Вы находитесь на странице: 1из 14

REVIEWS

Immunopathology of multiple sclerosis


Calliope A.Dendrou1, Lars Fugger1,2 and Manuel A.Friese3
Abstract | Two decades of clinical experience with immunomodulatory treatments for
multiple sclerosis point to distinct immunological pathways that drive disease relapses and
progression. In light of this, we discuss our current understanding of multiple sclerosis
immunopathology, evaluate long-standing hypotheses regarding the role of the immune
system in the disease and delineate key questions that are still unanswered. Recent and
anticipated advances in the field of immunology, and the increasing recognition of
inflammation as an important component of neurodegeneration, are shaping our
conceptualization of disease pathophysiology, and we explore the potential implications
for improved healthcare provision to patients in the future.

Approximately 2.5 million people worldwide are afflicted symptoms and the development of other autoimmune
Demyelination
Damage to the myelin sheath
with multiple sclerosis, a chronic neuroinflammatory disorders to malignancies and even fatal opportun-
surrounding nerves in the brain disease of the brain and spinal cord that is a common istic infections such as progressive multifocal leuko
and spinal cord, which affects cause of serious physical disability in young adults1, encephalopathy 4 (see Supplementary information S1
the function of the nerves especially women. Multiple sclerosis poses a major per- (table)), indicating the need to identify more specific
involved. Demyelination occurs
sonal and socioeconomic burden: the average age of therapeutic targets that can be efficaciously modulated
in multiple sclerosis and in
experimental autoimmune disease onset is 30years a time that is decisive for but without inducing such significant adverse reactions.
encephalomyelitis, which is work and family planning and 25years after diag- Concomitantly, it has been increasingly acknowl-
an animal model of multiple nosis, approximately 50% of patients require permanent edged that although the long-standing treatments
sclerosis.
use of a wheelchair. The condition has a heterogeneous approved for multiple sclerosis can reduce relapses, they
presentation (BOX1) that can include sensory and visual do not substantially halt the disease4,5, and neuroaxonal
disturbances, motor impairments, fatigue, pain and damage with ensuing physical disability continues
cognitive deficits1. The variation in clinical manifesta- to accumulate and become permanent. This supports
1
Nuffield Department of
Clinical Neurosciences,
tions correlates with the spatiotemporal dissemination the concept that there is some degree of discord between
Division of Clinical Neurology of lesional sites of pathology within the central nervous the processes driving overt relapses and those driving
and MRC Human Immunology system (CNS)2. These lesions are a hallmark of multiple chronic progression. Indeed, secondary progressive
Unit, Weatherall Institute of sclerosis (BOX2) and are caused by immune cell infiltra- disease may not be a temporally distinct phase of the
Molecular Medicine,
tion across the bloodbrain barrier (BBB) that promotes condition arising as a direct consequence of the relaps-
John Radcliffe Hospital,
University of Oxford, inflammation, demyelination, gliosis and neuroaxonal ing-remitting disease but may instead be the outcome of
Oxford OX3 9DS, UK. degeneration, leading to disruption of neuronal signal- other underlying pathophysiological mechanisms. This
2
Clinical Institute, ling 3. Tcells appear early in lesion formation, and the is also in keeping with the existence of the relapse-free,
Aarhus University Hospital, disease is considered to be autoimmune, initiated by primary progressive form of multiple sclerosis (BOX1).
DK8200 Aarhus, Denmark.
3
Institut fr
autoreactive lymphocytes that mount aberrant responses The inferred uncoupling of relapses and disability
Neuroimmunologie und against CNS autoantigens, the precise nature of which, progression has considerable ramifications for our
Multiple Sklerose, however, remains enigmatic. understanding of disease pathways and for therapeu-
Zentrum fr Molekulare Infiltration of immune cells from the periphery tic design, as there are currently no drugs approved
Neurobiologie Hamburg,
which is particularly prominent in the common, to specifically treat primary or secondary progressive
Universittsklinikum
Hamburg-Eppendorf, relapsing-remitting form of the disease has been the multiple sclerosis5. Although disease progression is not
20246 Hamburg, Germany. main target of currently available therapies for multiple greatly influenced by the available immunomodulatory
Correspondence to L.F. sclerosis (see Supplementary information S1 (table)). therapies, which target peripheral immune cell activa-
email: Although these broad-spectrum immunomodulatory tion and entry into the CNS, immunological involve-
lars.fugger@imm.ox.ac.uk
doi:10.1038/nri3871
drugs reduce immune cell activity and entry into the ment is implicated in this process: there is an additional
Published online CNS and decrease relapse frequency, they are often inflammatory component residing in the CNS that is
7 August 2015 associated with side effects. These range from flu-like only marginally influenced by peripheral immune

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | SEPTEMBER 2015 | 545

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Gliosis control and that contributes to gradual neuroaxonal Dissecting the distinct roles of the immune system
The proliferation and activation loss and demise of myelin-producing oligodendro- in the events that trigger multiple sclerosis develop-
of glial cells (microglia, cytes68. This CNS-resident inflammatory arm of the ment and those that contribute to disease progression is
oligodendrocytes and disease is less well defined but is likely to involve con- thus complicated by the multicellular pathophysiology
astrocytes) in response to
damage in the central nervous
tinuous activation of innate immune cells; these cells associated with infiltrating adaptive and innate immune
system. have been found to predominate in demyelinated areas, cells, as well as CNS-resident innate immune cells with
but they are also present diffusely throughout normal- inflammatory capacity and by the chronic nature of the
appearing white matter, and their numbers correlate disease that unfolds over a period of many decades.
with tissue damage9. In this Review, we evaluate how our understanding
of the involvement of the immune system in driving
the development of multiple sclerosis is being shaped
Box 1 | The heterogeneity of multiple sclerosis by the ongoing interrogation of genetic predisposition
and environmental influences. We discuss the changing
The disease course and symptomatology of multiple sclerosis are heterogeneous,
role of peripheral immune cells including effector and
although efforts to categorize patients by general patterns of disease presentation
have allowed several disease subtypes to be recognized. The most common form, regulatory lymphocytes and innate immune cells in
affecting approximately 85% of patients, is relapsing-remitting multiple sclerosis. promoting pathogenesis as the disease takes its course,
It is characterized by an initial episode of neurological dysfunction (clinically isolated and we point to CNS-resident innate cells as emerging
syndrome), followed by a remission period of clinical recovery and then recurring bouts key contributors to chronic inflammation. In consid-
of relapse and remission (see the figure; black line). Relapses coincide with focal central ering our current view of multiple sclerosis immuno
nervous system (CNS) inflammation and demyelination that are typically discernible, pathology, we highlight the outstanding clinical needs
using magnetic resonance imaging, as white matter lesions. Eventually, improvement and the imminent biomedical challenges for thefuture.
during each remission wanes as disability accumulates, and approximately 80% of
patients go on to develop secondary progressive multiple sclerosis, one to two decades
What causes multiple sclerosis?
post diagnosis. In secondary progressive disease, inflammatory lesions are no longer
The exact cause of multiple sclerosis, and whether this
characteristic, and progressive neurological decline is instead accompanied by CNS
atrophy; that is, decreased brain volume and increased axonal loss (see the figure; red varies from one patient to the next, still remains elusive,
solid and dashed lines, respectively). Approximately 10% of patients with multiple but the disease is thought to arise in genetically suscep-
sclerosis are diagnosed with primary progressive disease, which features progressive tible individuals, with stochastic events and environ-
decline from the outset and an absence of relapses (see the figure; blue line). mental factors influencing disease penetrance. Genetic
No significant geographical differences have been reported in the relative prevalence variation accounts for approximately 30% of the over-
of the three main disease subtypes1. In Asian populations, however, a different form of all disease risk, and with the advent of genome-wide
relapsing-remitting disease known as opticospinal multiple sclerosis is thought to association studies (GWASs), more than 100 distinct
be common, although it is debated whether this disease should in fact be classified as a genetic regions have been identified as being associ-
neuromyelitis optica spectrum disorder rather than a form of multiple sclerosis112. Rarer
ated with multiple sclerosis, collectively explaining
variations of multiple sclerosis have also been reported1, such as progressive-relapsing
approximately one-third of the genetic component of
and paediatric disease, as well as tumefactive multiple sclerosis, which includes the
particularly severe Marburg variant. the condition10. Despite the fact that non-genetic fac-
As the characterization of multiple sclerosis heterogeneity improves, a key challenge is tors have a proportionately larger contribution than
to determine whether or not differing disease presentations can truly be classified as a genetic factors to immunological heterogeneity in
single disease, and what implications diagnostic accuracy will have on the understanding general11, comparatively less progress has been made
of shared and distinct pathophysiological mechanisms and on therapeutic targeting. in elucidating environmental determinants of multiple
sclerosis, perhaps reflecting the difficulty of accurately
interpreting complex, and sometimes confounding,
Secondary progressive disease epidemiologicaldata12.
Relapsing-remitting disease Primary Without a known predominant exogenous risk fac-
Pre-symptomatic progressive tor, it is an open question whether multiple sclerosis is
disease disease
triggered in the periphery or in the CNS. In the CNS-
extrinsic (peripheral) model, autoreactive Tcells that
are activated at peripheral sites potentially through
Disability

Clinically isolated
syndrome molecular mimicry 1315, bystander activation or the
Clinical threshold
coexpression of Tcell receptors (TCRs) with different
specificities16 traffic to the CNS along with activated
Bcells and monocytes (FIG.1). This model is consistent
with the method used to induce the multiple sclerosis-
like disease experimental autoimmune encephalo-
myelitis (EAE) in animals: emulsified CNS antigen is
administered along with immune stimulants, resulting
30 40 Average age (years) in the generation of pathogenic CD4+ T helper 1 (TH1)
cells and TH17 cells in the draining lymph nodes. These
Inammatory Neurological cells then enter the circulation and ultimately exert their
Axonal loss
relapses dysfunction
effector functions within the CNS, having crossed the
Underlying Primary BBB or the bloodcerebrospinal fluid (CSF) barrier at
Brain volume
disease progression progressive disease
the choroid plexus (BOX3).

Nature Reviews | Immunology


546 | SEPTEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Tumefactive multiple Alternatively, CNS-intrinsic events may trigger dis- Genetic predisposition. The majority of multiple sclerosis-
sclerosis ease development, with the infiltration of autoreactive associated candidate genes are thought to be immuno-
A subtype of multiple sclerosis lymphocytes occurring as a secondary phenomenon. logical. Consequently, the notable overlap in associ-
characterized by atypical, It is unclear what these CNS-intrinsic events might be, ated genomic regions between multiple sclerosis and
large demyelinated lesions
that appear tumour-like and
although postulated mechanisms include inflammatory other autoimmune diseases is not surprising 10 and may
oedematous and can exert responses to an as yet unknown CNS viral infection suggest some sharing of predisposing immunological
pressure on the surrounding a hypothesis based partly on the emerging apprecia- processes, thereby supporting the peripheral model of
central nervous system tissue tion of CNS immune surveillance17,18 (BOX3) or to multiple sclerosis initiation. However, in some cases
due to their size.
processes leading to primary neurodegeneration, similar this is only an apparent overlap: for instance, the same
Molecular mimicry to those that have been implicated in Alzheimer dis- variant in the gene region encoding tumour necro-
A mechanism by which a ease or Parkinson disease19. However, drawing support sis factor receptor 1 (TNFR1) confers susceptibility
peptide from a foreign antigen for either model of multiple sclerosis aetiology from to multiple sclerosis but promotes protection against
that is presented to a Tcell other diseases warrants a closer consideration of how ankylosing spondylitis, consistent with the opposing
closely resembles part of a
self-protein, thereby triggering
known multiple sclerosis risk factors compare to those effects of drugs targeting the TNFR1 pathway, which
an autoimmune reaction. for other common autoimmune and neurodegenerative exacerbate multiple sclerosis relapses but show efficacy
conditions. in ankylosing spondylitis20. Despite this caveat, efforts
Choroid plexus
The site of production of
cerebrospinal fluid in the
adult brain. It is formed by Box 2 | The pathology of multiple sclerosis
invagination of ependymal cells
Multiple sclerosis pathology is characterized by confluent demyelinated areas in the white and grey matter of the brain
into the ventricles, which
become highly vascularized.
and spinal cord that are called plaques or lesions and that indicate a loss of myelin sheaths and oligodendrocytes (see the
figure). Although axons and neurons are mostly preserved in early multiple sclerosis, ongoing disease results in gradual
Primary neurodegeneration neuroaxonal loss that correlates with patient disability, and the brain atrophy that occurs is accompanied by ventricular
The process of progressive enlargement (see the coronal view in the figure). Astrocytes form multiple sclerotic glial scars in white matter lesions.
dysfunction and loss of axons Demyelinated areas in the white matter can be partially repaired by remyelination. Demyelination is also found in the
and neurons, triggered by grey matter of the cortex, nuclei and spinal cord9.
mechanisms involving central Inflammation is present at all stages of multiple sclerosis, but it is more pronounced in acute phases than in chronic
nervous system-resident cells,
phases. Early lesions show invading peripheral immune cells and leakage of the bloodbrain barrier. Macrophages
as opposed to cells infiltrating
dominate the infiltrate, followed by CD8+ Tcells, whereas lower numbers of CD4+ Tcells, Bcells and plasma cells can also
from the periphery.
be found. In early disease, there is little overt damage to the brain and spinal cord in the areas outside the focal lesions,
Candidate genes termed normal-appearing white matter, although general brain atrophy has been noted104.
Genes assumed to be affected As the disease continues, diffuse inflammatory T cell and Bcell infiltrates, microglia and astrocyte activation,
by disease-associated genetic and diffuse myelin reduction and axonal injury are evident. This results in a more pronounced atrophy of the grey and
polymorphisms, based on their white matter9. Although the Tcell composition of infiltrates does not differ as the disease develops, the relative
functional relevance and/or proportion of Bcells and plasma cells increases3. Microglia and macrophages remain in a chronic state of
their physical proximity to the activation throughout the disease113.
polymorphisms in question.
Eventually the inflammation becomes organized inside the central nervous system, with fewer invading cells observed in
The determination of whether
the lesions during progression. In secondary progressive disease, tertiary lymphoid structures have been found to form in the
the assigned candidates are
truly affected by the
meninges, and these inflammatory aggregates may contribute to cortical demyelination and tissue injury at later stages71.
polymorphisms and how they
influence disease susceptibility
typically requires functional
followup investigations at the Relapsing-
molecular, cellular and Healthy remitting disease
systemic levels.
Cervical
Coronal brain view
Thoracic

Transverse Lumbar
spinal cord view
White Grey Demyelinated
matter matter lesions

Relapsing-
Healthy remitting disease Progressive disease
Atrophy at site
Grey of prior lesion
matter Demyelination in
deep grey matter

Ventricle
White White matter Cortical Ventricular enlargement
matter demyelination demyelination with increasing atrophy

Nature Reviews | Immunology


NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | SEPTEMBER 2015 | 547

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

to obtain a more comprehensive interpretation of the These findings are consistent with pre-GWAS con-
genetic data have led to the construction of interactome cepts regarding immunological mechanisms in multiple
networks21 using the presumed candidate genes assigned sclerosis, but the more substantial use of GWAS data to
to each associated region. For multiple sclerosis, such dissect disease pathways requires more indepth inves-
analyses implicate the involvement of interleukin2 tigations. Epigenetic23,24, transcriptomic25 and immuno-
(IL2), interferons (IFNs) and nuclear factorB signal- profiling 26,27 analyses are just beginning to shed light
ling, among numerous other immunological pathways, on how the variants correlate with immune cell subset-
in disease predisposition22. specific differences in the regulation of gene expression,

Periphery Blood CNS


Thymus Autoreactive T cells can circulation
escape central tolerance and
be released into the periphery

Lymph node Bidirectional


exchange
BCR

Peripheral tolerance TCR


breakdown due to TReg cell
defects and/or eector cell Memory
resistance may contribute to CD4+ T cell B cell B cell
multiple sclerosis development
CD8+ T cell 41 integrin
TReg cell
Mucosal surface Inltration
TH1 cell
Autoreactive
Peptide CD4+ T cell
Macrophage
MHC
TH17 cell
Bacteria
Goblet cell CD8+
MAIT cell Monocyte

Activated innate
Viruses immune cell
Autoreactive adaptive immune
cells can be activated by molecular
Smoke mimicry, novel autoantigen, release
constituents of sequestered CNS antigen or
stimulated by bystander activation

Thymic egress

Peripheral homeostatic proliferation

Exposure to environmental factors

Time
Figure 1 | Immune system dysregulation outside the CNS. During the sequestered CNS antigen released into the periphery or bystander
establishment of central tolerance in the thymus, most autoreactive Tcells activation. Genetic and environmental factors,Nature
including infectious
Reviews agents
| Immunology
are deleted; however, this process is imperfect, and some autoreactive and smoke constituents, contribute to these events. Once activated,
Tcells are released into the periphery. In health, peripheral tolerance CD8+ Tcells, differentiated CD4+ T helper 1 (TH1) and TH17 cells, Bcells
mechanisms keep these cells in check. If this tolerance is broken and innate immune cells can infiltrate the CNS, leading to inflammation and
through the reduced function of regulatory T (TReg) cells and/or the increased tissue damage. Bcells trafficking out of the CNS can undergo affinity
resistance of effector B cells and Tcells to suppressive mechanisms maturation in the lymph nodes before reentering the target organ
central nervous system (CNS)-directed autoreactive Bcells and Tcells can and promoting further damage. Dashed arrows indicate differentiation.
be activated in the periphery to become aggressive effector cells by BCR, Bcell receptor; CD8+ MAIT cell, CD8+ mucosa-associated invariant
molecular mimicry, novel autoantigen presentation, recognition of Tcell; TCR, Tcell receptor.

548 | SEPTEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Interactome networks as most associated genetic variants are non-coding and HLADRB1*15:01 variants28,29, and the genes encoding
Maps of molecular interactions, many colocalize with gene enhancers or repressors in the chains of the IL2 and IL7 receptors (IL2R and
often segregated by cell type, immune cells23. However, correlations do not necessar- IL7R, respectively)3033. The data implicate central
and used as a framework to ily reflect causality. To date, a more detailed, but not tolerance mechanisms, as well as peripheral differences
simplify cellular organization
and to help address systems
definitive, understanding of genetically determined in effector Tcell function due to altered cytokine
biology questions at the disease pathways has been attained for only a hand- responsiveness, cytokine production and homeostatic
cellular level. These networks ful of associated loci, such as the HLAA*02:01 and proliferation, in multiple sclerosis predisposition.
may reflect sets of physical
intermolecular interactions as
well as other molecules that
indirectly act together in Box 3 | Breaching CNS barriers: peripheral attack, a CSF Trojan horse or an inflammatory uprising?
specific pathways.
The central nervous system (CNS) has been considered to be an immune-privileged site: administration of immunogenic
Central tolerance agents into the CNS parenchyma typically fails to elicit an adaptive immune response. This has previously been attributed
Self-tolerance that is created to the presence of endothelial and epithelial barriers that restrict leukocyte trafficking (including the bloodbrain barrier
at the level of the central (BBB), and the bloodcerebrospinal fluid (CSF) and bloodleptomeningeal barriers114), the parenchymal anti-inflammatory
lymphoid organs. Developing milieu and the anatomical isolation of the CNS from the lymphatic system. A recent study in mice, however, provides
Tcells (in the thymus) and initial evidence for CNS lymphatic vessels lining the dural sinuses, suggesting that the dogma regarding the lack of
Bcells (in the bone marrow)
anatomical connectivity between the CNS and lymphatic system may require re-evaluation18, and warrants further
that strongly recognize
investigation in humans.
self-antigen must undergo
further rearrangement of
The relatively immune-privileged status of the CNS has been considered as a proponent for the peripheral initiation
antigen-receptor genes to of adaptive immune responses against CNS antigens with ensuing CNS barrier infiltration (see part a in the figure),
become self-tolerant, or they but even in the healthy CNS, memory Tcells traffic through the CSF, indicating a capacity for CNS-intrinsic immune
face deletion. surveillance17,114. The isolation of the CNS from the adaptive immune system is not absolute, thereby providing some
potential for the initiation of an adaptive immune response in the CNS in the absence of a BBB breach. Consistent with
Immune-privileged site CNS immune surveillance, blood-derived innate immune cells in the meningeal, perivascular and ventricular spaces are
An area in the body with a appropriately localized to present antigen to Tcells patrolling through the CSF (see part b in the figure), and they may
decreased immune response
activate CNS antigen-restricted Tcells under highly inflammatory conditions52.
to foreign antigens, including
Behind the BBB, the CNS parenchyma contains innate, yolk sac-derived microglial cells115 (see part c in the figure).
tissue grafts. These sites
include the brain, eye, testis
These tissue-resident macrophages have diverse pro-inflammatory and anti-inflammatory roles, although their
and placenta. primary function is not antigen presentation. Notably, microglial cells are implicated in the pathophysiological
chronic, low-grade inflammation that is associated with Alzheimer disease19, and thus they may have a similar
Dural sinuses role in multiple sclerosis.
Venous channels located
between layers of the brain b CSF Trojan horse: activation of
dura mater. These sinuses patrolling CSF lymphocytes
receive blood from both CNS interstital uid drains to
internal and external brain the CSF or to the circulation
veins, and cerebrospinal fluid via arachnoid villi
from the subarachnoid space,
and empty into the jugular Dura mater
vein. a Peripheral attack: immune cell Blood CSF
inltration from the periphery

T cell Lymphocyte
activation

Ventricle

From the
Macrophage CNS parenchyma
periphery
or dendritic Choroid
cell Skull
plexus
c Inammatory uprising:
Subarachnoid CNS-resident innate cells
space with inammatory capacity
Cribriform plate
Aerent
lymphatic

Cervical
lymph node
CSF to
CNS-derived soluble antigens spinal cord Microglial Astrocyte
are carried in the CSF across cell
the cribriform plate to the
nasal mucosa and then drain
to cervical lymph nodes

Nature Reviews | Immunology


NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | SEPTEMBER 2015 | 549

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Although still limited, the present view regarding the pathogen-associated molecular patterns38,39, and pro-
functional implications of multiple sclerosis-associated inflammatory cytokines at high concentrations in the
genetic polymorphisms is that the HLA variants primar- circulation can be transported across the BBB and can
ily define the CNS specificity of the disease by affect- induce signalling in perivascular macrophages40. The
ing the Tcell repertoire, whereas the non-HLA variants outcome of this immune system-toCNS communica-
more broadly influence the threshold of immune cell tion seems to typically involve the pro-inflammatory
activation, thereby ultimately altering the likelihood of activation of microglial cells. This raises the provocative
a CNS-directed autoimmune response being mounted. question of whether, in some cases, multiple sclerosis
Overall, strikingly few genetic associations are shared can result indirectly from peripheral inflammation that
between multiple sclerosis and other neurodegenerative drives microglia-dependent neurodegeneration, without
conditions such as Alzheimer disease34 and Parkinson the need for a CNS-directed autoreactive response to be
disease35. This indicates that non-immunological, pri- mounted.
mary neurodegenerative processes are less likely to Considering that the numerous non-HLA genetic
promote the initiation of multiple sclerosis, although risk factors for multiple sclerosis probably affect a multi-
genetic contributions to disease severity or subpheno- tude of immunological pathways, environmental factors
types of the disease may yet reveal a role for neurological that influence any one of these different pathways may
genes. Intriguingly, however, disease risk associations in also contribute to disease development. Based on this
the HLA region have also been observed for the other parallel, there may be just as many different environmen-
neurodegenerative conditions34,35, even though Tcell tal determinants of multiple sclerosis as there are genetic
infiltration is uncharacteristic for these diseases, and risk variants. To date, the reported environmental fac-
thus further investigation is needed to determine the tors implicated in multiple sclerosis variably, but not
significance of these findings. exclusively, include vitaminD, human cytomegalovirus
The genetic architecture of multiple sclerosis empha- infection12 and circadian disruption41. However, smok-
sizes the prominent role of the immune system in ing and EpsteinBarr virus (EBV) infection remain the
disease predisposition. The clinical relevance of deter- best-confirmed environmental contributors12, although
mining the specific phenotypic consequences of multi- it is notable that the modest impact of their individual
ple sclerosis-associated polymorphisms has now begun effects on overall multiple sclerosis risk is comparable to
to be recognized20, and this plethora of variants can that of any single associated genetic variant.
serve as a platform for interrogating human immune There is robust evidence that high levels of EBV-
system diversity: to help to fine-tune our understand- specific antibodies correlate with increased multiple
ing of disease immunopathogenesis, to identify more sclerosis risk, as does a history of infectious mono-
targeted treatment approaches and to even uncover nucleosis12,42. Several mechanisms for the role of EBV
novel immunological pathways that can be harnessed infection in multiple sclerosis development have been
for therapeutic benefit. proposed. One hypothesis is that inadequate regula-
tion of latent EBV infection leads to viral reactivation
Environmental factors. In line with the perceived dis- in the CNS, resulting in EBV-transformed Bcells in the
tinct roles of multiple sclerosis genetic risk factors in meningeal and perivascular space expressing viral pro-
the direct triggering of autoreactivity and in the broader teins that could activate effector Tcells43. Furthermore,
modulation of thresholds of immune cell activation, chronic viral infection can lead to an increase in the
the environmental factors that contribute to disease numbers of virus-specific memory Tcells44, and this
development may also fall into two similar categories. increase may be accentuated in multiple sclerosis;
Those environmental factors more directly involved indeed, homeostatic peripheral Tcell proliferation in
in the triggering of autoreactive Tcells are often postu- response to an accelerated thymic involution has been
lated to be viral or microbial in nature and mediate their observed in patients with relapsing-remitting dis-
effects through molecular mimicry 1315. Tolerance break- ease45. However, there is conflicting evidence regarding
down may also arise through the environmental factor- whether EBV RNA or protein is present in the CNS of
driven generation of novel autoantigens36. In addition patients with multiple sclerosis43,46, and this hypothesis
to directly providing or modifying relevant antigens, thus remains controversial. A second hypothesis sug-
environmental determinants such as CNS-tropic infec- gests that EBV may instead have a more general role
tious agents may also promote the release of sequestered in immune system dysregulation, which is in keeping
CNS antigens into the periphery, as has been observed with the correlation of EBV infection with the risk
in a model of Theilers murine encephalomyelitis virus of developing other autoimmune diseases, such as
infection14,37 (FIG.1). systemic lupus erythematosus14.
Environmental influences with a more modula- As the vastness of the human virome is just beginning
tory role may indirectly alter the activation thresholds to be appreciated47, our understanding of viral involve-
of autoreactive Tcells by triggering a pro-inflamma- ment in multiple sclerosis is still in its infancy. This is
tory milieu. Intriguingly, peripheral inflammation due equally true for the bacterial microbiome, the genome of
to infection may also have a direct influence on the which is approximately 100times larger than the human
CNS: locally secreted cytokines can activate afferent genome, and which fluctuates in composition based on
nerve endings, circumventricular organ and choroid environmental factors such as diet 48. EAE studies have
plexus innate immune cells can respond to circulating demonstrated that changes to the gut microbiota, for

550 | SEPTEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Epitope spreading example, can alter the incidence and severity of CNS been demonstrated to be recognized by circulating
This term is used to describe inflammation and ensuing disease49. However, a direct CD4+ Tcells in patients with multiple sclerosis but also
how a self-directed immune link between the microbiota and multiple sclerosis in in healthy individuals, and there is conflicting evidence
response induced by a single humans has yet to be demonstrated. regarding potential differences in the frequency and
peptide (or epitope) could
spread to include other
Although identifying the many environmental factors avidity of these cells between the two groups50,51. This
peptides (or epitopes) not only that may alter multiple sclerosis risk and comprehend- controversy, as well as the absence of a dominant Tcell
on the same autoantigen ing their mode of action poses a particularly significant autoantigen in multiple sclerosis, may be attributed to
(intramolecular spreading) but challenge, the putative ease of modifying exogenous technical limitations in detecting such autoantigens, to
also on other self-molecules
influences and human behaviour to reduce disease risk inter-patient variation, or to epitope spreading52,53, but
clustered in close vicinity within
the target cell (intermolecular
or severity is an attractive prospect for future medical unbiased combinatorial library screening methods54
spreading). intervention. and antigen-tolerizing approaches55 may help to further
elucidate anti-myelin immune responses in the disease.
Diapedesis Chronic multicellular disease development In EAE, infiltrating CD4+ Tcells are reactivated in
The migration of leukocytes
across the endothelium, which
The multifactorial nature of multiple sclerosis involving the CNS by antigen-presenting cells (APCs), including
occurs by leukocytes squeezing a potential deluge of different geneenvironment inter CD11c+ dendritic cells (DCs), with the resulting inflam-
through the junctions between actions at its inception unfolds through a complex, matory response leading to monocyte recruitment into
adjacent endothelial cells. highly multicellular pathophysiological process that evolves the CNS, as well as naive CD4+ Tcell activation through
throughout the duration of the disease course (FIGS1,2). epitope spreading that further fuels the inflammation52.
TH1 cells and TH17 cells are the main CD4+ Tcell sub-
Autoreactive Tcells. The presence of Tcells within CNS sets implicated in disease, and thus skewing of Tcell
lesions is detectable in the early stages of multiple sclero- differentiation away from these subsets and towards a
sis9, and the long-appreciated HLA associations with the TH2 cell phenotype has been a prominent therapeutic
disease are thought to reflect the presentation of specific concept and is considered to be a mechanism of action of
CNS autoantigens to autoreactive Tcells. As demyelina- the first-line, disease-modifying therapies IFN56, glati-
tion is a key feature of multiple sclerosis neuropathology, ramer acetate (Copaxone; Teva and SanofiAventis)57
myelin protein-derived antigens have been hypoth- and dimethyl fumarate (Tecfidera; Biogen)58.
esized to be the main autoreactive targets. Myelin basic However, the relative importance of TH1 cells versus
protein (MBP), proteolipid protein and myelin oligo TH17 cells in multiple sclerosis pathogenesis is conten-
dendrocyte glycoprotein (MOG), for example, have tious: conflicting studies variably report the predomi-
nance of one cell type over the other at initial diagnosis
and during subsequent relapses and progression59,60, and
Box 4 | The promises and pitfalls of animal models of multiple sclerosis compared with controls, patient myelin-reactive periph-
The induction of experimental autoimmune encephalomyelitis (EAE) can be performed eral CD4+ Tcells expressing CCchemokine receptor 6
in a range of different species, although rodents are most common and have greatly (CCR6) show enhanced expression of both the respec-
contributed to our understanding of autoimmunity and of inflammation-induced tive TH1 and TH17 cell signature cytokines IFN and
neurodegeneration. For example, the development of multiple sclerosis drug IL17A61. Furthermore, some lesional CD4+ Tcells have
natalizumab resulted directly from EAE experiments: natalizumab effectively an intermediate phenotype, expressing IFN and IL17A
blocked 41 integrin, which is involved in leukocyte adhesion and diapedesis at simultaneously 62. Despite these inconsistencies, the fail-
the bloodbrain barrier116. ure of a PhaseII clinical trial in patients with relapsing-
Next-generation transgenic and genetic engineering technologies, in combination remitting multiple sclerosis following the administration
with advances in imaging, for example, are now expanding the potential utility of
of ustekinumab (Stelara; Janssen)63 an antibody that
EAE. Thus, EAE will remain an essential tool for preclinical and mechanistic research,
enabling findings from simpler invitro systems such as stem cell-based cocultures targets the p40 subunit that is shared by the IL12 and
to be interrogated invivo. However, EAE is a reductive model that needs to be used IL23 cytokines, which are involved in TH1 and TH17
and interpreted with care. Several key aspects of the model have to be considered cell differentiation, respectively was not anticipated.
when translating EAE results to multiple sclerosis: Suggested explanations have included a putative inability
Disease induction: harsh induction regimens using adjuvants remain a major criticism. of the drug to cross the BBB and exert an effect directly
Despite genetically engineered mice that show spontaneous disease development in the CNS, and a diminished importance for IL12
(see Supplementary information S2 (table)), their use has remained limited. and/or IL23 at later stages of disease63. The very premise
Disease course: most mouse strains, including the commonly used C57BL/6 mice, for the ustekinumab trial, based partly on EAE studies,
show a monophasic disease course. However, other available strains can show relapsing- has also been questioned; although EAE models (see
remitting and more progressive disease courses (see Supplementary information S2 Supplementary information S2 (table)) are indispensa-
(table)) and need to be used in concordance with the scientific question. ble for studying disease mechanisms invivo, interspecies
Location of central nervous system inflammation: most EAE models show focused immunological differences have been recognized (BOX4),
inflammation in the spinal cord, whereas multiple sclerosis is usually dominated by including the essential requirement for IL23 in TH17 cell
brain inflammation. induction in mice but not in humans64,65. In addition, the
Immune cell infiltrate: due to the immunization regimen, Tcell responses are heavily function of TH17 cells seems to differ between mice and
biased towards CD4+ Tcells (see Supplementary information S2 (table)), whereas humans. TH17 cell-mediated granulocytemacrophage
CD8+ Tcell responses dominate in multiple sclerosis. colony-stimulating factor (GMCSF) production contrib-
Interspecies immune differences: genetic and phenotypic differences in the immune utes to chronic inflammation in EAE66, whereas TH1 cells
systems of mice and humans have been well documented and can have implications and other cell subsets are the primary producers of this
for the relevance of EAE findings to the human disease117.
cytokine in humans67.

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | SEPTEMBER 2015 | 551

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

CD8+ Tcells are found in higher frequency than In the absence of known autoantigens, the mecha-
CD4+ Tcells in the white matter and in grey matter cor- nisms controlling Bcell activation, selection and affinity
tical demyelinating lesions, and their numbers closely maturation have been a matter of speculation. However,
correlate with axonal damage3. Consistent with a key the recent application of next-generation sequencing
role for these cells in disease pathogenesis, myelin- technologies to analyse Bcell receptor diversity has
specific CD8+ Tcells are readily activated by epitope allowed for the characterization of Bcell clonotypes in
spreading, even in a CD4+ Tcell-driven EAE model, the peripheral compartments and the CSF of patients
with this being aided by antigen cross-presentation by with multiple sclerosis, and such studies indicate that
monocyte-derived DCs in the CNS 53. Non-myelin antigen-experienced Bcells can undergo maturation
astrocyte-derived antigen can also trigger spontane- in draining cervical lymph nodes before transmigra-
ous relapsing-remitting disease in mice by driving the tion to the CNS76,77. These data imply a therapeutic
establishment of non-recirculating resident memory- potential for the peripheral modulation of specific
like CD8+ Tcells within the CNS68. The disease course Bcell subtypes76,77. Currently, PhaseII clinical trials
and pathology in this model was modulated by Bcells have shown that CD20specific monoclonal antibodies
and by viral triggering, suggesting that complex multi- rituximab (MabThera; Roche)78 or ocrelizumab (Roche
Cross-presentation cellular and environmental interactions can contribute and Biogen)79 are efficacious in reducing relapse rates.
The initiation of a CD8+ Tcell to disease heterogeneity. In humans, up to a quarter of These drugs deplete the majority of Bcell subsets but
response to an antigen that
CD8+ Tcells in the active lesions of patients with mul- not autoantibody-producing terminally differentiated
is not present within
antigen-presenting cells (APCs). tiple sclerosis can produce IL17 and are thus thought plasma cells, and they may thus serve to effectively
This exogenous antigen must to be mucosa-associated invariant T cells (MAIT cells)69. reduce Bcell-mediated antigen presentation and other
be taken up by APCs and then Efficacious autologous haematopoietic stem cell trans- non-autoantibody-associated pathogenic contributions
rerouted to the MHC classI plantation in patients with highly active disease results such as pro-inflammatory IL6 production80.
pathway of antigen
presentation.
in the long-lasting depletion of these cells, suggesting
that they have an important role in disease patho Defective regulatory cells. The emergence and action of
Mucosa-associated genesis70. The precise contribution of CD8+ Tcells, autoreactive B cells and Tcells in multiple sclerosis may
invariant T cells compared with that of CD4 + Tcells and other cell be due to the defective functions of regulatory cells, such
(MAIT cells). A type of
types, following autologous haematopoietic stem cell as forkhead box P3 (FOXP3)-expressing CD4+ regulatory
CD8+ Tcell that is enriched
at mucosal sites and is transplantation and the relative importance of target- T (TReg) cells81 and IL10producing Tregulatory type1
characterized by the ing these cells in the therapeutic efficacy of broad- (TR1) cells82. Although few such cells are present in the
expression of a semi-invariant spectrum drugs such as natalizumab (Tysabri; Biogen CNS of patients83, disease-associated HLA classII vari-
Tcell receptor (a dimer of and Elan), alemtuzumab (Campath1H; Genzyme) ants could skew thymic selection such that the regulatory
V7.2 in combination with
J12, J20 or J33) and is
or fingolimod (FTY720 and Gilenya; Novartis)4 is Tcells that are released into the periphery inadequately
restricted by the not entirely clear and requires further study; however, suppress autoreactive effector Tcells81. Alternatively,
non-polymorphic, highly based on evidence published to date, the exploration of dysfunction of peripheral suppressor cells could be indi-
evolutionarily conserved CD8+ Tcell-specific therapies in the future is warranted. rectly driven by the dysregulation of tolerogenic APCs,
MHC classIb molecule, MR1.
as shown in EAE84. Non-HLA genetic associations, such
Tertiary lymphoid structures Autoreactive Bcells. Compared with Tcells, infiltrat- as variation in the BACH2 gene region10, may also be
Organized lymphocytic ing Bcell numbers in the CNS vary more throughout implicated in altering TReg cell function, as the transcrip-
aggregates that form in sites of disease progression. Clonally expanded Bcells can tion factor BACH2 has an essential role in the develop-
chronic inflammation. Typically, be found in the meninges, parenchyma and CSF, and ment of these cells85 and acts as a super-enhancer for Tcell
Bcell- and Tcell-rich zones are
segregated, and dendritic cells
intrathecal Bcells produce antibodies that are detect- identity 86. However, patients with immunodysregulation,
(DCs), germinal centres with able in the CSF and are of diagnostic value. Numbers of polyendocrinopathy, enteropathy, Xlinked syndrome
follicular DC networks and antibody-secreting plasma cells are increased with age in (IPEX), who have a FOXP3 deficiency, do not develop
specialized endothelial cells patients with primary or secondary progressive multiple CNS-directed autoimmunity 87, and therefore TReg cell
are present.
sclerosis3. The meninges of patients with secondary pro- dysfunction in patients with multiple sclerosis may be
Super-enhancer gressive disease often contain tertiary lymphoid structures an acquired rather than a primarydefect.
A cluster of regulatory of aggregated plasma cells, Bcells, Tcells and follicular Studies have variably reported a decreased fre-
elements within a genomic DCs (FDCs)71, which are a product of long-term inflam- quency and/or suppressive capacity of TReg cells88,89, as
region, often particularly mation as observed in other chronic inflammatory or well as an altered frequency of specific TReg cell subsets
enriched in sites that bind
transcriptional co-activators.
infectious diseases72. By contrast, primary progressive (such as CD39+ cells90), in the periphery of patients
disease is characterized by diffuse meningeal infiltra- with multiple sclerosis compared with controls. Such
Immunodysregulation, tion without such structures73. Despite initial reports that defects have been attributed to reduced frequencies
polyendocrinopathy, certain autoantigens including MOG, neurofascin, of naive circulating TReg cells of recent thymic origin
enteropathy, Xlinked
contactin and the ATP-dependent inwardly rectifying (identified as CD45RA+CD31+), along with the com-
syndrome
(IPEX). A disease caused by potassium channel KIR4.1 are recognized by patho- pensatory but ineffective expansion of the memory
mutations in FOXP3 (which genic Bcells in subgroups of patients, these findings still TReg cell population88. Another possibility is the skew-
encodes forkhead box P3) and await verification74. Moreover, other antibody-mediated ing of TReg cells towards an IFN-secreting TH1 celllike
characterized by refractory neurological diseases, such as myasthenia gravis, neuro phenotype in patients, which is reversible upon IFN
enteritis and, in some patients,
autoimmune endocrinopathies,
myelitis optica and autoimmune encephalitis, show a therapy 91. An alternative but non-mutually exclusive
autoimmune diabetes and clinical uniformity 75 that is absent in the subgroup of explanation for the action of autoreactive effector
thyroiditis. patients with antibody-positive multiple sclerosis. Tcells in multiple sclerosis is that, rather than being

552 | SEPTEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

the outcome of inadequate peripheral suppression, to this resistance 92,93. Such resistance mechanisms
the effector Tcells themselves are actively resistant emphasize the putative caveat of studies that document
to suppressive mechanisms, with the suggestion that patient TReg cell dysfunction using autologous effector
IL6induced signal transducer and activator of tran- Tcells, as these reports may in fact reflect increased
scription 3 (STAT3)-mediated signalling contributes effector Tcell resistance.

Box 5 | Key neurodegenerative processes as a consequence of chronic inflammation


Chronic inflammation in multiple sclerosis results in the production of reactive oxygen species (ROS) and reactive nitrogen
species (RNS) that probably promote mitochondrial injury as a result of the accumulation of detrimental mitochondrial
DNA mutations113,118,119 (see part a in the figure). This promotes metabolic stress, protein misfolding in the endoplasmic
reticulum (ER), energy deficiency and a loss of neuronal fitness, which have critical implications due to the fact that
transport along axons to maintain normal neuroaxonal function is highly energy demanding. Emphasizing the importance
of this cascade of events in disease progression, several single-gene mitochondrial and neurometabolic disorders present
as multiple sclerosis phenocopies based on certain clinical and radiographical features120.
Several different neuronal ion channels such as acid-sensing ion channel 1 (ASIC1), transient receptor potential cation
channel subfamily M member 4 (TRPM4) and voltage-gated sodium channels (Nav1.2 and Nav1.6) display a compensatory
redistribution along demyelinated neurons in multiple sclerosis to help to maintain ionic homeostasis6,7,110,121. However, this
redistribution, along with the excess accumulation of glutamate which is the main excitatory neurotransmitter in the
central nervous system and which is excessively released during neuronal injury promotes an ionic imbalance that only
serves to perpetuate tissue damage (see part b in the figure). From the initial site of axonal injury, these degenerative
mechanisms can spread backwards towards the neuronal cell body (termed retrograde degeneration or neuronal dying back)
or towards the distal axon terminal (termed anterograde degeneration or Wallerian degeneration) and can also influence
nearby presynaptic and postsynaptic neurons, respectively, eventually leading to neuronal apoptosis or necrosis.
A range of buffering mechanisms are triggered to counterbalance neuroaxonal injury, such as an upregulation of the
expression of pro-survival genes and the action of the cannabinoid system, but eventually the increasingly destructive
inflammatory milieu overrides them and fundamental neuroaxonal damage follows6. Therefore, although there has been
an interest in therapeutically boosting neuroprotective pathways as well as remyelination in multiple sclerosis, such
putative therapies would probably be most efficacious when administered along with anti-inflammatory agents.
a b
Nucleus DNA pH and ionic
damage imbalance
Ion H+
exchanger ATPase ASIC1
TRPM4
ER stress ROS
or RNS
Autophagy Damage
Translation Na+ Ca2+
Na+ Na
+
Ca2+
inhibition Ca2+ Caspases Proteolytic Cell
ER-associated Oxidative enzyme swelling
protein phosphorylation activation Ca2+ Ca2+ Na+
degradation
Death
receptor
Apoptosis Necrosis
Glutamate Nav
Voltage-dependent receptor channel
Ca2+ channel
Glutamate

Retrograde Anterograde Loss of


Nucleus degeneration degeneration neuronal
connectivity
Demyelination
Myelin
sheath

Mitochondrion Injured Node of


ER
axon Ranvier

Neuroaxonal damage promotes a decit in the highly Presynaptic energy failure and lipid
energy-demanding axonal transport processes, and this peroxidation driven by ROS and RNS can
decit in turns contributes to further energy deciency and lead to postsynaptic neuronal apoptosis
metabolic stress as mitochondria and other motor protein by promoting excessive postsynaptic
cargo are not transported to distal parts of the axon stimulation by neurotransmitters

Nature Reviews | Immunology


NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | SEPTEMBER 2015 | 553

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

In addition to CD4+ TReg cells, CD8+ regulatory Tcells Figure 2 | Immune system dysregulation inside the CNS
have been implicated in EAE94. These cells have also in early and late multiple sclerosis. Immune cell
been found in patients with multiple sclerosis in whom infiltration from the periphery is a prominent feature of
HLAErestricted CD8+ Tcells display a less regula- early-stage multiple sclerosis (top panel) and can occur
from the meningeal blood vessels by direct crossing of the
tory phenotype than those in healthy individuals95, and
bloodbrain barrier (denoted 1 in the figure) or the
neuroantigen-specific CD8+ Tcells may have less sup- subarachnoid space (SAS; denoted 2), or from the choroid
pressive capacity during relapses96. Enhanced cytotoxic plexus across the bloodcerebrospinal fluid (CSF) barrier
CD8+ regulatory Tcell function has also been observed (denoted 3). Peripheral innate and adaptive immune cells
in patients following glatiramer acetate therapy 97, and can accumulate in perivascular spaces and enter the
expansion of a putative regulatory CD103+CD8+ Tcell central nervous system (CNS) parenchyma. These cells,
subset has been reported in some patients treated with along with activated CNS-resident microglia and
natalizumab98. Drug administration may also influ- astrocytes, promote demyelination and oligodendrocyte
ence regulatory Bcells: IFN therapy correlates with (ODC) and neuroaxonal injury through direct cell
an increase in the numbers of IL10producing regula- contact-dependent mechanisms and through the action of
soluble inflammatory and neurotoxic mediators. Later on in
tory CD19+CD24hiCD38hi transitional Bcells in treated
the disease (bottom panel), immune cell infiltration wanes,
patients with multiple sclerosis99. Other regulatory Bcell perhaps due to adaptive immune cell exhaustion from
subsets, such as those secreting IL35, have also been chronic antigen exposure. However, chronic CNS-intrinsic
implicated in recovery fromEAE100. inflammation and neurodegeneration continue. Meningeal
Collectively, dysregulation of effectorregulatory cell tertiary lymphoid-like structures, which have specifically
interactions occurs in multiple sclerosis, ultimately result- been documented in secondary progressive disease, may
ing in the emergence of autoreactive adaptive immune contribute to late-stage inflammation in patients with this
cells that are capable of infiltrating and promoting dam- form of multiple sclerosis. The action of the CNS-resident
age within the CNS. The skewing of effectorregulatory innate cells may contribute to chronic inflammation
cell interactions may provide some therapeutic benefit irrespective of the precise disease subtype. Stimulated by
the microglia, astrocytes can produce CC-chemokine
but may not be sufficient to prevent neurodegeneration.
ligand 2 (CCL2) and granulocytemacrophage
colony-stimulating factor (GMCSF), leading to even
Inflammation in progressive neurodegeneration further microglial recruitment and activation, and the
As currently available immunomodulatory therapies astrocytes can prevent remyelination at sites of
decrease relapse rates but not necessarily long-term neuroaxonal injury by inhibiting progenitor cells from
multiple sclerosis progression, it has been suggested that developing into mature ODCs. APC, antigen-presenting
autoimmune response-instigated neuroaxonal injury cell; CD8+ MAIT cell, CD8+ mucosa-associated invariant
triggers a potentially self-sustaining chronic neuro Tcell; FDC, follicular dendritic cell; IFN, interferon;
degenerative process. This proceeds even in the absence IL17, interleukin17; NO, nitric oxide; RNS, reactive
of continued immune cell infiltration from the periphery, nitrogen species; ROS, reactive oxygen species;
TH1 cell, T helper 1 cell.
which eventually wanes regardless of therapy, possibly
due to immune cell exhaustion associated with chronic
antigenic exposure101. Although neurodegeneration in
multiple sclerosis is thought to be the culmination of of microglia and macrophages can be observed in lesions
a cascade of events occurring in axons and neurons and in the normal-appearing white matter 9,102. In addi-
including oxidative stress responses, energy deficiencies, tion, as neuroaxonal degeneration disseminates (BOX5),
ionic imbalances, and the failure of neuroprotective microglia in the vicinity of axons emanating from dis-
and regenerative mechanisms6 (BOX5) chronic CNS tally damaged neurons may become activated; these cells
inflammation may fuel these processes through the may hence serve as the nucleus of new lesion formation103
action of cells that have become or are already resident and might also contribute to the general brain atrophy
within the CNS (FIG.2). that is observed in early disease104. Notably, the relative
Previously infiltrating adaptive immune cells may con- role of microglia versus monocyte-derived macrophages
tribute to long-term inflammation in multiple sclerosis throughout the course of multiple sclerosis has not been
through the eventual establishment of tertiary lymphoid fully elucidated owing to the difficulty in distinguish-
structures within the CNS71. However, it is becoming ing these two morphologically and functionally similar
increasingly apparent that CNS-resident cells that sense celltypes.
homeostatic disturbances, mainly microglia and astro- Some insights have been gained from transgenic
cytes, can also produce a range of neurotoxic inflammatory EAE models, which have enabled these cell types to be
mediators (such as cytokines, chemokines and reactive studied in distinction: these studies have shown that
Exhaustion
oxygen species) that promote and sustain neuroaxonal at disease onset monocyte-derived macrophages ini-
Non-responsiveness of the
immune system resulting from damage and thus neurodegeneration6,19 (FIG.2). tiate demyelination, whereas microglia may be more
the deletion of specific Moreover, these cells are likely to have a role in mul- involved in debris clearing 105. This suggests that micro-
thymocytes (central tolerance) tiple sclerosis-associated CNS inflammation not only glia may have some neuroprotective capacity by helping
and the deletion or functional during the later stages of the disease when immune cell to resolve inflammation105, as well as by actively displac-
inactivation of specific Tcells
in the periphery (peripheral
infiltration from the periphery subsides but also from ing specific neuronal synapses to maintain CNS homeo-
tolerance) in the presence of the outset (FIG.2). Even after the very first manifestation stasis106 and by producing neurotrophic factors (such as
large quantities of antigen. of disease, increases in the numbers and activation status brain-derived neurotrophic factor) to aid the repair of

554 | SEPTEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Immune cell Early disease


inltration from
the periphery SAS
CNS parenchyma
Soluble
mediators
recruit
immune cells Soluble mediators
1
CD8+ promote inammation
T cell at distal sites

Perivascular DC
or macrophage Perivascular immune
T cell cell accumulation
reactivation
Myelin
ODC sheath
Antibody
Vessel Neuron
CD4+
activation
T cell CD8+ TH1 cell IFN Axon
MAIT IL-17 Degraded
cell myelin protein
TH17 cell
Monocyte GM-CSF
Demyelination
2

Phagocytosis Complement
proteins Ependyma
B cell Choroid
Plasma Microglial Capillary
Pericyte plexus
cell cell
Astrocyte
Choroid plexus
Meningeal macrophage
vessel 3
T cell reactivation by choroid CSF
plexus and meningeal APCs
Pial basement membrane Glia limitans
Time

Late disease

Meningeal tertiary
lymphoid-like structures
promote glia limitans damage
and astrocyte dysfunction

FDC
ODC progenitor

Clonally
expanded CCL2, Metabolic
B cells GM-CSF stress

Osteopontin,
NO Energy
deciency
Glutamate ROS,
accumulation RNS Ionic
imbalance

Neuroaxonal and
ODC damage
and death

Neurodegenerative
processes promote
further damage
at distal sites
Chronic CNS
inammation

Nature Reviews | Immunology


NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | SEPTEMBER 2015 | 555

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

neuroaxonal damage. Primary neurodegeneration in In multiple sclerosis, the additional inflammation


conditions such as Alzheimer disease is partly attributed induced and/or enhanced by peripheral immune cell
to inadequate neuroprotective microglial action19, and filtration and by CNS-resident innate-like immune
this functional failure may also be relevant to multiple cells may effectively contribute to the acceleration of
sclerosis development. the inevitable ageing processes in the CNS, and as the
Conversely, healthy neurons constitutively express subjugating burden of the stress response is too great for
inhibitors that block the phagocytic capacity of micro- homeostasis to be adequately maintained, pronounced
glia107, implying that if left unchecked, microglia may progressive neurodegenerative decline follows.
promote tissue injury, driving a feedback loop of pro-
gressive neuroaxonal damage. Moreover, specific trans- Concluding remarks
genic targeting of microglia has also been reported to Our understanding of multiple sclerosis immunopathol-
reduce EAE-associated CNS inflammation108. In addi- ogy has been consistently modified since the approval
tion, activated microglia can promote astrocyte dys- of the first immunomodulatory therapy for the condi-
function. Similarly to microglia, astrocytes can display tion, and therefore questions regarding the mechanisms
both pro-inflammatory and anti-inflammatory proper- underscoring the triggers and long-term development of
ties, and they have a crucial CNS barrier function by the disease remain to be definitively answered, although
forming the glia limitans that lines the neuronal tissue. these questions are now better defined.
Their dysfunction can permit and even facilitate periph- Perhaps most significantly, the appreciation of mul-
eral immune cell infiltration early in multiple sclerosis tiple sclerosis as the pathophysiological intersection
through the production of chemokines. Moreover, upon between interlinked but not entirely interdepend-
activation by stimulated microglia, astrocytes can pro- ent autoimmune and neurodegenerative processes
duce CCchemokine ligand 2 (CCL2) and GMCSF, has set imminent research challenges. There is a dire
leading to even further microglial recruitment and need to meaningfully integrate rapidly emerging
activation, and they can prevent remyelination at sites technologies and data with existing neuroimmuno-
of neuroaxonal injury by inhibiting the generation of logical clinical concepts in order to interrogate the
mature oligodendrocytes8 (FIG.2). Therefore, targeting multicellular interplay that unfolds within the CNS
pro-inflammatory mediators produced by astrocytes throughout disease progression. Underscoring this is
may serve to inhibit both peripheral immune cell infil- the requirement to further decompartmentalize the
tration and the continuous inflammation within the study of immunology and neurology in multiple scle-
CNS, and may thus be of therapeuticvalue. rosis through the investigation of neuroinflammation
The incompletely resolved role of CNS-resident as a whole in order to better delineate which inflam-
innate-like immune cells in multiple sclerosis immuno- matory and neurodegenerative mechanisms are truly
pathology in dampening down inflammation and/or distinct but occur in parallel and which are inextrica-
actively contributing to it may reflect our only partial bly associated, so as to aid the design of more effective
understanding of how the function of these cells varies therapeutic strategies.
across different regions of the CNS and throughout the A goal for future treatment of multiple sclerosis may
course of the disease. Notably, the pro-inflammatory thus be the simultaneous, early targeting of peripheral
action of CNS-resident innate-like immune cells in pro- immune cell function and of CNS-intrinsic inflam-
gressive neurodegeneration may be intrinsically linked mation, potentially through combinatorial therapies
to multiple sclerosis chronicity. Inflammation in the CNS designed to effectively and specifically modulate these
may be viewed as a stress response to maintain tissue two immunological arms of the disease, along with
homeostasis109 particularly as both the innate immune the provision of neuroprotective or neuroregenerative
and neuronal compartments of the CNS are specialized drugs. Improved disease prognosis and potential patient
to sense an array of stressors including ions, low pH110, stratification for more directed healthcare provision
temperature changes, hormones and cytokines and are also much-anticipated prospects and may become
even in the absence of disease, CNS-derived inflamma- tangible as we move into the immune informatics era
tory processes increase as a function of time, eventually and as large-scale, organized health resources become
promoting ageing-associated neurodegeneration111. increasingly accessible.

1. Compston,A. & Coles,A. Multiple sclerosis. 6. Friese,M.A., Schattling,B. & Fugger,L. Mechanisms of 11. Brodin,P. etal. Variation in the human immune
Lancet 372, 15021517 (2008). neurodegeneration and axonal dysfunction in multiple system is largely driven by non-heritable influences.
2. Kearney,H. etal. Cervical cord lesion load is sclerosis. Nat. Rev. Neurol. 10, 225238 (2014). Cell 160, 3747 (2015).
associated with disability independently from 7. Schattling,B. etal. TRPM4 cation channel mediates A comprehensive study of the contribution of
atrophy in MS. Neurology 84, 367373 (2015). axonal and neuronal degeneration in experimental non-genetic factors to the variation of more than
3. Frischer,J.M. etal. The relation between autoimmune encephalomyelitis and multiple sclerosis. 200 immunological parameters.
inflammation and neurodegeneration in multiple Nat. Med. 18, 18051811 (2012). 12. Belbasis,L., Bellou,V., Evangelou,E., Ioannidis,J.P.A.
sclerosis brains. Brain 132, 11751189 (2009). 8. Mayo,L. etal. Regulation of astrocyte activation & Tzoulaki,I. Environmental risk factors and multiple
4. Haghikia,A., Hohlfeld,R., Gold,R. & Fugger,L. by glycolipids drives chronic CNS inflammation. sclerosis: an umbrella review of systematic reviews and
Therapies for multiple sclerosis: translational Nat. Med. 20, 11471156 (2014). meta-analyses. Lancet Neurol. 14, 263273 (2015).
achievements and outstanding needs. 9. Popescu,B.F. & Lucchinetti,C.F. Pathology of 13. Harkiolaki,M. etal. Tcell-mediated autoimmune
Trends Mol. Med. 19, 309319 (2013). demyelinating diseases. Annu. Rev. Pathol. 7, disease due to low-affinity crossreactivity to common
5. Feinstein,A., Freeman,J. & Lo,A.C. Treatment of 185217 (2012). microbial peptides. Immunity 30, 348357 (2009).
progressive multiple sclerosis: what works, what does 10. Beecham,A.H. etal. Analysis of immune-related loci 14. Muenz,C., Luenemann,J.D., Getts,M.T. & Miller,S.D.
not, and what is needed. Lancet Neurol. 14, 194207 identifies 48 new susceptibility variants for multiple Antiviral immune responses: triggers of or triggered by
(2015). sclerosis. Nat. Genet. 45, 13531360 (2013). autoimmunity? Nat. Rev. Immunol. 9, 246258 (2009).

556 | SEPTEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

15. Olson,J.K., Croxford,J.L., Calenoff,M.A., 39. Vitkovic,L. etal. Cytokine signals propagate through 64. Ghoreschi,K. etal. Generation of pathogenic
Dal Canto,M.C. & Miller,S.D. A virus-induced the brain. Mol. Psychiatry 5, 604615 (2000). TH17 cells in the absence of TGF- signalling.
molecular mimicry model of multiple sclerosis. 40. Dantzer,R., OConnor,J.C., Freund,G.G., Nature 467, 967971 (2010).
J.Clin. Invest. 108, 311318 (2001). Johnson,R.W. & Kelley,K.W. From inflammation to 65. Zielinski,C.E. etal. Pathogen-induced human
16. Ji,Q., Perchellet,A. & Goverman,J.M. Viral infection sickness and depression: when the immune system TH17 cells produce IFN- or IL10 and are regulated
triggers central nervous system autoimmunity via subjugates the brain. Nat. Rev. Neurosci. 9, 4657 by IL1. Nature 484, 514518 (2012).
activation of CD8+ Tcells expressing dual TCRs. (2008). 66. Codarri,L. etal. RORt drives production of the
Nat. Immunol. 11, 628634 (2010). 41. Hedstrom,A.K., Akerstedt,T., Hillert,J., Olsson,T. & cytokine GMCSF in helper Tcells, which is essential for
17. Ransohoff,R.M. & Engelhardt,B. The anatomical and Alfredsson,L. Shift work at young age is associated the effector phase of autoimmune neuroinflammation.
cellular basis of immune surveillance in the central with increased risk for multiple sclerosis. Nat. Immunol. 12, 560567 (2011).
nervous system. Nat. Rev. Immunol. 12, 623635 Ann. Neurol. 70, 733741 (2011). 67. Noster,R. etal. IL17 and GMCSF expression are
(2012). 42. Operskalski,E.A., Visscher,B.R., Malmgren,R.M. & antagonistically regulated by human T helper cells.
18. Louveau,A. etal. Structural and functional features Detels,R. A case-control study of multiple sclerosis. Sci. Transl. Med. 6, 241ra280 (2014).
of central nervous system lymphatic vessels. Neurology 39, 825829 (1989). 68. Sasaki,K. etal. Relapsingremitting central nervous
Nature 523, 337341 (2015). 43. Lossius,A. etal. High-throughput sequencing of system autoimmunity mediated by GFAP-specific
19. Heneka,M.T., Kummer,M.P. & Latz,E. Innate TCR repertoires in multiple sclerosis reveals CD8 Tcells. J.Immunol. 192, 30293042 (2014).
immune activation in neurodegenerative disease. intrathecal enrichment of EBV-reactive CD8+ Tcells. 69. Willing,A. etal. CD8+ MAIT cells infiltrate into the
Nat. Rev. Immunol. 14, 463477 (2014). Eur. J.Immunol. 44, 34393452 (2014). CNS and alterations in their blood frequencies
20. Gregory,A.P. etal. TNF receptor 1 genetic risk 44. Snyder,C.M. etal. Memory inflation during chronic correlate with IL18 serum levels in multiple sclerosis.
mirrors outcome of anti-TNF therapy in multiple viral infection is maintained by continuous production Eur. J.Immunol. 44, 31193128 (2014).
sclerosis. Nature 488, 508511 (2012). of short-lived, functional Tcells. Immunity 29, 70. Abrahamsson,S.V. etal. Non-myeloablative
This study demonstrates how investigating the 650659 (2008). autologous haematopoietic stem cell transplantation
functional consequences of disease-associated 45. Duszczyszyn,D.A. etal. Thymic involution and expands regulatory cells and depletes IL17 producing
genetic variation can help to predict therapeutic proliferative Tcell responses in multiple sclerosis. mucosal-associated invariant Tcells in multiple
outcome. J.Neuroimmunol. 221, 7380 (2010). sclerosis. Brain 136, 28882903 (2013).
21. Menche,J. etal. Uncovering disease-disease 46. Sargsyan,S.A. etal. Absence of Epstein-Barr virus in 71. Howell,O.W. etal. Meningeal inflammation is
relationships through the incomplete interactome. the brain and CSF of patients with multiple sclerosis. widespread and linked to cortical pathology in
Science 347, 1257601 (2015). Neurology 74, 11271135 (2010). multiple sclerosis. Brain 134, 27552771 (2011).
22. Tasan,M. etal. Selecting causal genes from genome- 47. Virgin,H.W. The virome in mammalian physiology 72. Drayton,D.L., Liao,S., Mounzer,R.H. &
wide association studies via functionally coherent and disease. Cell 157, 142150 (2014). Ruddle,N.H. Lymphoid organ development: from
subnetworks. Nat. Methods 12, 154159 (2015). 48. Yatsunenko,T. etal. Human gut microbiome viewed ontogeny to neogenesis. Nat. Immunol. 7, 344353
23. Farh,K.K. etal. Genetic and epigenetic fine mapping across age and geography. Nature 486, 222227 (2006).
of causal autoimmune disease variants. Nature 518, (2012). 73. Choi,S.R. etal. Meningeal inflammation plays a role
337343 (2014). 49. Berer,K. etal. Commensal microbiota and myelin in the pathology of primary progressive multiple
24. Kundaje,A. etal. Integrative analysis of 111 reference autoantigen cooperate to trigger autoimmune sclerosis. Brain 135, 29252937 (2012).
human epigenomes. Nature 518, 317330 (2015). demyelination. Nature 479, 538541 (2011). 74. Brickshawana,A. etal. Investigation of the KIR4.1
25. Raj,T. etal. Polarization of the effects of autoimmune 50. Bielekova,B. etal. Expansion and functional potassium channel as a putative antigen in patients
and neurodegenerative risk alleles in leukocytes. relevance of high-avidity myelin-specific CD4+ Tcells with multiple sclerosis: a comparative study.
Science 344, 519523 (2014). in multiple sclerosis. J.Immunol. 172, 38933904 Lancet Neurol. 13, 795806 (2014).
26. Ye,C.J. etal. Intersection of population variation and (2004). 75. Leypoldt,F., Armangue,T. & Dalmau,J.
autoimmunity genetics in human Tcell activation. 51. Hellings,N. etal. Tcell reactivity to multiple myelin Autoimmune encephalopathies. Ann. NY Acad. Sci.
Science 345, 1254665 (2014). antigens in multiple sclerosis patients and healthy 1338, 94114 (2015).
27. Roederer,M. etal. The genetic architecture of controls. J.Neurosci. Res. 63, 290302 (2001). 76. Palanichamy,A. etal. Immunoglobulin class-switched
the human immune system: a bioresource for 52. McMahon,E.J., Bailey,S.L., Castenada,C.V., Bcells form an active immune axis between CNS
autoimmunity and disease pathogenesis. Waldner,H. & Miller,S.D. Epitope spreading initiates and periphery in multiple sclerosis. Sci. Transl. Med.
Cell 161, 387403 (2015). in the CNS in two mouse models of multiple sclerosis. 6, 248ra106 (2014).
An extensive immunophenotyping study that Nat. Med. 11, 335339 (2005). 77. Stern,J.N.H. etal. Bcells populating the multiple
provides a bioresource for linking genetic control 53. Ji,Q., Castelli,L. & Goverman,J.M. MHC classI sclerosis brain mature in the draining cervical lymph
elements associated with normal immunological restricted myelin epitopes are cross-presented by nodes. Sci. Transl. Med. 6, 248ra107 (2014).
traits to common autoimmune and infectious Tip-DCs that promote determinant spreading to References 76 and 77 use Bcell receptor
diseases. CD8+ Tcells. Nat. Immunol. 14, 254261 (2013). sequencing to investigate the relationship between
28. Friese,M.A. etal. Opposing effects of HLA classI 54. Siewert,K. etal. Unbiased identification of target Bcell subsets in the periphery and the CNS,
molecules in tuning autoreactive CD8+ Tcells in antigens of CD8+ Tcells with combinatorial libraries supporting the peripheral targeting of Bcells for
multiple sclerosis. Nat. Med. 14, 12271235 (2008). coding for short peptides. Nat. Med. 18, 824828 multiple sclerosis treatment.
29. Gregersen,J.W. etal. Functional epistasis on a (2012). 78. Hauser,S.L. etal. Bcell depletion with rituximab in
common MHC haplotype associated with multiple 55. Lutterotti,A. etal. Antigen-specific tolerance by relapsingremitting multiple sclerosis. N.Engl.
sclerosis. Nature 443, 574577 (2006). autologous myelin peptide-coupled cells: a phase1 J.Med. 358, 676688 (2008).
30. Hartmann,F.J. etal. Multiple sclerosis-associated trial in multiple sclerosis. Sci. Transl. Med. 5, 79. Kappos,L. etal. Ocrelizumab in relapsing-remitting
IL2RA polymorphism controls GMCSF production in 188ra175 (2013). multiple sclerosis: a phase2, randomised, placebo-
human TH cells. Nat. Commun. 5, 5056 (2014). 56. Kozovska,M.E. etal. Interferon beta induces controlled, multicentre trial. Lancet 378, 17791787
31. Dendrou,C.A. etal. Cell-specific protein phenotypes Thelper 2 immune deviation in MS. Neurology 53, (2011).
for the autoimmune locus IL2RA using a genotype- 16921697 (1999). 80. Barr,T.A. etal. Bcell depletion therapy ameliorates
selectable human bioresource. Nat. Genet. 41, 57. Miller,A. etal. Treatment of multiple sclerosis with autoimmune disease through ablation of IL6
10111015 (2009). copolymer1 (Copaxone): implicating mechanisms of producing Bcells. J.Exp. Med. 209, 10011010
32. Gregory,S.G. etal. Interleukin 7 receptor chain Th1 to Th2/Th3 immune-deviation. J.Neuroimmunol. (2012).
(IL7R) shows allelic and functional association with 92, 113121 (1998). 81. Venken,K. etal. Natural naive CD4+CD25+CD127low
multiple sclerosis. Nat. Genet. 39, 10831091 58. Zoghi,S. etal. Cytokine secretion pattern in treatment regulatory Tcell (Treg) development and function are
(2007). of lymphocytes of multiple sclerosis patients with disturbed in multiple sclerosis patients: recovery of
33. Lundstrom,W. etal. Soluble IL7R potentiates IL7 fumaric acid esters. Immunol. Invest. 40, 581596 memory Treg homeostasis during disease progression.
bioactivity and promotes autoimmunity. Proc. Natl (2011). J.Immunol. 180, 64116420 (2008).
Acad. Sci. USA 110, E1761E1770 (2013). 59. Frisullo,G. etal. IL17 and IFN production by 82. Martinez-Forero,I. etal. IL10 suppressor activity
34. Lambert,J.C. etal. Meta-analysis of 74,046 peripheral blood mononuclear cells from clinically and exvivo Tr1 cell function are impaired in multiple
individuals identifies 11 new susceptibility loci for isolated syndrome to secondary progressive multiple sclerosis. Eur. J.Immunol. 38, 576586 (2008).
Alzheimers disease. Nat. Genet. 45, 14521458 sclerosis. Cytokine 44, 2225 (2008). 83. Fritzsching,B. etal. Intracerebral human regulatory
(2013). 60. Tzartos,J.S. etal. Interleukin17 production in Tcells: Analysis of CD4+CD25+FOXP3+ Tcells in
35. Nalls,M.A. etal. Large-scale meta-analysis of central nervous system-infiltrating Tcells and glial brain lesions and cerebrospinal fluid of multiple
genome-wide association data identifies six new risk cells is associated with active disease in multiple sclerosis patients. PLoS ONE 6, e17988 (2011).
loci for Parkinsons disease. Nat. Genet. 46, 989993 sclerosis. Am. J.Pathol. 172, 146155 (2008). 84. Yogev,N. etal. Dendritic cells ameliorate
(2014). 61. Cao,Y. etal. Functional inflammatory profiles autoimmunity in the CNS by controlling the
36. Scally,S.W. etal. A molecular basis for the distinguish myelin-reactive Tcells from patients with homeostasis of PD1 receptor+ regulatory Tcells.
association of the HLADRB1 locus, citrullination, and multiple sclerosis. Sci. Transl. Med. 7, 287ra274 Immunity 37, 264275 (2012).
rheumatoid arthritis. J.Exp. Med. 210, 25692582 (2015). 85. Roychoudhuri,R. etal. BACH2 represses effector
(2013). 62. Kebir,H. etal. Preferential recruitment of interferon-- programs to stabilize Treg-mediated immune
37. Miller,S.D. etal. Persistent infection with Theilers expressing TH17 cells in multiple sclerosis. homeostasis. Nature 498, 506510 (2013).
virus leads to CNS autoimmunity via epitope Ann. Neurol. 66, 390402 (2009). 86. Vahedi,G. etal. Super-enhancers delineate disease-
spreading. Nat. Med. 3, 11331136 (1997). 63. Segal,B.M. etal. Repeated subcutaneous injections associated regulatory nodes in Tcells. Nature 520,
38. Quan,N., Whiteside,M. & Herkenham,M. Time of IL12/23 P40 neutralising antibody, ustekinumab, 558562 (2015).
course and localization patterns of interleukin1 in patients with relapsing-remitting multiple sclerosis: 87. Bennett,C.L. etal. The immune dysregulation,
messenger RNA expression in brain and pituitary a phaseII, double-blind, placebo-controlled, polyendocrinopathy, enteropathy, Xlinked syndrome
after peripheral administration of lipopolysaccharide. randomised, dose-ranging study. Lancet Neurol. (IPEX) is caused by mutations of FOXP3. Nat. Genet.
Neuroscience 83, 281293 (1998). 7, 796804 (2008). 27, 2021 (2001).

NATURE REVIEWS | IMMUNOLOGY VOLUME 15 | SEPTEMBER 2015 | 557

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

88. Venken,K. etal. Compromised CD4+CD25high 100. Shen,P. etal. IL35producing Bcells are critical 113. Fischer,M.T. etal. NADPH oxidase expression in
regulatory Tcell function in patients with relapsing- regulators of immunity during autoimmune and active multiple sclerosis lesions in relation to oxidative
remitting multiple sclerosis is correlated with a infectious diseases. Nature 507, 366370 (2014). tissue damage and mitochondrial injury. Brain 135,
reduced frequency of FOXP3positive cells and 101. Wherry,E.J. Tcell exhaustion. Nat. Immunol. 12, 886899 (2012).
reduced FOXP3 expression at the single-cell level. 492499 (2011). 114. Shechter,R., London,A. & Schwartz,M.
Immunology 123, 7989 (2008). 102. Giannetti,P. etal. Increased PK11195PET binding in Orchestrated leukocyte recruitment to immune-
89. Feger,U. etal. Increased frequency of CD4+CD25+ normal-appearing white matter in clinically isolated privileged sites: absolute barriers versus educational
regulatory Tcells in the cerebrospinal fluid but syndrome. Brain 138, 110119 (2015). gates. Nat. Rev. Immunol. 13, 206218 (2013).
not in the blood of multiple sclerosis patients. 103. Kolasinski,J. etal. A combined post-mortem magnetic 115. Perdiguero,E.G. etal. Tissue-resident macrophages
Clin. Exp. Immunol. 147, 412418 (2007). resonance imaging and quantitative histological originate from yolk-sac-derived erythro-myeloid
90. Fletcher,J.M. etal. CD39+Foxp3+ regulatory Tcells study of multiple sclerosis pathology. Brain 135, progenitors. Nature 518, 547551 (2015).
suppress pathogenic Th17 cells and are impaired in 29382951 (2012). 116. Yednock,T.A. etal. Prevention of experimental
multiple sclerosis. J.Immunol. 183, 76027610 104. Chard,D.T. etal. Brain atrophy in clinically early autoimmune encephalomyelitis by antibodies against
(2009). relapsing-remitting multiple sclerosis. Brain 125, 41 integrin. Nature 356, 6366 (1992).
91. Dominguez-Villar,M., Baecher-Allan,C.M. & 327337 (2002). 117. Ben-Nun,A. etal. From classic to spontaneous and
Hafler,D.A. Identification of T helper type1like, 105. Yamasaki,R. etal. Differential roles of microglia and humanized models of multiple sclerosis: impact on
Foxp3+ regulatory Tcells in human autoimmune monocytes in the inflamed central nervous system. understanding pathogenesis and drug development.
disease. Nat. Med. 17, 673675 (2011). J.Exp. Med. 211, 15331549 (2014). J.Autoimmun. 54, 3350 (2014).
92. Schneider,A. etal. In active relapsing-remitting A study that used transgenic technology, scanning 118. Campbell,G.R. etal. Clonally expanded
multiple sclerosis, effector Tcell resistance to adaptive electron microscopy and gene expression profiling mitochondrial DNA deletions within the choroid
Tregs involves IL6mediated signaling. Sci. Transl. Med. to demonstrate a differential role of microglia and plexus in multiple sclerosis. Acta Neuropathol. 124,
5, 170ra115 (2013). monocytes in EAE. 209220 (2012).
93. Bhela,S. etal. Nonapoptotic and extracellular activity 106. Chen,Z. etal. Microglial displacement of inhibitory 119. Haider,L. etal. Oxidative damage in multiple sclerosis
of granzyme B mediates resistance to regulatory Tcell synapses provides neuroprotection in the adult brain. lesions. Brain 134, 19141924 (2011).
(Treg) suppression by HLADRCD25hiCD127lo Tregs in Nat. Commun. 5, 4486 (2014). 120. Weisfeld-Adams,J.D., Katz Sand,I.B., Honce,J.M. &
multiple sclerosis and in response to IL6. J.Immunol. 107. Neher,J.J. etal. Inhibition of microglial phagocytosis Lublin,F.D. Differential diagnosis of Mendelian and
194, 21802189 (2015). is sufficient to prevent inflammatory neuronal death. mitochondrial disorders in patients with suspected
94. Hu,D. etal. Analysis of regulatory CD8 Tcells in J.Immunol. 186, 49734983 (2011). multiple sclerosis. Brain 138, 517539 (2015).
Qa1deficient mice. Nat. Immunol. 5, 516523 108. Goldmann,T. etal. A new type of microglia gene 121. Craner,M.J. etal. Molecular changes in neurons in
(2004). targeting shows TAK1 to be pivotal in CNS autoimmune multiple sclerosis: altered axonal expression of
95. Pannemans,K. etal. HLAE restricted CD8+ Tcell inflammation. Nat. Neurosci. 16, 16181626 (2013). Nav1.2 and Nav1.6 sodium channels and Na+/Ca2+
subsets are phenotypically altered in multiple sclerosis 109. Kotas,M.E. & Medzhitov,R. Homeostasis, exchanger. Proc. Natl Acad. Sci. USA 101,
patients. Mult. Scler. 20, 790801 (2014). inflammation, and disease susceptibility. Cell 160, 81688173 (2004).
96. Baughman,E.J. etal. Neuroantigen-specific 816827 (2015).
CD8+ regulatory Tcell function is deficient during An intriguing review on the parallels between Acknowledgements
acute exacerbation of multiple sclerosis. homeostatic and inflammatory control mechanisms This work was supported by the Wellcome Trust, the Medical
J.Autoimmun. 36, 115124 (2011). in health and disease. Research Council, the Alan and Babette Sainsbury Charitable
97. Tennakoon,D.K. etal. Therapeutic induction of 110. Friese,M.A. etal. Acid-sensing ion channel1 Fund, and the Rosetrees Trust (L.F.).
regulatory, cytotoxic CD8+ Tcells in multiple sclerosis. contributes to axonal degeneration in autoimmune
J.Immunol. 176, 71197129 (2006). inflammation of the central nervous system. Competing interests statement
98. Mattoscio,M. etal. Hematopoietic mobilization: Nat. Med. 13, 14831489 (2007). The authors declare no competing interests.
potential biomarker of response to natalizumab in 111. Baruch,K. etal. Aging-induced typeI interferon
multiple sclerosis. Neurology 84, 14731482 response at the choroid plexus negatively affects
(2015). brain function. Science 346, 8993 (2014). SUPPLEMENTARY INFORMATION
99. Schubert,R.D. etal. IFN- treatment requires Bcells 112. Kira,J. Neuromyelitis optica and Asian phenotype of See online article: S1 (table) | S2 (table)
for efficacy in neuroautoimmunity. J.Immunol. 194, multiple sclerosis. Ann. NY Acad. Sci. 1142, 5871 ALL LINKS ARE ACTIVE IN THE ONLINE PDF
21102116 (2015). (2008).

558 | SEPTEMBER 2015 | VOLUME 15 www.nature.com/reviews/immunol

2015 Macmillan Publishers Limited. All rights reserved

Вам также может понравиться