You are on page 1of 10

Autoimmunity Reviews 19 (2020) 102430

Contents lists available at ScienceDirect

Autoimmunity Reviews
journal homepage: www.elsevier.com/locate/autrev

Multiple sclerosis, the microbiome, TLR2, and the hygiene hypothesis T


a b a,c,⁎
Nicholas J. Wasko , Frank Nichols , Robert B. Clark
a
Department of Immunology, The University of Connecticut Health Center, Farmington, CT 06032, USA
b
School of Dental Medicine, Department of Oral Health and Diagnostic Sciences, The University of Connecticut Health Center, Farmington, CT 06032, USA
c
Department of Medicine,The University of Connecticut Health Center, Farmington, CT 06032, USA

A R T I C LE I N FO A B S T R A C T

Keywords: The pathophysiology of autoimmune diseases such as Multiple Sclerosis (MS) involves a complex interaction
Multiple sclerosis between genetic and environmental factors. Studies of monozygotic twins suggest a significant role for en-
Microbiome vironmental factors in susceptibility to MS. Numerous studies, driven by the “Hygiene Hypothesis,” have focused
TLR2 on the role of environmental factors in allergic and autoimmune diseases. The hygiene hypothesis postulates that
TLR tolerance
individuals living in environments that are too “clean” lack the requisite exposure to “immune-tolerizing” mi-
Remyelination
Hygiene hypothesis
crobial products, resulting in poorly regulated immune systems and increased immune-mediated diseases.
Interestingly, few studies have linked MS with the hygiene hypothesis. Similarly, although numerous studies
have examined the role of the microbiome in autoimmune diseases, there has been no consistent documentation
of disease-specific alterations in the MS microbiome. In this review, we present evidence that integrating the
hygiene hypothesis and the microbiome allows for the identification of novel pathophysiologic mechanisms in
MS.
Our central hypothesis is that the microbiome in MS represents a “defective environment” that fails to provide
normal levels of “TLR2-tolerizing” bacterial products to the systemic immune system. Consistent with the hy-
giene hypothesis, we posit that this defective microbiome function results in abnormally regulated systemic
innate immune TLR2 responses that play a critical role in both the inflammatory and defective remyelinative
aspects of MS. We have completed proof of concept studies that support the inflammatory, remyelinating, and
human immune response components of this paradigm. Our studies suggest that induction of TLR2 tolerance
may represent a novel approach to treating MS, inhibiting autoimmune inflammation while simultaneously
facilitating remyelination.

1. Introduction variables that contribute to pathogenesis, disease onset, and therapeutic


response. Genetic factors lay the foundation for autoimmune diseases,
The study of autoimmune diseases is complicated by the multiple but studies such as those in monozygotic twins demonstrate the

Abbreviations: APC, Antigen presenting cell; Arg1, Arginase 1.; CNS, Central nervous system.; DAMP, Damage associated molecular pattern.; EAE, Experimental
autoimmune encephalomyelitis.; ELISA, Enzyme-linked immunosorbent assay.; EM, Electron microscopy.; GI, Gastrointestinal.; IBA1, Ionized calcium binding
adaptor molecule 1.; iNOS, Inducible nitric oxide synthase.; IRAK, Interleukin-1 receptor-associated kinase.; i.v., Intravenous.; L654, Lipid 654.; LPS,
Lipopolysaccharide.; MRM-mass spectrometry, Multiple reaction monitoring mass spectrometry.; MS, Multiple sclerosis.; MyD88, Myeloid differentiation primary
response protein MyD88.; NF-κB, nuClear factor kappa-light-chain-enhancer of activated B cells.; OL, Oligodendrocyte.; OPC, Oligodendrocyte precursor cell.; P2C,
Pam2CSK4, a synthetic diacylated lipopeptide.; P3C, Pam3CSK4, a synthetic triacylated lipopeptide.; PAMP, Pathogen associated molecular pattern.; PBMC,
Peripheral blood mononuclear cells.; PLP, Proteolipid protein.; PPMS, Primary progressive multiple sclerosis.; PRR, Pattern recognition receptor.; RRMS, Relapsing
remitting multiple sclerosis.; SPMS, Secondary progressive multiple sclerosis.; TID, Type 1 diabetes.; TIR, Toll/interleukin-1 receptor (TIR) homology domain.; TLR,
Toll-like receptor.; Traf6, TNF Receptor-associated factor 6.; Treg, Regulatory T cell.; TRIF, TIR-domain-containing adapter-inducing interferon-β.; TNFα, Tumor
necrosis alpha.; VC, Vehicle control.; Animal welfare, All mice were maintained under specific pathogen-free conditions in accordance with the guidelines for the
Center for Comparative Medicine at UConn Health. All procedures were performed in compliance with Institutional Animal Care and Use Committee-approved
protocols.; Informed consent, Informed consent was obtained on all human subjects after the nature and possible consequences of the studies had been fully
explained.

Corresponding author at: Department of Immunology, Department of Medicine, Room E3013, University of Connecticut Health Center, 263 Farmington Avenue,
Farmington, CT 06030, USA.
E-mail addresses: wasko@uchc.edu (N.J. Wasko), nichols@uchc.edu (F. Nichols), rclark@uchc.edu (R.B. Clark).

https://doi.org/10.1016/j.autrev.2019.102430
Received 7 July 2019; Accepted 11 July 2019
Available online 15 November 2019
1568-9972/ © 2019 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/BY/4.0/).
N.J. Wasko, et al. Autoimmunity Reviews 19 (2020) 102430

significance of non-genetic factors in the etiology of these diseases. underlying these findings [5]. They found that the level of environ-
Studies of monozygotic twins document the variable heritability for mental PAMPs directly affected the phenotype and responses of cells of
different autoimmune diseases, ranging from high concordance among the innate immune system. They documented that the population with
twins with psoriasis to lower concordance (approximately 30%) among less PAMP exposure (i.e., the “cleaner” environment with more asthma)
twins with multiple sclerosis (MS) [1,2]. The twin concordance studies demonstrated stronger innate immune responses (e.g., higher LPS-sti-
in MS suggest that genetics can only partly explain the susceptibility for mulated cytokine secretion), while the population with more PAMP
developing MS and that environmental factors contribute significantly exposure (i.e., the “dirtier” environment with less asthma) demon-
to disease occurrence. In this way, environmental factors also take on strated weaker innate immune responses and also demonstrated a more
great significance as potential targets for new therapeutic approaches. “suppressed” peripheral blood monocyte phenotype (e.g., lower ex-
However, the “environment” involves many potentially relevant and pression of HLA-DR molecules) [5]. The studies of Schuijs et al. [4] and
varied components such as prior infections, geography, toxin exposures Stein et al. [5] highlight that the level of exposure to environmental
(including smoking), and diet, but discerning the relative importance of PAMPs plays a role in regulating the level of innate immune responses
such factors has proven difficult. Recently, much attention has been in the host, and in this way might be a critical factor, along with ge-
focused on environmental factors and their role in allergic and auto- netics, in determining the incidence of immune-mediated diseases.
immune diseases based on two major paradigms: 1) the “Hygiene Hy-
pothesis” and; 2) the role of the microbiome. However, these two 3. Multiple sclerosis and the hygiene hypothesis
concepts have rarely been integrated as a lens for understanding these
immune-mediated diseases. In this review, we present evidence for MS is a chronic neuroinflammatory disease affecting 2.3 million
using this integrated approach to identify novel pathophysiologic me- people worldwide [11] characterized by immune-mediated damage to
chanisms underlying MS. the myelin sheath surrounding neuronal axons [12]. Roughly 80% of
cases present as relapsing-remitting MS (RRMS), with periodic clinical
2. The hygiene hypothesis attacks punctuating periods of stability. Many such cases transition into
secondary progressive MS (SPMS), in which neurological disability
One clue to the identity of relevant environmental factors may be accumulates more consistently [12]. Alternatively, primary progressive
found in the observation that the frequency of both autoimmune dis- MS (PPMS) occurs in 15% of cases, manifesting as continuous dete-
eases and allergic diseases (including asthma) have been significantly rioration of neurological function [13]. The disease pathogenesis in
increasing in industrialized nations over the past few decades. This has RRMS is believed to involve a T lymphocyte-directed attack on myelin
led to the “Hygiene Hypothesis,” which postulates that the increase in or oligodendrocytes, the myelin producing cells, resulting in areas of
these immune-related allergic and autoimmune diseases is a result of an demyelination in the central nervous system (CNS). In addition, pa-
environment that is too clean and fails to appropriately stimulate, tients with MS have a defect in their ability to repair (remyelinate) their
educate, and thus regulate the immune system [3]. The “cleanliness” of demyelinated lesions. Consistent with this concept of T cell-directed
the environment in most cases refers to the relative paucity of microbes damage, therapeutic approaches that target the immune system can
and microbial-derived molecules (pathogen-associated molecular pat- reduce symptomatic flares in RRMS. In contrast, the pathogenesis of
terns or “PAMPs”) that are routinely encountered in one's normal daily both SPMS and PPMS is less clear and no existing therapies demonstrate
activities. Although the mechanisms underlying the hygiene hypothesis consistent efficacy in either slowing the progression of the disease or in
have been somewhat elusive, recent studies have suggested that PAMP- repairing the myelin damage [14–16]. However, as reviewed by
exposure that is chronically “too low” results in poorly regulated and Fraussen et al., the identification of meningeal ectopic B cell follicles in
thus potentially over-reactive innate immune responses [4–6]. SPMS and the successful use of B cell-depleting therapy in PPMS have
As highlighted by Stein et al. [5], a number of epidemiologic studies underlined the importance of B cells in progressive MS” [17].
conducted in Europe have shown significant protection from asthma While extensive research has been conducted over decades at-
and allergic disease in children raised on traditional dairy farms with tempting to identify epidemiologic and environmental factors that in-
high microbial and farm animal exposures [7–10]. In attempting to fluence the incidence of MS, very little has been conclusively docu-
identify relevant mechanisms underlying this association, Schuijs et al. mented. The most frequently cited MS-relevant environmental factors
reported that chronic exposure to low dose lipopolysaccharide (“LPS”; include: the incidence of MS increases as one moves farther away from
“endotoxin”) or farm dust extract protected mice from house dust mite- the equator; the incidence of MS increases with a chronic decrease in
induced asthma [4]. After toll-like receptor (TLR4) signaling, nuclear exposure to sunlight; and the incidence of MS increases in those with a
translocation of NF-kB induces both pro-inflammatory genes and genes low level of serum vitamin D [18]. Other environmental/epidemiologic
for molecules such as A20 that attenuate signaling [4]. Schuijs et al. factors that have historically been implicated in susceptibility to MS
generated mice that lacked A20 in lung epithelium and found that include a history of early contraction or severe infection with Epstein
treatment of mice with low dose LPS or farm dust extract no longer Barr virus [18]. Importantly, little has been published connecting MS to
suppressed asthma in these mice. These studies suggested that treat- the hygiene hypothesis.
ment with low dose LPS or farm dust extract mediated suppression from As summarized by Wendel-Haga and Celius [19] and by Correale
asthma through A20 and attenuation of TLR4 signaling. Consistent with and Gaitán [20], for many years the risk of MS has been suggested to be
their mouse results, these investigators documented that a single nu- higher in individuals with high hygienic standards during childhood,
cleotide polymorphism in the gene encoding A20 was associated with but relevant specific factors have not been identified. Conflicting results
allergy and asthma risk in children growing up on farms [4]. have been reported regarding number of siblings, attendance in a day
Stein et al. investigated the incidence of asthma in farming popu- care center, and exposure to animals during childhood in relation to MS
lations that are genetically similar but differ in their farming techniques risk, but common childhood infections and vaccinations do not seem to
such that exposure to environmental PAMPs (specifically LPS in farm influence the risk of MS. In line with the hygiene hypothesis, two large
dust) differ significantly between the populations [5]. These in- meta-analyses have recently shown that infection with Helicobacter
vestigators found that the population with chronically high PAMP ex- pylori is negatively correlated with MS [21,22].
posures (i.e., “dirtier” environments) had a significantly lower in-
cidence of asthma compared to the population with the chronically low 4. The microbiome
PAMP exposures (i.e., “cleaner” environments). They concluded that
the more microbial products in the environment the less asthma in the The microbiome, a term used to describe the trillions of commensal
population, and they next went on to explore potential mechanisms microorganisms that occupy niches on or within the human body, has

2
N.J. Wasko, et al. Autoimmunity Reviews 19 (2020) 102430

become an area of intense research focus because of its potential as a [37,38] and thus a deficiency in these bacteria and these fatty acids
regulator or modifier of human health, disease, and therapeutic re- could play a role in a decreased functional suppressive capacity of
sponsiveness. Microbiome species colonize several body sites ranging regulatory T cells in RRMS patients [36].
from the oral cavity to the skin to the vagina, but the largest population Overall, there is as yet little consensus in identifying MS-specific
of microorganisms reside within the gastrointestinal (GI) tract [23–28]. microbiome profiles. As summarized by Tremlett et al., most studies
Studies assessing the role of the microbiome in human health and pa- report subtle, rather than large, differences in the MS gut microbiota
thology are ongoing, though preliminary studies suggest that disturbing composition, and it is likely that further longitudinal studies of the
a “normal” microbiome can lead to disease-related alterations in im- microbiome in MS will be necessary to understand the influences of
mune function and metabolism. The composition of the gut microbiome ongoing inflammation, concomitant medications, and issues such as
has been shown to modulate immune cell activity in the host [3], with whether any microbiome changes pre-date or are a consequence of MS
segmented filamentous bacteria inducing intestinal Th17 production or its treatment [39].
[29] and strains of Clostridium and other short chain fatty acid-produ-
cing bacteria facilitating the accumulation of regulatory T cells (Tregs) 6. TLR2 signaling connects MS to both the microbiome and the
in the gut [30]. Important questions that remain unanswered in mi- hygiene hypothesis
crobiome research include the identification of what constitutes a
“normal” microbiome [23,25] and the identification of mechanisms Over the past decade our laboratory has investigated whether MS
that underlie the effect of the microbiome on both the local and sys- could have any connection to the hygiene hypothesis and/or to the
temic immune and metabolic status of the host. microbiome. The studies outlined below strongly suggest that there is
While most mechanistic studies have focused on the effect of spe- indeed a connection and that TLR2 signaling is the key that ties MS to
cific bacterial species and bacterial products on the local (i.e., GI) im- both the hygiene hypothesis and the microbiome. We next present four
mune system, a more difficult and potentially more important question “proof of concept” studies that we believe support this paradigm in MS.
is how the GI microbiome mediates effects on the immune system
outside of the GI tract, i.e., on the entire systemic immune system. At
present, only a small number of studies have suggested mechanisms by 7. First proof of concept: A microbiome-derived PAMP is lower is
which the GI microbiome can affect the systemic immune system. For MS blood
example, it has been reported that specific immune cells, (e.g., Tregs),
initially interacting with and potentially programmed by the micro- In 2013, while studying a family of unique bacterial lipopeptides
biome while in the GI tract, can subsequently leave the GI tract and (“serine lipids”) derived from bacteria in the oral and GI microbiome,
mediate functions in the systemic immune system [31–33]. This me- (produced by several Bacteroides species [40]), we asked whether these
chanism describes an effect of the local environment (GI microbiome) lipopeptides had the ability to be recognized by human immune cells
that then bridges to a more systemic effect via trafficking of cells out of and whether these lipopeptides left their microbiome site and entered
the local environment. A second mechanism involving entry of micro- the human systemic circulation. We found and reported that this family
biome-derived PAMPs into the systemic circulation is discussed in de- of lipopeptides, and specifically one family member, Lipid 654 (L654)
tail below. (Fig. 1A), has the ability to stimulate innate immune cells through TLR2
[40]. Next, to investigate whether L654 can leave its microbiome sites
5. Multiple sclerosis and the microbiome and enter the human systemic circulation, we analyzed human serum
samples for L654. This lipidomic analysis used sophisticated mass
Over the past five years, extensive research has focused on the mi- spectrometry analysis (multiple reaction monitoring [MRM] mass
crobiome in MS. Numerous studies have catalogued the GI bacterial spectrometry). MRM-mass spectrometry allows for unequivocal
populations in patients with MS versus controls. While most studies find
that MS patients have alterations in the composition of their GI mi-
crobiome compared to healthy controls (GI microbial dysbiosis), overall
there has been little consensus in identifying MS-specific microbiome
profiles.
Jangi et al. reported that alterations in microbiome bacterial po-
pulations in MS include increases in Methanobrevibacter and
Akkermansia and decreases in Butyricimonas. Furthermore, these in-
vestigators found that patients on disease-modifying treatment show
increased abundances of Prevotella and Sutterella, and decreased
Sarcina, compared with untreated patients. They concluded that “fur-
ther study is required to assess whether the observed alterations in the
gut microbiome play a role in, or are a consequence of, MS pathogen-
esis” [34]. Chen et al. found an increased abundance of Pseudomonas,
Mycoplana, Haemophilus, Blautia, and Dorea genera in MS patients,
whereas the control group showed increased abundance of Para-
bacteroides, Adlercreutzia, and Prevotella genera. These authors con-
cluded that MS patients have GI microbial dysbiosis and that further
study was needed to understand the role of this dysbiosis in the pa-
thogenesis of MS [35]. Calvo-Barreiro et al., reviewed potential func-
tional links between microbiome alterations and MS, specifically ex-
amining immunoregulatory effects of microbiome changes reported for
RRMS patients [36]. These authors point out that bacterial species Fig. 1. Lipid 654 (L654) is a bacterial-derived lipopeptide found at lower serum
belonging to the Clostridium genus, found in some studies to be un- concentrations in MS patients. (A). Structure of Lipid 654. (B). L654 in Serum.
derrepresented in RRMS patients, are known to be producers of short Serum lipids were derived from 17 MS patients and 13 healthy controls and
chain fatty acids. Short chain fatty acids are reported to promote reg- analyzed for L654 by MRM-mass spectrometry. Results are expressed as ion
ulatory T cells and enhance the production of IL-10 in studies of EAE abundance, Log base 2. [41].

3
N.J. Wasko, et al. Autoimmunity Reviews 19 (2020) 102430

identification and quantification of L654, and through its unique bac- 9. TLR tolerance
terial-associated structures, allows it to be differentiated from lipids of
mammalian (human) origin. Evolutionarily, the fact that the expression of TLR2 is highly con-
Analyzing serum samples from healthy individuals, we were sur- served signifies the importance of TLR2 signaling in immunity to in-
prised to identify L654 in all serum samples studied. This suggested that fections. However, as discussed above, evidence from a number of la-
this microbiome-derived TLR2-stimulating bacterial PAMP was routi- boratories has documented that TLR2 signaling plays a critical disease-
nely and chronically circulating in everyone's bloodstream along with related role in the pathophysiology of MS. These reports, together with
the “normal” mammalian lipids. Most importantly, when we compared our documentation that a large percentage of MS patients have sig-
the serum samples from healthy individuals with those of MS patients, nificantly enhanced responses to TLR2 ligands [60] (see below), sug-
we found that healthy individuals demonstrated levels of L654 that gests a novel therapeutic goal. In this goal, TLR2-responsiveness in
ranged from high to low values while, in contrast, MS patients had patients with MS would be “tuned-down” to levels equal to or less than
levels of L654 that were clustered only in the low range [41] (Fig. 1B). that seen in healthy individuals, while still maintaining the response
This finding was striking and initially surprising given that MS is an level required for protection against infectious agents. This therapeutic
autoimmune disease associated with inflammation of the CNS. Because goal would be difficult to achieve with most inhibitor-based (e.g., an-
L654 has the capacity to stimulate cells in a pro-inflammatory manner tibody) or genetic interference-based approaches that tend to be “all or
via TLR2, we expected serum levels of L654 to be equal or higher in MS none” in their inhibition. However, through evolution, biology has
patients compared to controls. However, we were also aware that TLR2 developed its own “tuning-down” mechanism termed “TLR tolerance”
signaling results not only in an initial release of pro-inflammatory cy- [61–63]. In TLR2 tolerance, cells down-regulate TLR2 responsiveness
tokines but also induces a state of TLR2 tolerance in which subsequent after being exposed repeatedly to an adequate TLR2 stimulus. As dis-
TLR2 responses are dampened or inhibited. cussed below, in recent proof of concept studies, we document that TLR
tolerance can be harnessed to treat autoimmune disease.
TLR tolerance (originally termed “endotoxin tolerance”) is a phe-
8. TLR2 signaling in MS and Experimental Autoimmune nomenon in which prior exposure to TLR-stimulating PAMPs results in
Encephalomyelitis (EAE) a transient state of refractoriness to subsequent challenge [61]. Phe-
notypically, TLR tolerance is associated with increased phagocytic
Our findings with L654 prompted us first to ask what role, if any, ability coupled with reduced antigen presentation in macrophages [64],
does TLR2 signaling play in the pathophysiology of MS. TLRs are a as well as decreased expression of pro-inflammatory cytokines like
family of pattern recognition receptors (PRRs) activated by different TNFα and increased expression of anti-inflammatory cytokines like
PAMPs, which are evolutionarily conserved compounds generated by interleukin-10 (IL-10) [65]. Intracellularly, TLR tolerance induces up-
bacteria, viruses, fungi, and other infectious species [42,43]. TLRs also regulation of negative regulators of MyD88 signaling, including IRAK-
recognize damage-associated molecular patterns (DAMPs) that arise M, A20, BCL3, and microRNAs that target parts of the MyD88 pathway
endogenously from sterile injury or cell turnover [43]. Different TLRs [65–67]. Because many TLRs converge on the MyD88 signaling
sense different PAMPs and DAMPs, such as TLR4 that responds to LPS pathway, induction of tolerance to one TLR ligand can induce cross-
from Gram-negative bacteria [44] and TLR5 that responds to bacterial tolerance to ligands that stimulate other TLRs [67,68]. TLR tolerance is
flagellin [45]. TLR2 is capable of being stimulated by triacylated bac- well documented in monocytes where repeated ligation of TLRs results
terial lipopeptides when heterodimerized with TLR1 or diacylated in subsequent signal dampening and a state of TLR tolerance that pre-
bacterial lipopeptides when heterodimerized with TLR6 [46], as well as vents successive ligations from signaling normally and changes down-
a wide variety of other non-lipopeptidic PAMPs and DAMPs [47]. stream effector molecules to a more regulatory phenotype [61,65]. TLR
When ligated, most TLRs activate the myeloid differentiation pri- tolerance has also been demonstrated in other cell types including en-
mary-response protein 88 (MyD88)-dependent pathway, with the ex- dothelial cells [69] and, as noted above in studies of asthma and A20, in
ceptions of TLR3, which signals via the Toll/interleukin 1 receptor lung epithelial cells [4].
(TIR)-domain-containing adapter-inducing interferon-β (TRIF), and
TLR4, which utilizes both MyD88 and TRIF signaling pathways 10. Our central hypothesis
[42,46,48]. MyD88 activation triggers recruitment of tumor-necrosis-
factor-receptor-associated factor 6 (TRAF6) and members of the IL-1R- With the concept of TLR2 tolerance in mind, in 2013 [41] we pos-
associated kinases (IRAK) family, which allows NF-κB to translocate tulated that the presence of L654 in all serum samples we studied,
into the nucleus [42,46–48]. Nuclear NF-κB increases production of coupled with the findings of lower levels of L654 in MS serum suggested
pro-inflammatory cytokines like tumor necrosis factor α (TNFα) and that: 1) relatively small amounts of L654 (and potentially other mi-
other immune mediators like cyclooxygenase-2 [42,47,48]. crobiome-derived PAMPs) chronically enter the systemic circulation
Studies have reported that the expression of TLR2 is enhanced in from the microbiome; 2) circulating L654 interacts on a chronic basis
both MS and its murine model, EAE [49–51]. While the role of TLR2 with TLR2 on innate immune cells and, at levels found in healthy in-
signaling in the pathophysiology of MS is somewhat unclear, overall the dividuals, induces a relative state of TLR2 tolerance in the innate im-
literature supports a disease-promoting role for TLR2 signaling mune system; 3) this normal state of TLR2 “tolerance” is not sufficient
[52–56]. Key studies include the report that TLR2-deficient mice de- to completely inhibit TLR2 signaling, but rather serves as a micro-
velop attenuated EAE [50,52,54–56] and that TLR2 signaling inhibits biome-driven regulator, dampening or “rheostating down” the magni-
CNS remyelination [51], a defect seen in patients with MS. Further- tude of systemic TLR2 responses without completely inhibiting TLR2
more, there is some evidence that endogenous TLR2 ligands, primarily responses; 4) this microbiome-driven regulation of TLR2 responsiveness
in the form of DAMPs, account for the disease-promoting role of TLR2 has evolved to protect the host from over-reacting to endogenous TLR2
signaling in EAE and MS [57–59]. Support for this concept includes stimulants (e.g., DAMPs), while preserving the ability to respond in an
documentation that inhibiting DAMP signaling through the use of an- appropriate pro-inflammatory manner to the strong-signaling and high
tibodies to DAMPs can attenuate EAE [58,59]. Expression of DAMPs is concentration of TLR2 PAMPs present during an infection; 5) the low
sporadically enhanced when tissue damage or inflammation is present, level of L654 found in the serum of MS patients suggests that one
thus providing a source of endogenous TLR2 ligands to mediate this normal function of the microbiome - i.e., to seed the systemic circula-
disease-promoting effect. tion with levels of L654 that are adequate to “rheostat” down the level
of response to TLR2 - is abnormal in MS; and 6) the subsequent en-
hanced responsiveness to endogenous TLR2 stimulants, including

4
N.J. Wasko, et al. Autoimmunity Reviews 19 (2020) 102430

Fig. 2. Central Hypothesis: in Healthy Individuals: GI micro-


biome-derived PAMPs, such as L654, chronically enter the
systemic circulation at low levels, interact with PRRs such as
TLR2 on immune cells, and mediate a level of TLR tolerance
that serves to “rheostat” down the response to subsequent li-
gation. Through evolution, this level of tolerance has been
selected to diminish the stimulation potentially occurring
through intermittent DAMP expression (and thus decrease the
risk for auto-inflammation and autoimmunity), while preser-
ving an appropriate pro-inflammatory response to the en-
hanced concentration and strength of ligands presented by
infectious agents. In Patients with MS: there is inadequate
seeding of relevant and functional GI microbiome-derived
PAMPs into the systemic circulation resulting in inadequate
induction of innate immune TLR tolerance. This inadequate
level of TLR tolerance allows for a significant pro-in-
flammatory response through intermittent DAMP expression
and stimulation, which in turn increases the risk for auto-
immune diseases such as MS. In addition, the enhanced TLR2
responses result in defective remyelination, as discussed
below.

DAMPs, plays an integral role in the inflammatory and defective re- that a paucity of functionally adequate “environmental” bacterial
myelination pathophysiology of MS (Fig. 2). PAMPs (dust-derived in the asthma studies; microbiome-derived in the
T1D study) resulted in inadequate regulation of systemic innate im-
11. Recent literature support for our 1st proof of concept and mune TLR responses and enhanced autoimmunity.
central hypothesis Overall, this T1D microbiome study supports our central hypothesis,
establishing the microbiome as an “environment” that supplies the
Our hypothesis posits that the microbiome is a critical component of systemic innate immune system with a chronic low-dose of PAMPs that
our “environment” and as such, plays an important role as a source of regulate the immune response and aid in controlling the development
environmental PAMPs in the context of the hygiene hypothesis. Put of autoimmune disease.
forth in 2013 [41], our central hypothesis gained support in the 2016
report documenting that differences in the function of the GI micro- 12. Second proof of concept: Systemic TLR2 tolerance induction
biome-derived PAMP, LPS, could alter the incidence of the autoimmune inhibits a T-cell transfer model of EAE
disease type 1 diabetes (T1D) [6].
In this study, Vatanen et al. studied the GI microbiome of infants To begin to probe the association of TLR2 tolerance and MS, we
from Finland and Estonia and compared them with the GI microbiome conducted proof of concept studies using EAE [70]. We specifically used
of infants from nearby Russia [6]. Finland and Estonia are known to the SJL-proteolipid protein (PLP) model of adoptive transfer (AT) EAE
have a significantly higher incidence of allergy, autoimmunity, and in which disease is mediated in naïve mice by adoptive transfer of ac-
early onset T1D compared to the nearby region of Russia. These in- tivated T cells from PLP-immunized mice. This model avoids using
vestigators analyzed the microbiome of these subjects and found dif- exogenous adjuvants that contain TLR2 ligands. We found we could
ferences in the microbiome bacterial composition between the Finnish/ enhance the level of TLR2 tolerance beyond the level theoretically
Estonian subjects and the Russian subjects. Importantly, they found that mediated by the microbiome by administering a low dose of the TLR2
the Finnish/Estonian microbiome-derived LPS differed in structure and ligand Pam2Cys (P2C) intravenously (i.v.) for as few as three days,
function from the Russian microbiome-derived LPS. The LPS produced which we termed “TLR2 tolerance induction” [70]. Tolerance was
by the Russian (low incidence of autoimmunity) microbiome func- documented by a diminished serum TNFα response to administration of
tioned efficiently in inducing TLR4 tolerance. In contrast, the LPS de- a larger dose of a second TLR2 ligand after three days of treatment with
rived from the Finnish/Estonian (higher incidence of autoimmunity) P2C. Most importantly, administering low dose P2C for 14 days sig-
microbiome was incapable of inducing TLR4 tolerance. Additionally, nificantly attenuated AT EAE compared to mice receiving vehicle
when tested in the diabetes-prone NOD strain of mice, the Russian LPS control (VC) for 14 days (Fig. 3A). The TLR2-tolerized mice with atte-
induced TLR4 tolerance in vivo and inhibited the onset of diabetes, nuated EAE demonstrated significantly fewer T cells in the CNS com-
while the Finnish/Estonian LPS exhibited neither of these effects. Based pared to control mice [70]. EAE attenuation was also seen when tol-
on their in vitro TLR-stimulation results, their NOD mice studies, and erance was induced with L654 instead of P2C. We further documented
the published hygiene hypothesis-related asthma studies, Vatanen et al. that in vivo induction of TLR2 tolerance cross-tolerized to other TLRs,
postulated that PAMPs emanating from the microbiome “environment” enhancing the efficiency of this mode of immune regulation [70].
have the capacity to “tolerize” cells of the innate immune system and In this EAE proof of concept study, we found that TLR2 tolerance
thereby down-regulate enhanced immune responses underlying auto- and EAE attenuation was associated with significant decreases in CD80-
immune and allergic diseases. This study concluded that lack of ex- expressing macrophages and pro-inflammatory F4/80+CD11c+ mac-
posure to microbiome-derived LPS capable of inducing TLR4 tolerance rophages in the CNS (Fig. 3B). We postulated that one mechanism
resulted in inadequate regulation of systemic innate immune TLR re- underlying the decreased T cell numbers in the CNS is diminished T cell
sponses and enhanced autoimmunity in the form of T1D. In this way, re-stimulation in the CNS by tolerized antigen presenting cells (APCs).
this T1D study paralleled the asthma studies discussed above, finding However, the role of specific CNS APC populations in EAE pathogenesis

5
N.J. Wasko, et al. Autoimmunity Reviews 19 (2020) 102430

Fig. 3. Administration of low-dose P2C attenuates AT EAE. (A). Naïve SJL/J mice were injected intraperitoneally with 29 × 106 PLP139–151-reactive lymph node
cells on day 0. EAE disease severity was recorded daily for each mouse and compiled as a daily mean disease score for each cohort. Injections of vehicle control (VC)
or 0.35 μg Pam2Cys (P2C) began at day −5 and continued through day +8 post cell transfer; 2 independent experiments are compiled. n = 6–10 per group; disease
incidence was VC: 9/10 and P2C: 1/6; p < 0.0001 Mann-Whitney U. (B). Percentage of spinal cord F4/80+ cells that are positive for CD80 expression; (left: n = 9;
*p = 0.0248 by Student‘s t-test) and percentage of spinal cord mononuclear cells that are F4/80 + CD11c+; (right: n = 13; ****p = 0.0001 by Student ‘s t-test [70].

remains controversial and uncertainty remains regarding which cells stimulation [60]. Interestingly, the enhanced TLR2 responders included
are most critical in presenting antigen to CNS-migrating T cells. In our a significant fraction of those with progressive forms of MS, a subset of
study [70], we identified alterations in CNS macrophage populations patients considered largely unresponsive to adaptive immune system-
but not in microglial or classic dendritic cell populations in TLR2-tol- targeting therapies (Fig. 4A and B). The results of this study are con-
erized mice. We concluded that the decrease in CNS T cells and CNS sistent with our central hypothesis suggesting that there is defective
APC activation markers supports the working hypotheses that TLR2 regulation of TLR2 responses in patients with MS and further highlight
tolerance induction inhibits a stage of disease relating to T cell-APC the presence of a pathologically relevant TLR2-related innate immune
interactions affecting T cell re-stimulation and thus T cell survival and abnormality in patients with both relapsing-remitting and progressive
proliferation in the CNS. forms of MS.

13. Third proof of concept: enhanced TLR2 responses in multiple 14. Fourth proof of concept: systemic TLR2 tolerance induction
sclerosis enhances remyelination in the cuprizone model of CNS
demyelination
Based on the lower levels of serum L654 in MS patients (first proof
of concept), the therapeutic efficacy of TLR2 tolerance in EAE (second MS involves not only an autoimmune CNS inflammatory process but
proof of concept), and our hypothesis that TLR2 tolerance is defective also a defect in the ability to remyelinate the neurons demyelinated in
in MS patients, we next postulated that defective TLR2 tolerance will this inflammatory process [14,71,72]. It is known that in MS there is a
result in TLR2 hyper-responsiveness in MS patients. Studies of TLR loss of the myelin-producing cells, the oligodendrocytes (OLs) [73].
responses in patients with MS have been conflicting. Importantly, most Although demyelination can theoretically be repaired by differentiation
of these investigations have focused on the response to TLR4 ligation of oligodendrocyte precursor cells (OPCs) into mature OLs, for reasons
and few have characterized TLR2 responses in MS. In this third proof of as yet unknown, remyelination is defective in MS [71,73–75], and di-
concept study, our goal was to use multiple approaches to characterize minished differentiation of OPCs into mature OLs may contribute to this
TLR2 responses of peripheral blood mononuclear cells (PBMCs) and defect in remyelination [14,71,72].
CD14+ monocytes derived from MS patients. Studying a total of 26 MS Enhancing remyelination has become an increasingly important
patients and 32 healthy controls, we documented for the first time that goal of MS research. While studies have suggested that signaling
50% of MS patients demonstrate enhanced responsiveness to TLR2 through TLR2 plays a critical role in the inflammatory pathogenesis of

6
N.J. Wasko, et al. Autoimmunity Reviews 19 (2020) 102430

Fig. 4. MS patients exhibit enhanced TLR2 responsiveness compared to healthy controls. (A). CD14+ monocytes from MS patients (15 patients; 16 total responses)
and healthy controls (13 individuals; 15 total responses), were cultured at 105/ml, stimulated with 2 μg/ml of Pam 2Cys (P2C) or Pam3Cys (P3C) for 4 h, and TNFα in
the culture supernatants was measured by ELISA. The level of TNFα representing the upper threshold of control responses [based on the control interquartile range
(IQR)] is depicted by the dashed line. The percentage of responses above the threshold is depicted for control and total MS patients. Medians are depicted. (a) Red
icons represent P2C stimulation and black icons represent P3C stimulation; (b) same results as in (a), but green icons represent patients with progressive MS. The
mean TNFα level for the control cohort was 104 ρg/ml and the mean TNFα level for the total MS cohort was 247 ρg/ml; P = 0.0712 via Mann–Whitney. The
percentage of responses above threshold: 57% of progressive MS patients, 33% of RRMS patients. (B). Compilation of percentages of control and MS patients with
enhanced TLR2 responses as assessed in 3 studies using: peripheral blood mononuclear cells (PBMC), CD14+ whole population, or CD14+ single-cell analyses. These
compiled studies encompass 26 MS patients and 32 healthy controls. The percentages of progressive MS patients (Prog MS) with enhanced responses
(mean = 60.13%) were significantly greater than those of the control patients (mean = 6.87%); P = 0.0098 via Kruskal–Wallis followed by Dunn's multiple
comparisons test. The percentages of total MS patients (51.55%) and RRMS patients (41.65%) with enhanced responses were greater than those of the control
patients, but did not reach statistical significance. Medians are depicted [60].

MS and EAE [70,76,77], only few studies have investigated a role for the recovery period resulted in a significant enhancement in re-
TLR2 signaling in remyelination. Critically, the majority of these studies myelination as documented by electron microscopy (EM) evaluation of
have demonstrated an inhibitory effect of TLR2 signaling on re- both percentage of unmyelinated axons and myelin thickness (g-ratios:
myelination [51,78,79]. Prior studies have reported that the in vitro the ratio of the inner axonal diameter to the total outer diameter) [81]
maturation of OPCs to mature OLs is inhibited by TLR2 ligation, and (Fig. 5A, B). As shown in Fig. 5 A–B, results based on evaluation of the
that hyaluronate, an endogenous TLR2 ligand (i.e., a DAMP), may be a EM images demonstrated that the percentage of unmyelinated axons
relevant mediator of this inhibition of remyelination [51]. Furthermore, was significantly less in the TLR2 tolerized mice and the myelin
mice in which TLR2 was genetically deleted (TLR2−/− mice) demon- thickness was significantly greater in the TLR2 tolerized mice (evi-
strated enhanced remyelination in vivo after lysolecithin-induced de- denced by a lower g-ratio). In fact, at the two week time period of
myelination [51]. recovery, the degree of remyelination in TLR2 tolerized mice was
Based on studies relating TLR2 signaling to the inhibition of re- equivalent to that normally seen only after four-to-five weeks of re-
myelination [51,78,79] and the potential for therapeutic intervention covery in control mice, and was identical to that seen in mice that had
in EAE and MS via TLR2 tolerance induction [60,70], we next tested the never undergone demyelination.
role of TLR2 tolerance induction in the process of CNS remyelination. In confirmation of these results, we documented that TLR2−/− mice
Since we previously documented that TLR2 tolerance induction could showed a significant enhancement in remyelination using the same EM
attenuate the CNS inflammation in EAE [70], in this study we used the evaluation parameters [81]. In line with previous studies [78], TLR2−/

non-inflammatory cuprizone model of demyelination [80]. mice lost myelin at levels similar to WT mice when examined im-
Mice were fed cuprizone for five weeks to induce a non-in- mediately after five weeks of cuprizone exposure. However, when given
flammatory demyelination, followed by a two-week feeding with two weeks to recover on normal chow (without any intervention),
normal chow to allow for remyelination. We asked whether diminishing TLR2−/− mice exhibited a significant enhancement in remyelination,
TLR2 signaling by inducing systemic TLR2 tolerance during the two- phenocopying the effect seen in TLR2-tolerized WT mice [81]. These
week recovery period would enhance remyelination. TLR2 tolerance results suggest that TLR2 tolerance induces a state of functional TLR2
was induced by daily administration of low dose P2C beginning when deficiency, allowing myelin to recover without the interference medi-
the cuprizone feeding was stopped and continuing for the two-week ated by endogenous TLR2 signaling.
recovery period. Results showed that inducing TLR2 tolerance during Using immunohistofluorescent approaches, we found that the TLR2

7
N.J. Wasko, et al. Autoimmunity Reviews 19 (2020) 102430

Fig. 5. Induction of TLR2 tolerance enhances remyelination during recovery from cuprizone feeding. (A) Percentage of unmyelinated axons and (B) myelin thickness
(g-ratios) as calculated from EM images. VC: mice treated with vehicle control during recovery; P2C: mice treated with the TLR2 ligand, P2C. during trecovery. Data
points represent individual mice. N = 8–10 mice per experimental condition. Statistical differences were assessed by 2-way ANOVA. (C) Percentage of IBA1+ cells
that are iNOS+, Arg1- (D) Percentage of IBA1+ cells that are Arg1+, iNOS-. Error bars represent the mean ± SEM. Statistical differences were assessed by 2-way
ANOVA, * = p < 0.05; ** = p < 0.01; *** = p < 0.001; **** = p < 0.0001 (adapted from [81]).

tolerance-induced enhancement in remyelination was not associated 50% of MS patients [60], the results of this remyelination study further
with changes in the number of OPCs or mature OLs, but rather was support our central hypothesis that inducing TLR2 tolerance in vivo
associated with a change in the balance of microglial (IBA1+) pheno- mediates functional changes in TLR2 signaling that have physiologic
types from pro-inflammatory M1 (iNOS+ Arg1−) to non-inflammatory and disease-relevant effects. By demonstrating the relevance of TLR2
M2 (iNOS− Arg1+) [81] (Fig. 5C, D). Functionally, microglia (like signaling in remyelination, the present study also confirms earlier stu-
macrophages) can be divided into different states of activation. Though dies suggesting that TLR2 signaling plays a critical role in the re-
likely an oversimplification, M1 microglia are described as “pro-in- myelination defect noted in patients with MS.
flammatory” with regards to their cytokine secretion while M2 micro-
glia, or “alternatively activated” microglia, are associated with extra- 15. Summary and conclusions
cellular matrix remodeling, progenitor cell differentiation, and tissue
regeneration [82–84]. Many components of the pathophysiology of MS remain unclear.
These results suggest that the enhanced remyelination seen with Although extensive research has focused on the relationship between
TLR2 tolerance results, at least in part, from effects on microglial ac- autoimmune diseases and the hygiene hypothesis or autoimmune dis-
tivation phenotype, and specifically on an increase in the percentage of eases and the microbiome, as yet no conclusive breakthroughs in our
microglia expressing an M2 phenotype. While there is not yet a clear understanding of disease mechanisms in MS have come from these
understanding of how M2 microglial polarization results in enhanced approaches. In this review, we present a novel paradigm that connects
remyelination, proposed mechanisms include facilitating OPC to OL the pathophysiology of MS to both the hygiene hypothesis and the
differentiation [85,86] and increasing phagocytic clearance of myelin microbiome.
debris [87,88]. In sum, this study presents evidence that the innate A key to our paradigm is the concept that abnormally enhanced
immune system, and TLR2 specifically, has a role in the process of re- TLR2 signaling is a critical player in both the autoreactive CNS in-
myelination. Moreover, together with our previous findings of lower flammation and defective remyelination seen in MS. An abnormally
serum L654 levels in MS patients [41], TLR2 tolerance-induced at- enhanced TLR2 responsiveness is, in turn, dependent on both a mi-
tenuation of EAE [70], and enhanced in vitro TLR2 responsiveness in crobiome that is defective in providing adequate TLR2-tolerizing

8
N.J. Wasko, et al. Autoimmunity Reviews 19 (2020) 102430

PAMPs to the systemic immune system and the hygiene hypothesis that sclerosis. Nature 1993;364(6434):243–7.
posits that environmental PAMPs are required to adequately regulate [3] Bach JF. The hygiene hypothesis in autoimmunity: the role of pathogens and
commensals. Nat Rev Immunol 2018;18(2):105–20.
and “tune” innate immune responses. In terms of the hygiene hypoth- [4] Schuijs MJ, Willart MA, Vergote K, Gras D, Deswarte K, Ege MJ, et al. Farm dust and
esis and MS, the microbiome is the relevant “environment” and its endotoxin protect against allergy through A20 induction in lung epithelial cells.
defective delivery of “environmental” PAMPs to the systemic immune Science 2015;349(6252):1106–10.
[5] Stein MM, Hrusch CL, Gozdz J, Igartua C, Pivniouk V, Murray SE, et al. Innate
system makes it functionally “too clean.” The result is innate immune immunity and asthma risk in Amish and Hutterite farm children. N Engl J Med
over-responsiveness that we postulate is critical to the pathophysiology 2016;375(5):411–21.
of MS. [6] Vatanen T, Kostic AD, d’Hennezel E, Siljander H, Franzosa EA, Yassour M, et al.
Variation in microbiome LPS immunogenicity contributes to autoimmunity in hu-
In outlining our postulate, we present four proof of concept studies mans. Cell 2016;165(6):1551.
that support the inflammatory component (EAE studies), the re- [7] Braun-Fahrländer C, Riedler J, Herz U, Eder W, Waser M, Grize L, et al.
myelinating component (cuprizone studies), and the human immune Environmental exposure to endotoxin and its relation to asthma in school-age
children. N Engl J Med 2002;347(12):869–77.
response component (serum L654 levels and peripheral blood monocyte
[8] Ege MJ, Frei R, Bieli C, Schram-Bijkerk D, Waser M, Benz MR, et al. Not all farming
TLR2 responses) of our paradigm. It should be noted that we do not environments protect against the development of asthma and wheeze in children. J
believe the one PAMP we have studied in MS, L654, is necessarily the Allergy Clin Immunol 2007;119(5):1140–7.
only or most relevant circulating PAMP in this paradigm. However, [9] Ege MJ, Mayer M, Normand AC, Genuneit J, Cookson WO, Braun-Fahrländer C,
et al. Exposure to environmental microorganisms and childhood asthma. N Engl J
Calvo-Barreiro et al. note that L654 is produced by Bacteroides species Med 2011;364(8):701–9.
and the proportion of several Bacteroides genera is reported to be re- [10] Ege MJ, Strachan DP, Cookson WO, Moffatt MF, Gut I, Lathrop M, et al. Gene-
duced in the microbiome of RRMS patients, suggesting that “…defi- environment interaction for childhood asthma and exposure to farming in Central
Europe. J Allergy Clin Immunol 2011;127(1). [138–44, 44.e1–4].
ciency of this naturally occurring innate immune regulation could lead [11] Browne P, Chandraratna D, Angood C, Tremlett H, Baker C, Taylor BV, et al. Atlas of
to the escape of autoreactive immune responses and could represent a multiple sclerosis 2013: a growing global problem with widespread inequity.
mechanism linking the commensal microbiome and MS…” [36]. We Neurology 2014;83(11):1022–4.
[12] Yong H, Chartier G, Quandt J. Modulating inflammation and neuroprotection in
believe that our proof of concept studies lay the foundation for con- multiple sclerosis. J Neurosci Res 2018 Jun;96(6):927–50. https://doi.org/10.
tinued documentation of the relevance of this paradigm in MS and also 1002/jnr.24090. Epub 2017 Jun 5.
for more basic studies confirming the relationship between the micro- [13] Ransohoff RM, Hafler DA, Lucchinetti CF. Multiple sclerosis-a quiet revolution. Nat
Rev Neurol 2015;11(3):134–42.
biome production of systemically “tolerizing” PAMPs and innate im- [14] Hanafy KA, Sloane JA. Regulation of remyelination in multiple sclerosis. FEBS Lett
mune regulation in health and disease. 2011;585(23):3821–8.
Finally, our studies to date highlight the dual role of TLR2-signaling [15] Shirani A, Okuda DT, Stuve O. Therapeutic advances and future prospects in pro-
gressive forms of multiple sclerosis. Neurotherapeutics 2016;13(1):58–69.
in autoimmune CNS inflammation and defective CNS remyelination,
[16] Gholamzad M, Ebtekar M, Ardestani MS, Azimi M, Mahmodi Z, Mousavi MJ, et al. A
and suggest that induction of TLR2 tolerance may represent a novel comprehensive review on the treatment approaches of multiple sclerosis: currently
two-pronged approach to treating MS, inhibiting autoimmune in- and in the future. Inflamm Res 2019;68(1):25–38.
flammation while simultaneously facilitating myelin repair. [17] Fraussen J, de Bock L, Somers V. B cells and antibodies in progressive multiple
sclerosis: contribution to neurodegeneration and progression. Autoimmun Rev
2016;15(9):896–9.
Funding source [18] Ascherio A. Environmental factors in multiple sclerosis. Expert Rev Neurother
2013;13(12 Suppl):3–9.
[19] Wendel-Haga M, Celius EG. Is the hygiene hypothesis relevant for the risk of
Connecticut Bio Innovations Fund (CBIF, Grant #517) funded this multiple sclerosis? Acta Neurol Scand 2017;136(Suppl. 201):26–30.
research. CBIF had no role in study design; in the collection, analysis [20] Correale J, Gaitán MI. Multiple sclerosis and environmental factors: the role of
and interpretation of data; in the writing of the report; and in the de- vitamin D, parasites, and Epstein-Barr virus infection. Acta Neurol Scand
2015;132(199):46–55.
cision to submit the article for publication. [21] Jaruvongvanich V, Sanguankeo A, Jaruvongvanich S, Upala S. Association between
helicobacter pylori infection and multiple sclerosis: a systematic review and meta-
Declaration of Competing Interest analysis. Mult Scler Relat Disord 2016;7:92–7.
[22] Yao G, Wang P, Luo XD, Yu TM, Harris RA, Zhang XM. Meta-analysis of association
between Helicobacter pylori infection and multiple sclerosis. Neurosci Lett
None. 2016;620:1–7.
This manuscript is not under consideration for publication else- [23] Cho I, Yamanishi S, Cox L, Methe BA, Zavadil J, Li K, et al. Antibiotics in early life
alter the murine colonic microbiome and adiposity. Nature 2012;488(7413):621–6.
where, that its publication is approved by all authors and tacitly or
[24] Hou YP, He QQ, Ouyang HM, Peng HS, Wang Q, Li J, et al. Human gut microbiota
explicitly by the responsible authorities at the University of Connecticut associated with obesity in Chinese children and adolescents. Biomed Res Int
Health Center, and if accepted, it will not be published elsewhere in the 2017;2017:7585989.
same form, in English or in any other language, including electronically [25] Lambrecht BN, Hammad H. The immunology of the allergy epidemic and the hy-
giene hypothesis. Nat Immunol 2017;18(10):1076–83.
without the written consent of the copyrightholder. [26] Selber-Hnatiw S, Rukundo B, Ahmadi M, Akoubi H, Al-Bizri H, Aliu AF, et al.
Human gut microbiota: toward an ecology of disease. Front Microbiol 2017;8:1265.
Acknowledgements [27] Sochocka M, Donskow-Lysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J.
The gut microbiome alterations and inflammation-driven pathogenesis of
Alzheimer’s disease-a critical review. Mol Neurobiol 2019;56(3):1841–51.
The authors would like to thank Dr. Glenn Matsushima, University [28] Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J
of North Carolina School of Medicine, for his expert advice on using the 2017;474(11):1823–36.
[29] Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, et al. Induction of
cuprizone model, Ms. Maya Yankova of the UConn Health Electron intestinal Th17 cells by segmented filamentous bacteria. Cell 2009;139(3):485–98.
Microscopy Core, and Dr. Evan Jellison of the UConn Health Flow [30] Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction
Cytometry Core for their expert technical assistance. We also thank all of colonic regulatory T cells by indigenous Clostridium species. Science
2011;331(6015):337–41.
the healthy volunteers and the MS patients for their participation in our
[31] Ochoa-Reparaz J, Mielcarz DW, Wang Y, Begum-Haque S, Dasgupta S, Kasper DL,
study as well as the staff of the Lowell P. Weicker Jr. Clinical Research et al. A polysaccharide from the human commensal Bacteroides fragilis protects
Center, UConn Health. against CNS demyelinating disease. Mucosal Immunol 2010;3(5):487–95.
[32] Wang Y, Begum-Haque S, Telesford KM, Ochoa-Reparaz J, Christy M, Kasper EJ,
et al. A commensal bacterial product elicits and modulates migratory capacity of
References CD39(+) CD4 T regulatory subsets in the suppression of neuroinflammation. Gut
Microbes 2014;5(4):552–61.
[1] Generali E, Ceribelli A, Stazi MA, Selmi C. Lessons learned from twins in auto- [33] Mangalam A, Shahi SK, Luckey D, Karau M, Marietta E, Luo N, et al. Human gut-
immune and chronic inflammatory diseases. J Autoimmun 2017;83:51–61. derived commensal bacteria suppress CNS inflammatory and demyelinating dis-
[2] Utz U, Biddison WE, McFarland HF, McFarlin DE, Flerlage M, Martin R. Skewed T- ease. Cell Rep 2017;20(6):1269–77.
cell receptor repertoire in genetically identical twins correlates with multiple [34] Jangi S, Gandhi R, Cox LM, Li N, von Glehn F, Yan R, et al. Alterations of the human
gut microbiome in multiple sclerosis. Nat Commun 2016;7:12015.

9
N.J. Wasko, et al. Autoimmunity Reviews 19 (2020) 102430

[35] Chen J, Chia N, Kalari KR, Yao JZ, Novotna M, Paz Soldan MM, et al. Multiple molecular mechanisms and relevance to disease. J Endotoxin Res
sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci 2006;12(3):133–50.
Rep 2016;6:28484. [62] Xiong Y, Medvedev AE. Induction of endotoxin tolerance in vivo inhibits activation
[36] Calvo-Barreiro L, Eixarch H, Montalban X, Espejo C. Combined therapies to treat of IRAK4 and increases negative regulators IRAK-M, SHIP-1, and A20. J Leukoc Biol
complex diseases: the role of the gut microbiota in multiple sclerosis. Autoimmun 2011;90(6):1141–8.
Rev 2018;17(2):165–74. [63] Xiong Y, Pennini M, Vogel SN, Medvedev AE. IRAK4 kinase activity is not required
[37] Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, de Roos P, et al. for induction of endotoxin tolerance but contributes to TLR2-mediated tolerance. J
Metabolites produced by commensal bacteria promote peripheral regulatory T-cell Leukoc Biol 2013;94(2):291–300.
generation. Nature 2013;504(7480):451–5. [64] del Fresno C, Garcia-Rio F, Gomez-Pina V, Soares-Schanoski A, Fernandez-Ruiz I,
[38] Haghikia A, Jörg S, Duscha A, Berg J, Manzel A, Waschbisch A, et al. Dietary fatty Jurado T, et al. Potent phagocytic activity with impaired antigen presentation
acids directly impact central nervous system autoimmunity via the small intestine. identifying lipopolysaccharide-tolerant human monocytes: demonstration in iso-
Immunity. 2015;43(4):817–29. lated monocytes from cystic fibrosis patients. J Immunol 2009;182(10):6494–507.
[39] Tremlett H, Waubant E. The multiple sclerosis microbiome? Ann Transl Med [65] Biswas SK, Lopez-Collazo E. Endotoxin tolerance: new mechanisms, molecules and
2017;5(3):53. clinical significance. Trends Immunol 2009;30(10):475–87.
[40] Clark RB, Cervantes JL, Maciejewski MW, Farrokhi V, Nemati R, Yao X, et al. Serine [66] Nahid MA, Benso LM, Shin JD, Mehmet H, Hicks A, Ramadas RA. TLR4, TLR7/8
lipids of porphyromonas gingivalis are human and mouse toll-like receptor 2 li- agonist-induced miR-146a promotes macrophage tolerance to MyD88-dependent
gands. Infect Immun 2013;81(9):3479–89. TLR agonists. J Leukoc Biol 2016;100(2):339–49.
[41] Farrokhi V, Nemati R, Nichols FC, Yao X, Anstadt E, Fujiwara M, et al. Bacterial [67] Nahid MA, Satoh M, Chan EK. MicroRNA in TLR signaling and endotoxin tolerance.
lipodipeptide, lipid 654, is a microbiome-associated biomarker for multiple Cell Mol Immunol 2011;8(5):388–403.
sclerosis. Clin Transl Immunol 2013;2(11):e8. [68] Dalpke AH, Lehner MD, Hartung T, Heeg K. Differential effects of CpG-DNA in toll-
[42] Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol like receptor-2/-4/-9 tolerance and cross-tolerance. Immunology.
2004;4(7):499–511. 2005;116(2):203–12.
[43] Li J, Wang X, Zhang F, Yin H. Toll-like receptors as therapeutic targets for auto- [69] Koch SR, Lamb FS, Hellman J, Sherwood ER, Stark RJ. Potentiation and tolerance of
immune connective tissue diseases. Pharmacol Ther 2013;138(3):441–51. toll-like receptor priming in human endothelial cells. Transl Res 2017
[44] Park BS, Song DH, Kim HM, Choi BS, Lee H, Lee JO. The structural basis of lipo- Feb;180:53–67. https://doi.org/10.1016/j.trsl.2016.08.001. Epub 2016 Aug 8.
polysaccharide recognition by the TLR4-MD-2 complex. Nature [70] Anstadt EJ, Fujiwara M, Wasko N, Nichols F, Clark RB. TLR tolerance as a treatment
2009;458(7242):1191–5. for central nervous system autoimmunity. J Immunol 2016;197(6):2110–8.
[45] Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett DR, et al. The innate [71] Kremer D, Aktas O, Hartung HP, Kury P. The complex world of oligodendroglial
immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature differentiation inhibitors. Ann Neurol 2011;69(4):602–18.
2001;410(6832):1099–103. [72] Kuhlmann T, Miron V, Cui Q, Wegner C, Antel J, Bruck W. Differentiation block of
[46] O’Neill LA, Golenbock D, Bowie AG. The history of Toll-like receptors-redefining oligodendroglial progenitor cells as a cause for remyelination failure in chronic
innate immunity. Nat Rev Immunol 2013;13(6):453–60. multiple sclerosis. Brain 2008;131(Pt 7):1749–58.
[47] Kawai T, Akira S. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int [73] McTigue DM, Tripathi RB. The life, death, and replacement of oligodendrocytes in
Immunol 2009;21(4):317–37. the adult CNS. J Neurochem 2008;107(1):1–19.
[48] Fiebich BL, Batista CRA, Saliba SW, Yousif NM, de Oliveira ACP. Role of microglia [74] Franklin RJ, Ffrench-Constant C. Remyelination in the CNS: from biology to
TLRs in neurodegeneration. Front Cell Neurosci 2018;12:329. therapy. Nat Rev Neurosci 2008;9(11):839–55.
[49] Hasheminia SJ, Zarkesh-Esfahani SH, Tolouei S, Shaygannejad V, Shirzad H, [75] Goldman SA, Nedergaard M, Windrem MS. Glial progenitor cell-based treatment
Hashemzadeh CM. Toll like receptor 2 and 4 expression in peripheral blood and modeling of neurological disease. Science 2012;338(6106):491–5.
mononuclear cells of multiple sclerosis patients. Iran J Immunol 2014;11(2):74–83. [76] Kdirby TO, Ochoa-Reparaz J. The gut microbiome in multiple sclerosis: a potential
[50] Prinz M, Garbe F, Schmidt H, Mildner A, Gutcher I, Wolter K, et al. Innate immunity therapeutic avenue. Med Sci (Basel) 2018;6(3).
mediated by TLR9 modulates pathogenicity in an animal model of multiple [77] Wang Y, Telesford KM, Ochoa-Reparaz J, Haque-Begum S, Christy M, Kasper EJ,
sclerosis. J Clin Invest 2006;116(2):456–64. et al. An intestinal commensal symbiosis factor controls neuroinflammation via
[51] Sloane JA, Batt C, Ma Y, Harris ZM, Trapp B, Vartanian T. Hyaluronan blocks oli- TLR2-mediated CD39 signalling. Nat Commun 2014;5:4432.
godendrocyte progenitor maturation and remyelination through TLR2. Proc Natl [78] Esser S, Gopfrich L, Bihler K, Kress E, Nyamoya S, Tauber SC, et al. Toll-like re-
Acad Sci U S A 2010;107(25):11555–60. ceptor 2-mediated glial cell activation in a mouse model of Cuprizone-induced
[52] Herrmann I, Kellert M, Schmidt H, Mildner A, Hanisch UK, Bruck W, et al. demyelination. Mol Neurobiol 2018;55(8):6237–49.
Streptococcus pneumoniae infection aggravates experimental autoimmune en- [79] Gyetvai G, Roe C, Heikal L, Ghezzi P, Mengozzi M. Leukemia inhibitory factor in-
cephalomyelitis via Toll-like receptor 2. Infect Immun 2006;74(8):4841–8. hibits erythropoietin-induced myelin gene expression in oligodendrocytes. Mol Med
[53] Miranda-Hernandez S, Baxter AG. Role of toll-like receptors in multiple sclerosis. 2018;24(1):51.
Am J Clin Exp Immunol 2013;2(1):75–93. [80] Matsushima GK, Morell P. The neurotoxicant, cuprizone, as a model to study de-
[54] Miranda-Hernandez S, Gerlach N, Fletcher JM, Biros E, Mack M, Korner H, et al. myelination and remyelination in the central nervous system. Brain Pathol
Role for MyD88, TLR2 and TLR9 but not TLR1, TLR4 or TLR6 in experimental 2001;11(1):107–16.
autoimmune encephalomyelitis. J Immunol 2011;187(2):791–804. [81] Wasko NJ, Kulak MH, Paul D, Nicaise AM, Yeung ST, Nichols FC, et al. Systemic
[55] Reynolds JM, Pappu BP, Peng J, Martinez GJ, Zhang Y, Chung Y, et al. Toll-like TLR2 tolerance enhances central nervous system remyelination. J
receptor 2 signaling in CD4(+) T lymphocytes promotes T helper 17 responses and Neuroinflammation 2019;16(1):158.
regulates the pathogenesis of autoimmune disease. Immunity 2010;32(5):692–702. [82] Amici SA, Dong J, Guerau-de-Arellano M. Molecular mechanisms modulating the
[56] Shaw PJ, Barr MJ, Lukens JR, McGargill MA, Chi H, Mak TW, et al. Signaling via the phenotype of macrophages and microglia. Front Immunol 2017;8:1520.
RIP2 adaptor protein in central nervous system-infiltrating dendritic cells promotes [83] Cherry JD, Olschowka JA, O’Banion MK. Neuroinflammation and M2 microglia: the
inflammation and autoimmunity. Immunity 2011;34(1):75–84. good, the bad, and the inflamed. J Neuroinflammation 2014;11:98.
[57] Andersson A, Covacu R, Sunnemark D, Danilov AI, Dal BA, Khademi M, et al. [84] Nakagawa Y, Chiba K. Diversity and plasticity of microglial cells in psychiatric and
Pivotal advance: HMGB1 expression in active lesions of human and experimental neurological disorders. Pharmacol Ther 2015;154:21–35.
multiple sclerosis. J Leukoc Biol 2008;84(5):1248–55. [85] Miron VE, Boyd A, Zhao JW, Yuen TJ, Ruckh JM, Shadrach JL, et al. M2 microglia
[58] Robinson AP, Caldis MW, Harp CT, Goings GE, Miller SD. High-mobility group box and macrophages drive oligodendrocyte differentiation during CNS remyelination.
1 protein (HMGB1) neutralization ameliorates experimental autoimmune en- Nat Neurosci 2013;16(9):1211–8.
cephalomyelitis. J Autoimmun 2013;43:32–43. [86] Miron VE, Franklin RJ. Macrophages and CNS remyelination. J Neurochem
[59] Uzawa A, Mori M, Taniguchi J, Masuda S, Muto M, Kuwabara S. Anti-high mobility 2014;130(2):165–71.
group box 1 monoclonal antibody ameliorates experimental autoimmune en- [87] Laflamme N, Cisbani G, Prefontaine P, Srour Y, Bernier J, St-Pierre MK, et al. mCSF-
cephalomyelitis. Clin Exp Immunol 2013;172(1):37–43. induced microglial activation prevents myelin loss and promotes its repair in a
[60] Fujiwara M, Anstadt EJ, Flynn B, Morse K, Ng C, Paczkowski P, et al. Enhanced mouse model of multiple sclerosis. Front Cell Neurosci 2018;12:178.
TLR2 responses in multiple sclerosis. Clin Exp Immunol 2018;193(3):313–26. [88] Lloyd AF, Davies CL, Miron VE. Microglia: origins, homeostasis, and roles in myelin
[61] Medvedev AE, Sabroe I, Hasday JD, Vogel SN. Tolerance to microbial TLR ligands: repair. Curr Opin Neurobiol 2017;47:113–20.

10

You might also like