2 affecting its capability to bind phospholipids
12 The Neuroscientist 00(0)

(Kenniston and Lemmon 2010) and impairing CME, memory, one of the most fundamental brain functions.
clathrin-independent endocytosis and membrane traffick- As DSPs are required in a number of these mechanisms,
ing (Liu and others 2011). Additionally, CMT-mutations it can be said that their GTP-ase activity is a key factor
in dynamin-2 enhance the dynamin-2/microtubule asso- modulating the strength of the synaptic response and
ciation, impairing microtubules instability (Tanabe and consequently allowing memory formation. While at the
Takei 2009). presynapses DSPs support endocytic-SV recycling and
Another inherited neuromuscular disorder, centronu- neurotransmitter release (Fig. 2), at postsynapses, they
clear myopathy (CNM), is caused by mutations in the are critical in controlling receptor-availability at the sur-
DNM2 gene. Its clinical phenotype includes muscle face membranes. Particularly in excitatory glutamatergic
weakness, fatigability and progressive atrophy of distal synapses, classical dynamins orchestrate the local traf-
skeletal muscles (Bitoun and others 2009; Bitoun and ficking of AMPAR to and from PSDs (Fig. 3) supporting
others 2005; Fischer and others 2006; Jeannet and others excitatory synaptic plasticity. Furthermore, DRPs impli-
2004). DNM2 mutations linked to CNM are mainly cated in mitochondrial dynamics (Fig. 3) are important
located in the middle and PH domains of dynamin-2. modulators of structural and functional synaptic plastic-
Most of them have shown to increase dynamin oligomer- ity. It is not surprising then that on conditions affecting
ization and basal GTP-ase activity in in vitro experiments expression and/or functionality of DSPs synaptic trans-
(Kenniston and Lemmon 2010; Wang and others 2010) mission and plasticity are severely compromised, leading
although it is still unclear their effect on the enzymatic to different pathologies of the nervous system (Table 1).
activity in a cell-context. As reported by Liu and collabo- Better understanding the processes in which DSPs par-
rators, clathrin-independent endocytosis and membrane ticipate is a major challenge in neuroscience, with a view
trafficking, but not CME, are mechanisms affected by to using them as therapeutic targets in prevalent
CNM-causing dynamin-2 mutations (Liu and others synaptopathies.
2011). On the contrary, significant defects in CME have
been published by other authors (Ali and others 2019; Acknowledgments
Bitoun and others 2009; Koutsopoulos and others 2011).
We thanks Mr. Felipe Serrano and his company Illustrative-
Moreover, as we described in 2017, CNM-causing dyna- science (www.illustrative-science.com) for constructing the
min-2 mutations in muscle cells impair cytoskeletal actin schemes presented in Figures 1, 2 and 3.
remodeling and insulin-induced trafficking of the glucose
transporter GLUT4 (Gonzalez-Jamett and others 2017)
pointing out that actin dynamics and receptor trafficking Author Contributions
are important mechanisms affected in the CNM context. All authors listed have made a substantial intellectual contribu-
Despite the fact that CNM is intrinsically a skeletal tion to this work and approved it for publication.
muscular disorder, several clinical reports describe cogni-
tive impairments manifested as learning disabilities, lim- Declaration of Conflicting Interests
ited intelligence quotient and even borderline “mental The author(s) declared no potential conflicts of interest with
retardation” in dynamin-2-related CNM patients (Bohm respect to the research, authorship, and/or publication of this
and others 2012; Echaniz-Laguna and others 2007; article.
Fischer and others 2006; Jeannet and others 2004)
although the underlying mechanisms have never been Funding
studied. Certainly, understanding signaling cascades The author(s) disclosed receipt of the following financial sup-
leading to cognitive defects in patients with CNM could port for the research, authorship, and/or publication of this arti-
have valuable implications to better understand the role cle: This work was supported by ANID 22200154 (to JA-D),
of dynamin-2 in synaptic transmission and plasticity. Fondecyt 11180731 (to AMG-J), Fondecyt 1201342 (to AOA),
In summary, DSPs are relevant regulation points in ICM-ANID ICN09-022, CINV (to AMG-J, AOA, and AMCD)
which converge many of the synaptic mechanisms under-
lying neurotransmission. When the activity of these pro- ORCID iD
teins is deregulated these mechanisms are substantially Arlek M. Gonzalez-Jamett https://orcid.org/0000-0003
affected, endangering synaptic health and leading to neu- -2357-0260
rological diseases.
References
Concluding Remarks Aidaralieva NJ, Kamino K, Kimura R, Yamamoto M, Morihara
T, Kazui H, and others. 2008. Dynamin 2 gene is a novel
Mechanisms mediating synaptic transmission and plas- susceptibility gene for late-onset Alzheimer disease in non-
ticity constitute the molecular basis of learning and APOE-epsilon4 carriers. J Hum Genet 53(4):296–302.
Arriagada-Diaz et al. 13

Ali T, Bednarska J, Vassilopoulos S, Tran M, Diakonov IA, Alzheimer’s disease with apolipoprotein E type 4 on chro-
Ziyadeh-Isleem A, and others. 2019. Correlative SICM- mosome 19. Lancet 342(8871):625.
FCM reveals changes in morphology and kinetics of Bosch M, Castro J, Saneyoshi T, Matsuno H, Sur M, Hayashi
endocytic pits induced by disease-associated mutations in Y. 2014. Structural and molecular remodeling of dendritic
dynamin. FASEB J 33(7):8504–18. spine substructures during long-term potentiation. Neuron
Anantharam A, Bittner MA, Aikman RL, Stuenkel EL, Schmid 82(2):444–59.
SL, Axelrod D, and others. 2011. A new role for the dyna- Byers CE, Barylko B, Ross JA, Southworth DR, James NG,
min GTPase in the regulation of fusion pore expansion. Taylor CAt, and others. 2015. Enhancement of dyna-
Mol Biol Cell 22(11):1907–18. min polymerization and GTPase activity by Arc/Arg3.1.
Anggono V, Huganir RL. 2012. Regulation of AMPA recep- Biochim Biophys Acta 1850(6):1310–8.
tor trafficking and synaptic plasticity. Curr Opin Neurobiol Calabrese B, Halpain S. 2015. Differential targeting of dyna-
22(3):461–9. min-1 and dynamin-3 to nerve terminals during chronic sup-
Antonny B, Burd C, De Camilli P, Chen E, Daumke O, pression of neuronal activity. Mol Cell Neurosci 68:36–45.
Faelber K, and others. 2016. Membrane fission by dyna- Cao H, Garcia F, McNiven MA. 1998. Differential distribution
min: what we know and what we need to know. EMBO J of dynamin isoforms in mammalian cells. Mol Biol Cell
35(21):2270–84. 9(9):2595–609.
Ashby MC, De La Rue SA, Ralph GS, Uney J, Collingridge Cardenas AM, Marengo FD. 2010. Rapid endocytosis and ves-
GL, Henley JM. 2004. Removal of AMPA receptors icle recycling in neuroendocrine cells. Cell Mol Neurobiol
(AMPARs) from synapses is preceded by transient endocy- 30(8):1365–70.
tosis of extrasynaptic AMPARs. J Neurosci 24(22):5172–6. Carroll RC, Beattie EC, Xia H, Luscher C, Altschuler Y, Nicoll
Assia Batzir N, Bhagwat PK, Eble TN, Liu P, Eng CM, Elsea RA, and others. 1999. Dynamin-dependent endocytosis of
SH, and others. 2019. De novo missense variant in the ionotropic glutamate receptors. Proc Natl Acad Sci U S A
GTPase effector domain (GED) of DNM1L leads to static 96(24):14112–7.
encephalopathy and seizures. Cold Spring Harb Mol Case Cha MY, Han SH, Son SM, Hong HS, Choi YJ, Byun J, and
Stud 5(3):a003673. others. 2012. Mitochondria-specific accumulation of amy-
Baek SH, Park SJ, Jeong JI, Kim SH, Han J, Kyung JW, and loid beta induces mitochondrial dysfunction leading to
others. 2017. Inhibition of Drp1 ameliorates synaptic apoptotic cell death. PLoS One 7(4):e34929.
depression, Aβ deposition, and cognitive impairment in an Chappie JS, Acharya S, Liu YW, Leonard M, Pucadyil TJ,
Alzheimer’s disease model. J Neurosci 37(20):5099–110. Schmid SL. 2009. An intramolecular signaling element that
Bear MF, Malenka RC. 1994. Synaptic plasticity: LTP and modulates dynamin function in vitro and in vivo. Mol Biol
LTD. Curr Opin Neurobiol 4(3):389–99. Cell 20(15):3561–71.
Berthet A, Margolis EB, Zhang J, Hsieh I, Zhang J, Hnasko TS, Chappie JS, Mears JA, Fang S, Leonard M, Schmid SL,
and others. 2014. Loss of mitochondrial fission depletes Milligan RA, and others. 2011. A pseudoatomic model
axonal mitochondria in midbrain dopamine neurons. J of the dynamin polymer identifies a hydrolysis-dependent
Neurosci 34(43):14304–17. powerstroke. Cell 147(1):209–22.
Bhatnagar A, Willins DL, Gray JA, Woods J, Benovic JL, Roth Chen MS, Obar RA, Schroeder CC, Austin TW, Poodry CA,
BL. 2001. The dynamin-dependent, arrestin-independent Wadsworth SC, and others. 1991. Multiple forms of dyna-
internalization of 5-hydroxytryptamine 2A (5-HT2A) min are encoded by shibire, a Drosophila gene involved in
serotonin receptors reveals differential sorting of arrestins endocytosis. Nature 351(6327):583–6.
and 5-HT2A receptors during endocytosis. J Biol Chem Cheung G, Cousin MA. 2019. Synaptic vesicle generation from
276(11):8269–77. activity-dependent bulk endosomes requires a dephos-
Bitoun M, Durieux AC, Prudhon B, Bevilacqua JA, Herledan phorylation-dependent dynamin-syndapin interaction. J
A, Sakanyan V, and others. 2009. Dynamin 2 mutations Neurochem 151(5):570–583.
associated with human diseases impair clathrin-mediated Chowdhury S, Shepherd JD, Okuno H, Lyford G, Petralia RS,
receptor endocytosis. Hum Mutat 30(10):1419–27. Plath N, and others. 2006. Arc/Arg3.1 interacts with the
Bitoun M, Maugenre S, Jeannet PY, Lacene E, Ferrer X, Laforet endocytic machinery to regulate AMPA receptor traffick-
P, and others. 2005. Mutations in dynamin 2 cause domi- ing. Neuron 52(3):445–59.
nant centronuclear myopathy. Nat Genet 37(11):1207–9. Claeys KG, Zuchner S, Kennerson M, Berciano J, Garcia A,
Bohm J, Biancalana V, Dechene ET, Bitoun M, Pierson CR, Verhoeven K, and others. 2009. Phenotypic spectrum of
Schaefer E, and others. 2012. Mutation spectrum in the dynamin 2 mutations in Charcot-Marie-Tooth neuropathy.
large GTPase dynamin 2, and genotype-phenotype correla- Brain 132(Pt 7):1741–52.
tion in autosomal dominant centronuclear myopathy. Hum Clark SG, Shurland DL, Meyerowitz EM, Bargmann CI,
Mutat 33(6):949–59. van der Bliek AM. 1997. A dynamin GTPase mutation
Bonekamp NA, Vormund K, Jacob R, Schrader M. 2010. causes a rapid and reversible temperature-inducible loco-
Dynamin-like protein 1 at the Golgi complex: a novel com- motion defect in C. elegans. Proc Natl Acad Sci U S A
ponent of the sorting/targeting machinery en route to the 94(19):10438–43.
plasma membrane. Exp Cell Res 316(20):3454–67. Clayton EL, Cousin MA. 2008. Differential labelling of bulk
Borgaonkar DS, Schmidt LC, Martin SE, Kanzer MD, Edelsohn endocytosis in nerve terminals by FM dyes. Neurochem Int
L, Growdon J, and others. 1993. Linkage of late-onset 53(3-4):51–5.
14 The Neuroscientist 00(0)

Damke H, Baba T, Warnock DE, Schmid SL. 1994. Induction targeting and insertion into the postsynaptic membrane.
of mutant dynamin specifically blocks endocytic coated EMBO J 25(8):1623–34.
vesicle formation. J Cell Biol 127(4):915–34. Glebov OO, Tigaret CM, Mellor JR, Henley JM. 2015. Clathrin-
Delgado R, Maureira C, Oliva C, Kidokoro Y, Labarca P. 2000. independent trafficking of AMPA receptors. J Neurosci
Size of vesicle pools, rates of mobilization, and recycling 35(12):4830–6.
at neuromuscular synapses of a Drosophila mutant, shibire. Gonzalez-Jamett AM, Baez-Matus X, Hevia MA, Guerra MJ,
Neuron 28(3):941–53. Olivares MJ, Martinez AD, and others. 2010. The associa-
Divakaruni SS, Van Dyke AM, Chandra R, LeGates TA, tion of dynamin with synaptophysin regulates quantal size
Contreras M, Dharmasri PA, and others. 2018. Long-term and duration of exocytotic events in chromaffin cells. J
potentiation requires a rapid burst of dendritic mitochon- Neurosci 30(32):10683–91.
drial fission during induction. Neuron 100(4):860–875.e7. Gonzalez-Jamett AM, Guerra MJ, Olivares MJ, Haro-Acuna V,
Echaniz-Laguna A, Nicot AS, Carre S, Franques J, Tranchant Baez-Matus X, Vasquez-Navarrete J, and others. 2017. The
C, Dondaine N, and others. 2007. Subtle central and F-actin binding protein cortactin regulates the dynamics of
peripheral nervous system abnormalities in a family with the exocytotic fusion pore through its SH3 domain. Front
centronuclear myopathy and a novel dynamin 2 gene muta- Cell Neurosci 11:130.
tion. Neuromuscul Disord 17(11-12):955–9. Gonzalez-Jamett AM, Haro-Acuna V, Momboisse F, Caviedes
Ehlers MD. 2000. Reinsertion or degradation of AMPA recep- P, Bevilacqua JA, Cardenas AM. 2014. Dynamin-2 in ner-
tors determined by activity-dependent endocytic sorting. vous system disorders. J Neurochem 128(2):210–23.
Neuron 28(2):511–25. Gonzalez-Jamett AM, Momboisse F, Guerra MJ, Ory S, Baez-
Evergren E, Zotova E, Brodin L, Shupliakov O. 2006. Matus X, Barraza N, and others. 2013a. Dynamin-2 regu-
Differential efficiency of the endocytic machinery in tonic lates fusion pore expansion and quantal release through a
and phasic synapses. Neuroscience 141(1):123–31. mechanism that involves actin dynamics in neuroendocrine
Fa M, Staniszewski A, Saeed F, Francis YI, Arancio O. 2014. chromaffin cells. PLoS One 8(8):e70638.
Dynamin 1 is required for memory formation. PLoS One Gonzalez-Jamett AM, Momboisse F, Haro-Acuna V, Bevilacqua
9(3):e91954. JA, Caviedes P, Cardenas AM. 2013b. Dynamin-2 func-
Faelber K, Posor Y, Gao S, Held M, Roske Y, Schulze D, and tion and dysfunction along the secretory pathway. Front
others. 2011. Crystal structure of nucleotide-free dynamin. Endocrinol (Lausanne) 4:126.
Nature 477(7366):556–60. Graham ME, O’Callaghan DW, McMahon HT, Burgoyne
Ferguson SM, Brasnjo G, Hayashi M, Wolfel M, Collesi C, RD. 2002. Dynamin-dependent and dynamin-independent
Giovedi S, and others. 2007. A selective activity-dependent processes contribute to the regulation of single vesicle
requirement for dynamin 1 in synaptic vesicle endocytosis. release kinetics and quantal size. Proc Natl Acad Sci U S
Science 316(5824):570–4. A 99(10):7124–9.
Ferguson SM, De Camilli P. 2012. Dynamin, a membrane- Granseth B, Odermatt B, Royle SJ, Lagnado L. 2006. Clathrin-
remodelling GTPase. Nat Rev Mol Cell Biol 13(2):75–88. mediated endocytosis is the dominant mechanism of vesicle
Fischer D, Herasse M, Bitoun M, Barragan-Campos HM, retrieval at hippocampal synapses. Neuron 51(6):773–86.
Chiras J, Laforet P, and others. 2006. Characterization of Groc L, Heine M, Cognet L, Brickley K, Stephenson FA,
the muscle involvement in dynamin 2-related centronuclear Lounis B, and others. 2004. Differential activity-dependent
myopathy. Brain 129(Pt 6):1463–9. regulation of the lateral mobilities of AMPA and NMDA
Ford MG, Jenni S, Nunnari J. 2011. The crystal structure of receptors. Nat Neurosci 7(7):695–6.
dynamin. Nature 477(7366):561–6. Gu C, Yaddanapudi S, Weins A, Osborn T, Reiser J, Pollak M,
Fortin DA, Srivastava T, Soderling TR. 2012. Structural and others. 2010. Direct dynamin-actin interactions regu-
modulation of dendritic spines during synaptic plasticity. late the actin cytoskeleton. EMBO J 29(21):3593–606.
Neuroscientist 18(4):326–41. Hamao K, Morita M, Hosoya H. 2009. New function of the
Fu ZX, Tan X, Fang H, Lau PM, Wang X, Cheng H, and others. proline rich domain in dynamin-2 to negatively regulate its
2017. Dendritic mitoflash as a putative signal for stabiliz- interaction with microtubules in mammalian cells. Exp Cell
ing long-term synaptic plasticity. Nat Commun 8(1):31. Res 315(7):1336–45.
Fujii S, Tanaka H, Hirano T. 2017. Detection and character- Hanley JG. 2014. Actin-dependent mechanisms in AMPA
ization of individual endocytosis of AMPA-type gluta- receptor trafficking. Front Cell Neurosci 8:381.
mate receptor around postsynaptic membrane. Genes Cells Hayashi M, Raimondi A, O’Toole E, Paradise S, Collesi C,
22(6):583–90. Cremona O, and others. 2008. Cell- and stimulus-depen-
Fukazawa Y, Saitoh Y, Ozawa F, Ohta Y, Mizuno K, Inokuchi dent heterogeneity of synaptic vesicle endocytic recycling
K. 2003. Hippocampal LTP is accompanied by enhanced mechanisms revealed by studies of dynamin 1-null neu-
F-actin content within the dendritic spine that is essential rons. Proc Natl Acad Sci U S A 105(6):2175–80.
for late LTP maintenance in vivo. Neuron 38(3):447–60. Henley JR, Krueger EW, Oswald BJ, McNiven MA. 1998.
Gaiarsa JL, Caillard O, Ben-Ari Y. 2002. Long-term plasticity Dynamin-mediated internalization of caveolae. J Cell Biol
at GABAergic and glycinergic synapses: mechanisms and 141(1):85–99.
functional significance. Trends Neurosci 25(11):564–70. Hinshaw JE, Schmid SL. 1995. Dynamin self-assembles into
Gerges NZ, Backos DS, Rupasinghe CN, Spaller MR, Esteban rings suggesting a mechanism for coated vesicle budding.
JA. 2006. Dual role of the exocyst in AMPA receptor Nature 374(6518):190–2.
Arriagada-Diaz et al. 15

Ishida N, Nakamura Y, Tanabe K, Li SA, Takei K. 2011. during chemically induced long-term potentiation. J
Dynamin 2 associates with microtubules at mitosis Neurosci 26(7):2000–9.
and regulates cell cycle progression. Cell Struct Funct Koutsopoulos OS, Koch C, Tosch V, Bohm J, North KN,
36(2):145–54. Laporte J. 2011. Mild functional differences of dynamin
Jaskolski F, Mayo-Martin B, Jane D, Henley JM. 2009. 2 mutations associated to centronuclear myopathy and
Dynamin-dependent membrane drift recruits AMPA recep- Charcot-Marie-Tooth peripheral neuropathy. PLoS One
tors to dendritic spines. J Biol Chem 284(18):12491–503. 6(11):e27498.
Jeannet PY, Bassez G, Eymard B, Laforet P, Urtizberea JA, Kreitzer G, Marmorstein A, Okamoto P, Vallee R, Rodriguez-
Rouche A, and others. 2004. Clinical and histologic find- Boulan E. 2000. Kinesin and dynamin are required for
ings in autosomal centronuclear myopathy. Neurology post-Golgi transport of a plasma-membrane protein. Nat
62(9):1484–90. Cell Biol 2(2):125–7.
Jena BP, Cho SJ, Jeremic A, Stromer MH, Abu-Hamdah Kroll JR, Wong KG, Siddiqui FM, Tanouye MA. 2015.
R. 2003. Structure and composition of the fusion pore. Disruption of endocytosis with the dynamin mutant shi-
Biophys J 84(2 Pt 1):1337–43. birets1 suppresses seizures in Drosophila. Genetics
Jones SM, Howell KE, Henley JR, Cao H, McNiven MA. 1998. 201(3):1087–102.
Role of dynamin in the formation of transport vesicles from Lanzetti L. 2007. Actin in membrane trafficking. Curr Opin
the trans-Golgi network. Science 279(5350):573–7. Cell Biol 19(4):453–8.
Kabbani N, Jeromin A, Levenson R. 2004. Dynamin-2 asso- Lasic E, Stenovec M, Kreft M, Robinson PJ, Zorec R. 2017.
ciates with the dopamine receptor signalplex and regu- Dynamin regulates the fusion pore of endo- and exocytotic
lates internalization of activated D2 receptors. Cell Signal vesicles as revealed by membrane capacitance measure-
16(4):497–503. ments. Biochim Biophys Acta Gen Subj 1861(9):2293–303.
Kamagata E, Kudo T, Kimura R, Tanimukai H, Morihara T, Lee HK, Kirkwood A. 2011. AMPA receptor regulation during
Sadik MG, and others. 2009. Decrease of dynamin 2 lev- synaptic plasticity in hippocampus and neocortex. Semin
els in late-onset Alzheimer’s disease alters Aβ metabolism. Cell Dev Biol 22(5):514–20.
Biochem Biophys Res Commun 379(3):691–5. Lee HK, Takamiya K, Han JS, Man H, Kim CH, Rumbaugh G,
Kandimalla R, Thirumala V, Reddy PH. 2017. Is Alzheimer’s and others. 2003. Phosphorylation of the AMPA receptor
disease a type 3 diabetes? A critical appraisal. Biochim GluR1 subunit is required for synaptic plasticity and reten-
Biophys Acta Mol Basis Dis 1863(5):1078–89. tion of spatial memory. Cell 112(5):631–43.
Kawasaki F, Hazen M, Ordway RW. 2000. Fast synaptic fatigue Lee JE, Westrate LM, Wu H, Page C, Voeltz GK. 2016.
in shibire mutants reveals a rapid requirement for dynamin Multiple dynamin family members collaborate to drive
in synaptic vesicle membrane trafficking. Nat Neurosci mitochondrial division. Nature 540(7631):139–43.
3(9):859–60. Li H, Alavian KN, Lazrove E, Mehta N, Jones A, Zhang P, and
Kelly BL, Vassar R, Ferreira A. 2005. Beta-amyloid-induced others. 2013. A Bcl-xL-Drp1 complex regulates synaptic
dynamin 1 depletion in hippocampal neurons. A potential vesicle membrane dynamics during endocytosis. Nat Cell
mechanism for early cognitive decline in Alzheimer dis- Biol 15(7):773–85.
ease. J Biol Chem 280(36):31746–53. Li YY, Chen XN, Fan XX, Zhang YJ, Gu J, Fu XW, and others.
Kenniston JA, Lemmon MA. 2010. Dynamin GTPase regula- 2015. Upregulated dynamin 1 in an acute seizure model
tion is altered by PH domain mutations found in centro- and in epileptic patients. Synapse 69(2):67–77.
nuclear myopathy patients. EMBO J 29(18):3054–67. Li Z, Okamoto K, Hayashi Y, Sheng M. 2004. The importance
Kijima K, Numakura C, Izumino H, Umetsu K, Nezu A, Shiiki of dendritic mitochondria in the morphogenesis and plas-
T, and others. 2005. Mitochondrial GTPase mitofusin 2 ticity of spines and synapses. Cell 119(6):873–87.
mutation in Charcot-Marie-Tooth neuropathy type 2A. Lin B, Kramar EA, Bi X, Brucher FA, Gall CM, Lynch G. 2005.
Hum Genet 116(1-2):23–7. Theta stimulation polymerizes actin in dendritic spines of
Kim CH, Lisman JE. 1999. A role of actin filament in synap- hippocampus. J Neurosci 25(8):2062–9.
tic transmission and long-term potentiation. J Neurosci Lin HC, Barylko B, Achiriloaie M, Albanesi JP. 1997.
19(11):4314–24. Phosphatidylinositol (4,5)-bisphosphate-dependent activa-
Koenig JH, Ikeda K. 1989. Disappearance and reformation tion of dynamins I and II lacking the proline/arginine-rich
of synaptic vesicle membrane upon transmitter release domains. J Biol Chem 272(41):25999–6004.
observed under reversible blockage of membrane retrieval. Linares-Clemente P, Rozas JL, Mircheski J, Garcia-Junco-
J Neurosci 9(11):3844–60. Clemente P, Martinez-Lopez JA, Nieto-Gonzalez JL, and
Kokotos AC, Cousin MA. 2015. Synaptic vesicle genera- others. 2015. Different dynamin blockers interfere with
tion from central nerve terminal endosomes. Traffic distinct phases of synaptic endocytosis during stimulation
16(3):229–40. in motoneurones. J Physiol 593(13):2867–88.
Kong L, Sochacki KA, Wang H, Fang S, Canagarajah B, Liu YW, Lukiyanchuk V, Schmid SL. 2011. Common mem-
Kehr AD, and others. 2018. Cryo-EM of the dyna- brane trafficking defects of disease-associated dynamin 2
min polymer assembled on lipid membrane. Nature mutations. Traffic 12(11):1620–33.
560(7717):258–62. Loerke D, Mettlen M, Yarar D, Jaqaman K, Jaqaman H,
Kopec CD, Li B, Wei W, Boehm J, Malinow R. 2006. Danuser G, and others. 2009. Cargo and dynamin regulate
Glutamate receptor exocytosis and spine enlargement clathrin-coated pit maturation. PLoS Biol 7(3):e57.
16 The Neuroscientist 00(0)

Lou X, Fan F, Messa M, Raimondi A, Wu Y, Looger LL, and Mooren OL, Kotova TI, Moore AJ, Schafer DA. 2009.
others. 2012. Reduced release probability prevents vesicle Dynamin2 GTPase and cortactin remodel actin filaments.
depletion and transmission failure at dynamin mutant syn- J Biol Chem 284(36):23995–4005.
apses. Proc Natl Acad Sci U S A 109(8):E515–23. Mulugeta E, Vallortigara J, Francis PT, Hye A, Kalaria RN,
Lu J, Helton TD, Blanpied TA, Racz B, Newpher TM, Perry EK, and others. 2014. Dynamin protein in stroke and
Weinberg RJ, and others. 2007a. Postsynaptic posi- vascular dementia. Neurosci Lett 563:118–22.
tioning of endocytic zones and AMPA receptor cycling Newman-Smith ED, Shurland DL, van der Bliek AM. 1997.
by physical coupling of dynamin-3 to Homer. Neuron Assignment of the dynamin-1 gene (DNM1) to human
55(6):874–89. chromosome 9q34 by fluorescence in situ hybridization
Lu W, Ma H, Sheng ZH, Mochida S. 2009. Dynamin and activ- and somatic cell hybrid analysis. Genomics 41(2):286–9.
ity regulate synaptic vesicle recycling in sympathetic neu- Newton AJ, Kirchhausen T, Murthy VN. 2006. Inhibition
rons. J Biol Chem 284(3):1930–7. of dynamin completely blocks compensatory synap-
Lu W, Man H, Ju W, Trimble WS, MacDonald JF, Wang YT. tic vesicle endocytosis. Proc Natl Acad Sci U S A
2001. Activation of synaptic NMDA receptors induces 103(47):17955–60.
membrane insertion of new AMPA receptors and LTP in Nicoziani P, Vilhardt F, Llorente A, Hilout L, Courtoy PJ,
cultured hippocampal neurons. Neuron 29(1):243–54. Sandvig K, and others. 2000. Role for dynamin in late
Lu Y, Allen M, Halt AR, Weisenhaus M, Dallapiazza RF, endosome dynamics and trafficking of the cation-inde-
Hall DD, and others. 2007b. Age-dependent requirement pendent mannose 6-phosphate receptor. Mol Biol Cell
of AKAP150-anchored PKA and GluR2-lacking AMPA 11(2):481–95.
receptors in LTP. EMBO J 26(23):4879–90. Noakes PG, Chin D, Kim SS, Liang S, Phillips WD. 1999.
Lustbader JW, Cirilli M, Lin C, Xu HW, Takuma K, Wang Expression and localisation of dynamin and syntaxin dur-
N, and others. 2004. ABAD directly links Abeta to ing neural development and neuromuscular synapse forma-
mitochondrial toxicity in Alzheimer’s disease. Science tion. J Comp Neurol 410(4):531–40.
304(5669):448–52. Okamoto K, Bosch M, Hayashi Y. 2009. The roles of CaMKII
Ma H, Cai Q, Lu W, Sheng ZH, Mochida S. 2009. KIF5B motor and F-actin in the structural plasticity of dendritic spines: a
adaptor syntabulin maintains synaptic transmission in sym- potential molecular identity of a synaptic tag? Physiology
pathetic neurons. J Neurosci 29(41):13019–29. (Bethesda) 24:357–66.
Maeda K, Nakata T, Noda Y, Sato-Yoshitake R, Hirokawa N. Okamoto PM, Gamby C, Wells D, Fallon J, Vallee RB. 2001.
1992. Interaction of dynamin with microtubules: its struc- Dynamin isoform-specific interaction with the shank/
ture and GTPase activity investigated by using highly puri- ProSAP scaffolding proteins of the postsynaptic density
fied dynamin. Mol Biol Cell 3(10):1181–94. and actin cytoskeleton. J Biol Chem 276(51):48458–65.
Mahapatra S, Fan F, Lou X. 2016. Tissue-specific dynamin-1 Park M, Penick EC, Edwards JG, Kauer JA, Ehlers MD. 2004.
deletion at the calyx of Held decreases short-term depres- Recycling endosomes supply AMPA receptors for LTP.
sion through a mechanism distinct from vesicle resupply. Science 305(5692):1972–5.
Proc Natl Acad Sci U S A 113(22):E3150–8. Park M, Salgado JM, Ostroff L, Helton TD, Robinson CG,
Malenka RC, Bear MF. 2004. LTP and LTD: an embarrassment Harris KM, and others. 2006. Plasticity-induced growth
of riches. Neuron 44(1):5–21. of dendritic spines by exocytic trafficking from recycling
Man HY, Lin JW, Ju WH, Ahmadian G, Liu L, Becker LE, endosomes. Neuron 52(5):817–30.
and others. 2000. Regulation of AMPA receptor-mediated Petrini EM, Lu J, Cognet L, Lounis B, Ehlers MD, Choquet D.
synaptic transmission by clathrin-dependent receptor inter- 2009. Endocytic trafficking and recycling maintain a pool
nalization. Neuron 25(3):649–62. of mobile surface AMPA receptors required for synaptic
Mattson MP. 2007. Mitochondrial regulation of neuronal plas- potentiation. Neuron 63(1):92–105.
ticity. Neurochem Res 32(4-5):707–15. Poodry CA, Edgar L. 1979. Reversible alteration in the neu-
Maurer I, Zierz S, Moller HJ. 2000. A selective defect of cyto- romuscular junctions of Drosophila melanogaster bear-
chrome c oxidase is present in brain of Alzheimer disease ing a temperature-sensitive mutation, shibire. J Cell Biol
patients. Neurobiol Aging 21(3):455–62. 81(3):520–7.
Mears JA, Lackner LL, Fang S, Ingerman E, Nunnari J, Praefcke GJ, McMahon HT. 2004. The dynamin superfamily:
Hinshaw JE. 2011. Conformational changes in Dnm1 sup- universal membrane tubulation and fission molecules? Nat
port a contractile mechanism for mitochondrial fission. Nat Rev Mol Cell Biol 5(2):133–47.
Struct Mol Biol 18(1):20–6. Raimondi A, Ferguson SM, Lou X, Armbruster M, Paradise
Min L, Leung YM, Tomas A, Watson RT, Gaisano HY, Halban S, Giovedi S, and others. 2011. Overlapping role of dyna-
PA, and others. 2007. Dynamin is functionally coupled to min isoforms in synaptic vesicle endocytosis. Neuron
insulin granule exocytosis. J Biol Chem 282(46):33530–6. 70(6):1100–14.
Mishra P, Chan DC. 2016. Metabolic regulation of mitochon- Rappoport JZ, Simon SM. 2003. Real-time analysis of clath-
drial dynamics. J Cell Biol 212(4):379–87. rin-mediated endocytosis during cell migration. J Cell Sci
Montgomery JM, Selcher JC, Hanson JE, Madison DV. 116(Pt 5):847–55.
2005. Dynamin-dependent NMDAR endocytosis dur- Richards DA, Guatimosim C, Rizzoli SO, Betz WJ. 2003.
ing LTD and its dependence on synaptic state. BMC Synaptic vesicle pools at the frog neuromuscular junction.
Neurosci 6:48. Neuron 39(3):529–41.
Arriagada-Diaz et al. 17

Salim K, Bottomley MJ, Querfurth E, Zvelebil MJ, Gout I, dynamin block an intermediate stage in coated vesicle for-
Scaife R, and others. 1996. Distinct specificity in the rec- mation. J Cell Biol 122(3):553–63.
ognition of phosphoinositides by the pleckstrin homology Vanstone JR, Smith AM, McBride S, Naas T, Holcik M, Antoun
domains of dynamin and Bruton’s tyrosine kinase. EMBO G, and others. 2016. DNM1L-related mitochondrial fission
J 15(22):6241–50. defect presenting as refractory epilepsy. Eur J Hum Genet
Samasilp P, Chan SA, Smith C. 2012. Activity-dependent fusion 24(7):1084–8.
pore expansion regulated by a calcineurin-dependent dyna- Verstreken P, Ly CV, Venken KJ, Koh TW, Zhou Y, Bellen HJ.
min-syndapin pathway in mouse adrenal chromaffin cells. 2005. Synaptic mitochondria are critical for mobilization
J Neurosci 32(30):10438–47. of reserve pool vesicles at Drosophila neuromuscular junc-
Selkoe DJ. 2001. Alzheimer’s disease: genes, proteins, and tions. Neuron 47(3):365–78.
therapy. Physiol Rev 81(2):741–66. von Spiczak S, Helbig KL, Shinde DN, Huether R, Pendziwiat
Selkoe DJ. 2002. Alzheimer’s disease is a synaptic failure. M, Lourenco C, and others. 2017. DNM1 encepha-
Science 298(5594):789–91. lopathy: a new disease of vesicle fission. Neurology
Shields LY, Kim H, Zhu L, Haddad D, Berthet A, Pathak D, 89(4):385–394.
and others. 2015. Dynamin-related protein 1 is required Wang L, Barylko B, Byers C, Ross JA, Jameson DM, Albanesi
for normal mitochondrial bioenergetic and synaptic JP. 2010. Dynamin 2 mutants linked to centronuclear
function in CA1 hippocampal neurons. Cell Death Dis myopathies form abnormally stable polymers. J Biol Chem
6:e1725. 285(30):22753–7.
Shin W, Ge L, Arpino G, Villarreal SA, Hamid E, Liu H, and Wang W, Ju YY, Zhou QX, Tang JX, Li M, Zhang L, and oth-
others. 2018. Visualization of membrane pore in live cells ers. 2017. The small GTPase Rac1 Contributes to extinction
reveals a dynamic-pore theory governing fusion and endo- of aversive memories of drug withdrawal by facilitating
cytosis. Cell 173(4):934–945.e12. GABAA receptor endocytosis in the vmPFC. J Neurosci
Sidiropoulos PN, Miehe M, Bock T, Tinelli E, Oertli CI, Kuner 37(30):7096–110.
R, and others. 2012. Dynamin 2 mutations in Charcot- Warnock DE, Schmid SL. 1996. Dynamin GTPase, a force-gen-
Marie-Tooth neuropathy highlight the importance of erating molecular switch. Bioessays 18(11):885–93.
clathrin-mediated endocytosis in myelination. Brain 135(Pt Watanabe S, Liu Q, Davis MW, Hollopeter G, Thomas N,
5):1395–411. Jorgensen NB, and others. 2013. Ultrafast endocytosis at
Singh M, Denny H, Smith C, Granados J, Renden R. 2018. Caenorhabditis elegans neuromuscular junctions. Elife
Presynaptic loss of dynamin-related protein 1 impairs syn- 2:e00723.
aptic vesicle release and recycling at the mouse calyx of Watanabe S, Trimbuch T, Camacho-Perez M, Rost BR,
Held. J Physiol 596(24):6263–87. Brokowski B, Sohl-Kielczynski B, and others. 2014.
Soykan T, Kaempf N, Sakaba T, Vollweiter D, Goerdeler F, Clathrin regenerates synaptic vesicles from endosomes.
Puchkov D, and others. 2017. Synaptic vesicle endocytosis Nature 515(7526):228–33.
occurs on multiple timescales and is mediated by formin- Watanabe T, Iwasaki K, Takasaki K, Yamagata N, Fujino M,
dependent actin assembly. Neuron 93(4):854–866.e4. Nogami A, and others. 2010. Dynamin 1 depletion and
Tanabe K, Takei K. 2009. Dynamic instability of microtubules memory deficits in rats treated with Aβ and cerebral isch-
requires dynamin 2 and is impaired in a Charcot-Marie- emia. J Neurosci Res 88(9):1908–17.
Tooth mutant. J Cell Biol 185(6):939–48. Waterham HR, Koster J, van Roermund CW, Mooyer PA,
Tanifuji S, Funakoshi-Tago M, Ueda F, Kasahara T, Mochida S. Wanders RJ, Leonard JV. 2007. A lethal defect of
2013. Dynamin isoforms decode action potential firing for mitochondrial and peroxisomal fission. N Engl J Med
synaptic vesicle recycling. J Biol Chem 288(26):19050–9. 356(17):1736–41.
Tardin C, Cognet L, Bats C, Lounis B, Choquet D. 2003. Direct Wenger J, Klinglmayr E, Frohlich C, Eibl C, Gimeno A,
imaging of lateral movements of AMPA receptors inside Hessenberger M, and others. 2013. Functional mapping
synapses. EMBO J 22(18):4656–65. of human dynamin-1-like GTPase domain based on x-ray
Tilokani L, Nagashima S, Paupe V, Prudent J. 2018. structure analyses. PLoS One 8(8):e71835.
Mitochondrial dynamics: overview of molecular mecha- Wenzel EM, Morton A, Ebert K, Welzel O, Kornhuber
nisms. Essays Biochem 62(3):341–60. J, Cousin MA, and others. 2012. Key physiological
Todorova V, Blokland A. 2017. Mitochondria and synaptic parameters dictate triggering of activity-dependent
plasticity in the mature and aging nervous system. Curr bulk endocytosis in hippocampal synapses. PLoS One
Neuropharmacol 15(1):166–73. 7(6):e38188.
Tsuboi T, McMahon HT, Rutter GA. 2004. Mechanisms of Williams M, Kim K. 2014. From membranes to organelles:
dense core vesicle recapture following “kiss and run” emerging roles for dynamin-like proteins in diverse cellular
(“cavicapture”) exocytosis in insulin-secreting cells. J Biol processes. Eur J Cell Biol 93(7):267–77.
Chem 279(45):47115–24. Wong MY, Cavolo SL, Levitan ES. 2015. Synaptic neuropep-
van Dam EM, Stoorvogel W. 2002. Dynamin-dependent trans- tide release by dynamin-dependent partial release from cir-
ferrin receptor recycling by endosome-derived clathrin- culating vesicles. Mol Biol Cell 26(13):2466–74.
coated vesicles. Mol Biol Cell 13(1):169–82. Wu Y, Chen M, Jiang J. 2019. Mitochondrial dysfunction in
van der Bliek AM, Redelmeier TE, Damke H, Tisdale EJ, neurodegenerative diseases and drug targets via apoptotic
Meyerowitz EM, Schmid SL. 1993. Mutations in human signaling. Mitochondrion 49:35–45.
18 The Neuroscientist 00(0)

Wu Y, O’Toole ET, Girard M, Ritter B, Messa M, Liu X, and Yudowski GA, Puthenveedu MA, Leonoudakis D, Panicker S,
others. 2014. A dynamin 1-, dynamin 3- and clathrin-inde- Thorn KS, Beattie EC, and others. 2007. Real-time imaging
pendent pathway of synaptic vesicle recycling mediated by of discrete exocytic events mediating surface delivery of
bulk endocytosis. Elife 3:e01621. AMPA receptors. J Neurosci 27(41):11112–21.
Yamada H, Abe T, Satoh A, Okazaki N, Tago S, Kobayashi K, Zhang R, Lee DM, Jimah JR, Gerassimov N, Yang C, Kim
and others. 2013. Stabilization of actin bundles by a dyna- S, and others. 2020. Dynamin regulates the dynamics
min 1/cortactin ring complex is necessary for growth cone and mechanical strength of the actin cytoskeleton as a
filopodia. J Neurosci 33(10):4514–26. multifilament actin-bundling protein. Nat Cell Biol
Yamashita T, Hige T, Takahashi T. 2005. Vesicle endocytosis 22(6):674–88.
requires dynamin-dependent GTP hydrolysis at a fast CNS Zheng N, Jeyifous O, Munro C, Montgomery JM, Green WN.
synapse. Science 307(5706):124–7. 2015. Synaptic activity regulates AMPA receptor traffick-
Yan J, Liu XH, Han MZ, Wang YM, Sun XL, Yu N, and others. ing through different recycling pathways. Elife 4:e06878.
2015. Blockage of GSK3β-mediated Drp1 phosphorylation Zhou Q, Homma KJ, Poo MM. 2004. Shrinkage of dendritic
provides neuroprotection in neuronal and mouse models of spines associated with long-term depression of hippocam-
Alzheimer’s disease. Neurobiol Aging 36(1):211–27. pal synapses. Neuron 44(5):749–57.
Yang Z, Li H, Chai Z, Fullerton MJ, Cao Y, Toh BH, and oth- Zuchner S, Noureddine M, Kennerson M, Verhoeven K, Claeys
ers. 2001. Dynamin II regulates hormone secretion in neu- K, De Jonghe P, and others. 2005. Mutations in the pleck-
roendocrine cells. J Biol Chem 276(6):4251–60. strin homology domain of dynamin 2 cause dominant
Yoon G, Malam Z, Paton T, Marshall CR, Hyatt E, Ivakine intermediate Charcot-Marie-Tooth disease. Nat Genet
Z, and others. 2016. Lethal disorder of mitochondrial fis- 37(3):289–94.
sion caused by mutations in DNM1L. J Pediatr 171:313– Zucker RS, Regehr WG. 2002. Short-term synaptic plasticity.
316.e1-2. Annu Rev Physiol 64:355–405.

You might also like