You are on page 1of 14

RESEARCH ARTICLE

Alterations of Juxtaparanodal Domains in


Two Rodent Models of CNS Demyelination
Lida Zoupi,1,2 Kyriaki Markoullis,3 Kleopas A. Kleopa,3 and Domna Karagogeos1,2

The segregation of myelinated fibers into distinct domains around the node of Ranvier—the perinodal areas—is crucial for
nervous system homeostasis and efficient nerve conduction. Perinodal areas are formed by axo-glial interactions, namely the
interaction of molecules between the axon and the myelinating glia. In a variety of demyelinating pathologies including multiple
sclerosis, the molecular architecture of the myelinated fiber is disrupted, leading to axonal degeneration. In this study we have
analyzed the alterations of TAG-1, Caspr2, and voltage-gated potassium channels (VGKCs), forming the juxtaparanodal tripartite
complex, in relation to adjacent paranodal and nodal molecules, in two different models of CNS demyelination, the experimental
autoimmune encephalomyelitis (EAE) and the cuprizone model of toxic demyelination. We found extensive alterations of the
juxtaparanodal molecular architecture under de- and remyelinating conditions. Inflammation alone was sufficient to disrupt the
borders between the domains leading to the diffusion of juxtaparanodal components to the adjacent paranodal area. EAE
induction and cuprizone-induced demyelination resulted initially in paranodal domain elongation with subsequent diffusion of
the juxtaparanodal components and the reduction of their expression levels. At later stages, with decreasing inflammation and
spontaneous remyelination there was a partial restoration of the paranodal domain but not sufficient re-organization of the
juxtaparanodes. The latter were re-formed only when complete remyelination was allowed in the cuprizone model, indicating
that juxtaparanodal domain reorganization is a later event that may remain incomplete in a hostile inflammatory milieu.

GLIA 2013;61:1236–1249
Key words: nodes of Ranvier; paranodes; EAE; cuprizone; demyelination

Introduction lesions has been put forward (Barnett et al., 2006; Bradl and Lass-
mann, 2010; Franklin et al., 2012; Haider et al., 2011; Lassmann,
M ultiple Sclerosis (MS) is a chronic, inflammatory, neurode-
generative disease of the central nervous system (CNS). MS
pathology includes multifocal demyelinated plaques accompanied
2001, 2003; Lassmann et al., 2001; Lassmann and Lucchinetti,
2008). However, in spite of many advances in the field, the mech-
by the formation of an active glial scar (Lassmann, 2001). In anisms of disease initiation and progression remain obscure.
actively demyelinating lesions, macrophages containing fragments A healthy myelinated fiber is periodically interrupted at
of myelin, microglia, and infiltrating T-cells are associated with the nodes of Ranvier. The anchored sodium channels are re-
lesion formation in the white matter, although recent studies sponsible for the generation and efficient propagation of axon
point out at alterations in gray matter areas as well (Barnett and potentials. Next to the node, a physical barrier to the lateral
Prineas, 2004; Lassmann, 2001, 2008; Lucchinetti et al., 2011; diffusion of nodal components is formed by the close apposi-
Prineas and Parratt, 2012). Lesion formation is a multi-factorial tion of the myelin membrane to the axon and the interaction
process that includes cytolytic factors like cytokines and proteases, of glial NF155 with the axonal immunoglobulin superfamily
secreted by inflammatory cells, complement activation, energy de- (IgSF) molecule Contactin-1 and the Neurexin superfamily
privation, oxidative, and nitrative damage. Recently a hypothesis protein Caspr/Paranodin (Charles et al., 2002; Labasque and
implicating oligodendrocyte alterations or damage as preceding Faivre-Sarrailh, 2010; Sherman et al., 2005; Susuki and Ras-
macrophage and microglia recruitment in newly formed MS band, 2008). Juxtaparanodes form adjacent to paranodes and

View this article online at wileyonlinelibrary.com. DOI: 10.1002/glia.22511

Published online Jul 5, 2013 in Wiley Online Library (wileyonlinelibrary.com). Received Jan 8, 2013, Accepted for publication Mar 20, 2013.

Address correspondence to Dr. O. Karagogeos, Domna Karagogeos, Department of Basic Science, Faculty of Medicine, University of Crete and Institute of Molecular
Biology and Biotechnology–Foundation for Research and Technology, Vassilika Vouton, 71110, Heraklion, Crete, Greece. E-mail: karagoge@imbb.forth.gr

From the 1Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion, Greece; 2Institute of Molecular Biology and BiotechnologyFoundation for
Research and Technology, Vassilika Vouton, 71110, Heraklion, Crete, Greece; 3Neuroscience Laboratory and Neurology Clinics, The Cyprus Institute of Neurology and
Genetics (CING), P.O. Box 23462, 1683 Nicosia, Cyprus.

Additional Supporting Information may be found in the online version of this article.

1236 V
C 2013 Wiley Periodicals, Inc.
Zoupi et al.: Juxtaparanodal Alterations in CNS-Demyelinating Models

are comprised of a tripartite complex between the Shaker- ing 16 to 18 g were purchased from HarlanTM (San Pietro Al Nati-
type voltage gated potassium channels (VGKCs), Caspr2 sone, Italy) and kept in our mouse facility for 2 weeks before EAE
(another member of the Neurexin Superfamily) on the axon induction at the age of 8 weeks. Animals were kept under controlled
conditions of temperature (21–23 C), humidity, air-exchange and
and the IgSF molecule TAG-1/Contactin-2 both on the axon
light cycle (12/12 h light/dark) and provided with standardized
and the glial cell (Labasque and Faivre-Sarrailh, 2010; Poliak
mouse diet and drinking water ad libitum. For disease induction,
and Peles, 2003; Poliak et al., 2003; Savvaki et al., 2008; mice were anaesthetized with an intraperitoneal injection of Tribro-
Traka et al., 2002, 2003). These molecular interactions medi- moethanol solution (AvertinV R, Sigma-Aldrich, Germany) and
ate the axo-glial contact that is essential for the organization injected subcutaneously in the dorsal area of the tail base with 160
of axonal domains and axonal homeostasis. lL of emulsion containing 200 lg (end volume: 2.5 mg/mL) of
Humoral responses were recently found to severely affect mouse recombinant myelin oligodendrocyte glycoprotein (rMOG)
the organization of white matter myelinated fibers in human and an equal volume of Complete Freund’s Adjuvant (CFA) mixed
MS lesions, initiating the disruption of the paranodal area and with 5 mg/mL desiccated M. tuberculosis (Difco Laboratories,
resulting in a progressive axonal denudation, and nerve conduc- Detroit, USA). rMOG was produced as previously described (Price
tion impairment (Coman et al., 2006; Howell et al., 2006, et al., 2002). Simultaneously with rMOG as well as 48 h later, each
2010; Wolswijk and Balesar, 2003). Moreover, axo-glial contact mouse received intraperitoneally 200 lL Pertussis Toxin (PTX) (1
lg/mL in PBS) (List Biological Laboratories, CA). Control mice (n
components (Neurofascin isoforms and Contactin-2/ TAG-1)
¼ 6) were injected with CFA mixed with M. tuberculosis and PTX
were also identified as auto-antigens in a subset of MS patients,
in the same way and time points as for EAE. A group of littermate
suggesting that a specific T cell-mediated immune response is
female mice (n ¼ 6) were used as naive controls. Mice were
directed against perinodal components, during the course of weighted and evaluated daily for clinical changes using the following
the disease (Derfuss et al., 2009; Mathey et al., 2007). These scale: 0 ¼ no clinical signs, 1 ¼ paralysis of the tail, 2 ¼ parapare-
results highlight the association of MS pathology with altered sis/monoplegia, 3 ¼ paraplegia/hemiparesis, 4 ¼ hemiplegia/quadri-
molecular organization of the myelinated fiber. paresis, 5 ¼ quadriplegia/moribund. Groups of mice (n ¼ 7 each)
To further clarify the nature and time course of these were sacrificed at 14 days post immunization (dpi), 2–3 days after
perinodal molecular changes in MS-related pathologies we per- the onset of the disease (MOG-EAE and CFA controls) and at 28
formed a detailed analysis of the tripartite complex of the jux- dpi (MOG-EAE and naive controls).
taparanodal domain under de-and remyelinating conditions, in
two different rodent models of CNS demyelination: the experi- Cuprizone Administration
mental autoimmune encephalomyelitis (EAE) that is character- Eight-week-old male mice (C57B110/CBA) were fed with 0.2%
ized as one of the rodent model of MS and the cuprizone cuprizone [oxalic bis(cyclohexylidenehydrazide)] (Sigma-Aldrich,
model of toxic demyelination. In these models we analyzed the USA) mixed with the animal’s standard chow. As previously
effects of myelin disruption on the localization and expression described (Matsushima and Morell, 2001), cuprizone toxin induces
of juxtaparanodal proteins, with or without the involvement of full demyelination of the corpus callosum when administered contin-
uously for 6 weeks, while toxin removal from the chow allows
a T-cell mediated immune response at different time points.
remyelination to occur. Juxtaparanodal complex formation was tested
We identified severe juxtaparanodal alterations under demyeli-
after 4.5 (demyelination can be visualized as distinct lesions) and 6
nating conditions in both models. The expression levels of all
weeks of cuprizone treatment (demyelination and partial remyelina-
three juxtaparanodal proteins were severely decreased compared tion). Additionally, 6 weeks of treatment with subsequent toxin re-
with control animals, with disruption of normal localization moval for 3 weeks (referred as 9 weeks) was considered as remyelina-
and diffusion. In both models, partial paranodal reorganization tion time point. All groups (3–4 animals/group) were compared
was evident correlating with spontaneous remyelination and with age and sex matched untreated controls.
decreasing inflammation, while juxtaparanodal reorganization
occurred only with complete remyelination in the cuprizone Immunohistochemistry
model. Our study provides insights into juxtaparanodal protein Spinal cords and cortices were harvested from mice after transcardial
complex involvement under demyelinating and remyelinating perfusion with 4% paraformaldehyde (PFA) in 0.1 M phosphate
conditions in relation to paranodal and nodal proteins in two buffer saline (PBS). Cervical and thoracic spinal cord segments were
different rodent models of CNS demyelination. isolated from MOG-EAE mice, for the present analysis. Tissues were
then post-fixed in the same fixative for 1 h (h), cryo-protected over-
night in 20% sucrose in 0.1 M PBS, embedded in OCT compound
Materials and Methods and stored at 80oC. Quantification of demyelination, inflamma-
EAE Induction tion and axonal loss was performed in transverse sections of the na-
For the purpose of the present analysis we used four different groups ive, CFA-14dpi and MOG-EAE cervical and thoracic segments as
of animals that have been previously described by Markoullis et al., previously described (Markoullis et al., 2012). Longitudinal white
(2012). Six-week-old female C57BL/6, wild type (WT) mice weight- matter cryosections from the posterior spinal cord (through the

August 2013 1237


dorsal columns) and corpus callosum (CC) of 10 lm were obtained 7.0, 150 mM NaCl, 2 mM EDTA, 50 mM Sodium fluoride, 1%
and mounted on SuperfrostV R Plus microscope slides (O. Kindler, NP-40, 1% Sodium deoxycholate and 0.1% SDS) with an addition of
Germany). For immunohistochemistry, sections were post-fixed in protease inhibitor cocktail (Roche) followed by a brief sonication on
ice cold acetone for 10 min at 20oC, blocked and incubated with ice. The total protein concentration in each sample was quantified with
primary and secondary antibodies in 5% BSA (Sigma-Aldrich, USA) the Bradford kit (Bio-Rad Laboratories). Protein extracts were analyzed
0.5 % Triton-X in 0.1 M PBS. Slides were mounted using by SDS poly-acrylamide gel electrophoresis and transferred to Hybond-
MOWIOL mounting medium (Calbiochem, Germany). Samples C extra membrane (GE Healthcare Bio-Sciences) for 1 hour using a
were visualized and photographed via confocal microscopy (TCS wet transfer unit (Bio-Rad Laboratories,). Following 1 hour blocking
SP2, Leica Microsystems, Germany). (5% powdered skim milk and 0.1% Tween-20 in 0.1 M PBS); the
The following antibodies were used for immunohistochemistry: membrane was incubated overnight with the primary antibodies. After
rabbit polyclonal antibodies against TAG-1 (1:1000), against Caspr2 washing three times for 10 min in 0.1 % Tween-20 in 0.1 M PBS,
and Caspr (kind gifts by Dr Laurence Goutebroze, Inserm 536, Paris, samples were incubated for 1 h at room temperature with horseradish
both antibodies 1:800); against Kv1.2 (Alomone, APC-010, Jerusalem, peroxidase–coupled secondary antibodies, and proteins were visualized
1:200); against IbaI (Biocare Medical CP290A, USA, 1:500), against by enhanced chemiluminescence (ECL Plus, GE Healthcare Bio-Scien-
Nav1.6 (ASC009, Alomone Labs, Jerusalem, 1:100). Mouse monoclo- ces). The following antibodies were used for Western blot analysis: rab-
nal PanNav (clone K58/35, S8809, Sigma-Aldrich, Germany, 1:50); bit polyclonal antibodies against TAG-1 (1:4000), rabbit polyclonal
Caspr (kind gift by Dr Elior Peles, Weizmann Institute of Science, antibodies against Caspr2 and Caspr (kind gifts by Dr Laurence Gou-
Israel, 1:1000), CC-1 (Merk Millipore OP80, Billerica, MA 01821, tebroze, both antibodies 1:3000), rabbit polyclonal antibody against
1:100); RT97 (Developmental Studies Hybridoma Bank, 1:1,000) and Kv1.2 (Alomone, APC-010, Jerusalem, 1:1000), mouse monoclonal
rat monoclonal antibody against Myelin Basic Protein (MBP - AbD antibody against GAPDH (internal control, Santa Cruz, SC32233,
Serotec, MCA409S, UK, 1:200) and against Proteolipid Protein (PLP- 1:4000) and horseradish peroxidase–coupled secondary antibodies (all
Prof. Reynold’ s lab,1:10). Sections were then incubated with fluoro- from Jackson ImmunoResearch Laboratories, 1:5000-1:10,000). Band
chrome-labeled secondary antibodies Alexa Fluor 488 and 555 (all intensity of comparable animal groups (n ¼ 3 from each group) was
from Molecular Probes, Eugene, OR, 1:800) and counterstained either quantified with Tinascan version 2.07d and normalized with the re-
with 40, 60-diamidino-2-phenylindole (DAPI) (Sigma-Aldrich) or To- spective GAPDH intensity levels as loading control. Expression values
Pro 3 iodide (Life Technologies, 642/661) to visualize cell nuclei. are shown as % percentage considering naive animal values as 100%.
Data are depicted as means 6 standard deviation of the mean and sta-
Quantification of Perinodal Pathology tistical analysis was performed, using nonparametric, one-way ANOVA
For quantification of pathological changes in perinodal molecular with additional Bonferroni post-test for multiple comparisons (P <
architecture in MOG-EAE, longitudinal posterior white matter cryo- 0.05 was considered statistically significant).
sections from cervical and thoracic spinal cord were immunostained
with combinations of antibodies against Caspr, Caspr2, Kv1.2, and Results
TAG-1, to label paranodes and juxtaparanodes, respectively. We
counted the percentage of paranodes with/without stained juxtapar-
Juxtaparanodal Protein Alterations Upon EAE
anodes within a square area of 95  95 lm in images obtained at
Induction
the confocal microscope. We examined five to six different areas
Our analysis of the juxtaparanodal complex alterations in
from each mouse and three different animals per group. In the same EAE stages consists of comparisons between MOG-EAE (14
areas, we calculated the total juxtaparanodal length by adding the days post induction [dpi] and 28dpi), Complete Freund’s ad-
length of each juxtaparanodal segment of the myelinated fiber, in juvant (CFA-14dpi) and naive control animals. In animals
immunostained spinal cord cryosections against Caspr / Caspr2 and treated with CFA, an increased CNS inflammation occurs
Caspr/TAG-1 (Fig. 2A inset image). ImageJ software was used for but without the formation of demyelinating lesions. In our
the calculation of the length, width and area of juxtaparanodes. Peri- MOG-EAE model, the disease peaks at 21 days post immuni-
nodal pathology was also quantified in respective areas of the corpus zation (dpi) and exhibits partial remission at 28dpi. We exam-
callosum of cuprizone treated animals and depicted as a column ined animals at 14dpi, when total demyelination is higher and
graph of the number of clustered domains versus the total number at 28dpi, when inflammation remits and demyelinating lesions
of perinodal areas detected in each group. Data are expressed as
are reduced (partial remyelination). The overall axonal loss in
means 6 standard deviation of the mean or as scatter plots. Statisti-
all MOG-EAE time points was 20% in spinal cord white
cal analysis of measured values between the different animal groups
was performed, using nonparametric, one-way ANOVA with addi-
matter (Markoullis et al., 2012). Details of clinical scores and
tional Bonferroni post-test for multiple comparisons between the quantification of pathology in MOG-EAE are summarized in
animal groups (P < 0.05 was considered statistically significant). Table 1, Supp. Info. Fig. 1 and Supp. Info. Fig. 2.
VGKCs were the first component of the juxtaparanodal
Western Blot Analysis complex we examined via immunohistochemistry on longitudi-
Tissues were collected from freshly sacrificed adult mice and homoge- nal cryosections of posterior spinal cord, with antibodies against
nized directly in ice cold RIPA buffer (10 mM Sodium phosphate pH paranodal Caspr and the VGKC subtype Kv1.2 (Fig. 1A–D,

1238 Volume 61, No. 8


Zoupi et al.: Juxtaparanodal Alterations in CNS-Demyelinating Models

FIGURE 1: Perinodal complex disruption during the course of MOG-EAE. A–D: Paranodal Caspr (red) and juxtaparanodal VGKCs (green)
immunohistochemistry in posterior spinal cord white matter cryosections from naive (A), CFA-14dpi (B), 14dpi (C), and 28dpi (D) MOG
induced EAE animals. E–H: Paranodal Caspr (red) and juxtaparanodal Caspr2 (green) immunohistochemistry in posterior spinal cord
white matter cryosections from naive (E), CFA-14dpi (F), 14dpi (G) and 28dpi (H) MOG-induced EAE animals. I–L: Nodal sodium channel
subunit 1.6 (red) and juxtaparanodal VGKCs (green) immunohistochemistry in posterior spinal cord white matter cryosections from naive
(I), CFA-14dpi (J), 14dpi (K) and 28dpi (L) MOG induced EAE animals. Juxtaparanodal proteins are normally positioned next to paranodal
Caspr and nodal Nav1.6 in control fibers (A, E, insets below merged images correspond to single fibers, asterisks). CFA inflammation
did not result in any apparent alteration of perinodal protein localization but induced the diffusion of juxtaparanodal Caspr2 to the para-
nodal area; 14 days post injection (B, F, J insets). At the peak of MOG-EAE (14dpi), juxtaparanodal protein clustering is severely
reduced, albeit in a lesser extent than that of paranodal Caspr (C, G, insets). Moreover, the nodal density is strongly affected (K, insets).
At a later MOG-EAE stage (28 dpi) the perinodal protein disorganization is still evident, albeit the focal remyelination efforts observed
at this stage (D, H, L, asterisks in merged images represent properly clustered perinodal domains). [Color figure can be viewed in the
online issue, which is available at wileyonlinelibrary.com.]

insets represent separate channels and merged images of single ized next to paranodal Caspr, occupying distinct areas on the
fibers). In naive animals (Fig. 1A, insets, asterisks represent myelinated fiber (insets and asterisks). In CFA-14dpi spinal
properly clustered perinodal domains), as well as in CFA-14dpi cord white matter, inflammatory infiltrates were prominent,
(Fig. 1B, insets), VGKCs are clustered at juxtaparanodes next to although without the presence of demyelinating lesions (Mar-
paranodal Caspr. In contrast, during the peak of demyelination, koullis et al., 2012). We observed the majority of Caspr2
a disrupted paranodal phenotype was evident in MOG-EAE staining properly aggregated at the juxtaparanodal domain
14dpi (Fig. 1C, insets). VGKCs formed reduced overall clusters although occasionally appeared diffused in the Caspr-positive
at the juxtaparanodal area with apparent diffusion of the paranodal region (Fig. 1F, insets). At the same stage we did
remaining VGKC aggregates to the adjacent domains (Fig. 1C, not observe any alterations in Caspr localization (Fig. 1F,
insets). At MOG-EAE 28dpi, extensive VGKC diffusion was insets). In MOG-EAE spinal cord, during the peak of inflam-
still evident (Fig. 1D, insets) in spite of the partial reorganiza- mation and demyelination at 14dpi, the paranodal organiza-
tion of the paranodal domains at this stage. tion was disrupted, with the Caspr signal either absent or
Taking these results into account we asked whether the diffused along the myelinated fiber (Fig. 1G, insets). Juxtapar-
other two molecules of the juxtaparanodal tripartite complex anodal Caspr2 localization was also affected upon MOG-EAE
display similar phenotypes as VGKCs. Under physiological induction. At MOG-EAE 28dpi, when the inflammatory
conditions (Naive, Fig. 1E), juxtaparanodal Caspr2 was local- infiltrates decrease and spontaneous re-myelination may

August 2013 1239


TABLE 1: Clinical and Pathological Features of Animals Examined in the MOG-EAE Study

% white matter covered % of white matter


with inflammatory cells demyelinated axons % axonal loss
(AVG 6 SEM) (AVG 6 SEM) (AVG 6 SEM)

Clinical score*
Animal group (AVG 6 SEM) Cervical Thoracic Cervical Thoracic Cervical Thoracic

Naive (n ¼ 6) 0 0.6 6 0.11 0.64 6 0.02 0 0 0 0


CFA (n ¼ 6) 0 1.63 6 0.31 1.69 6 0.33 0 0 16.97 6 3.3 7.38 6 1.03
EAE 14 dpi (n ¼ 7) 0.5 6 0.28 4.2 6 1.2 5.26 6 1.27 2.83 6 1.45 7.29 6 3.17 17.47 6 6.49 23.17 6 6.33
EAE 28 dpi (n ¼ 7) 1.63 6 0.35 2.44 6 0.89 4.28 6 1.55 1.1 6 0.4 3.88 6 1.91 32.33 6 1.82 20.91 6 3.86
Clinical scores represent average weekly scores.

occur, partial reclustering of paranodal Caspr and juxtapara- and may reflect the percentage of axonal loss of observed in
nodal Caspr2 was evident (Fig. 1H, insets). these stages (Markoullis et al., 2012).
We similarly analyzed the expression of TAG-1, which
is expressed both by axons and myelinating glia in CNS and Juxtaparanodal Protein Diffusion in MOG- EAE
PNS, and plays a key role in the organization and mainte- Stages
nance of the juxtaparanodal complex (Savvaki et al., 2008, Our following aim concerned the evaluation of the immuno-
2010; Traka et al., 2002, 2003). Immunohistochemical analy- histochemical results. More specifically, our interest was
sis with specific antibodies for Caspr and TAG-1 showed jux- mainly focused to the less well analyzed molecules of the jux-
taparanodal localization of the latter in naive myelinated taparanodal complex, Caspr2 and TAG-1. Protein staining
fibers (Supp. Info. Fig. 3A, insets and asterisks). In CFA- distribution at the juxtaparanodal area was quantified by
14dpi mice, the non-specific immune response caused a clear measuring the total length of the stained domain (Caspr2 or
diffusion of TAG-1 protein from the juxtaparanodes to par- TAG-1- positive) in each myelinated fiber, in relation to the
anodes (Supp. Info. Fig. 3B, insets). At MOG-EAE 14dpi respective width (Fig. 2A, inset picture). This measurement
where Caspr staining was disrupted (Supp. Info. Fig. 3C, was performed in all animal groups and presented as separate
insets), TAG-1 juxtaparanodal localization was also reduced scatter plots (Fig. 2A–H). In addition, the average length and
similar to the other two juxtaparanodal proteins. At 28dpi, the total area were also measured and presented as column
juxtaparanodal clustering of TAG-1 was incomplete, since the graphs in Fig. 2I–L.
detected signal remained diffused (Supp. Info. Fig. 3D, Under control conditions, Caspr2-positive areas were
insets), while clustered localization was observed only in a characterized by a specific range of length, width and total
small percentage of fibers (asterisks) or in heminodal-like area values (Fig. 2A,I,K). In CFA-14dpi animals, Caspr2 dif-
structures (Supp. Info. Fig. 3D, insets). fusion towards the paranodes did not alter the overall area
The differences in the clustering between paranodal and length according to the respective scatter plot measurements
juxtaparanodal components prompted us to investigate the (Fig. 2B) while the average length and area (Fig. 2I,K, respec-
effect on nodal clustering between 14dpi and 28dpi MOG- tively), showed a nonsignificant increase compared to naive
EAE stages, in relation to the juxtaparanodal component. To animals. In contrast, MOG-EAE induction resulted in an
this end, we immunostained for the mature form of sodium obvious clustering reduction of Caspr2 protein compared to
channels (Nav1.6), a form mainly affected in demyelinating naive or CFA myelinated fibers (Fig. 1C). Moreover, the
diseases (Craner et al., 2003; Waxman et al., 2004), along remaining clustered juxtaparanodes diffused, as indicated by
with juxtaparanodal VGKCs (Fig. 1I–L, insets). CNS inflam- the significant increase in both the length and the area of
mation in CFA-14dpi did not have any apparent effect in the Caspr2-positive signal (Fig. 2C,I,K, P < 0.05* and P <
nodal density of the white matter areas (Fig. 1J, insets and 0.01**). Finally, despite the apparent focal perinodal re-orga-
asterisks). In contrast, at MOG-EAE 14dpi, there is an nization (Fig. 1D,H), analysis of the diffusion status of
obvious reduction in the nodal density of the white matter Caspr2 at 28dpi points at only minor and non-significant dif-
(Fig. 1K) which is sustained at MOG-EAE 28dpi (Fig. 1L) ferences compared with 14dpi animals (Fig. 2D,I,K).

1240 Volume 61, No. 8


Zoupi et al.: Juxtaparanodal Alterations in CNS-Demyelinating Models

FIGURE 2: Diffusion of juxtaparanodal components at different MOG-EAE stages. Scatter plot analysis of the distribution of Caspr2 (A–
D) and TAG-1 staining (E–H) at juxtaparanodes as the length in relation to the corresponding width of the juxtaparanodal protein1 area
(A: the lines in the inset picture show the measured area). Quantification of the average length and area of Caspr2 and TAG-1 protein
staining is depicted in graphs I and J, respectively. CNS inflammation (CFA-14dpi) transiently disrupted the borders between the
domains leading both Caspr2 and TAG-1 proteins to the paranodal domain, whilst with no significant alteration of the length and area
of the stained segment (B, F, I–K). At MOG-EAE 14dpi, the fiber organization was impaired. Caspr2 and TAG-1 aggregates that were
still detected at this stage were diffused towards the adjacent areas (C, G, I–K). At EAE-28dpi, there is an obvious reorginazation of par-
anodal domains and not of juxtaparanodal proteins as indicated bythe unchanged values in the length and area of Caspr2 and TAG-1
staining, compared to MOG-EAE 14dpi (D, H, I–K)–; P < 0.05 *, P < 0.01 **).

For the TAG-1 protein, a similar measurement was per- EAE Effects on Protein Expression Levels of the
formed (Fig. 2E–H,J,L). In CFA-14dpi, the respective scatter Perinodal Components
plot (Fig. 2F) showed a shift to higher length values indicat- The activation of the immune system and the subsequent de-
ing TAG-1 diffusion but without alterations in the average myelination of the white matter fibers caused altered localization
juxtaparanodal length and occupied area compared with naive of the perinodal proteins. We next asked whether the domain
animals (Fig. 2E,F,J,L). At the peak of demyelination (MOG- disorganization is coupled to alterations of protein expression
EAE 14dpi), TAG-1 diffusion was obvious with a subsequent levels (Fig. 3). Caspr protein levels were insignificantly reduced
increase in the total length and area (Fig. 2G,J,L, P < 0.05) in CFA-14dpi spinal cord, compared to naive animals. In con-
that was not altered at the 28dpi stage (Fig. 2H,J,L). Finally, trast, MOG-EAE 14dpi mice showed significantly reduced lev-
we also observed a subsequent increase of the juxtaparanodal els (P < 0.05), while a robust increase of Caspr expression was
area width in both MOG-EAE stages compared to control found at MOG-EAE 28dpi coinciding with the remission of
animals (Fig. 2A–H). inflammation (Fig. 3, panel A, P < 0.001).

August 2013 1241


FIGURE 3: MOG-EAE effects on protein expression levels of the perinodal components. Western blot analysis of spinal cord lysates
from naive, CFA-14dpi, MOG-EAE 14dpi and 28dpi animals. Band intensity quantification of the percentage of protein expression levels
normalized to the corresponding GAPDH levels A: Paranodal Caspr displayed reduced although not statistically significant protein levels
in both CFA and MOG-EAE 14dpi, compared with naive animals. At 28 dpi the protein levels were significantly increased compared with
all the groups analyzed. B: Western blot analysis for the potassium channel subunit 1.2 (VGKCs) revealed a significant decrease in CFA
and MOG-EAE 14dpi and 28dpi spinal cords. C: Caspr2 protein levels were significantly reduced in both CFA and MOG-EAE 14dpi ani-
mals. At 28dpi Caspr2 levels slightly increased but not significantly, compared to the other animal groups. D: TAG-1 expression levels
significantly decreased in MOG-EAE 14dpi- but not in CFA animals- a reduction that persisted in MOG-EAE 28dpi. (P < 0.05*, P <
0.01**, P < 0.001 ***).

Similar to Caspr, juxtaparanodal VGKC, Caspr2 and 20% at the peak of EAE demyelination (P < 0.01), while
TAG-1 protein levels were significantly reduced in MOG- remaining at similar levels at 28dpi (Fig. 3, panel B). Caspr2
EAE 14dpi samples. However, in contrast to Caspr, there was protein was decreased in both CFA and MOG-EAE 14dpi (P
no alteration of the juxtaparanodal protein levels at MOG- < 0.01) but the subsequent increase at 28dpi was not signifi-
EAE 28dpi spinal cords (Fig. 3, panels B–D). Specifically, cant (Fig. 3, panel C, P > 0.05). Finally, TAG-1 levels were
VGKCs protein levels were strongly affected by the immune also affected, although to a lesser extent, exhibiting a 40%
response initiation and EAE induction, with protein levels decrease at MOG-EAE14dpi (P < 0.05) and an additional
dropping to 40% of the naive levels in CFA (P < 0.01) and 20% reduction at 28dpi (Fig. 3, panel D, P < 0.001).

1242 Volume 61, No. 8


Zoupi et al.: Juxtaparanodal Alterations in CNS-Demyelinating Models

FIGURE 4: Quantification of the perinodal domain clustering at different MOG-EAE stages. A: EAE induction resulted in a decrease of
the perinodal protein clustering at 14dpi, compared with naive and CFA animals that did not recover until 28dpi (P < 0.01 **, P <
0.001***) B: Nodal density measurement of white matter areas in the posterior spinal cord indicated a significant reduction of mature
nodes in both MOG-EAE stages (P < 0.05 *, P < 0.01 **). C: Quantification of heminodal formations between naive, 14dpi and 28dpi
MOG-EAE animals (P < 0.01 **, P < 0.001***). D: Total diagram of paranodal and juxtaparanodal distribution in the EAE stages. E: Peri-
nodal protein redistribution at 14 and 28dpi EAE animals. At 14dpi, paranodes were more susceptible to damage than the juxtaparano-
des (Caspr1 paranodes, light gray). On the contrary, juxtaparanodes were less affected since the three proteins of the juxtaparanodal
tripartite complex displayed higher levels of clustering with no significant differences between them. At 28dpi (dark gray), paranodal
clustering increased while juxtaparanodal staining decreased.

Perinodal Domain Clustering at Different EAE inflammation in CFA-14dpi was not able to alter the white
Stages matter nodal density (Fig. 4B). At MOG-EAE 14dpi, quanti-
So far we have reported alterations in the expression levels, fication of the axonal loss ranged between 15 and 20% com-
localization, length, and area of clustering of the perinodal pared with naive animals (Supp. Info. Fig. 1K). Surprisingly,
proteins, focusing on the juxtaparanodal tripartite complex the number of nodal clusters was reduced approximately by
components. These differences led us to further examine the 50% at this stage and it was significantly reduced compared
differences on protein clustering between paranodes and jux- to naive (P < 0.05) and to CFA-14dpi animals (P < 0.01)
taparanodes, as the disease progresses. (Fig. 4B). These results indicate extensive nodal damage upon
First we wanted to evaluate the extent of damage in the demyelination that is not restored following remission of
perinodal clustering by quantifying the percentage of physiolog- inflammation at 28dpi.
ically clustered perinodal domains (Supp. Info. Fig 2a) in spinal In parallel with the nodal and perinodal clustering during
cord white matter cryosections, at the two MOG-EAE stages the course of MOG-EAE, we have also quantified the amount
compared to control animal areas (Fig. 4A and Supp. Info. Fig. of heminodal structure formation in naive, 14 and 28 dpi
4a). The overall clustering of perinodal proteins was markedly MOG-EAE. Heminodes are structures found at the borders of
reduced during the peak of demyelination (MOG-EAE 14dpi) each myelin segment that fuse to give rise to mature nodes dur-
by 50% compared with naive and CFA-14dpi animals (Fig. 4A, ing the process of myelination (Feinberg et al., 2010). We
P < 0.001, P < 0.01 respectively). Similarly, our quantification found a significant increase in the number of heminodes in spi-
at 28dpi did not show any significant difference when com- nal cord white matter at MOG-EAE 28dpi, indicating an
pared to 14dpi indicating a sustained clustering defect of peri- ongoing remyelination attempt (Fig. 4C, P ¼ 0.004).
nodal components in both MOG-EAE stages (Fig. 4A). The immunohistochemical analysis of spinal cord white
We additionally quantified the number of nodal sodium matter revealed specific phenotypes of perinodal clustering in
channel clusters per microscopic area in CFA-14dpi, MOG- the two MOG-EAE stages (Supp. Info. Fig. 4b and c). In
EAE 14dpi, MOG-EAE 28dpi and naive animals by perform- particular, we encountered paranodes without the presence of
ing multiple comparisons between groups (Fig. 4B). CNS juxtaparanodes and vice versa. Taking into account these

August 2013 1243


clustering defects, we have quantified the ratio of each corpus callosum were evident (Fig. 5B). Moreover, the nodal
abnormality to the total number of detected perinodal density was obviously reduced after 4.5 and 6 weeks of cupri-
domains for each protein of interest (Fig. 4D). At MOG- zone administration and nodal domain elongation of denuded
EAE 14dpi paranodal Caspr was either diffused along myelin- axons was also observed (Fig. 5F,G arrows indicate disrupted
ated fibers or disrupted. In addition, all three juxtaparanodal phenotypes). In this model, we also quantified the overall my-
proteins were affected in terms of their localization. Consider- elinated fiber clustering in the corpus callosum, performed as in
ing that paranodal area elongation is thought to be an early the case of the MOG-EAE model (Fig 4A). During demyelin-
event before demyelination (Coman et al., 2006; Howell ation the number of properly organized fibers was severely
et al., 2006; Howell et al., 2010) we examined separately par- reduced (Fig. 5Q,R). The remaining Caspr localization was
anodal and juxtaparanodal integrity at the peak of demyelin- characterized by disruptions, shrinkage or diffusion along the
ation. Indeed, at MOG-EAE 14dpi, paranodes were more myelinated fiber (Fig. 5F,J,N, arrows), phenotypes that were also
affected than juxtaparanodes (increased numbers of clustered sustained after 6 weeks of cuprizone treatment (Fig. 5G,K,O,
juxtaparanodes with disrupted paranodes), confirming the arrows). By quantifying Caspr-positive paranodal numbers in the
increased paranodal susceptibility to damage, upon demyelin- different groups, we observed a significant reduction of parano-
ation (Fig. 4D, E). At 28dpi, remission of inflammation and des after 4.5 weeks of cuprizone administration, compared to
spontaneous remyelination permitted the re-clustering of the untreated animals (Fig. 5Q). Slightly increased paranodal forma-
paranodal components (increased numbers of clustered paran- tion was observed at 6 weeks of treatment, characterized by
odes with diffused juxtaparanodes), as indicated by the extensive demyelination followed by spontaneous remyelination
increased levels of Caspr protein expression (Fig. 3, panel A) (Fig. 5G,K,O). Further nodal and paranodal recovery occurs at
and clustering (Fig. 4D,E). In contrast, juxtaparanodal pro- 9 weeks with full remyelination (Fig. 5H,L,P,Q).
teins remained diffused, similar to 14dpi (Fig. 4D,E). Juxtaparanodal proteins displayed various alterations in the
cuprizone model similar to the ones observed in the MOG-EAE
Perinodal Protein Clustering is Affected in the model. Caspr2 protein signal was progressively diffused (at 4.5
Cuprizone Model of Toxic Demyelination weeks) and lost (at 6 weeks) (Fig. 5J,K, arrows). A similar pheno-
The results from the MOG-EAE model highlight the perino- type was observed also for TAG-1 protein (data not shown).
dal disorganization during the peak of EAE and at a subse- Finally, VGKCs appeared diffused along the fiber at both treat-
quent stage when the immune T cell infiltrates are decreasing ment stages (Fig. 5N,O, arrows). However, toxin removal fol-
(28dpi). Although reorganization of the paranodal domain lowed by remyelination restored the uniform MBP staining (Fig.
was observed upon decreased inflammation, it was not 5D), increased the presence of mature oligodendrocytes in the
adequate to restore the aggregation of the juxtaparanodal white matter and reduced the occurrence of activated microglia
components. We wished to examine whether juxtaparanodal in the area (Supp. Info. Fig. 5D), in contrast to the EAE situa-
proteins were affected upon demyelination and remyelination tion where activated microglia persisted at 28dpi (Supp. Info.
without the implication of the immune system. For this rea- Fig. 2). In addition, both paranodal and juxtaparanodal proteins
son, we used the cuprizone model of toxic demyelination reclustered, leading to restoration of axonal domains (Fig. 5L,P).
(Matsushima and Morell, 2001). After 4.5 weeks of treatment, the clustering of the
Corpus callosum cryosections were initially immuno- domains was severely reduced compared with untreated ani-
stained for MBP, for the discrimination of well-defined mals and persisted also after 6 weeks (Fig. 5R, P < 0.05 and P
lesions (L) (Fig. 5A–D) followed by immunohistochemical < 0.01, respectively). In contrast, when the toxin was removed
analysis using antibodies against mature oligodendrocytes and remyelination was allowed, axonal domains were fully
(CC-1) and activated microglia (Iba1) for the detection of restored and were indistinguishable from the untreated ani-
innate immunity (Supp. Info. Fig. 5). Antibodies against mals. Thus, although our results indicate a similar pathological
nodal sodium channels (Nav, Fig. 5E–H), paranodal Caspr pattern of domain disruption in both models during demyelin-
and juxtaparanodal proteins Caspr2 (Fig. 5I–L) and VGKCs ation, remyelination in the cuprizone model resulted in resto-
(Fig. 5M–P) were subsequently used. ration of both paranodal and juxtaparanodal areas, while in the
Untreated animals showed uniform MBP staining of the MOG-EAE model juxtaparanodal domain clustering remained
corpus callosum (Fig. 5A), normal numbers of mature oligoden- incomplete even after paranodal reorganization.
drocyte and microglial populations (Supp. Info. Fig. 5A) and
proper myelinated fiber organization with the juxtaparanodal
protein staining to be adjacent to the paranodal Caspr and to Discussion
nodal Nav (Fig. 5A,E,I,M). After 4.5 weeks of cuprizone treat- In this paper we provide a detailed analysis of perinodal pro-
ment MBP staining was disrupted and distinct lesions in the tein alterations in two different models of CNS

1244 Volume 61, No. 8


Zoupi et al.: Juxtaparanodal Alterations in CNS-Demyelinating Models

FIGURE 5: Perinodal protein clustering is affected in the cuprizone model of toxic demyelination. Immunohistochemical analysis of cor-
pus callosum with antibodies against MBP (Myelin Basic Protein- A–D), nodal sodium channels (Nav) and perinodal proteins Caspr,
Caspr2 and VGKCs (E–P). Four different groups of animals were analyzed: untreated controls, animals treated with cuprizone toxin for
4.5 weeks, 6 weeks, and 6 weeks followed by 3 weeks of toxin removal for remyelination to occur (9 weeks). Untreated animals showed
normal MBP staining (A) They also displayed proper clustering of nodal Nav, paranodal Caspr and juxtaparanodal proteins (E, L, M). Af-
ter 4.5 and 6 weeks of Cuprizone treatment MBP staining was significantly reduced, an obvious lesion (L) was formed (B, C). Perinodal
protein clustering was strongly affected in demyelinated areas with reduced nodal density and the appearance of elongated nodes (F,G)
and Caspr staining appearing severely reduced, diffused or completely absent (F,J,N,G,K,O,Q,R, arrows indicate disrupted phenotypes).
In addition, both juxtaparanodal protein clustering was strongly affected exhibiting Caspr2 loss and VGKC diffusion towards the interno-
des (J, N, K, O, R, arrows). Toxin removal led to remyelination (D). Remyelination resulted in the restoration of perinodal protein local-
ization with reformed nodes, paranodes and re-clustered juxtaparanodes (H, L, P, Q). Quantification of the clustering status between
the groups is shown in as the ratio of fibers with clustered domains per total number of perinodal domains (R) while the average number
of paranodes per area is presented in Q (P < 0.05*, P < 0.01 **). [Color figure can be viewed in the online issue, which is available at
wileyonlinelibrary.com.]

demyelination, highlighting for the first time the molecular nodal protein organization is closely associated to myelinated
pathology of the juxtaparanodal domain of the myelinated fiber homeostasis and efficient signal transmission along the
fibers. During the last decade it has become evident that peri- axons. Analysis of different mouse mutants has offered

August 2013 1245


valuable information about the importance of axo-glial inter- transient overlap of paranodal NF155 with VGKCs in CFA-
actions in maintaining perinodal domain integrity. Paranodal 10dpi animals. In addition, VGKC diffusion is evident in
protein mutants (Boyle et al., 2001; Rios et al., 2003; Sher- EAE animals and post-mortem human MS lesions (Coman
man et al., 2005; Tait et al., 2000; Zonta et al., 2008) display et al., 2006; Howell et al., 2010). Our analysis supports and
disrupted paranodal junctions, perinodal protein diffusion extends these studies since in CFA-14dpi animals the overlap
and domain elongation. Disrupted paranodes are also related of Caspr2 or TAG-1 with paranodal Caspr is evident.
to axonal pathology, since elimination of the paranodal bar- Domain border disruption may be induced by the
rier and loss of axo-glial contact led to neurofilament disorga- inflammatory milieu alone as an early and direct event of
nization and swelling of the nodes and paranodes (Griffiths inflammation; this phenomenon may be restored by perinodal
et al., 1998; Mathis et al., 2001; Pillai et al., 2009). In addi- protein recycling and border restoration as time passes. CNS
tion to paranodal mutants, mice lacking either Caspr2 or inflammation is also sufficient to affect juxtaparanodal protein
TAG-1 protein exhibit specific disorganization of the juxta- expression levels, causing a significant reduction of Caspr2
paranodal domain with subsequent diffusion of potassium and VGKC levels. In contrast to the other two members of
channels towards the internode (Poliak et al., 2003; Traka the complex, TAG-1 protein expression was not significantly
et al., 2003). Moreover, Tag-1-/- animals display significant reduced. Since TAG-1 is a key molecule of the juxtaparanodal
hypomyelination of the optic nerve and impaired gait, motor complex, interacting with both Caspr2 and VGKCs, one pos-
co-ordination and memory deficits (Chatzopoulou et al., sibility for this discrepancy may be that it is more stable than
2008; Savvaki et al., 2008). These studies emphasize the its partners and thus less susceptible to degradation upon
necessity of intact axo-glial organization for the establishment mild or temporary changes in the domain architecture. For
of efficient nerve conduction and CNS function. the above mentioned reasons it would be interesting to test
Apart from studies in mutant animals, recent reports perinodal protein expression levels in MS patients, while jux-
show the involvement of perinodal domains in human MS taparanodal protein recycling should be further examined.
pathology (Coman et al., 2006; Derfuss et al., 2009; Howell MOG-EAE induction results in altered distribution and
et al., 2006; Howell et al., 2010; Mathey et al., 2007). More- expression of juxtaparanodal proteins (Fig. 6A). During the
over, specific perinodal proteins (Neurofascin and TAG-1) peak of inflammation/demyelination, a severe reduction of the
were identified as auto-antigens in MS patients (Derfuss total clustering of perinodal domains is observed in spinal cord
et al., 2009; Mathey et al., 2007). Nodal sodium channels white matter. Paranodes are primarily affected consistent with
and NF186 redistribution was evident in active demyelinating their localization on the myelinated fiber that makes them
and chronic lesions in post-mortem MS brain (Craner et al., more susceptible to damage than the less exposed juxtaparano-
2004a,b; Howell et al., 2006). In addition, paranodal Caspr des. However, juxtaparanodes are also disorganized during
(expressed by the axon) and NF155 (expressed by glia) were MOG-EAE. All three components of the juxtaparanodal com-
shown to be affected in demyelinating MS lesions and in plex display reduced protein levels and disrupted clustering
rodent models of MS (EAE). Domain border disruption and (Fig. 6A). Previous studies (Howell et al., 2010) have shown a
subsequent area elongation is now thought to be an early profound diffusion of VGKCs even at 10dpi in MOG-EAE
event before the initiation of demyelination (Coman et al., spinal cords. The observed increase of the length occupied by
2006; Howell et al., 2006, 2010). While nodal and paranodal the signal and the respective area for both Caspr2 and TAG-1
areas have been previously analyzed, less attention has been during MOG-EAE, indicates diffusion of the proteins and
given to the adjacent juxtaparanodal components. This is the elongation of the juxtaparanodal domain. Domain width is also
first study that analyzes all proteins of the juxtaparanodal tri- an important factor as width augmentation has been related to
partite complex in relation to paranodal and nodal compo- axonal pathology that also takes place under demyelinating
nents in two different rodent models of CNS demyelination, conditions (Soulika et al., 2009; Wolswijk and Balesar, 2003).
the EAE and the cuprizone model. Having demonstrated that severe CNS inflammation
Our results showing juxtaparanodal alterations in these and subsequent demyelination leads to disruption of the orga-
two models add new information to the working model of nization of myelinated fibers, we analyzed 28dpi animals,
perinodal domain distribution under demyelinating condi- when inflammation is diminishing. At this stage, paranodal
tions (summarized in Fig. 6). We have shown that CNS Caspr protein levels increase and a marked reclustering of the
inflammation alone (CFA animals) is sufficient to disrupt the respective domain is evident. In addition, increased numbers
barriers between the perinodal domains, resulting in the diffu- of heminodal-like structures are detected. In contrast, juxta-
sion of the juxtaparanodal components into the paranodal paranodal protein levels and localization are similar to 14dpi
area. This phenomenon has been previously reported by (Fig. 6A). Our results confirm the notion of sequential and
Howell and colleagues (Howell et al., 2010), who noticed the progressive disorganization of myelinated fibers under

1246 Volume 61, No. 8


Zoupi et al.: Juxtaparanodal Alterations in CNS-Demyelinating Models

FIGURE 6: Perinodal protein distribution in the two rodent models of CNS demyelination. Schematic representation of the nodal, para-
nodal and juxtaparanodal component distribution in the different MOG-EAE stages (A) and in the cuprizone model of toxic demyelin-
ation (B). A: CFA immunization alone disrupts the boundaries between paranodal and juxtaparanodal domains leading to the diffusion
of the juxtaparanodal proteins into the paranodal area. At MOG-EAE 14dpi, destruction of the myelin sheath causes perinodal domain
disorganization and the diffusion of perinodal proteins and the reduction of the nodal density. At MOG-EAE 28dpi, paranodes begin to
reform without the subsequent reclustering of the juxtaparanodal components. B: After 4.5 weeks of cuprizone treatment there is for-
mation of demyelinating lesions and disruption of perinodal architecture. This phenotype persists after 6 weeks of treatment although
an increase in the number of paranodal clustering is observed. Juxtaparanodal protein re-organization is evident only when extensive
remyelination takes place.

pathological conditions, even though the observed paranodal Adaptive immunity with invasion of activated T lym-
protein aggregation and heminodal formation reflect the phocytes against myelin components in the CNS is a key
ongoing remyelination process. A putative functional signifi- player in the induction and progression of MS pathology,
cance of delayed reorganization of the juxtaparanodes may be while the role of innate immunity is being increasingly appre-
the partial or incomplete level of remyelination that is ciated (Howell et al., 2010). In order to assess the changes of
observed in MOG-EAE 28dpi and possibly in shadow pla- juxtaparanodal organization in the absence of induced inflam-
ques in the human MS brain. Additionally, our MOG-EAE mation, we studied another rodent model of CNS demyelin-
model is characterized by significant axonal loss that persists ation, the cuprizone model of toxic demyelination (Groebe
and even increases at 28dpi, likely contributing to persistently et al., 2009; Kipp et al., 2009; Matsushima and Morell
reduced axonal protein levels despite remyelination. 2001). At demyelinating stages, total perinodal clustering is

August 2013 1247


significantly reduced compared to naive animals. Phenotypi- ber: 2010-14; Grant sponsors: IMBB\-FoRTH Scholarship
cally, Caspr is progressively diffused and disrupted. Moreover and University of Crete, Manasaki Scholarship.
the nodal density is severely reduced and elongated nodal The authors would like to thank Irene Sargiannidou
areas are obvious. Finally, juxtaparanodal Caspr2 is gradually and Natasha Schiza for help with western blots and immuno-
lost while VGKC clustering appears severely diffused towards staining and Dr Richard Reynolds for providing recombinant
both paranodal and internodal regions. However, by the 6th MOG and advice on EAE induction. They also would like to
week of cuprizone treatment, paranodal re-formation is evi- thank Maria Savvaki and Maura Strigini for comments on
dent devoid of juxtaparanodal re-organization, which occurs the manuscript and Dr C. Linington for helpful discussion.
only when efficient remyelination is allowed and innate im-
munity is withdrawn (Fig. 6B). In contrast to MOG-EAE
animals, in the cuprizone model, paranodal regions re-organ- References
ize first, followed by juxtaparanodal ones. Barnett MH, Henderson AP, Prineas JW. 2006. The macrophage in MS: Just
Our findings are relevant for the effects of dalfampridine a scavenger after all? Pathology and pathogenesis of the acute MS lesion.
Mult Scler 12:121–132.
(4-AP), a known potassium channel blocker, in MS. Myelin
Barnett MH, Prineas JW. 2004. Relapsing and remitting multiple sclerosis:
damage may expose VGKCs at juxtaparanodal regions which Pathology of the newly forming lesion. Ann Neurol 55:458–468.
are covered by myelin in healthy axons (Bostock et al., 1981;
Bostock H, Sears TA, Sherratt RM. 1981. The effects of 4-aminopyridine and
Chiu and Ritchie, 1980, 1981; Haghighi et al., 1995; Karimi- tetraethylammonium ions on normal and demyelinated mammalian nerve
Abdolrezaee et al., 2004; Nashmi et al., 2000; Sherratt et al., fibres. J Physiol 313:301–315.

1980). The potassium efflux through fast potassium channels Boyle ME, Berglund EO, Murai KK, Weber L, Peles E, Ranscht B. 2001.
Contactin orchestrates assembly of the septate-like junctions at the paranode
may cause axonal conduction block by preventing sufficient in myelinated peripheral nerve. Neuron 30:385–397.
depolarization and initiation of the action potential at the node Bradl M, Lassmann H. 2010. Oligodendrocytes: Biology and pathology. Acta
of Ranvier. 4-AP has been shown to restore action potential Neuropathol 119:37–53.
conduction in demyelination models (Bostock et al., 1981; Charles P, Tait S, Faivre-Sarrailh C, Barbin G, Gunn-Moore F, Denisenko-
Nehrbass N, Guennoc AM, Girault JA, Brophy PJ, Lubetzki C. 2002.
Leung, 2012). In addition to myelin-dependent alterations of Neurofascin is a glial receptor for the paranodin/Caspr-contactin axonal
potassium channel expression, as described here, myelin-inde- complex at the axoglial junction. Curr Biol 12:217–220.
pendent somatodendritic Kv2.1 are also altered in EAE (Juk- Chatzopoulou E, Miguez A, Savvaki M, Levasseur G, Muzerelle A, Muriel MP,
kola et al., 2012), indicating that pharmacological actions of 4- Goureau O, Watanabe K, Goutebroze L, Gaspar P, et al. 2008. Structural
requirement of TAG-1 for retinal ganglion cell axons and myelin in the
AP may occur at different levels and may be difficult to corre- mouse optic nerve. J Neurosci 28:7624–7636.
late with the sequential changes at the juxtaparanodes alone. Chiu SY, Ritchie JM. 1980. Potassium channels in nodal and internodal axonal
Our study provides a detailed analysis and therefore membrane of mammalian myelinated fibres. Nature 284:170–171.

highlights juxtaparanodal dis-and re-organization under path- Chiu SY, Ritchie JM. 1981. Evidence for the presence of potassium channels
in the paranodal region of acutely demyelinated mammalian single nerve
ological conditions. Demyelination leads to the gradual disor- fibres. J Physiol 313:415–437.
ganization initially of the paranodal area, followed by disrup- Coman I, Aigrot MS, Seilhean D, Reynolds R, Girault JA, Zalc B, Lubetzki C. 2006.
tion of juxtaparanodal and nodal domains. Expression levels Nodal, paranodal and juxtaparanodal axonal proteins during demyelination and
of perinodal proteins are also affected by demyelination. As remyelination in multiple sclerosis. Brain 129(Part 12):3186–3195.

inflammation diminishes, paranodal expression levels increase Craner MJ, Hains BC, Lo AC, Black JA, Waxman SG. 2004a. Co-localization
of sodium channel Nav1.6 and the sodium-calcium exchanger at sites of
and paranodes are reformed in order to restore the disrupted axonal injury in the spinal cord in EAE. Brain 127(Part 2):294–303.
nerve conduction and protect the axon from further damage. Craner MJ, Lo AC, Black JA, Waxman SG. 2003. Abnormal sodium channel
Juxtaparanodal restoration does not occur in parallel with par- distribution in optic nerve axons in a model of inflammatory demyelination.
Brain 126(Part 7):1552–1561.
anodal reorganization. This implies that restoration of the
Craner MJ, Newcombe J, Black JA, Hartle C, Cuzner ML, Waxman SG.
juxtaparanodal complex is either a later event or a process 2004b. Molecular changes in neurons in multiple sclerosis: altered axonal
that remains incomplete possibly due to the persistence of a expression of Nav1.2 and Nav1.6 sodium channels and Naþ/Ca2þ
exchanger. Proc Natl Acad Sci USA 101:8168–8173.
hostile inflammatory environment as the disease progresses.
Derfuss T, Parikh K, Velhin S, Braun M, Mathey E, Krumbholz M, Kumpfel T,
Moldenhauer A, Rader C, Sonderegger P, et al. 2009. Contactin-2/TAG-1-
directed autoimmunity is identified in multiple sclerosis patients and mediates
Acknowledgment gray matter pathology in animals. Proc Natl Acad Sci USA 106:8302–8307.
Grant sponsor: ELA Foundation; Grant number: 2007- Feinberg K, Eshed-Eisenbach Y, Frechter S, Amor V, Salomon D, Sabanay H,
02512C; Grant sponsor: MS UK; Grant number: 847/07; Dupree JL, Grumet M, Brophy PJ, Shrager P, et al. 2010. A glial signal
consisting of gliomedin and NrCAM clusters axonal Naþ channels during the
Grant sponsor: Cyprus Research Promotion Foundation; formation of nodes of Ranvier. Neuron 65:490–502.
Grant numbers: HEALTH/BIOS/0308/01, HEALTH/BIOS/ Franklin RJ, Ffrench-Constant C, Edgar JM, Smith KJ. 2012. Neuroprotection
0609/BIE/09; Grant sponsor: Cyprus Telethon; Grant num- and repair in multiple sclerosis. Nat Rev Neurol 8:624–634.

1248 Volume 61, No. 8


Zoupi et al.: Juxtaparanodal Alterations in CNS-Demyelinating Models
Griffiths I, Klugmann M, Anderson T, Yool D, Thomson C, Schwab MH, Nashmi R, Jones OT, Fehlings MG. 2000. Abnormal axonal physiology is
Schneider A, Zimmermann F, McCulloch M, Nadon N, et al. 1998. Axonal associated with altered expression and distribution of Kv1.1 and Kv1.2 Kþ
swellings and degeneration in mice lacking the major proteolipid of myelin. channels after chronic spinal cord injury. Eur J Neurosci 12:491–506.
Science 280:1610–1613.
Pillai AM, Thaxton C, Pribisko AL, Cheng JG, Dupree JL, Bhat MA. 2009.
Groebe A, Clarner T, Baumgartner W, Dang J, Beyer C, Kipp M. 2009. Spatiotemporal ablation of myelinating glia-specific neurofascin (Nfasc NF155)
Cuprizone treatment induces distinct demyelination, astrocytosis, and microglia in mice reveals gradual loss of paranodal axoglial junctions and concomitant
cell invasion or proliferation in the mouse cerebellum. Cerebellum 8:163–174. disorganization of axonal domains. J Neurosci Res 87:1773–1793.
Haghighi SS, Pugh SL, Perez-Espejo MA, Oro JJ. 1995. Effect of 4- Poliak S, Peles E. 2003. The local differentiation of myelinated axons at
aminopyridine in acute spinal cord injury. Surg Neurol 43:443–447. nodes of Ranvier. Nat Rev Neurosci 4:968–980.
Haider L, Fischer MT, Frischer JM, Bauer J, Hoftberger R, Botond G, Poliak S, Salomon D, Elhanany H, Sabanay H, Kiernan B, Pevny L, Stewart CL,
Esterbauer H, Binder CJ, Witztum JL, Lassmann H. 2011. Oxidative damage Xu X, Chiu SY, Shrager P, et al. 2003. Juxtaparanodal clustering of Shaker-
in multiple sclerosis lesions. Brain 134(Part 7):1914–1924. like Kþ channels in myelinated axons depends on Caspr2 and TAG-1. J Cell
Biol 162:1149–1160.
Howell OW, Palser A, Polito A, Melrose S, Zonta B, Scheiermann C, Vora AJ,
Brophy PJ, Reynolds R. 2006. Disruption of neurofascin localization reveals Price JC, Kelley DE, Ryan CM, Meltzer CC, Drevets WC, Mathis CA, Mazumdar
early changes preceding demyelination and remyelination in multiple S, Reynolds CFIII. 2002. Evidence of increased serotonin-1A receptor binding in
sclerosis. Brain 129(Part 12):3173–3185. type 2 diabetes: A positron emission tomography study. Brain Res 927:97–103.
Howell OW, Rundle JL, Garg A, Komada M, Brophy PJ, Reynolds R. 2010. Prineas JW, Parratt JD. 2012. Oligodendrocytes and the early multiple
Activated microglia mediate axoglial disruption that contributes to axonal sclerosis lesion. Ann Neurol 72:18–31.
injury in multiple sclerosis. J Neuropathol Exp Neurol 69:1017–1033.
Rios JC, Rubin M, St Martin M, Downey RT, Einheber S, Rosenbluth J,
Jukkola PI, Lovett-Racke AE, Zamvil SS, Gu C. 2012. Kþ channel alterations Levinson SR, Bhat M, Salzer JL. 2003. Paranodal interactions regulate
in the progression of experimental autoimmune encephalomyelitis. Neurobiol expression of sodium channel subtypes and provide a diffusion barrier for the
Dis 47:280–293. node of Ranvier. J Neurosci 23:7001–7011.
Karimi-Abdolrezaee S, Eftekharpour E, Fehlings MG. 2004. Temporal and Savvaki M, Panagiotaropoulos T, Stamatakis A, Sargiannidou I, Karatzioula P,
spatial patterns of Kv1.1 and Kv1.2 protein and gene expression in spinal Watanabe K, Stylianopoulou F, Karagogeos D, Kleopa KA. 2008. Impairment
cord white matter after acute and chronic spinal cord injury in rats: of learning and memory in TAG-1 deficient mice associated with shorter CNS
Implications for axonal pathophysiology after neurotrauma. Eur J Neurosci internodes and disrupted juxtaparanodes. Mol Cell Neurosci 39:478–490.
19:577–589.
Savvaki M, Theodorakis K, Zoupi L, Stamatakis A, Tivodar S, Kyriacou K,
Kipp M, Clarner T, Dang J, Copray S, Beyer C. 2009. The cuprizone animal Stylianopoulou F, Karagogeos D. 2010. The expression of TAG-1 in glial cells is
model: New insights into an old story. Acta Neuropathol 118:723–736. sufficient for the formation of the juxtaparanodal complex and the phenotypic
rescue of tag-1 homozygous mutants in the CNS. J Neurosci 30:13943–13954.
Labasque M, Faivre-Sarrailh C. 2010. GPI-anchored proteins at the node of
Ranvier. FEBS Lett 584:1787–1792. Sherman DL, Tait S, Melrose S, Johnson R, Zonta B, Court FA, Macklin WB,
Meek S, Smith AJ, Cottrell DF, et al. 2005. Neurofascins are required to
Lassmann H. 2001. Classification of demyelinating diseases at the interface
establish axonal domains for saltatory conduction. Neuron 48:737–742.
between etiology and pathogenesis. Curr Opin Neurol 14:253–258.
Sherratt RM, Bostock H, Sears TA. 1980. Effects of 4-aminopyridine on
Lassmann H. 2003. Axonal injury in multiple sclerosis. J Neurol Neurosurg
normal and demyelinated mammalian nerve fibres. Nature 283:570–572.
Psychiatry 74:695–697.
Soulika AM, Lee E, McCauley E, Miers L, Bannerman P, Pleasure D. 2009.
Lassmann H. 2008. Models of multiple sclerosis: New insights into
Initiation and progression of axonopathy in experimental autoimmune
pathophysiology and repair. Curr Opin Neurol 21:242–247.
encephalomyelitis. J Neurosci 29:14965–14979.
Lassmann H, Bruck W, Lucchinetti C. 2001. Heterogeneity of multiple
Susuki K, Rasband MN. 2008. Molecular mechanisms of node of Ranvier
sclerosis pathogenesis: Implications for diagnosis and therapy. Trends Mol
formation. Curr Opin Cell Biol 20:616–623.
Med 7:115–121.
Tait S, Gunn-Moore F, Collinson JM, Huang J, Lubetzki C, Pedraza L,
Lassmann H, Lucchinetti CF. 2008. Cortical demyelination in CNS
Sherman DL, Colman DR, Brophy PJ. 2000. An oligodendrocyte cell adhesion
inflammatory demyelinating diseases. Neurology 70:332–333.
molecule at the site of assembly of the paranodal axo-glial junction. J Cell
Leung YM. 2012. Involvement of C-type inactivation gating in the actions of Biol 150:657–666.
voltage-gated Kþ channel inhibitors. Pharmacol Ther 133:151–158.
Traka M, Dupree JL, Popko B, Karagogeos D. 2002. The neuronal adhesion
Lucchinetti CF, Popescu BF, Bunyan RF, Moll NM, Roemer SF, Lassmann protein TAG-1 is expressed by Schwann cells and oligodendrocytes and is
H, Bruck W, Parisi JE, Scheithauer BW, Giannini C, et al. Inflammatory localized to the juxtaparanodal region of myelinated fibers. J Neurosci 22:
cortical demyelination in early multiple sclerosis. N Engl J Med 2011;365: 3016–3024.
2188–2197.
Traka M, Goutebroze L, Denisenko N, Bessa M, Nifli A, Havaki S, Iwakura Y,
Markoullis K, Sargiannidou I, Gardner C, Hadjisavvas A, Reynolds R, Kleopa Fukamauchi F, Watanabe K, Soliven B, et al. 2003. Association of TAG-1 with
KA. 2012. Disruption of oligodendrocyte gap junctions in experimental Caspr2 is essential for the molecular organization of juxtaparanodal regions
autoimmune encephalomyelitis. Glia 60:1053–1066. of myelinated fibers. J Cell Biol 162:1161–1172.
Mathey EK, Derfuss T, Storch MK, Williams KR, Hales K, Woolley DR, Al-Hayani Waxman SG, Craner MJ, Black JA. 2004. Naþ channel expression along
A, Davies SN, Rasband MN, Olsson T, et al. 2007. Neurofascin as a novel target axons in multiple sclerosis and its models. Trends Pharmacol Sci 25:584–591.
for autoantibody-mediated axonal injury. J Exp Med 204:2363–2372.
Wolswijk G, Balesar R. 2003. Changes in the expression and localization of
Mathis C, Denisenko-Nehrbass N, Girault JA, Borrelli E. 2001. Essential role the paranodal protein Caspr on axons in chronic multiple sclerosis. Brain
of oligodendrocytes in the formation and maintenance of central nervous 126(Part 7):1638–1649.
system nodal regions. Development 128:4881–4890.
Zonta B, Tait S, Melrose S, Anderson H, Harroch S, Higginson J, Sherman
Matsushima GK, Morell P. 2001. The neurotoxicant, cuprizone, as a model to DL, Brophy PJ. 2008. Glial and neuronal isoforms of Neurofascin have
study demyelination and remyelination in the central nervous system. Brain distinct roles in the assembly of nodes of Ranvier in the central nervous
Pathol 11:107–116. system. J Cell Biol 181:1169–1177.

August 2013 1249

You might also like