ArticlePDF Available

Abstract and Figures

Epidemiological data suggest the notion that in Multiple Sclerosis (MS) is an acquired autoimmune disease and the cause may be an environmental factor(s), probably infectious, in genetically susceptible individuals. Several cases of viral induced demyelinatimg encephalomyelitis in human beings and in experimental models as well as the presence of IgG oligoclonal bands in the cerebrospinal fluid indicate that the infectious factor may be viral. However, the absence of a specific virus identification in MS central nervous system may hardly support this notion. On the other hand, the partial response of patients with MS to immunosuppressive and immunomodulatory therapy support the evidence of an autoimmune etiology for MS. However, the autoimmune hypothesis shares the same criticism with the infectious one in that no autoantigen(s) specific to and causative for MS has ever been identified. Nevertheless, the absence of identifiable infectious agent, especially viral does not rule out its presence at a certain time--point and the concomitant long term triggering of an autoimmune cascade of events thereafter. Several concepts have emerged in an attempt to explain the autoimmune mechanisms and ongoing neurodegeneration in MS on the basis of the infectious--viral hypothesis.
Content may be subject to copyright.
BioMed Central
Page 1 of 8
(page number not for citation purposes)
Journal of Autoimmune Diseases
Open Access
Review
Virus-mediated autoimmunity in Multiple Sclerosis
Nikolaos Grigoriadis
1
and Georgios M Hadjigeorgiou*
2
Address:
1
B' Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, 1 Stilp
Kyriakidi Street, Aristotle University of Thessaloniki, Thessaloniki, 54636 Thessaloniki, Greece and
2
Department of Neurology, Neurogenetics
Unit, Medical School, University of Thessaly, 22 Papakyriazi Street, 41222 Larissa, Greece
Email: Nikolaos Grigoriadis - grigoria@med.auth.gr; Georgios M Hadjigeorgiou* - gmhadji@med.uth.gr
* Corresponding author
Abstract
Epidemiological data suggest the notion that in Multiple Sclerosis (MS) is an acquired autoimmune
disease and the cause may be an environmental factor(s), probably infectious, in genetically
susceptible individuals. Several cases of viral induced demyelinatimg encephalomyelitis in human
beings and in experimental models as well as the presence of IgG oligoclonal bands in the
cerebrospinal fluid indicate that the infectious factor may be viral. However, the absence of a
specific virus identification in MS central nervous system may hardly support this notion. On the
other hand, the partial response of patients with MS to immunosuppressive and
immunomodulatory therapy support the evidence of an autoimmune etiology for MS. However, the
autoimmune hypothesis shares the same criticism with the infectious one in that no autoantigen(s)
specific to and causative for MS has ever been identified. Nevertheless, the absence of identifiable
infectious agent, especially viral does not rule out its presence at a certain time – point and the
concomitant long term triggering of an autoimmune cascade of events thereafter. Several concepts
have emerged in an attempt to explain the autoimmune mechanisms and ongoing
neurodegeneration in MS on the basis of the infectious – viral hypothesis.
Background
Multiple sclerosis (MS) is widely believed to be an
autoimmune disorder characterized by multifocal lesions
of the CNS myelin and accumulating clinical signs due to
axonal damage [1]. The aetiology of MS has been debated
several times since the disease was first described. Myelin
is damaged due to an immune attack consisted of several
pathways and molecules, leading to impaired nerve func-
tion. Autoantibodies and autoreactive T cells activated
against myelin antigens such as myelin basic protein
(MBP), proteolipid protein (PLP), and myelin oli-
godendrocyte glycoprotein (MOG), have been detected in
MS patients [2]. The majority of researchers consider MS
as a CD4
+
T-helper 1 (Th1)-mediated inflammatory
demyelinating disease [3,4]. Several data indicate this
consideration, such as the cellular composition of brain
and cerebrospinal fluid (CSF)-infiltrating cells and data
from studies in a widely used animal model for MS, the
Experimental Alergic Encephalomyelitis (EAE) [5]. In the
EAE model, myelin components emulsified in complete
Freund's adjuvant (CFA) and injected in susceptible ani-
mals lead to a CD4
+
-mediated autoimmune disease that
shares clinical, immunological and pathological similari-
ties with MS. CFA creates an artificial inflammatory milieu
that does not reflect the natural environment in which self
or mimic peptides would be normally encountered. EAE
may also be induced passively by transferring anti-myelin
activated T-cells to naive animals (transfer EAE), a finding
Published: 19 February 2006
Journal of Autoimmune Diseases 2006, 3:1 doi:10.1186/1740-2557-3-1
Received: 31 October 2005
Accepted: 19 February 2006
This article is available from: http://www.jautoimdis.com/content/3/1/1
© 2006 Grigoriadis and Hadjgeorgiou; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0
),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Journal of Autoimmune Diseases 2006, 3:1 http://www.jautoimdis.com/content/3/1/1
Page 2 of 8
(page number not for citation purposes)
that clearly indicates the autoimmune component of the
disease.
Studies on EAE indicated that cytokines, chemokines and
adhesion molecules induce the recruitment of leukocytes
from periphery to CNS throughout a disrupted blood
brain barrier (BBB) and a cascade of inflammatory events
is established within the CNS. Eventually, axonal degener-
ation and loss is the hallmark in the disease process lead-
ing to a long-term disability [6]. Although EAE may not be
the ideal animal model for the disease, the model itself in
combination with several other experimental and clinical
data indicate that MS is an autoimmune disease [7]. How-
ever, the major criticism of the autoimmune hypothesis is
that autoantigen(s) specific to and causative for MS has
never been identified. In addition, although inflamma-
tion is considered to be a primary feature of demyelianat-
ing plaques thus favouring the autoimmune component
in this process, recent reports indicate that demyelination
may precede inflammation [8] On the other hand, there
are reports proposing that MS is not an autoimmune dis-
ease but a genetically determined disorder characterized
by metabolically dependent neurodegeneration [9]. The
latter may imply that immune reaction in MS may be a
secondary one to the ongoing degeneration of axons and
neurons. Nevertheless, while the autoimmune model may
not explain every aspect of MS, it is difficult to ignore the
considerable evidence that immunity plays a major role in
MS. Another intriguing idea regarding the aetiology of MS
may be that the immune response in MS could result from
a chronic viral infection rather than autoimmunity in the
usual sense [10-12].
The possible involvement of viruses in the aetiology of MS
is a rather controversial issue. Based on immigration data,
it has been suggested that environmental factor(s) may
trigger MS before the age of adolescence, while the disease
is clinically silent until years later. It is also apparent that
there is a genetic susceptibility related at least to the HLA
system [13-15] Among monozygotic twins there is a 70%
disconcordance of MS suggesting that an exogenous factor
causes the disease[14]. Evidently, these indications lead to
the hypothesis that MS is a disease triggered by an envi-
ronmental factor in genetically susceptible individuals
during childhood [12,16-20]. Moreover, it has been spec-
ulated that the environmental factor in MS could be a
virus [21-23] In addition, abnormal immune response to
a variety of viruses in MS patients as well as analogy with
animal models and other human diseases in which
viruses can cause diseases with long incubation periods,
relapse, and demyelination, further support the concept
that viruses may be implicated in the MS aetiology.
Although to date no virus has been recognized as a causa-
tive factor of MS, the possibility that both autoimmunity
and neurodegeneration in MS may coexist and commonly
be explained following a viral infection is reviewed here.
Experimental and clinical evidence for a virus-
related aetiology of MS
Animal models
Various viruses have been found to induce demyelination
in laboratory animals following various infection proto-
cols. The most studied experimental demyelination is
infection of mice with Theiler's murine encephalomyelitis
virus (TMEV) [24]. TMEV (a higly cytolytic picovarious)
infection in mice serves as a model to explain infectious
and parainfectious mechanisms underlying CNS demyeli-
nation. TMEV infection of oligodendrocytes is productive,
resulting in cell lysis and liberation of more virions. By
contrast, TMEV infection in macrophages is restricted, and
results in apoptosis of macrophages. TMEV antigen is
abundant in the cytoplasm of apoptotic macrophages.
Small amounts of TMEV are liberated from persistently
infected macrophages leading to infection of more macro-
phages as well as oligodendrocytes. A persistent CNS
infection is established as virus spreads from macrophage
to macrophage. Virus released from macrophages can
infect and damage more oligodendrocytes, thus adding to
immunopathological destruction of myelin [25].
Another animal model of virus-induced demyelination is
the one established in BALB/c mice following infection
with JHM virus (coronavirus). This strain infects predom-
inantly oligodendrocytes, and the induced demyelination
is not preceded by inflammation or any immune – related
mechanism [26,27]. The infected oligodendrocytes con-
tain intracisternal virions [28] and this model may be con-
sidered as a case of demyelination resulting simply from a
direct virus induced cytopathology of oligodendrocytes.
Autoimmune responses to myelin antigens are observed
following infection with TMEV. Despite the fact that this
autoimmunity to myelin components may not play a
major role in the initiation of demyelination, it may prob-
ably contribute to lesion progression in chronically dis-
eased animals [29].
CNS demyelination in host animals may also occur fol-
lowing infection of other viruses such as in mice with JHM
or MHV-4 (coronaviruses), dogs with canine distemper
virus, and sheep and goats with Visna virus and caprine
arthritis-encephalitis virus. Each of these viruses is capable
of establishing a persistent infection in their host, such
that there is continuous virus replication over a long
period without killing the host. Another virus-induced
demyelination animal model is Semliki Forest virus (SFV)
infection of mice [30,31]. The initial immune-mediated
demyelination may be due to targeting of SFV-infected
Journal of Autoimmune Diseases 2006, 3:1 http://www.jautoimdis.com/content/3/1/1
Page 3 of 8
(page number not for citation purposes)
oligodendrocytes by cytotoxic T-cells. SFV induces
repaired acute demyelination with no relapses.
Clinical studies supporting a role of virus in MS
pathogenesis
The application of modern sophisticated laboratory tech-
niques have led to a growing number of viruses associated
with MS albietno such a pathogen has been accepted as
the canditate causal agent in MS. In addition, interferon
beta, a currently applied treatment in MS patients [32],
was originally proposed as being capable of increasing the
resistance of host tissues against viral infections. However,
no scientific data to date support viral inhibition as one of
the underlying mechanisms of action interferon beta in
MS.
Several clinical studies have suggested that MS in general
as well as episodes of disease exacerbation are associated
with concomitant viral or microbial infections[12,33].
Upper respiratory tract infections can trigger acute
relapses of MS, resulting in an increase in the risk of clin-
ical exacerbations during the weeks that follow the onset
of virus infection[34] Most importantly, when recurrent,
these viral infections are associated with neurological pro-
gression [35,36].
Many of the studies related to the virus infection in MS are
serological and involve the demonstration of increased
antibody titers against a particular virus[12]. In addition,
in a number of studies, isolation of virus from MS mate-
rial has also been reported [37]. Antibody levels to various
viruses are elevated in MS patients, but it has not been
clarified whether this elevation is related to the aetiology
of MS or is a concomitant phenomenon. Many viruses
have been detected in CNS autopsy tissue from MS
patients [38,39]. Throughout the last decades (see table 1
for selected references), among the viral agents related to
demyelination were considered the measles virus [40],
parainfluenza virus [41], canine distemper [42], Epstein-
Barr virus [43], human herpes virus-6 (HHV-6) [44] and
retroviruses [45]. It is generally accepted that despite the
sensitive methods used, still there is no convincing evi-
dence that viruses are related to MS aetiology, mainly due
to controversies among the related studies. HHV-6 in par-
ticular, is a typical example of a virus recently tested in
details with various assays in order to investigate any rela-
tion to MS aetiology[46]. HHV-6 was subjected to detec-
tion both in MS patients and healthy controls or patients
suffering from other neurological disorders. Patients'
material examined was brain tissue [47-49], CSF [50-52],
serum/plasma [53-55], and peripheral blood mononu-
clear cells [53-57] using PCR [47,48,53-57], immunohis-
tochemistry [58-60], or in vitro virus culture assays [58].
Despite such a detailed investigation there is a lack of a
conclusive remark on whether HHV-6 is associated to
MS[46]. The controversy that is evident throughout the
various studies may be attributed either to differences in
the sensitivity of the applied methods or the patient selec-
tion, different methodology applied, etc. Hence, no mat-
ter whether there the viral aetiology of MS is not yet
Table 1: Selected studies exploring the relation of multiple sclerosis with human herpes virus-6
Material used Method followed Relation to MS Year of publication
Brain tissue Viral DNA-PCR, immunohistochemistry Positive 1995[44]
Viral DNA-PCR Negative 1996[49]
Viral DNA-PCR, immunohistochemistry Positive 1999[47]
Immunohistochemistry Positive 2000[58]
Viral DNA-PCR Positive 2000[59]
Viral DNA-PCR, immunohistochemistry Positive 2003[60]
Viral DNA-PCR Uncertain 2003[48]
CSF Viral DNA-PCR Negative 1999[50]
Viral DNA-PCR, antibodies titer Negative 2000[51]
Viral DNA-PCR, antibodies titer Positive 2002[52]
Serum Viral DNA-PCR Negative 1999[50]
Viral DNA-PCR, antibodies titer Positive 2000[52]
Viral DNA-PCR Positive 2000[53]
Viral DNA-PCR Positive 2001[54]
Viral DNA-PCR Negative 2002[55]
Viral DNA-PCR, antibodies titer Positive 2003[56]
Peripheral blood mononuclear cells Viral DNA-PCR Negative 2000[51]
Viral DNA-PCR Positive 2000[53]
Viral DNA-PCR Negative 2000[57]
Viral DNA-PCR Positive 2001[54]
Viral DNA-PCR Positive 2002[55]
Viral DNA-PCR Positive 2003[56]
Journal of Autoimmune Diseases 2006, 3:1 http://www.jautoimdis.com/content/3/1/1
Page 4 of 8
(page number not for citation purposes)
clarified, it should be emphasized that the absence of evi-
dence does not necessary imply the evidence of absence
[61].
Mechanisms of virus-induced CNS
autoimmunity
Following a virus infection, there may be two potential
options: the virus might reactivate after a long term
latency, up to years and lyse oligodendrocytes, or could
initiate a rather acute or subacute demyelinating immun-
opathology. Examples of the first option might be pro-
gressive multifocal leucoencephalopathy (PML) through
the infection of JC virus (a human papova virus) whereas
the later is the case of TMEV encephalomyelitis model, as
well as infections with corona viruses, and lenti viruses
[62].
Among the major indications for the association of demy-
elination with viral infection and a destructive host
immune response to autoantigens is the case of post-
infectious encephalomyelitis, a complication mainly
noticed following smallpox vaccination or measles virus
and to a lesser extend, varicella and rubella infection. The
underlying pathology share similarities with the one
induced in EAE [63]. Another example of virus-induced
demyelination with concomitant autoimmunity is the
one following infection with murine coronavirus in rats.
At the acute stage of this animal model demyelination is
restricted and related to the infection of glial cells from the
virus. However, at later stages, at a time when animals
recover from the viral infection, perivascular infiltrates
and extented demyelination are present. A transfer EAE
was performed by injecting in vitro activated anti-myelin
lymphocytes harvested from infected rats at a time that
the animals recovered from the initial infection to naive
recipients. The resulted EAE was mild with no evidence of
demyelination [64]. This finding may indicate that the
pre-exposure of the animal to virus may be necessary for
the induction of autoimmune demyelination. Similarly,
the canine distemper virus (paramyxovirus) – induced
demyelination has been reported to be associated with
perivascular infiltrations at the late phases following ini-
tial infection [65].
The example of TMEV – induced encephalomyelitis is
probably the best currently used model of virus-induced
immune-mediated demyelination in susceptible mice.
TMEV is divided into two subgroups: high-neurovirulent
strains, including GDVII and FA, which cause fatal
encephalitis, and low-neurovirulent strains, such as DA
and BeAn, which cause persistent infection and demyeli-
nation in mice [66,67]. The demyelinating phase is pre-
ceded by an inflammatory one consisted of macrophages
and MHC II – restricted T-cells. However, during the
demyelinating phase, cytotoxic and suppressor MHC I –
restricted T-cells gradually replace the initial inflamma-
tory subpopulation. In particular, the prevailing opinion
is that B- and T-lymphocytes play paradoxical functions in
the TMEV-induced CNS disease since they may participate
in the virus clearance in CNS cells during the acute phase
of the disease and aggravate the demyelinating process in
the chronic phase of the disease, thereafter. Therefore, in
this model, inflammatory cells are constant components
of the underlying demyelinating process [68]. The large
number of B- and T-lymphocytes in demyelinating areas
suggest that recruitment of these cells into the CNS is an
important step in the process of myelin destruction [69-
71]. In addition, the response to immunomodulatory or
immunosupressent agents [24,72,73] as well as the
expression of Ia antigens in glial cells [74], indicate the
immune-mediated mechanism of demyelination in this
animal model. It was hypothesized that the specificity of
primary white matter destruction in the TMEV model
depends on immune-sensitised cells, which interact with
viral antigen plus MHC antigens on the surfaces of oli-
godendrocytes or myelin sheaths[75]. The whole underly-
ing pathology shares similarities with the one in MS,
except that in the later no viruses are detected in oli-
godendrocytes as in TMEV encephalomyelitis [76].
It was suggested that virus infection may initiate or exac-
erbate organ-specific autoimmune diseases [77]. There is
growing evidence about possible mechanisms by which
virus infection can trigger autoimmunity. Among them
are: (a) molecular mimicry, (b) bystander activation, and
(c) epitope spreading (figure 1).
(a) Molecular mimicry involves the de novo activation of
autoreactive T cells due to the cross-reactivity between self
epitopes and viral epitopes during a virus infection [78].
Hence, an immune response of the host to a viral epitope
will recognize as nonself the crossreacting host epitope
even when the virus is no longer present. The concept of
molecular mimicry is among the most popular theories
about how virus may induce autoimmunity. Accordingly,
Proposed scheme for virus-mediated autoimmunity in multi-ple sclerosisFigure 1
Proposed scheme for virus-mediated autoimmunity in multi-
ple sclerosis.
Journal of Autoimmune Diseases 2006, 3:1 http://www.jautoimdis.com/content/3/1/1
Page 5 of 8
(page number not for citation purposes)
a molecular mimicry has been reported between anti-
TMEV antibody responses in TMEV-infected mice and the
myelin component galactocerebroside [67]. In TMEV-
induced demyelination, CD4+ T cell responses to myelin
epitopes arising via epitope spreading after initial CNS
damage approximately 45–60 days post-infection [29].
Interestingly, a molecular mimicry model for initiation of
autoimmune demyelination was developed following
virus infection with nonpathogenic TMEV which was con-
taining a self myelin epitope such as native or mimic
sequences of the immunodominant PLP139-151 epitope.
Infection of SJL mice infection with such a virus express-
ing a self epitope mimic, directly induced autoreactive T
cells with pathologic potential in the absence of CFA [79].
The later may be considered as a big advantage of this
molecular mimicry model since CFA is a chemical com-
pound imposing artificial inflammatory environment.
Alternatively, what is necessary i.e. the CFA, in EAE as a
model for autoimmunity in MS, is "provided" in the virus-
induced model by the virus per se. The antigen presenting
cells (macrophages, dendritic cells, microglia) as well as
the capability of the mimic peptide being processed from
the native pathogen protein, are two key factors that play
important role in the molecular mimicry mechanism dur-
ing the induction of autoimmunity. In addition, the
nature of the innate immune response to the pathogen
which determines the immunopathologic potential of the
induced cross-reactive T cells, the site(s) of the primary
infection in the host and the ability of the pathogen to
persist, and finally the potential requirement for multiple
infections with the same or different pathogens, are all
considered as contributing factors determining the mech-
anism of molecular mimicry [80].
MBP is among the most important targets in the immun-
opathogenesis of MS. In addition, the MBP(85–99) pep-
tide is a T-cell target for patients with the HLA-DR2
haplotype while the MBP(88–102) peptide may be a tar-
get for patients with other HLA-DR haplotypes. The HLA
antigen and the T-cell receptor (TCR) are key factors in the
constitution of the trimolecular complex (antigen pre-
senting cell – myelin antigen – T-cell) during lymphocyte
activation. Peptide-binding studies determined which
MBP peptide residues were important for binding to DR2
and which ones to TCR. These criteria have been applied
to generate a minimal molecular mimicry peptide by
searching a sequence database for viral and bacterial mim-
icry peptides of MBP(85–99). Finally, 8 yielded peptides
performed biological activity and stimulated MBP(85–
99)-specific T-cell clones. These peptides did not show
any significant linear homology to MBP(85–99), and they
were derived from human pathogens such as EBV, HSV,
CMV, influenza virus, and adenovirus [81].
(b) Bystander activation is the nonspecific activation of
autoreactive T cells resulting from the direct inflammatory
and/or necrotic effects of virus infection on tissue in the
target organ [82]. This mechanism requires destruction of
specific tissue such as CNS, release of sequestered antigen
such as those of myelin and increased local immune
inflammation. Lymphocytes would be recruited to the
injured CNS and those reactive to the released myelin
antigen would in turn be restimulated in the inflamma-
tory response. Consequently, autorreactive lymphocytes
would gain access to the target tissue without being
directly involved in the initial vira! insult or reactive to
viral antigens. Successive targeted viral infections over a
lifetime would fulfill the requirement for generation, acti-
vation and recruitment of autoimmune lymphocytes. The
role of virus in this mechanism is not only to select the tis-
sue, but also to induce a strong inflammatory response
[83].
(c) Epitope spreading is characterized by a widening of the
immune response from an initiating antigenic epitope to
different epitopes on the same molecule (intramolecular
spreading) or to a epitopes on a different antigenic mole-
cule (intermolecular spreading). The addition of func-
tional immunogenic myelin epitopes to the original viral
epitopes in TMEV infection, represents a classical example
of intermolecular epitope spreading [29,84]. In particular,
cells responding to the major PLP epitope139-152, iso-
lated from lymph nodes of TMEV infected mice, have the
ability to demyelinate organotypic cultures of spinal cord.
No similar results were obtained when the cells were stim-
ulated with MBP. These results suggest that in animals
infected with TMEV, the spreading of the immune
response from TMEV to PLP has functional significance,
and is specific[85] T cells specific for a secondary, non-
cross-reactive epitope, PLP178-191, have been reported to
mediate the primary clinical relapse [86]. This phenome-
non has been described in a number of autoimmune dis-
eases, TMEV included [87-90]. More importantly, naive T-
cells enter the inflammed CNS and are activated by local
antigen presenting cells to initiate epitope spreading [91].
Axonal degeneration in MS: is there any value
for viruses?
Injured axons are common in the lesions of multiple scle-
rosis, and axonal transection may be the pathologic corre-
late of the irreversible neurologic impairment in this
disease [92]. Axonal degeneration has been identified as
the major determinant of irreversible neurological disabil-
ity in patients with MS. Evidence for the axonal injury –
related hypothesis is provided by animal models with pri-
mary myelin or axonal pathology, and from pathological
or magnetic resonance studies on MS patients [93]. Dis-
ruption of axons is observed both in EAE and TMEV mod-
els [94].
Journal of Autoimmune Diseases 2006, 3:1 http://www.jautoimdis.com/content/3/1/1
Page 6 of 8
(page number not for citation purposes)
A correlation between inflammation and axonal loss with
neurological disability has been reported in chronic-
relapsing EAE. At the acute stage, CNS inflammation, but
not axon loss, correlated with the degree of neurological
disability. In contrast, fixed neurological impairment in
chronic EAE correlated with axon loss. As proposed for
MS, these observations imply a causal relationship
between inflammation, axon loss, and irreversible neuro-
logical disability [95,96]. It has also been demonstrated in
TMEV model that demyelination in the spinal cord is fol-
lowed by a loss of medium to large myelinated fibres. By
measuring spinal cord areas, motor-evoked potentials,
and motor coordination and balance, axonal loss follow-
ing demyelination was determined to be associated with
electrophysiological abnormalities and correlated
strongly with reduced motor coordination and spinal
cord atrophy. These findings demonstrate that axonal loss
can follow primary, immune-mediated demyelination in
the CNS and that the severity of axonal loss correlates
almost perfectly with the degree of spinal cord atrophy
and neurological deficits [97,98].
Axonal injury begins early at disease onset and correlates
with the degree of inflammation within lesions, indicat-
ing that inflammatory demyelination influences axon
pathology during relapsing-remitting MS (RR-MS) [93].
However, axonal injury exists even in the normal appear-
ing white matter [99] where inflammation may be mini-
mal or absent. In addition, the fact that currently applied
immunomodulators and immunosuppresants may
hardly reverse or even halt the long term disability and the
underlying neurodegeneration, may additionally indicate
that there is no absolute relation between the level of
inflammation and the extend of axonal degeneration and
loss. Moreover, evidence for widespread axonal damage at
the earliest clinical stage of MS lessens the validity of the
concept that the axonal pathology of MS is the end-stage
result of repeated inflammatory events [100].
Interesting information about a possible mechanism
under which axonal injury may exist without concomitant
demyelination, may emerge from TMEV animal model.
The extent and location of axonal injury following infec-
tions with both DA and GDVII, TMEV strains, was investi-
gated [101]. In DA virus infection, axonal injury was
detected as early as 1 week after infection. The number of
damaged axons increased throughout time. During the
subclinical phase, 2 and 3 weeks after infection, axonal
injury was associated with parenchymal infiltration of
microglia and T cells, and viral antigen and damaged
axons were present within intact myelin sheaths. How-
ever, vigorous inflammatory demyelinating lesions were
not seen until the chronic phase (4 weeks after infection).
In GDVII virus infection, extensive axonal injury was
noted 1 week after infection without association with
inflammation, virus, or demyelination. The distribution
of injured or damaged axons in both GDVII virus infec-
tion and the early phase of DA virus infection corre-
sponded to regions where subsequent demyelination
occurred during the chronic phase of DA virus infection.
These findings indicate that axonal injury may not follow
but rather herald demyelination in some virus models.
Somebody may therefore hypothesize that axonal injury
noticed during the earliest stages of MS or in the normal
appearing white matter, could be attributed to the activa-
tion of an as yet unidentified virus. The same or similar
virus may further contribute through induced autoimmu-
nity to axonal injury during later stages of the finally
established inflammatory demyelinating process.
There are two main animal models currently used in MS
research: EAE and TMEV. Both models contributed to a
greater understanding of MS and the development of clin-
ical therapies [6]. Although from a first point of view they
may represent fanatic supporters of either the autoim-
mune or the viral hypothesis on MS aetiology, it becomes
clear later on that the two models complement each other.
Both systems are powerful tools for an in-depth study of
the neuroinflammatory mechanisms potentially involved
in MS pathophysiology. Analysing therapeutic successes
and failures with both models may also help the develop-
ment of more directed, positive treatments for MS that
have fewer negative effects [102].
References
1. Keegan BM, Noseworthy JH: Multiple sclerosis. Annu Rev Med
2002, 53:285-302.
2. Garren H, Steinman L, Lock C: The specificity of the antibody
response in multiple sclerosis. Ann Neurol 1998, 43:4-6.
3. Martin R, McFarland HF, McFarlin DE: Immunological aspects of
demyelinating diseases. Annu Rev Immunol 1992, 10:153-187.
4. Hafler DA: Multiple sclerosis. J Clin Invest 2004, 113:788-794.
5. Zamvil SS, Steinman L: The T lymphocyte in experimental aller-
gic encephalomyelitis. Annu Rev Immunol 1990, 8:579-621.
6. Grigoriadis N, Tselios T, Deraos S, Orologas A, Deraos G, Matsoukas
J, Mavromatis I, Milonas I: Animal models of central nervous sys-
tem immune-mediated diseases: therapeutic interventions
with bioactive peptides and mimetics. Curr Med Chem 2005,
12:1513-1519.
7. Weiner HL: Multiple sclerosis is an inflammatory T-cell-medi-
ated autoimmune disease. Arch Neurol 2004, 61:1613-1615.
8. Barnett MH, Prineas JW: Relapsing and remitting multiple scle-
rosis: pathology of the newly forming lesion. Ann Neurol 2004,
55:458-468.
9. Chaudhuri A, Behan PO: Multiple sclerosis is not an autoim-
mune disease. Arch Neurol 2004, 61:1610-1612.
10. Roach ES: Is multiple sclerosis an autoimmune disorder? Arch
Neurol 2004, 61:1615-1616.
11. Gilden DH: Multiple sclerosis exacerbations and infection.
Lancet Neurol 2002, 1:145.
12. Gilden DH: Infectious causes of multiple sclerosis. Lancet Neurol
2005, 4:195-202.
13. Lock C, Hermans G, Pedotti R, Brendolan A, Schadt E, Garren H,
Langer-Gould A, Strober S, Cannella B, Allard J, Klonowski P, Austin
A, Lad N, Kaminski N, Galli SJ, Oksenberg JR, Raine CS, Heller R,
Steinman L: Gene-microarray analysis of multiple sclerosis
lesions yields new targets validated in autoimmune enceph-
alomyelitis. Nat Med 2002, 8:500-508.
14. Dyment DA, Ebers GC, Sadovnick AD: Genetics of multiple scle-
rosis. Lancet Neurol 2004, 3:104-110.
Journal of Autoimmune Diseases 2006, 3:1 http://www.jautoimdis.com/content/3/1/1
Page 7 of 8
(page number not for citation purposes)
15. Ibrahim SM, Gold R: Genomics, proteomics, metabolomics:
what is in a word for multiple sclerosis? Curr Opin Neurol 2005,
18:231-235.
16. Gaudet JP, Hashimoto L, Sadovnick AD, Ebers GC: Is sporadic MS
caused by an infection of adolescence and early adulthood? A
case-control study of birth order position. Acta Neurol Scand
1995, 91:19-21.
17. James WH: Further evidence in support of the hypothesis that
one cause of multiple sclerosis is childhood infection. Neu-
roepidemiology 1988, 7:130-133.
18. Kurland LT: The evolution of multiple sclerosis epidemiology.
Ann Neurol 1994, 36 Suppl:S2-5.
19. Kurtzke JF: Epidemiologic evidence for multiple sclerosis as an
infection. Clin Microbiol Rev 1993, 6:382-427.
20. Sotgiu S, Pugliatti M, Fois ML, Arru G, Sanna A, Sotgiu MA, Rosati G:
Genes, environment, and susceptibility to multiple sclerosis.
Neurobiol Dis 2004, 17:131-143.
21. Poser CM: Pathogenesis of multiple sclerosis. A critical reap-
praisal. Acta Neuropathol (Berl) 1986, 71:1-10.
22. Allen I, Brankin B: Pathogenesis of multiple sclerosis--the
immune diathesis and the role of viruses. J Neuropathol Exp
Neurol 1993, 52:95-105.
23. Fazakerley JK, Buchmeier MJ: Pathogenesis of virus-induced
demyelination. Adv Virus Res 1993, 42:249-324.
24. Lipton HL, Dal Canto MC: Theiler's virus-induced demyelina-
tion: prevention by immunosuppression. Science 1976,
192:62-64.
25. Schlitt BP, Felrice M, Jelachich ML, Lipton HL: Apoptotic cells,
including macrophages, are prominent in Theiler's virus-
induced inflammatory, demyelinating lesions. J Virol 2003,
77:4383-4388.
26. Weiner LP: Pathogenesis of demyelination induced by a
mouse hepatitis. Arch Neurol 1973, 28:298-303.
27. Lampert PW, Sims JK, Kniazeff AJ: Mechanism of demyelination
in JHM virus encephalomyelitis. Electron microscopic stud-
ies. Acta Neuropathol (Berl) 1973, 24:76-85.
28. Powell HC, Lampert PW: Oligodendrocytes and their myelin-
plasma membrane connections in JHM mouse hepatitis virus
encephalomyelitis. Lab Invest 1975, 33:440-445.
29. Miller SD, Vanderlugt CL, Begolka WS, Pao W, Yauch RL, Neville KL,
Katz-Levy Y, Carrizosa A, Kim BS: Persistent infection with
Theiler's virus leads to CNS autoimmunity via epitope
spreading. Nat Med 1997, 3:1133-1136.
30. Atkins GJ, Sheahan BJ, Dimmock NJ: Semliki Forest virus infec-
tion of mice: a model for genetic and molecular analysis of
viral pathogenicity. J Gen Virol 1985, 66 ( Pt 3):395-408.
31. Atkins GJ, Sheahan BJ, Liljestrom P: The molecular pathogenesis
of Semliki Forest virus: a model virus made useful? J Gen Virol
1999, 80 ( Pt 9):2287-2297.
32. Grigoriadis N: Interferon beta treatment in relapsing-remit-
ting multiple sclerosis. A review. Clin Neurol Neurosurg 2002,
104:251-258.
33. Buljevac D, Flach HZ, Hop WC, Hijdra D, Laman JD, Savelkoul HF,
van Der Meche FG, van Doorn PA, Hintzen RQ: Prospective study
on the relationship between infections and multiple sclerosis
exacerbations. Brain 2002, 125:952-960.
34. Kriesel JD, Sibley WA: The case for rhinoviruses in the patho-
genesis of multiple sclerosis. Mult Scler 2005, 11:1-4.
35. Panitch HS: Influence of infection on exacerbations of multiple
sclerosis. Ann Neurol 1994, 36 Suppl:S25-8.
36. Edwards S, Zvartau M, Clarke H, Irving W, Blumhardt LD: Clinical
relapses and disease activity on magnetic resonance imaging
associated with viral upper respiratory tract infections in
multiple sclerosis. J Neurol Neurosurg Psychiatry 1998, 64:736-741.
37. Clark DA: Human herpesvirus 6 and multiple sclerosis. Herpes
1999, 6(3):73-77.
38. Haahr S, Sommerlund M, Christensen T, Jensen AW, Hansen HJ,
Moller-Larsen A: A putative new retrovirus associated with
multiple sclerosis and the possible involvement of Epstein-
Barr virus in this disease. Ann N Y Acad Sci 1994, 724:148-156.
39. Christensen T, Dissing Sorensen P, Riemann H, Hansen HJ, Moller-
Larsen A: Expression of sequence variants of endogenous ret-
rovirus RGH in particle form in multiple sclerosis. Lancet
1998, 352:1033.
40. Field EJ, Cowshall S, Narang HK, Bell TM: Viruses in multiple scle-
rosis? Lancet 1972, 2:280-281.
41. ter Meulen V, Koprowski H, Iwasaki Y, Kackell YM, Muller D: Fusion
of cultured multiple-sclerosis brain cells with indicator cells:
presence of nucleocapsids and virions and isolation of parain-
fluenza-type virus. Lancet 1972, 2:1-5.
42. Cook SD, Dowling PC: A possible association between house
pets and multiple sclerosis. Lancet 1977, 1:980-982.
43. Ascherio A, Munger KL, Lennette ET, Spiegelman D, Hernan MA,
Olek MJ, Hankinson SE, Hunter DJ: Epstein-Barr virus antibodies
and risk of multiple sclerosis: a prospective study. Jama 2001,
286:3083-3088.
44. Challoner PB, Smith KT, Parker JD, MacLeod DL, Coulter SN, Rose
TM, Schultz ER, Bennett JL, Garber RL, Chang M, et al.: Plaque-asso-
ciated expression of human herpesvirus 6 in multiple sclero-
sis. Proc Natl Acad Sci U S A 1995, 92:7440-7444.
45. Perron H, Garson JA, Bedin F, Beseme F, Paranhos-Baccala G, Komu-
rian-Pradel F, Mallet F, Tuke PW, Voisset C, Blond JL, Lalande B, Sei-
gneurin JM, Mandrand B: Molecular identification of a novel
retrovirus repeatedly isolated from patients with multiple
sclerosis. The Collaborative Research Group on Multiple
Sclerosis. Proc Natl Acad Sci U S A 1997, 94:7583-7588.
46. Clark D: Human herpesvirus type 6 and multiple sclerosis.
Herpes 2004, 11 Suppl 2:112A-119A.
47. Friedman JE, Lyons MJ, Cu G, Ablashl DV, Whitman JE, Edgar M, Kos-
kiniemi M, Vaheri A, Zabriskie JB: The association of the human
herpesvirus-6 and MS. Mult Scler 1999, 5:355-362.
48. Cermelli C, Berti R, Soldan SS, Mayne M, D'Ambrosia J M, Ludwin SK,
Jacobson S: High frequency of human herpesvirus 6 DNA in
multiple sclerosis plaques isolated by laser microdissection.
J Infect Dis 2003, 187:1377-1387.
49. Sanders VJ, Felisan S, Waddell A, Tourtellotte WW: Detection of
herpesviridae in postmortem multiple sclerosis brain tissue
and controls by polymerase chain reaction. J Neurovirol 1996,
2:249-258.
50. Mirandola P, Stefan A, Brambilla E, Campadelli-Fiume G, Grimaldi LM:
Absence of human herpesvirus 6 and 7 from spinal fluid and
serum of multiple sclerosis patients. Neurology 1999,
53:1367-1368.
51. Taus C, Pucci E, Cartechini E, Fie A, Giuliani G, Clementi M, Menzo
S: Absence of HHV-6 and HHV-7 in cerebrospinal fluid in
relapsing-remitting multiple sclerosis. Acta Neurol Scand 2000,
101:224-228.
52. Tejada-Simon MV, Zang YC, Hong J, Rivera VM, Killian JM, Zhang JZ:
Detection of viral DNA and immune responses to the human
herpesvirus 6 101-kilodalton virion protein in patients with
multiple sclerosis and in controls. J Virol 2002, 76:6147-6154.
53. Akhyani N, Berti R, Brennan MB, Soldan SS, Eaton JM, McFarland HF,
Jacobson S: Tissue distribution and variant characterization of
human herpesvirus (HHV)-6: increased prevalence of HHV-
6A in patients with multiple sclerosis. J Infect Dis 2000,
182:1321-1325.
54. Tomsone V, Logina I, Millers A, Chapenko S, Kozireva S, Murovska M:
Association of human herpesvirus 6 and human herpesvirus
7 with demyelinating diseases of the nervous system. J Neu-
rovirol 2001, 7:564-569.
55. Alvarez-Lafuente R, Martin-Estefania C, de Las Heras V, Castrillo C,
Picazo JJ, Varela de Seijas E, Gonzalez RA: Active human herpesvi-
rus 6 infection in patients with multiple sclerosis. Arch Neurol
2002, 59:929-933.
56. Chapenko S, Millers A, Nora Z, Logina I, Kukaine R, Murovska M:
Correlation between HHV-6 reactivation and multiple scle-
rosis disease activity. J Med Virol 2003, 69:111-117.
57. Ferrante P, Mancuso R, Pagani E, Guerini FR, Calvo MG, Saresella M,
Speciale L, Caputo D: Molecular evidences for a role of HSV-1
in multiple sclerosis clinical acute attack. J Neurovirol 2000, 6
Suppl 2:S109-14.
58. Knox KK, Brewer JH, Henry JM, Harrington DJ, Carrigan DR:
Human herpesvirus 6 and multiple sclerosis: systemic active
infections in patients with early disease. Clin Infect Dis 2000,
31:894-903.
59. Blumberg BM, Mock DJ, Powers JM, Ito M, Assouline JG, Baker JV,
Chen B, Goodman AD: The HHV6 paradox: ubiquitous com-
mensal or insidious pathogen? A two-step in situ PCR
approach. J Clin Virol 2000, 16:159-178.
60. Goodman AD, Mock DJ, Powers JM, Baker JV, Blumberg BM: Human
herpesvirus 6 genome and antigen in acute multiple sclerosis
lesions. J Infect Dis 2003, 187:1365-1376.
Journal of Autoimmune Diseases 2006, 3:1 http://www.jautoimdis.com/content/3/1/1
Page 8 of 8
(page number not for citation purposes)
61. Fazakerley JK, Walker R: Virus demyelination. J Neurovirol 2003,
9:148-164.
62. Buchmeier MJ, Lane TE: Viral-induced neurodegenerative dis-
ease. Curr Opin Microbiol 1999, 2:398-402.
63. Lampert PW: Autoimmune and virus-induced demyelinating
diseases. A review. Am J Pathol 1978, 91:176-208.
64. Watanabe R, Wege H, ter Meulen V: Adoptive transfer of EAE-
like lesions from rats with coronavirus-induced demyelinat-
ing encephalomyelitis. Nature 1983, 305:150-153.
65. Krakowka S, McCullough B, Koestner A, Olsen R: Myelin-specific
autoantibodies associated with central nervous system
demyelination in canine distemper virus infection. Infect
Immun 1973, 8:819-827.
66. Tsunoda IFRS: Theiler's murine encephalomyelitis virus. In
Pesistent viral infections Edited by: R Ahmed IC. Chichester, John Wiley
& Sons Ltd; 1999:517 -5536.
67. Tsunoda I, Fujinami RS: Two models for multiple sclerosis:
experimental allergic encephalomyelitis and Theiler's
murine encephalomyelitis virus. J Neuropathol Exp Neurol 1996,
55:673-686.
68. Dal Canto MC, Lipton HL: Primary demyelination in Theiler's
virus infection. An ultrastructural study. Lab Invest 1975,
33:626-637.
69. Yauch RL, Kim BS: A predominant viral epitope recognized by
T cells from the periphery and demyelinating lesions of SJL/
J mice infected with Theiler's virus is located within
VP1(233-244). J Immunol 1994, 153:4508-4519.
70. Kurtz CI, Sun XM, Fujinami RS: B-lymphocyte requirement for
vaccine-mediated protection from Theiler's murine enceph-
alomyelitis virus-induced central nervous system disease. J
Virol 1995, 69:5152-5155.
71. Rodriguez M, Pavelko KD, Njenga MK, Logan WC, Wettstein PJ: The
balance between persistent virus infection and immune cells
determines demyelination. J Immunol 1996, 157:5699-5709.
72. Roos RP, Firestone S, Wollmann R, Variakojis D, Arnason BG: The
effect of short-term and chronic immunosuppression on
Theiler's virus demyelination. J Neuroimmunol 1982, 2:223-234.
73. Rodriguez M, Lafuse WP, Leibowitz J, David CS: Partial suppres-
sion of Theiler's virus-induced demyelination in vivo by
administration of monoclonal antibodies to immune-
response gene products (Ia antigens). Neurology 1986,
36:964-970.
74. Rodriguez M, Pierce ML, Howie EA: Immune response gene
products (Ia antigens) on glial and endothelial cells in virus-
induced demyelination. J Immunol 1987, 138:3438-3442.
75. Rodriguez M, Oleszak E, Leibowitz J: Theiler's murine encephalo-
myelitis: a model of demyelination and persistence of virus.
Crit Rev Immunol 1987, 7:325-365.
76. Rodriguez M, Leibowitz JL, Lampert PW: Persistent infection of
oligodendrocytes in Theiler's virus-induced encephalomyeli-
tis. Ann Neurol 1983, 13:426-433.
77. Olson JK, Croxford JL, Miller SD: Virus-induced autoimmunity:
potential role of viruses in initiation, perpetuation, and pro-
gression of T-cell-mediated autoimmune disease. Viral Immu-
nol 2001, 14:227-250.
78. Fujinami RS, Oldstone MB: Amino acid homology between the
encephalitogenic site of myelin basic protein and virus:
mechanism for autoimmunity. Science 1985, 230:1043-1045.
79. Olson JK, Croxford JL, Calenoff MA, Dal Canto MC, Miller SD: A
virus-induced molecular mimicry model of multiple sclero-
sis. J Clin Invest 2001, 108:311-318.
80. Olson JK, Ludovic Croxford J, Miller SD: Innate and adaptive
immune requirements for induction of autoimmune demy-
elinating disease by molecular mimicry. Mol Immunol 2004,
40:1103-1108.
81. Wucherpfennig K: T cell mediated autoimmunity in multiple
sclerosis. In Molecular biology of multiple sclerosis Edited by: WC R.
Chichester, John Wiley & Sons, Ltd; 1997:191-200.
82. Horwitz MS, Bradley LM, Harbertson J, Krahl T, Lee J, Sarvetnick N:
Diabetes induced by Coxsackie virus: initiation by bystander
damage and not molecular mimicry. Nat Med 1998, 4:781-785.
83. Horwitz MS, Sarvetnick N: Viruses, host responses, and autoim-
munity. Immunol Rev 1999, 169:241-253.
84. Vanderlugt CL, Miller SD: Epitope spreading in immune-medi-
ated diseases: implications for immunotherapy. Nat Rev Immu-
nol 2002, 2:85-95.
85. Dal Canto MC, Calenoff MA, Miller SD, Vanderlugt CL: Lym-
phocytes from mice chronically infected with Theiler's
murine encephalomyelitis virus produce demyelination of
organotypic cultures after stimulation with the major
encephalitogenic epitope of myelin proteolipid protein.
Epitope spreading in TMEV infection has functional activity.
J Neuroimmunol 2000, 104:79-84.
86. McRae BL, Vanderlugt CL, Dal Canto MC, Miller SD: Functional evi-
dence for epitope spreading in the relapsing pathology of
experimental autoimmune encephalomyelitis. J Exp Med
1995, 182:75-85.
87. Katz-Levy Y, Neville KL, Padilla J, Rahbe S, Begolka WS, Girvin AM,
Olson JK, Vanderlugt CL, Miller SD: Temporal development of
autoreactive Th1 responses and endogenous presentation of
self myelin epitopes by central nervous system-resident
APCs in Theiler's virus-infected mice. J Immunol 2000,
165:5304-5314.
88. Miller SD, Katz-Levy Y, Neville KL, Vanderlugt CL: Virus-induced
autoimmunity: epitope spreading to myelin autoepitopes in
Theiler's virus infection of the central nervous system. Adv
Virus Res 2001, 56:199-217.
89. Vanderlugt CL, Begolka WS, Neville KL, Katz-Levy Y, Howard LM,
Eagar TN, Bluestone JA, Miller SD: The functional significance of
epitope spreading and its regulation by co-stimulatory mol-
ecules. Immunol Rev 1998, 164:63-72.
90. Tuohy VK, Yu M, Yin L, Kawczak JA, Kinkel RP: Spontaneous
regression of primary autoreactivity during chronic progres-
sion of experimental autoimmune encephalomyelitis and
multiple sclerosis. J Exp Med 1999, 189:1033-1042.
91. McMahon EJ, Bailey SL, Castenada CV, Waldner H, Miller SD:
Epitope spreading initiates in the CNS in two mouse models
of multiple sclerosis. Nat Med 2005, 11:335-339.
92. Trapp BD, Peterson J, Ransohoff RM, Rudick R, Mork S, Bo L: Axonal
transection in the lesions of multiple sclerosis. N Engl J Med
1998, 338:278-285.
93. Bjartmar C, Wujek JR, Trapp BD: Axonal loss in the pathology of
MS: consequences for understanding the progressive phase
of the disease. J Neurol Sci 2003, 206:165-171.
94. Bjartmar C, Yin X, Trapp BD: Axonal pathology in myelin disor-
ders. J Neurocytol 1999, 28:383-395.
95. Wujek JR, Bjartmar C, Richer E, Ransohoff RM, Yu M, Tuohy VK,
Trapp BD: Axon loss in the spinal cord determines permanent
neurological disability in an animal model of multiple sclero-
sis. J Neuropathol Exp Neurol 2002, 61:23-32.
96. Grigoriadis N, Ben-Hur T, Karussis D, Milonas I: Axonal damage in
multiple sclerosis: a complex issue in a complex disease. Clin
Neurol Neurosurg 2004, 106:211-217.
97. McGavern DB, Murray PD, Rivera-Quinones C, Schmelzer JD, Low
PA, Rodriguez M: Axonal loss results in spinal cord atrophy,
electrophysiological abnormalities and neurological deficits
following demyelination in a chronic inflammatory model of
multiple sclerosis. Brain 2000, 123 Pt 3:519-531.
98. Ure D, Rodriguez M: Extensive injury of descending neurons
demonstrated by retrograde labeling in a virus-induced
murine model of chronic inflammatory demyelination. J Neu-
ropathol Exp Neurol 2000, 59:664-678.
99. Evangelou N, Esiri MM, Smith S, Palace J, Matthews PM: Quantita-
tive pathological evidence for axonal loss in normal appear-
ing white matter in multiple sclerosis. Ann Neurol 2000,
47:391-395.
100. Filippi M, Bozzali M, Rovaris M, Gonen O, Kesavadas C, Ghezzi A,
Martinelli V, Grossman RI, Scotti G, Comi G, Falini A: Evidence for
widespread axonal damage at the earliest clinical stage of
multiple sclerosis. Brain 2003, 126:433-437.
101. Tsunoda I, Kuang LQ, Libbey JE, Fujinami RS: Axonal injury heralds
virus-induced demyelination. Am J Pathol 2003, 162:1259-1269.
102. Nelson AL, Bieber AJ, Rodriguez M: Contrasting murine models
of MS. Int MS J 2004, 11:95-99.
... Human demyelinating diseases of known viral etiology can also serve as models for MS. Progressive multifocal leukoencephalopathy (PML), a disease with high mortality, is the only known human demyelinating disease with an established viral etiology [20]: after long periods of latency, the John Cunningham virus (JC virus) becomes active during periods of relative immunosuppression and induces astrocytic infection and oligodendrocyte lysis [52]. This neurovirulence could be due to interviral recombination with EBV [53]. ...
... PML supports the hypothesis of an interaction between virus and host, and it suggests a probable viral trigger for autoimmune reactions. In this regard, the TMEV model does not follow the same pathogenesis but instead causes (sub)acute demyelination [52] that is more reminiscent of MS, which manifests with slower clinical deterioration than PML. ...
... Indeed, a recent rare example of a patient sensitized with their own brain cells resulted in an MS-like disease that fulfilled all the pathological diagnostic criteria, direct evidence that self-peptides can induce MS-like disease [63]. The efficacy of this mechanism depends on the APCs and the peptide obtained from a pathogen that resembles the self-myelin peptide [52]. The first proof-of-concept molecular mimicry study was in a murine model of autoimmune herpes stromal keratitis, in which UL-6, a herpes simplex virus (HSV)-1 protein, was recognized by autoreactive T cells, while mutant viral strains not recognized by these T cells failed to induce the disease [64]. ...
Article
Full-text available
Viruses are directly or indirectly implicated in multiple sclerosis (MS). Here, we review the evidence on the virus-related pathophysiology of MS, introduce common experimental models, and explore the ways in which viruses cause demyelination. By emphasizing knowledge gaps, we highlight future research directions for effective MS diagnostics and therapies: (i) identifying biomarkers for at-risk individuals, (ii) searching for direct evidence of specific causative viruses, (iii) establishing the contribution of host genetic factors and viruses, and (iv) investigating the contribution of immune regulation at extra-CNS sites. Research in these areas is likely to be facilitated by the application of high-throughput technologies, the development of systems-based bioinformatic approaches, careful selection of experimental models, and the acquisition of high-quality clinical material for tissue-based research.
... In response to the question regarding T-cell mediation of PR etiopathogenesis, we would like to cite the example of Theiler's murine encephalomyelitis virus (TMEV)-induced encephalomyelitis (8). According to this model, lymphocyte subtypes play a paradoxical role in the TMEV disease process; that is, they participate in virus clearance during the acute phase of the disease and aggravate the demyelinating process in the chronic phase of the disease (8). ...
... In response to the question regarding T-cell mediation of PR etiopathogenesis, we would like to cite the example of Theiler's murine encephalomyelitis virus (TMEV)-induced encephalomyelitis (8). According to this model, lymphocyte subtypes play a paradoxical role in the TMEV disease process; that is, they participate in virus clearance during the acute phase of the disease and aggravate the demyelinating process in the chronic phase of the disease (8). In view of these findings, it might be hypothesized that an extensive peripheral priming of polyfunctional cross-reactive T cells during symptomatic primary DENV infection due to high viral loads and continuous restimulation could potentially establish and maintain a distinct repertoire of virus-specific T cells, which might lead to epitope spreading and could create a predisposition to PR development (9). ...
... Intramolecular spreading refers to epitopes on the same molecule, whereas intermolecular spreading refers to epitopes on different molecules. This can also occur upon CNS damage, which provides plenty new self-antigens, after a persistent virus infection (Grigoriadis and Hadjigeorgiou 2006;Tuohy et al. 1997). On the basis of that mechanism, PLP was experimentally shown to stimulate lymphocytes from mice, chronically infected with Theiler's murine encephalomyelitis virus (TMEV), leading them to provoke myelin sheath destruction in organotypic cultures (Dal Canto et al. 2000). ...
Article
Full-text available
The purpose of this review is to examine whether there is a possible (etiological/triggering) relationship between infection with various Coronaviruses, including Severe Acute Respiratory Syndrome-related Coronavirus-2 (SARS-CoV-2), the virus responsible for the Coronavirus disease-19 (Covid-19) pandemia, and Multiple Sclerosis (MS), and whether an increase of the prevalence of MS after the current Covid-19 pandemia should be expected, examining new and preexisting data. Although the exact pathogenesis of MS remains unknown, environmental agents seem to greatly influence the onset of the disease, with viruses being the most popular candidate. Existing data support this possible etiological relationship between viruses and MS, and experimental studies show that Coronaviruses can actually induce an MS-like demyelinating disease in animal models. Findings in MS patients could also be compatible with this coronaviral MS hypothesis. More importantly, current data from the Covid-19 pandemia show that SARS-CoV-2 can trigger autoimmunity and possibly induce autoimmune diseases, in the Central Nervous System as well, strengthening the viral hypothesis of MS. If we accept that Coronaviruses can induce MS, it is reasonable to expect an increase in the prevalence of MS after the Covid-19 pandemia. This knowledge is of great importance in order to protect the aging groups that are more vulnerable against autoimmune diseases and MS specifically, and to establish proper vaccination and health policies.
... MBP toxicity is due to the fact that this protein is an intrinsically nonstructural protein with a very positive charge [11], and upon release from the myelin cell membrane begins to interact with various molecules, including negatively charged lipids, sialic acids, polyanionic proteins, and neuron plasmas [12], which in turn causes inflammation and death of nerve cells [23]. Virus-mediated autoimmunity seen in multiple sclerosis and Theiler's virus infection was reported to cause T cell-mediated autoimmune disease related to demyelination [22]. The development of a myelin-specific autoimmune response may be a relatively important cause of demyelination in patients with Japanese encephalitis and other viral infections [23]. ...
Article
Full-text available
Objective: The aim: To study the peculiarities of demyelination by detection of changes in the levels of myelin basic protein (MBP) in CSF of patients with acute herpesviral meningitis (M) and meningoencephalitis (ME). Patients and methods: Materials and methods: A total of 136 CSF samples from 68 patients with herpesviral M and ME were collected. The control group consisted of patients with acute respiratory infection and meningismus. MBP level in CSF was identified at the admission and after 10-12 days of treatment. Analysis of MBP concentrations in CSF was performed using an enzyme immunoassay. Results: Results: Examination of patients on the first day of hospitalization showed the presence of a significant increase of MBP in the CSF in all patients with viral M/ME compared with the indicators of the comparison group (р<0.01). In all groups of patients with ME, the level of MBP in CSF was significantly higher than the indicators of comparison group and M groups of the suitable etiology of the disease (p<0.01). In patients with lethal outcome, the MBP level was significantly higher (p<0.01) than in all meningitis groups, but we did not find a significant difference with the patients with ME (p>0.05). Conclusion: Conclusions: The increase of MBP level identified in patients with acute M/ME confirms the presence of the demyelinating process that occurs in all patients, but it is more pronounced in patients with ME.
... In human multiple sclerosis or murine experimental autoimmune encephalomyelitis, CD4 + T cells are critical drivers of autoimmunity against the central nervous system. [3][4][5]. Experimental autoimmune encephalomyelitis (EAE) is usually employed as an ideal animal model of MS [6] and Th1 and Th17 cells have long been considered to be the most important pathogenic CD4 + Th cells during the progress of MS/EAE [7]. ...
Article
Full-text available
MicroRNA 182 is important for the clonal expansion of CD4+ T cells (Th) following IL‐2 stimulation and is a potential therapeutic target for autoimmune diseases. In the present study, we investigated the role of microRNA 182 in the differentiation of pro‐inflammatory CD4+ T helper cell by overexpressing or silencing microRNA 182 expression both in in vivo and in vitro settings. We report that in the studied Chinese cohort, microRNA 182 is upregulated in patients with relapse and remitting multiple sclerosis (RRMS) and this upregulation is associated with increased IFN‐γ producing CD4+ Th1 cells in the circulation. In the murine experimental autoimmune encephalomyelitis (EAE) model, global microRNA 182 overexpression exacerbates clinical symptoms and results in augmented CD4+IFN‐γ+ Th1 and CD4+IL‐17+ Th17 differentiation in vivo. Addition of microRNA 182 mimics in vitro represses both the protein expression and transcriptional activity of hypoxia induced factor 1α (HIF‐1α) but increases the level of IFN‐γ transcripts in sorted murine CD4+ T cells. Together, our results provide evidence that microRNA 182 may be one of the transitional hubs contribution to regulate Th cells expansion in response to self‐antigens and differentiation of antigen specific Th cells during the progression of autoimmune inflammations. This article is protected by copyright. All rights reserved
... The etiology of MS remains unknown. Several environmental factors, including microbial agents, have been considered potential inducers of the disease [3]. ...
Article
Full-text available
Background Multiple sclerosis (MS) is an autoimmune demyelinating disorder. The etiology of MS remains unknown exactly. Helicobacter pylori heat shock proteins were suggested as a potential trigger of immune system causing MS. Objectives The aim of this study was to assess the level of anti-Helicobacter pylori heat shock proteins 60 (Hp hsp60) antibodies at patients of MS and to correlate it with various epidemiological and clinical data. Subjects and methods This study design was a cross-sectional case control one. A total of 65 patients with multiple sclerosis diagnosed according to 2010 revised McDonald criteria and other 65 age- and sex-matched healthy controls were included in this study. All participants were subjected to full history taking, complete neurological examination including Expanded Disability Status Scale (EDSS) for the patients, measurement of serum level of anti-Hp hsp60 IgG using ELISA technique, and MRI brain for all the patients, being a goldstone for inclusion in the study. Results There was statistically significant high level of anti-Hp hsp60 IgG at MS patients especially secondary progressive multiple sclerosis (SPMS) patients. Moreover, a positive statistically significant correlation was found between it and age of patients, duration of illness, and EDSS. Conclusion We conclude that hsp60 of Hp may be a useful biomarker for attesting course progression in MS.
... 14 Although the evidence for a causative viral aetiology for MS in humans remains inconclusive, viruses appear to play a role in modulating the neuro-immunological system of genetically susceptible individuals to cause MS. For instance, IgG antibodies against several viruses including varicella zoster virus (VZV), cytomegalovirus (CMV), measles, rubella, mumps and herpes simplex virus (HSV-1) have been identified in the cerebrospinal fluid (CSF) of patients with MS. 15,16 More recently, other viruses have attracted attention including Saffold virus (a novel human cardiovirus). 17,18 With this in mind, this review provides an indepth discussion of the viruses implicated in MS pathogenesis. ...
Article
Full-text available
Multiple sclerosis is an immune-mediated disease with an environmental component. According to a long-standing but unproven hypothesis dating to initial descriptions of multiple sclerosis (MS) at the end of the 19th century, viruses are either directly or indirectly implicated in MS pathogenesis. Whether viruses in MS are principally causal or simply contributory remains to be proven, but many viruses or viral elements-predominantly Epstein-Barr virus, human endogenous retroviruses (HERVs) and human herpesvirus 6 (HHV-6) but also less common viruses such as Saffold and measles viruses-are associated with MS. Here, we present an up-to-date and comprehensive review of the main candidate viruses implicated in MS pathogenesis and summarize how these viruses might cause or lead to the hallmark demyelinating and inflammatory lesions of MS. We review data from epidemiological, animal and in vitro studies and in doing so offer a transdisciplinary approach to the topic. We argue that it is crucially important not to interpret "absence of evidence" as "evidence of absence" and that future studies need to focus on distinguishing correlative from causative associations. Progress in the MS-virus field is expected to arise from an increasing body of knowledge on the interplay between viruses and HERVs in MS. Such interactions suggest common HERV-mediated pathways downstream of viral infection that cause both neuroinflammation and neurodegeneration. We also comment on the limitations of existing studies and provide future research directions for the field.
Chapter
In recent decades, several neurodegenerative diseases have been shown to be exacerbated by systemic inflammatory processes. There is a wide range of literature that demonstrates a clear but complex relationship between the central nervous system (CNS) and the immunological system, both under naïve or pathological conditions. In diseased brains, peripheral inflammation can transform “primed” microglia into an “active” state, which can trigger stronger pathological responses. Demyelinating diseases are a group of neurodegenerative diseases characterized by inflammatory lesions associated with demyelination, which in turn induces axonal damage, neurodegeneration, and progressive loss of function. Among them, the most important are multiple sclerosis (MS) and neuromyelitis optica (NMO). In this review, we will analyze the effect of specific peripheral inflammatory stimuli in the progression of demyelinating diseases and discuss their animal models. In most cases, peripheral immune stimuli are exacerbating.
Article
Full-text available
Multiple sclerosis is a complex genetic disease associated with inflammation in the CNS white matter thought to be mediated by autoreactive T cells. Clonal expansion of B cells, their antibody products, and T cells, hallmarks of inflammation in the CNS, are found in MS. This review discusses new methods to define the molecular pathology of human disease with high-throughput examination of germline DNA haplotypes, RNA expression, and protein structures that will allow the generation of a new series of hypotheses that can be tested to develop better understanding of and therapies for this disease.
Article
Full-text available
Summary The role of epitope spreading in the pathology of relapsing-remitting experimental autoimmune encephalomyelitis (R-EAE) was examined. Using peripherally induced immunologic tolerance as a probe to analyze the neuropathologic T cell repertoire, we show that the majority of the immunopathologic reactivity during the acute phase of R-EAE in SJL/J mice induced by active immunization with the intact proteolipid (PLP) molecule is directed at the PLP139-151 epitope and that responses to secondary encephalitogenic PLP epitopes may contribute to the later relapsing phases of disease. Intermolecular epitope spreading was demonstrated by showing the develop- ment of T cell responses to PLP139-151 after acute disease in mice in which R-EAE was initiated by the transfer ofT cells specific for the non-cross-reactive MBP84-104 determinant. Intramolecular epitope spreading was demonstrated by showing that endogenous host T cells specific for a sec- ondary encephalitogenic PLP epitope (PLP178-191) are demonstrable by both splenic T cell prolifer- ative and in vivo delayed-type hypersensitivity responses in mice in which acute central nervous system damage was initiated by T cells reactive with the immunodominant, non-cross-reactive PLP139-151 sequence. The PLP178-191-specific responses are activated as a result of and correlate with the degree of acute tissue damage, since they do not develop in mice tolerized to the initi- ating epitope before expression of acute disease. Most importantly, we show that the PLP178- 191-specific responses are capable of mediating R-EAE upon adoptive secondary transfer to naive recipient mice. Furthermore, induction of tolerance to intact PLP (which inhibits responses to both the initiating PLP139-151 epitope and to the PLP178-191 epitope) after the acute disease episode is sufficient to prevent relapsing disease. These results strongly support a contributory role ofT cell responses to epitopes released as a result of acute tissue damage to the immunopatho- genesis of relapsing clinical episodes and have important implications for the design of antigen- specific immunotherapies for the treatment of chronic autoimmune disorders in humans.
Article
One of the characteristics of multiple sclerosis is the unpredictable occurrence of exacerbations and remissions. These fluctuations in disease activity are related to alterations in (auto‐)immune activity. Exacerbations lead to short‐term morbidity, but may also influence long‐term disability. This longitudinal study in 73 patients with relapsing–remitting multiple sclerosis assessed the contribution of systemic infections to the natural course of exacerbations. In addition, we analysed whether infections lead to an increase in the number of gadolinium‐enhancing lesions. A total of 167 infections and 145 exacerbations were observed during 6466 patient weeks. During a predefined at‐risk period (ARP) of 2 weeks before until 5 weeks after the onset of a clinical infection (predominantly upper airway infections), there was an increased risk of exacerbations (rate ratio 2.1), which is in accordance with previous studies. Exacerbations with onset during the ARP led more frequently to sustained deficit [increase of ≥1 Expanded Disability Status Scale (EDSS) point or ≥0.5 above EDSS 5.5 for >3 months] than exacerbations with onset outside the ARP, with a rate ratio of 3.8. Minor and major exacerbations were equally distributed between the ARP and non‐ARP onset groups. ARP exacerbations were associated with significantly higher plasma levels of the inflammatory marker soluble intracellular adhesion molecule 1 than non‐ARP exacerbations, indicating relatively enhanced immune activation during ARP relapses. Three serial MRI scans were performed after the onset of an infection over a 6‐week period. There was no difference in the number of gadolinium‐enhancing lesions between the three time points. In conclusion, exacerbations in the context of a systemic infection lead to more sustained damage than other exacerbations. There is no indication that this effect occurs through enhanced opening of the blood–brain barrier.
Article
Although axonal pathology is recognized as one of the major pathological features of multiple sclerosis, it is less clear how early in its course it occurs and how it correlates with MRI‐visible lesion loads. To assess this early axonal pathology, we quantified the concentration of whole‐brain N ‐acetylaspartate (WBNAA) in a group of patients at the earliest clinical stage of the disease and compared the results with those from healthy controls. Conventional brain MRI and WBNAA using unlocalized proton magnetic resonance spectroscopy were obtained from 31 patients at presentation with clinically isolated syndromes suggestive of multiple sclerosis and paraclinical evidence of dissemination in space, and from 16 matched controls. An additional conventional MRI scan was obtained in all patients 4–6 months later to detect dissemination of lesions in time. The mean WBNAA concentration was significantly lower in patients compared with the controls ( P < 0.0001). It was not significantly different between patients with and without enhancing lesions at the baseline MRI or between patients with and without lesion dissemination in time. No correlation was found between WBNAA concentrations and lesion volumes. Widespread axonal pathology, largely independent of MRI‐visible inflammation and too extensive to be completely reversible, occurs in patients even at the earliest clinical stage of multiple sclerosis. This finding lessens the validity of the current concept that the axonal pathology of multiple sclerosis is the end‐stage result of repeated inflammatory events, and argues strongly in favour of early neuroprotective intervention.
Article
Epitope spreading is a process whereby epitopes distinct from and non-cross-reactive with an inducing epitope become major targets of an ongoing immune response. This phenomenon has been defined in experimental and natural situations as a consequence of acute or persistent infection and secondary to chronic tissue destruction that occurs during progressive autoimmune disease. We have investigated the functional significance of this process in the chronic stages of both autoimmune and virus-induced central nervous system (CNS) demyelinating disease models in the SJL/J mouse. During the relapsing-remitting course of experimental autoimmune encephalomyelitis (R-EAE) induced with defined encephalitogenic myelin peptides, CD4+ T cells specific for endogenous epitopes on both the initiating myelin protein (intramolecular epitope spreading) and distinct myelin proteins (intermolecular epitope spreading) are primed secondary to myelin destruction during acute disease and play a major functional role in mediating disease relapses. Similarly, epitope spreading to endogenous myelin epitopes appears to play a major functional role in the chronic-progressive course of Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD), a virus-induced CD4+ T-cell-mediated immunopathology. In TMEV-IDD, myelin destruction is initiated by virus-specific CD4+ T cells which target virus epitopes persisting in CNS-derived antigen-presenting cells. However, the chronic stage of this progressive disease is associated with the activation of CD4+ T cells specific for multiple myelin epitopes. In both models, the temporal course of T-cell activation occurs in a hierarchical order of epitope dominance, spreading first to the most immunodominant epitope and progressing to lesser immunodominant epitopes. In addition, epitope spreading in R-EAE is regulated predominantly by CD28/B7-1 co-stimulatory interactions, as antagonism of B7-1-mediated co-stimulation using anti-B7-1 F(ab) fragments is an effective ameliorative therapy for ongoing disease. The process of epitope spreading bas obvious important implications for the design of antigen-specific therapies for the treatment of autoimmune disease since these therapies will have to identify and target endogenous self epitopes associated with chronic tissue destruction.
Article
Birth order position was examined in 164 cases with sporadic multiple sclerosis (MS), i.e. no other family members with MS, and spousal controls, matched for sibship size, socioeconomic status and opposite sex. The results did not find an association between birth order position and the subsequent development of MS and thus do not support the concept of an infectious cause for MS where “early exposure” is protective and exposure to the infection is a single event of short duration.
Article
Conceptually, the initiation of autoimmune disease can be described as a three-stage process involving both genetic and environmental influences. This process begins with the development of an autoimmune cellular repertoire, followed by activation of these autoreactive cells in response to a localized target and, finally, the immune system's failure to regulate these self reactive constituents. Viruses have long been associated with inciting autoimmune disorders. Two mechanisms have been proposed to explain how a viral infection can overcome immunological tolerance to self components and initiate an organ specific autoreactive process, these mechanisms arc molecular mimicry and bystander activation. Both pathways, as discussed here, could play pivotal roles in the development of autoimmunity without necessarily excluding each other. Transgene technology has allowed us and others to examine more closely the roles of these mechanisms in mice and to dissect the requirements for initiating disease. These results demonstrate that bystander activation is the must likely explanation fur disease development. Additional evidence suggests a further role for viruses in the reactivation and chronicity of autoimmune diseases. In this scenario, a second invasion by a previously infecting virus may restimulate already existing autoreactive lymphocytes and thereby contribute to the diversity of the immune response.
Article
Myelination provides extrinsic trophic signals that influence normal maturation and long-term survival of axons. The extent of axonal involvement in diseases affecting myelin or myelin forming cells has traditionally been underestimated. There are, however, many examples of axon damage as a consequence of dysmyelinating or demyelinating disorders. More than a century ago, Charcot described the pathology of multiple sclerosis (MS) in terms of demyelination and relative sparing of axons. Recent reports demonstrate a strong correlation between inflammatory demyelination in MS lesions and axonal transection, indicating axonal loss at disease onset. Disruption of axons is also observed in experimental allergic encephalomyelitis and in Theiler's murine encephalomyelitis virus disease, two animal models of inflammatory demyelinating CNS disease. A number of dysmyelinating mouse mutants with axonal pathology have provided insights regarding cellular and molecular mechanisms of axon degeneration. For example, the myelin-associated glycoprotein and proteolipid protein have been shown to be essential for mediating myelin-derived trophic signals to axons. Patients with the inherited peripheral neuropathy Charcot-Marie Tooth disease type 1 develop symptomatic progressive axonal loss due to abnormal Schwann cell expression of peripheral myelin protein 22. The data summarized in this review indicate that axonal damage is an integral part of myelin disease, and that loss of axons contributes to the irreversible functional impairment observed in affected individuals. Early neuroprotection should be considered as an additional therapeutic option for these patients.