N-Acetylglucosamine drives myelination by triggering oligodendrocyte precursor cell differentiation

Page created by Karl Martin
 
CONTINUE READING
N-Acetylglucosamine drives myelination by triggering oligodendrocyte precursor cell differentiation
JBC Papers in Press. Published on September 25, 2020 as Manuscript RA120.015595
       The latest version is at https://www.jbc.org/cgi/doi/10.1074/jbc.RA120.015595

       N-Acetylglucosamine drives myelination by triggering
            oligodendrocyte precursor cell differentiation

    Michael Sy1, Alexander U. Brandt1,2,3, Sung-Uk Lee1, Barbara L. Newton1, Judy Pawling4,
      Autreen Golzar1, Anas A. Rahman6, Zhaoxia Yu7, Graham Cooper2,3, Michael Scheel3,
                Friedemann Paul2,3,8, James W. Dennis4,6, Michael Demetriou1,5$

                                                                                                 Downloaded from http://www.jbc.org/ by guest on November 9, 2020
1
  Department of Neurology, University of California Irvine, 208 Sprague Hall, Mail Code 4032,
Irvine, CA 92697, USA.
2
  Experimental and Clinical Research Center, Charité – Universitätsmedizin Berlin and Max-
Delbrueck-Center for Molecular Medicine, Lindenberger Weg 80, 13125 Berlin, Germany.
3
  NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, corporate member
of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health,
Charitéplatz 1, 10117 Berlin, Germany.
4
  Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 600 University Ave, Toronto, ON
M5G 1X5, Canada.
5
  Department of Microbiology and Molecular Genetics, University of California Irvine.
6
  Department of Molecular Genetics, University of Toronto, ON M5S 1A8, Canada.
7
  Department of Statistics, Donald Bren School of Information and Computer Sciences
University of California Irvine, Bren Hall 2019, Irvine, CA 92697, USA
8
  Department of Neurology, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin,
Germany.
$
 Correspondence to: Michael Demetriou, Department of Neurology, University of California
Irvine, 208 Sprague Hall, Irvine, CA 92697, USA; Phone: (949) 824-9775, Email:
mdemetriou@uci.edu.

Running title: N-acetylglucosamine and myelination

Keywords: N-glycan branching, N-acetylglucosamine, oligodendrocytes, myelination, myelin
repair, multiple sclerosis.

                                              1
N-Acetylglucosamine drives myelination by triggering oligodendrocyte precursor cell differentiation
Abstract                                             oligodendrocytes is often incomplete despite
                                                     the presence of abundant oligodendrocyte
Myelination plays an important role in               precursor cells (OPC) throughout the brain
cognitive development and in demyelinating           (6-10). The molecular mechanisms that
diseases like multiple sclerosis (MS), where         block     re-myelination     in    MS      are
failure     of     re-myelination    promotes        incompletely understood and there is a lack
permanent          neuro-axonal       damage.        of therapies to promote myelin repair.
Modification of cell surface receptors with          Failure to adequately re-myelinate is
branched N-glycans coordinates cell growth           influenced by the microenvironment of the
and      differentiation     by    controlling       MS lesion, where reactive astrocytes,
glycoprotein clustering, signaling and               microglia, and macrophages produce various
endocytosis. N-acetylglucosamine (GlcNAc)            inhibitory factors leading to disruption in
is a rate-limiting metabolite for N-glycan           OPC       differentiation,    oligodendrocyte
branching. Here we report that GlcNAc and            migration,      process    outgrowth,     and
N-glycan             branching         trigger       attachment to axons (11). Multiple studies
oligodendrogenesis from precursor cells by           have identified molecules that limit OPC

                                                                                                      Downloaded from http://www.jbc.org/ by guest on November 9, 2020
inhibiting PDGF receptor-α cell endocytosis.         differentiation into myelin producing cells
Supplying oral GlcNAc to lactating mice              including      LINGO-1       (12),    various
drives primary myelination in newborn pups           extracellular matrix proteins (13,14), and
via secretion in breast milk, while                  myelin debris (15). Thus, increasing OPC
genetically blocking N-glycan branching              differentiation has become an important
markedly inhibits primary myelination. In            strategy for promoting remyelination in MS
adult mice with toxin (cuprizone) induced            and other demyelinating diseases (16).
demyelination, oral GlcNAc prevents neuro-
axonal damage by driving myelin repair. In           Cell surface and secreted proteins are co-
MS patients, endogenous serum GlcNAc                 and post-translationally modified on Asn(N)
levels inversely correlated with imaging             by the addition of carbohydrates (N-glycans)
measures        of      demyelination      and       in the endoplasmic reticulum and
microstructural damage. Our data identifies          subsequently remodeled in the Golgi. The
N-glycan branching and GlcNAc as critical            degree of N-acetylglucosamine (GlcNAc)
regulators of primary myelination and                branching in N-glycans promotes binding to
myelin repair and suggests oral GlcNAc               galectins, a family of sugar-binding proteins
may be neuro-protective in demyelinating             (Fig. S1A). Poly-valent galectin –
diseases like MS                                     glycoprotein interactions at the cell surface
                                                     form a macromolecular lattice that
Introduction                                         simultaneously controls the movement,
                                                     clustering and/or endocytosis of multiple
Myelination of axons by oligodendrocytes in          receptors and transporters to control
the central nervous system plays an critical         signaling, cell growth, differentiation and
role in normal cognitive development and             death (17-24). For example, N-glycan
function as well as in demyelinating disease         branching controls epithelial cell growth by
such as multiple sclerosis (MS) (1,2). In            regulating     receptor   tyrosine    kinases
addition to speeding conduction of the               endocytosis (17-20), promotes glucose
action potential, myelination supports axon          uptake in mesenchymal and pancreatic b
health and survival (3-5). In MS, re-                cells by inhibiting glucose transporter
myelination of demyelinated axons by                 endocytosis (19,25) and reduces T cells, B

                                                 2
N-Acetylglucosamine drives myelination by triggering oligodendrocyte precursor cell differentiation
cell and neutrophil pro-inflammatory                 oligodendrocyte progenitor/precursor cells
responses by co-regulating the clustering            (OPC)(42). In epithelial cells, N-glycan
and/or endocytosis of multiple glycoproteins         branching deficiency reduces PDGFRα
(17,23,26-29). These mechanisms in turn              surface expression by enhancing loss via
impact cancer, type II diabetes and                  endocytosis, leading to reduced signaling
autoimmunity (20). For example, reductions           (18). Thus, here we examine the hypothesis
in N-glycan branching are associated with            that GlcNAc may provide an oral
MS and promote both inflammatory                     therapeutic to raise N-glycan branching in
demyelination and neurodegeneration in               OPC’s, promote myelination and reduce the
mice, the latter by an unknown mechanism             potential for neurodegeneration by initiating
(17,30-36).                                          oligodendrocyte differentiation via enhanced
                                                     PDGFRα surface expression and signaling
Given the diverse and pleiotropic effects of         in OPC’s.
N-glycan branching, identifying and
manipulating regulatory mechanisms may               Results
provide new insights into disease

                                                                                                     Downloaded from http://www.jbc.org/ by guest on November 9, 2020
pathogenesis    and     opportunities    for         GlcNAc and N-glycan branching trigger
therapeutic intervention. In this regard,            oligodendrogenesis  by      inhibiting
metabolism is a critical regulator of N-             PDGFRα endocytosis
glycan branching by controlling availability
of the sugar-nucleotide UDP-GlcNAc, the              We examined oligodendrogenesis in vitro
substrate used by the Mgat family of N-              using mouse neural stem cells (NSC)
glycan branching enzymes (19,20,24,37,38).           derived from the medial ganglionic
UDP-GlcNAc is generated in the                       eminence of E12.5 mouse embryos, where
hexosamine pathway de novo from glucose              OPC’s first appear. GlcNAc treatment of
or by salvage from N-acetylglucosamine               NSCs for 48hrs in growth media lacking
(GlcNAc). Extracellular GlcNAc enters                exogenous differentiation cytokines (i.e. no
cells through micropinocytosis, with                 PDGF-AA, T3, CNTF) significantly
supplementation of cells or mice with                increased N-glycan branching and PDGFRα
GlcNAc inhibiting pro-inflammatory T-cell            surface expression (Fig. 1A,B), the former
responses    and    murine    models      of         assessed by L-PHA (Phaseolus vulgaris
inflammatory demyelination by enhancing              leukoagglutinin) flow cytometry (17,22).
N-glycan branching (19,31,37,38).                    Consistent with increased PDGFRα surface
                                                     expression, GlcNAc also promoted pre-
Targeted deletion of galectin-3, a ligand for        oligodendrocyte differentiation as evidenced
N-glycan branching, leads to decreased               by       augmented        expression       of
production of oligodendrocytes, poor                 Oligodendrocyte      Transcription     Factor
myelination of axons and reduced ability to          (OLIG2) and increased numbers of O4 and
re-myelinate after injury (39). In humans,           GalC positive cells (Fig. 1A-C, S1B,C).
loss of function mutations in PGM3, a gene           Double staining for PDGFRα and O4
required to generate branched N-glycans              revealed     that     GlcNAc       promoted
from GlcNAc, display reduced branching               development       of   pre-oligodendrocytes
and severe CNS hypomyelination (40).                 (PDGFRα+O4+) with no change in the
Platelet derived growth Factor – AA plays a          number of OPC’s (PDGFRα+O4-) (Fig. 1D).
critical role in oligodendrogenesis (41), with       Thus, after only two days of culture,
its receptor (PDGFRα) expressed in                   GlcNAc initiated oligodendrogenesis from

                                                 3
NSC’s despite the absence of exogenous               Next, we examined whether oral GlcNAc
differentiation cytokines such as PDGF-AA.           can cross the blood-brain barrier to promote
Remarkably, GlcNAc in growth media was               oligodendrocyte         differentiation      and
more potent than differentiation media               myelination in vivo. Adult mice (n=6) and
containing exogenous PDGF-AA at                      lactating     mothers        were       provided
                                                                           13
initiating oligodendrogenesis (Fig. S1D).            with/without         C -labelled        GlcNAc
Combining GlcNAc with PDGF-AA also                   ([U13C]GlcNAc) (10) in their drinking water
enhanced NSC differentiation to O4+ pre-             and metabolites derived from perfused
oligodendrocytes (Fig. S1E).                         brains     were      analyzed      by     Liquid
                                                     chromatography         -      tandem       mass
To confirm a role for N-glycan branching in          spectroscopy (LC-MS/MS). Although this
oligodendrogenesis,      we     first    used        method does not resolve stereoisomers of N-
kifunensine to inhibit N-glycan branching            Acetylhexosamines (ie. GlcNAc versus
(23) in NSC induced to differentiate by              GalNAc),         a reversible       4-epimerase
exogenous PDGF-AA. Reducing branching                (GALE) equilibrates UDP-GlcNAc               and
in NSC’s using kifunensine significantly             UDP-GalNAc in vivo (19). LC-MS/MS

                                                                                                        Downloaded from http://www.jbc.org/ by guest on November 9, 2020
reduced PDGFRα surface expression and the            identified                      UDP-[U13C]-N-
number of O4+ cells induced by PDGF-AA               Acetylhexosamines (UDP-[U13C]-HexNAc)
differentiation media (Fig. S1F). To confirm         in treated adult female mouse brains as well
this result genetically, we utilized Mgat5-/-        as in the brains of their suckling pups (Fig.
and doxycycline inducible Mgat1f/f/tetO-             2A). This demonstrates that orally delivered
cre/ROSA-rtTA        mice (17,22). Mgat5             GlcNAc is not only able to cross the blood
deletion mildly reduces N-glycan branching           brain barrier and be metabolized to UDP-
while Mgat1 deletion completely blocks N-            GlcNAc by CNS cells, it is also secreted at
glycan branching (Fig. S1A). In vitro                sufficient levels in breast milk to raise UDP-
doxycycline treatment of NSC from                    GlcNAc in the brains of suckling pups.
Mgat1f/f/tetO-cre/ROSA-rtTA mice readily
induced deletion of Mgat1, as measured by            To assess whether oral GlcNAc promotes
loss of L-PHA binding (Fig. S1G). Mgat5              oligodendrogenesis in vivo in the absence of
and Mgat1 deleted NSC decreased surface              inflammation, we examined primary
levels of PDGFRα and were markedly                   myelination in mice during the early peri-
reduced in their ability to differentiate into       natal period. We provided GlcNAc or
O4+ pre-oligodendrocytes in response to              vehicle to pregnant/lactating female mice
PDGF-AA differentiation media (Fig 1E,               from E12.5, post-natal day 3 (P3) or P5
S1G). In Mgat5 heterozygous NSC’s, small             through to P8. Indeed, oral GlcNAc
reductions in N-glycan branching also                increased N-glycan branching in PDGFRα+
inhibited oligodendrocyte differentiation            cells as well as the number of pre-
(Fig. 1E). Thus, subtle changes in N-glycan          oligodendrocytes (PDGFRα+O4+), immature
branching       can      markedly      impact        oligodendrocytes      (PDGFRα-O4+)       and
oligodendrocyte differentiation from NSC in          mature oligodendrocytes (MBP+), with little
vitro.                                               effect on the number of OPC’s
                                                     (PDGFRα+O4-) (Fig. 2B, S2A). The lack of
GlcNAc and N-glycan branching promote                change in OPC number is consistent with
primary myelination in mice.                         our in vitro data (Fig. 1D) and suggests that
                                                     GlcNAc promotes OPC self-renewal and/or
                                                     NSC differentiation to OPC, resulting in a

                                                 4
stable number of OPC’s. Consistent with              tamoxifen induces deletion of Mgat1 in
increased oligodendrogenesis, oral GlcNAc            OPC’s but not mature oligodendrocytes.
also increased primary myelination when              Indeed, eight weeks but not two weeks after
provided to pups from P3-8, as assessed by           tamoxifen treatment, deletion of Mgat1 in
increased levels of staining for myelin basic        OPC’s significantly reduced levels of MBP,
protein (MBP) and myelin (as measured by             myelin (fluoromyelin) and number of total
fluoromyelin) (Fig. 2C).                             (Olig2+)     and    mature    (Olig2+CC1+)
                                                     oligodendrocytes along with increased
To confirm that N-glycan branching                   numbers      of   immature     (Olig2+CC1-)
promotes myelination in the absence of               oligodendrocytes (Fig 2E, S2E). Tamoxifen
inflammation in vivo, we generated mice              has     been     reported    to     promote
with tamoxifen inducible deletion of Mgat1           myelination(47), however Mgat1 deletion
only in OPC’s and oligodendrocytes, namely           reduced myelination despite potential
Mgat1f/fPlp1-cre/ERTc+ mice. As proteolipid          positive effects of tamoxifen. Together,
protein (PLP) promoter driven Cre                    these data demonstrate that GlcNAc and N-
expression only becomes restricted to the            glycan     branching    promote     primary

                                                                                                    Downloaded from http://www.jbc.org/ by guest on November 9, 2020
oligodendrocyte      lineage     (OPC     and        myelination in mice by driving OPC
oligodendrocyte) at P28 (43), we focused on          differentiation.
adult mice. OPC’s continue to proliferate
and generate significant new myelin in               GlcNAc prevents damage to demyelinated
adulthood, with myelination gradually                axons by promoting myelin repair.
doubling from ~2 to 10 months (44-46).
Tamoxifen readily induced Mgat1 deletion             To explore whether GlcNAc can promote re-
in O4+ oligodendrocytes but not 04— cells in         myelination in adult mice following myelin
vivo, as determined by loss of L-PHA                 injury, we utilized the cuprizone model of
binding by flow cytometry (Fig. S2B).                non-immune induced de-myelination/re-
Consistent with slow accumulation of new             myelination on Mgat5+/- and wildtype
myelin from OPC’s during adulthood, two              C57BL/6 mice. Cuprizone at 0.2% induces
weeks following tamoxifen treatment                  demyelination in the corpus callosum by 3
(Mgat1 deletion) adult Mgat1f/fPlp1-                 weeks, with maximum demyelination at 5-6
cre/ERTc+ mice did not display significant           weeks. Partial re-myelination via maturation
differences in brain levels of MBP or myelin         of OPC’s begins at the height of
(fluoromyelin) relative to tamoxifen treated         demyelination and becomes complete ~3-5
controls (Fig. S2C). However, eight weeks            weeks after cuprizone withdrawal. Given
after initial tamoxifen treatment, Mgat1             this, we examined four different treatment
deletion resulted in significant reductions in       regimens (Fig. 3A). When GlcNAc was
levels of MBP, myelin (fluoromyelin) and             concurrently provided during the final 3
number of total (Olig2+) and mature                  weeks of a 6-week cuprizone (0.2%)
(Olig2+CC1+) oligodendrocytes along with             exposure in wildtype mice, GlcNAc
increased numbers of immature (Olig2+CC1-)           prevented loss of motor function (as
oligodendrocytes (Fig. 2D, S2D). To                  measured using rotarod fall latency) while
confirm that these results primarily arose           increasing MBP levels and reducing axonal
from a defect in new myelin formation from           damage (as measured by reduced
OPC’s, rather than a defect in mature                accumulation of amyloid precursor protein
oligodendrocytes,         we        generated        (APP)) in the corpus callosum (Fig. 3C). To
       f/f             +
Mgat1 Pdgfra-creER            mice      where        address potential confounding effects of

                                                 5
GlcNAc on inhibiting demyelination by               patients to correlate endogenous serum
cuprizone during concurrent treatment, we           HexNAc levels with measures of white
initiated treatment of wildtype mice for 2          matter damage by magnetic resonance
weeks or Mgat5+/- mice for 1 or 4 weeks of          imaging (MRI) of the brain. Increased T2w
GlcNAc only after cuprizone was stopped             lesion volume and count on brain MRI are
(Fig. 3A). This revealed that GlcNAc                measures of the extent and frequency of
enhanced levels of MBP, myelin                      demyelination, respectively. T2w lesion
(fluoromyelin) and mature oligodendrocytes          volume correlated with lower HexNAc
(CC1+Olig2+) while reducing the amount of           serum levels (Fig. 4A, p=0.020), whereas
degraded          MBP        (dMBP)/myelin          T2w lesion count did not (p=0.387).
degeneration within the corpus callosum             Likewise, patients with contrast enhancing
(Fig. 3D-F). dMBP was detected by an                lesions, a marker of active inflammation in
antibody that specifically recognizes areas         MS, had similar serum HexNAc levels to
of myelin degeneration (48). Electron               those without (p=0.866), suggesting
microscopy analysis confirmed these results,        GlcNAc primarily impacts the extent of
revealing that GlcNAc enhanced the number           permanent demyelination rather than

                                                                                                   Downloaded from http://www.jbc.org/ by guest on November 9, 2020
of myelinated axons, and the degree of              initiation of inflammatory demyelination.
myelination as measured by the g-ratio,             T1w/T2w ratio maps (50) reflect
while also reducing axon loss and the               microstructural integrity of myelin/axons in
number         of     degenerating       and        normal appearing white matter (NAWM)
dystrophic/swollen axons (Fig. 3G, S3A).            (51) and cortical grey matter(52,53). With
GlcNAc also enhanced the number of                  age and gender as covariates, low serum
paranodes,      which    increase    with re-       HexNAc levels were strongly associated
myelination (Fig. S3A)(49). Enhancement             with lower T1w/T2w ratios indicating
of myelination by GlcNAc depends on time,           microstructural damage of myelin/axons in
as the increase in fluoromyelin staining in         both normal appearing white matter (r2=0.18,
Mgat5+/- mice was ~2-fold greater with 4            p=2.25×10−5) and grey matter (r2=0.23,
versus 1 week of GlcNAc treatment (Fig.             p=1.32×10−6), (Fig. 4B,C). Together, these
3D, F). Importantly, the subtle reductions in       data are consistent with our mouse data and
N-glycan branching induced in Mgat5+/-              suggest that GlcNAc may promote
mice did not alter baseline levels of myelin        myelination in MS.
yet reduced re-myelination following
cuprizone induced injury relative to                Discussion
Mgat5+/+ control mice (Fig. S3B, C).
Together, these data indicate that GlcNAc           Here we report a novel pathway for
and N-glycan branching promotes myelin              regulating oligodendrogenesis, primary
repair and provides neuro-protection to             myelination and myelin repair by N-glycan
axons following demyelination.                      branching and GlcNAc. Our data
                                                    demonstrates that GlcNAc and N-glycan
A marker of serum GlcNAc inversely                  branching     are    neuroprotective   for
associates with imaging markers of                  demyelinated      axons   by     promoting
myelin-axon damage.                                 oligodendrogenesis and myelination from
                                                    OPC’s. The association of low endogenous
To explore whether alterations in GlcNAc            GlcNAc with increased myelin-axon
may impact myelination status in MS                 microstructural damage in MS patients
patients, we used a cohort of 180 MS                suggests this mechanism is relevant to

                                                6
pathogenesis of MS. This hypothesis is               drive myelination and promote axonal health.
consistent with recent data suggesting that          For example, cell motility is significantly
some MS patients are blocked in their ability        enhanced by N-glycan branching via
to generate new myelin from progenitors (9).         reduced clustering of integrins (58,59).
The         mechanisms         that      drive       Such activity in OPC’s would enhance their
neurodegeneration in MS are poorly                   ability to traffic to sites of demyelination
understood and our data raise the possibility        and promote myelin repair.          N-glycan
that alterations in N-glycan branching and/or        branching      also     stimulates   glucose
GlcNAc        availability    may     promote        transporter surface retention, to enhance
neurodegeneration          by         blocking       glucose uptake (19,25). Glucose-transporter-
remyelination. Indeed, we find that low              1 (GLUT1) in oligodendrocytes promotes
levels of serum GlcNAc in MS patients is             axonal health and function by increasing
associated with a progressive disease course,        transfer of lactate to axons via increased
clinical disability and multiple neuroimaging        glucose supply to the glycolytic pathway in
measures of neurodegeneration (unpublished           oligodendrocytes (60). Thus, part of the
data).                                               neuroprotective effect of GlcNAc following

                                                                                                      Downloaded from http://www.jbc.org/ by guest on November 9, 2020
                                                     myelin repair may be through enhanced
Providing oral GlcNAc to lactating female            transport of glucose into oligodendrocytes.
mice increased primary myelination in
nursing pups via delivery of GlcNAc in               GlcNAc and/or N-glycan branching also
breast milk. In humans, GlcNAc is a major            play important roles in suppressing T cell
component of breast milk oligosaccharides            and B cell mediated inflammatory
(~1.5 to ~0.6mg/ml from term to 13                   demyelination (17,28,30-33,35,37,38). . In
weeks)(54), and can be released as a                 T cells, GlcNAc and N-glycan branching
monosaccharide by infant microbiota (55).            suppress activation signaling via the T cell
Thus, breast fed newborns consume ~0.5 -             receptor     (17,22,26),      inhibit    pro-
1.5g of GlcNAc per day or ~100-                      inflammatory TH1 and TH17 differentiation
300mg/kg/day for a 5kg infant. This is               (24,38) and enhance anti-inflammatory T
similar to the ~160mg/kg/day dose that we            regulatory cell differentiation (24). B cell
observed to promote myelination in adult             depletion is a potent therapy in MS,
mice. In contrast to human breast milk,              predominantly acting by suppressing innate
GlcNAc is not a significant component of             antigen presenting cell function rather than
commercial baby formula.         Breast-fed          via antibody production (61,62). N-glycan
infants display increased myelination and            branching in B cells reduces pro-
cognitive function compared to formula fed           inflammatory innate signaling via toll-like
infants (56,57), but the mechanism is                receptors and inhibits antigen presenting cell
unknown. Our data suggests that GlcNAc in            activity, yet promotes adaptive responses
human breast-milk may be a major driver of           through the B cell receptor(28). Thus, oral
this effect.                                         GlcNAc is uniquely positioned as a
                                                     therapeutic to reverse three major targets
GlcNAc and N-glycan branching markedly               driving MS pathogenesis, namely pro-
enhanced cell surface expression of                  inflammatory T cell responses, pro-
PDGFRα, a critical initiator of OPC                  inflammatory innate B cell activity and
differentiation. However, GlcNAc and N-              myelin repair. No current MS therapy has
glycan branching likely impact other cell            such diverse mechanisms of action.
surface receptors/transporters in OPC’s to

                                                 7
Concentrations of GlcNAc required to raise           Mice were bred and utilized as approved by
N-glycan branching in vivo are markedly              the University of California, Irvine
lower than those required for in vitro               Institutional Animal Care and Use
activity (37,38). This is largely driven by          Committee. Dorsal forebrain cortical tissue
GlcNAc entering cells by macropinocytosis,           was dissected from the medial ganglionic
and therefore both time and rate of                  eminence (MGE) at embryonic day 12.5
membrane        turnover     can     influence       (E12) of CD1 mice (Charles River) or
concentrations of GlcNAc required to raise           Mgat5-/- C57BL/6 mice and their wildtype
N-glycan branching (37,38). Thus, short-             littermates and placed in dissection buffer:
term in vitro experiments require high               PBS, 0.6% glucose, 50 U/mL Pen/Strep.
concentrations to raise intracellular GlcNAc         Tissue from multiple embryos within the
levels quickly while primary cells remain            same litter were pooled, and a subsequent
viable. In contrast, cells can be exposed to         culture from a single litter was considered a
GlcNAc over a longer time period in vivo,            biological repeat. The tissue was dissociated
allowing lower concentrations to be                  using 0.05% Trypsin-EDTA at 37o C for 10
effective at raising N-glycan branching. The         min, followed by treatment with soybean

                                                                                                     Downloaded from http://www.jbc.org/ by guest on November 9, 2020
rate of macropinocytosis may also be much            trypsin inhibitor (Life Technologies).
higher in vivo compared to in vitro.                 Dissociated cells were re-suspended in
                                                     proliferation medium containing DMEM, 1x
GlcNAc is also known to be highly safe in            B27, 1x N2, 1 mM sodium pyruvate, 2 mM
humans. In addition to breastfed infants             L-glutamine, 1 mM N-acetylcysteine, 20
consuming significant quantities, large              ng/mL EGF (PeproTech), 10 ng/mL bFGF
intravenous doses of GlcNAc (20g and 100g)           (PeproTech), and 2 µg/mL heparin and
in humans demonstrated no toxicity issues            seeded at 150,000 cells/mL (non-tissue
and no alterations in blood glucose or               culture treated plastic plates) and grown as
insulin (63,64). Moreover, treatment with            non-adherent spheres. Cell cultures were
insulin had no effect on the serum half-life         passaged approximately every 3 days using
of GlcNAc (63). Oral GlcNAc (3-6g/day)               enzyme-free        NeuroCult        Chemical
has also been used in 12 children with               Dissociation Kit (Mouse) (StemCell
inflammatory bowel disease for ~2 years              Technologies). Cultures were passaged at
without reported toxicities and/or side              least once prior to experimental use. For
effects (65). In rats, chronic systematic            experiments, passaged cells were cultured in
toxicological studies at doses of 2323-              proliferation media (bFGF and EGF) or
2545mg/kg/day for up to 114 weeks found              differentiation media (bFGF (10ng/ml) and
no toxicity (66,67). Coupled with                    PDGF-AA (10ng/ml); Life technologies) for
availability as a dietary supplement, oral           48 hours with or without the presence of
GlcNAc may provide a potent, inexpensive             GlcNAc (Ultimate Glucosamine, Wellesley
and safe therapy for MS. Large double-blind          Therapeutics) or kifunensine (GlycoSyn).
placebo-controlled trials are warranted to           Neurospheres were dispersed using the
investigate this hypothesis.                         enzyme-free NeuroCult kit before being
                                                     analyzed by flow cytometry using one or
Experimental procedures                              more of the following antibodies: anti-
                                                     CD140a/PDGF-RA PE conjugate (1:200,
Mouse brain and neural stem               cell       A15785, Molecular Probes), anti-O4 Alexa
isolation and analysis                               Fluor 488 conjugated (1:200, FAB1326G,
                                                     RnD systems), Anti-GalC Alexa Fluor 647

                                                 8
(1:200, MAB342-AF647, Millipore), Anti-             of 20mg/mL. Mgat1f/fPlp1-cre/ERTc+ and
Olig2 (1:200, AB9610, Millipore) with anti-         Mgat1f/fPdgfra-creER+ (mean age: P71.24,
rabbit Alexa Fluor 488 (1:200, thermofisher).       std. dev. 1.393) mice and their control
GlcNAc treatment of mouse pups - GlcNAc             Mgat1f/f     littermates   were     injected
(1mg/mL) in drinking water was provided to          intraperitoneally with tamoxifen (75mg/kg)
pregnant PLJ mothers or mothers who                 daily for 3 days starting on day 0 and
recently delivered pups and were nursing            sacrificed at two weeks or re-treated with
their young. After the treatment period, pups       tamoxifen and sacrificed at 8 weeks. Mice
were anestheszied with isofluorane and              were sacrificed following anesthesia and
cardiac perfused with PBS. Pup and fetal            cardiac perfusion with phosphate buffered
brains were removed and homogenized by              saline. Brains examined by flow cytometry
trituration using glass pipettes in PBS with        were first homogenized by trituration using
5% Fetal Bovine Serum (FBS). Cells were             glass pipettes in PBS with 5% FBS. Brains
then stained with antibodies and analyzed by        examined for myelin content were drop
flow cytometry using antibodies described           fixed in 4% paraformaldehyde overnight.
above. For immunofluorescence analysis of

                                                                                                     Downloaded from http://www.jbc.org/ by guest on November 9, 2020
pup brains, pups were quickly decapitated,          Cuprizone induced demyelination
brains harvested and fixed in 4%
paraformaldehyde overnight.                         Cuprizone at 0.2% induces demyelination in
                                                    the corpus callosum by 3 weeks, with
[U13C]GlcNAc treatment of mice                      maximum demyelination at 5-6 weeks(68).
                                                    8-week-old C57BL/6 mice purchased from
[U13C] GlcNAc was purchased from                    Jackson Laboratories or 8-week old Mgat5+/-
Omicron Biochemicals and put in the                 C57BL/6 mice were treated with 0.2%
drinking water at 1mg/mL of female mice             Cuprizone (Sigma) mixed into milled rodent
aged 8 weeks for 3 days. Fresh solution of          chow for 6 weeks for the active phase
[U13C] GlcNAc in drinking water was                 treatment and 5 weeks for the recovery
provided each day. After 3 days, mice were          treatment. During active phase treatment,
anesthetized with isofluorane and underwent         GlcNAc (1mg/mL) in drinking water or just
cardiac perfusion with 50mL of PBS. Brains          drinking water (control) was provided for
were harvested and snap frozen in liquid            the last 3 weeks of Cuprizone treatment. For
nitrogen. Tissues were cut into 0.04g pieces        the recovery phase treatment, GlcNAc in
and crushed mechanically before undergoing          drinking water or control was provided after
extraction as described below (Targeted LC-         Cuprizone treatment had been stopped.
MS/MS). Levels of UDP-[U13C]GlcNAc                  Mice were anesthetized and underwent
were measured by LC–MS/MS analysis as               cardiac       perfusion        with       4%
described below (Targeted LC-MS/MS).                paraformaldehyde      in   PBS      or    4%
                                                    paraformaldehyde plus 0.5% glutaraldehyde
Tamoxifen induced deletion of Mgat1                 in    Sodium      cacodylate    buffer     for
                                                    immunofluorescence          or       electron
Mgat1f/fPlp1-cre/ERTc+ and Mgat1f/fPdgfra-          microscopic analysis respectively. Brains
creER+ were generated by crossing our               were then fixed overnight in perfusion
Mgat1f/f mice with Plp1-cre/ERTc+ and               solution.
Pdgfra-creER+ lines       from     Jackson’s
Laboratory. Tamoxifen was dissolved in              Accelerated Rotarod
corn oil overnight at 37°C at a concentration

                                                9
One day prior to Cuprizone treatment, mice             anti-rat    Alexa-fluor     488     (1:200,
were trained on the rotarod by allowing                Thermofisher), goat anti-rabbit TxRd (1:200,
them to run three 5-minute trials at a                 Thermofisher). APP is a marker for neuro-
constant 30 rotations per minute (RPM).                axonal damage(69-71) while co-staining for
Mice then underwent weekly testing during              CC1 and Olig2 are markers of mature
Cuprizone and GlcNAc treatment on an                   oligodendrocytes(72,73). In order to
accelerating rotarod starting at 4 rpm                 examine the amount of myelin, slices were
increasing to 40 rpm over 5 minutes.                   incubated in Fluoromyelin (1:300; F34651,
Latency for mice to fall was recorded. If a            Thermofisher) for 45 minutes. Images were
mouse was not running on the rotarod by                acquired on a Keyence fluorescence
holding on for 3 turns, this was considered a          microscope. Mean fluorescence intensity of
fall. For the active phase treatment, one trial        the medial corpus callosum was measured
was run every week. For the recovery phase             using ImageJ.
treatment, 3 trials were run for each mouse
each week and latencies were averaged. As              Electron Microscopy
expected      with    Cuprizone     treatment

                                                                                                       Downloaded from http://www.jbc.org/ by guest on November 9, 2020
performance       degraded     as   treatment          Three mice from each treatment group
progressed. Mice whose performance did                 (control and GlcNAc) were selected
not drop below a pre-determined threshold              randomly for EM analysis (before other
(200 seconds) were not used in analysis.               investigations were performed). Portions of
                                                       these brains from 0 to -1 bregma were rinsed
Immunofluorescence analysis                            in 0.1 M cacodylate buffer overnight and
                                                       again for 15 min. the next day. 2 x 1 mm
For NSC immunoflouresence, whole                       blocks of the corpus callosum were
neurospheres were seeded onto laminin-                 dissected out and post-fixed with 1 %
coated coverslips (Neuvitro) in proliferation          osmium tetroxide in 0.1 M cacodylate buffer
medium. After 24 hours, proliferation media            for 1 hour, rinsed in ddH20, dehydrated in
was     removed     and    replaced     with           increasing serial dilutions of ethanol (70%,
differentiation medium (same components                85%, 95%, 100% × 2) for 10 min each, put
as proliferation medium but excluding EGF,             in propylene oxide (intermediate solvent) for
bFGF, and heparin) to induce differentiation.          2 x 10 min, incubated in propylene
For analysis of mouse brains, brains were              oxide/Spurr's resin (1:1 mix) for 1 h, and
incubated in 30% sucrose for at least 72               then in Spurr's resin overnight. The blocks
hours, embedded in OCT (Tissue-tek),                   were put in a fresh change of resin in flat
frozen for at least 48 hours at -80οC, and             embedding molds the next day and
then cut at 40 microns on a cryostat.                  polymerized overnight at 60 ° Celsius. The
Multiple sections from -1 bregma to -2.5               blocks were sectioned at 1 µm using a Leica
bregma were then stained with antibodies               Ultracut UCT ultramicrotome. Floating
for MBP (1:100; MAB386, Millipore),                    sections were stained in toluidine blue
Olig2 (1:200; AB9610, Millipore), CC1                  (1 % toluidine blue and 2 % sodium borate
(1:100; OP90, Millipore), degraded MBP(48)             in ddH20) at 60 ° C for 3 min., mounted on
(1:200, AB5864, Millipore) and Amyloid                 slides and cover-slipped. Ultrathin sections
Precursor Protein (APP, 1:200; clone                   were sectioned at 70 nm using a Leica
22C11). After overnight incubation with                Ultracut UCT ultramicrotome. Sections
primary antibody, tissues were washed and              were mounted on 150 mesh copper grids,
incubated with secondary antibodies: goat              stained with uranyl acetate and lead citrate

                                                  10
and viewed using a JEOL 1400 electron                MRI was performed at 1.5 Tesla using three-
microscope. Images were captured using a             dimensional T1-weighted magnetization
Gatan digital camera. A blinded rater                prepared rapid acquisition and multiple
analyzed images by calculating the g-ratio           gradient echo sequences (MPRAGE; T1w)
(ratio of the diameter of the axon excluding         and axial T2-weighted (T2w) sequences.
the myelin sheath divided by the axon                Images were either acquired on a Sonata
diameter including the myelin sheath) as             MRI (Siemens Medical Systems, Erlangen,
well as counting the number of total axons,          Germany) with TE 4.38 ms, TR 2,110 ms,
myelinated axons, dystrophic axons (defined          TI 1.1 ms, flip angle 15° and isotropic
as axon diameter >0.7µm), degenerating               resolutions 1 mm3 for T1w, and Multiecho
axons and paranodes. Degenerating axons              TSE with TE 81 ms, TR 5,780 ms, 150° flip
were identified as axonal swellings                  angle, resolution 0.5x0.5x3 mm, no gap for
containing more than 5 clustered dark                T2w, or on an Avanto MRI (Siemens
mitochondria and lysosomes. Paranodes                Medical Systems, Erlangen, Germany) with
were identified as axons with close                  TE 3.09 ms, TR 1,900 ms, TI 1.1 ms, flip
proximity of the axolemma with the inner             angle 15° and isotropic resolutions 1 mm3

                                                                                                   Downloaded from http://www.jbc.org/ by guest on November 9, 2020
membrane of the myelin sheath with a                 for T1w, and 3D TSE with TE 175 ms, TR
surrounding      cytoplasmic    portion   of         3,000 ms, flip angle 120°, isotropic
oligodendrocyte.                                     resolutions 1 mm3 for T2w. Conventional
                                                     spin-echo T1-weighted images (TR 1060 ms,
MS patient cohort                                    TE 14 ms, 3 mm slice thickness, no gap and
                                                     44 contiguous axial slices) were obtained
MS patients were recruited from the                  before and 5 minutes after injection of 0.1
neuroimmunology clinical trial unit at the           mmol/kg Gd-DTPA (Magnevist, Bayer-
NeuroCure Clinical Research Center,                  Schering, Berlin, Germany).
Charité - Universitätsmedizin Berlin (Table
S1). Inclusion criteria were MS based on the         T2w lesion segmentation was performed as
2010 revised McDonald criteria2, stable              previously described(75) using a semi-
immunomodulatory therapy (RRMS) or no                automated procedure including image co-
treatment (PPMS and SPMS). Exclusion                 registration using FLIRT (FMRIB Software
criteria were acute relapse and/or                   Library, Oxford, UK) and inhomogeneity
corticosteroids within 6 months prior to             correction as embedded into the MedX
inclusion. Disease course was determined             v3.4.3 software package (Sensor Systems
under strict adherence to the 1996 Lublin            Inc., Sterling, VA, USA). Bulk white matter
criteria(74). Blood draws were fasting. The          lesion load and lesion count of T2w scans
study was approved by the local ethics               were routinely measured using MedX.
committee of Berlin (Landesamt für
Gesundheit und Soziales (LAGeSo)). All               For calculation of T1w/T2w ratio maps,
study participants gave written informed             MPRAGE, FLAIR and T2w scans were
consent. Studies were conducted in                   reoriented to standard space, bias field
conformity with the 1964 Declaration of              corrected and cropped to a robust field of
Helsinki in its currently applicable version.        view using FSL 5.0.9(76). The MPRAGE
                                                     and FLAIR scans were then linearly co-
Magnetic Resonance Imaging (MRI)                     registered to T2w using FSL FLIRT and
                                                     then registered to MNI (Montreal
                                                     Neurological Institute) space and brain

                                                11
extracted using the Brain Extraction                 spectrometer (AB Sciex 4000Qtrap, Toronto,
Toolbox (BET)(76). T2w lesions were then             ON, Canada). MultiQuant software (AB
automatically segmented by applying the              Sciex, Version 2.1) was used for peak
lesion prediction algorithm (LPA) to FLAIR           analysis and manual peak confirmation. The
scans, implemented in the Lesion                     results, expressed as area ratio (area of
Segmentation Toolbox version 2.0.15(77)              analyte/area of internal standard), were
for SPM. GM, WM and brain masks were                 exported to Excel, and analyzed with
then extracted from the MPRAGE. The                  MetaboAnalyst 3.0. Standard curves were
lesion mask was subtracted from these                prepared      by      adding    increasing
masks to remove any lesion effects. The              concentrations of GlcNAc or N-Acetyl-D-
T1w/T2w ratio was created by dividing the            [UL-13C6]glucosamine ([UL13C6] GlcNAc)
processed MPRAGE scans by the processed              (Omicron Biochemicals, Indiana) to 50 µl
T2w scans. Median T1w/T2w ratios were                aliquot of control serum (Fig. S4A). This
extracted from the normal appearing WM,              way we were able to create a calibration
GM and brain masks.                                  curve for HexNAc serum levels, obtaining
                                                     absolute values rather than relative

                                                                                                     Downloaded from http://www.jbc.org/ by guest on November 9, 2020
Targeted LC-MS/MS                                    concentrations (Fig. S4B). Analysts were
                                                     blinded in regard to sample origin (HC or
Serum samples for metabolomics analysis              MS).
were prepared as described previously(78).
Briefly, 50 µL serum (stored at -80oC) and           Statistical Analysis
200 µl ice cold extraction solvent (40%
acetonitrile: 40% methanol: 20% H2O), were           Statistical analyses for the in vitro and
vortexed for 2 minutes, then shaken in an            animal experiments were done with
Eppendorf shaker (Thermomixer R) at 1400             Graphpad Prism by t-tests, ANOVA with
rpm, 4°C for 1 hour and centrifuged at 4°C           Sidak’s post-test correction or comparing
for 10 minutes at ~18,000 x g in an                  best fit curves from non-linear regression
Eppendorf microfuge. Supernatants were               (Y=Bmax*X/(Kd + X) as described in the
transferred to a clean tube and evaporated in        relevant figure legends. Statistical analyses
a Speedvac (Acid-Resistant CentriVap                 for the clinical part were performed with R
Vacuum Concentrators, Labconco). Dried               Project version 3.5.3. Correlation between
samples were stored at -80°C. Samples were           serum HexNAc levels and lesion
resuspended in 100 µl of water containing            measurements were analyzed using non-
the Internal Standards D7-Glucose at 0.2             parametric Spearman’s Rho analyses.
mg/mL       and     H-Tyrosine     at    0.02        Correlations between HexNAc levels and
mg/ml. Samples were resolved by LC-                  T1w/T2w-ration        measurements      were
MS/MS, in negative mode at the optimum               analyzed using linear regression models
polarity in MRM mode on an electrospray              with HexNAc levels as an independent
ionization (ESI) triple-quadrupole mass              variable.

Data Availability
All data are contained within the manuscript.

Acknowledgements

                                                12
We thank Cynthia Kraut and Susan Pikol for MRI assistance and Bibiane Seeger and Carey F. Li
for laboratory assistance. We thank Oswald Steward and Ilse Sears-Kraxberger for providing
electron microscopy services.

Author Contributions
MS (Sy) conceived and coordinated the study with AUB and MD, performed the in vitro and in
vivo animal experiments along with SUL, and wrote the manuscript together with MD and AUB.
AUB conceived and coordinated the study with MD and MS, participated in clinical data
collection, contributed to image analysis and statistical analysis, and wrote the manuscript
together with MS and MD.
SUL maintained animal colonies and performed in vivo animal experiments

                                                                                                    Downloaded from http://www.jbc.org/ by guest on November 9, 2020
BLN assisted with study coordination.
JP performed mass spectroscopy.
AG helped perform in vitro experiments and was the blinded rater for in vivo experiments
AAR developed the LC-MS/MS analysis to detect serum GlcNAc.
ZY performed statistical planning of the interventional study and contributed to statistical
analysis of the whole study.
GC calculated the T1w/T2w ratio measurements.
MS (Scheel) coordinated MRI measurements and setup MRI protocols
FP planned and supervised the clinical study.
JWD planned and coordinated mass spectroscopy measurements and analysis.
MD conceived and coordinated the study with AUB and MS, coordinated the serum analysis,
supervised the mouse studies and wrote the manuscript together with MS and AUB.

Funding
Research was supported by a Marilyn Hilton innovator award to MS as well as R01AT007452
and R01AI144403 to MD. The MS cohort and MRI study was supported by DFG grant Exc. 257
to FP. The content is solely the responsibility of the authors and does not necessarily represent
the official views of the National Institutes of Health.

Conflict of Interest

                                                 13
AUB, FP, JD and MD are named as inventors on a patent that describes GlcNAc as a biomarker
for multiple sclerosis. JD and MD are named as inventors on a patent for use of GlcNAc in MS
and were co-founders of Glixis Therapeutics, a company developing analogs of GlcNAc for MS
and other autoimmune diseases.

References
1.     Reich, D. S., Lucchinetti, C. F., and Calabresi, P. A. (2018) Multiple Sclerosis. N Engl J
       Med 378, 169-180
2.     Fields, R. D. (2008) White matter in learning, cognition and psychiatric disorders. Trends
       Neurosci 31, 361-370
3.     Lee, Y., Morrison, B. M., Li, Y., Lengacher, S., Farah, M. H., Hoffman, P. N., Liu, Y.,
       Tsingalia, A., Jin, L., Zhang, P. W., Pellerin, L., Magistretti, P. J., and Rothstein, J. D.
       (2012) Oligodendroglia metabolically support axons and contribute to neurodegeneration.

                                                                                                      Downloaded from http://www.jbc.org/ by guest on November 9, 2020
       Nature 487, 443-448
4.     Fünfschilling, U., Supplie, L. M., Mahad, D., Boretius, S., Saab, A. S., Edgar, J.,
       Brinkmann, B. G., Kassmann, C. M., Tzvetanova, I. D., Möbius, W., Diaz, F., Meijer, D.,
       Suter, U., Hamprecht, B., Sereda, M. W., Moraes, C. T., Frahm, J., Goebbels, S., and
       Nave, K. A. (2012) Glycolytic oligodendrocytes maintain myelin and long-term axonal
       integrity. Nature 485, 517-521
5.     Mei, F., Lehmann-Horn, K., Shen, Y. A., Rankin, K. A., Stebbins, K. J., Lorrain, D. S.,
       Pekarek, K., S, A. S., Xiao, L., Teuscher, C., von Budingen, H. C., Wess, J., Lawrence, J.
       J., Green, A. J., Fancy, S. P., Zamvil, S. S., and Chan, J. R. (2016) Accelerated
       remyelination during inflammatory demyelination prevents axonal loss and improves
       functional recovery. eLife 5
6.     Patrikios, P., Stadelmann, C., Kutzelnigg, A., Rauschka, H., Schmidbauer, M., Laursen,
       H., Sorensen, P. S., Brück, W., Lucchinetti, C., and Lassmann, H. (2006) Remyelination
       is extensive in a subset of multiple sclerosis patients. Brain 129, 3165-3172
7.     Goldschmidt, T., Antel, J., König, F. B., Brück, W., and Kuhlmann, T. (2009)
       Remyelination capacity of the MS brain decreases with disease chronicity. Neurology 72,
       1914-1921
8.     Patani, R., Balaratnam, M., Vora, A., and Reynolds, R. (2007) Remyelination can be
       extensive in multiple sclerosis despite a long disease course. Neuropathology and applied
       neurobiology 33, 277-287
9.     Yeung, M. S. Y., Djelloul, M., Steiner, E., Bernard, S., Salehpour, M., Possnert, G.,
       Brundin, L., and Frisen, J. (2019) Dynamics of oligodendrocyte generation in multiple
       sclerosis. Nature 566, 538-542
10.    Jakel, S., Agirre, E., Mendanha Falcao, A., van Bruggen, D., Lee, K. W., Knuesel, I.,
       Malhotra, D., Ffrench-Constant, C., Williams, A., and Castelo-Branco, G. (2019) Altered
       human oligodendrocyte heterogeneity in multiple sclerosis. Nature 566, 543-547
11.    Galloway, D. A., Gowing, E., Setayeshgar, S., and Kothary, R. (2020) Inhibitory milieu
       at the multiple sclerosis lesion site and the challenges for remyelination. Glia 68, 859-877
12.    Mi, S., Hu, B., Hahm, K., Luo, Y., Kam Hui, E. S., Yuan, Q., Wong, W. M., Wang, L.,
       Su, H., Chu, T. H., Guo, J., Zhang, W., So, K. F., Pepinsky, B., Shao, Z., Graff, C.,

                                               14
Garber, E., Jung, V., Wu, E. X., and Wu, W. (2007) LINGO-1 antagonist promotes spinal
      cord remyelination and axonal integrity in MOG-induced experimental autoimmune
      encephalomyelitis. Nature medicine 13, 1228-1233
13.   Stoffels, J. M., de Jonge, J. C., Stancic, M., Nomden, A., van Strien, M. E., Ma, D.,
      Siskova, Z., Maier, O., Ffrench-Constant, C., Franklin, R. J., Hoekstra, D., Zhao, C., and
      Baron, W. (2013) Fibronectin aggregation in multiple sclerosis lesions impairs
      remyelination. Brain 136, 116-131
14.   Sloane, J. A., Batt, C., Ma, Y., Harris, Z. M., Trapp, B., and Vartanian, T. (2010)
      Hyaluronan blocks oligodendrocyte progenitor maturation and remyelination through
      TLR2. Proceedings of the National Academy of Sciences of the United States of America
      107, 11555-11560
15.   Kotter, M. R., Li, W. W., Zhao, C., and Franklin, R. J. (2006) Myelin impairs CNS
      remyelination by inhibiting oligodendrocyte precursor cell differentiation. J Neurosci 26,
      328-332
16.   Mi, S., Miller, R. H., Tang, W., Lee, X., Hu, B., Wu, W., Zhang, Y., Shields, C. B.,
      Zhang, Y., Miklasz, S., Shea, D., Mason, J., Franklin, R. J., Ji, B., Shao, Z., Chedotal, A.,

                                                                                                      Downloaded from http://www.jbc.org/ by guest on November 9, 2020
      Bernard, F., Roulois, A., Xu, J., Jung, V., and Pepinsky, B. (2009) Promotion of central
      nervous system remyelination by induced differentiation of oligodendrocyte precursor
      cells. Ann Neurol 65, 304-315
17.   Demetriou, M., Granovsky, M., Quaggin, S., and Dennis, J. W. (2001) Negative
      regulation of T-cell activation and autoimmunity by Mgat5 N-glycosylation. Nature 409,
      733-739
18.   Partridge, E. A., Le Roy, C., Di Guglielmo, G. M., Pawling, J., Cheung, P., Granovsky,
      M., Nabi, I. R., Wrana, J. L., and Dennis, J. W. (2004) Regulation of cytokine receptors
      by Golgi N-glycan processing and endocytosis. Science 306, 120-124
19.   Lau, K. S., Partridge, E. A., Grigorian, A., Silvescu, C. I., Reinhold, V. N., Demetriou,
      M., and Dennis, J. W. (2007) Complex N-glycan number and degree of branching
      cooperate to regulate cell proliferation and differentiation. Cell 129, 123-134
20.   Dennis, J. W., Nabi, I. R., and Demetriou, M. (2009) Metabolism, cell surface
      organization, and disease. Cell 139, 1229-1241
21.   Chen, I. J., Chen, H. L., and Demetriou, M. (2007) Lateral compartmentalization of T cell
      receptor versus CD45 by galectin-N-glycan binding and microfilaments coordinate basal
      and activation signaling. J Biol Chem 282, 35361-35372
22.   Zhou, R. W., Mkhikian, H., Grigorian, A., Hong, A., Chen, D., Arakelyan, A., and
      Demetriou, M. (2014) N-glycosylation bidirectionally extends the boundaries of
      thymocyte positive selection by decoupling Lck from Ca(2)(+) signaling. Nature
      immunology 15, 1038-1045
23.   Mkhikian, H., Mortales, C. L., Zhou, R. W., Khachikyan, K., Wu, G., Haslam, S. M.,
      Kavarian, P., Dell, A., and Demetriou, M. (2016) Golgi self-correction generates
      bioequivalent glycans to preserve cellular homeostasis. eLife 5
24.   Araujo, L., Khim, P., Mkhikian, H., Mortales, C. L., and Demetriou, M. (2017)
      Glycolysis and glutaminolysis cooperatively control T cell function by limiting
      metabolite supply to N-glycosylation. eLife 6
25.   Ohtsubo, K., Takamatsu, S., Minowa, M. T., Yoshida, A., Takeuchi, M., and Marth, J. D.
      (2005) Dietary and genetic control of glucose transporter 2 glycosylation promotes
      insulin secretion in suppressing diabetes. Cell 123, 1307-1321

                                               15
26.   Chen, I. J., Chen, H. L., and Demetriou, M. (2007) Lateral compartmentalization of T cell
      receptor versus CD45 by galectin-N-glycan binding and microfilaments coordinate basal
      and activation signaling. J Biol Chem 282, 35361-35372
27.   Mortales, C. L., Lee, S. U., and Demetriou, M. (2020) N-Glycan Branching Is Required
      for Development of Mature B Cells. J Immunol 205, 630-636
28.   Mortales, C. L., Lee, S. U., Manousadjian, A., Hayama, K. L., and Demetriou, M. (2020)
      N-Glycan Branching Decouples B Cell Innate and Adaptive Immunity to Control
      Inflammatory Demyelination. iScience 23, 101380
29.   Bahaie, N. S., Kang, B. N., Frenzel, E. M., Hosseinkhani, M. R., Ge, X. N., Greenberg,
      Y., Ha, S. G., Demetriou, M., Rao, S. P., and Sriramarao, P. (2011) N-Glycans
      differentially regulate eosinophil and neutrophil recruitment during allergic airway
      inflammation. J Biol Chem 286, 38231-38241
30.   Lee, S. U., Grigorian, A., Pawling, J., Chen, I. J., Gao, G., Mozaffar, T., McKerlie, C.,
      and Demetriou, M. (2007) N-glycan processing deficiency promotes spontaneous
      inflammatory demyelination and neurodegeneration. J Biol Chem 282, 33725-33734
31.   Mkhikian, H., Grigorian, A., Li, C. F., Chen, H. L., Newton, B., Zhou, R. W., Beeton, C.,

                                                                                                     Downloaded from http://www.jbc.org/ by guest on November 9, 2020
      Torossian, S., Tatarian, G. G., Lee, S. U., Lau, K., Walker, E., Siminovitch, K. A.,
      Chandy, K. G., Yu, Z., Dennis, J. W., and Demetriou, M. (2011) Genetics and the
      environment converge to dysregulate N-glycosylation in multiple sclerosis. Nat Commun
      2, 334
32.   Li, C. F., Zhou, R. W., Mkhikian, H., Newton, B. L., Yu, Z., and Demetriou, M. (2013)
      Hypomorphic MGAT5 polymorphisms promote multiple sclerosis cooperatively with
      MGAT1 and interleukin-2 and 7 receptor variants. Journal of neuroimmunology
33.   Brynedal, B., Wojcik, J., Esposito, F., Debailleul, V., Yaouanq, J., Martinelli-Boneschi,
      F., Edan, G., Comi, G., Hillert, J., and Abderrahim, H. (2010) MGAT5 alters the severity
      of multiple sclerosis. J Neuroimmunol 220, 120-124
34.   Ye, Z., and Marth, J. D. (2004) N-glycan branching requirement in neuronal and
      postnatal viability. Glycobiology 14, 547-558
35.   Grigorian, A., and Demetriou, M. (2011) Mgat5 deficiency in T cells and experimental
      autoimmune encephalomyelitis. ISRN Neurol 2011, 374314
36.   Backer-Koduah, P., Infante-Duarte, C., Ivaldi, F., Uccelli, A., Bellmann-Strobl, J.,
      Wernecke, K. D., Sy, M., Demetriou, M., Dorr, J., Paul, F., and Ulrich Brandt, A. (2020)
      Effect of vitamin D supplementation on N-glycan branching and cellular
      immunophenotypes in MS. Ann Clin Transl Neurol
37.   Grigorian, A., Lee, S. U., Tian, W., Chen, I. J., Gao, G., Mendelsohn, R., Dennis, J. W.,
      and Demetriou, M. (2007) Control of T Cell-mediated autoimmunity by metabolite flux
      to N-glycan biosynthesis. J Biol Chem 282, 20027-20035
38.   Grigorian, A., Araujo, L., Naidu, N. N., Place, D., Choudhury, B., and Demetriou, M.
      (2011) N-acetylglucosamine inhibits T-helper 1 (Th1) / T-helper 17 (Th17) responses and
      treats experimental autoimmune encephalomyelitis. The Journal of biological chemistry
39.   Pasquini, L. A., Millet, V., Hoyos, H. C., Giannoni, J. P., Croci, D. O., Marder, M., Liu,
      F. T., Rabinovich, G. A., and Pasquini, J. M. (2011) Galectin-3 drives oligodendrocyte
      differentiation to control myelin integrity and function. Cell death and differentiation 18,
      1746-1756
40.   Zhang, Y., Yu, X., Ichikawa, M., Lyons, J. J., Datta, S., Lamborn, I. T., Jing, H., Kim, E.
      S., Biancalana, M., Wolfe, L. A., DiMaggio, T., Matthews, H. F., Kranick, S. M., Stone,

                                              16
K. D., Holland, S. M., Reich, D. S., Hughes, J. D., Mehmet, H., McElwee, J., Freeman,
       A. F., Freeze, H. H., Su, H. C., and Milner, J. D. (2014) Autosomal recessive
       phosphoglucomutase 3 (PGM3) mutations link glycosylation defects to atopy, immune
       deficiency, autoimmunity, and neurocognitive impairment. The Journal of allergy and
       clinical immunology 133, 1400-1409, 1409 e1401-1405
41.    Hu, J. G., Fu, S. L., Wang, Y. X., Li, Y., Jiang, X. Y., Wang, X. F., Qiu, M. S., Lu, P. H.,
       and Xu, X. M. (2008) Platelet-derived growth factor-AA mediates oligodendrocyte
       lineage differentiation through activation of extracellular signal-regulated kinase
       signaling pathway. Neuroscience 151, 138-147
42.    Rivers, L. E., Young, K. M., Rizzi, M., Jamen, F., Psachoulia, K., Wade, A., Kessaris, N.,
       and Richardson, W. D. (2008) PDGFRA/NG2 glia generate myelinating
       oligodendrocytes and piriform projection neurons in adult mice. Nature neuroscience 11,
       1392-1401
43.    Michalski, J. P., Anderson, C., Beauvais, A., De Repentigny, Y., and Kothary, R. (2011)
       The proteolipid protein promoter drives expression outside of the oligodendrocyte lineage
       during embryonic and early postnatal development. PloS one 6, e19772

                                                                                                      Downloaded from http://www.jbc.org/ by guest on November 9, 2020
44.    Young, K. M., Psachoulia, K., Tripathi, R. B., Dunn, S. J., Cossell, L., Attwell, D.,
       Tohyama, K., and Richardson, W. D. (2013) Oligodendrocyte dynamics in the healthy
       adult CNS: evidence for myelin remodeling. Neuron 77, 873-885
45.    Sturrock, R. R. (1980) Myelination of the mouse corpus callosum. Neuropathol Appl
       Neurobiol 6, 415-420
46.    Hughes, E. G., Orthmann-Murphy, J. L., Langseth, A. J., and Bergles, D. E. (2018)
       Myelin remodeling through experience-dependent oligodendrogenesis in the adult
       somatosensory cortex. Nat Neurosci 21, 696-706
47.    Gonzalez, G. A., Hofer, M. P., Syed, Y. A., Amaral, A. I., Rundle, J., Rahman, S., Zhao,
       C., and Kotter, M. R. N. (2016) Tamoxifen accelerates the repair of demyelinated lesions
       in the central nervous system. Sci Rep 6, 31599
448.   Cantoni, C., Bollman, B., Licastro, D., Xie, M., Mikesell, R., Schmidt, R., Yuede, C. M.,
       Galimberti, D., Olivecrona, G., Klein, R. S., Cross, A. H., Otero, K., and Piccio, L.
       (2015) TREM2 regulates microglial cell activation in response to demyelination in vivo.
       Acta neuropathologica 129, 429-447
49.    Prineas, J. W., and McDonald, W. I. (1997) Demyelinating diseases, Arnold, London,
       United Kingdom
50.    Glasser, M. F., and Van Essen, D. C. (2011) Mapping human cortical areas in vivo based
       on myelin content as revealed by T1- and T2-weighted MRI. The Journal of neuroscience
       : the official journal of the Society for Neuroscience 31, 11597-11616
51.    Beer, A., Biberacher, V., Schmidt, P., Righart, R., Buck, D., Berthele, A., Kirschke, J.,
       Zimmer, C., Hemmer, B., and Muhlau, M. (2016) Tissue damage within normal
       appearing white matter in early multiple sclerosis: assessment by the ratio of T1- and T2-
       weighted MR image intensity. Journal of neurology 263, 1495-1502
52.    Nakamura, K., Chen, J. T., Ontaneda, D., Fox, R. J., and Trapp, B. D. (2017) T1-/T2-
       weighted ratio differs in demyelinated cortex in multiple sclerosis. Annals of neurology
       82, 635-639
53.    Righart, R., Biberacher, V., Jonkman, L. E., Klaver, R., Schmidt, P., Buck, D., Berthele,
       A., Kirschke, J. S., Zimmer, C., Hemmer, B., Geurts, J. J. G., and Muhlau, M. (2017)

                                               17
Cortical pathology in multiple sclerosis detected by the T1/T2-weighted ratio from
      routine magnetic resonance imaging. Annals of neurology 82, 519-529
54.   Miller, J. B., Bull, S., Miller, J., and McVeagh, P. (1994) The oligosaccharide
      composition of human milk: temporal and individual variations in monosaccharide
      components. J Pediatr Gastroenterol Nutr 19, 371-376
55.   Lawson, M. A. E., O'Neill, I. J., Kujawska, M., Gowrinadh Javvadi, S., Wijeyesekera, A.,
      Flegg, Z., Chalklen, L., and Hall, L. J. (2020) Breast milk-derived human milk
      oligosaccharides promote Bifidobacterium interactions within a single ecosystem. ISME J
      14, 635-648
56.   Deoni, S. C., Dean, D. C., 3rd, Piryatinsky, I., O'Muircheartaigh, J., Waskiewicz, N.,
      Lehman, K., Han, M., and Dirks, H. (2013) Breastfeeding and early white matter
      development: A cross-sectional study. Neuroimage 82, 77-86
57.   Isaacs, E. B., Fischl, B. R., Quinn, B. T., Chong, W. K., Gadian, D. G., and Lucas, A.
      (2010) Impact of breast milk on intelligence quotient, brain size, and white matter
      development. Pediatr Res 67, 357-362
58.   Granovsky, M., Fata, J., Pawling, J., Muller, W. J., Khokha, R., and Dennis, J. W. (2000)

                                                                                                     Downloaded from http://www.jbc.org/ by guest on November 9, 2020
      Suppression of tumor growth and metastasis in Mgat5-deficient mice. Nature medicine 6,
      306-312
59.   Demetriou, M., Nabi, I. R., Coppolino, M., Dedhar, S., and Dennis, J. W. (1995) Reduced
      contact-inhibition and substratum adhesion in epithelial cells expressing GlcNAc-
      transferase V. The Journal of cell biology 130, 383-392
60.   Saab, A. S., Tzvetavona, I. D., Trevisiol, A., Baltan, S., Dibaj, P., Kusch, K., Mobius, W.,
      Goetze, B., Jahn, H. M., Huang, W., Steffens, H., Schomburg, E. D., Perez-Samartin, A.,
      Perez-Cerda, F., Bakhtiari, D., Matute, C., Lowel, S., Griesinger, C., Hirrlinger, J.,
      Kirchhoff, F., and Nave, K. A. (2016) Oligodendroglial NMDA Receptors Regulate
      Glucose Import and Axonal Energy Metabolism. Neuron 91, 119-132
61.   Bar-Or, A., Fawaz, L., Fan, B., Darlington, P. J., Rieger, A., Ghorayeb, C., Calabresi, P.
      A., Waubant, E., Hauser, S. L., Zhang, J., and Smith, C. H. (2010) Abnormal B-cell
      cytokine responses a trigger of T-cell-mediated disease in MS? Annals of neurology 67,
      452-461
62.   Cross, A. H., Stark, J. L., Lauber, J., Ramsbottom, M. J., and Lyons, J. A. (2006)
      Rituximab reduces B cells and T cells in cerebrospinal fluid of multiple sclerosis patients.
      J Neuroimmunol 180, 63-70
63.   Levin, R. M., Krieger, N. N., and Winzler, R. J. (1961) Glucmsumine and
      acetylglucosamine tolerance in man. The Journal of laboratory and clinical medicine 58,
      927-932
64.   Gaulden, E. C., and Keating, W. C. (1964) The Effect of Intravenous N-Acetyl-D-
      Glucosamine on the Blood and Urine Sugar Concentrations of Normal Subjects.
      Metabolism: clinical and experimental 13, 466-472
65.   Salvatore, S., Heuschkel, R., Tomlin, S., Davies, S. E., Edwards, S., Walker-Smith, J. A.,
      French, I., and Murch, S. H. (2000) A pilot study of N-acetyl glucosamine, a nutritional
      substrate for glycosaminoglycan synthesis, in paediatric chronic inflammatory bowel
      disease. Aliment Pharmacol Ther 14, 1567-1579
66.   Lee, K. Y., Shibutani, M., Takagi, H., Arimura, T., Takigami, S., Uneyama, C., Kato, N.,
      and Hirose, M. (2004) Subchronic toxicity study of dietary N-acetylglucosamine in F344

                                              18
You can also read