You are on page 1of 12

Journal of Neuroimmunology 309 (2017) 88–99

Contents lists available at ScienceDirect

Journal of Neuroimmunology
journal homepage: www.elsevier.com/locate/jneuroim

B cells from patients with multiple sclerosis induce cell death via apoptosis MARK
in neurons in vitro
Robert P. Lisaka,b,⁎,1, Liljana Nedelkoskaa, Joyce A. Benjaminsa,b,1, Dana Schalkc,2,
Beverly Bealmeara, Hanane Touild,e,3, Rui Lid,e,3, Gillian Muirheadd, Amit Bar-Ord,e,⁎⁎,1,3
a
Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
b
Department of Microbiology, Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, MI, USA
c
Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
d
Department of Neurology, McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
e
Department of Neurology and Center for Neuroinflammation and Neurotherapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA

A R T I C L E I N F O A B S T R A C T

Keywords: B cells mediate multiple sclerosis (MS) pathogenesis by mechanisms unrelated to immunoglobulin (Ig). We
B cells reported that supernatants (Sup) from cultured B cells from blood of relapsing remitting MS (RRMS) patients, but
Cell death not normal controls (NC), were cytotoxic to rat oligodendrocytes (OL). We now show that RRMS blood B cells,
Cytokines not stimulated in vitro, secrete factor/s toxic to rat and human neurons. Cytotoxicity is independent of Ig and
Multiple sclerosis
multiple cytokines, not complement-mediated, and involves apoptosis. The factor/s have an apparent mw
Neurons
Oligodendrocytes
of > 300 kDa. B cells could contribute to damage within the central nervous system by secreting molecules toxic
to OL and neurons.

1. Introduction course of the disease (Absinta et al., 2011; Bo et al., 2007; Calabrese
et al., 2009; Kutzelnigg et al., 2005; Lucchinetti et al., 2011; Peterson
Studies in multiple sclerosis (MS) have continued to highlight the et al., 2001; Rocca et al., 2005). The subpial pattern of cortical
complexities of the pathogenesis of this disorder, particularly mechan- involvement appears to be a major component of gray matter injury
isms underlying relentless progressive disease. MS was often described highlighted in recent years as likely representing a major pathologic
as an inflammatory demyelinating disease of the central nervous system substrate of (non-relapsing) progressive MS, and correlating with both
(CNS) mainly affecting focal perivascular white matter regions, invol- motor and cognitive decline better than the focal perivascular deep
ving microglial activation, astrocyte proliferation and hyperplasia; white matter lesions (Fisher et al., 2008; Fisniku et al., 2008; Rudick
secondary axonal changes were thought to occur as a late phenomenon. and Trapp, 2009). This subpial pathology is not perivascular and
We have relearned that damage to axons occurs in the earliest stages involves injury of both oligodendrocytes (OL) and neurons, evidenced
even of relapsing remitting MS (Ferguson et al., 1997; Trapp et al., in part by apoptosis of these cells in the superficial layers of cortex (Bo
1998). Gray matter involvement including the cerebral cortex was also et al., 2006; Kutzelnigg et al., 2005; Peterson et al., 2001; Serafini et al.,
viewed as involved late in disease, yet we now know that cortical gray 2004). Of growing interest has been the potential contribution to such
matter damage occurs not only in chronic disease but also early in the injury by immune cell collections in the meninges, some of which are

Abbreviations: CNS, central nervous system; DMEM, Dulbecco's modified essential medium; DMT, disease modifying therapy; EBV, Epstein-Barr virus; FGF, fibroblast growth factor; GM-
CSF, granulocyte macrophage-colony stimulating factor; IFN, interferon; Ig, immunoglobulin; IL, interleukin; kDa, kilodalton; LT, lymphotoxin; MIP, macrophage inflammatory protein;
MMP, matrix metalloproteinase; MS, multiple sclerosis; mw, molecular weight; NC, normal control; OL, oligodendrocyte; OPC, oligodendrocyte precursor cell; PBS, phosphate buffered
saline; PPMS, primary progressive multiple sclerosis; RRMS, relapsing remitting multiple sclerosis; SPMS, secondary progressive multiple sclerosis; Sup, supernatant; TGF, transforming
growth factor; TNF, tumor necrosis factor; TUNEL, terminal deoxynucleotidyl dUTP nick end labeling

Correspondence to: R.P. Lisak, Department of Neurology, Wayne State University School of Medicine, 8D, University Health Center 4201 St Antoine, Detroit, MI 48201, USA.
⁎⁎
Correspondence to: A. Bar-Or, Perelman Center for Advanced Medicine (PCAM) South Pavilion, 7th Floor; Rm 7633400 Civic Center Blvd, Philadelphia, PA 19104, USA
E-mail addresses: rlisak@med.wayne.edu (R.P. Lisak), lnedelk@med.wayne.edu (L. Nedelkoska), jbenjami@med.wayne.edu (J.A. Benjamins),
DLS2DA@hscmail.mcc.virginia.edu (D. Schalk), bbealmea@med.wayne.edu (B. Bealmear), hanane.touil@mail.med.upenn.edu (H. Touil), rui.li@mail.med.upenn.edu (R. Li),
g.muirhead@gmail.com (G. Muirhead), amitbar@upenn.edu (A. Bar-Or).
1
Drs. Lisak, Benjamins and Bar-Or contributed equally to this study.
2
Current address: Department of Medicine, Division of Hematology/Oncology, University of Virginia, Charlottesville, VA, USA.
3
Current address: Center for Neuroinflammation and Neurotherapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.

http://dx.doi.org/10.1016/j.jneuroim.2017.05.004
Received 7 February 2017; Received in revised form 9 May 2017; Accepted 10 May 2017
0165-5728/ © 2017 Elsevier B.V. All rights reserved.
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

described as ‘B cell rich’ (Choi et al., 2012; Kutzelnigg et al., 2005; Table 1
Magliozzi et al., 2007; Serafini et al., 2004). Cytokines and factors assayed in B cell supernatants from NC and RRMS.
We have been interested in the effector role of B cells in causing
Cytokine (14-PLEX) Cytokine (25-PLEX) Individual ELISAs
damage to OL/myelin and neurons/axons independent of involvement
of immunoglobulin (Ig) and/or complement. We previously reported GM-CSF Eotaxin IL-10 CD40
that secretory products from cultures of B cells (including unstimulated IFNα FGF (basic) ILp40p70 CD40 ligand
IL-1β GM-CSF IL-13 Fas
B cells) from blood of untreated patients with RRMS are cytotoxic for
IL-4 IFN-α IL-15 Fas ligand
OL in vitro in mixed rat glial cultures, whereas Sup from B cells from IL-6 IFN-γ IL-17 Granzyme B
normal controls (NC) are not cytotoxic (Lisak et al., 2012). We IL-10 IL-1β IP-10 IL-16
hypothesize that B cells entering the meninges and cerebral spinal IL-12p40 IL-1RA MCP-1 IL-23
fluid (CSF) from the peripheral immune system could secrete factors IL-12p60 IL-2 MIG (CXCL9) LT-α
MIP-1α (CCL3) IL-2R MIP-1α (CCL3) LT-β
that lead to damage characteristic of MS (Fischer et al., 2013; Haider
MIP-1β IL-4 MIP-1β MMP-9
et al., 2016; Lassmann, 2012; Lassmann, 2014) in the underlying TGF-α IL-5 RANTES (CCL5) TGF-β
cortical gray matter. TNF-α IL-6 TNF-α
In the current study we tested whether Sup from blood-derived B TNF-β IL-7 VEGF
VEGF IL-8
cells of patients with RRMS seen at the Montreal Neurological Institute/
McGill University also compromise neuronal viability in comparison to Abbreviations: FGF, fibroblast growth; GM-CSF, granulocyte-colony stimulating factor;
Sup of B cells from NC and examined the mechanism underlying such IFN, interferon; IL, interleukin; IL-1RA, interleukin 1 receptor; IL-2R, interleukin-2
toxicity if observed. We hypothesized that neuronal cell death would be receptor; LT, lymphotoxin; MCP, monocyte chemoattractant protein; MIG, monocyte-
induced by the MS patient-derived B cell Sup and this death would be induced by gamma interferon; MIP, macrophage inflammatory protein; MMP-9, matrix
due to apoptosis. We examined the effects of B cell Sup from RRMS metalloproteinase; TGFβ, transforming growth factor-alpha; TNF, tumor necrosis factor;
RANTES, regulated on activation, normal T cell expressed and secreted; VEGF, vascular
patients and NC on both rodent and human neurons in vitro. We
endothelial growth factor.
measured levels of IgM and IgG and an expanded number of cytokines
and related protein secretory products of inflammatory cells in the B
2.3. Human neuronal cultures
cell culture Sup.

Coverslips previously coated with poly-D-lysine (1 mg/10 ml) were


2. Methods
placed in a 24 well plate and coated overnight with laminin (10 μg/ml);
the excess solution was removed just prior to plating the cells. One vial
2.1. Materials
of cryopreserved primary human neurons (Sciencell) containing
1 × 106 cells was thawed and suspended in 14 ml of complete neuronal
Human neurons (#1520, medium (#1521), growth supplement
medium, per the supplier's instructions (Sciencell), then plated in 700 μl
(#1562), and penicillin streptomycin solution (#0503) were purchased
aliquots. After 4 days in culture, fresh medium was added, and the
from Sciencell (San Diego CA). Poly-D-lysine (P7405) and laminin
neurons were used the next day to test the B cell Sup.
(L2020) were from Sigma (St. Louis MO).
ELISA kits were obtained as follows: 14-PLEX magnetic bead custom
array (HCYTOMAG60K-14) from EMD Millipore; 25-PLEX non-mag- 2.4. Rat mixed glial cell cultures
netic cytokine array (LHC00009) from Life Technologies/Invitrogen;
transforming growth factor-β (TGF-β), LEGEND MAX (436707) and Some of the B cell supernatants from RRMS patients and NC used in
human interleukin (IL)-35 heterodimer, LEGEND MAX (439507) from this study were tested previously for capacity to kill OL in rat mixed
BioLegend; lymphotoxin-α (LT-α) (MBS268485) and lymphotoxin-β glial cell cultures (Lisak et al., 2012). We tested additional newly
(LT-β) (MBS261430) from MyBiosource; IL-16 and IL-23 2-PLEX produced supernatants from RRMS patients (n = 6) and NC (n = 6) to
(HCYP2MAG-62K-02) from EMD Millipore; and matrix metalloprotei- ascertain whether these supernatants were also toxic to OL. Cultures
nase-9 (MMP-9) (DMP900) from R & D. ELISA assays for Fas (apo-1; enriched in differentiated OL were prepared from neonatal rat brain,
ab100513); Fas ligand (ab100515); CD40 (ab119516); CD40 ligand using a modification of the shakeoff method (McCarthy and de Vellis,
(ab196268) and granzyme B (ab46142) were from Abcam. The 1980); cultures from the 3rd–5th shakeoffs were used, resulting in
complete list of cytokines and biological modifier proteins examined cultures containing on average 35–40% OL, 35–40% astroglia and 15%
are listed in Table 1. microglia. The methods for preparation of cultures have been pre-
viously described in detail (Lisak et al., 2006; Lisak et al., 2012).
2.2. Rat neuronal cultures
2.5. B cell cultures
Neurons were cultured from neonatal rat brain based on the method
of Eide and McMurray (Eide and McMurray, 2005), modified as Blood was obtained with informed consent from patients with RRMS
described (Lisak et al., 2011). For each explant, three forebrains were and age matched controls at the Montreal Neurological Institute/McGill
used; each half forebrain was minced in 2 ml of HEPES buffered saline University. B cells were obtained by positive selection for CD19 from
containing 2 mg/ml of papain, then combined with the other tissue for peripheral blood of consenting patients with RRMS (Polman et al.,
the subsequent steps. The final cell pellet was triturated 5–7 times in 2011) and from NC, as previously described (Bar-Or et al., 2010; Bar-Or
3 ml of plating medium; 50 μl were plated on each coverslip, with 10 et al., 2001; Duddy et al., 2007; Duddy et al., 2004) and as approved by
coverslips per 60 mm dish. After overnight attachment at 370C, 2 ml of the Ethics Review Board of the Montreal Neurological Institute and
plating medium with 100 ng/ml nerve growth factor was added to each McGill University. B cells were isolated by density centrifugation using
dish. Cytosine arabinoside, final concentration 1 × 10− 5 M, was added Ficoll (GE Healthcare). CD19 beads (Miltenyi Biotech) were used to
the next day; after 5 days, the medium was changed to B-27 feeding positively select B cells according to the manufacturer's protocol. Purity
medium containing NGF and cytosine arabinoside. Neuronal cultures of the cells was assessed using flow cytometry with an enrichment
were used to test toxicity of B cell Sup between 5 and 7 days after of > 98%. B cells were then cultured without any stimulation for 3 days
explant. Cultures were 85–90% neurons, with the remaining cells (Bar-Or et al., 2010; Bar-Or et al., 2001; Duddy et al., 2004; Lisak et al.,
astrocytes and microglia, and no detectable OL based on phenotypic 2012); Sup were harvested and frozen at -80° C until testing for
markers (Lisak et al., 2011; Lisak et al., 2012). cytotoxicity studies performed at Wayne State University School of

89
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

Medicine. 31146). Detection was carried out with goat anti-human IgG horse
RRMS patients consisted of 9 women and 4 men with a mean age of radish peroxidase conjugate (Invitrogen H10007) or goat anti-human
39.8 years of age (range 18–60 years), and the healthy NC were 8 IgM horse radish peroxidase conjugate (Invitrogen H15007) and TMB
women and 5 men with mean age of 35.5 years of age (range substrate (Thermo 34021).
22–63 years). All MS patients had RRMS (at least one relapse in the
prior 12 months) with the exception of one patient who presented as
clinically isolated syndrome (and was subsequently confirmed to have 2.9. Cytokines and related proteins
RRMS). At the time of blood studies, 10 patients were stable (no new
symptoms or signs to suggest relapse in the prior 3 months). None had The human specific cytokines and related protein biological modi-
received corticosteroids for at least 3 months, none had received fiers examined are listed in Table 1.
disease modifying therapy (DMT) for at least 24 months and 9 had
never received any DMT. Of the 13 NC and 13 MS Sup analyzed for
2.10. Estimation of apparent molecular weight of toxic factors
their effects on neurons in this study, 7 NC and 7 MS were used in our
previous study that assessed OL toxicity (Lisak et al., 2012).
B cell medium and B cell Sup from NC and RRMS were analyzed
using centrifugal filtration units with molecular weight cutoff mem-
2.6. Cytotoxicity assays
branes. The membranes were pre-washed, 250 μl samples added and
units centrifuged according to manufacturers' instructions. Units with
Sup from B cell cultures were diluted with either rat or human
membrane cutoffs of 30 kilodalton (kDa) and 100 kDa were purchased
neuronal medium (1:4). The diluted B cell Sup were incubated with
from EMD Millipore (Temecula CA), and 300 kDa units (NanoSep
neuronal cultures for 72 h. As control medium, we used the B cell
300 K) from Pall Corporation (Ann Arbor, MI).
culture medium (not conditioned with B cells), diluted 1:4 with the
respective neuronal medium. Neuronal death was assessed by uptake of
trypan blue as we employed for OL, oligodendrocyte precursors (OPC) 2.11. Statistical analysis
and neurons (Benjamins et al., 2014; Lisak et al., 2012; Lisak et al.,
2011). For all experiments with rat neuronal cultures, the cultures were B cell Sup were tested for cell death or apoptosis on duplicate
analyzed in a blinded fashion. Experiments were performed with mixed coverslips in each experiment; the means of the values for NC and
glial cell cultures to assess whether newly obtained B cell supernatants RRMS were analyzed using the Students' t-test or using one way ANOVA
(6 NC and 6 MS) were also toxic for OL; Sup were diluted 1:4 with glial with Tukey post test, as indicated in the figure legends.
cell culture medium as previously reported (Lisak et al., 2012). All
bovine sourced serum used in culture medium for B cells, neurons and
glia were heat inactivated as done in past experiments. 3. Results

2.7. Detection of apoptosis 3.1. Impact of B cell secreted products on neuronal and glial cell survival

Attached cells on coverslips were fixed and stained to assess Our prior publication reported OL cytotoxic effects of Sup from 7
apoptosis utilizing the Apoptag In Situ Apoptosis Kit (EM Millipore, RRMS, with little to no toxicity by Sup from 3 out of 4 NC (Lisak et al.,
Billerica MA) to visualize terminal deoxynucleotidyl dUTP nick end 2012). We have now expanded our findings to an additional 15 RRMS
labeling (TUNEL), as described previously (Skoff et al., 1998). We and 16 NC Sup and find similar results. Data for OL toxicity of the 13
quantitated the percentage of TUNEL positive rat neurons at 24, 48 and RRMS and 13 NC Sup that we subsequently tested for neuronal toxicity,
72 h of incubation with supernatants from B cell cultures from 3 NC and are shown in Fig. 1. Substantiating our previous observations, B cell Sup
4 RRMS. Additional supernatants from NC and RRMS were tested on from all RRMS patients (but not from NC) induced on average 58% OL
both rat neurons and human neurons at 72 h. Surface appearance of the death compared to 4% for NC B cells Sup. None of the B cell Sup
apoptotic marker annexin was used to assess apoptosis in OL. Live cells appeared to have toxic effects on astrocytes or microglia within the
on coverslips were treated for 15 min at room temperature with mixed glial cultures.
Alexa488-labeled annexin V (Thermo-Fisher, Waltham MA), diluted We next observed that Sup from the unstimulated B cell cultures of
1:20 with annexin-binding buffer. Coverslips were washed once gently all 13 patients with RRMS (at a 1:4 dilution with neuronal culture
in Dulbecco's minimal essential medium (DMEM), fixed, then mounted medium) induced cytotoxicity of rat neurons, while similarly diluted
for subsequent viewing by fluorescence microscopy. For immunostain- Sup from 13 NC induced little or no neuronal death above background
ing of activated caspase-3, cells were fixed, permeabilized with 0.05% (Fig. 2A, B). Background neuronal cell death induced by the control
Triton for 20 min, blocked with 10% bovine serum albumin for 30 min, medium alone at 1:4 ranged between 11 and 14% neuronal death and
then incubated overnight at 4° C with a 1:200 dilution of antibody to was subtracted for each experiment from total trypan blue positive
activated caspase 3 (Thermo-Fisher, Waltham MA). Antibody binding cells. The mean cell death above background for RRMS patient B cell
was visualized with FITC-labeled donkey anti-rabbit IgG. Sup was 47.5% ± 2.7% S.E.M compared to 9.6% ± 2.4% S.E.M. for
cell death induced by Sup from NC B cells (Fig. 1C).
2.8. Immunoglobulins To examine whether the cytotoxic potential of MS patient-derived B
cell Sup was limited to rodent neurons, we assessed the effects of B cell
Human specific IgG and IgM enzyme-linked immunosorbent (ELISA) Sup derived from RRMS patients and NC on human-derived neurons. B
assays were performed in the immunoanalysis core of the Bone Marrow cell Sup (at 1:4 dilution with human neuronal medium) from 4 patients
Transplant/Immunotherapy Program of the Karmanos Cancer Institute with RRMS were toxic to human neurons while similarly diluted Sup
and Wayne State University Department of Oncology, using a method from 3 NC induced little or no neuronal death (Fig. 3A). In each
developed by the core. ELISA plates were coated with goat anti-human experiment, background neuronal cell death induced by control med-
IgG (Invitrogen H10000) or goat anti-human IgM (Invitrogen H15000). ium alone at 1:4 was assayed as was done for rat neurons and
The blocking solution was 3% bovine serum albumin + 5% fetal bovine subtracted as above from total trypan blue positive cells for RRMS
serum in 1× phosphate buffered saline (PBS), and the washing solution and NC. The mean cell death above background for RRMS patient B cell
was 1× PBS + 0.05% Tween 20. The standards were human IgG, Sup was 55.9% ± 1.9% S.E.M. compared to 8.8% ± 0.8% S.E.M cell
whole molecule (Pierce 31154) or human IgM, whole molecule (Pierce death induced by Sup from NC (Fig. 3B).

90
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

Fig. 1. Secreted products of B cells from RRMS patients are cytotoxic to rat OL compared to those from normal individuals. B cell Sup in addition to those previously tested (Lisak et al.,
2012) were assayed for OL toxicity prior to assays on neuronal cultures, mixed glial cultures were treated for 3 days with Sup from B cells (not stimulated in vitro) from 9 NC and 12
RRMS, diluted 1:4 with rat OL medium; and OL death was measured by trypan blue uptake; OL death in 1:4 B cell medium alone averaged 13%; this value was subtracted from NC and
RRMS values. A. OL death in cultures treated with 1:4 B cell Sup from 9 NC; NC 1, 2 and 4 were tested previously; B. OL death in cultures treated with 1:4 B cell supernatants from 12
RRMS patients; MS 3, 4, 6 and 7 were tested previously (Lisak et al., 2012); C. Comparison of mean % OL death induced by B cell supernatants from NC versus RRMS: NC,
4.1 ± 2.6 S.E.M; RRMS, 58.1 ± 3.6 S.E.M.; p < 0.0001, Student's t-test.

3.2. The impact of MS B cell supernatants on rat and human neuronal and 3.3. Levels of immunoglobulins, cytokines and related proteins in B cell
OL survival involves apoptotic cell death culture supernatants

Sup from B cell cultures of 3 patients with RRMS and 3 NC were We found very low levels of IgG and IgM in these short-term
tested to determine if apoptosis contributed to death of rat neurons. cultured B cell Sup, and no significant difference in the mean levels
When stained with antibodies to NF-H, the neurons exposed to the MS B between the NC and RRMS samples. Average levels ± S.E.M. for IgG
cell Sup appeared damaged and showed process retraction compared to were 0.060 ± 0.016 mg/dl in NC and 0.052 ± 0.016 mg/dl for MS,
neurons exposed to control Sup (Fig. 4A). At different time points in p < 0.70. Average levels ± S.E.M. for IgM were 0.017 ± 0.006 mg/dl
vitro as assessed by percent of neurofilament-H (NF-H)+ neurons that in NC and 0.014 ± 0.003 mg/dl for MS, p < 0.63. There was no
stained with TUNEL, B cell Sup from RRMS patients induced significant correlation between cytotoxicity and the IgG or IgM levels in either
apoptosis at 72 h with little if any apoptosis induced by Sup from NC group (Fig. 5A, B).
(Fig. 4B). Less TUNEL staining was observed at 24 h and 48 h. Levels of most cytokines and chemokines were low or undetectable
Additional experiments were carried out with 3 more NC and 3 more in the Sup of these unstimulated B cells, both from NC and RRMS.
RRMS Sup at 72 h (Fig. 4C). The conclusion that TUNEL staining Analysis of the values obtained using a 14-PLEX magnetic array (Tables
represents apoptosis rather than DNA fragmentation due to DNA repair 1 and 2) showed no statistical differences between the concentrations of
is consistent with the trypan blue results and our observation that 64% any of the cytokines/chemokines/biologic modifiers in the Sup ob-
of OL treated for 3 days with Sup from RRMS are positive for annexin V tained from B cells of NC and RRMS, with the exception of LT-β which
staining compared to only 15% for NC Sup; similar results were found appeared reduced in RRMS B cell Sup. There was a trend for an
for immunostaining for activated caspase 3 (data not shown). increased level of the chemokine MIP-1α in Sup of RRMS patients, as
Cell death induced in the human neurons by Sup of B cells from well a trend for decreased IL-6 compared to the B cell Sup of NC. There
RRMS patients also involved apoptosis. The human neurons exposed to was no correlation between the concentrations of any of these proteins
the MS B cell Sup appeared damaged, exhibiting greater process and neuronal cell death mediated by these respective B cell Sup.
retraction (Fig. 4D) and significantly greater apoptosis by TUNEL Examination of values obtained using a 25-PLEX non-magnetic im-
staining (Fig. 4E–F) as compared to human neurons treated with B cell munoarray analysis (Tables 1 and 2) showed that only 12 of the
Sup from NC B cells. cytokines assayed were detectable in Sup from either NC, RRMS or both
for some of the samples, while 15 of the cytokines were absent or below
detectable levels. There was no significant difference in values for any

Fig. 2. Secreted products of B cells from RRMS patients are cytotoxic to rat neurons compared to those from normal individuals. Neuronal cultures were treated for 3 days with Sup from B
cells (not stimulated in vitro) from NC or RRMS, diluted 1:4 with rat neuronal medium; and neuronal death was measured by trypan blue uptake; neuronal death in 1:4 B cell medium
alone averaged 13%; this value was subtracted from NC and RRMS values. A. Neuronal death in cultures treated with 1:4 B cell Sup from 13 NC. B. Neuronal death in cultures treated with
1:4 B cell Sup from 13 RRMS patients. C. Comparison of mean neuronal death induced by B cell Sup from NC versus RRMS. NC, 9.6% ± 2.4% S.E.M.; RRMS, 47.5% ± 2.7% S.E.M.;
p < 0.001, Student's t-test.

91
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

Fig. 3. Secreted products of B cells from RRMS patients are cytotoxic to human neurons compared to Sup of B cells obtained from normal individuals. A. Neuronal cultures were treated
for 3 days with Sup from B cells (not stimulated in vitro) from 3 NC samples and 4 RRMS samples, each diluted 1:4 with human neuronal medium; neuronal death was measured by trypan
blue uptake. Neuronal death in 1:4 B cell medium alone averaged 12%; this value was subtracted from RRMS values. B. Comparison of mean neuronal death induced by B cell Sup from
NC versus RRMS. NC, 8.8% ± 0.8% S.E.M.; RRMS, 55.9% ± 1.9% S.E.M.; p < 0.0001, Student's t-test.

of the detected cytokines between NC and RRMS B cell Sup. granzyme B (Table 1); these proteins were not detectable in most of the
Six cytokines of potential interest that were not represented on the samples, and again, there were no differences between NC and MS
arrays were analyzed by individual ELISAs (Tables 1 and 2), with low samples (data not shown).
levels of MMP-9, TGF-β and IL-16 detected in some samples. There was When levels of IL-16, LT-α, LT-β, TNF-α (14-PLEX), MIP-1α (14-
no difference in the levels of MMP-9 or TGF-β in the B cell Sup of NC PLEX), MIP-1β (14-PLEX) and GM-CSF are compared with neuronal cell
and RRMS patients. There was also no difference between NC and death induced by B cell.
RRMS B cell Sup levels of IL-16 when results were analyzed with the Sup from RRMS and/or NC (Fig. 5C–I), there were no correlations
one NC outlier included (p = 0.33); with the outlier removed, mean between any given cytokine concentration and death of neurons. We
levels of IL-16 were slightly increased in RRMS compared to NC performed similar analyses for MMP-9, IL-6 (values from both cytokine
(p < 0.04). Five additional proteins of interest, related to B cell assays), interferon (IFN)-α, IFN-γ, ΤΝF-α, and fibroblast growth factor
biology, were also assayed: CD40, CD40 ligand, Fas, Fas ligand and (FGF), and there was no correlation between protein levels in the Sup

Fig. 4. Apoptosis is prominent in neurons treated with secreted products of B cells from RRMS patients compared to normal individuals. A. Representative fields of rat neuronal cultures
treated for 3 days with B cell Sup from NC or RRMS, immunostained for NF-H (green) or TUNEL (red). B. Time course shows % of apoptotic rat neurons at 1, 2 and 3 days for 3 NC and 4
MS Sup. *p < 0.01, NC 3d vs. RRMS 3d. C. Apoptosis in rat neurons was measured by immunostaining for TUNEL and NF-H after 3 days of treatment with 1:4 B cell Sup from an
additional 3 NC and 3 RRMS. *p < 0.001, NC 3d vs. RRMS 3d. D. Representative fields of human neuronal cultures treated for 3 days with B cell Sup from NC or RRMS, immunostained
for NF-H (green) or TUNEL (red). E. Apoptosis in human neurons was measured by immunostaining for TUNEL and NF-H after 3 days of treatment with B cell Sup from 4 NC and 4 RRMS.
F. Comparison of mean apoptotic human neuronal death induced by B cell Sup from NC versus RRMS. NC, 5.5% ± 1.6%; RRMS, 53.3% ± 1.0% S.E.M.; p < 0.0001, Student's t-test.
(For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

92
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

Fig. 5. Levels of human IgG, IgM and cytokines are similar in supernatants from RRMS and NC, and do not correlate with death of cultured rat neurons. A. Comparison of % neuronal
death with the levels of IgG in the B cell Sup showed no significant correlation for either NC or RRMS samples, as analyzed by Pearson's correlation coefficient. NC vs. IgG levels, r = 0.39,
p < 0.0.30; RRMS vs, IgG levels, r = 0.44, p < 0.20. B. Comparison of % neuronal death with the levels of IgM in the B cell Sup showed no significant correlation for either NC or RRMS
samples. NC vs. IgM levels, r = 0.41, p < 0.27; RRMS vs, IgM levels, r = 0.46, p < 0.50. C - I. Comparison of % neuronal death with the concentration of levels of GM-CSF, IL-16, LT-α,
LT-β, MIP-1α, MIP-1β, or TNF-α in the B cell Sup showed no significant correlation for either NC or RRMS samples, as analyzed by Pearson's correlation coefficient.

and neuronal cell death (data not shown). In addition, combining levels the NC Sup showed no toxicity. In subsequent experiments B cell Sup
of ‘proinflammatory proteins’ or of ‘downregulatory anti-inflammatory’ from 6 NC and 6 RRMS were fractionated with a 300 kDa cutoff
proteins did not suggest any combination that correlated with neuronal membrane filter. The 300 kDa retentates from RRMS Sup induced OL
cell death in our system (data not shown). death whereas retentates from control medium or NC Sup did not
(Fig. 6A). None of the filtrates showed toxicity to OL. After centrifuga-
tion of the RRMS > 300 kDa fraction at 2,000 × g for 30 min to remove
3.4. Toxic factor/s in RRMS B cell supernatants are enriched in high
cell debris, the toxic factor/s remained in the supernatant. In subse-
molecular weight fractions
quent experiments, > 300 kDa retentates and filtrates from NC and
RRMS Sup were tested for toxicity on neuronal cultures. As with OL, the
Our experiments determining levels of cytokines and related
toxic factors were concentrated in the > 300 kDa fractions from RRMS
proteins as well as Ig levels did not identify a likely toxin or toxins.
Sup (Fig. 6B); the filtrates from RRMS Sup also exhibited a low but
Therefore as the next step in identifying the molecule/s responsible for
statistically significant toxicity compared to filtrates from the NC Sup.
toxicity, control medium and B cell Sup from NC and RRMS were
analyzed using centrifugal filtration units with molecular weight cutoff
membranes. We used both neurons and OL as targets in these assays. 4. Discussion
After filtration, the filtrate and retentate, each in a final volume of
250 μl of B cell medium, were diluted 1:4 with either neuronal or OL The role of B cells, their progeny (plasmablasts and plasma cells)
medium and tested for toxicity on neuron or OL cultures, respectively. and secreted Ig have been the subject of study in MS for many years
Cell death was analyzed at the end of 3 days using trypan blue uptake. (Cross and Wu, 2010; Kabat et al., 1942; Link and Huang, 2006; Miller
Initial experiments with OL using 30 kDa and 100 kDa cutoff mem- et al., 1983). Studies of serum, cerebrospinal fluid (CSF) and eluates of
branes showed that the toxic activity was retained in the higher Ig from CNS of MS patients attempting to identify one or more
molecular weight fraction of RRMS Sup, with no activity in the RRMS antibodies to specific target antigens (either CNS constituents or
filtrate. The filtrates and retentates from the control medium alone, or specific microbial antigens using increasingly sophisticated techni-

93
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

Table 2
Cytokines and other factors detected in B cell supernatants from NC and RRMS.

Cytokine Controls Mean ± S.E.M. Detected/assayed RRMS Mean ± S.E.M. Detected/assayed p Value
pg/ml pg/ml (t-test)

FGF, basicb 0–1.86 0.50 ± 0.27 5/6 0–0.70 0.16 ± 0.12 2/6 ns p = 0.20
GM-CSFa 0–8.9 2.21 ± 1.02 6/9 0–3.5 0.79 ± 0.39 5/10 ns p = 0.25
IFN-αb 0–10.6 5.31 ± 1.94 4/6 0–10.6 3.5 ± 1.77 3/6 ns p = 0.52
IFN-γb 3.7–9.0 0.89 ± 0.28 4/6 0–0.90 0.89 ± 0.28 3/6 ns p = 1.0
IL-4a 0–32.2 1.14 ± 0.99 2/9 0–9.1 1.48 ± 0.99 4/10 ns p = 0.81
IL-6a 0–151 27.8 ± 16.8 5/9 0–5.36 1.81 ± 0.68 5/10 ns p = 0.11
IL-6b 0–2.34 1.19 ± 0.37 5/6 0.68–3.4 1.48 ± 0.45 6/6 ns p = 0.62
IL-12p60a 0–2.76 0.71 ± 0.39 3/9 0–3.70 1.13 ± 0.48 5/10 ns p = 0.52
IL-16c 89–361 153.4 ± 29 9/9 102–272 189 ± 20 10/10 ns p = 0.33
(p = 0.045)⁎
LT-αc 0–39 19.4 ± 6.0 7/9 0–53 20.3 ± 5.4 6/9 ns p < 0.9
LT-βc 0–29 12.4 ± 2.7 8/9 0–13.3 4.8 ± 1.3 9/10 p = 0.018
MCP-1b 3.0–101 19.3 ± 16.3 2/6 0–80.7 24.1 ± 12.3 5/6 ns p = 0.82
MIP1αa 0–108 16.3 ± 12.2 2/9 0–332 83.7 ± 31.7 8/10 ns p = 0.074
MIP1β a 0–548 122 ± 63 6/9 0–424 162 ± 43 9/10 ns p = 0.69
MIP1βb 8–87.1 29.1 ± 11.9 6/6 8–62.6 29.3 ± 8.4 6/6 ns p = 0.98
MMP-9c 0–3895 830 ± 634 3/6 0–3570 800 ± 639 5/6 ns p = 0.97
TGFβc 0–1031 245 ± 173 2/6 0–1250 231 ± 204 3/6 ns p = 0.96
TNFαa 0–100 21.1 ± 10.9 8/9 2.8–26.8 11.1 ± 2.4 10/10 ns p = 0.36

a
B cell Sup from 6 to 9 NC and 6–10 RRMS were analyzed on a Luminex 14-plex magnetic immunoarray.
b
B cell Sup from 6 NC and 6 RRMS were analyzed on a 27-plex non-magnetic immunoarray.
c
B cell Sup from 6 to 9 NC and 6–9 RRMS were analyzed by individual ELISA kits.

p Value for IL-16 with one NC outlier value removed.

Fig. 6. Cytotoxicity of secretory factors from RRMS B cells is retained in a fraction with > 300 kDa molecular weight. (A) B cell Sup from 3 NC and 3 RRMS were fractionated with a
300 kDa cutoff membrane filter. The > 300 kDa retentates from RRMS Sup induced OL death whereas NC did not, compared to the medium control. Values represent averages of
duplicates. Comparison of mean % OL death induced by > 300 kDa retentates from NC versus RRMS: NC, 7.7 ± 0.7 S.E.M; RRMS, 44.0 ± 4.6 S.E.M.; **p < 0.0015, Student's t-test. No
toxicity above baseline was found in the filtrates or in the pellets (2,000 × g, 30 min) from the retentates for either NC or RRMS samples. The experiment was repeated on the same
samples with similar results. (B) B cell Sup from 5 NC and 5 RRMS were fractionated with a 300 kDa cutoff membrane filter. The > 300 kDa retentates from RRMS Sup induced neuronal
death whereas NC did not, compared to the medium control. Values represent averages of duplicates. Comparison of mean % neuronal death induced by the > 300 kDa retentates from
NC versus RRMS was NC 23.8 ± 1.7 S.E.M., RRMS 67.0 ± 2.5,** p < 0.0001, Student's t-test. Neuronal death with the filtrates was NC 20.4 ± 1.8 S.E.M; RRMS, 31.7 ± 1.6 S.E.M.;
*p < 0.002.

ques), have largely failed to identify disease-implicated target antigens Hauser, 2015). Some therapeutic benefit of anti-CD20 treatment has
(Blauth et al., 2015b; Brandle et al., 2016; Genain et al., 1999; Gilden, also been reported in subsets of patients with primary progressive MS
2005; Lisak et al., 1968). (PPMS) (Hawker et al., 2009; Montalban et al., 2017).
Recent clinical trials with agents directed against CD20 (expressed B cells in MS migrate from the periphery into the meninges, CSF and
by B cells) have demonstrated substantial benefit in patients with RRMS the CNS parenchyma of patients (Levinson et al., 1983; Sandberg et al.,
(Hauser et al., 2017; Hauser et al., 2008). The onset of therapeutic 1986), and such trafficking seems to be bidirectional (Stern et al., 2014;
effects in these patients was noted early, before any effect on serum von Budingen et al., 2012), with activation of B cells both outside
immunoglobulin levels (Bar-Or et al., 2008; Hauser et al., 2017; Hauser (Sandberg-Wollheim et al., 1986; Stern et al., 2014) and within the
et al., 2008). Notably, such treatment had no effect on the abnormal IgG neuraxis (Levinson et al., 1983; Sandberg et al., 1986; Monson et al.,
present in CSF of these patients (Piccio et al., 2010). Together, these 2005; Qin et al., 1998). Cloning techniques show that the same
findings clearly demonstrate an important role for B cells in mediating populations of B cells documented in the CNS of patients can also be
new MS disease activity, but also indicate that their contribution to detected in the peripheral immune compartment (Lovato et al., 2011;
such disease activity is independent of Ig secretion (Blauth et al., 2015a; Palanichamy et al., 2014; Hauser, 2015). Therefore studying B cells

94
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

from peripheral blood of patients may contribute to understanding the axonal damage and death in the white matter (Bjartmar et al., 2003;
role of B cells in the meninges as well as in CSF and CNS parenchyma in Jeffery et al., 2000; Jurgens et al., 2016), it also seems clear that some
MS pathogenesis. of the damage within the cortical gray matter is not directly related to
There is increasing understanding and interest in the complexity of lesions within the underlying white matter (Bo et al., 2007; Fischer
B cell subtypes and their functions (Mamula, 2013), with active et al., 2013; Haider et al., 2016; Lassmann et al., 2007). It has been
research on the roles of these different subsets of B cells in the postulated that some of the subpial the damage results from diffusion of
pathogenesis of MS (Antel and Bar-Or, 2006; Hauser, 2015). B cells cytotoxic factors, perhaps cytokines, from the meninges and CSF into
act as antigen presenting cells (APC) to T cells, which has been well the cortical gray matter (Serafini et al., 2007; Magliozzi et al., 2010;
demonstrated using the experimental autoimmune encephalomyelitis Howell et al., 2011; Lisak et al., 2012; Lassmann, 2014) and perhaps, by
model (Molnarfi et al., 2013; Weber et al., 2011). Molnarfi and extension, to the cerebellum as well (Kutzelnigg et al., 2007). The exact
colleagues also demonstrated that B cell APC function and formation distribution of meningeal inflammatory lesions is still not fully under-
of meningeal B cells aggregates was not dependent on myelin-specific stood in RRMS, SPMS and PPMS and so factors secreted by B cells might
antibody production (Molnarfi et al., 2013). Among the functions of well be able to circulate for short distances in the CSF and cause
particular interest in our studies are the antibody-independent effector damage at short distance from the meningeal inflammatory lesions. In
functions of B cells including production of cytokines and other non-Ig addition it is clear that inflammation in the meninges is not limited to
biological modifiers (Li et al., 2015; Lino et al., 2016). areas where the atypical germinal follicles are seen. And of course some
Subpopulations of B cells include effector B cells, some of which can changes in the cortical gray matter may be the result of lesions
secrete proinflammatory cytokines such as GM-CSF (Bhattacharya damaging axons and neurons downstream and at a distance from the
et al., 2015; Li et al., 2015), as well as what have been described as cortex, as mentioned above (Bjartmar et al., 2003; Jeffery et al., 2000;
‘killer B cells’ which can also serve regulatory functions (Lundy, 2009). Jurgens et al., 2016).
Is it possible that these cells might secrete factors that can injure other It has been suggested that other cells in the meninges, including
cell types? Additional studies will examine B cells from the blood of CD8 T cells reacting to activated EBV in B cells, release toxic factors
patients with progressive forms of MS to determine if Sup from B cell that contribute to cortical damage (Meier et al., 2012; Serafini et al.,
cultures from such patients also are cytotoxic to OL and neurons. Other 2007); however, several groups have not been able to demonstrate EBV
studies will include testing Sup from B cell cultures of patients with in CNS B cells in patients with MS (Lassmann et al., 2011; Sargsyan
other immune-mediated neurological diseases including myasthenia et al., 2010; Lassmann et al., 2011; Sargsyan et al., 2010; Willis et al.,
gravis (MG) and chronic inflammatory demyelinating polyneuropathy 2009). In other studies, in vitro killing of human neurons in culture by T
(CIDP) and systemic immune-mediated diseases like rheumatoid ar- cells seemed to require direct cell to cell contact (Giuliani et al., 2003),
thritis (RA) and systemic lupus erythematosis (SLE), and testing B cell yet cortical pathology in MS is notable for relative paucity of T cells
Sup from patients with MS and NC to determine if they produce death within the tissue (Fischer et al., 2013; Howell et al., 2011; Lassmann,
of Schwann cells, the myelin forming cells of the peripheral nervous 2012; Peterson et al., 2001).
system (PNS). Even if B cells from patients with MG, CIDP, RA or SLE For these reasons, we have considered whether soluble products,
secrete factors that damage OL or CNS neurons, or B cells from patients and particularly those secreted by meningeal B cells, may be toxic to the
with MS damage Schwann cells, this in no way diminishes the potential underlying cortex. We have shown that B cells obtained from patients
relevance of our findings in MS. These B cells from patients with MS with RRMS but not from NC produce one or more factors that cause
may secrete factors acting locally where B cells are present in the CNS. death of OL in our rat mixed glial cell cultures, but do not impact
Thus the difference might well be that in MS there are chemokine astrocytes or microglia. This killing of OL does not seem to be caused by
signals that attract and hold B cells in the CNS including the meninges IgM or IgG and is independent of complement activation (Lisak et al.,
and those factors are not present in the PNS or thymus of patients with 2012). The lack of apparent involvement of Ig or complement in this in
MS. It is likely a combination of where the B cells that mediate vitro toxicity to OL parallels the in vivo findings of type III lesions within
cytotoxicity go in which diseases. We are also performing studies to the cortical gray matter particularly in progressive forms of MS.
determine which subpopulations of B cells are producing the putative In the current study, we first extend our findings and demonstrate
toxic factors including testing Sup from memory (CD27+) and naïve that secretory products from B cells obtained from patients with RRMS
(CD27-) B cells (Duddy et al., 2007). (not receiving disease modifying therapy or any recent corticosteroids)
Studies of cortical pathology in patients with secondary progressive and cultured in vitro, produce one or more factors that induce death of
MS (SPMS) reveal meningeal inflammation that can be rich in B cells rat CNS neurons, and that the same effect is seen on human neurons in
(some of which have been described to have follicle-like properties), culture, demonstrating that this is not a toxic effect directed exclusively
with underlying demyelination and axonal/neuronal loss and a paucity at rat cells or even just against rat neurons or rat OL. A recent study
of inflammatory cells within cortex itself, particularly in (subpial) Type reported in abstract form showed that B cells from the blood of patients
III lesions (Howell et al., 2011; Kutzelnigg et al., 2005; Magliozzi et al., with MS induced apoptosis of neurons in cerebellar slice cultures
2007; Peterson et al., 2001). There is no evidence of Ig or significant (Harlow et al., 2016) supporting our earlier work on OL (Lisak et al.,
complement activation (Brink et al., 2005) in the type III lesions, lesions 2012) and our current study of rat and importantly human neurons.
that involve layers 1–4, although they may merge with deeper Type I Using a different assay for IgG and IgM than in our paper on OL
leukocortical lesions. Intense meningeal inflammation containing B cytotoxicity where we did not detect any measurable IgM and very low
cells has been reported in some patients with primary progressive MS levels of IgG in a minority of tested Sup (Lisak et al., 2012), we detected
(PPMS) (Choi et al., 2012) and in biopsies from some patients with very IgG and IgM at very low levels in the Sup we tested for neuronal
early RRMS in which B cells may also be found within the cortex cytotoxicity. As in our study of induction of OL death, there was no
(Lucchinetti et al., 2011). Meningeal inflammation has also been shown correlation between neuronal death and levels of IgG or IgM in the Sup
to correlate with underlying cerebellar atrophy (Kutzelnigg et al., of B cells from patients with RRMS or NC. As was true in our studies of
2007). In each case the degree of damage to the underlying cortical OL cell death, serum used in B cell cultures and neuronal cultures was
OL/myelin and neurons/axons is reportedly directly proportional to the heat inactivated, eliminating complement activity. Death of both rat
intensity of the overlying active meningeal disease (Calabrese et al., and human neurons in our system seems to be due in part to apoptosis
2009; Howell et al., 2011; Lucchinetti et al., 2011; Magliozzi et al., which is not what one would expect with death induced by antigen-
2010; Popescu and Lucchinetti, 2012), although one group did not antibody mediated complement activation.
observe this relationship (Kooi et al., 2009). While some have suggested In our previous study of induction of OL death by B cell secretory
much of this damage is the result of downstream demyelination and products, we measured levels of several cytokines associated with B

95
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

cells: TNF-α, LT-α, IL-6, IL-10 and TGF-β (Lisak et al., 2012). We did molecular weight components. For neurons, the toxicity in the
not detect a difference in levels of any of these cytokines in super- > 300 kDa fraction from MS Sup was 47% compared to 11% in the
natants from either nonstimulated or unstimulated B cells of patients < 300 kDa fraction, when corrected for the medium control. If
with RRMS and NC. There was no correlation of OL death with levels of confirmed with a larger sample size, this suggests that MS Sup may
any single cytokine or with any combination of cytokines including contain low levels of factors < 300 kDa that are toxic to neurons but
TNF-α + LT-α + IL-6, so-called ‘proinflammatory’ cytokines or IL- not OL. The apparent large size of the toxic factor(s) raises several
10 + TGF-β, considered to be ‘anti-inflammatory’ or ‘downregulatory’. possibilities; 1) cytokines or other proteins that are complexed together
TNF-α and LT-α have been reported to be toxic for OL and cause in a manner that they cannot be fully quantitated with immunologic
demyelination in vitro (Selmaj et al., 1991a; Selmaj et al., 1991b; Selmaj techniques like ELISA assays and Luminex chips, or 2) larger proteins
and Raine, 1988). In this study we examined levels of a larger number including IgM or aggregated IgG. This latter possibility seems unlikely
of cytokines and other protein biologic mediators. With the exception of given the evidence for killing, at least in part, by apoptosis, as
LT-β which may have been reduced in RRMS Sup, we did not find determined by three separate assays for apoptosis, and the lack of
significant differences in the levels of any of the cytokines or protein activated complement in our system. It does not appear to be simply
biological modifiers in the Sup of cultures of the B cells from RRMS cell debris since the pellet containing cell debris when resuspended was
patients compared to NC. The lack of an increase in the levels of pro- not toxic. If the toxicity is in exosomes or related extracellular vesicles
inflammatory molecules secreted from MS vs. NC B cells in our current in the > 300 kDa fraction, then miRNA or lipids in addition to proteins
study (in contrast to such previously reported differences, Bar-Or et al., within the vesicles are potential toxic factors. Extracellular vesicles
2010; Lino et al., 2016; Lisak et al., 2012) may reflect the fact that here including exosomes have been analyzed in serum and CSF of MS
we did not examine Sup from B cells stimulated in vitro whereas in patients. Their roles in MS pathogenesis and usefulness as biomarkers
earlier experiments Sup from B cells stimulated in vitro were also or therapeutic targets are active areas of investigation (Saenz-Cuesta
examined (Bar-Or et al., 2010; Lino et al., 2016; Lisak et al., 2012). et al., 2014; Carandini et al., 2015; Pusic et al., 2014; Selmaj et al.,
Analysis of the 14-PLEX array data (Table 2) showed a trend in Sup 2017). For example, proteomic analysis of CSF exosomes identified
for B cells from NC for increased levels of IL-6, a proinflammatory distinctive patterns for MS and neuromyelitis optica (Lee et al., 2016).
cytokine produced by many inflammatory cell types including B cells. Lipidomic analysis of exosome-enriched fractions from plasma showed
In Sup of B cells from RRMS patients, there was a trend for increased increased levels of sulfatide in MS samples compared to controls
levels of the chemokine MIP-1α, which primarily results in attraction of (Moyano et al., 2016). Profiling of miRNA patterns in plasma and
polymorphonuclear cells. That trend was not seen for MIP-1α using the CSF has revealed multiple differences between MS and controls;
25-PLEX assay (Table 2), with no differences in levels of MIP-1β. There extracellular vesicles are likely the major carriers for these miRNAs
were no differences in levels of MCP-1, IFN-α or IFN-γ. IL-16 levels (Gandhi, 2015; Jadot and Davoust, 2016; Miyazaki et al., 2014; Selmaj
were modestly increased in Sup from RRMS compared to NC when the et al., 2017). While extracellular vesicles released by normal B cells
one outlier among the NC is eliminated (Table 2). IL-16 is a proin- have been characterized in detail (Choi et al., 2013; Danger et al., 2014;
flammatory cytokine, produced by many inflammatory cell types McLellan, 2009), little is known about those released by B cells from MS
including B cells, that is important in the pathogenesis of experimental patients.
autoimmune encephalomyelitis and may be involved in the pathogen- The cargo of extracellular vesicles and exosomes may have toxic or
esis of MS (Skundric et al., 2015). B cells from patients with MS have protective effects on neurons and OL. Exosome-like particles can deliver
increased expression of MMP-9 (Aung et al., 2015), a protease that is neurotoxic or misfolded proteins or neuroprotective factors (Janas
important in opening of the blood brain barrier in inflammatory lesions et al., 2016; Yuyama and Igarashi, 2016). Some miRNAs can bind to
allowing peripheral immune cells to enter the parenchyma(Stuve et al., toll-like receptors and induce regulatory or cytotoxic effects in the
1996). We did not find an increase in levels of secreted MMP-9 in Sup of target cells (Chen et al., 2013). Astrocytes (Wang et al., 2012) and
B cells from patients with RRMS compared to NC, and no correlation cytokine-treated glioma cells (Podbielska et al., 2016) release ceramide-
between levels of MMP9 and neuronal cell death. enriched exosomes that can induce cell killing. It has been reported that
In earlier work we showed that B cells in the CSF of patients with ceramides are enriched in CSF of MS patients and can cause neuronal
MS were more activated than B cells in the blood of the same patients mitochondrial dysfunction trhough oxidative stress and damage to
(Levinson et al., 1983). There is also evidence that the blood B cells, not axons (Vidaurre et al., 2014). Under some conditions, immune cells
stimulated in culture, as well as other mononuclear inflammatory cells, release exosomes that promote CNS myelination and regulate inflam-
are in a higher state of activation than seen in normal individuals mation (Pusic and Kraig, 2014; Pusic et al., 2016). Mesenchymal stem
(Mathias et al., 2016). Thus these more activated B cells may be cell-derived exosomes can improve recovery from experimental trau-
producing and secreting greater amounts of one or more B cells matic brain injury (Zhang et al., 2015) and deliver miRNAs that
products when they are placed into culture. In our prior study (Lisak enhance neuronal differentiation (Lee et al., 2014). Exosomes from
et al., 2012) in vitro stimulation had a variable affect on B cell Sup from Schwann cells have been reported to regulate neuron/glia communica-
RRMS patients to kill OL when compared to B cells from the same tion and promote axonal regeneration (Ching and Kingham, 2015;
subject that had not undergone in vitro stimulation. In addition in vitro Lopez-Leal and Court, 2016). OL-derived exosome-like particles can
stimulation of B cells from NC further reduced any minimal toxic effects inhibit CNS myelin membrane sheath formation (Bakhti et al., 2011)
of Sup on OL (Lisak et al., 2012). but also regulate neuronal firing rate, signal transduction and gene
We found no correlation between the level of any single cytokine or regulation (Frohlich et al., 2014).
other protein tested and induction of cell death of rat neurons in vitro in Current studies to identify the toxic factor or factors secreted by B
these experiments, as illustrated by several examples in Fig. 5. Analysis cells of patients with RRMS are underway, focusing primarily on
of combinations of cytokine levels, either proinflammatory or down- the > 300 kDa retentates of the Sup, employing proteomics and other
regulatory, failed to reveal any combinations that showed any correla- approaches. It is possible that the toxic factor/s is/are not proteins. It is
tion with neuronal cell death. These results indicate that none of the also possible that B cells from RRMS patients compared to those from
cytokines analyzed are likely candidates for neurotoxic factors present NC secrete less of one or more factors necessary for OL and neuronal
in the RRMS B cell Sup, or as protective factors, absent in the RRMS health in vitro.
Sup, but present in the NC Sup.
Our studies on the molecular weight of the toxic factor or factors 5. Conclusions
indicate that they are primarily > 300 kDa in size. In the case of OL, no
significant toxicity was found in the filtrate containing the lower RRMS B cells secrete factor/s in vitro toxic to neurons and OL

96
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

independent of Ig, not complement-mediated, and involving apoptosis. pathology of multiple sclerosis is location-dependent: no significant complement
activation is detected in purely cortical lesions. J. Neuropathol. Exp. Neurol. 64,
The factor/s toxic to neurons and oligodendrocytes have an apparent 147–155.
mw of > 300 kDa and modest neuronal toxicity may also be induced by von Budingen, H.C., Kuo, T.C., Sirota, M., van Belle, C.J., Apeltsin, L., Glanville, J., Cree,
factor/s of < 300 kDa. These results provide significant new informa- B.A., Gourraud, P.A., Schwartzburg, A., Huerta, G., Telman, D., Sundar, P.D., Casey,
T., Cox, D.R., Hauser, S.L., 2012. B cell exchange across the blood-brain barrier in
tion about a functional difference in B cells from multiple sclerosis multiple sclerosis. J. Clin. Invest. 122, 4533–4543.
patients compared to controls, and elucidate a novel mechanism by Calabrese, M., Rocca, M.A., Atzori, M., Mattisi, I., Bernardi, V., Favaretto, A., Barachino,
which B cells could contribute to disease. L., Romualdi, C., Rinaldi, L., Perini, P., Gallo, P., Filippi, M., 2009. Cortical lesions in
primary progressive multiple sclerosis: a 2-year longitudinal MR study. Neurology 72,
1330–1336.
Funding Carandini, T., Columbo, F., Finardi, A., Casella, G., Garzetti, L., Verderio, C., Furlan, R.,
2015. Microvesicles: what is the role in mutliple sclerosis. Front. Neurol. 6, 1–7.
Chen, X., Liang, H., Zhang, J., Zen, K., Zhang, C.Y., 2013. MicroRNAs are ligands of Toll-
Funding is provided by the National Multiple Sclerosis Society USA,
like receptors. RNA 19, 737–739.
Grant #4929A2/3 (RPL, JAB, AB-O), the Research Foundation of the Ching, R., Kingham, P., 2015. The role of exosomes in peripheral nerve regeneration.
MS Society of Canada, Grant #204209 (AB-O), the Parker Webber Chair Neural Regen. Res. 10, 743–747.
in Neurology Endowment (DMC Foundation/Wayne State University Choi, S.R., Howell, O.W., Carassiti, D., Magliozzi, R., Gveric, D., Muraro, P.A., Nicholas,
R., Roncaroli, F., Reynolds, R., 2012. Meningeal inflammation plays a role in the
School of Medicine; RPL) and DMC Foundation, Grant #2015-1242 pathology of primary progressive multiple sclerosis. Brain 135, 2925–2937.
(JAB). HT is supported by a doctoral studentship from the MS Society of Choi, D.-S., Kim, D.-K., Kim, Y.-K., Gho, Y., 2013. Proteomics, transcriptomics and
Canada. RL is supported by a Banque National Translational Research lipidomics of exosomes and ectosomes. Proteomics 13, 1554–1571.
Cross, A.H., Wu, G.F., 2010. Multiple sclerosis: oligoclonal bands still yield clues about
Fellowship of the Montreal Neurological Institute. multiple sclerosis. Nat. Rev. Neurol. 6, 588–589.
Danger, R., Braza, F., Giral, M., Soulillou, J.-P., Brouard, S., 2014. MicroRNAs, major
Acknowledgments players in B cells homeostasis and function. Front. Immunol. 5, 1–9.
Duddy, M.E., Alter, A., Bar-Or, A., 2004. Distinct profiles of human B cell effector
cytokines: a role in immune regulation? J. Immunol. 172, 3422–3427.
We are grateful to all MS patients and controls who provided Duddy, M., Niino, M., Adatia, F., Hebert, S., Freedman, M., Atkins, H., Kim, H.J., Bar-Or,
samples and would like to thank the members of the Experimental A., 2007. Distinct effector cytokine profiles of memory and naive human B cell
subsets and implication in multiple sclerosis. J. Immunol. 178, 6092–6099.
Therapeutics Program, including Dr. Boli Fan, as well as the Clinical
Eide, L., McMurray, C.T., 2005. Culture of adult mouse neurons. BioTechniques 38,
Research Unit (CRU) team, at the Montreal Neurological Institute for 99–104.
their support in sample procurement and sample processing. We also Ferguson, B., Matyszak, M.K., Esiri, M.M., Perry, V.H., 1997. Axonal damage in acute
multiple sclerosis lesions. Brain 120, 393–399.
thank Dr. Paul Stemmer, Institute of Environmental Health Sciences,
Fischer, M.T., Wimmer, I., Hoftberger, R., Gerlach, S., Haider, L., Zrzavy, T., Hametner,
Director of the Proteomics Core at Wayne State University for helpful S., Mahad, D., Binder, C.J., Krumbholz, M., Bauer, J., Bradl, M., Lassmann, H., 2013.
discussions. Disease-specific molecular events in cortical multiple sclerosis lesions. Brain 136,
1799–1815.
Fisher, E., Lee, J.C., Nakamura, K., Rudick, R.A., 2008. Gray matter atrophy in multiple
References sclerosis: a longitudinal study. Ann. Neurol. 64, 255–265.
Fisniku, L.K., Chard, D.T., Jackson, J.S., Anderson, V.M., Altmann, D.R., Miszkiel, K.A.,
Absinta, M., Rocca, M.A., Moiola, L., Copetti, M., Milani, N., Falini, A., Comi, G., Filippi, Thompson, A.J., Miller, D.H., 2008. Gray matter atrophy is related to long-
M., 2011. Cortical lesions in children with multiple sclerosis. Neurology 76, 910–913. termdisability in multiple sclerosis. Ann. Neurol. 64, 247–254.
Antel, J., Bar-Or, A., 2006. Roles of immunoglobulins and B cells in multiple sclerosis: Frohlich, D., Kuo, W.P., Fruhbeis, C., Sun, J.J., Zehendner, C.M., Luhmann, H.J., Pinto, S.,
from pathogenesis to treatment. J. Neuroimmunol. 180, 3–8. Toedling, J., Trotter, J., Kramer-Albers, E.M., 2014. Multifaceted effects of
Aung, L.L., Mouradian, M.M., Dhib-Jalbut, S., Balashov, K.E., 2015. MMP-9 expression is oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal
increased in B lymphocytes during multiple sclerosis exacerbation and is regulated by transduction and gene regulation. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 369.
microRNA-320a. J. Neuroimmunol. 278, 185–189. Gandhi, R., 2015. miRNA in multiple sclerosis: search for novel biomarkers. Mult. Scler.
Bakhti, M., Winter, C., Simons, M., 2011. Inhibition of myelin membrane sheath 21, 1095–1103.
formation by oligodendrocyte-derived exosome-like vesicles. J. Biol. Chem. 286, Genain, C.P., Cannella, B., Hauser, S.L., Raine, C.S., 1999. Identification of autoantibodies
787–796. associated with myelin damage in multiple sclerosis [see comments]. Nat. Med. 5,
Bar-Or, A., Oliveira, E.M., Anderson, D.E., Krieger, J.I., Duddy, M., O'Connor, K.C., Hafler, 170–175.
D.A., 2001. Immunological memory: contribution of memory B cells expressing Gilden, D.H., 2005. Infectious causes of multiple sclerosis. Lancet Neurol. 4, 195–202.
costimulatory molecules in the resting state. J. Immunol. 167, 5669–5677. Giuliani, F., Goodyer, C.G., Antel, J.P., Yong, V.W., 2003. Vulnerability of human neurons
Bar-Or, A., Calabresi, P.A., Arnold, D., Markowitz, C., Shafer, S., Kasper, L.H., Waubant, to T cell-mediated cytotoxicity. J. Immunol. 171, 368–379.
E., Gazda, S., Fox, R.J., Panzara, M., Sarkar, N., Agarwal, S., Smith, C.H., 2008. Haider, L., Zrzavy, T., Hametner, S., Hoftberger, R., Bagnato, F., Grabner, G., Trattnig, S.,
Rituximab in relapsing-remitting multiple sclerosis: a 72-week, open-label, phase I Pfeifenbring, S., Bruck, W., Lassmann, H., 2016. The topograpy of demyelination and
trial. Ann. Neurol. 63, 395–400. neurodegeneration in the multiple sclerosis brain. Brain 139, 807–815.
Bar-Or, A., Fawaz, L., Fan, B., Darlington, P.J., Rieger, A., Ghorayeb, C., Calabresi, P.A., Harlow, D., Blauth, K., Saul, K., Selva, S., Niedzielko, T., Jackson, L., Macklin, W.,
Waubant, E., Hauser, S.L., Zhang, J., Smith, C.H., 2010. Abnormal B-cell cytokine Vollmer, T., 2016. B lymphoctes from MS patients induce CNS pathology in a murine
responses a trigger of T-cell-mediated disease in MS? Ann. Neurol. 67, 452–461. brain slice model. Abstracts on line American Academy of Neurology P5, 321.
Benjamins, J.A., Nedelkoska, L., Lisak, R.P., 2014. Adrenocorticotropin hormone 1-39 Hauser, S.L., 2015. The charcot lecture | Beating MS: a story of B cells, with twists and
promotes proliferation and differentiation of oligodendroglial progenitor cells and turns. Mult. Scler. 21, 8–21.
protects from excitotoxic and inflammation-related damage. J. Neurosci. Res. 92, Hauser, S.L., Waubant, E., Arnold, D.L., Vollmer, T., Antel, J., Fox, R.J., Bar-Or, A.,
1243–1251. Panzara, M., Sarkar, N., Agarwal, S., Langer-Gould, A., Smith, C.H., 2008. B-cell
Bhattacharya, P., Thiruppathi, M., Elshabrawy, H.A., Alharshawi, K., Kumar, P., depletion with rituximab in relapsing-remitting multiple sclerosis. N. Engl. J. Med.
Prabhakar, B.S., 2015. GM-CSF: an immune modulatory cytokine that can suppress 358, 676–688.
autoimmunity. Cytokines. Hauser, S.L., Bar-Or, A., Comi, G., Giovannoni, G., Hartung, H.P., Hemmer, B., Lublin, F.,
Bjartmar, C., Wujek, J.R., Trapp, B.D., 2003. Axonal loss in the pathology of MS: Montalban, X., Rammohan, K.W., Selmaj, K., Traboulsee, A., Wolinsky, J.S., Arnold,
consequences for understanding the progressive phase of the disease. J. Neurol. Sci. D.L., Klingelschmitt, G., Masterman, D., Fontoura, P., Belachew, S., Chin, P., Mairon,
206, 165–171. N., Garren, H., Kappos, L., Opera, I., Investigators, O.I.C., 2017. Ocrelizumab versus
Blauth, K., Soltys, J., Matschulat, A., Reiter, C.R., Ritchie, A., Baird, N.L., Bennett, J.L., interferon beta-1a in relapsing multiple sclerosis. N. Engl. J. Med. 376, 221–234.
Owens, G.P., 2015. Antibodies produced by clonally expanded plasma cells in Hawker, K., O'Connor, P., Freedman, M.S., Calabresi, P.A., Antel, J., Simon, J., Hauser, S.,
multiple sclerosis cerebrospinal fluid cause demyelination of spinal cord explants. Waubant, E., Vollmer, T., Panitch, H., Zhang, J., Chin, P., Smith, C.H., 2009.
Acta Neuropathol. 130, 765–781. Rituximab inpatients with primary progressive multiple sclerosis: results of a
Bo, L., Geurts, J.J., Mork, S.J., van der Valk, P., 2006. Grey matter pathology in multiple randomized double-blind placebo-controlled multicenter trial. Ann. Neurol. 66,
sclerosis. Acta Neurol. Scand. Suppl. 183, 48–50. 460–471.
Bo, L., Geurts, J.J., van der Valk, P., Polman, C., Barkhof, F., 2007. Lack of correlation Howell, O.W., Reeves, C.A., Nicholas, R., Carassiti, D., Radotra, B., Gentleman, S.M.,
between cortical demyelination and white matter pathologic changes in multiple Serafini, B., Aloisi, F., Roncaroli, F., Magliozzi, R., Reynolds, R., 2011. Meningeal
sclerosis. Arch. Neurol. 64, 76–80. inflammation is widespread and linked to cortical pathology in multiple sclerosis.
Brandle, S., Obermeier, B., Senel, M., Bruder, J., Mentele, R., Khademi, M., Olsson, T., Brain 134, 27552771.
Tumani, H., Kristoferitsch, W., Lottspeich, F., Wekerle, H., Hohlfeld, R., Dornmair, K., Jadot, F., Davoust, N., 2016. Is it worth considering circulating microRNAs in multiple
2016. Distinct oligoclonal band antibodies in multiple sclerosis recognize ubiquitous sclerosis? Front. Immunol. 7, 129.
self-proteins. Proc. Natl. Acad. Sci. U. S. A. 113, 7864–7869. Janas, A.M., Sapon, K., Janas, T., Stowell, M.H., Janas, T., 2016. Exosomes and other
Brink, B.P., Veerhuis, R., Breij, E.C., van der Valk, P., Dijkstra, C.D., Bo, L., 2005. The extracellular vesicles in neural cells and neurodegenerative diseases. Biochim.

97
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

Biophys. Acta 1858, 1139–1151. 203–217.


Jeffery, D.R., Absher, J., Pfeiffer, F.E., Jackson, H., 2000. Cortical deficits in multiple Meier, U.C., Giovannoni, G., Tzartos, J.S., Khan, G., 2012. Translational mini-review
sclerosis on the basis of subcortical lesions. Mult. Scler. 6, 50–55. series on B cell subsets in disease. B cells in multiple sclerosis: drivers of disease
Jurgens, T., Jafari, M., Kreutzfeldt, M., Bahn, E., Bruck, W., Kerschensteiner, M., Merkler, pathogenesis and Trojan horse for Epstein-Barr virus entry to the central nervous
D., 2016. Reconstruction of single cortical projection neurons reveals primary spine system? Clin. Exp. Immunol. 167, 1–6.
loss in multiple sclerosis. Brain 139, 39–46. Miller, J.R., Burke, A.M., Bever, C.T., 1983. Occurrence of oligoclonal bands in multiple
Kabat, E.A., Moore, D.H., Landow, H., 1942. An electrophoretic study of the protein sclerosis and other CNS diseases. Ann. Neurol. 13, 53–58.
components in cerebrospinal fluid and their relationship to the serum proteins. J. Miyazaki, Y., Li, R., Rezk, A., Misirliyan, H., Moore, C., Farooqi, N., Solis, M., Goiry, L.G.,
Clin. Invest. 21, 571–577. de FariaJunior, O., Dang, V.D., Colman, D., Dhaunchak, A.S., Antel, J., Gommerman,
Kooi, E.J., Geurts, J.J., van Horssen, J., Bo, L., van der Valk, P., 2009. Meningeal J., Prat, A., Fillatreau, S., Bar-Or, A., C.M.N.E.T.G.i.C. Autoimmunity, M.C.B.c.i.M.
inflammation is not associated with cortical demyelination in chronic multiple Team, 2014. Anovel microRNA-132-sirtuin-1 axis underlies aberrant B-cell cytokine
sclerosis. J. Neuropathol. Exp. Neurol. 68, 1021–1028. regulation in patients with relapsing-remitting multiple sclerosis [corrected]. PLoS
Kutzelnigg, A., Lucchinetti, C.F., Stadelmann, C., Bruck, W., Rauschka, H., Bergmann, M., One 9, e105421.
Schmidbauer, M., Parisi, J.E., Lassmann, H., 2005. Cortical demyelination and diffuse Molnarfi, N., Schulze-Topphoff, U., Weber, M.S., Patarroyo, J.C., Prod'homme, T., Varrin-
white matter injury in multiple sclerosis. Brain 128, 2705–2712. Doyer, M., Shetty, A., Linington, C., Slavin, A.J., Hidalgo, J., Jenne, D.E., Wekerle, H.,
Kutzelnigg, A., Faber-Rod, J.C., Bauer, J., Lucchinetti, C.F., Sorensen, P.S., Laursen, H., Sobel, R.A., Bernard, C.C., Shlomchik, M.J., Zamvil, S.S., 2013. MHC class II-
Stadelmann, C., Bruck, W., Rauschka, H., Schmidbauer, M., Lassmann, H., 2007. dependent B cell APC function is required for induction of CNS autoimmunity
Widespread demyelination in the cerebellar cortex in multiple sclerosis. Brain Pathol. independent of myelin-specific antibodies. J. Exp. Med. 210, 2921–2937.
17, 38–44. Monson, N.L., Brezinschek, H.P., Brezinschek, R.I., Mobley, A., Vaughan, G.K., Frohman,
Lassmann, H., 2012. Cortical lesions in multiple sclerosis: inflammation versus E.M., Racke, M.K., Lipsky, P.E., 2005. Receptor revision and atypical mutational
neurodegeneration. Brain 135, 2904–2905. characteristics in clonally expanded B cells from the cerebrospinal fluid of recently
Lassmann, H., 2014. Multiple sclerosis: lessons from molecular neuropathology. Exp. diagnosed multiple sclerosis patients. J. Neuroimmunol. 158, 170–181.
Neurol. 262PA, 2–7. Montalban, X., Hauser, S.L., Kappos, L., Arnold, D.L., Bar-Or, A., Comi, G., de Seze, J.,
Lassmann, H., Bruck, W., Lucchinetti, C.F., 2007. The immunopathology of multiple Giovannoni, G., Hartung, H.P., Hemmer, B., Lublin, F., Rammohan, K.W., Selmaj, K.,
sclerosis: an overview. Brain Pathol. 17, 210–218. Traboulsee, A., Sauter, A., Masterman, D., Fontoura, P., Belachew, S., Garren, H.,
Lassmann, H., Niedobitek, G., Aloisi, F., Middeldorp, J.M., 2011. Epstein-Barr virus in the Mairon, N., Chin, P., Wolinsky, J.S., Investigators, O.C., 2017. Ocrelizumab versus
multiple sclerosis brain: a controversial issue–report on a focused workshop held in placebo in primary progressive multiple sclerosis. N. Engl. J. Med. 376, 209–220.
the Centre for Brain Research of the Medical University of Vienna, Austria. Brain. Moyano, A.L., Li, G., Boullerne, A.I., Feinstein, D.L., Hartman, E., Skias, D., Balavanov, R.,
134, 2772–2786. vanBreemen, R.B., Bongarzone, E.R., Mansson, J.E., Givogri, M.I., 2016. Sulfatides in
Lee, H., Finnis, S., Cazacu, S., Xiang, C., Broddie, C., 2014. Mesenchymal stem cells extracellular vesicles isolated from plasma of multiple sclerosis patients. J. Neurosci.
deliver exogenous miRNAs to neural stem cells and induce their differentiation and Res. 94, 1579–1587.
glutamate transporter expression. Stem Cells Dev. 23, 2851–2861. Palanichamy, A., Apeltsin, L., Kuo, T.C., Sirota, M., Wang, S., Pitts, S.J., Sundar, P.D.,
Lee, J., McKinney, K.Q., Pavlopoulos, A.J., Han, M.H., Kim, S.H., Kim, H.J., Hwang, S., Telman, D., Zhao, L.Z., Derstine, M., Abounasr, A., Hauser, S.L., von Budingen, H.C.,
2016. Exosomal proteome analysis of cerebrospinal fluid detects biosignatures of 2014. Immunoglobulin class-switched B cells form an active immune axis between
neuromyelitis optica and multiple sclerosis. Clin. Chim. Acta 462, 118–126. CNS and periphery in multiple sclerosis. Sci. Transl. Med. 6, 248ra106.
Levinson, A.I., Sandberg-Wollheim, M., Lisak, R.P., Zweiman, B., Sjogren, K., Laramore, Peterson, J.W., Bo, L., Mork, S., Chang, A., Trapp, B.D., 2001. Transected neurites,
C., Moskovitz, A.R., Hedstrom, S.A., 1983. Analysis of B-cell activation of apoptotic neurons, and reduced inflammation in cortical multiple sclerosis lesions.
cerebrospinal fluid lymphocytes in multiple sclerosis. Neurology 33, 1305–1310. Ann. Neurol. 50, 389–400.
Li, R., Rezk, A., Miyazaki, Y., Hilgenberg, E., Touil, H., Shen, P., Moore, C.S., Michel, L., Piccio, L., Naismith, R.T., Trinkaus, K., Klein, R.S., Parks, B.J., Lyons, J.A., Cross, A.H.,
Althekair, F., Rajasekharan, S., Gommerman, J.L., Prat, A., Fillatreau, S., Bar-Or, A., 2010. Changes in B-and T-lymphocyte and chemokine levels with rituximab
B.c.i.M.S.T.Canadian, 2015. Proinflammatory GM-CSF-producing B cells in multiple treatment in multiple sclerosis. Arch. Neurol. 67, 707–714.
sclerosis and B cell depletion therapy. Sci. Transl. Med. 7, 310ra166. Podbielska, M., Szulc, Z.M., Kurowska, E., Hogan, E.L., Bielawski, J., Bielawska, A., Bhat,
Link, H., Huang, Y.M., 2006. Oligoclonal bands in multiple sclerosis cerebrospinal fluid: N.R., 2016. Cytokine-induced release of ceramide-enriched exosomes as a mediator
an update on methodology and clinical usefulness. J. Neuroimmunol. 180, 17–28. of cell death signaling in an oligodendroglioma cell line. J. Lipid Res.
Lino, A.C., Dorner, T., Bar-Or, A., Fillatreau, S., 2016. Cytokine-producing B cells: a Polman, C.H., Reingold, S.C., Banwell, B., Clanet, M., Cohen, J.A., Filippi, M., Fujihara,
translational view on their roles in human and mouse autoimmune diseases. K., Havrdova, E., Hutchinson, M., Kappos, L., Lublin, F.D., Montalban, X., O'Connor,
Immunol. Rev. 269, 130–144. P., Sandberg-Wollheim, M., Thompson, A.J., Waubant, E., Weinshenker, B.,
Lisak, R.P., Heinze, R.G., Falk, G.A., Kies, M.W., 1968. Search for anti-encephalitogen Wolinsky, J.S., 2011. Diagnostic criteria for multiple sclerosis: 2010 revisions to the
antibody in human demyelinative diseases. Neurology 18, 122–128. McDonaldcriteria. Ann. Neurol. 69, 292–302.
Lisak, R.P., Benjamins, J.A., Bealmear, B., Yao, B., Land, S., Nedelkoska, L., Skundric, D., Popescu, B.F., Lucchinetti, C.F., 2012. Meningeal and cortical grey matter pathology in
2006. Differential effects of Th1, monocyte/macrophage and Th2 cytokine mixtures multiple sclerosis. BMC Neurol. 12, 11.
on early gene expression for immune-related molecules by central nervous system Pusic, A., Kraig, R., 2014. Youth and environmental enrichment generate serum exosomes
mixed glial cell cultures. Mult. Scler. 12, 149–168. containing miR-219 that promote CNS myelination. Glia 62, 284–299.
Lisak, R.P., Nedelkoska, L., Studzinski, D., Bealmear, B., Xu, W., Benjamins, J.A., 2011. Pusic, A., Lusic, K., Kraig, R., 2014. What are exosomes and how can they be used in
Cytokines regulate neuronal gene expression: differential effects of Th1, Th2 and multiple sclerosis therapy? Expert. Rev. Neurother. 14, 353–355.
monocyte/macrophage cytokines. J. Neuroimmunol. 238, 19–33. Pusic, K., Pusic, A., Kraig, R., 2016. Envirnomental enrichment stimulates immune cell
Lisak, R.P., Benjamins, J.A., Nedelkoska, L., Barger, J.L., Ragheb, S., Fan, B., Ouamara, N., secretion of exosomes that promote CNS and may regulate inflammation. Cell. Mol.
Johnson, T.A., Rajasekharan, S., Bar-Or, A., 2012. Secretory products of multiple Neurobiol. 36, 313–325.
sclerosis B cells are cytotoxic to oligodendroglia in vitro. J. Neuroimmunol. 246, Qin, Y., Duquette, P., Zhang, Y., Talbot, P., Poole, R., Antel, J., 1998. Clonal expansion
85–95. and somatic hypermutation of V(H) genes of B cells from cerebrospinal fluid in
Lopez-Leal, R., Court, F.A., 2016. Schwann cell exosomes mediate neuron-glia multiple sclerosis. J. Clin. Invest. 102, 1045–1050.
communication and enhance axonal regeneration. Cell. Mol. Neurobiol. 36, 429–436. Rocca, M.A., Mezzapesa, D.M., Ghezzi, A., Falini, A., Martinelli, V., Scotti, G., Comi, G.,
Lovato, L., Willis, S.N., Rodig, S.J., Caron, T., Almendinger, S.E., Howell, O.W., Reynolds, Filippi, M., 2005. A widespread pattern of cortical activations in patients at
R., O'Connor, K.C., Hafler, D.A., 2011. Related B cell clones populate the meninges presentation with clinically isolated symptoms is associated with evolution to definite
and parenchyma of patients with multiple sclerosis. Brain 134, 534–541. multiple sclerosis. AJNR Am. J. Neuroradiol. 26, 1136–1139.
Lucchinetti, C.F., Popescu, B.F., Bunyan, R.F., Moll, N.M., Roemer, S.F., Lassmann, H., Rudick, R.A., Trapp, B.D., 2009. Gray-matter injury in multiple sclerosis. N. Engl. J. Med.
Bruck, W., Parisi, J.E., Scheithauer, B.W., Giannini, C., Weigand, S.D., Mandrekar, J., 361, 1505–1506.
Ransohoff, R.M., 2011. Inflammatory cortical demyelination in early multiple Saenz-Cuesta, M., Osorio-Querejeta, I., Otaegui, D., 2014. Extracellular vesicles in
sclerosis. N. Engl. J. Med. 365, 2188–2197. multiple sclerosis:What are they telling us? Front. Cell. Neurosci. 28, 100.
Lundy, S., 2009. Killer B lymphocytes: the evidence and the potential. Inflamm. Res. 58, Sandberg, M., Levinson, A.I., Zweiman, B., Lisak, R.P., 1986. B cell activation in multiple
345–357. sclerosis. Acta Neurol. Scand. 74, 417–424.
Magliozzi, R., Howell, O., Vora, A., Serafini, B., Nicholas, R., Puopolo, M., Reynolds, R., Sandberg-Wollheim, M., Zweiman, B., Levinson, A.I., Lisak, R.P., 1986. Humoral immune
Aloisi, F., 2007. Meningeal B-cell follicles in secondary progressive multiple sclerosis responses within the human central nervous system following systemic
associate with early onset of disease and severe cortical pathology. Brain 130, immunization. J. Neuroimmunol. 11, 205–214.
10891104. Sargsyan, S.A., Shearer, A.J., Ritchie, A.M., Burgoon, M.P., Anderson, S., Hemmer, B.,
Magliozzi, R., Howell, O.W., Reeves, C., Roncaroli, F., Nicholas, R., Serafini, B., Aloisi, F., Stadelmann, C., Gattenlohner, S., Owens, G.P., Gilden, D., Bennett, J.L., 2010.
Reynolds, R., 2010. A gradient of neuronal loss and meningeal inflammation in Absence of Epstein-Barr virus in the brain and CSF of patients with multiple sclerosis.
multiple sclerosis. Ann. Neurol. 68, 477–493. Neurology 74, 1127–1135.
Mamula, M., 2013. Editorial: B cells: not just making immunogloblulin anymore. Arthritis Selmaj, K.W., Raine, C.S., 1988. Tumor necrosis factor mediates myelin and
Rheum. 66, 2–5. oligodendrocyte damage in vitro. Ann. Neurol. 23, 339–346.
Mathias, A., Perriard, G., Canales, M., Soneson, C., Delorenzi, M., Schluep, M., Selmaj, K., Raine, C.S., Cannella, B., Brosnan, C.F., 1991a. Identification of lymphotoxin
DuPasquier, R.A., 2016. Increased ex Vivo Antigen Presentation Profile of B Cells in and tumor necrosis factor in multiple sclerosis lesions. J. Clin. Invest. 87, 949–954.
Multiple Sclerosis. (Mult Scler). Selmaj, K., Raine, C.S., Farooq, M., Norton, W.T., Brosnan, C.F., 1991b. Cytokine
McCarthy, K.D., de Vellis, J., 1980. Preparation of separate astroglial and cytotoxicity against oligodendrocytes. Apoptosis induced by lymphotoxin. J.
oligodendroglial cell cultures from rat cerebral tissue. J. Cell Biol. 85, 890–902. Immunol. 147, 1522–1529.
McLellan, A.D., 2009. Exosome release by primary B cells. Crit. Rev. Immunol. 29, Selmaj, I., Mycko, M.P., Raine, C.S., Selmaj, K.W., 2017. The role of exosomes in CNS

98
R.P. Lisak et al. Journal of Neuroimmunology 309 (2017) 88–99

inflammation and their involvement in multiple sclerosis. J. Neuroimmunol. 306, transection in the lesions of multiple sclerosis [see comments]. N. Engl. J. Med. 338,
1–10. 278–285.
Serafini, B., Rosicarelli, B., Magliozzi, R., Stigliano, E., Aloisi, F., 2004. Detection of Vidaurre, O.G., Haines, J.D., Katz Sand, I., Adula, P., Huynh, J.L., McGraw, C.A., Zhang,
ectopic B-cell follicles with germinal centers in the meninges of patients with F., Varghese, M., Sotirchos, E., Bhargava, P., Bandaru, V.V., Pasinetti, G., Zhang, W.,
secondary progressive multiple sclerosis. Brain Pathol. 14, 164–174. Inglese, M., Calabresi, P.A., Wu, G., Miller, A.E., Haughey, N.J., Lublin, F.D.,
Serafini, B., Rosicarelli, B., Franciotta, D., Magliozzi, R., Reynolds, R., Cinque, P., Casaccia, P., 2014. Cerebrospinal fluid ceramides from patients with multiple
Andreoni, L., Trivedi, P., Salvetti, M., Faggioni, A., Aloisi, F., 2007. Dysregulated sclerosis impair neuronal bioenergetics. Brain 137, 2271–2286.
Epstein-Barr virus infection in the multiple sclerosis brain. J. Exp. Med. 204, Wang, G., Dinkins, M., He, Q., Zhu, G., Poirier, C., Campbell, A., Mayer-Proschel, M.,
2899–2912. Bieberich, E., 2012. Astrocytes secrete exosomes enriched with proapoptotic
Skoff, A.M., Lisak, R.P., Bealmear, B., Benjamins, J.A., 1998. TNF-alpha and TGF-beta act ceramide and prostate apoptosis response 4 (PAR-4): potential mechanism of
synergistically to kill Schwann cells. J. Neurosci. Res. 53, 747–756. apoptosis induction in Alzheimer disease (AD). J. Biol. Chem. 287, 21384–21395.
Skundric, D.S., Cruikshank, W.W., Montgomery, P.C., Lisak, R.P., Tse, H.Y., 2015. Weber, M.S., Hemmer, B., Cepok, S., 2011. The role of antibodies in multiple sclerosis.
Emerging role of IL-16 in cytokine-mediated regulation of multiple sclerosis. Biochim. Biophys. Acta 1812, 239–245.
Cytokine 75, 234–248. Willis, S.N., Stadelmann, C., Rodig, S.J., Caron, T., Gattenloehner, S., Mallozzi, S.S.,
Stern, J.N., Yaari, G., Vander Heiden, J.A., Church, G., Donahue, W.F., Hintzen, R.Q., Roughan, J.E., Almendinger, S.E., Blewett, M.M., Brück, W., Hafler, D.A., O'Connor,
Huttner, A.J., Laman, J.D., Nagra, R.M., Nylander, A., Pitt, D., Ramanan, S., Siddiqui, K.C., 2009. Epstein-Barr virus infection is not a characteristic feature of multiple
B.A., Vigneault, F., Kleinstein, S.H., Hafler, D.A., O'Connor, K.C., 2014. B cells sclerosis brain. Brain 132, 3318–3328.
populating the multiple sclerosis brain mature in the draining cervical lymph nodes. Yuyama, K., Igarashi, Y., 2016. Physiological and pathological roles of exosomes in the
Sci. Transl. Med. 6, 248ra107. nervous system. Biomol. Concepts. 7, 53–68.
Stuve, O., Dooley, N.P., Uhm, J.H., Antel, J.P., Francis, G.S., Williams, G., Yong, V.W., Zhang, Y., Chopp, M., Meng, Y., Katakowski, M., Xin, H., Mahmood, A., Xiong, Y., 2015.
1996. Interferon beta-1b decreases the migration of T lymphocytes in vitro: effects on Effect of exosomes derived from multipluripotential stromal cells on functional
matrix metalloproteinase-9. Ann. Neurol. 40, 853–863. recovery and neurovascular plasticity in rats after traumatic brain injury. J.
Trapp, B.D., Peterson, J., Ransohoff, R.M., Rudick, R., Mork, S., Bo, L., 1998. Axonal Neurosurg. 122, 856–867.

99

You might also like