You are on page 1of 14

Handbook of Clinical Neurology, Vol.

184 (3rd series)


Neuroplasticity: From Bench to Bedside
A. Quartarone, M.F. Ghilardi, and F. Boller, Editors
https://doi.org/10.1016/B978-0-12-819410-2.00024-2
Copyright © 2022 Elsevier B.V. All rights reserved

Chapter 30

Multiple sclerosis: Inflammation, autoimmunity and plasticity


MARIO STAMPANONI BASSI1, ENNIO IEZZI1, AND DIEGO CENTONZE1,2*

1
Unit of Neurology & Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
2
Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy

Abstract
In recent years, experimental studies have clarified that immune system influences the functioning of the
central nervous system (CNS) in both physiologic and pathologic conditions. The neuro-immune crosstalk
plays a crucial role in neuronal development and may be critically involved in mediating CNS response to
neuronal damage. Multiple sclerosis (MS) represents a good model to investigate how the immune system
regulates neuronal activity. Accordingly, a growing body of evidence has demonstrated that increased
levels of pro-inflammatory mediators may significantly impact synaptic mechanisms, influencing overall
neuronal excitability and synaptic plasticity expression. In this chapter, we provide an overview of pre-
clinical data and clinical studies exploring synaptic functioning noninvasively with transcranial magnetic
stimulation (TMS) in patients with MS. Moreover, we examine how inflammation-driven synaptic dys-
function could affect synaptic plasticity expression, negatively influencing the MS course. Contrasting
CSF inflammation together with pharmacologic enhancement of synaptic plasticity and application of
noninvasive brain stimulation, alone or in combination with rehabilitative treatments, could improve
the clinical compensation and prevent the accumulating deterioration in MS.

INTRODUCTION
and remissions, the disease dramatically shifts towards a
Multiple sclerosis (MS) is an autoimmune-mediated slowly increasing accumulation of disability without
inflammatory and degenerative disorder of the central clear relapses (secondary progressive, SP-MS). In a
nervous system (CNS), representing one of the most small portion of patients, the disease shows an essentially
common causes of neurologic disability among young progressive course from the onset (primary progressive,
adults. The clinical course can vary from benign to PP-MS).
rapidly evolving forms, with most patients showing a The ability of the CNS to compensate for new demy-
relapsing–remitting course (RR-MS), characterized by elinating lesions or neuronal loss importantly affects the
recurrent manifestations of acute neurologic deficits fol- course of MS and could contribute to explain the differ-
lowed by complete or partial recovery. Clinical relapses ent disease phenotypes and the discrepancies observed
are correlated with the appearance of focal lesions dis- between MRI findings and disability. Long-term poten-
closed by magnetic resonance imaging (MRI), although tiation (LTP) is one of the main neurophysiologic synap-
newly developed lesions could be clinically silent. Nev- tic mechanisms involved in clinical recovery after
ertheless, the association between the degree of brain damage (Centonze et al., 2007a). Accordingly, it has
damage detectable at MRI and disability is fairly weak been proposed that efficiency of LTP-like plasticity
(Filippi et al., 1995; Barkhof, 1999; Kappos et al., could critically influence the clinical course of MS
1999). In some cases, after a variable period of relapses (Weiss et al., 2014; Stampanoni Bassi et al., 2019).

*Correspondence to: Diego Centonze, Unit of Neurology & Neurorehabilitation, IRCCS Neuromed, Via Atinense 18, 86077
Pozzilli, (IS), Italy. Tel: +39-0865-929170, Fax: +39-0865-929259, E-mail: centonze@uniroma2.it
458 M. STAMPANONI BASSI ET AL.
Experimental studies in animal models (i.e., experi- homeostatic plasticity (i.e. synaptic scaling) (Schneider
mental autoimmune encephalomyelitis, EAE) and in et al., 1998; Beattie, 2002; Avital et al., 2003;
MS patients have shown that specific proinflammatory Stellwagen and Malenka, 2006). In pathologic condi-
cytokines involved in the pathogenesis of MS also alter tions, both acutely or chronically increased levels of
synaptic transmission and plasticity, negatively affecting pro-inflammatory mediators may significantly impact
the disease course (Centonze et al., 2009; Di Filippo synaptic functioning, influencing the overall neuronal
et al., 2013; Nisticò et al., 2013; Stampanoni Bassi excitability and synaptic plasticity expression.
et al., 2019). MS could therefore represent a useful model
to investigate how the immune system regulates neuronal
SYNAPTIC INFLAMMATION IN MS:
functioning and vice-versa.
PRECLINICAL STUDIES
Alterations of synaptic transmission have been
NEURO-IMMUNE CROSSTALK IN MS
described in the striatum of EAE mice starting from
MS has been classically considered a white matter the early phases, together with increased concentrations
pathology characterized by inflammatory demyelin- of pro-inflammatory molecules and microglial activa-
ation, while atrophy is observed in late phases in tion (Centonze et al., 2009). In particular, enhanced
response to diffuse axonal transection and retrograde excitatory synaptic transmission has been associated
neuronal death. Adaptive immune response has a pivotal with amplified expression and function of the a-amino-
role in the pathogenesis of MS and autoreactive T cells 3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)
and autoantibodies represent the key players that induce glutamate receptor (Centonze et al., 2009). Synaptic
and maintain the inflammatory process and mediate hyperexcitability has been related to excitotoxic neuro-
demyelination (Chitnis, 2007; Arneth, 2019). Specific nal damage and dendritic spine loss, suggesting that
inflammatory mediators, including tumor necrosis factor neurodegeneration in MS could primarily hinge on
(TNF), interleukin (IL)-1b, and IL-6, released by acti- inflammatory synaptopathy independently of demye-
vated T lymphocytes and resident immune cells promote lination. The proinflammatory cytokine TNF has been
the entry of infiltrating lymphocytes into the CNS, alter identified as the major player of the excitotoxic-induced
the blood brain barrier and amplify the immune response neurodegeneration. Accordingly, in vitro TNF incuba-
(Larochelle et al., 2011; Arneth, 2019). tion reproduced the synaptic alterations observed in
Studies in animal models and in MS patients have EAE and the administration of TNF inhibitors or AMPA
demonstrated that the same inflammatory molecules receptor blockers reversed both the neurophysiologic alter-
significantly affect synaptic transmission and plasticity ations and dendritic loss (Centonze et al., 2009). The pro-
(Centonze et al., 2009; Stampanoni Bassi et al., 2017a), inflammatory molecule IL-1b has also been consistently
suggesting that inflammatory synaptopathy may repre- associated to the synaptic alterations of EAE. One study
sent a specific feature of MS (Mandolesi et al., 2015). showed that IL-1b pathologically enhances glutamatergic
In recent years, experimental studies have contributed transmission in the cerebellum of EAE mice altering
to clarify the relationship between the immune system the expression of the glutamate–aspartate transporter/
and the CNS. Although peripheral immune cells have excitatory amino acid transporter 1 (GLAST/EAAT1) on
limited access to the brain under physiologic conditions, cerebellar astrocytes, thus reducing glutamate reuptake
the relatively recent discovery of its own lymphatic (Mandolesi et al., 2013). In addition, impaired inhibitory
drainage system, represented particularly in the menin- synaptic transmission has been reported in EAE. Specifi-
ges, has led to a revision of the notion of CNS immune cally, TNF and IL-1b have been associated with reduced
privilege (Meyer et al., 2017; De Mesquita et al., 2018). It GABAergic transmission (Rossi et al., 2011a; Mandolesi
has been observed that different cytokines and chemo- et al., 2012), thus confirming the role of these molecules
kines, released by resident immune cells and lympho- in promoting hyperexcitability.
cytes, regulated neuronal functioning, suggesting that Inflammation also plays a role in the alteration of
immune mediators may represent a major communica- synaptic plasticity observed in EAE. Different forms of
tion system in the CNS together with neurotransmitters synaptic plasticity have been described and LTP repre-
and neurotrophins (Kipnis, 2016; De Mesquita et al., sents the most studied. LTP consists in a long-lasting
2018). This neuro-immune crosstalk plays a crucial role enhancement of synaptic excitability and has been linked
in neuronal development and can be critically involved in to learning and memory processes (Bliss and Lømo,
mediating the CNS response to neuronal damage. 1973, see also Chapter 2), as well as to clinical recovery
It has been shown that TNF and IL-1b regulate the (Centonze et al., 2007a). LTP is associated to increased
physiologic expression of different forms of synaptic size, density, and clustering of synaptic spines (Desmond
plasticity, including Hebbian plasticity (i.e. LTP) and and Levy, 1986; Engert and Bonhoeffer, 1999;
MULTIPLE SCLEROSIS: INFLAMMATION, AUTOIMMUNITY AND PLASTICITY 459
De Roo et al., 2008). LTP requires the activation of of excitatory and inhibitory intracortical circuits
the N-methyl-D-aspartate (NMDA) receptors (Bliss and (Rossini et al., 2015).
Collingridge, 1993) and could be induced by high- Studies in patients with MS have demonstrated asso-
frequency repetitive stimulation (Bliss and Lømo, ciations between different TMS parameters and clinical/
1973). Conversely, low-frequency stimulation is associ- demographic characteristics. For instance, prolonged
ated to long-term depression (LTD) of synaptic activity central motor conduction time (CMCT) and increased
(Mulkey and Malenka, 1992; Kirkwood and Bear, 1994). resting motor threshold (RMT) have been previously
Altered LTP induction has been demonstrated in the reported particularly in patients with progressive MS
hippocampus during the acute phase of EAE. One study phenotype (Conte et al., 2009; Ayache et al., 2015;
reported that impaired LTP induction is paralleled by Neva et al., 2016). CMCT tests the integrity of the
hippocampal microglia activation and increased expres- cortico-spinal tract (Rossini et al., 2015), whereas
sion of IL-1b (Di Filippo et al., 2013). Pathologically RMT provides information on pyramidal cells excitabil-
increased LTP expression has also been described in ity within M1 including the influence of neighboring
EAE: IL-1b incubation in control hippocampal slices cortical neurons and the excitability of spinal motor
induced paradoxical LTP in response to an LTD-inducing neurons (Rossini et al., 1999).
protocol, and this finding has been possibly related to Moreover, increased cortical silent period (CSP) dura-
reduced GABAergic signaling (Nisticò et al., 2013). It tion and reduced short-interval intracortical inhibition
has also been shown that other pro-inflammatory mole- (SICI) have been related to structural damage, clinical
cules modulate synaptic plasticity expression in vitro. disability, MS phenotype and the presence of fatigue
For example, IL-6 is a major pro-inflammatory cytokine (Conte et al., 2009; Ayache et al., 2015; Nantes et al.,
that has also been associated to LTP disruption when 2016a,b; Chaves et al., 2019). CSP, measured as an inter-
incubated in mice hippocampal slices (Gruol, 2015; ruption of the voluntary electromyographic activity after
Stampanoni Bassi et al., 2019). the TMS pulse, partly reflects the inhibitory activity
Studies in EAE have shown that, in addition to within M1 that is mediated by GABA-B receptors
LTP and LTD, other forms of synaptic plasticity, partic- (Fuhr et al., 1991; Inghilleri et al., 1993; Siebner et al.,
ularly synaptic scaling, can be altered by inflammatory 1998) and SICI, tested with a TMS paired-pulse para-
cytokines. Synaptic scaling depends on increased or digm, evaluates the activity of GABA-A receptors
decreased expression of AMPA receptors and represents (Kujirai et al., 1993; Di Lazzaro et al., 2000). Other
a homeostatic form of plasticity aimed at maintaining paired-pulse TMS protocols are commonly used to test
neuronal excitability within physiologic ranges (Lissin the excitatory interactions within M1. Intracortical
et al., 1998; Turrigiano et al., 1998). In EAE, TNF signal- facilitation (ICF) engages glutamatergic transmission
ing has been associated to pathologically increased requiring the activation of NMDA receptors (Kujirai
upscaling of excitatory synapses, leading to glutamate- et al., 1993; Ziemann et al., 1998; Schwenkreis et al.,
mediated excitotoxicity and synaptic loss (Centonze 1999). In addition, short-interval intracortical facilitation
et al., 2009; Rizzo et al., 2018). In addition, elevated (SICF) could be mediated by facilitatory I-waves interac-
TNF levels have been found in the cerebrospinal fluid tions (Tokimura et al., 1996; Ziemann et al., 1998, 2015;
(CSF) of patients with progressive MS (Rossi et al., Hanajima et al., 2003), although the exact mechanisms
2014a). Notably, it has been demonstrated that incubat- of SICF have not been completely elucidated (Paulus
ing the CSF of patients with progressive MS phenotype et al., 2008; Van den Bos et al., 2018). In MS, a negative
in slices of control mice induced an increase of gluta- correlation between clinical disability and SICF has been
matergic transmission and neuronal swelling caused by found (Mori et al., 2013a). Although these measures
TNF (Rossi et al., 2014a). have clearly shown that also in patients with MS there
are alterations of inhibitory and excitatory transmission,
several differences between studies about MS pheno-
type, disability, and disease phase, complicate the inter-
SYNAPTIC ACTIVITY
pretation of the results.
IN PATIENTS WITH MS
Further results have convincingly suggested that
Synaptic activity and plasticity can be explored noninva- altered TMS measures of cortical excitability and synap-
sively in humans with transcranial magnetic stimulation tic activity in MS could be specifically linked to central
(TMS) (Rossini and Rossi, 2007; Ziemann et al., 2008; inflammation. One study reported both reduced CSP
Rossini et al., 2015). Different TMS protocols can be duration and SICI in patients with relapsing MS, indicat-
used to probe the excitability of the primary motor ing that acute inflammation could specifically impair M1
cortex (M1) to test the functionality of the cortico-spinal inhibitory activity (Caramia et al., 2004). Similarly, it has
tract and corpus callosum and to investigate the activity been demonstrated that CSF inflammation could affect
460 M. STAMPANONI BASSI ET AL.
M1 excitatory transmission. Enhanced IL-1b signaling of the paradoxical LTP-like response elicited by the
in patients with relapsing MS was positively correlated cTBS protocol (Mori et al., 2014a). These findings are
with the extent of ICF, suggesting a link between inflam- in line with the preclinical studies showing paradoxical
mation and glutamatergic transmission (Rossi et al., LTP induction in presence of IL-1b (Nisticò et al.,
2012). Furthermore, higher CSF levels of the pro- 2013), suggesting that this cytokine subverts LTP-like
inflammatory molecule RANTES have been associated plasticity mechanisms. Another proinflammatory cyto-
to increased ICF and reduced SICI in patients with remit- kine, IL-6, has also been recently associated to altered
ting RR-MS (Mori et al., 2016). Conversely, it has been synaptic plasticity in patients with MS, as the CSF levels
shown that the CSF levels of the anti-inflammatory mol- of this cytokine negatively correlated with the amount of
ecule IL-13 could exert beneficial effects, restoring SICI PAS-induced LTP-like plasticity (Stampanoni Bassi
(Rossi et al., 2011b). Overall, these data are in line et al., 2019). Conversely, other neurotrophins such as
with findings in EAE animal models and suggest that the platelet-derived growth factor (PDGF) could enhance
exacerbated central inflammation could promote cortical the paradoxical LTP induction by cTBS in patients with
hyperexcitability also in patients with MS. RR-MS, as the CSF levels of PDGF correlated with the
amount of LTP-like induced plasticity by cTBS (Mori
et al., 2013b).
SYNAPTIC PLASTICITY IN PATIENTS
It is worth mentioning that also the inflammation-
WITH MS
driven alteration of the metabolism of amyloid b (Ab)
TMS has been widely used to explore synaptic plasticity could contribute to the dysfunctional plasticity found
in humans, particularly in the motor cortex (Ziemann in patients with MS (Lassmann, 2011; Gentile et al.,
et al., 2008). The paired associative stimulation (PAS) 2015; Stampanoni Bassi et al., 2017b). Indeed, exacer-
protocol, which combines electrical stimulation of the bated inflammatory milieu has been specifically associ-
peripheral nerve with TMS of M1, elicits either LTP-like ated to altered Ab homeostasis, as in the CSF, levels of
or LTD-like modifications depending on the time inter- different pro-inflammatory molecules positively corre-
vals between the two types of stimuli (Stefan et al., late with the Ab1–42 levels, while anti-inflammatory mol-
2000) (see also Chapter 5). Another TMS protocol com- ecules showed a negative correlation with Ab1–42 levels
monly used to explore M1 plasticity is theta burst (Stampanoni Bassi et al., 2017b, 2019). Accordingly,
stimulation (TBS): in particular, continuous TBS (cTBS) reduced Ab1–42 CSF levels in patients with MS have
induces LTD-like effects, whereas intermittent TBS been linked to the presence of MRI gadolinium-
(iTBS) is associated to long-lasting increase of M1 excit- enhancing lesions. In addition, a negative correlation
ability that resembles LTP-like plasticity (Di Lazzaro was observed between the CSF levels of Ab1–42 and
et al., 2005; Huang et al., 2005) (see also Chapter 5). the amount of iTBS-induced LTP-like plasticity (Mori
TMS studies testing M1 plasticity in MS (Fig. 30.1A et al., 2011a).
and B) have shown that both PAS-induced LTP-like plas-
ticity and cTBS-induced LTD-like plasticity were similar
SYNAPTIC PLASTICITY EXPRESSION
in patients with remitting MS and controls (Zeller et al.,
AND CLINICAL RECOVERY IN MS
2010, 2012). Conversely, a simple repetitive motor task
produced a lower increase of MEP amplitude in patients It has been demonstrated that synaptic plasticity phe-
with RR-MS compared to healthy subjects (Stampanoni nomena, especially LTP, critically contributes to the
Bassi et al., 2020). compensation of symptoms following brain damage. In
Nevertheless, altered synaptic plasticity expression animal models, clinical recovery after experimental brain
has been associated to disease relapses, to CSF inflam- ischemia is strongly predicted by the amount of long-
mation and has been reported in progressive MS pheno- lasting increase of synaptic activity occurring in the
types (Fig. 30.1C–E). Accordingly, in patients with neighboring, spared neurons (Centonze et al., 2007a).
relapsing MS, iTBS protocols elicited LTP-like plasticity A similar role of LTP-like plasticity mechanisms has
that was reduced compared to controls but fully recov- been demonstrated in humans using TMS. In patients
ered after 3-month treatment with interferon b-1a with ischemic brain stroke, the efficiency of iTBS-
(Mori et al., 2012). Moreover, a study reported that a induced LTP-like plasticity in the acute phase correlated
PAS protocol inducing LTD produced a paradoxical with the amount of clinical recovery tested 6 months
LTP-like plasticity response in patients with relapsing later (Di Lazzaro et al., 2010). Similar results have also
MS (Wirsching et al., 2018). Specific inflammatory cyto- been reported in patients with relapsing RR-MS using the
kines have been implicated in the alterations of synaptic PAS protocol (Mori et al., 2014b).
plasticity in patients with MS. In patients with RR-MS, Physical rehabilitation represents the most useful
CSF levels of IL-1b positively correlated to the extent approach to promote recovery after neurologic injuries.
MULTIPLE SCLEROSIS: INFLAMMATION, AUTOIMMUNITY AND PLASTICITY 461

Fig. 30.1. Summary of TMS studies exploring synaptic plasticity in MS. LTP-like and LTD-like effects of different plasticity-
inducing protocols, TBS (black) and PAS (red). (A) Normal LTP-like plasticity: iTBS-induced and PAS-induced LTP-like plas-
ticity of patients with remitting MS was comparable to controls (Zeller et al., 2010; Mori et al., 2013b). (B) Normal LTD-like
plasticity: cTBS-induced LTD-like plasticity was similar in both patients with remitting MS and controls (Zeller et al., 2012).
(C) Reduced/absent LTP-like plasticity: in patients with relapsing MS, iTBS-induced LTP-like plasticity was reduced (Mori
et al., 2012) and correlated with reduced CSF Ab1–42 (Mori et al., 2011a). In patients with RR-MS, reduced PAS-induced
LTP-like plasticity correlated with increased CSF IL-6 levels (Stampanoni Bassi et al., 2019). In progressive MS patients
LTP-like plasticity induced by both iTBS and PAS was absent (Mori et al., 2013b; Nicoletti et al., 2019). (D) Abnormal LTP-like
plasticity: in remitting MS patients paradoxical LTP-like in response to an LTD-inducing protocol (cTBS) was correlated with
increased CSF PDGF levels (Mori et al., 2013b). In RR-MS patients paradoxical cTBS-induced LTP-like response was also cor-
related with increased CSF IL-1b levels (Mori et al., 2014a). In relapsing MS patients an LTD-inducing protocol (PAS10) elicited
LTP-like plasticity (Wirsching et al., 2018). (E) Absent LTD-like plasticity: no effect of an LTD-inducing plasticity protocol (cTBS)
in progressive MS patients (Mori et al., 2013b). Abbreviations: Ab1–42, amyloid-beta-1-42; CSF, cerebrospinal fluid; cTBS, contin-
uous TBS; IL, interleukin; iTBS, intermittent TBS; LTD, long-term depression; LTP, long-term potentiation; MS, multiple sclerosis;
PAS, paired associative stimulation; PDGF, platelet-derived growth factor; RR, relapsing–remitting; TBS, theta burst stimulation;
TMS, transcranial magnetic stimulation.
462 M. STAMPANONI BASSI ET AL.
The beneficial effects of rehabilitation on clinical recov- synaptic compensatory mechanisms, causing increased
ery have been previously explored in animal models. disability load even without new lesions (Mori et al.,
Exposing rats with focal brain ischemic damage to 2013b; Stampanoni Bassi et al., 2017a).
enriched rehabilitation strongly improved motor func-
tion with a specific increase in dendritic arborization
POSSIBLE APPROACHES TO BOOST
and number of spines in surviving neurons (Biernaskie
SYNAPTIC PLASTICITY IN MS
and Corbett, 2001). Similarly, physical exercise dramat-
ically improved the clinical impact of EAE, reducing the The key role of LTP in favoring clinical recovery after
dendritic spine loss and the typical synaptic alterations neurologic injuries makes LTP modulation an appealing
(Rossi et al., 2009). The relationship between motor treatment option. The observation that physical exercise
activity and LTP-like plasticity in humans has been can enhance LTP by influencing the expression of neuro-
explored with TMS: following motor practice, PAS- trophins, cannabinoids, and inflammatory cytokines,
induced LTP plasticity did not develop, suggesting a sat- strongly supports the use of physical rehabilitation as
uration effect due to metaplasticity (Ziemann et al., 2004; an active treatment, in both acute relapses and progres-
Stefan et al., 2006). Remarkably, it has been highligh- sive MS phenotypes. It has been recently suggested
ted that motor exercise interacted with PAS-induced that practice-dependent LTP in M1 following motor
LTP-like plasticity only in presence of motor learning learning of a simple task is reduced, in nondisabled
(Ziemann et al., 2004), suggesting that the beneficial patients in early stages of RR-MS (Stampanoni Bassi
effects of physical rehabilitation could be linked to et al., 2020). This highlights the need for additional inter-
practice-dependent LTP. ventions specifically aimed at increasing the expression
of LTP in MS.
In different neurologic conditions, it has been shown
INFLAMMATION NEGATIVELY
that pharmacologic enhancement of LTP could promote
INFLUENCES THE COURSE OF MS
recovery alone or in combination with rehabilitative
There is good evidence that inflammation that alters treatments (Dam et al., 1996; Scheidtmann et al., 2001;
the expression of synaptic plasticity may influence the R€osser et al., 2008). Synaptic plasticity mechanisms
mechanisms of brain damage compensation, therefore are regulated by different signaling pathways that have
worsening the disease course. Different studies have been extensively investigated in experimental studies,
shown that elevated CSF levels of specific pro- including NMDA receptors and endocannabinoids. Both
inflammatory molecules at the time of diagnosis, includ- in vitro and in vivo studies have clearly demonstrated
ing IL-1b, IL-6, and IL8, were associated to enhanced that stimulation of NMDA receptors is critical for induc-
disease activity and prospective disability (Rossi et al., ing LTP (see Chapter 2) (Bliss and Collingridge, 1993).
2014b, 2015; Stampanoni Bassi et al., 2019). In a group Accordingly, the administration of memantine, a NMDA
of patients with RR-MS, elevated CSF levels of IL-6 at receptor antagonist, causes blockade of iTBS-induced
the time of diagnosis predicted a greater number of symp- LTP in healthy humans (Huang et al., 2007) and the reap-
tomatic relapses, thus indicating an impaired ability to pearance of clinical signs and symptoms in patients with
compensate for the appearance of new brain lesions RR-MS (Villoslada et al., 2009). NMDA receptors activ-
(Stampanoni Bassi et al., 2019). Notably, the detrimental ity is strongly regulated by the D-amino D-aspartic acid
effect of IL-6 was accompanied by specific disruption of (D-Asp). Preclinical studies have shown that D-Asp is
LTP-like plasticity induced by PAS. Conversely, it has involved in LTP induction and dendritic spines formation
been proposed that some anti-inflammatory molecules in the hippocampus (D’Aniello et al., 1993; Errico et al.,
and neurotrophins could exert positive effects on the 2014). It has been recently suggested that D-Asp admin-
MS course (Sarchielli et al., 2002; Astier et al., 2006; istration may represent a useful approach to promote LTP
Vana et al., 2007; Mayo et al., 2016). In fact, elevated expression in MS patients. Accordingly, in patients with
CSF levels of PDGF could favor a stable disease course progressive MS, a 4-week administration of D-Asp par-
(Stampanoni Bassi et al., 2018): PDGF can promote LTP tially restored LTP-like plasticity explored with PAS
induction both in vitro (Peng et al., 2010) and in patients (Nicoletti et al., 2019), thus opening the way for inves-
with RR-MS (Mori et al., 2013b), while PDGF expres- tigating its clinical effectiveness to limit the disability
sion is absent in the CSF of patients with progressive progression in these patients.
MS (Mori et al., 2013b). Indeed, in patients with progres- The cannabinoid system is also involved in the
sive MS, LTP-like plasticity is absent with different regulation of LTP mechanisms. In particular, stimulation
TMS protocols (Mori et al., 2013b; Nicoletti et al., of cannabinoid type 1 receptors (CB1Rs) is required for
2019). Therefore, it is likely that, in progressive MS, the induction of LTP, which is absent in mice lacking the
the absence of LTP may express the disruption of CB1Rs (Heifets and Castillo, 2009; Madroñal et al., 2012).
Table 30.1
Studies using noninvasive brain stimulation to treat MS-related symptoms

Study Patients Protocol Area Symptom Main clinical finding

Nielsen et al. (1996) RR-MS (remitting) rTMS (25 Hz) 7-days Thoracic spinal cord Spasticity rTMS reduced spasticity
Centonze et al. (2007b) RR-MS (remitting) rTMS (5 Hz) 10-days Leg M1 Spasticity rTMS induced long-lasting improvement of spasticity
Centonze et al. (2007c) RR-MS (remitting) rTMS (5 Hz) 10-days Leg M1 Urinary rTMS improved urinary symptoms with beneficial
effects on the bladder voiding phase
Koch et al. (2008) RR-MS (remitting) rTMS (5 Hz) single M1 Hand dexterity rTMS improved hand dexterity in patients with
session cerebellar symptoms
Mori et al. (2010a) RR-MS (remitting) TDCS 5-days M1 Pain Anodal TDCS reduced chronic central pain
Mori et al. (2010b) RR-MS (remitting) iTBS 10-days Leg M1 Spasticity iTBS reduced spasticity
Mori et al. (2011b) RR-MS (remitting) iTBS  ET 10-days Leg M1 Spasticity iTBS + ET reduced spasticity and improved quality of
life and functional measures more consistently than
iTBS alone
Mori et al. (2013c) RR-MS (remitting) TDCS 5-days Somatosensory Sensory Anodal TDCS improved sensory deficits
Deficits
Leocani et al. (2012) Progressive MS rTMS (20 Hz) Leg M1 Spasticity 3-week treatment with rTMS using H-coil over
11 sessions bilateral leg M1 improved spasticity and walking
Ferrucci et al. (2014) RR-MS TDCS 5-days M1 Fatigue Bilateral anodal TDCS reduced fatigue
Tecchio et al. (2015) RR-MS (remitting) TDCS 5-days Somatosensory Fatigue Bilateral anodal TDCS reduced fatigue
Saiote et al. (2014) RR-MS (remitting) TDCS 5-days Left DLPFC Fatigue Anodal TDCS had no effect on fatigue
Mattioli et al. (2015) RR-MS (remitting) TDCS 10 sessions Left DLPFC Cognitive Anodal TDCS during cognitive training improved
attention and executive functions
Burhan et al. (2015) RR-MS rTMS (6-Hz) 4 Left DLPFC Gait rTMS improved gait parameters in a single patient
sessions
Tecchio et al. (2015) RR-MS (remitting) TDCS 5-days Somatosensory/hand Fatigue Bilateral whole-body somatosensory stimulation
sensorimotor reduced fatigue
Iodice et al. (2015) RR-MS (remitting) TDCS 5-days M1 Spasticity Anodal tDCS had no effect on spasticity
Ayache et al. (2016) RR/SP/PP-MS TDCS 3-days Left DLPFC Pain Anodal tDCS reduced neuropathic pain
Elzamarany et al. RR/SP-MS rTMS (5 Hz) M1 Hand dexterity rTMS improved hand dexterity in patients with
(2016) 2- sessions cerebellar symptoms
Boutière et al. (2017) RR-MS iTBS 13-days Leg M1 Spasticity iTBS reduced spasticity
(remitting)/
SP-MS
Chalah et al. (2017a) RR-MS (remitting) TDCS 4-weeks Left DLPFC Fatigue Anodal tDCS reduced fatigue in a single patient
Chalah et al. (2017b) RR-MS (remitting) TDCS 5-days Left DLPFC/Right Fatigue Anodal tDCS over DLPFC reduced fatigue
PPC

Continued
Table 30.1
Continued

Study Patients Protocol Area Symptom Main clinical finding

Charvet et al. (2018a) RR-MS TDCS 10/20-sessions Left DLPFC Fatigue Anodal tDCS reduced fatigue
(remitting)/
SP/PP
Charvet et al. (2018b) RR (remitting)/ TDCS + CT Left DLPFC Cognitive Anodal tDCS improved complex attention and
SP/PP 10-sessions response variability
Cancelli et al. (2018) RR-MS (remitting) TDCS 5-days Somatosensory Fatigue Bilateral anodal whole-body somatosensory
stimulation reduced fatigue
Fiene et al. (2018) RR-MS TDCS single session Left DLPFC Fatigue Anodal tDCS reduced fatigue-related deterioration in
(remitting)/ cognitive performance
SP-MS
Cosentino et al. (2018) RR/SP-MS TDCS 5-days Right swallowing Dysphagia Anodal tDCS improved dysphagia
motor cortex
Restivo et al. (2019) RR (remitting)/ TDCS 5-days Dominant pharyngeal Dysphagia Anodal tDCS improved dysphagia in patients with
SP-MS motor cortex brainstem involvement
Korzhova et al. (2019) SP-MS rTMS (20 Hz)/iTBS Leg M1 Spasticity rTMS and iTBS reduced spasticity with a
10-days longer-lasting effect of iTBS
Berra et al. (2019) RR (remitting)/ TSDCS 10-days Thoracic spinal cord Pain Anodal tsDCS reduced neuropathic pain
SP/PP-MS

Abbreviations: CT, cognitive training; DLPFC, dorsolateral prefrontal cortex; ET, exercise therapy; iTBS, intermittent theta burst stimulation; MS, multiple sclerosis; M1, primary motor cortex (M1); PPC,
posterior parietal cortex; PP, primary progressive; RR, relapsing–remitting; rTMS, repetitive transcranial magnetic stimulation; SP, secondary progressive; TDCS, transcranial direct current stimulation;
TSDCS, transcutaneous spinal direct current stimulation.
MULTIPLE SCLEROSIS: INFLAMMATION, AUTOIMMUNITY AND PLASTICITY 465
In addition, the administration of a cannabis-based med- Serono, Novartis, Roche, Sanofi-Genzyme, and Teva.
icine containing tetrahydrocannabinol and cannabidiol He is also the principal investigator in clinical trials for
(Sativex), was able to change in a group of RR-MS Bayer Schering, Biogen, Merck Serono, Mitsubishi,
patients the expected cTBS-induced inhibition into Novartis, Roche, Sanofi-Genzyme, and Teva. His preclin-
LTP-like effect (Koch et al., 2009). It has been demon- ical and clinical research was supported by grants from
strated that the endocannabinoid system is involved in Bayer Schering, Biogen Idec, Celgene, Merck Serono,
mediating the beneficial effects of exercise (Heyman Novartis, Roche, Sanofi-Genzyme and Teva. The funders
et al., 2012; Raichlen et al., 2012). Notably, a polymor- had no role in the design of the study; in the collection,
phism in the CB1R gene (long AAT) that reduces CB1R analyses, or interpretation of data; in the writing of the
protein expression, is also associated with reduced iTBS- manuscript, or in the decision to publish the results.
induced LTP and limited effect of motor rehabilitation M.S.B.: no conflict of interest.
(Mori et al., 2014c). E.I.: no conflict of interest.
Another interesting approach to increase synaptic
plasticity is the application of noninvasive brain stimula- REFERENCES
tion techniques. In the last decade, a number of studies
Arneth BM (2019). Impact of B cells to the pathophysiology of
have explored the potential beneficial effect of repetitive
multiple sclerosis. J Neuroinflammation 16 (1): 128.
TMS and transcranial direct current stimulation on sev- https://doi.org/10.1186/s12974-019-1517-1.
eral MS-related symptoms such as spasticity, pain, sensory Astier AL, Meiffren G, Freeman S et al. (2006). Alterations in
deficits, fatigue, cognitive disturbances and dysphagia, CD46-mediated Tr1 regulatory T cells in patients with mul-
with promising results (Table 30.1). In particular, it has tiple sclerosis. J Clin Invest 116 (12): 3252–3257. https://
been shown that noninvasive brain stimulation techniques doi.org/10.1172/JCI29251. PMID: 17099776.
can prime the effects of physical rehabilitation. Accord- Avital A, Goshen I, Kamsler A et al. (2003). Impaired
ingly, iTBS combined with exercise therapy led to a interleukin-1 signaling is associated with deficits in hippo-
greater improvement in motor function, fatigue, and qual- campal memory processes and neural plasticity. Hippocam-
ity of life, than exercise alone (Mori et al., 2011b). pus 13 (7): 826–834. https://doi.org/10.1002/hipo.10135.
Ayache SS, Creange A, Farhat WH et al. (2015). Cortical
excitability changes over time in progressive multiple scle-
CONCLUSIONS rosis. Funct Neurol 30: 257–263. PMID: 26727704.
Preclinical and clinical studies have shown that inflam- Ayache SS, Palm U, Chalah MA et al. (2016). Prefrontal
tDCS decreases pain in patients with multiple sclerosis.
matory synaptopathy alters synaptic plasticity mecha-
Front Neurosci 10: 147. https://doi.org/10.3389/fnins.2016.
nisms negatively influencing the ability to compensate 00147.
for new demyelinating lesions or neuronal loss in MS. Barkhof F (1999). MRI in multiple sclerosis: correlation with
Restoring synaptic plasticity could therefore represent expanded disability status scale (EDSS). Mult Scler 5:
a useful therapeutic strategy to promote a stable disease 283–286. PMID: 10467389.
course in patients with MS. Contrasting CSF inflammation Beattie EC (2002). Control of synaptic strength by glial TNFa.
together with pharmacologic enhancement of synaptic Science 295 (5563): 2282–2285. https://doi.org/10.1126/
plasticity and application of noninvasive brain stimulation, science.1067859. PMID: 11910117.
alone or in combination with rehabilitative treatments, Berra E, Bergamaschi R, De Icco R et al. (2019). The effects of
could improve the clinical compensation and prevent the transcutaneous spinal direct current stimulation on neuro-
accumulating deterioration in MS. pathic pain in multiple sclerosis: clinical and neurophysio-
logical assessment. Front Hum Neurosci 13: 31. https://doi.
org/10.3389/fnhum.2019.00031.
ACKNOWLEDGMENTS Biernaskie J, Corbett D (2001). Enriched rehabilitative train-
This work was supported by the Italian Ministry of Health ing promotes improved forelimb motor function and
enhanced dendritic growth after focal ischemic injury.
(Ricerca Corrente—IRCCS Neuromed and 5 x 1000—
J Neurosci 21 (14): 5272–5280. PMID: 11438602.
IRCCS Neuromed) and by FISM—Fondazione Italiana Bliss TV, Collingridge GL (1993). A synaptic model of mem-
Sclerosi Multipla- cod. 2019/S/1 to DC. ory: long-term potentiation in the hippocampus. Nature 361:
31–39. https://doi.org/10.1038/361031a0. PMID: 8421494.
CONFLICT OF INTEREST STATEMENT Bliss TV, Lømo T (1973). Long-lasting potentiation of synap-
tic transmission in the dentate area of the anaesthetized rab-
D.C. is an Advisory Board member of Almirall, Bayer bit following stimulation of the perforant path. J Physiol
Schering, Biogen, GW Pharmaceuticals, Merck Serono, 232: 331–356. PMID: 4727084.
Novartis, Roche, Sanofi-Genzyme, and Teva and received Boutière C, Rey C, Zaaraoui W et al. (2017). Improvement of
honoraria for speaking or consultation fees from Almirall, spasticity following intermittent theta burst stimulation
Bayer Schering, Biogen, GW Pharmaceuticals, Merck in multiple sclerosis is associated with modulation of
466 M. STAMPANONI BASSI ET AL.
resting-state functional connectivity of the primary motor Chitnis T (2007). The role of CD4 T cells in the pathogenesis of
cortices. Mult Scler 23 (6): 855–863. https://doi.org/ multiple sclerosis. Int Rev Neurobiol 79: 43–72PMID:
10.1177/1352458516661640. 17531837.
Burhan AM, Subramanian P, Pallaveshi L et al. (2015). Conte A, Lenzi D, Frasca V et al. (2009). Intracortical
Modulation of the left prefrontal cortex with high fre- excitability in patients with relapsing-remitting and
quency repetitive transcranial magnetic stimulation facili- secondary progressive multiple sclerosis. J Neurol 256:
tates gait in multiple sclerosis. Case Rep Neurol Med 2015: 933–938. https://doi.org/10.1007/s00415-009-5047-0.
251829. https://doi.org/10.1155/2015/251829. PMID: 19252788.
Cancelli A, Cottone C, Giordani A et al. (2018). Personalized, Cosentino G, Gargano R, Bonura G et al. (2018). Anodal tDCS
bilateral whole-body somatosensory cortex stimulation to of the swallowing motor cortex for treatment of dysphagia
relieve fatigue in multiple sclerosis. Mult Scler 24 (10): in multiple sclerosis: a pilot open-label study. Neurol Sci
1366–1374. https://doi.org/10.1177/1352458517720528. 39 (8): 1471–1473. https://doi.org/10.1007/s10072-018-
Caramia MD, Palmieri MG, Desiato MT et al. (2004). 3443-x.
Brain excitability changes in the relapsing and remit- D’Aniello A, Vetere A, Petrucelli L (1993). Further study on
ting phases of multiple sclerosis: a study with trans- the specificity of D-amino acid oxidase and D-aspartate
cranial magnetic stimulation. Clin Neurophysiol 115: oxidase and time course for complete oxidation of
956–965. https://doi.org/10.1016/j.clinph.2003.11.024. D-amino acids. Comp Biochem Physiol B 105: 731–734.
PMID: 15003779. Dam M, Tonin P, De Boni A et al. (1996). Effects of fluoxetine
Centonze D, Rossi S, Tortiglione A et al. (2007a). Synaptic and maprotiline on functional recovery in poststroke hemi-
plasticity during recovery from permanent occlusion of plegic patients undergoing rehabilitation therapy. Stroke
the middle cerebral artery. Neurobiol Dis 27 (1): 44–53. 27 (7): 1211–1214. https://doi.org/10.1161/01.str.27.7.
PMID: 17490888. 1211. PMID: 8685930.
Centonze D, Koch G, Versace V et al. (2007b). Repetitive tran- De Mesquita S, Fu Z, Kipnis J (2018). The meningeal lym-
scranial magnetic stimulation of the motor cortex amelio- phatic system: a new player in neurophysiology. Neuron
rates spasticity in multiple sclerosis. Neurology 68 (13): 100 (2): 375–388. https://doi.org/10.1016/j.neuron.2018.
1045–1050. https://doi.org/10.1212/01.wnl.0000257818. 09.022. PMID: 30359603.
16952.62. PMID: 17389310. De Roo M, Klauser P, Muller D (2008). LTP promotes a selec-
Centonze D, Petta F, Versace V et al. (2007c). Effects of motor tive long-term stabilization and clustering of dendritic
cortex rTMS on lower urinary tract dysfunction in multiple spines. PLoS Biol 6: e219. https://doi.org/10.1371/jour-
sclerosis. Mult Scler 13 (2): 269–271. https://doi.org/ nal.pbio.0060219. PMID: 18788894.
10.1177/135245850607072917439897. Desmond NL, Levy WB (1986). Changes in the postsynaptic
Centonze D, Muzio L, Rossi S et al. (2009). Inflammation density with long-term potentiation in the dentate gyrus.
triggers synaptic alteration and degeneration in experimen- J Comp Neurol 253: 476–482. PMID: 3025273.
tal autoimmune encephalomyelitis. J Neurosci 29 (11): Di Filippo M, Chiasserini D, Gardoni F et al. (2013). Effects
3442–3452. https://doi.org/10.1523/JNEUROSCI.5804-08. of central and peripheral inflammation on hippocampal
2009. PMID: 19295150. synaptic plasticity. Neurobiol Dis 52: 229–236. https://
Chalah MA, Lefaucheur JP, Ayache SS (2017a). Long-term doi.org/10.1016/j.nbd.2012.12.009. PMID: 23295855.
effects of tDCS on fatigue, mood and cognition in multiple Di Lazzaro V, Oliviero A, Meglio M et al. (2000). Direct
sclerosis. Clin Neurophysiol 128 (11): 2179–2180. https:// demonstration of the effect of lorazepam on the excitabi-
doi.org/10.1016/j.clinph.2017.08.004. lity of the motor cortex. Clin Neurophysiol 111: 794–799.
Chalah MA, Riachi N, Ahdab R et al. (2017b). Effects of left https://doi.org/10.1016/s1388-2457(99)00314-4. PMID:
DLPFC versus right PPC tDCS on multiple sclerosis 10802448.
fatigue. J Neurol Sci 372: 131–137. https://doi.org/10.1016/ Di Lazzaro V, Pilato F, Saturno E et al. (2005). Theta-burst
j.jns.2016.11.015. repetitive transcranial magnetic stimulation suppresses
Charvet LE, Dobbs B, Shaw MT et al. (2018a). Remotely specific excitatory circuits in the human motor cortex.
supervised transcranial direct current stimulation for the J Physiol 565: 945–950. PMID: 15845575.
treatment of fatigue in multiple sclerosis: results from a ran- Di Lazzaro V, Profice P, Pilato F et al. (2010). Motor
domized, sham-controlled trial. Mult Scler 24 (13): cortex plasticity predicts recovery in acute stroke. Cereb
1760–1769. https://doi.org/10.1177/1352458517732842. Cortex 20: 1523–1528. https://doi.org/10.1093/cercor/bhp
Charvet L, Shaw M, Dobbs B et al. (2018b). Remotely super- 216. PMID: 19805417.
vised transcranial direct current stimulation increases the Elzamarany E, Afifi L, El-Fayoumy NM et al. (2016). Motor
benefit of at-home cognitive training in multiple sclerosis. cortex rTMS improves dexterity in relapsing-remitting and
Neuromodulation 21 (4): 383–389. https://doi.org/10.1111/ secondary progressive multiple sclerosis. Acta Neurol Belg
ner.12583. 116 (2): 145–150. https://doi.org/10.1007/s13760-015-
Chaves AR, Kelly LP, Moore CS et al. (2019). Prolonged cor- 0540-y.
tical silent period is related to poor fitness and fatigue, but Engert F, Bonhoeffer T (1999). Dendritic spine changes
not tumor necrosis factor, in multiple sclerosis. Clin associated with hippocampal longterm synaptic plasticity.
Neurophysiol 130 (4): 474–483. https://doi.org/10.1016/ Nature 399: 66–70. https://doi.org/10.1038/19978. PMID:
j.clinph.2018.12.015. PMID: 30771724. 10331391.
MULTIPLE SCLEROSIS: INFLAMMATION, AUTOIMMUNITY AND PLASTICITY 467
Errico F, Nisticò R, Di Giorgio A et al. (2014). Free multiple sclerosis: a meta-analysis. Gadolinium MRI Meta-
D-aspartate regulates neuronal dendritic morphology, syn- analysis Group. Lancet 353: 964–969. PMID: 10459905.
aptic plasticity, gray matter volume and brain activity in Kipnis J (2016). Multifaceted interactions between adaptive
mammals. Transl Psychiatry 4: e417. immunity and the central nervous system. Science 353:
Ferrucci R, Vergari M, Cogiamanian F et al. (2014). 766–771.
Transcranial direct current stimulation (tDCS) for fatigue Kirkwood A, Bear MF (1994). Homosynaptic long-term
in multiple sclerosis. NeuroRehabilitation 34 (1): 121–127. depression in the visual cortex. J Neurosci 14:
https://doi.org/10.3233/NRE-131019. 3404–3412. PMID: 8182481.
Fiene M, Rufener KS, Kuehne M et al. (2018). Koch G, Rossi S, Prosperetti C et al. (2008). Improvement of
Electrophysiological and behavioral effects of frontal tran- hand dexterity following motor cortex rTMS in multiple
scranial direct current stimulation on cognitive fatigue in sclerosis patients with cerebellar impairment. Mult Scler
multiple sclerosis. J Neurol 265 (3): 607–617. https://doi. 14 (7): 995–998. https://doi.org/10.1177/13524585080
org/10.1007/s00415-018-8754-6. 88710. PMID: 18573820.
Filippi M, Paty DW, Kappos L et al. (1995). Correlations between Koch G, Mori F, Codecà C et al. (2009). Cannabis-based treat-
changes in disability and T2-weighted brain MRI activity in ment induces polarity-reversing plasticity assessed by theta
multiple sclerosis: a follow-up study. Neurology 45: burst stimulation in humans. Brain Stimul 2 (4): 229–233.
255–260. https://doi.org/10.1212/wnl.45.2.255. PMID: Korzhova J, Bakulin I, Sinitsyn D et al. (2019). High-
7854522. frequency repetitive transcranial magnetic stimulation
Fuhr P, Agostino R, Hallett M (1991). Spinal motor neuron and intermittent theta-burst stimulation for spasticity
excitability during the silent period after cortical stimula- management in secondary progressive multiple sclerosis.
tion. Electroencephalogr Clin Neurophysiol 81: 257–262. Eur J Neurol 26 (4): 680–e44. https://doi.org/10.1111/
PMID: 1714819. ene.13877.
Gentile A, Mori F, Bernardini S et al. (2015). Role of amyloid- Kujirai T, Caramia MD, Rothwell JC et al. (1993).
b CSF levels in cognitive deficit in MS. Clin Chim Corticocortical inhibition in human motor cortex. J Physiol
Acta 449: 23–30. https://doi.org/10.1016/j.cca.2015.01. 471: 501–519. PMID: 8120818.
035. PMID: 25659291. Larochelle C, Alvarez JI, Prat A (2011). How do immune cells
Gruol DL (2015). IL-6 regulation of synaptic function in overcome the blood-brain barrier in multiple sclerosis?
the CNS. Neuropharmacology 96: 42–54. https://doi.org/ FEBS Lett 585 (23): 3770–3780. https://doi.org/10.1016/
10.1016/j.neuropharm.2014.10.023. j.febslet.2011.04.066. PMID: 21550344.
Hanajima R, Furubayashi T, Iwata NK et al. (2003). Further Lassmann H (2011). Mechanisms of neurodegeneration shared
evidence to support different mechanisms underlying intra- between multiple sclerosis and Alzheimer’s disease.
cortical inhibition of the motor cortex. Exp Brain Res J Neural Transm 118: 747–752. https://doi.org/10.1007/
151: 427–434. https://doi.org/10.1007/s00221-003-1455-z. s00702-011-0607-8.
PMID: 1283034. Leocani L, Nuara A, Formenti A et al. (2012). Deep rTMS
Heifets BD, Castillo PE (2009). Endocannabinoid signaling with H-coil associated with rehabilitation enhances
and long-term synaptic plasticity. Annu Rev Physiol 71: improvement of walking abilities in patients with pro-
283–306. gressive multiple sclerosis: randomized, controlled, double
Heyman E, Gamelin FX, Goekint M et al. (2012). Intense exer- blind study. Neurology 78: S49.007-S49.007. Epub ahead
cise increases circulating endocannabinoid and BDNF of print 26 April 2012. https://doi.org/10.1212/WNL.
levels in humans—possible implications for reward and 78.1_MeetingAbstracts.S49.007.
depression. Psychoneuroendocrinology 37: 844–851. Lissin DV, Gomperts SN, Carroll RC et al. (1998). Activity
Huang YZ, Edwards MJ, Rounis E et al. (2005). Theta burst differentially regulates the surface expression of synaptic
stimulation of the human motor cortex. Neuron 45: AMPA and NMDA glutamate receptors. Proc Natl Acad
201–206. PMID: 15664172. Sci USA 95: 7097–7102. https://doi.org/10.1073/pnas.95.
Huang YZ, Chen RS, Rothwell JC et al. (2007). The aftereffect 12.7097.
of human theta burst stimulation is NMDA receptor Madroñal N, Gruart A, Valverde O et al. (2012). Involvement
dependent. Clin Neurophysiol 118 (5): 1028–1032. of cannabinoid CB1 receptor in associative learning and in
Inghilleri M, Berardelli A, Cruccu G et al. (1993). Silent period hippocampal CA3-CA1 synaptic plasticity. Cereb Cortex
evoked by transcranial stimulation of the human cortex and 22 (3): 550–566.
cervicomedullary junction. J Physiol 466: 521–534. PMID: Mandolesi G, Grasselli G, Musella A et al. (2012). GABAergic
8410704. signaling and connectivity on Purkinje cells are impaired in
Iodice R, Dubbioso R, Ruggiero L et al. (2015). Anodal tran- experimental autoimmune encephalomyelitis. Neurobiol
scranial direct current stimulation of motor cortex does not Dis 46 (2): 414–424. https://doi.org/10.1016/j.nbd.2012.
ameliorate spasticity in multiple sclerosis. Restor Neurol 02.005. PMID: 22349452.
Neurosci 33 (4): 487–492. https://doi.org/10.3233/RNN- Mandolesi G, Musella A, Gentile A et al. (2013). Interleukin-
150495. 1b alters glutamate transmission at purkinje cell synapses
Kappos L, Moeri D, Radue EW et al. (1999). Predictive value in a mouse model of multiple sclerosis. J Neurosci 33
of gadolinium-enhanced magnetic resonance imaging for (29): 12105–12121. https://doi.org/10.1523/JNEUROSCI.
relapse rate and changes in disability or impairment in 5369-12.2013. PMID: 23864696.
468 M. STAMPANONI BASSI ET AL.
Mandolesi G, Gentile A, Musella A et al. (2015). Synaptopathy Mori F, Kusayanagi H, Nicoletti CG et al. (2014b). Cortical
connects inflammation and neurodegeneration in multiple plasticity predicts recovery from relapse in multiple sclero-
sclerosis. Nat Rev Neurol 11 (12): 711–724. https://doi.org/ sis. Mult Scler 20 (4): 451–457. https://doi.org/10.1177/
10.1038/nrneurol.2015.222. PMID: 26585978. 1352458513512541. PMID: 24263385.
Mattioli F, Bellomi F, Stampatori C et al. (2015). Mori F, Ljoka C, Nicoletti CG et al. (2014c). CB1
Neuroenhancement through cognitive training and anodal receptor affects cortical plasticity and response to phy-
tDCS in multiple sclerosis. Mult Scler J 22 (2): 222–230. siotherapy in multiple sclerosis. Neurol Neuroimmunol
https://doi.org/10.1177/1352458515587597. Neuroinflamm 1 (4): e48.
Mayo L, Cunha AP, Madi A et al. (2016). IL-10-dependent Mori F, Nisticò R, Nicoletti CG et al. (2016). RANTES corre-
Tr1 cells attenuate astrocyte activation and ameliorate lates with inflammatory activity and synaptic excitability in
chronic central nervous system inflammation. Brain 139: multiple sclerosis. Mult Scler 22 (11): 1405–1412. https://
1939–1957. https://doi.org/10.1093/brain/aww113. PMID: doi.org/10.1177/1352458515621796. PMID: 26733422.
27246324. Mulkey RM, Malenka RC (1992). Mechanisms underlying
Meyer C, Martin-Blondel G, Liblau RS (2017). Endothelial induction of homosynaptic longterm depression in area
cells and lymphatics at the interface between the immune CA1 of the hippocampus. Neuron 9: 967–975. https://doi.
and central nervous systems: implications for multiple scle- org/10.1016/0896-6273(92)90248-c. PMID: 1419003.
rosis. Curr Opin Neurol 30 (3): 222–230. https://doi.org/ Nantes JC, Zhong J, Holmes SA et al. (2016a). Cortical
10.1097/WCO.0000000000000454. PMID: 28323646. damage and disability in multiple sclerosis: relation to
Mori F, Codecà C, Kusayanagi H et al. (2010a). Effects of intracortical inhibition and facilitation. Brain Stimul 9:
anodal transcranial direct current stimulation on chronic 566–573. https://doi.org/10.1016/j.brs.2016.01.003.
neuropathic pain in patients with multiple sclerosis. Nantes JC, Zhong J, Holmes SA et al. (2016b). Intracortical
J Pain 11 (5): 436–442. https://doi.org/10.1016/j.jpain. inhibition abnormality during the remission phase of mul-
2009.08.011. PMID: 20018567. tiple sclerosis is related to upper limb dexterity and lesions.
Mori F, Codecà C, Kusayanagi H et al. (2010b). Effects of Clin Neurophysiol 127: 1503–1511. https://doi.org/10.
intermittent theta burst stimulation on spasticity in patients 1016/j.clinph.2016.11.022. PMID: 26394909.
with multiple sclerosis. Eur J Neurol 17 (2): 295–300. Neva JL, Lakhani B, Brown KE et al. (2016). Multiple
https://doi.org/10.1111/j.1468-1331.2009.02806.x. measures of corticospinal excitability are associated with
Mori F, Rossi S, Sancesario G et al. (2011a). Cognitive and cor- clinical features of multiple sclerosis. Behav Brain Res
tical plasticity deficits correlate with altered amyloid-b CSF 297: 187–195. https://doi.org/10.1016/j.bbr.2015.10.015.
levels in multiple sclerosis. Neuropsychopharmacology 36 Nicoletti CG, Monteleone F, Marfia GA et al. (2019). Oral
(3): 559–568. https://doi.org/10.1038/npp.2010.187. PMID: D-Aspartate enhances synaptic plasticity reserve in progres-
20944553. sive multiple sclerosis. Mult Scler 7: 1352458519828294.
Mori F, Ljoka C, Magni E et al. (2011b). Transcranial mag- https://doi.org/10.1177/1352458519828294. [Epub ahead of
netic stimulation primes the effects of exercise therapy in print] PMID: 30730244.
multiple sclerosis. J Neurol 258 (7): 1281–1287. https:// Nielsen JF, Sinkjaer T, Jakobsen J (1996). Treatment of spas-
doi.org/10.1007/s00415-011-5924-1. ticity with repetitive magnetic stimulation; a double-blind
Mori F, Kusayanagi H, Buttari F et al. (2012). Early treatment placebo-controlled study. Mult Scler 5: 227–232. https://
with high-dose interferon beta-1a reverses cognitive and doi.org/10.1177/135245859600200503. PMID: 9050361.
cortical plasticity deficits in multiple sclerosis. Funct Nisticò R, Mango D, Mandolesi G et al. (2013). Inflammation
Neurol 27 (3): 163–168. PMID: 23402677. subverts hippocampal synaptic plasticity in experimental
Mori F, Kusayanagi H, Monteleone F et al. (2013a). multiple sclerosis. PLoS One 8 (1): e54666. https://doi.
Short interval intracortical facilitation correlates with the org/10.1371/journal.pone.0054666. PMID: 23355887.
degree of disability in multiple sclerosis. Brain Stimul Paulus W, Classen J, Cohen LG et al. (2008). State of the art:
6 (1): 67–71. https://doi.org/10.1016/j.brs.2012.02.001. pharmacologic effects on cortical excitability measures
PMID: 22425067. tested by transcranial magnetic stimulation. Brain Stimul
Mori F, Rossi S, Piccinin S et al. (2013b). Synaptic plasticity 1: 151–163. https://doi.org/10.1016/j.brs.2008.06.002.
and PDGF signaling defects underlie clinical progression PMID: 20633382.
in multiple sclerosis. J Neurosci 33 (49): 19112–19119. Peng F, Yao H, Bai X et al. (2010). Platelet-derived growth
https://doi.org/10.1523/JNEUROSCI.2536-13.2013. factor-mediated induction of the synaptic plasticity gene
PMID: 24305808. Arc/Arg3.1. J Biol Chem 285: 21615–21624. https://doi.
Mori F, Nicoletti CG, Kusayanagi H et al. (2013c). org/10.1074/jbc.M110.107003. PMID: 20452974.
Transcranial direct current stimulation ameliorates tactile Raichlen DA, Foster AD, Gerdeman GL et al. (2012). Wired to
sensory deficit in multiple sclerosis. Brain Stimul 6 (4): run: exercise-induced endocannabinoid signaling in
654–659. https://doi.org/10.1016/j.brs.2012.10.003. humans and cursorial mammals with implications for the
Mori F, Nisticò R, Mandolesi G et al. (2014a). Interleukin-1b ’runner’s high. J Exp Biol 215 (Pt. 8): 1331–1336.
promotes long-term potentiation in patients with multiple Restivo DA, Alfonsi E, Casabona A et al. (2019). A pilot study
sclerosis. Neuromolecular Med 16 (1): 38–51. https://doi. on the efficacy of transcranial direct current stimulation
org/10.1007/s12017-013-8249-7. PMID: 23892937. applied to the pharyngeal motor cortex for dysphagia
MULTIPLE SCLEROSIS: INFLAMMATION, AUTOIMMUNITY AND PLASTICITY 469
associated with brainstem involvement in multiple sclero- Saiote C, Goldschmidt T, Tim€aus C et al. (2014). Impact
sis. Clin Neurophysiol 130 (6): 1017–1024. https://doi.org/ of transcranial direct current stimulation on fatigue in
10.1016/j.clinph.2019.04.003. multiple sclerosis. Restor Neurol Neurosci 32 (3):
Rizzo FR, Musella A, De Vito F et al. (2018). Tumor necrosis 423–436. https://doi.org/10.3233/RNN-130372.
factor and interleukin-1b modulate synaptic plasticity dur- Sarchielli P, Greco L, Stipa A et al. (2002). Brain-
ing neuroinflammation. Neural Plast 2018: 8430123. derived neurotrophic factor in patients with multiple
https://doi.org/10.1155/2018/8430123. PMID: 29861718. sclerosis. J Neuroimmunol 132 (1–2): 180–188. PMID:
R€
osser N, Heuschmann P, Wersching H et al. (2008). 12417449.
Levodopa improves procedural motor learning in Scheidtmann K, Fries W, M€ uller F et al. (2001). Effect of levo-
chronic stroke patients. Arch Phys Med Rehabil 89 (9): dopa in combination with physiotherapy on functional
1633–1641. https://doi.org/10.1016/j.apmr.2008.02.030. motor recovery after stroke: a prospective, randomised,
PMID: 18760148. double-blind study. Lancet 358 (9284): 787–790. PMID:
Rossi S, Furlan R, De Chiara V et al. (2009). Exercise attenu- 11564483.
ates the clinical, synaptic and dendritic abnormalities of Schneider H, Pitossi F, Balschun D et al. (1998).
experimental autoimmune encephalomyelitis. Neurobiol A neuromodulatory role of interleukin-1beta in the hippo-
Dis 36 (1): 51–59. https://doi.org/10.1016/j.nbd.2009. campus. Proc Natl Acad Sci USA 95 (13): 7778–7783.
06.013. PMID: 19591937. https://doi.org/10.1073/pnas.95.13.7778.
Rossi S, Muzio L, De Chiara V et al. (2011a). Impaired striatal Schwenkreis P, Witscher K, Janssen F et al. (1999). Influence
GABA transmission in experimental autoimmune enceph- of the N-methyl-D-aspartate antagonist memantine on
alomyelitis. Brain Behav Immun 25 (5): 947–956. https:// human motor cortex excitability. Neurosci Lett270:
doi.org/10.1016/j.bbi.2010.10.004. PMID: 20940040. 137–140. https://doi.org/10.1016/s0304-3940(99)00492-9.
Rossi S, Mancino R, Bergami A et al. (2011b). Potential PMID: 10462113.
role of IL-13 in neuroprotection and cortical excitability Siebner HR, Dressnandt J, Auer C et al. (1998). Continuous
regulation in multiple sclerosis. Mult Scler 17 (11): intrathecal baclofen infusions induced a marked increase
1301–1312. https://doi.org/10.1177/1352458511410342. of the transcranially evoked silent period in a patient with
PMID: 21677024. generalized dystonia. Muscle Nerve 21: 1209–1212.
Rossi S, Furlan R, De Chiara V et al. (2012). Interleukin-1b https://doi.org/10.1002/(sici)1097-4598(199809)21:9<1209
causes synaptic hyperexcitability in multiple sclerosis. ::aid-mus15>3.0.co;2-m.
Ann Neurol 71 (1): 76–83. https://doi.org/10.1002/ana. Stampanoni Bassi M, Mori F, Buttari F et al. (2017a).
22512. PMID: 22275254. Neurophysiology of synaptic functioning in multiple scle-
Rossi S, Motta C, Studer V et al. (2014a). Tumor necrosis rosis. Clin Neurophysiol 128 (7): 1148–1157. https://doi.
factor is elevated in progressive multiple sclerosis and org/10.1016/j.clinph.2017.04.006. PMID: 28511127.
causes excitotoxic neurodegeneration. Mult Scler 20 Stampanoni Bassi M, Garofalo S, Marfia GA et al. (2017b).
(3): 304–312. https://doi.org/10.1177/1352458513498128. Amyloid-b homeostasis bridges inflammation, synaptic
PMID: 23886826. plasticity deficits and cognitive dysfunction in multiple
Rossi S, Studer V, Motta C et al. (2014b). Cerebrospinal sclerosis. Front Mol Neurosci 10: 390. https://doi.org/
fluid detection of interleukin-1b in phase of remission 10.3389/fnmol.2017.00390. PMID: 29209169.
predicts disease progression in multiple sclerosis. Stampanoni Bassi M, Iezzi E, Marfia GA et al. (2018). Platelet-
J Neuroinflammation 11: 32. https://doi.org/10.1186/ derived growth factor predicts prolonged relapse-
1742-2094-11-32. free period in multiple sclerosis. J Neuroinflammation 15
Rossi S, Motta C, Studer V et al. (2015). Subclinical central (1): 108. https://doi.org/10.1186/s12974-018-1150-4.
inflammation is risk for RIS and CIS conversion to MS. Stampanoni Bassi M, Iezzi E, Mori F et al. (2019).
Mult Scler 21 (11): 1443–1452. https://doi.org/10.1177/ Interleukin-6 disrupts synaptic plasticity and impairs
1352458514564482. tissue damage compensation in multiple sclerosis. Neuro-
Rossini PM, Rossi S (2007). Transcranial magnetic stimulation: rehabil Neural Repair 33: 825–835. https://doi.org/
diagnostic, therapeutic, and research potential. Neurology 10.1177/1545968319868713. Epub 2019 Aug 20. PMID:
68: 484–488. PMID: 17296913. 31431121.
Rossini PM, Berardelli A, Deuschl G et al. (1999). Stampanoni Bassi M, Buttari F, Maffei P et al. (2020).
Applications of magnetic cortical stimulation. The Practice-dependent motor cortex plasticity is reduced
International Federation of Clinical Neurophysiology. in non-disabled multiple sclerosis patients. Clin Neurop-
Electroencepha hysiol 131 (2): 566–573. https://doi.org/10.1016/j.clinph.
logr Clin Neurophysiol 52: 171–185. PMID: 10590986. 2019.10.023.
Rossini PM, Burke D, Chen R et al. (2015). Non-invasive elec- Stefan K, Kunesch E, Cohen LG et al. (2000). Induction of
trical and magnetic stimulation of the brain, spinal cord, plasticity in the human motor cortex by paired associative
roots and peripheral nerves: basic principles and procedures stimulation. Brain 123: 572–584. PMID: 10686179.
for routine clinical and research application. An updated Stefan K, Wycislo M, Gentner R et al. (2006). Temporary
report from an I.F.C.N. Committee. Clin Neurophysiol occlusion of associative motor cortical plasticity by prior
126 (6): 1071–1107. https://doi.org/10.1016/j.clinph.2015. dynamic motor training. Cereb Cortex 16 (3): 376–385.
02.001. PMID: 25797650. https://doi.org/10.1093/cercor/bhi116. PMID: 15930370.
470 M. STAMPANONI BASSI ET AL.
Stellwagen D, Malenka RC (2006). Synaptic scaling mediated Neurosci Biobehav Rev 43: 88–99. https://doi.org/10.1016/
by glial TNF-a. Nature 440: 1054–1059. PMID: 16547515. j.neubiorev.2014.03.02324726576.
Tecchio F, Cancelli A, Cottone C et al. (2015). Brain plasti- Wirsching I, Buttmann M, Odorfer T et al. (2018). Altered
city effects of neuromodulation against multiple sclerosis motor plasticity in an acute relapse of multiple sclerosis.
fatigue. Front Neurol 6: 141. https://doi.org/10.3389/fneur. Eur J Neurosci 47 (3): 251–257. https://doi.org/10.1111/
2015.00141. ejn.13818. PMID: 29285814.
Tokimura H, Ridding MC, Tokimura Y et al. (1996). Short Zeller D, aufm Kampe K, Biller A et al. (2010). Rapid-
latency facilitation between pairs of threshold magnetic stim- onset central motor plasticity in multiple sclerosis.
uli applied to human motor cortex. Electroencephalogr Clin Neurology 74: 728–735. https://doi.org/10.1212/WNL.
Neurophysiol 101: 263–272. PMID: 8761035. 0b013e3181d31dcf. PMID: 20194911.
Turrigiano GG, Leslie KR, Desai NS et al. (1998). Zeller D, Dang SY, Weise D et al. (2012). Excitability decreas-
Activity-dependent scaling of quantal amplitude in neo- ing central motor plasticity is retained in multiple sclerosis
cortical neurons. Nature 391: 892–896. https://doi.org/ patients. BMC Neurol 12: 92. https://doi.org/10.1186/
10.1038/36103. 1471-2377-12-92. PMID: 22974055.
Van den Bos MAJ, Menon P, Howells J et al. (2018). Ziemann U, Tergau F, Wassermann EM et al. (1998).
Physiological processes underlying short interval intra- Demonstration of facilitatory I-wave interaction in
cortical facilitation in the human motor cortex. Front the human motor cortex by paired transcranial magnetic
Neurosci 12: 240. https://doi.org/10.3389/fnins.2018.00 stimulation. J Physiol 511: 181–190. PMID: 9679173.
240. PMID: 29695952. Ziemann U, Ilic TV, Pauli C et al. (2004). Learning modifies
Vana AC, Flint NC, Harwood NE et al. (2007). Platelet- subsequent induction of long-term potentiation-like and
derived growth factor promotes repair of chronically demye- long-term depression-like plasticity in human motor cor-
linated white matter. J Neuropathol Exp Neurol 66: tex. J Neurosci 24 (7): 1666–1672. PMID: 14973238.
975–988. https://doi.org/10.1097/NEN.0b013e3181587d46. Ziemann U, Paulus W, Nitsche MA et al. (2008). Consensus:
PMID: 17984680. motor cortex plasticity protocols. Brain Stimul 1: 164–182.
Villoslada P, Arrondo G, Sepulcre J et al. (2009). Memantine https://doi.org/10.1016/j.brs.2008.06.006. PMID: 20633383.
induces reversible neurologic impairment in patients with Ziemann U, Reis J, Schwenkreis P et al. (2015). TMS
MS. Neurology 72 (19): 1630–1633. and drugs revisited 2014. Clin Neurophysiol 126 (10):
Weiss S, Mori F, Rossi S et al. (2014). Disability in multiple 1847–1868. https://doi.org/10.1016/j.clinph.2014.08.028.
sclerosis: when synaptic long-term potentiation fails. PMID: 25534482.

You might also like