Neuroscience & Medicine, 2012, 3, 203-224
http://dx.doi.org/10.4236/nm.2012.33026 Published Online September 2012 (http://www.SciRP.org/journal/nm)
203
Initiation and Regulation of CNS Autoimmunity: Balancing
Immune Surveillance and Inflammation in the CNS
Melissa G. Harris1,2, Zsuzsanna Fabry1
1Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, USA;
2Neuroscience Training Program, School of Medicine and Public Health, University of Wisconsin, Madison, USA.
Email: zfabry@facstaff.wisc.edu
Received June 7th, 2012; revised July 4th, 2012; accepted July 15th, 2012
ABSTRACT
While the central nervous system (CNS) was once thought to be immune privileged, more recent data support that cer-
tain areas of the healthy CNS are routinely patrolled by immune cells. Further, antigen drainage is another means by
which the adaptive arm of the immune system can gain information about the health of the CNS. Altogether these en-
sure that the CNS is not beyond the scope of immune protection against viruses and tumors. However, immune surveil-
lance in the CNS has to be tightly regulated, as CNS autoimmune disease and inflammation may arise from increased
immune cell infiltration. In this review we discuss the concept and implications of CNS immune surveillance and in-
troduce the CNS antigen-presenting cells (APCs) that potentially regulate neuroinflammation and autoimmunity. We
also discuss novel animal models in which CNS disease initiation and the role of APCs in disease regulation can be
tested.
Keywords: CNS; Immune Surveillance, Autoimmunity; APCs; DCs; Oligodendrocyte Death; DAMPs; Initiation;
Regulation
1. Introduction
The immune system has evolved to help the body fight
foreign pathogens and harmful self-intruders, such as
tumors. Immunity requires continual surveillance of the
body by immune cells, primarily tissue-resident macro-
phages and dendritic cells (DCs), which initiate inflam-
matory responses that result in the recruitment of T cells
and other leukocytes to the site of infection or damage.
This process is highly restricted in the healthy central
nervous system (CNS) due to several regulatory factors
that preclude the infiltration of activated T cells from the
blood into the CNS parenchyma, thus contributing to the
immune privileged status of the CNS. In spite of this
regulation, it has been shown that limited surveillance by
T cells still promotes the health of this tissue. However,
increased immunological surveillance and immune cell
infiltration into the CNS may lead to inflammation and
autoimmune disease [1,2]. One idea is that immune cells,
particularly DCs, which accumulate in the CNS under in-
flammatory conditions may pick up and deliver myelin
antigens to lymph nodes for the priming of adaptive im-
mune responses. This process could lead to the initiation
or exacerbation of CNS autoimmune disease. Thus, an
understanding of how adaptive immunity is generated
against CNS self-antigens and how it is regulated is nec-
essary for treating diseases such as multiple sclerosis
(MS). In this review we will focus on T cell-mediated
adaptive immunity and will discuss conceptual changes
in our understanding of CNS surveillance and the role of
CNS antigen-presenting cells (APCs) in regulating adap-
tive immunity in the CNS. We will further discuss cur-
rent models of CNS autoimmune disease initiation, and
consider the potential contribution of damage-associated
molecules to the exacerbation of CNS autoimmunity.
2. CNS Immunity: Balancing Surveillance
and Autoimmunity
The CNS has historically been considered immune privi-
leged. It was originally thought that antigens within the
CNS parenchyma went unnoticed by circulating immune
cells, mainly because the blood-brain barrier (BBB) kept
them out (reviewed in [3]). This was thought to explain
how foreign tissue grafts could survive in the CNS for
long periods of time. It is now generally accepted that
immune privilege is only afforded to the parenchyma,
and not to the cerebrospinal fluid (CSF)-exposed parts of
the CNS (i.e., leptomeninges, choroid plexus, circum-
ventricular organs, and ventricles) [3]. The developed
BBB (referred to as the neurovascular unit, or NVU)
consists of not only vascular endothelial cells, but also
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
204
the basement membrane of these cells and that of the
astrocytic endfeet (i.e., the glia limitans), which delineate
the CSF-filled perivascular space (PVS) that drains into
the leptomeningeal subarachnoid space (SAS; Figure 1)
(reviewed in [4]). Additionally, pericytes of the PVS, as
well as neurons and extracellular matrix, are all part of
the NVU (reviewed in [5]). Important to our discussion,
the SAS is an active immunological niche that is crucial
in the development and maintenance of CNS autoim-
mune disease.
Under homeostatic conditions T lymphocytes are re-
stricted in their capacity to cross endothelial cells of the
quiescent parenchymal BBB. However, activated T cells
may cross into the SAS by binding adhesion molecules
(namely, P-selectin via PSGL-1, but also ICAM-1) and
cytokines (e.g., CCL20 via CCR6) constitutively ex-
pressed by endothelial cells of the meningeal BBB or
epithelial cells of the choroid plexus, which form the
blood-cerebrospinal fluid barrier (BCSFB; [6]; reviewed
in [4,7]). Thus, activated T cells present in normal CSF
and stroma of the choroid plexus and meninges are
thought to carry out routine immune surveillance in the
SAS [6,8], which is rich in CNS antigens that drain into
the CSF from interstitial fluid. This process is vital to the
health of the CNS, as clinical reports have described pa-
tients developing opportunistic viral infections in the
CNS and subsequent progressive multifocal leukoen-
cephalopathy when T cell transmigration is inhibited
[9-11]. Of further clinical relevance, the SAS is believed
to be the initial site of CCR6-mediated entry by patho-
genic Th17 cells [12], which regulate the initiation of
experimental autoimmune encephalomyelitis (EAE), the
animal model of MS [13]. Interestingly, Th17 cells also
mediate the formation of ectopic lymphoid follicles
(eLFs) [14], which have been observed in the meninges
of both mice with EAE [14,15] and patients with secon-
dary (chronic) progressive MS [16,17]. These structures
(discussed later) are thought to be important for main-
taining chronic inflammation and may be key determi-
nants for relapse and progression in the case of CNS
autoimmune disease [17].
While it now seems clear that the CNS is immu-
nologically monitored by activated T cells, which can be
beneficial, the question remains: how do they become
activated in the first place? With the exception of the
CNS, every tissue in the body is connected to a complex
network of lymphatic vessels. One of the key functions
of this lymphatic system is to allow for drainage/homing
of both soluble antigen and antigen-bearing cells (espe-
cially DCs) to peripheral lymphoid tissues to engage the
Figure 1. Anatomical locations supporting immune cell surveillance and entry into the CNS. Routine immune surveillance of
the CNS is thought to occur by activated memory T cells in the subarachnoid space (SAS) of the leptomeninges, which is
comprised of the arachnoid mater and the pia mater (left picture). These T cells may enter into the CSF-filled SAS either
from postcapillary venules of the leptomeninges or across the epithelial cells of the choroid plexus, which form the
blood-cerebrospinal fluid barrier (not shown). Together with neurons, the neurovascular unit (right picture) is comprised of
the endothelial cells of the blood-brain barrier (BBB) and their underlying basement membrane, the perivascular space
(PVS), and the basement membrane of the astrocytic endfeet forming the glia limitans. In the presence of neural inflamma-
tion, activated T cells may cross the endothelial cells of the BBB into the PVS, presumably where they are re-primed by cog-
nate antigen-bearing APCs that allow them to enter the CNS parenchyma.
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS 205
adaptive immune system. However, without draining
lymphatics, how do antigens that are sequestered behind
the BBB generate peripheral T cell responses? In the
context of infection, it was proposed that bacterial or
viral pathogens are allowed to persist in the CNS paren-
chyma without eliciting adaptive immune responses,
which can only be generated following peripheral subcu-
taneous challenge with certain viruses [18-21]. In con-
trast, it is not known how normal, non-pathogenic CNS
self-antigens (such as those from myelin) might drain
and become the targets of autoimmune attack, despite the
apparent immune privilege afforded to CNS pathogens.
One possible explanation is that autoreactive T cell re-
sponses might develop following delayed-type hypersen-
sitivity response to CNS infection, due to generation of
new antigens created by bystander myelin damage [19].
However, in the absence of infection, it is hard to under-
stand how this might occur.
The traditional method used to study soluble antigen
drainage from the CNS has been intracerebral antigen
injection. Using this technique, it was established that,
despite the CNS not having conventional lymphatics,
protein antigens injected intracerebrally into different
parenchymal regions (i.e., caudate nucleus, internal cap-
sule, and midbrain) and into CSF could be largely recov-
ered in the cervical lymph nodes (CLNs) [22,23]. As
reviewed by Cserr and Knopf [22], antigens may drain
from the CNS into the blood by exiting the SAS through
the arachnoid villi, which protrude into the dural sinus.
The second exit is along the cranial nerves, in particular,
along the olfactory pathway and nasal lymphatics to the
CLNs [22]. To support the functional significance of this
drainage process, it was shown that protein antigens that
drain to the periphery are capable of eliciting adaptive
immune responses [24,25], and may even be more im-
munogenic (as reflected in higher antibody titers) than
the same antigens introduced peripherally [24]. We have
also shown that both soluble and cell-bound intracere-
brally injected antigens drain to the CLNs, and this is
followed by the preferential recruitment of primed re-
sponder T cells to the CNS [26-29]. This accumulation of
effector T cells in the CNS may be important because it
has the potential to initiate and/or exacerbate autoim-
mune disease.
The technique of intracerebral antigen injection is a
relatively easy and straightforward procedure in which
antigen can be stereotaxically injected into desired loca-
tions of the brain. It is also fairly quantitative, giving the
investigator control over the concentration of antigen or
number of antigen-pulsed cells introduced. We previ-
ously reported a titration effect, in which the number of
antigen-pulsed dendritic cells (DCs) injected into the
brain positively correlated with their accumulation in the
CLNs, as well as with the numbers of activated antigen-
specific T cells recruited to the brain [26]. However, it is
difficult to control for blood-brain barrier disruption, as
minor tissue trauma from the needle injection may lead
to inadvertent but minor immune cell activation and re-
cruitment to the CNS [22]. Another limitation of this
technique is that it disconnects antigen release and cellu-
lar signals associated with cell death (i.e., damage-asso-
ciated molecular patterns, or DAMPs), thereby preclude-
ing study of the potentially immunogenic role of DAMPs
in priming CNS antigen-specific adaptive immune re-
sponses.
To overcome the above listed limitations, newer mod-
els have more recently been created to test the critical
questions of how CNS cell-specific neoantigens are rec-
ognized by the peripheral immune system, and how this
recognition leads to CNS pathology. Notably, neural
cell-specific neoantigen models have been created in
which expression of immunogenic antigens is restricted
to neurons [30,31], astrocytes [32], oligodendrocytes
(ODCs) [33,34], and both ODCs and Schwann cells [35],
and is achieved either by using Cre driver transgenic
mouse lines or by having the neoantigens under direct
neural cell-specific promoter control. These models are
being used to understand mechanisms of immune toler-
ance and reactivity to CNS antigens, the findings of
which are summarized in Table 1. We created mice with
myelinating glial cell-specific expression of major histo-
compatibility complex (MHC) class I- and MHC class
II-restricted ovalbumin neoepitopes in order to study the
mechanism of myelinating cell-specific antigen recogni-
tion by immune cells and requirements for antigen-spe-
cific CD4+ or CD8+ T cell infiltration in the CNS under
normal and inflammatory conditions. Our findings sug-
gest that myelinating cell-specific neoantigen expression
itself is not sufficient to induce neoantigen-specific T cell
accumulation into the CNS. Other signals induced by
neuroinflammation are required for the accumulation of
neoantigen-specific CD8+ T cells in the CNS. We also
found that ovalbumin neoantigen-specific CD8+ T cells
exacerbate myelin oligodendrocyte glycoprotein (MOG)-
induced inflammation in EAE (manuscript in prepara-
tion). This supports the overall hypothesis that increased
T cell surveillance could contribute to the initiation and
maintenance of CNS autoimmunity.
3. Antigen-Presenting C e l l s i n t h e C N S :
Locations, Function, and Subtypes
As discussed above, activated T cells routinely survey
the SAS of the healthy CNS, yet how and where
CNS-infiltrating T cells are initially primed in humans
and the conditions under which this leads to autoimmu-
nity are unknown. Once primed, however, these T cells
must then re-encounter their antigen in the appropriate
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
206
Table 1. Neural cell-specific neoantige n mode ls.
Cell type Promoter or
Cre driver line
Neoantigen or
Neoantigen/Lox
line
Neoantigen
localization
(and/or site of
normal protein
expression
[Ref])
Major Finding Additional comments Reference
(s)
ODCs MOGi-Cre
Influenza
hemagglutinin
(Rosa26tm(HA)1Lib1
mice)
unspecified
(myelin sheath
[34])
ODC-targeted attack by
pre-activated
neoantigen-specific CTLs
and neurological disease
Ignorance of neoantigen by
neoantigen-specific CD8+ T
cells in Lox/Cre x CL4-TCR
mice
[34,37]
ODCs MBP
floxed ovalbumin
(ODC-OVA mice)
cytosolic
(cytoplasmic
side of
membrane [38])
Neoantigen-specific CD8+ T
cells induce ODC death and
neurological disease in
ODC-OVA x OT-I mice
Ignorance of neoantigen by
neoantigen-specific
CD4+ T cells in ODC-OVA x
OT-II mice
[33,39]
ODCs/Schwann
cells
PLP-CreERT2
(inducible)
LCMV- and
β-gal-derived CD8+
T cell neoepitopes
(ST33.396 mice)
Unspecified
(myelin sheath
[40])
Endogenous CD8+ T cell
tolerance to neoantigens
induced by DCs
- [35,41]
astrocytes and
enteric glial cells GFAP influenza
hemagglutinin cytoplasm [42]
Neoantigen-specific CD4+ T
cell ignorance in GFAP-HA
x HNT-TCR mice
- [32]
neurons NSE ovalbumin
cell
surface/
membrane [43]
intracerebral infection with
Listeria monocyto-
genes-ovalbumin induces
neurological disease, medi-
ated by
SIINFEKL-specific CD8+ T
cells
No endogenous OVA323-339
CD4+ response [30]
neurons CamK-iCre
influenza
hemagglutinin
(Rosa26tm(HA)1Lib1
mice)
unspecified
(cytosolic or
anchored to
cytoskeleton,
depending on
isoform [44])
transient encephalomyelitis
but development of chronic
diabetes
insipidus due to
destruction of
hypothalamic neurons by
neoantigen-specific CTLs
- [31,45]
ODCs = oligodendrocytes; MOG = myelin oligodendrocyte glycoprotein; MBP = myelin basic protein; PLP = proteolipid protein; GFAP = glial fibrillary acidic
protein; NSE = neuron-specific enolase promoter; CamK = calcium/calmodulin-dependent kinase; CTL = cytotoxic T lymphocyte; TCR = T cell receptor; HA
= hemagglutinin.
MHC context by a functional (and as yet uncharacterized)
APC before entering the CNS parenchyma and initiating
disease [7,46]. Naïve T cells that might indiscriminately
enter the CNS once inflammation is established also re-
quire antigen in order to become activated in situ in the
CNS [47]. It was proposed that this process is limited in
the healthy CNS, however, as there is no clear evidence
that DCs, the only cells capable of activating naïve T
cells, are present in the healthy CNS parenchyma in de-
tectable numbers [3]. However, cells that carry common
features of DCs, such as OX62 [48,49], MHC class II,
and the integrin alpha X molecule CD11c [8,50], have
been shown in the healthy rodent meninges and choroid
plexus. These meningeal/choroid plexus APCs might
represent a unique subpopulation of DCs that can con-
tribute to the development and regulation of CNS auto-
immunity. Additionally, DCs do accumulate in the in-
flamed CNS [48,51-53], indicating that these cells are
important for the maintenance of chronic inflammation in
these tissues.
In this section we focus on microglia, astrocytes, and
DCs as in situ CNS APC candidates, potentially capable
of regulating the initiation of neuroinflammation. Things
to consider in evaluation of their APC candidacy are 1)
how efficiently they activate T cells through upregulation
of co-stimulatory molecules (in particular, CD40, CD80
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS 207
(B7-1), CD86 (B7-2), and ICAM-1) and also MHC class
I and II; and 2) their ability to process and present CNS
self-antigen. As CD8+ T cells are the dominant subset of
inflammatory infiltrates observed in MS lesions [54], the
ability of the candidate APCs to cross-present exoge-
nously-derived antigen to CD8+ T cells will also be
given special consideration apart from activation of
CD4+ T cells, which govern EAE pathology. Addition-
ally, CNS-associated macrophages (found in the choroid
plexus, meninges, and perivascular space), as well as
endothelial cells and pericytes, may also be involved in
antigen presentation and disease pathology [55-60], but
they will not be discussed here.
3.1. Microglia: Providers of Neuronal Support
and Parenchymal Surveillance
The “oldest” APCs that were proposed to contribute to
CNS immunity were the microglia and were first de-
scribed by Pio del Rio-Hortega (reviewed in [61]). Mi-
croglia are abundant everywhere in the CNS [61] and are
considered CNS-resident macrophages that arise from a
primitive myeloid progenitor population in the extra-
embryonic yolk sac that enters the embryonic brain as
blood vessels begin to develop (E9.5) [62]. The different-
tiation of the shared common myeloid progenitor into the
granulocyte-monocyte progenitor (which further differ-
entiates into monocytes, DCs, and macrophages) takes
place during another developmental wave in the fetal
liver (“definitive hematopoiesis”), and eventually occurs
in the bone marrow (reviewed in [63]). Additionally,
microglia can renew in situ without contribution from
circulating hematopoietic cells [62]. While microglia are
known phagocytes of cellular material in health and dis-
ease [64], one of their primary functions in the healthy
CNS is to maintain neuronal synapses, which is in part
reflected by their ramified (non-macrophage-like) mor-
phology [65]. Likewise, they are under tight regulatory
control by active neurons (reviewed in [66,67]). However,
and of relevance in terms of innate immunity, microglia
also provide routine and active surveillance of the nerv-
ous tissue and are thus immediate responders to danger.
Their production of chemokines and proinflammatory
cytokines allows for the recruitment and entry of immune
cells from the periphery [66].
In terms of their antigen-presenting capacity, resting
microglia (i.e., CD11b+ CD45low) express very low levels
of MHC class I and II and the costimulatory molecules
CD40, CD80, CD86, and ICAM-1, making them less
efficient at priming naïve T cells compared to DCs [68].
This was demonstrated in earlier in vitro experiments, in
which resting microglia isolated from neonatal mouse
brains required signaling through B7/CD28 (endowed by
the addition of IFN-
and granulocyte-macrophage col-
ony-stiumulating factor, GM-CSF) and CD40/CD40L
in order to serve as more efficient “professional” APCs
[69]. In contrast, peripheral DCs were demonstrated to be
much more efficient APCs than activated microglia in
their ability to prime naïve T cells; however, microglia
were just as effective as DCs in their ability to prime
helper T (Th) cell lines [70]. In vivo, microglia have a
relatively limited capacity to “pick up” and present anti-
gens to naïve T cells infiltrating the inflamed CNS. Re-
sults from S. Miller’s group show that microglia isolated
from the CNS of mice with relapsing-EAE (R-EAE) re-
main relatively poor stimulators of naïve proteolipid
protein PLP139-151-specific CD4+ T cells ex vivo, and only
become strong stimulators upon addition of exogenous
antigen at a very high APC:T cell ratio (1:1) [71]. How-
ever, they could stimulate CD4+ T cell lines much more
efficiently. Recently, microglia isolated from the brain of
naïve adult wild type mice have also been shown to be
capable of TAP-dependent cross-presentation of soluble
antigen in vitro and also intracerebrally injected antigen
ex vivo to naïve CD8+ T cells and T cell lines [72]. This
ability was enhanced in microglia when stimulated with
GM-CSF or CpG oligodeoxynucleotide. However, these
results were obtained using very high concentrations of
antigen (100 - 200 M in vitro) at a very high APC:T
cell ratio (2:1), and likely do not reflect how much anti-
gen is normally taken up in vivo, which (as the data in
[71] suggests) is probably very little. Additionally, in all
of these studies it is difficult to compare the efficiency
with which microglia can stimulate effector T cell line-
ages that develop in vivo with their ability to stimulate T
cell lines in vitro, since T cell lines require very minimal
stimulation in order to become activated.
While activated microglia (i.e., CD11b+ CD45high)
characterize most neuroinflammatory diseases, their ac-
tual contribution to CNS injury or protection is contro-
versial. In response to neuroinflammation, activated mi-
croglia can upregulate all of the co-stimulatory molecules
needed for facilitating adaptive immune responses in the
CNS, which have been found in human MS tissue ([73],
and the references therein). Prior to the manifestation of
EAE clinical symptoms, microglia activation and prolif-
eration have been noted [74], and temporally coincide
with the entry of IL-17- and IFN-
-producing T cells into
the brain [75]. At this time microglia also start to up-
regulate MHC class II, CD40, CD80, and CD86 mole-
cules, suggesting that their primary role in this early
phase is to re-activate entering T cells or to prime naïve
T cells indiscriminately entering the inflamed CNS [75].
Indeed, EAE was greatly attenuated in ganciclovir (GCV)-
treated bone marrow chimeric CD11b-HSVTK mice (i.e.,
having CD11b promoter-driven, GCV-responsive herpes
simplex virus thymidine kinase), which have paralyzed
microglia but otherwise functional CD11b-expressing
monocytic/macrophage cells [76]. Additionally, there were
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
208
fewer lymphocytes in the CNS of these mice. These
studies illustrate that microglia contribute to the devel-
opment of CNS autoimmune disease. However, micro-
glia have also been shown to have suppressive functions,
as in vitro activated microglia presenting the immuno-
genic myelin basic protein (MBP) Ac1-11 peptide have
been shown to induce T cell anergy and death [69,77].
This tolerogenic outcome (as opposed to T cell activation)
in vivo would depend on both T cell avidity for self-an-
tigen (i.e., the number of peptide/MHC complexes, as
well as the affinity of the T cell receptor for the pep-
tide/MHC complex), which may increase under neuroin-
flammatory conditions [7], and also the local cytokine
milieu, which influences the activation level of APCs in
the CNS. Undoubtedly, the function of microglia needs
to be further studied, as these cells might be important in
both the induction and regulation of CNS autoimmunity.
Cytokines have been shown to exert a strong influence
on the phenotype acquired by peripheral monocyte-de-
rived macrophages, which have been classified as either
classically activated (pro-inflammatory M1; polarized by
lipopolysaccharide and IFN-
) or alternatively activated
(anti-inflammatory M2; polarized by IL-4, IL-10, IL-13,
and TGF-
) (reviewed in [78], and the references
therein). These phenotypes may apply to microglia as
well; however, this has not been demonstrated in vivo. It
was shown that microglia with a constitutive IL-4-driven
alternatively activated (M2-like) phenotype are actively
involved with suppression of neuroinflammation, as EAE
is very severe in bone marrow chimeric mice that lack
IL-4 cytokine in the CNS compared to control chimeric
mice [79]. Interestingly, M2 macrophages can be re-po-
larized to an M1 phenotype in vivo at the site of injury in
a spinal cord injury model [80], suggesting that factors
within the lesion itself contributed to macrophage phe-
notype polarization with corresponding pro- or anti-in-
flammatory function. Additionally, regional differences
in local microenvironmental factors may influence macro-
phage phenotype within the same lesion (or between le-
sions). This is supported by immunostaining of CNS tis-
sue from MS patients, which showed higher numbers of
macrophage/microglia with a more M2-like phenotype
(characterized by expression of CD163) within acute
active lesions and on the edge of chronic active lesions,
i.e., sites of active inflammation, than in the center of
chronic active lesions [81]. These cells also expressed
MHC class II and stained positive for myelin, indicating
that they had the capacity to present myelin antigen to T
cells and perhaps could induce a regulatory T cell re-
sponse. Collectively, these results suggest that the type of
adaptive response that is promoted by microglia is, in
part, determined by local microenvironmental cues.
New data cast doubt on the role of microglia in
chronic disease progression. Using an elegant parabiosis
technique combined with irradiation, in which blood cir-
culation from one mouse is allowed to naturally enter the
blood circulation of the irradiated partner, Ajami et al.
recently demonstrated that while activated microglia are
present at EAE disease onset, it is the monocytes that are
recruited to the CNS from the blood after this initial
phase that are required for disease progression [82]. In-
stead, at this late stage, microglia may be more involved
with tissue repair or inhibiting further T cell activation. A
recent study examined the CNS expression profiles of
various co-stimulatory signals required for T cell activa-
tion during the different phases of EAE [83]. The authors
found that B7.2 expression on non-ramified cells during
the inductive and peak phases of EAE was largely re-
stricted to areas around blood vessels, whereas ramified
B7.2+ microglia were found in increasing numbers dur-
ing the recovery phase in the perivascular areas and
somewhat in the parenchyma. Additionally, they found
an accumulation of CTLA-4+ cells near the blood vessels
in the recovery phase of EAE. These results suggest that
T cells entering the parenchyma during EAE may be in-
duced to undergo anergy, as there is very little
co-stimulatory B7.2 expression in this area at this time.
At later phases, microglia may induce inhibition in ef-
fector cells entering from the blood vessels through
CTLA-4 signaling, which has been shown to negatively
regulate T cells (reviewed in [84]). Therefore, microglia
might play a dual role in inhibiting lymphocytes and
promoting tolerance during CNS autoimmune disease.
3.2. Astrocytes: Not Antigen-Presenting Cells in
Vivo but Contributors to BBB Integrity
Astrocytes are one of the two types of CNS-resident
macroglia (the other being the oligodendrocytes) and
arise from the neuroectoderm during embryonic devel-
opment. They have many roles—chief among them is
facilitating neuronal synaptic transmission by removing
and recycling excess neurotransmitters from the ex-
tracellular space [85]. Their communication with active
neurons and close connection with blood vessels pene-
trating the CNS parenchyma also allows them to regulate
blood flow by signaling to smooth muscle cells within
the vessel walls [86]. Unlike microglia, astrocytes are not
considered immune cells. However, astrocytic endfeet
form the glia limitans, which is an essential component
of the neurovascular unit (described above) and another
barrier to T cell infiltration. The question of whether as-
trocytes can present antigens to T cells will next be con-
sidered.
It is generally accepted that non-stimulated astrocytes
are very poor APCs and, like microglia, express very low
or no constitutive levels of costimulatory and MHC
molecules. However, activated astrocytes express in-
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS 209
creased levels of MHC class II and B7-1 molecules in
vitro, but not B7-2 and CD40, which remain at baseline
levels [87]. This suggests that they would be weak in-
ducers of T cell activation at least in vitro, if not also in
vivo. Indeed, compared to activated astrocytes, naïve
astrocytes from primary cultures have almost no ability
to induce proliferation or IFN-
production in PLP139-151-
specific CD4+ effector T cell lines [87]. Activated astro-
cytes are only slightly better in their capacity to induce
proliferation of effector CD4+ T cells from T cell lines
than are naïve astrocytes, but cannot induce proliferation
or effector cytokine production (e.g., IL-2, IL-4, IFN-
)
in naïve CD4+ T cells at all [70]. However, activated
astrocytes have been shown to induce low IFN-
produc-
tion by effector T cells [70,87], though they seem to be
even better at inducing IL-4 and IL-10 cytokine produc-
tion, and so they may have a role in forming a Th2-po-
larizing environment [68,70].
While activated astrocytes have some capacity to
process myelin protein and present encephalitogenic
myelin antigens in vitro [88, 89], their pathogenic role in
the CNS autoimmune disease process is dubious at best.
Mice in which astrocytes constitutively express the tran-
scription factor CIITA (required for MHC class II protein
expression in astrocytes) have a similar EAE disease
course to wild type mice, despite these astrocytes having
upregulated message levels of CIITA (the transcription
factor associated with MHC class II expression) and the
components that are involved in the assembly of pep-
tide/MHC class II complexes (invariant chain (Ii) and
H-2M molecules) [90]. Finally, astrocytes have been
shown to present soluble viral peptide antigen to naïve
CD8+ T cells in vitro [91]. While astrocytes engineered
to express viral neoantigen may activate effector CD8+ T
cells in vivo and initiate disease [92], true cross-presen-
tation of viral or myelin antigens by astrocytes in vivo
has never been tested. Given their poor ability to phago-
cytose myelin antigen compared to microglia [93], it
seems unlikely that they have any significant contribu-
tion in the presentation of self-antigen to T cells recruited
to the CNS during neuroinflammation.
Based on these studies, there is no major role for as-
trocytes in inducing adaptive immune responses in the
CNS in vivo. However, undoubtedly, these cells play a
critical role in supporting BBB and neuron functions
(reviewed in [94]).
3.3. Dendritic Cells: Major Players in CNS
Immunity
In the last few years, interest has focused on DCs in the
initiation of CNS autoimmunity, as these cells have
emerged as potential targets for modulating immune dis-
eases of the nervous tissue. DCs are highly specialized,
professional APCs that reside in tissues in an immature
state, where they capture and process antigens. Anti-
gen-bearing DCs mature en route to the peripheral lym-
phoid tissues, where they play a crucial role in T cell
activation and differentiation, as well as tolerization.
They are the most efficient of all the professional APCs
at priming naïve T cells, given their ability to migrate
and rapidly upregulate the necessary costimulatory and
MHC molecules. While this knowledge comes from
studying DCs in peripheral, non-CNS tissues, until re-
cently little was known about their immunosurveillant
role in the CNS. In one of the first studies to characterize
the presence of DCs in various non-lymphoid tissues, it
was suggested that their absence in the normal rat paren-
chyma contributed to CNS immune privilege [95]. Thus,
cells within the CNS (e.g., microglia and astrocytes)
were studied for their role in autoimmune disease patho-
genesis. But then, almost three decades after their initial
description by Steinman and Cohn [96], DCs were iden-
tified in the perivascular space, choroid plexus, and
meninges in rodents [48,49], where they localized in dif-
ferent aspects of these tissues (i.e., CSF-exposed) than
macrophages [97]. Since then, both plasmacytoid and
myeloid DC subsets have been found in CSF in humans
[98].
The identification of this professional APC in
CSF-exposed parts of the CNS has radically reshaped our
ideas about the immune-privileged nature of the CNS
and the mechanism of CNS autoimmune disease initia-
tion. Upon their discovery, it was proposed that DCs in
these non-parenchymal CNS tissue areas might acquire
antigens obtained from CSF, exit the CNS via the olfac-
tory pathway and nasal lymphatics, and stimulate the
appropriate T cells within the CLNs [19,49]. Experimen-
tal support for the crucial role of DCs in the CNS auto-
immune disease process came in a landmark study, in
which Greter et al. used mice that had DC-restricted ex-
pression of MHC class II to show that DCs within the
meninges and CNS blood vessels, but not parenchymal
MHC class II+ cells (i.e., microglia or astrocytes), were
necessary and sufficient to induce EAE [99]. DCs have
also been shown to regulate the process of epitope
spreading in the CNS, in which naïve T cells are primed
against antigens that are different than the one used to
induce disease [71]. This process is thought to underlie
the relapses that patients suffer in relapsing-remitting MS.
In this study, DCs isolated from the inflamed CNS of
mice with R-EAE (induced with PLP178-191) were able to
cross-present non-immunizing epitopes that they had
picked up in vivo to prime naïve PLP139-151-specific T
cells ex vivo [71]. Additionally, studies in our lab have
demonstrated that DCs can promote T-T cell interactions,
which facilitates their entry into the CNS [100]. Thus, the
evidence is growing that DCs are critical regulators of
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
210
adaptive immune responses in the CNS. Special consid-
eration of the role of DCs in tolerance and immunity to
CNS self-antigens will be given later in this review.
The discovery of DCs in the CSF-exposed parts of the
healthy CNS suggested that these cells might be impor-
tant in the regulation of adaptive immune responses
within the CNS. The CNS was now viewed as being
relatively, rather than absolutely, immune privileged. In
terms of disease mechanisms, the potential for develop-
ment of CNS autoimmunity arises should the DCs be
presenting neural self-antigens, yet it would be simplistic
to think that this would be the inevitable outcome. There
exist many central and peripheral tolerance mechanisms
and cellular regulators of immunity (for example, induc-
tion of T cell anergy; clonal deletion; induction of/regu-
lation by regulatory T cells, myeloid-derived suppressor
cells, and alternatively activated macrophages) that pre-
vent self-reactive T cells from becoming activated
[101-103]. The conditions in which antigens draining
from the CNS induce and/or break peripheral tolerance
are unknown but are essential to understanding how pe-
ripherally located CNS antigen-specific T cells become
pathogenic and/or contribute to autoimmune disease.
A remaining unanswered question is whether CNS an-
tigen-specific naïve T cells are primed by DCs in the
CLNs, or whether they are primed by DCs in the SAS.
McMahon and colleagues have shown that while initial
priming of naïve CD4+ T cells occurs in the periphery as
a result of the R-EAE immunization protocol, those spe-
cific for non-immunizing epitope that have newly arrived
in the CNS are primed locally by DCs [71]. However,
Walter and Albert reported that cross-presentation of
membrane-bound antigens on splenocytes lacking MHC
class I first occurs in the CLNs before responder T cell
recruitment to the CNS [104]. Collectively, these results
suggest that antigen drainage to the CLNs is one of the
first steps regulating activated T cell recruitment to the
CNS and, thus, is a key component of CNS surveillance,
but that local stimulation in the CNS may be crucial to
disease progression. For example, DCs in the SAS may
contribute both to priming of Th17 cells and to the for-
mation of eLFs by producing CXCL13 [15,17], which
binds to CXCR5 expressed on Th17 cells that may as-
sume follicular helper T cell-like functions.
Finally, there has been some recent investigation into
the origin of CNS DCs that are present under neuroin-
flammatory conditions, namely, whether they originate
from a precursor in the CNS or enter from the blood.
Microglia, for instance, can take on a DC-like phenotype
when exposed to GM-CSF, or a more macrophage-like
phenotype when exposed to M-CSF (macrophage col-
ony-stimulating factor) [105]. Mice with CD11c pro-
moter-driven expression of GFP [106] and EYFP [107]
have been used to probe the presence and distribution of
DCs in the normal CNS parenchyma [108,109], where
the reporter cells seem to have definite DC morphology
and functionality. Bulloch and colleagues identified stel-
late EYFP+ cells in areas of the normal brain lacking a
BBB (e.g., circumventricular organs), which they pro-
posed was perhaps indicative of the role of these cells in
immunosurveillance and antigen presentation [108]. In
the other model, the GFP+ cells were also identified in
several parenchymal areas, but their dendritic processes
were found to directly connect with the glia limitans,
which, again, may be related to their role in antigen
presentation in the perivascular space [109]. Functionally,
IFN-
-activated EYFP+ cells were able to migrate and
upregulate MHC class II in vivo, and were better at
stimulating naïve CD4+ T cells than EYFP- microglia ex
vivo, consistent with established DC properties [110].
However, it must be noted that CD11c expression is not
restricted to classical myeloid DCs, and, thus, alone it
may not reliably be used to determine lineage [111]. The
data from the reporter studies show that parenchymal
GFP+ and EYFP+ reporter cells colocalize with micro-
glia/macrophage markers F4/80, and Iba-1 and myeloid
marker CD11b, suggesting that parenchymal CD11c+
cells may be a subset of resident microglia [108,109].
Using the CD11c-EYFP reporter mice, it has recently
been shown that EYFP+ cells only in the meninges and
choroid plexus (but not in the parenchyma) of the healthy
CNS expand in response to FMS-like tyrosine kinase
receptor 3 ligand (Flt3L), which is necessary for DC
lineage commitment [50]. Additionally, these DCs re-
sembled splenic DCs in terms of their mRNA profile of
several cell surface/lineage markers and transcription
factors, as well as in their ability to stimulate naïve T
cells, and were thus distinctly different from parenchy-
mal microglia. These new findings support that pre-DCs
in the meninges and choroid plexus enter from the blood
and differentiate into mature DCs in situ. Against the
idea that DCs found in the inflamed CNS are of micro-
glia origin, bone marrow chimeric mice have previously
been used to show that the majority (i.e., more than
eighty percent) of DCs present in the CNS of mice with
R-EAE are from the bone marrow [112].
In summary, DCs in the perivascular space and men-
inges are the choice cellular candidate responsible for
priming naïve and restimulating CNS antigen-specific T
cells. Once inside the parenchyma proper, activated T
cells can then interact with CNS-resident astrocytes and
microglia. Microglia may present antigen to infiltrating
effector T cells and are perhaps more involved with
negative regulation of disease.
4. Initiation of CNS Autoimmunity: The
Perfect Immunological “Storm”
Many of our ideas about the pathogenic mechanisms
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
Copyright © 2012 SciRes. NM
211
underlying chronic neuroinflammatory diseases, such as
MS, come from studying the EAE animal model. In this
model, disease may be initiated by either immunizing the
animal with a myelin antigen (“active” induction), or by
adoptively transferring activated, encephalitogenic T
cells (“passive” induction). Under both experimental
conditions, activated T cells from the periphery infiltrate
the healthy CNS and contribute to disease. Yet in hu-
mans with MS, it is difficult to determine what drives the
entry of myelin antigen-specific T cells into the CNS.
Because of the preponderance of these and macrophage
cells in active demyelinating lesions and elevated levels
of chemokines and proinflammatory Th1 cytokines in
CSF, MS is widely accepted to be a T cell-mediated
autoimmune disease [113-115]. Thus, the standard model
is that myelin-reactive T cells initiate oligodendrocyte
death and mediate further myelin destruction by solicit-
ing the recruitment of macrophages. However, given the
different histopathological patterns displayed in active
MS lesions [116], the question arises whether MS is pri-
marily an autoimmune disease, or whether adaptive im-
munity is secondary to a different underlying cause. This
is tremendously important in terms of how the disease is
treated and is a subject of intense debate.
tion, pathogens display conserved molecules (i.e., pat-
tern-associated molecular patterns, PAMPs), which can
bind to pattern recognition receptors, such as Toll-like
receptors, on APCs and induce their activation, thus fa-
cilitating efficient presentation of the pathogenic/self
antigen. Mice infected with a neurotropic virus engi-
neered to express a myelin-self-antigen develop paralytic
demyelinating disease induced by cross-reactive CD4+ T
cells [117]. In a similar study, CD8+ T cell-mediated
attack of ovalbumin (OVA)-expressing neurons was ini-
tiated following intracerebral injection with OVA-secret-
ing Listeria monocytogenes [30]. It is important to note
that in this experiment, peripheral infection alone did not
induce disease, suggesting that CNS inflammation and
breakdown of the BBB are also required. While no spe-
cific virus has been causally linked to human MS, deep
sequencing technology is now being used to detect new
viruses from MS brain samples and will likely yield
strong correlational data [118].
Another model of CNS autoimmune disease initiation
has been proposed based on the histological observation
that oligodendrocyte (ODC) apoptosis precedes immune
cell infiltration in new lesions in several cases of relaps-
ing-remitting MS [119]. The idea is that some unknown
factor that causes ODC death also results in the direct or
indirect activation of CNS APCs, leading to their migra-
tion to CLNs and the priming of naïve T cells, which
then get recruited to the CNS (Figure 2) [120]. The
power of modeling primary ODC death as an initiator of
Infection is one of several potential triggers of CNS-
targeted adaptive immune responses and has received
much attention. An infectious pathogen might drive the
cross-activation of CNS antigen-specific T cells due to
similarities in epitope sequence homology or molecular
conformation (this is called molecular mimicry). In addi-
Figure 2. Model of initiation of CNS autoimmunity. 1) An initiating factor (red arrow) may cause damage/stress to oligoden-
drocytes (ODCs); 2) Antigens or danger signals released upon injury may be carried by interstitial fluid and drain into the
CSF and either enter the blood circulation via arachnoid villi that protrude into the dural sinus or exit along the cranial
nerves (in particular, the olfactory nerve) and reach the cervical lymph nodes (CLNs) via the nasal lymphatics; 3) In the
CLNs, naïve myelin antigen-specific T cells are primed by dendritic cells (DCs) and undergo clonal expansion. Primed T cells
are recruited to areas of CNS inflammation, as BBB endothelial cells now express the appropriate adhesion molecules and
chemokines; 4) Once inside the PVS, the primed T cells are restimulated with their cognate antigen and are able to enter the
CNS parenchyma to carry out their effector functions.
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
212
other ideas of disease initiation. For example, primary
autoimmunity is that it allows for the initial adaptive
ral/bacterial infection) or self-antigens (e.g., from minor
trauma), and therefore connects the standard model with
immune response to be against either non-self (e.g., vi
ODC death (as opposed to T cell-mediated death) itself
may result in autoimmunity to myelin antigens exposed
by death [120]. In seeming conflict with this idea, two
groups have recently and independently shown that
diphtheria toxin-induced ODC death does not elicit adap-
tive immune cell accumulation in the CNS [121,122]. In
these studies, ODCs did not die by apoptosis (the form of
cell death usually associated with MS and EAE) but in-
stead underwent vacuolation-induced death, which is
distinct from classical necrosis and apoptosis [123]. Ad-
ditionally, change in the BBB is thought to be a critical
and obligatory step for disease initiation [124], and in-
deed, contrast enhanced MRI has shown that disruption
of the BBB is one of the earliest events in patients with
MS [125]. However, BBB permeability was not altered
in the transgenic animals used in either study, presuma-
bly due to lack of inflammation, thus precluding T cell
entry. From these studies, we may conclude that vacuola-
tion-induced ODC death per se does not induce autoim-
munity; however, other types of cell death may.
Careful consideration must be given to the type of
ODC death that is induced and whether it has the ability
to initiate inflammation in the CNS (beyond microglia
activation), which determines T cell recruitment and en-
try, as well as APC activation. These factors likely de-
termine whether death induces autoimmunity or toler-
ance (or ignorance). ODC apoptosis, in particular, is im-
portant for EAE initiation, as inhibiting ODC apoptosis
has been shown to attenuate the incidence and severity of
disease [37,126]. (However, as a caveat, it could also
mean that T cell-mediated effects are blocked.) For a
long time apoptosis was considered to be an “immu-
nologically silent” form of cell death, and so precisely
how it could contribute to MS and EAE development
was (and remains) unknown and is rather interesting.
Work done by Matthew Albert and colleagues has dem-
onstrated that DCs can acquire processed antigen from
cells undergoing apoptosis and efficiently cross-present
the antigen to CD8+ T cells [127-129]. In terms of CNS
immunity, Meloni et al. demonstrated that DCs present-
ing antigens from apoptotic ODCs could stimulate IFN-
production and proliferation of MBP-specific T cell lines
[130]. Additionally, DCs have been shown to mediate
myelin epitope spreading in the CNS in vivo [71]; how-
ever, ODC cell death was not examined as a source of
spread (or non-immunizing) antigen in this study. Thus,
whether DCs acquire antigens from apoptotic ODCs and
stimulate myelin-reactive T cells in vivo remains an open
question.
In addition to dying cells being a source of immuno-
genic epitopes, they can potentially release internal
damage-associated molecular patterns (DAMPs) and
alarmins (reviewed in [131]). These signals might, in
turn, activate and mobilize APCs. In this way, ODC
death might be “sensed” by DCs and contribute to CNS
autoimmunity. Several DAMPs were described by Kono
and Rock [131], and here we present their association
with MS/EAE in Table 2. While the DAMPs presented
in Table 2 come from a variety of sources and have a
variety of actions, to the best of our knowledge they are
not released by ODC death. However, their role in di-
recting DC subtype specification and, thus, in polarizing
T cell responses against antigens released by ODC death
are only beginning to be understood. For example, heat
shock protein 70 has been shown to facilitate processing
of MBP in murine fibroblasts made to express HLA-DR
[132]. Future research will determine whether any of
these or (as yet) undiscovered DAMPs are released from
dying ODCs, or are just microenvironmental cues that
contribute to the perfect “immunological storm”.
One question that remains is why “normal” cell death
results in tolerance and not autoimmunity. Recently, it
has been shown that indoleamine 2,3-dioxygenase (IDO)
can be induced in marginal zone macrophages and is
necessary for tolerance to antigens from apoptotic cells
[133]. Importantly, IDO-/- mice injected with apoptotic
thymocytes resulted in increased autoantibody titers in
the serum, as well as lethal autoimmunity due to renal
failure [133]. Additionally, IDO has been shown to be
upregulated in activated microglia from primary cell
cultures [134]. These studies suggest that microglia may
play a role in tolerance to self-antigens exposed by cel-
lular apoptosis via IDO-dependent mechanisms. How-
ever, studies using MHC class I H-2Db-/- bone marrow
chimera mice have demonstrated that DCs are also im-
portant in establishing tolerance to CNS LCMV neo-
antigens [35]. In summary, the whole microenvironment-
tal context of cell death likely determines whether CNS
autoimmunity or tolerance results following injury.
5. Regulation of CNS Immunity: The Role of
Dendritic Cells
One of the greatest challenges in MS research is finding
ways to regulate aberrant immunity within the CNS. DCs
are critical at this juncture and can be matured and im-
printed by environmental cues both in vitro and in vivo to
be functionally either immunogenic (stimulatory) or
tolerogenic, which will be our focus in this section. Be-
cause of their functional plasticity, drugs that alter DC
functionality will enable the therapeutic use of these cells
(reviewed in [147]). There are two major DC subsets: clas-
sical myeloid DCs (mDCs) and plasmacytoid DCs (pDCs)
that arise from a common-DC progenitor (reviewed in
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS 213
Table 2. DAMPs associated with MS/EAE pathology.
DAMP Source or
localization Activity in MS/EAE
Suspected
autoantigen or
cross-reactivity
Receptor(s) Receptor
expression Reference(s)
HMGB1 microglia and
macrophage; CSF unknown -
RAGE; TLR2;
TLR4
active lesions;
blood and CSF [135]
Double-
stranded DNA
intracellular;
anti-dsDNA
antibodies in MS
active plaque and
periplaque regions
and B cells in CSF
anti-dsDNA antibodies
may promote MS lesion
formation
yes N/A N/A [136]
intracellular; both
CNS and systemic
tissue deposits of
cross-reactive
anti-dsDNA
antibodies
Areas of demyelination
observed in the CNS and
renal autoimmune
pathology
cross-reactivity
found with
MOG92-106
N/A N/A [137,138]
Extracellular
nucloeotides
(e.g., ATP,
ADP, UTP,
UDP)
intracellular
COX-2, iNOS, and
pro-inflammatory
cytokine production;
glial cell proliferation and
death
- P2
receptors
neurons, glial
cells, Schwann
cells,
CNS-infiltrating
leukocytes
reviewed in
[139]
Adenosine intracellular anti-inflammatory - P1
receptors
neurons,
microglia,
astrocytes,
leukocytes
reviewed in
[139]
Heat-shock
proteins intracellular
Hsp-CNP peptide can
protect against or
aggravate EAE,
depending on Th1 or Th2
immune response pattern
cross-reactivity
between
mycobacterial
Hsp65 and
CNP153-164
N/A N/A [140]
intracellular; binds to
certain MBP peptides
in vitro
Hsp 70 facilitates
autoantigen processing - N/A N/A [132]
S100 proteins
astrocytes and
subpopulation of
ODCs
unknown - - - [141]
Fibronectin
/Fibrinogen
ECM/plasma;
primarily localized to
microvessel walls
(fibronectin) and
lumen (fibrinogen),
but also on
mononuclear cells and
extracellular deposits
facilitate mononuclear
cell adhesion and
migration, myelin
phagocytosis, and
breakdown of BBB;
fibronectin can inhibit
myelination
- fibronectin
receptor macrophages [142-145]
Hyaluronan
(HA)
ECM component;
secreted by astrocytes
and microglia
LMW HA inhibits OPC
maturation via TLR2 - CD44; TLR2 ODCs; T cells [146-148]
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
214
Continued
Heparan sulfate
(proteoglycan)
ECM component
of basement
membrane
cleavage by heparanase
facilitates immune cell
migration through ECM
-
may act as a
chemokine
reservoir
N/A [149,150]
Laminin- and
collagen-derived
peptides
ECM components
of basement
membrane
cleavage by matrix
metalloproteinases
facilitates immune cell
migration through ECM
-
integrins (e.g.,
VLA-1,
VLA-2,
VLA-6)
lymphocytes
and monocytes [150,151]
Elastin-derived
peptides
component of the
dura mater;
degraded by
macrophage
metalloelastase
unknown - elastin recep-
tor
peripheral blood
lymphocytes [152-154]
Galectins
microglia and
macrophage,
astrocytes,
endothelial cells,
DCs
induction of tolerogenic
DCs; negative
regulation of effector T
cells
-
GM1
ganglioside;
glycoproteins
- [155-160]
[161]). These subsets can be distinguished by differences
in surface marker expression. mDCs are CD11c+ CD11b+.
In mice, pDCs are CD11clow CD11b B220+ Ly6C+,
whereas in humans, pDCs are CD11c CD4+ CD45RA+
IL-3R
+ ILT2+ ILT1 (reviewed in [162]). Additionally,
many more sub-subsets of mDCs have been found in
lymphoid and non-lymphoid tissues [161]. Distinct DC
subsets have been shown to accumulate in the CNS in
response to different environmental stimuli (bacterial,
viral, DAMP), and thereby promote the appropriate
adaptive immune response (reviewed in [163]). As we
will also discuss, pDCs may have a critical role in pro-
moting protection against CNS autoimmunity. It will
only be mentioned here in passing that mechanisms in-
ternal to the DC itself also contribute to immune regula-
tion by keeping the DC in an immature, non-stimulatory
state [164]. This was demonstrated recently, as DCs
lacking nuclear factor-
B1 (NF-
B1) were able to induce
autoimmune diabetes as a result of unchecked production
of TNF-
, which promoted cytotoxic CD8+ T cell pro-
duction of the apoptosis-inducing enzyme granzyme B
[164]. Finally, other cell types are involved in the sup-
pression of CNS autoimmune responses, such as regula-
tory T cells (Tregs) and myeloid-derived suppressor cells
(which are now beginning to be recognized for their con-
tribution to the resolution of CNS autoimmunity [103]),
but they will not be discussed further.
5.1. Environmental Imprinting of Myeloid DCs
Affects CNS Disease Outcome
5.1.1. Stimul a tory Roles for mDCs
DCs accumulate in the CNS when there is inflammation,
and so they are thought to be important for disease de-
velopment and maintenance. We have previously shown
that intracerebrally injected, antigen-loaded mDCs can
migrate to peripheral lymphoid tissues and induce the
homing of responder T cells to the CNS [26,100,165].
We found that intracerebral delivery of MOG35-55-pulsed
DCs lead to an increase in the frequency of activated
MOG35-55-specific (i.e., 2D2) effector T cells in the CNS,
which hastened the onset and increased the severity of
EAE [165]. Importantly, this effect was dependent upon
the functional status of the DCs. Mice intracerebrally
injected with stimulatory (i.e., LPS-stimulated) DCs had
more severe EAE and increased CNS accumulation of
pathogenic Th17 cells, whereas those that received
tolerogenic (i.e., TNF-
-stimulated) DCs had a much
lower disease incidence, as well as delayed onset and
decreased severity; tolerogenic DCs promoted IL-10
production in the periphery and suppressed IL-17 pro-
duction in the CNS. Our work therefore demonstrates
that depending on the functional status of DCs, the dis-
ease outcome can be better or worse. This illustrates the
capacity of DC functional status/phenotype to determine
the resulting adaptive immune response. Similarly, the
disease environment within the CNS also determines the
functional state of DCs. Deshpande et al. showed that
CD11c+ mDCs isolated right after EAE onset were bet-
ter at promoting 2D2 T cell activation and were markedly
more mature (i.e., displaying increased levels of
costimulatory molecules and the lymphoid tissue homing
receptor CCR7) than those isolated right before disease
remission [166]. Interestingly, while the mDCs isolated
early in EAE supported differentiation of both Th1 and
Th17 T cells, they simultaneously also supported Treg
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS 215
suppression of 2D2 activation [166]. Collectively, these
studies show that, depending on how they are imprinted
in vitro or in vivo, mDCs may either facilitate or suppress
ongoing inflammation.
The precise mechanisms by which DCs regulate CNS
disease processes are beginning to be unraveled. While
DCs are known for their unique ability to determine the
lineage commitment, and thus effector function, of naïve
T cells, effector T cells may also participate in perpetu-
ating their own lineage by directing monocytes to differ-
entiate into particular lineage-promoting DC subtypes
[167]. Particular attention has been paid to the differen-
tiation of Th17 cells, which are regarded as one of the
main pathogenic subsets involved in MS and EAE dis-
ease initiation. A recent study showed that Th17 produc-
tion of GM-CSF drove mDC production of IL-23, which,
in turn, had a positive feedback on Th17 lineage com-
mitment [168]. Thus, by assisting with the differentiation
of this pathogenic population, mDCs play a critical role
in stimulating ongoing inflammation and autoimmunity.
It has been shown that the ability of DCs to produce
IL-23 in response to GM-CSF depends on their expres-
sion of CC chemokine receptor 4 (CCR4), which is re-
quired for EAE initiation [169]. Finally, it was also
demonstrated that, mDCs, compared to pDCs and CNS
macrophages, induce Th17 polarization of naïve CD4+ T
cells specific for non-EAE-inducing epitope in the R-
EAE mouse model, thereby facilitating epitope spreading
[112]. Thus, there is strong evidence that mDCs contrib-
ute to CNS autoimmune disease initiation and relapse by
regulating the development of pathogenic Th17 cells.
5.1.2. Tolerogenic Roles for mDCs
There are many soluble factors that can promote the
tolerogenic phenotype of mDCs within the CNS and con-
tribute to the prevention and/or resolution of CNS auto-
immunity, including anti-inflammatory cytokines (e.g.,
IL-10; TGF-
), neuropeptides and hormones (e.g.,
vasoactive intestinal peptide, VIP;
-melanocyte-stimu-
lating hormone,
-MSH), as well as new molecular can-
didates that need to be considered (e.g., galectins).
However, we will only discuss a few examples here. For
a more thorough review of tolerogenic DCs, see [170].
Anti-inflammatory cytokines are well known for their
ability to promote tolerogenic immune responses by
competing effectively with pro-inflammatory cytokines
in an inflammatory setting; generally, they are thought to
activate Th2 and Treg cells and/or suppress Th1 cells and
inhibit pro-inflammatory cytokine synthesis (reviewed in
[171]). However, as recent evidence indicates, anti-in-
flammatory cytokines may also contribute to tolerance in
normal settings by preventing self-reactive T cell active-
tion [172]. Laouar et al. sought a mechanistic explana-
tion for the protective effect of TGF-
against EAE. Us-
ing a specialized set of bone marrow chimeric mice, they
were able to demonstrate that inhibiting TGF-
-mediated
signaling specifically in DCs could promote the devel-
opment of spontaneous and severe EAE that was accom-
panied by general features of inflammation (e.g., micro-
glia activation and increased levels of mRNA for pro-
inflammatory cytokines), as well as CD4+ T cell accu-
mulation in the CNS [172]. Interestingly, the majority of
MHC class II+ cells observed in the spinal cord were
CD45low, indicating that they were microglia. This sug-
gests that DCs exert their tolerogenic influence outside
the CNS. These results indicate that intact TGF-
R sig-
naling in DCs promotes suppression of autoimmunity,
perhaps by keeping resting DCs in an immature state.
Apart from cytokines, VIP, as mentioned above, is a
neuropeptide that induces tolerogenic function in mDCs
[173]. Lentiviral vectors were recently used to engineer
DCs to express VIP [174]. In this study, mice that re-
ceived a single injection of VIP-expressing DCs were
protected against disease development in both relapsing
and primary progressive models of EAE. Interestingly,
the VIP-expressing DCs were found to accumulate in
non-lymphoid peripheral tissues (e.g., liver and lung) in
higher numbers than in lymphoid tissues, a migration
pattern somewhat unexpected in the acute phase of EAE.
It would be interesting to see whether the anti-inflam-
matory cytokine profile observed in total RNA isolated
from the spinal cord at the peak of disease was due to
VIP-expressing DCs exerting their tolerogenic effects
systemically or within the CNS. However, the clear sup-
pression of EAE observed in two disease models illus-
trates that DCs may be used to deliver anti-inflammatory
agents for use in the treatment of CNS inflammatory
diseases.
As a final example, another soluble factor that works
as an anti-inflammatory cytokine in the traditional sense
is galectin-1 (Gal1), a glycoprotein that is a member of a
family of lectins. It promotes a tolerogenic phenotype in
DCs during their differentiation process. DCs differenti-
ated in presence of Gal1 or that express Gal1 endoge-
nously were shown to suppress the chronic phase of
MOG35-55-induced EAE, inhibit T cell proliferation and
pro-inflammatory cytokine production, and increase
IL-10 production via an IL-27-dependent pathway [102].
They also found that Gal1 levels were the highest at peak
and chronic phases of EAE. Additionally, immature, but
not mature, mDCs produced high levels of Gal1. When
subsequently cultured with tolerogenic stimuli (such as
VIP, IL-10, vitamin D3, and also apoptotic cells) these
immature mDCs significantly increased Gal1 expression,
whereas pro-inflammatory stimuli had the opposite effect.
This study is interesting because it shows that as the in-
flammatory milieu changes within the CNS, the matura-
tion status of DCs may render them more or less suscep-
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
216
tible to tolerogenic environmental factors, thereby influ-
encing their contribution to disease resolution.
5.2. Plasmacytoid DCs and Their Contribution
to Regulation of CNS Autoimmunity
While pDCs from the inflamed CNS are not as efficient
as mDCs at priming naïve or effector PLP peptide-spe-
cific T cells [175], they seem to play a crucial role in
negatively regulating EAE [175-177]. However, the
mechanisms by which they do this are only beginning to
be elucidated. In order to directly address the question of
whether protection against severe EAE was the result of
T cell priming by pDCs, Irla et al. used mutant chimeric
mice lacking MHC class II only in pDCs to show that
pDCs facilitated Treg priming and expansion in the
draining lymph nodes of mice with EAE in an anti-
gen-specific fashion [177]. A different mechanism was
proposed by Bailey-Bucktrout et al. [176], who found
that pDCs exerted their immunoregulatory effects by
suppressing mDC-induced CD4+ T cell production of
IFN-
, IL-17, and IL-10 cytokines, and not through
IDO-mediated inhibition of T cells. However, they did
observe that blocking IDO resulted in slightly increased
levels of IFN-
and IL-17 when CD4+ T cells were
stimulated with mDCs. This could be due to the inhibi-
tion of the natural Treg subpopulation [178], and would
also support that CNS mDCs might promote Treg prolif-
eration in the CNS, which was not found to be impaired
in the CNS of mutant chimeric mice in the Irla et al.
study [177].
6. Conclusion
The CNS is a precious tissue in which there must be a
fine balance between immune surveillance (i.e., immune
cell admittance) and privilege (i.e., immune cell exclu-
sion). We have discussed that while routine surveillance
of the subarachnoid space by T cells does occur in the
healthy CNS, increased surveillance results in autoim-
munity. While EAE initiation has been shown to be de-
pendent on DCs, it is still unknown where initial T cell
priming occurs. Based on our discussion, disease initia-
tion likely involves antigen drainage and priming of na-
ïve T cells in the CLNs, but disease progression likely
involves local restimulation by DCs and (to a lesser ex-
tent) perivascular macrophages within the PVS and ec-
topic lymphoid structures in the SAS. This is an ongoing
field of investigation. Several theories of CNS autoim-
mune disease initiation in humans have been presented,
including whether primary death of myelin-producing
cells in the CNS stimulates autoimmunity, which remains
controversial. Whether cell death induces immunity, tol-
erance, or ignorance is determined by the entire micro-
environmental context, which induces different func-
tional phenotypes of DCs. Therefore, DCs are probably
master regulators of the autoimmune disease process in
CNS.
7. Acknowledgements
The authors would like to thank Benjamin D. Clarkson
for critically reviewing this manuscript. This work was
funded by NIH/NIGMS grant T32-GM007507 (Neuro-
science Training Program) and NIH grant R01-NS37570
(Z. Fabry).
REFERENCES
[1] R. R. Caspi, “Immunotherapy of Autoimmunity and Can-
cer: The Penalty for Success,” Nature Reviews Immunol-
ogy, Vol. 8, No. 12, 2008, pp. 970-976.
doi:10.1038/nri2438
[2] O. Stuve, C. M. Marra, K. R. Jerome, L. Cook, P. D.
Cravens, S. Cepok, E. M. Frohman, J. T. Phillips, G. Ar-
endt, B. Hemmer, N. L. Monson and M. K. Racke, “Im-
mune Surveillance in Multiple Sclerosis Patients Treated
with Natalizumab,” Annals of Neurology, Vol. 59, No. 5,
2006, pp. 743-747. doi:10.1002/ana.20858
[3] I. Galea, I. Bechmann and V. H. Perry, “What Is Immune
Privilege (Not)?” Trends in Immunology, Vol. 28, No. 1,
2007, pp. 12-18. doi:10.1016/j.it.2006.11.004
[4] B. Engelhardt and C. Coisne, “Fluids and Barriers of the
CNS Establish Immune Privilege by Confining Immune
Surveillance to a Two-Walled Castle Moat Surrounding
the CNS Castle,” Fluids and Barriers of the CNS, Vol. 8,
No. 1, 2011, p. 4. doi:10.1186/2045-8118-8-4
[5] B. T. Hawkins and T. P. Davis, “The Blood-Brain Bar-
rier/Neurovascular Unit in Health and Disease,” Phar-
macological Reviews, Vol. 57, No. 2, 2005, pp. 173-185.
doi:10.1124/pr.57.2.4
[6] P. Kivisakk, D. J. Mahad, M. K. Callahan, C. Trebst, B.
Tucky, T. Wei, L. Wu, E. S. Baekkevold, H. Lassmann, S.
M. Staugaitis, J. J. Campbell and R. M. Ransohoff, “Hu-
man Cerebrospinal Fluid Central Memory CD4+ T Cells:
Evidence for Trafficking through Choroid Plexus and
Meninges via P-Selectin,” Proceedings of the National
Academy of Sciences of the United States of America, Vol.
100, No. 14, 2003, pp. 8389-8394.
doi:10.1073/pnas.1433000100
[7] J. Goverman, “Autoimmune T Cell Responses in the Cen-
tral Nervous System,” Nature Reviews Immunology, Vol.
9, No. 6, 2009, pp. 393-407. doi:10.1038/nri2550
[8] P. Kivisakk, J. Imitola, S. Rasmussen, W. Elyaman, B.
Zhu, R. M. Ransohoff and S. J. Khoury, “Localizing Cen-
tral Nervous System Immune Surveillance: Meningeal
Antigen-Presenting Cells Activate T Cells during Experi-
mental Autoimmune Encephalomyelitis,” Annals of Neu-
rology, Vol. 65, No. 4, 2009, pp. 457-469.
doi:10.1002/ana.21379
[9] B. K. Kleinschmidt-DeMasters and K. L. Tyler, “Pro-
gressive Multifocal Leukoencephalopathy Complicating
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS 217
treatment with Natalizumab and Interferon Beta-1a for
Multiple Sclerosis,” The New England Journal of Medi-
cine, Vol. 353, No. 4, 2005, pp. 369-374.
doi:10.1056/NEJMoa051782
[10] A. Langer-Gould, S. W. Atlas, A. J. Green, A. W. Bollen
and D. Pelletier, “Progressive Multifocal Leukoencepha-
lopathy in a Patient Treated with Natalizumab,” The New
England Journal of Medicine, Vol. 353, No. 4, 2005, pp.
375-381. doi:10.1056/NEJMoa051847
[11] G. Van Assche, M. Van Ranst, R. Sciot, B. Dubois, S.
Vermeire, M. Noman, J. Verbeeck, K. Geboes, W. Rob-
berecht and P. Rutgeerts, “Progressive Multifocal Leu-
koencephalopathy after Natalizumab Therapy for Crohn’s
Disease,” The New England Journal of Medicine, Vol.
353, No. 4, 2005, pp. 362-368.
doi:10.1056/NEJMoa051586
[12] A. Reboldi, C. Coisne, D. Baumjohann, F. Benvenuto, D.
Bottinelli, S. Lira, A. Uccelli, A. Lanzavecchia, B. Eng-
elhardt and F. Sallusto, “C-C Chemokine Receptor 6-
Regulated Entry of TH-17 Cells into the CNS through the
Choroid Plexus Is Required for the Initiation of EAE,”
Nature Immunology, Vol. 10, No. 5, 2009, pp. 514-523.
doi:10.1038/ni.1716
[13] C. L. Langrish, Y. Chen, W. M. Blumenschein, J.
Mattson, B. Basham, J. D. Sedgwick, T. McClanahan, R.
A. Kastelein and D. J. Cua, “IL-23 Drives a Pathogenic T
Cell Population That Induces Autoimmune Inflamma-
tion,” The Journal of Experimental Medicine, Vol. 201,
No. 2, 2005, pp. 233-240. doi:10.1084/jem.20041257
[14] A. Peters, L. A. Pitcher, J. M. Sullivan, M. Mitsdoerffer,
S. E. Acton, B. Franz, K. Wucherpfennig, S. Turley, M. C.
Carroll, R. A. Sobel, E. Bettelli and V. K. Kuchroo,
“Th17 Cells Induce Ectopic Lymphoid Follicles in Cen-
tral Nervous System Tissue Inflammation,” Immunity,
Vol. 35, No. 6, 2011, pp. 986-996.
doi:10.1016/j.immuni.2011.10.015
[15] R. Magliozzi, S. Columba-Cabezas, B. Serafini and F.
Aloisi, “Intracerebral Expression of CXCL13 and BAFF
Is Accompanied by Formation of Lymphoid Follicle-Like
Structures in the Meninges of Mice with Relapsing Ex-
perimental Autoimmune Encephalomyelitis,” Journal of
Neuroimmunology, Vol. 148, No. 1-2, 2004, pp. 11-23.
doi:10.1016/j.jneuroim.2003.10.056
[16] R. Magliozzi, O. Howell, A. Vora, B. Serafini, R. Nicho-
las, M. Puopolo, R. Reynolds and F. Aloisi, “Meningeal
B-Cell Follicles in Secondary Progressive Multiple Scle-
rosis Associate with Early Onset of Disease and Severe
Cortical Pathology,” Brain, Vol. 130, No. 4, 2007, pp.
1089-1104.
[17] B. Serafini, B. Rosicarelli, R. Magliozzi, E. Stigliano and
F. Aloisi, “Detection of Ectopic B-Cell Follicles with
Germinal Centers in the Meninges of Patients with Sec-
ondary Progressive Multiple Sclerosis,” Brain Pathology,
Vol. 14, No. 2, 2004, pp. 164-174.
doi:10.1111/j.1750-3639.2004.tb00049.x
[18] A. P. Byrnes, J. E. Rusby, M. J. Wood and H. M. Charl-
ton, “Adenovirus Gene Transfer Causes Inflammation in
the Brain,” Neuroscience, Vol. 66, No. 4, 1995, pp. 1015-
1024. doi:10.1016/0306-4522(95)00068-T
[19] M. K. Matyszak, “Inflammation in the CNS: Balance
between Immunological Privilege and Immune Re-
sponses,” Progress in Neurobiology, Vol. 56, No. 1, 1998,
pp. 19-35. doi:10.1016/S0301-0082(98)00014-8
[20] M. K. Matyszak and V. H. Perry, “Demyelination in the
Central Nervous System Following a Delayed-Type Hy-
persensitivity Response to Bacillus Calmette-Guerin,”
Neuroscience, Vol. 64, No. 4, 1995, pp. 967-977.
doi:10.1016/0306-4522(94)00448-E
[21] M. K. Matyszak, M. J. Townsend and V. H. Perry, “Ul-
trastructural Studies of an Immune-Mediated Inflamma-
tory Response in the CNS Parenchyma Directed against a
Non-CNS Antigen,” Neuroscience, Vol. 78, No. 2, 1997,
pp. 549-560. doi:10.1016/S0306-4522(96)00578-7
[22] H. F. Cserr and P. M. Knopf, “Cervical Lymphatics, the
Blood-Brain Barrier and the Immunoreactivity of the
Brain: A New View,” Immunology Today, Vol. 13, No.
12, 1992, pp. 507-512.
doi:10.1016/0167-5699(92)90027-5
[23] S. Yamada, M. DePasquale, C. S. Patlak and H. F. Cserr,
“Albumin Outflow into Deep Cervical Lymph from Dif-
ferent Regions of Rabbit Brain,” American Journal of
Physiology, Vol. 261, No. 4, 1991, pp. H1197-H1204.
[24] L. B. Gordon, P. M. Knopf and H. F. Cserr, “Ovalbumin
Is More Immunogenic When Introduced into Brain or
Cerebrospinal Fluid than Into Extracerebral Sites,” Jour-
nal of Neuroimmunology, Vol. 40, No. 1, 1992, pp. 81-87.
doi:10.1016/0165-5728(92)90215-7
[25] C. Harling-Berg, P. M. Knopf, J. Merriam and H. F. Cserr,
“Role of Cervical Lymph Nodes in the Systemic Humoral
Immune Response to Human Serum Albumin Microin-
fused into Rat Cerebrospinal Fluid,” Journal of Neuro-
immunology, Vol. 25, No. 2-3, 1989, pp. 185-193.
doi:10.1016/0165-5728(89)90136-7
[26] J. Karman, C. Ling, M. Sandor and Z. Fabry, “Initiation
of Immune Responses in Brain Is Promoted by Local
Dendritic Cells,” The Journal of Immunology, Vol. 173,
No. 4, 2004, pp. 2353-2361.
[27] C. Ling, M. Sandor and Z. Fabry, “In Situ Processing and
Distribution of Intracerebrally Injected OVA in the CNS,”
Journal of Neuroimmunology, Vol. 141, No. 1-2, 2003,
pp. 90-98. doi:10.1016/S0165-5728(03)00249-2
[28] C. Ling, M. Sandor, M. Suresh and Z. Fabry, “Traumatic
Injury and the Presence of Antigen Differentially Con-
tribute to T-Cell Recruitment in the CNS,” The Journal of
Neuroscience, Vol. 26, No. 3, 2006, pp. 731-741.
doi:10.1523/JNEUROSCI.3502-05.2006
[29] Z. Qing, D. Sewell, M. Sandor and Z. Fabry, “Antigen-
Specific T Cell Trafficking into the Central Nervous Sys-
tem,” Journal of Neuroimmunology, Vol. 105, No. 2,
2000, pp. 169-178. doi:10.1016/S0165-5728(99)00265-9
[30] M. Sanchez-Ruiz, L. Wilden, W. Muller, W. Stenzel, A.
Brunn, H. Miletic, D. Schluter and M. Deckert, “Molecu-
lar Mimicry between Neurons and an Intracerebral Patho-
gen Induces a CD8 T Cell-Mediated Autoimmune Dis-
ease,” The Journal of Immunology, Vol. 180, No. 12,
2008, pp. 8421-8433.
[31] T. Scheikl, B. Pignolet, C. Dalard, S. Desbois, D. Raison,
M. Yamazaki, A. Saoudi, J. Bauer, H. Lassmann, H. Har-
din-Pouzet and R. S. Liblau, “Cutting Edge: Neuronal
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
218
Recognition by CD8 T Cells Elicits Central Diabetes In-
sipidus,” The Journal of Immunology, Vol. 188, No. 10,
2012, pp. 4731-4735. doi:10.4049/jimmunol.1102998
[32] A. Cornet, T. C. Savidge, J. Cabarrocas, W. L. Deng, J. F.
Colombel, H. Lassmann, P. Desreumaux and R. S. Liblau,
“Enterocolitis Induced by Autoimmune Targeting of En-
teric Glial Cells: A Possible Mechanism in Crohn’s Dis-
ease?” Proceedings of the National Academy of Sciences
of the United States of America, Vol. 98, No. 23, 2001, pp.
13306-13311. doi:10.1073/pnas.231474098
[33] Y. Cao, C. Toben, S. Y. Na, K. Stark, L. Nitschke, A.
Peterson, R. Gold, A. Schimpl and T. Hunig, “Induction
of Experimental Autoimmune Encephalomyelitis in
Transgenic Mice Expressing Ovalbumin in Oligodendro-
cytes,” European Journal of Immunology, Vol. 36, No. 1,
2006, pp. 207-215. doi:10.1002/eji.200535211
[34] A. Saxena, J. Bauer, T. Scheikl, J. Zappulla, M. Audebert,
S. Desbois, A. Waisman, H. Lassmann, R. S. Liblau and
L. T. Mars, “Cutting Edge: Multiple Sclerosis-Like Le-
sions Induced by Effector CD8 T Cells Recognizing a
Sequestered Antigen on Oligodendrocytes,” The Journal
of Immunology, Vol. 181, No. 3, 2008, pp. 1617-1621.
[35] A. Schildknecht, H. C. Probst, K. D. McCoy, I. Miescher,
C. Brenner, D. P. Leone, U. Suter, P. S. Ohashi and M.
van den Broek, “Antigens Expressed by Myelinating Glia
Cells Induce Peripheral Cross-Tolerance of Endogenous
CD8+ T Cells,” European Journal of Immunology, Vol.
39, No. 6, 2009, pp. 1505-1515.
doi:10.1002/eji.200839019
[36] J. Jung and M. Michalak, “Cell Surface Targeting of
Myelin Oligodendrocyte Glycoprotein (MOG) in the Ab-
sence of Endoplasmic Reticulum Molecular Chaperones,”
Biochimica et Biophysica Acta, Vol. 1813, No. 5, 2011,
pp. 1105-1110. doi:10.1016/j.bbamcr.2010.12.014
[37] N. Hovelmeyer, Z. Hao, K. Kranidioti, G. Kassiotis, T.
Buch, F. Frommer, L. von Hoch, D. Kramer, L. Mini-
chiello, G. Kollias, H. Lassmann and A. Waisman,
“Apoptosis of Oligodendrocytes via Fas and TNF-R1 Is a
Key Event in the Induction of Experimental Autoimmune
Encephalomyelitis,” The Journal of Immunology, Vol.
175, No. 9, 2005, pp. 5875-5884.
[38] E. Barbarese, C. Barry, C. H. Chou, D. J. Goldstein, G. A.
Nakos, R. Hyde-DeRuyscher, K. Scheld and J. H. Carson,
“Expression and Localization of Myelin Basic Protein in
Oligodendrocytes and Transfected Fibroblasts,” Journal
of Neurochemistry, Vol. 51, No. 6, 1988, pp. 1737-1745.
doi:10.1111/j.1471-4159.1988.tb01153.x
[39] S. Y. Na, Y. Cao, C. Toben, L. Nitschke, C. Stadelmann,
R. Gold, A. Schimpl and T. Hunig, “Naive CD8 T-Cells
Initiate Spontaneous Autoimmunity to a Sequestered
Model Antigen of the Central Nervous System,” Brain,
Vol. 131, No. 9, 2008, pp. 2353-2365.
[40] V. S. Schwob, H. B. Clark, D. Agrawal and H. C.
Agrawal, “Electron Microscopic Immunocytochemical
Localization of Myelin Proteolipid Protein and Myelin
Basic Protein to Oligodendrocytes in Rat Brain during
Myelination,” Journal of Neurochemistry, Vol. 45, No. 2,
1985, pp. 559-571.
doi:10.1111/j.1471-4159.1985.tb04024.x
[41] D. P. Leone, S. Genoud, S. Atanasoski, R. Grausenburger,
P. Berger, D. Metzger, W. B. Macklin, P. Chambon and
U. Suter, “Tamoxifen-Inducible Glia-Specific Cre Mice
for Somatic Mutagenesis in Oligodendrocytes and Schwann
Cells,” Molecular and Cellular Neuroscience, Vol. 22,
No. 4, 2003, pp. 430-440.
doi:10.1016/S1044-7431(03)00029-0
[42] M. Jeffrey, G. A. Wells and A. W. Bridges, “An Immu-
nohistochemical Study of the Topography and Cellular
Localization of Three Neural Proteins in the Sheep Nerv-
ous System,” Journal of Comparative Pathology, Vol.
103, No. 1, 1990, pp. 23-35.
doi:10.1016/S0021-9975(08)80132-6
[43] P. J. Marangos and D. E. Schmechel, “Neuron Specific
Enolase, a Clinically Useful Marker for Neurons and
Neuroendocrine Cells,” Annual Review of Neuroscience,
Vol. 10, 1987, pp. 269-295.
doi:10.1146/annurev.ne.10.030187.001413
[44] K. Shen, M. N. Teruel, K. Subramanian and T. Meyer,
“CaMKIIbeta Functions as an F-Actin Targeting Module
That Localizes CaMKIIalpha/Beta Heterooligomers to
Dendritic Spines,” Neuron, Vol. 21, No. 3, 1998, pp.
593-606. doi:10.1016/S0896-6273(00)80569-3
[45] E. Casanova, S. Fehsenfeld, T. Mantamadiotis, T. Lem-
berger, E. Greiner, A. F. Stewart and G. Schutz, “A
CamKIIalpha iCre BAC Allows Brain-Specific Gene In-
activation,” Genesis, Vol. 31, No. 1, 2001, pp. 37-42.
doi:10.1002/gene.1078
[46] A. J. Slavin, J. M. Soos, O. Stuve, J. C. Patarroyo, H. L.
Weiner, A. Fontana, E. K. Bikoff and S. S. Zamvil, “Re-
quirement for Endocytic Antigen Processing and Influ-
ence of Invariant Chain and H-2M Deficiencies in CNS
Autoimmunity,” The Journal of Clinical Investigation,
Vol. 108, No. 8, 2001, pp. 1133-1139.
[47] M. L. Krakowski and T. Owens, “Naive T Lymphocytes
Traffic to Inflamed Central Nervous System, but Require
Antigen Recognition for Activation,” European Journal
of Immunology, Vol. 30, No. 4, 2000, pp. 1002-1009.
doi:10.1002/(SICI)1521-4141(200004)30:4<1002::AID-I
MMU1002>3.0.CO;2-2
[48] M. K. Matyszak and V. H. Perry, “The Potential Role of
Dendritic Cells in Immune-Mediated Inflammatory Dis-
eases in the Central Nervous System,” Neuroscience, Vol.
74, No. 2, 1996, pp. 599-608.
doi:10.1016/0306-4522(96)00160-1
[49] P. G. McMenamin, “Distribution and Phenotype of Den-
dritic Cells and Resident Tissue Macrophages in the Dura
Mater, Leptomeninges, and Choroid Plexus of the Rat
Brain as Demonstrated in Whole Mount Preparations,”
The Journal of Comparative Neurology, Vol. 405, No. 4,
1999, pp. 553-562.
doi:10.1002/(SICI)1096-9861(19990322)405:4<553::AID
-CNE8>3.0.CO;2-6
[50] N. Anandasabapathy, G. D. Victora, M. Meredith, R.
Feder, B. Dong, C. Kluger, K. Yao, M. L. Dustin, M. C.
Nussenzweig, R. M. Steinman and K. Liu, “Flt3L Con-
trols the Development of Radiosensitive Dendritic Cells
in the Meninges and Choroid Plexus of the Steady-State
Mouse Brain,” The Journal of Experimental Medicine,
Vol. 208, No. 8, 2011, pp. 1695-1705.
doi:10.1084/jem.20102657
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS 219
[51] H. G. Fischer, U. Bonifas and G. Reichmann, “Phenotype
and Functions of Brain Dendritic Cells Emerging during
Chronic Infection of Mice with Toxoplasma Gondii,” The
Journal of Immunology, Vol. 164, No. 9, 2000, pp. 4826-
4834.
[52] B. Serafini, S. Columba-Cabezas, F. Di Rosa and F.
Aloisi, “Intracerebral Recruitment and Maturation of
Dendritic Cells in the Onset and Progression of Experi-
mental Autoimmune Encephalomyelitis,” The American
Journal of Pathology, Vol. 157, No. 6, 2000, pp. 1991-
2002. doi:10.1016/S0002-9440(10)64838-9
[53] B. Serafini, B. Rosicarelli, R. Magliozzi, E. Stigliano, E.
Capello, G. L. Mancardi and F. Aloisi, “Dendritic Cells in
Multiple Sclerosis Lesions: Maturation Stage, Myelin
Uptake, and Interaction with Proliferating T Cells,” Jour-
nal of Neuropathology & Experimental Neurology, Vol.
65, No. 2, 2006, pp. 124-141.
doi:10.1097/01.jnen.0000199572.96472.1c
[54] M. A. Friese and L. Fugger, “Pathogenic CD8(+) T Cells
in Multiple Sclerosis,” Annals of Neurology, Vol. 66, No.
2, 2009, pp. 132-141. doi:10.1002/ana.21744
[55] S. Bourdoulous, E. Beraud, C. Le Page, A. Zamora, A.
Ferry, D. Bernard, A. D. Strosberg and P. O. Couraud,
“Anergy Induction in Encephalitogenic T Cells by Brain
Microvessel Endothelial Cells Is Inhibited by Inter-
leukin-1,” European Journal of Immunology, Vol. 25, No.
5, 1995, pp. 1176-1183. doi:10.1002/eji.1830250507
[56] B. O. Fabriek, E. S. Van Haastert, I. Galea, M. M. Polfliet,
E. D. Dopp, M. M. Van Den Heuvel, T. K. Van Den Berg,
C. J. De Groot, P. Van Der Valk and C. D. Dijkstra,
“CD163-Positive Perivascular Macrophages in the Hu-
man CNS Express Molecules for Antigen Recognition
and Presentation,” Glia, Vol. 51, No. 4, 2005, pp. 297-
305. doi:10.1002/glia.20208
[57] E. C. Henning, C. A. Ruetzler, M. R. Gaudinski, T. C. Hu,
L. L. Latour, J. M. Hallenbeck and S. Warach, “Feridex
Preloading Permits Tracking of CNS-Resident Macro-
phages after Transient Middle Cerebral Artery Occlu-
sion,” Journal of Cerebral Blood Flow & Metabolism,
Vol. 29, No. 7, 2009, pp. 1229-1239.
doi:10.1038/jcbfm.2009.48
[58] W. F. Hickey and H. Kimura, “Perivascular Microglial
Cells of the CNS Are Bone Marrow-Derived and Present
Antigen in Vivo,” Science, Vol. 239, No. 4837, 1988, pp.
290-292. doi:10.1126/science.3276004
[59] M. M. Polfliet, F. van de Veerdonk, E. A. Dopp, E. M.
van Kesteren-Hendrikx, N. van Rooijen, C. D. Dijkstra
and T. K. van den Berg, “The Role of Perivascular and
Meningeal Macrophages in Experimental Allergic En-
cephalomyelitis,” Journal of Neuroimmunology, Vol. 122,
No. 1-2, 2002, pp. 1-8.
doi:10.1016/S0165-5728(01)00445-3
[60] W. E. Thomas, “Brain Macrophages: On the Role of Peri-
cytes and Perivascular Cells,” Brain Research Reviews,
Vol. 31, No. 1, 1999, pp. 42-57.
doi:10.1016/S0165-0173(99)00024-7
[61] H. Kettenmann, U. K. Hanisch, M. Noda and A. Verk-
hratsky, “Physiology of Microglia,” Physiological Re-
views, Vol. 91, No. 2, 2011, pp. 461-553.
doi:10.1152/physrev.00011.2010
[62] F. Ginhoux, M. Greter, M. Leboeuf, S. Nandi, P. See, S.
Gokhan, M. F. Mehler, S. J. Conway, L. G. Ng, E. R.
Stanley, I. M. Samokhvalov and M. Merad, “Fate Map-
ping Analysis Reveals That Adult Microglia Derive from
Primitive Macrophages,” Science , Vol. 330, No. 6005,
2010, pp. 841-845. doi:10.1126/science.1194637
[63] S. H. Orkin and L. I. Zon, “Hematopoiesis: An Evolving
Paradigm for Stem Cell Biology,” Cell, Vol. 132, No. 4,
2008, pp. 631-644. doi:10.1016/j.cell.2008.01.025
[64] H. Neumann, M. R. Kotter and R. J. Franklin, “Debris
Clearance by Microglia: An Essential Link between De-
generation and Regeneration,” Brain, Vol. 132, No. 2,
2009, pp. 288-295.
[65] M. B. Graeber, “Changing Face of Microglia,” Science,
Vol. 330, No. 6005, 2010, pp. 783-788.
doi:10.1126/science.1190929
[66] F. Aloisi, “Immune Function of Microglia,” Glia, Vol. 36,
No. 2, 2001, pp. 165-179. doi:10.1002/glia.1106
[67] K. Saijo and C. K. Glass, “Microglial Cell Origin and
Phenotypes in Health and Disease,” Nature Reviews Im-
munology, Vol. 11, No. 11, 2011, pp. 775-787.
doi:10.1038/nri3086
[68] F. Aloisi, F. Ria and L. Adorini, “Regulation of T-Cell
Responses by CNS Antigen-Presenting Cells: Different
Roles for Microglia and Astrocytes,” Immunology Today,
Vol. 21, No. 3, 2000, pp. 141-147.
doi:10.1016/S0167-5699(99)01512-1
[69] M. K. Matyszak, S. Denis-Donini, S. Citterio, R. Longhi,
F. Granucci and P. Ricciardi-Castagnoli, “Microglia In-
duce Myelin Basic Protein-Specific T Cell Anergy or T
Cell Activation, According to Their State of Activation,”
European Journal of Immunology, Vol. 29, No. 10, 1999,
pp. 3063-3076.
doi:10.1002/(SICI)1521-4141(199910)29:10<3063::AID-
IMMU3063>3.0.CO;2-G
[70] F. Aloisi, F. Ria, S. Columba-Cabezas, H. Hess, G. Penna
and L. Adorini, “Relative Efficiency of Microglia, Astro-
cytes, Dendritic Cells and B Cells in Naive CD4+ T Cell
Priming and Th1/Th2 Cell Restimulation,” European
Journal of Immunology, Vol. 29, No. 9, 1999, pp. 2705-
2714.
doi:10.1002/(SICI)1521-4141(199909)29:09<2705::AID-
IMMU2705>3.0.CO;2-1
[71] E. J. McMahon, S. L. Bailey, C. V. Castenada, H. Wald-
ner and S. D. Miller, “Epitope Spreading Initiates in the
CNS in Two Mouse Models of Multiple Sclerosis,” Na-
ture Medicine, Vol. 11, No. 3, 2005, pp. 335-339.
doi:10.1038/nm1202
[72] [72] C. Beauvillain, S. Donnou, U. Jarry, M. Scotet, H.
Gascan, Y. Delneste, P. Guermonprez, P. Jeannin and D.
Couez, “Neonatal and Adult Microglia Cross-Present
Exogenous Antigens,” Glia, Vol. 56, No. 1, 2008, pp. 69-
77. doi:10.1002/glia.20565
[73] F. Aloisi, F. Ria, G. Penna and L. Adorini, “Microglia
Are More Efficient than Astrocytes in Antigen Processing
and in Th1 but Not Th2 Cell Activation,” The Journal of
Immunology, Vol. 160, No. 10, 1998, pp. 4671-4680.
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
220
[74] E. D. Ponomarev, L. P. Shriver, K. Maresz and B. N.
Dittel, “Microglial Cell Activation and Proliferation Pre-
cedes the Onset of CNS Autoimmunity,” Journal of Neu-
roscience Research, Vol. 81, No. 3, 2005, pp. 374-389.
doi:10.1002/jnr.20488
[75] A. C. Murphy, S. J. Lalor, M. A. Lynch and K. H. Mills,
“Infiltration of Th1 and Th17 Cells and Activation of Mi-
croglia in the CNS during the Course of Experimental
Autoimmune Encephalomyelitis,” Brain, Behavior, and
Immunity, Vol. 24, No. 4, 2010, pp. 641-651.
doi:10.1016/j.bbi.2010.01.014
[76] F. L. Heppner, M. Greter, D. Marino, J. Falsig, G.
Raivich, N. Hovelmeyer, A. Waisman, T. Rulicke, M.
Prinz, J. Priller, B. Becher and A. Aguzzi, “Experimental
Autoimmune Encephalomyelitis Repressed by Microglial
Paralysis,” Nature Medicine, Vol. 11, No. 2, 2005, pp.
146-152. doi:10.1038/nm1177
[77] A. L. Ford, E. Foulcher, F. A. Lemckert and J. D. Sedg-
wick, “Microglia Induce CD4 T Lymphocyte Final Ef-
fector Function and Death,” The Journal of Experimental
Medicine, Vol. 184, No. 5, 1996, pp. 1737-1745.
doi:10.1084/jem.184.5.1737
[78] F. Geissmann, M. G. Manz, S. Jung, M. H. Sieweke, M.
Merad and K. Ley, “Development of Monocytes, Macro-
phages, and Dendritic Cells,” Science, Vol. 327, No. 5966,
2010, pp. 656-661. doi:10.1126/science.1178331
[79] E. D. Ponomarev, K. Maresz, Y. Tan and B. N. Dittel,
“CNS-Derived Interleukin-4 Is Essential for the Regula-
tion of Autoimmune Inflammation and Induces a State of
Alternative Activation in Microglial Cells,” The Journal
of Neuroscience, Vol. 27, No. 40, 2007, pp. 10714-10721.
doi:10.1523/JNEUROSCI.1922-07.2007
[80] K. A. Kigerl, J. C. Gensel, D. P. Ankeny, J. K. Alexander,
D. J. Donnelly and P. G. Popovich, “Identification of
Two Distinct Macrophage Subsets with Divergent Effects
Causing Either Neurotoxicity or Regeneration in the In-
jured Mouse Spinal Cord,” The Journal of Neuroscience,
Vol. 29, No. 43, 2009, pp. 13435-13444.
doi:10.1523/JNEUROSCI.3257-09.2009
[81] Z. Zhang, Z. Y. Zhang, J. Schittenhelm, Y. Wu, R. Mey-
ermann and H. J. Schluesener, “Parenchymal Accumula-
tion of CD163+ Macrophages/Microglia in Multiple Scle-
rosis Brains,” Journal of Neuroimmunology, Vol. 237, No.
1-2, 2011, pp. 73-79. doi:10.1016/j.jneuroim.2011.06.006
[82] B. Ajami, J. L. Bennett, C. Krieger, K. M. McNagny and
F. M. Rossi, “Infiltrating Monocytes Trigger EAE Pro-
gression, but Do Not Contribute to the Resident Microglia
Pool,” Nature Neuroscience, Vol. 14, No. 9, 2011, pp.
1142-1149. doi:10.1038/nn.2887
[83] B. Almolda, B. Gonzalez and B. Castellano, “Activated
Microglial Cells Acquire an Immature Dendritic Cell
Phenotype and May Terminate the Immune Response in
an Acute Model of EAE,” Journal of Neuroimmunology,
Vol. 223, No. 1-2, 2010, pp. 39-54.
doi:10.1016/j.jneuroim.2010.03.021
[84] K. D. McCoy and G. Le Gros, “The Role of CTLA-4 in
the Regulation of T Cell Immune Responses,” Immunol-
ogy & Cell Biology, Vol. 77, No. 1, 1999, pp. 1-10.
doi:10.1046/j.1440-1711.1999.00795.x
[85] N. J. Allen and B. A. Barres, “Neuroscience: Glia More
than Just Brain Glue,” Nature, Vol. 457, No. 7230, 2009,
pp. 675-677. doi:10.1038/457675a
[86] G. C. Petzold and V. N. Murthy, “Role of Astrocytes in
Neurovascular Coupling,” Neuron, Vol. 71, No. 5, 2011,
pp. 782-797. doi:10.1016/j.neuron.2011.08.009
[87] P. A. Carpentier, W. S. Begolka, J. K. Olson, A. Elhofy,
W. J. Karpus and S. D. Miller, “Differential Activation of
Astrocytes by Innate and Adaptive Immune Stimuli,”
Glia, Vol. 49, No. 3, 2005, pp. 360-374.
doi:10.1002/glia.20117
[88] A. Fontana, W. Fierz and H. Wekerle, “Astrocytes Pre-
sent Myelin Basic Protein to Encephalitogenic T-Cell
lines,” Nature, Vol. 307, No. 5948, 1984, pp. 273-276.
doi:10.1038/307273a0
[89] J. M. Soos, J. Morrow, T. A. Ashley, B. E. Szente, E. K.
Bikoff and S. S. Zamvil, “Astrocytes Express Elements of
the Class II Endocytic Pathway and Process Central Nerv-
ous System Autoantigen for Presentation to Encephalito-
genic T Cells,” The Journal of Immunology, Vol. 161, No.
11, 1998, pp. 5959-5966.
[90] O. Stuve, S. Youssef, A. J. Slavin, C. L. King, J. C.
Patarroyo, D. L. Hirschberg, W. J. Brickey, J. M. Soos, J.
F. Piskurich, H. A. Chapman and S. S. Zamvil, “The Role
of the MHC Class II Transactivator in Class II Expression
and Antigen Presentation by Astrocytes and in Suscepti-
bility to Central Nervous System Autoimmune Disease,”
The Journal of Immunology, Vol. 169, No. 12, 2002, pp.
6720-6732.
[91] A. Cornet, E. Bettelli, M. Oukka, C. Cambouris, V.
Avellana-Adalid, K. Kosmatopoulos and R. S. Liblau,
“Role of Astrocytes in Antigen Presentation and Naive
T-Cell Activation,” Journal of Neuroimmunology, Vol.
106, No. 1-2, 2000, pp. 69-77.
doi:10.1016/S0165-5728(99)00215-5
[92] K. Baur, M. Rauer, K. Richter, A. Pagenstecher, J. Gotz,
J. Hausmann and P. Staeheli, “Antiviral CD8 T Cells
Recognize Borna Disease Virus Antigen Transgenically
Expressed in Either Neurons or Astrocytes,” Journal of
Virology, Vol. 82, No. 6, 2008, pp. 3099-3108.
doi:10.1128/JVI.02479-07
[93] K. Williams, E. Ulvestad, A. Waage, J. P. Antel and J.
McLaurin, “Activation of Adult Human Derived Micro-
glia by Myelin Phagocytosis in Vitro,” Journal of Neuro-
science Research, Vol. 38, No. 4, 1994, pp. 433-443.
doi:10.1002/jnr.490380409
[94] R. M. Ransohoff and M. A. Brown, “Innate Immunity in
the Central Nervous System,” The Journal of Clinical In-
vestigation, Vol. 122, No. 4, 2012, pp. 1164-1171.
doi:10.1172/JCI58644
[95] D. N. Hart and J. W. Fabre, “Demonstration and Charac-
terization of Ia-Positive Dendritic Cells in the Interstitial
Connective Tissues of Rat Heart and Other Tissues, but
Not Brain,” The Journal of Experimental Medicine, Vol.
154, No. 2, 1981, pp. 347-361.
doi:10.1084/jem.154.2.347
[96] R. M. Steinman and Z. A. Cohn, “Identification of a
Novel Cell Type in Peripheral Lymphoid Organs of Mice.
I. Morphology, Quantitation, Tissue Distribution,” The
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS 221
Journal of Experimental Medicine, Vol. 137, No. 5, 1973,
pp. 1142-1162. doi:10.1084/jem.137.5.1142
[97] P. G. McMenamin, R. J. Wealthall, M. Deverall, S. J.
Cooper and B. Griffin, “Macrophages and Dendritic Cells
in the Rat Meninges and Choroid Plexus: Three-Dimen-
sional Localisation by Environmental Scanning Electron
Microscopy and Confocal Microscopy,” Cell and Tissue
Research, Vol. 313, No. 3, 2003, pp. 259-269.
doi:10.1007/s00441-003-0779-0
[98] M. Pashenkov, Y. M. Huang, V. Kostulas, M. Haglund,
M. Soderstrom and H. Link, “Two Subsets of Dendritic
Cells Are Present in Human Cerebrospinal Fluid,” Brain,
Vol. 124, No. 3, 2001, pp. 480-492.
[99] M. Greter, F. L. Heppner, M. P. Lemos, B. M. Odermatt,
N. Goebels, T. Laufer, R. J. Noelle and B. Becher, “Den-
dritic Cells Permit Immune Invasion of the CNS in an
Animal Model of Multiple Sclerosis,” Nature Medicine,
Vol. 11, No. 3, 2005, pp. 328-334. doi:10.1038/nm1197
[100] J. Karman, H. H. Chu, D. O. Co, C. M. Seroogy, M.
Sandor and Z. Fabry, “Dendritic Cells Amplify T Cell-
Mediated Immune Responses in the Central Nervous
System,” The Journal of Immunology, Vol. 177, No. 11,
2006, pp. 7750-7760.
[101] J. M. Goverman, “Immune Tolerance in Multiple Sclero-
sis,” Immunological Reviews, Vol. 241, No. 1, 2011, pp.
228-240. doi:10.1111/j.1600-065X.2011.01016.x
[102] J. M. Ilarregui and G. A. Rabinovich, “Tolerogenic Den-
dritic Cells in the Control of Autoimmune Neuroinflam-
mation: An Emerging Role of Protein-Glycan Interac-
tions,” Neuroimmunomodulation, Vol. 17, No. 3, 2010,
pp. 157-160. doi:10.1159/000258712
[103] M. Ioannou, T. Alissafi, I. Lazaridis, G. Deraos, J. Mat-
soukas, A. Gravanis, V. Mastorodemos, A. Plaitakis, A.
Sharpe, D. Boumpas and P. Verginis, “Crucial Role of
Granulocytic Myeloid-Derived Suppressor Cells in the
Regulation of Central Nervous System Autoimmune Dis-
ease,” The Journal of Immunology, Vol. 188, No. 3, 2012,
pp. 1136-1146. doi:10.4049/jimmunol.1101816
[104] L. Walter and M. L. Albert, “Cutting Edge: Cross-Pre-
sented Intracranial Antigen Primes CD8+ T cells,” The
Journal of Immunology, Vol. 178, No. 10, 2007, pp.
6038-6042.
[105] L. Santambrogio, S. L. Belyanskaya, F. R. Fischer, B.
Cipriani, C. F. Brosnan, P. Ricciardi-Castagnoli, L. J.
Stern, J. L. Strominger and R. Riese, “Developmental
Plasticity of CNS Microglia,” Proceedings of the Na-
tional Academy of Sciences of the United States of Amer-
ica, Vol. 98, No. 11, 2001, pp. 6295-6300.
doi:10.1073/pnas.111152498
[106] S. Jung, D. Unutmaz, P. Wong, G. Sano, K. De los Santos,
T. Sparwasser, S. Wu, S. Vuthoori, K. Ko, F. Zavala, E.
G. Pamer, D. R. Littman and R. A. Lang, “In Vivo Deple-
tion of CD11c+ Dendritic Cells Abrogates Priming of
CD8+ T Cells by Exogenous Cell-Associated Antigens,”
Immunity, Vol. 17, No. 2, 2002, pp. 211-220.
doi:10.1016/S1074-7613(02)00365-5
[107] R. L. Lindquist, G. Shakhar, D. Dudziak, H. Wardemann,
T. Eisenreich, M. L. Dustin and M. C. Nussenzweig,
“Visualizing Dendritic Cell Networks in Vivo,” Nature
Immunology, Vol. 5, No. 12, 2004, pp. 1243-1250.
doi:10.1038/ni1139
[108] K. Bulloch, M. M. Miller, J. Gal-Toth, T. A. Milner, A.
Gottfried-Blackmore, E. M. Waters, U. W. Kaunzner, K.
Liu, R. Lindquist, M. C. Nussenzweig, R. M. Steinman
and B. S. McEwen, “CD11c/EYFP Transgene Illuminates
a Discrete Network of Dendritic Cells within the Embry-
onic, Neonatal, Adult, and Injured Mouse Brain,” The
Journal of Comparative Neurology, Vol. 508, No. 5, 2008,
pp. 687-710. doi:10.1002/cne.21668
[109] C. Prodinger, J. Bunse, M. Kruger, F. Schiefenhovel, C.
Brandt, J. D. Laman, M. Greter, K. Immig, F. Heppner, B.
Becher and I. Bechmann, “CD11c-Expressing Cells Re-
side in the Juxtavascular Parenchyma and Extend Proc-
esses into the Glia Limitans of the Mouse Nervous Sys-
tem,” Acta Neuropathologica, Vol. 121, No. 4, 2011, pp.
445-458. doi:10.1007/s00401-010-0774-y
[110] A. Gottfried-Blackmore, U. W. Kaunzner, J. Idoyaga, J. C.
Felger, B. S. McEwen and K. Bulloch, “Acute in Vivo
Exposure to Interferon-Gamma Enables Resident Brain
Dendritic Cells to Become Effective Antigen Presenting
Cells,” Proceedings of the National Academy of Sciences
of the United States of America, Vol. 106, No. 49, 2009,
pp. 20918-20923. doi:10.1073/pnas.0911509106
[111] F. Geissmann, S. Gordon, D. A. Hume, A. M. Mowat and
G. J. Randolph, “Unravelling Mononuclear Phagocyte
Heterogeneity,” Nature Reviews Immunology, Vol. 10,
No. 6, 2010, pp. 453-460. doi:10.1038/nri2784
[112] S. L. Bailey, B. Schreiner, E. J. McMahon and S. D.
Miller, “CNS Myeloid DCs Presenting Endogenous Mye-
lin Peptides ‘Preferentially’ Polarize CD4+ T(H)-17 Cells
in Relapsing EAE,” Nature Immunology, Vol. 8, No. 2,
2007, pp. 172-180. doi:10.1038/ni1430
[113] T. Olsson, “Multiple Sclerosis:Cerebrospinal Fluid,” An-
nals of Neurology, Vol. 36, 1994, pp. S100-S102.
doi:10.1002/ana.410360723
[114] T. L. Sorensen, M. Tani, J. Jensen, V. Pierce, C. Lucchi-
netti, V. A. Folcik, S. Qin, J. Rottman, F. Sellebjerg, R.
M. Strieter, J. L. Frederiksen and R. M. Ransohoff, “Ex-
pression of Specific Chemokines and Chemokine Recep-
tors in the Central Nervous System of Multiple Sclerosis
Patients,” The Journal of Clinical Investigation, Vol. 103,
No. 6, 1999, pp. 807-815. doi:10.1172/JCI5150
[115] H. L. Weiner, “Multiple Sclerosis Is an Inflammatory T-
Cell-Mediated Autoimmune Disease,” Archives of Neu-
rology, Vol. 61, No. 10, 2004, pp. 1613-1615.
doi:10.1001/archneur.61.10.1613
[116] C. Lucchinetti, W. Bruck, J. Parisi, B. Scheithauer, M.
Rodriguez and H. Lassmann, “Heterogeneity of Multiple
Sclerosis Lesions: Implications for the Pathogenesis of
Demyelination,” Annals of Neurology, Vol. 47, No. 6,
2000, pp. 707-717.
doi:10.1002/1531-8249(200006)47:6<707::AID-ANA3>3
.0.CO;2-Q
[117] J. K. Olson, T. N. Eagar and S. D. Miller, “Functional
Activation of Myelin-Specific T Cells by Virus-Induced
Molecular Mimicry,” The Journal of Immunology, Vol.
169, No. 5, 2002, pp. 2719-2726.
[118] J. D. Kriesel, M. R. Hobbs, B. B. Jones, B. Milash, R. M.
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
222
Nagra and K. F. Fischer, “Deep Sequencing for the De-
tection of Virus-Like Sequences in the Brains of Patients
with Multiple Sclerosis: Detection of GBV-C in Human
Brain,” PLoS One, Vol. 7, No. 3, 2012, Article ID:
e31886. doi:10.1371/journal.pone.0031886
[119] M. H. Barnett and J. W. Prineas, “Relapsing and Remit-
ting Multiple Sclerosis: Pathology of the Newly Forming
Lesion,” Annals of Neurology, Vol. 55, No. 4, 2004, pp.
458-468. doi:10.1002/ana.20016
[120] M. H. Barnett and I. Sutton, “The Pathology of Multiple
Sclerosis: A Paradigm Shift,” Current Opinion in Neu-
rology, Vol. 19, No. 3, 2006, pp. 242-247.
doi:10.1097/01.wco.0000227032.47458.cb
[121] G. Locatelli, S. Wortge, T. Buch, B. Ingold, F. Frommer,
B. Sobottka, M. Kruger, K. Karram, C. Buhlmann, I.
Bechmann, F. L. Heppner, A. Waisman and B. Becher,
“Primary Oligodendrocyte Death Does Not Elicit Anti-
CNS Immunity,” Nature Neuroscience, Vol. 15, No. 4,
2012, pp. 543-550. doi:10.1038/nn.3062
[122] H. B. Pohl, C. Porcheri, T. Mueggler, L. C. Bachmann, G.
Martino, D. Riethmacher, R. J. Franklin, M. Rudin and U.
Suter, “Genetically Induced Adult Oligodendrocyte Cell
Death Is Associated with Poor Myelin Clearance, Re-
duced Remyelination, and Axonal Damage,” The Journal
of Neuroscience, Vol. 31, No. 3, 2011, pp. 1069-1080.
doi:10.1523/JNEUROSCI.5035-10.2011
[123] T. Henics and D. N. Wheatley, “Cytoplasmic Vacuolation,
Adaptation and Cell Death: A View on New Perspectives
and Features,” Biology of the Cell, Vol. 91, No. 7, 1999,
pp. 485-498. doi:10.1016/S0248-4900(00)88205-2
[124] C. M. Poser, “The Role of Trauma in the Pathogenesis of
Multiple Sclerosis: A Review,” Clinical Neurology and
Neurosurgery, Vol. 96, No. 2, 1994, pp. 103-110.
doi:10.1016/0303-8467(94)90042-6
[125] A. G. Kermode, A. J. Thompson, P. Tofts, D. G.
MacManus, B. E. Kendall, D. P. Kingsley, I. F. Moseley,
P. Rudge and W. I. McDonald, “Breakdown of the
Blood-Brain Barrier Precedes Symptoms and Other MRI
Signs of New Lesions in Multiple Sclerosis. Pathogenetic
and Clinical Implications,” Brain, Vol. 113, No. 5, 1990,
pp. 1477-1489. doi:10.1093/brain/113.5.1477
[126] S. Hisahara, T. Araki, F. Sugiyama, K. Yagami, M. Su-
zuki, K. Abe, K. Yamamura, J. Miyazaki, T. Momoi, T.
Saruta, C. C. Bernard, H. Okano and M. Miura, “Targeted
Expression of Baculovirus p35 Caspase Inhibitor in Oli-
godendrocytes Protects Mice against Autoimmune-Me-
diated Demyelination,” The EMBO Journal, Vol. 19, No.
3, 2000, pp. 341-348. doi:10.1093/emboj/19.3.341
[127] M. L. Albert, S. F. Pearce, L. M. Francisco, B. Sauter, P.
Roy, R. L. Silverstein and N. Bhardwaj, “Immature Den-
dritic Cells Phagocytose Apoptotic Cells via Alphavbeta5
and CD36, and Cross-Present Antigens to Cytotoxic T
Lymphocytes,” The Journal of Experimental Medicine,
Vol. 188, No. 7, 1998, pp. 1359-1368.
doi:10.1084/jem.188.7.1359
[128] M. L. Albert, B. Sauter and N. Bhardwaj, “Dendritic
Cells Acquire Antigen from Apoptotic Cells and Induce
Class I-Restricted CTLs,” Nature, Vol. 392, No. 6671,
1998, pp. 86-89. doi:10.1038/32183
[129] N. E. Blachere, R. B. Darnell and M. L. Albert, “Apop-
totic Cells Deliver Processed Antigen to Dendritic Cells
for Cross-Presentation,” PLoS Biology, Vol. 3, No. 6,
2005, p. e185. doi:10.1371/journal.pbio.0030185
[130] F. Meloni, D. Accapezzato, C. Agresti, F. Aloisi, G. Ris-
tori, M. Salvetti, R. Furlan, G. Martino, V. Barnaba and
M. Paroli, “Dendritic Cells Loaded with Apoptotic Oli-
godendrocytes as a Source of Myelin T-Cell Epitopes in
Multiple Sclerosis,” Clinical Immunology, Vol. 129, No.
2, 2008, pp. 286-294. doi:10.1016/j.clim.2008.07.017
[131] H. Kono and K. L. Rock, “How Dying Cells Alert the
Immune System to Danger,” Nature Reviews Immunology,
Vol. 8, No. 4, 2008, pp. 279-289. doi:10.1038/nri2215
[132] M. P. Mycko, H. Cwiklinska, J. Szymanski, B. Szyman-
ska, G. Kudla, L. Kilianek, A. Odyniec, C. F. Brosnan
and K. W. Selmaj, “Inducible Heat Shock Protein 70 Pro-
motes Myelin Autoantigen Presentation by the HLA
Class II,” The Journal of Immunology, Vol. 172, No. 1,
2004, pp. 202-213.
[133] B. Ravishankar, H. Liu, R. Shinde, P. Chandler, B. Baban,
M. Tanaka, D. H. Munn, A. L. Mellor, M. C. Karlsson
and T. L. McGaha, “Tolerance to Apoptotic Cells Is
Regulated by Indoleamine 2,3-Dioxygenase,” Proceed-
ings of the National Academy of Sciences of the United
States of America, Vol. 109, No. 10, 2012, pp. 3909-3914.
doi:10.1073/pnas.1117736109
[134] Y. Wang, M. A. Lawson, K. W. Kelley and R. Dantzer,
“Primary Murine Microglia Are Resistant to Nitric Oxide
Inhibition of Indoleamine 2,3-Dioxygenase,” Brain, Be-
havior, and Immunity, Vol. 24, No. 8, 2010, pp. 1249-
1253. doi:10.1016/j.bbi.2010.04.015
[135] A. Andersson, R. Covacu, D. Sunnemark, A. I. Danilov,
A. Dal Bianco, M. Khademi, E. Wallstrom, A. Lobell, L.
Brundin, H. Lassmann and R. A. Harris, “Pivotal Ad-
vance: HMGB1 Expression in Active Lesions of Human
and Experimental Multiple Sclerosis,” Journal of Leuko-
cyte Biology, Vol. 84, No. 5, 2008, pp. 1248-1255.
doi:10.1189/jlb.1207844
[136] R. A. Williamson, M. P. Burgoon, G. P. Owens, O.
Ghausi, E. Leclerc, L. Firme, S. Carlson, J. Corboy, P. W.
Parren, P. P. Sanna, D. H. Gilden and D. R. Burton,
“Anti-DNA Antibodies Are a Major Component of the
Intrathecal B Cell Response in Multiple Sclerosis,” Pro-
ceedings of the National Academy of Sciences of the
United States of America, Vol. 98, No. 4, 2001, pp. 1793-
1798. doi:10.1073/pnas.031567598
[137] L. K. Peterson, T. Masaki, S. R. Wheelwright, I. Tsunoda
and R. S. Fujinami, “Cross-Reactive Myelin Antibody
Induces Renal Pathology,” Autoimmunity, Vol. 41, No. 7,
2008, pp. 526-536. doi:10.1080/08916930802128680
[138] L. K. Peterson, I. Tsunoda, T. Masaki and R. S. Fujinami,
“Polyreactive Myelin Oligodendrocyte Glycoprotein An-
tibodies: Implications for Systemic Autoimmunity in Pro-
gressive Experimental Autoimmune Encephalomyelitis,”
Journal of Neuroimmunology, Vol. 183, No. 1-2, 2007,
pp. 69-80. doi:10.1016/j.jneuroim.2006.11.024
[139] M. Cieslak, F. Kukulski and M. Komoszynski, “Emerg-
ing Role of Extracellular Nucleotides and Adenosine in
Multiple Sclerosis,” Purinergic Signalling, Vol. 7, No. 4,
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS 223
2011, pp. 393-402. doi:10.1007/s11302-011-9250-y
[140] G. Birnbaum, L. Kotilinek, P. Schlievert, H. B. Clark, J.
Trotter, E. Horvath, E. Gao, M. Cox and P. E. Braun,
“Heat Shock Proteins and Experimental Autoimmune
Encephalomyelitis (EAE): I. Immunization with a Peptide
of the Myelin Protein 2’,3’ Cyclic Nucleotide 3’ Phos-
phodiesterase That Is Cross-Reactive with a Heat Shock
Protein Alters the Course of EAE,” Journal of Neurosci-
ence Research, Vol. 44, No. 4, 1996, pp. 381-396.
doi:10.1002/(SICI)1097-4547(19960515)44:4<381::AID-
JNR10>3.0.CO;2-5
[141] M. Rickmann and J. R. Wolff, “S100 Immunoreactivity
in a Subpopulation of Oligodendrocytes and Ranvier’S
Nodes of Adult Rat Brain,” Neuroscience Letters, Vol.
186, No. 1, 1995, pp. 13-16.
doi:10.1016/0304-3940(95)11269-3
[142] R. A. Sobel and M. E. Mitchell, “Fibronectin in Multiple
Sclerosis Lesions,” The American Journal of Pathology,
Vol. 135, No. 1, 1989, pp. 161-168.
[143] R. A. Sobel, E. E. Schneeberger and R. B. Colvin, “The
Immunopathology of Acute Experimental Allergic Ence-
phalomyelitis. V. A Light Microscopic and Ultrastruc-
tural Immunohistochemical Analysis of Fibronectin and
Fibrinogen,” The American Journal of Pathology, Vol.
131, No. 3, 1988, pp. 547-558.
[144] P. Y. Paterson, C. S. Koh and H. C. Kwaan, “Role of the
Clotting System in the Pathogenesis of Neuroimmu-
nologic Disease,” Federation Proceedings, Vol. 46, No. 1,
1987, pp. 91-96.
[145] Z. Siskova, W. Baron, H. de Vries and D. Hoekstra, “Fi-
bronectin Impedes ‘Myelin’ Sheet-Directed Flow in Oli-
godendrocytes: A Role for a Beta 1 Integrin-Mediated
PKC Signaling Pathway in Vesicular Trafficking,” Mo-
lecular and Cellular Neuroscience, Vol. 33, No. 2, 2006,
pp. 150-159. doi:10.1016/j.mcn.2006.07.001
[146] S. A. Back, T. M. Tuohy, H. Chen, N. Wallingford, A.
Craig, J. Struve, N. L. Luo, F. Banine, Y. Liu, A. Chang,
B. D. Trapp, B. F. Bebo, Jr., M. S. Rao and L. S. Sherman,
“Hyaluronan Accumulates in Demyelinated Lesions and
Inhibits Oligodendrocyte Progenitor Maturation,” Nature
Medicine, Vol. 11, No. 9, 2005, pp. 966-972.
[147] Z. Fabry, H. A. Schreiber, M. G. Harris and M. Sandor,
“Sensing the Microenvironment of the Central Nervous
System: Immune Cells in the Central Nervous System
and Their Pharmacological Manipulation,” Current Opin-
ion in Pharmacology, Vol. 8, No. 4, 2008, pp. 496-507.
doi:10.1016/j.coph.2008.07.009
[148] J. A. Sloane, C. Batt, Y. Ma, Z. M. Harris, B. Trapp and
T. Vartanian, “Hyaluronan Blocks Oligodendrocyte Pro-
genitor Maturation and Remyelination through TLR2,”
Proceedings of the National Academy of Sciences of the
United States of America, Vol. 107, No. 25, 2010, pp.
11555-11560. doi:10.1073/pnas.1006496107
[149] A. M. de Mestre, M. A. Staykova, J. R. Hornby, D. O.
Willenborg and M. D. Hulett, “Expression of the Heparan
Sulfate-Degrading Enzyme Heparanase Is Induced in In-
filtrating CD4+ T Cells in Experimental Autoimmune
Encephalomyelitis and Regulated at the Level of Tran-
scription by Early Growth Response Gene 1,” Journal of
Leukocyte Biology, Vol. 82, No. 5, 2007, pp. 1289-1300.
doi:10.1189/jlb.0507315
[150] J. van Horssen, L. Bo, C. M. Vos, I. Virtanen and H. E.
de Vries, “Basement Membrane Proteins in Multiple
Sclerosis-Associated Inflammatory Cuffs: Potential Role
in Influx and Transport of Leukocytes,” Journal of Neu-
ropathology & Experimental Neurology, Vol. 64, No. 8,
2005, pp. 722-729.
doi:10.1097/01.jnen.0000173894.09553.13
[151] S. Miyake, T. Sakurai, K. Okumura and H. Yagita, “Iden-
tification of Collagen and Laminin Receptor Integrins on
Murine T Lymphocytes,” European Journal of Immu-
nology, Vol. 24, No. 9, 1994, pp. 2000-2005.
doi:10.1002/eji.1830240910
[152] J. T. Maikos, R. A. Elias and D. I. Shreiber, “Mechanical
Properties of Dura Mater from the Rat Brain and Spinal
Cord,” Journal of Neurotrauma, Vol. 25, No. 1, 2008, pp.
38-51. doi:10.1089/neu.2007.0348
[153] S. Chandler, J. Cossins, J. Lury and G. Wells, “Macro-
phage Metalloelastase Degrades Matrix and Myelin Pro-
teins and Processes a Tumour Necrosis Factor-Alpha Fu-
sion Protein,” Biochemical and Biophysical Research
Communications, Vol. 228, No. 2, 1996, pp. 421-429.
doi:10.1006/bbrc.1996.1677
[154] R. Debret, F. Antonicelli, A. Theill, W. Hornebeck, P.
Bernard, M. Guenounou and R. Le Naour, “Elastin-De-
rived Peptides Induce a T-Helper Type 1 Polarization of
Human Blood Lymphocytes,” Arteriosclerosis, Thrombo-
sis, and Vascular Biology, Vol. 25, No. 7, 2005, pp.
1353-1358. doi:10.1161/01.ATV.0000168412.50855.9f
[155] M. Stancic, J. van Horssen, V. L. Thijssen, H. J. Gabius,
P. van der Valk, D. Hoekstra and W. Baron, “Increased
Expression of Distinct Galectins in Multiple Sclerosis
Lesions,” Neuropathology and Applied Neurobiology,
Vol. 37, No. 6, 2011, pp. 654-671.
doi:10.1111/j.1365-2990.2011.01184.x
[156] J. M. Ilarregui, D. O. Croci, G. A. Bianco, M. A. Toscano,
M. Salatino, M. E. Vermeulen, J. R. Geffner and G. A.
Rabinovich, “Tolerogenic Signals Delivered by Dendritic
Cells to T Cells through a Galectin-1-Driven Immu-
noregulatory Circuit Involving Interleukin 27 and Inter-
leukin 10,” Nature Immunology, Vol. 10, No. 9, 2009, pp.
981-991. doi:10.1038/ni.1772
[157] J. Wang, Z. H. Lu, H. J. Gabius, C. Rohowsky-Kochan, R.
W. Ledeen and G. Wu, “Cross-Linking of GM1 Gan-
glioside by Galectin-1 Mediates Regulatory T Cell Activ-
ity Involving TRPC5 Channel Activation: Possible Role
in Suppressing Experimental Autoimmune Encephalo-
myelit is,” The Journal of Immunology, Vol. 182, No. 7,
2009, pp. 4036-4045. doi:10.4049/jimmunol.0802981
[158] A. Grigorian and M. Demetriou, “Mgat5 Deficiency in T
Cells and Experimental Autoimmune Encephalomyelitis,”
ISRN Neurology, Vol. 2011, 2011, Article ID: 374314.
doi:10.5402/2011/374314
[159] M. Demetriou, M. Granovsky, S. Quaggin and J. W.
Dennis, “Negative Regulation of T-Cell Activation and
Autoimmunity by Mgat5 N-Glycosylation,” Nature, Vol.
409, No. 6821, 2001, pp. 733-739.
doi:10.1038/35055582
Copyright © 2012 SciRes. NM
Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS
Copyright © 2012 SciRes. NM
224
[160] S. U. Lee, A. Grigorian, J. Pawling, I. J. Chen, G. Gao, T.
Mozaffar, C. McKerlie and M. Demetriou, “N-Glycan
Processing Deficiency Promotes Spontaneous Inflamma-
tory Demyelination and Neurodegeneration,” The Journal
of Biological Chemistry, Vol. 282, No. 46, 2007, pp.
33725-33734. doi:10.1074/jbc.M704839200
[161] K. Liu and M. C. Nussenzweig, “Origin and Develop-
ment of Dendritic Cells,” Immunological Reviews, Vol.
234, No. 1, 2010, pp. 45-54.
doi:10.1111/j.0105-2896.2009.00879.x
[162] M. Colonna, G. Trinchieri and Y. J. Liu, “Plasmacytoid
Dendritic Cells in Immunity,” Nature Immunology, Vol. 5,
No. 12, 2004, pp. 1219-1226. doi:10.1038/ni1141
[163] B. D. Clarkson, E. Heninger, M. G. Harris, J. Lee, M.
Sandor and Z. Fabry, “Innate-Adaptive Crosstalk: How
Dendritic Cells Shape Immune Responses in the CNS,”
Advances in Experimental Medicine and Biology, Vol.
946, 2012, pp. 309-333.
doi:10.1007/978-1-4614-0106-3_18
[164] D. Dissanayake, H. Hall, N. Berg-Brown, A. R. Elford, S.
R. Hamilton, K. Murakami, L. S. Deluca, J. L. Gommer-
man and P. S. Ohashi, “Nuclear Factor-Kappab1 Controls
the Functional Maturation of Dendritic Cells and Prevents
the Activation of Autoreactive T Cells,” Nature Medicine,
Vol. 17, No. 12, 2011, pp. 1663-1667.
doi:10.1038/nm.2556
[165] A. L. Zozulya, S. Ortler, J. Lee, C. Weidenfeller, M.
Sandor, H. Wiendl and Z. Fabry, “Intracerebral Dendritic
Cells Critically Modulate Encephalitogenic versus Regu-
latory Immune Responses in the CNS,” The Journal of
Neuroscience, Vol. 29, No. 1, 2009, pp. 140-152.
doi:10.1523/JNEUROSCI.2199-08.2009
[166] P. Deshpande, I. L. King and B. M. Segal, “Cutting Edge:
CNS CD11c+ Cells from Mice with Encephalomyelitis
Polarize Th17 Cells and Support CD25+ CD4+ T Cell-
Mediated Immunosuppression, Suggesting Dual Roles in
the Disease Process,” The Journal of Immunology, Vol.
178, No. 11, 2007, pp. 6695-6699.
[167] M. N. Alonso, M. T. Wong, A. L. Zhang, D. Winer, M.
M. Suhoski, L. L. Tolentino, J. Gaitan, M. G. Davidson,
T. H. Kung, D. M. Galel, K. C. Nadeau, J. Kim, P. J. Utz,
K. Soderstrom and E. G. Engleman, “T(H)1, T(H)2, and
T(H)17 Cells Instruct Monocytes to Differentiate into
Specialized Dendritic Cell Subsets,” Blood, Vol. 118, No.
12, 2011, pp. 3311-3320.
doi:10.1182/blood-2011-03-341065
[168] M. El-Behi, B. Ciric, H. Dai, Y. Yan, M. Cullimore, F.
Safavi, G. X. Zhang, B. N. Dittel and A. Rostami, “The
Encephalitogenicity of T(H)17 Cells Is Dependent on
IL-1- and IL-23-Induced Production of the Cytokine
GM-CSF,” Nature Immunology, Vol. 12, No. 6, 2011, pp.
568-575. doi:10.1038/ni.2031
[169] K. Poppensieker, D. M. Otte, B. Schurmann, A. Limmer,
P. Dresing, E. Drews, B. Schumak, L. Klotz, J. Raasch, A.
Mildner, A. Waisman, S. Scheu, P. Knolle, I. Forster, M.
Prinz, W. Maier, A. Zimmer and J. Alferink, “CC
Chemokine Receptor 4 Is Required for Experimental
Autoimmune Encephalomyelitis by Regulating GM-CSF
and IL-23 Production in Dendritic Cells,” Proceedings of
the National Academy of Sciences of the United States of
America, Vol. 109, No. 10, 2012, pp. 3897-3902.
doi:10.1073/pnas.1114153109
[170] R. A. Maldonado and U. H. von Andrian, “How Tolero-
genic Dendritic Cells Induce Regulatory T Cells,” Ad-
vances in Immunology, Vol. 108, 2010, pp. 111-165.
doi:10.1016/B978-0-12-380995-7.00004-5
[171] S. M. Opal and V. A. DePalo, “Anti-Inflammatory Cyto-
kines,” Chest, Vol. 117, No. 4, 2000, pp. 1162-1172.
doi:10.1378/chest.117.4.1162
[172] Y. Laouar, T. Town, D. Jeng, E. Tran, Y. Wan, V. K.
Kuchroo and R. A. Flavell, “TGF-Beta Signaling in Den-
dritic Cells Is a Prerequisite for the Control of Autoim-
mune Encephalomyelitis,” Proceedings of the National
Academy of Sciences of the United States of America, Vol.
105, No. 31, 2008, pp. 10865-10870.
doi:10.1073/pnas.0805058105
[173] M. Delgado, E. Gonzalez-Rey and D. Ganea, “The Neu-
ropeptide Vasoactive Intestinal Peptide Generates Tolero-
genic Dendritic Cells,” The Journal of Immunology, Vol.
175, No. 11, 2005, pp. 7311-7324.
[174] M. G. Toscano, M. Delgado, W. Kong, F. Martin, M.
Skarica and D. Ganea, “Dendritic Cells Transduced with
Lentiviral Vectors Expressing VIP Differentiate into
VIP-Secreting Tolerogenic-Like DCs,” Molecular Ther-
apy, Vol. 18, No. 5, 2010, pp. 1035-1045.
doi:10.1038/mt.2009.293
[175] S. L. Bailey, P. A. Carpentier, E. J. McMahon, W. S.
Begolka and S. D. Miller, “Innate and Adaptive Immune
Responses of the Central Nervous System,” Critical Re-
views in Immunology, Vol. 26, No. 2, 2006, pp. 149-188.
[176] S. L. Bailey-Bucktrout, S. C. Caulkins, G. Goings, J. A.
Fischer, A. Dzionek and S. D. Miller, “Cutting Edge:
Central Nervous System Plasmacytoid Dendritic Cells
Regulate the Severity of Relapsing Experimental Auto-
immune Encephalomyelitis,” The Journal of Immunology,
Vol. 180, No. 10, 2008, pp. 6457-6461.
[177] M. Irla, N. Kupfer, T. Suter, R. Lissilaa, M. Benkhoucha,
J. Skupsky, P. H. Lalive, A. Fontana, W. Reith and S.
Hugues, “MHC Class II-Restricted Antigen Presentation
by Plasmacytoid Dendritic Cells Inhibits T Cell-Mediated
Autoimmunity,” The Journal of Experimental Medicine,
Vol. 207, No. 9, 2010, pp. 1891-1905.
doi:10.1084/jem.20092627
[178] Y. Yan, G. X. Zhang, B. Gran, F. Fallarino, S. Yu, H. Li,
M. L. Cullimore, A. Rostami and H. Xu, “IDO Upregu-
lates Regulatory T Cells via Tryptophan Catabolite and
Suppresses Encephalitogenic T Cell Responses in Ex-
perimental Autoimmune Encephalomyelitis,” The Journal
of Immunology, Vol. 185, No. 10, 2010, pp. 5953-5961.
doi:10.4049/jimmunol.1001628