ArticlePDF Available

Distribution of the Extracellular Matrix in the Pararubral Area of the Rat

Authors:

Abstract and Figures

Previously we described similarities and differences in the organization and molecular composition of an aggrecan based extracellular matrix (ECM) in three precerebellar nuclei, the inferior olive, the prepositus hypoglossi nucleus and the red nucleus of the rat associated with their specific cytoarchitecture, connection and function in the vestibular system. The aim of present study is to map the ECM pattern in a mesencephalic precerebellar nucleus, the pararubral area, which has a unique function among the precerebellar nuclei with its retinal connection and involvement in the circadian rhythm regulation. Using histochemistry and immunohistochemistry we have described for the first time the presence of major ECM components, the hyaluronan, aggrecan, versican, neurocan, brevican, tenascin-R (TN-R), and the HAPLN1 link protein in the pararubral area. The most common form of the aggrecan based ECM was the diffuse network in the neuropil, but each type of the condensed forms was also recognizable. Characteristic perineuronal nets (PNNs) were only recognizable with Wisteria floribunda agglutinin (WFA) and aggrecan staining around some of the medium-sized neurons, whereas the small cells were rarely surrounded by a weakly stained PNNs. The moderate expression of key molecules of PNN, the hyaluronan (HA) and HAPLN1 suggests that the lesser stability of ECM assembly around the pararubral neurons may allow quicker response to the modified neuronal activity and contributes to the high level of plasticity in the vestibular system.
Content may be subject to copyright.
Distribution of the Extracellular Matrix in the Pararubral Area of the Rat
Do
´ra Szarvas,
a
Botond Gaa
´l,
a
Clara Matesz
a,b
and E
´va Ra
´cz
a,c
*
a
Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., Debrecen H-4032, Hungary
b
Division of Oral Anatomy, Faculty of Dentistry, University of Debrecen, Nagyerdei krt. 98., Debrecen H-4032, Hungary
c
MTA-DE Neuroscience Research Group, Nagyerdei krt. 98., Debrecen 4032, Hungary
Abstract—
Previously we described similarities and differences in the organization and molecular composition of
an aggrecan based extracellular matrix (ECM) in three precerebellar nuclei, the inferior olive, the prepositus hypo-
glossi nucleus and the red nucleus of the rat associated with their specific cytoarchitecture, connection and func-
tion in the vestibular system. The aim of present study is to map the ECM pattern in a mesencephalic precerebellar
nucleus, the pararubral area, which has a unique function among the precerebellar nuclei with its retinal connec-
tion and involvement in the circadian rhythm regulation. Using histochemistry and immunohistochemistry we
have described for the first time the presence of major ECM components, the hyaluronan, aggrecan, versican,
neurocan, brevican, tenascin-R (TN-R), and the HAPLN1 link protein in the pararubral area. The most common
form of the aggrecan based ECM was the diffuse network in the neuropil, but each type of the condensed forms
was also recognizable. Characteristic perineuronal nets (PNNs) were only recognizable with Wisteria floribunda
agglutinin (WFA) and aggrecan staining around some of the medium-sized neurons, whereas the small cells were
rarely surrounded by a weakly stained PNNs. The moderate expression of key molecules of PNN, the hyaluronan
(HA) and HAPLN1 suggests that the lesser stability of ECM assembly around the pararubral neurons may allow
quicker response to the modified neuronal activity and contributes to the high level of plasticity in the vestibular
system. Ó2018 IBRO. Published by Elsevier Ltd. All rights reserved.
Key words: perineuronal net, hyaluronan, chondroitin sulfate proteoglycans, tenascin-R, link protein, vestibular system.
INTRODUCTION
The intercellular space of central nervous system (CNS)
is filled with the extracellular matrix (ECM). The most
frequently occurring ECM molecules in the CNS are the
hyaluronan (HA), lecticans (aggrecan, brevican,
neurocan and versican), tenascin-R (TN-R) and the
HAPLN1 link protein. Although the majority of these
molecules appear as a diffuse network in the neuropil,
the condensed forms may also present as they
surround the neuronal cell body, dendrites and axon
initial segment as the perineuronal net (PNN), or form
the axonal coat around the presynaptic bouton, or
associated with the node of Ranvier as nodal ECM
(Celio et al., 1998; Carulli et al., 2006; Bruckner et al.,
2008; Bekku et al., 2009; Bekku and Oohashi, 2010;
Dityatev, 2010; Frischknecht and Seidenbecher, 2012;
Lendvai et al., 2012; Blosa et al., 2013). The ECM shows
an area-dependent distribution pattern, and its molecular
and structural heterogeneity is correlated with the mor-
phological and functional properties of the neurons
(Matesz et al., 2005; Szigeti et al., 2006; Meszar et al.,
2008; Morawski et al., 2009; Gati et al., 2010; Lendvai
et al., 2012; Morawski et al., 2012; Jager et al., 2013;
Gaal et al., 2014; Gaati et al., 2014; Racz et al., 2014,
2015b; Kecskes et al., 2015). The ECM molecules are
involved in the synaptic transmission as they are the
fourth components of synaptic machinery besides the
presynaptic and postsynaptic neurons as well as the
astroglia cell (Dityatev and Schachner, 2006; Dityatev
et al., 2006; Faissner et al., 2010; Dityatev and
Rusakov, 2011; Chelini et al., 2018). In addition, the
ECM stabilizes the synaptic connections, creates a
https://doi.org/10.1016/j.neuroscience.2018.10.027
0306-4522/Ó2018 IBRO. Published by Elsevier Ltd. All rights reserved.
*Correspondence to: E
´.Ra
´cz, Department of Anatomy, Histology and
Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei
krt. 98., Debrecen H-4032, Hungary. Fax: +36-52-255-115.
E-mail address: eva@anat.med.unideb.hu (E
´.Ra
´cz).
Abbreviations: AC, axonal coat; bHABP, biotinylated Hyaluronan
Binding Protein; bWFA, biotinylated Wisteria floribunda agglutinin;
BSA, bovine serum albumin; CNS, central nervous system; Cr,
cerebellum; CSPG, chondroitin sulfate proteoglycan; DAPI, 4’,6-
Diamidino-2-Phenylindole; ECM, extracellular matrix; GABA, gamma-
aminobutyric acid; GAD, glutamic acid decarboxylase; HA, hyaluronan;
HAPLN1, hyaluronan and proteoglycan link protein 1; IC, inferior
colliculus; LCN, lateral cerebellar nucleus; MAP2, microtubule-
associated protein 2; ml, medial lemniscus; MCN, medial cerebellar
nucleus; NeuN, neuronal nuclei; NDS, normal donkey serum; NGS,
normal goat serum; NRS, normal rabbit serum; PaR, pararubral
nucleus; PBS, phosphate buffered saline; PNN, perineuronal net;
pRN, parvicellular part of red nucleus; R, red nucleus; RT, room
temperature; SC, superior colliculus; scp, superior cerebellar peduncle;
TN-R, tenascin-R; WFA, Wisteria floribunda agglutinin; IV, fourth
ventricle.
NEUROSCIENCE
RESEARCH ARTICLE
D. Szarvas et al. / Neuroscience 394 (2018) 177–188
177
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
barrier against the formation of new synaptic contacts,
thereby it restricts the synaptic plasticity of adult CNS.
The molecular composition of ECM is changing parallel
to the neuronal activity in healthy brain and its modifica-
tion was described in different pathophysiological events
(Bradbury et al., 2002; Pizzorusso et al., 2002; Dityatev
and Schachner, 2003; Busch and Silver, 2007; Dityatev
and Fellin, 2008; Kwok et al., 2008; Morita et al., 2010;
Dityatev and Rusakov, 2011; Kwok et al., 2011;
Morawski et al., 2012, 2014; Sorg et al., 2016; Suttkus
et al., 2016; Bozzelli et al., 2018; Chelini et al., 2018;
Ferrer-Ferrer and Dityatev, 2018). In line with these
results it was observed that the lesion of vestibular recep-
tors and subsequent compensatory events resulted in
modification of ECM assembly in the vestibular nuclei of
brainstem suggesting the possible role of ECM in the
vestibular plasticity (Matesz et al., 2005; Deak et al.,
2012; Gaal et al., 2015a; Faralli et al., 2016). Since these
nuclei have widespread reciprocal connections with the
cerebellum, spinal cord and precerebellar nuclei, similar
alteration of ECM is expected in these structures. In order
to detect these possible changes, data on the ECM com-
position are needed in these areas. Previously we have
mapped the distribution and organization of an
aggrecan-based ECM in three precerebellar nuclei of
the vestibular neural circuits, the inferior olive (Kecskes
et al., 2015), the prepositus hypoglossi nucleus (Gaal
et al., 2015b), and the red nucleus (Racz et al., 2016)in
the rat. Besides the similarities we detected a number of
differences between the individual nuclei and their subnu-
clei associated with their specific functions in the vestibu-
lar system. These data are not yet available for the
mesencephalic pararubral area. The pararubral area is
located in the mesencephalic reticular formation, dorsolat-
eral to the red nucleus at the level of its caudal part
(Paxinos and Watson, 1998)(Fig. 1G). The GABA and
parvalbumin-positive small- and medium-sized pararubral
cells are excited by the sensorimotor cortex and terminate
on the rubrospinal neurons thereby they are involved in
the flexor motor execution (Liu et al., 2002). Although
the pararubral area is similar to the adjacent parvicellular
part of the red nucleus regarding the morphology, neuro-
chemistry, physiology, and connections, it has a unique
feature among the members of precerebellar nuclei as it
receives direct projection from the retina and involved in
the circadian rhythm regulation (Cooper et al., 1990;
Morcuende et al., 2002; Horowitz et al., 2004).
In this study we investigated the distribution and
organization of the major types of ECM molecules in the
pararubral nucleus of the rat. Using histochemical and
immunohistochemical methods, we have detected all
major ECM components in the pararubral area and the
staining pattern showed similarity, with some
differences, to the parvocellular part of the red nucleus.
EXPERIMENTAL PROCEDURES
Preparation of the rat brain for the histochemical and
immunohistochemical reactions
The protocol of the experiments was accepted by the
Animal Care Committee of the University of Debrecen,
Debrecen, Hungary and the national laws and EU
regulations (license number: 6/2017/DEMAB).
The study was performed on adult female (12–14-
week old) Wistar rats (n= 5) from Charles River
Laboratory (Strain Crl:WI), weighing from 250 to 300 g.
The animals were deeply anesthetized with
intraperitoneal injection of 10% urethane (1.3 ml/100 g
body weight; Reanal, Budapest, Hungary) and
transcardially perfused with physiological saline. The
brainstem was removed and immersed into Sainte-
Marie’s fixative (99% absolute ethanol and 1% glacial
acetic acid) for one day at 4 °C. The mesencephalon
was embedded in paraffin and sagittal sections were cut
with microtome at a thickness of 8 lm. The sections
were collected on silane-coated slides and left to dry
overnight at 37 °C. Following deparaffination, they were
rehydrated and washed in phosphate-buffered saline,
pH 7.4 (PBS) and treated with 3% H
2
O
2
dissolved in
bidistilled water for 10 min at room temperature (RT).
Histochemistry and immunohistochemistry
Detection of ECM molecules. Prior to histochemical
and immunohistochemical reactions, specimens were
blocked for 60 min at RT in 3% bovine serum albumin
(BSA) (for HA; Wisteria floribunda agglutinin, WFA;
versican), 3% BSA + 10% normal goat serum (NGS)
(for aggrecan), 3% BSA + 10% normal rabbit serum
(NRS) (for chondroitin sulfate proteoglycan, Clone Cat-
301; neurocan), 3% BSA + 10% normal donkey serum
(NDS) (for brevican, TN-R, HAPLN1). Reagents above
were dissolved in PBS.
Histochemical reactions. The distribution of HA was
detected by using biotinylated Hyaluronan Binding
Protein (bHABP; AMS Biotechnology, Abingdon, UK).
WFA histochemistry was carried out using biotinylated
Wisteria floribunda agglutinin (bWFA; Sigma-Aldrich, St.
Louis, MO, USA), a lectin that binds to N-
acetylgalactosamine residues of CSPG-
glycosaminoglycan chains and glycoproteins, as a
marker of PNNs (Hartig et al., 1992). bHABP and bWFA
were dissolved in PBS containing 1% BSA, sections were
incubated overnight at 4 °C. Visualization of labeling was
performed by incubating the samples with Streptavidin
AlexaFluor 555 (Life Technologies, Carlsbad, CA, USA)
for 1 h, diluted in 1:1000, in PBS.
Immunohistochemical reactions. Slides were
incubated in the following primary antibodies: rabbit
polyclonal anti-aggrecan (Merck Millipore, Billerica, MA,
USA), mouse monoclonal anti-chondroitin sulfate
proteoglycan (Clone Cat-301, Sigma-Aldrich), mouse
monoclonal anti-versican (12C5; Developmental Studies
Hybridoma Bank, DSHB, Iowa City, IA, USA), mouse
monoclonal anti-neurocan (1F6; DSHB), sheep
polyclonal anti-brevican (R&D Systems, Minneapolis,
MN, USA), goat polyclonal anti-TN-R (R&D Systems),
goat polyclonal anti-HAPLN1 (R&D Systems). For better
antigen exposure of aggrecan, Cat-301, versican and
brevican molecules, sections were digested with
178 D. Szarvas et al. / Neuroscience 394 (2018) 177–188
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
Fig. 1. (A, B) Distribution of hyaluronan reaction (red) in the pararubral nucleus. NeuN reaction (green) detects the neurons. Yellow arrow in (B)
shows thin, faintly stained perineuronal net. (C–F) Distribution of Wisteria floribunda agglutinin (WFA) reaction (red) in the pararubral nucleus. NeuN
reaction (green) in (C–F) detects the neurons. MAP2 immunostaining (green) in (F) labels dendrites (white arrowheads). Schematic drawing in (G)
point to the position of the pararubral nucleus in the mesencephalon. SC: superior colliculus; IC: inferior colliculus; Cr: cerebellum; IV: fourth
ventricle; scp: superior cerebellar peduncle; R: red nucleus; PaR: pararubral nucleus; ml: medial lemniscus. (For interpretation of the referencesto
color in this figure legend, the reader is referred to the web version of this article.)
D. Szarvas et al. / Neuroscience 394 (2018) 177–188 179
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
chondroitinase ABC (0.02 U/ml; Sigma-Aldrich) in specific
TRIS sodium-acetate buffer, pH 8 for 1 h at 37 °C.
Primary antibodies were diluted in PBS and 1% BSA
+ 3% NGS (aggrecan), 1% BSA (versican), 1% BSA
+ 3% NRS (Cat-301, neurocan), 1% BSA + 3% NDS
(brevican, TN-R, HAPLN1), overnight at 4 °C. The
incubation was followed by repeated rinsing in PBS.
More details of primary reagents are reported in Table 1.
The following secondary antibodies were used: goat-
anti-rabbit IgG AlexaFluor 555 (Life Technologies)
(aggrecan), rabbit-anti-mouse IgG AlexaFluor 555 (Life
Technologies) (Cat-301, versican, neurocan), donkey-
anti-sheep IgG AlexaFluor 555 (Life Technologies)
(brevican), and donkey-anti-goat IgG AlexaFluor 555
(Life Technologies) (TN-R, HAPLN1), for 1 h, all diluted
in 1:1000, in PBS.
Semiquantitative assessment of histochemical and
immunohistochemical reactions. After performing all
ECM reactions, slides containing the identical sagittal
sectional levels were selected from three animals.
Staining intensity of ECM reactions was evaluated on the
images recorded with the microscope using the same
settings for each section. Applying the same computer
screen we used a five-grade scaling (: no staining, +:
weak staining, ++: moderate staining, +++: strong
staining, ++++: very strong staining). The subjective
grading was performed independently by two authors (E
´.
R., B.G.) and checked by the others (C.M., D.Sz.).
Presence of axonal coats was evaluated accordingly:
: no ACs’ or ‘+: presence of ACs’ (Table 3).
Double fluorescent labeling. Double fluorescent
labeling was made using neuronal nuclei (NeuN),
neurofilament or microtubule-associated protein 2
(MAP2) antibodies in combination with specific ECM
markers. The following fluorescent labelings were
combined: rabbit polyclonal anti-NeuN (Merck Millipore)
+ bHABP, bWFA, versican, brevican, neurocan, TN-R
or HAPLN1; mouse monoclonal anti-NeuN (Merck
Millipore) + aggrecan; rabbit polyclonal anti-
neurofilament (Sigma-Aldrich) + versican; rabbit
polyclonal anti-MAP2 (Merck Millipore) + bWFA, Cat-
301, brevican or HAPLN1 (Table 2).
Prior to the incubation with NeuN, neurofilament or
MAP2 primary antibodies, specimens were blocked for
60 min at RT in 3% BSA + 10% NDS (for rabbit anti-
NeuN, rabbit anti-neurofilament, rabbit anti-MAP2), in
3% BSA + 10% NGS (for mouse anti-NeuN). Primary
antibodies were diluted in PBS with 1% BSA + 3%
NGS (for mouse anti-NeuN) and 1% BSA + 3% NDS
(for rabbit anti-NeuN, rabbit anti-neurofilament, rabbit
anti-MAP2). Visualization of reactions was by goat anti-
mouse IgG AlexaFluor 488 (mouse anti-NeuN; Life
Technologies) or donkey anti-rabbit IgG AlexaFluor 488
(rabbit anti-NeuN, rabbit anti-neurofilament, rabbit anti-
MAP2; Life Technologies).
Slides were coverslipped with ProLongÒDiamond
Antifade Mountant with DAPI (Life Technologies).
Images were recorded using Olympus CX31
epifluorescent light microscope with DP74 digital
Table 1. Probe, lectin and primary antibodies used for detection of ECM molecules
bHABP
a
bWFA
b
Anti-aggrecan CAT-301
c
Anti-
versican
Anti-
neurocan
Anti-brevican Anti-tenascin-R HAPLN1
d
Supplier,
cat. No.
AMS
Biotechnology;
AMS.HKD-BC41
Sigma-
Aldrich;
L1516
Merck Millipore;
AB1031
Sigma-
Aldrich;
MAB5284
DSHB; 12C5 DSHB; 1F6 R&D Systems;
AF4009
R&D Systems; AF3865 R&D Systems;
AF2608
Species of
origin,
type
Recombinant
human versican
G1 domain
expressed in
E. coli; biotinylated
Lectin
isolated from
Wisteria
floribunda;
biotinylated
Rabbit; polyclonal,
IgG
Mouse;
Monoclonal,
IgG1
Mouse;
monoclonal,
IgG1
Mouse;
monoclonal,
IgG1
Sheep; polyclonal,
IgG
Goat; polyclonal, IgG Goat; polyclonal, IgG
Immunogen GST fusion protein
containing amino
acids 1177–1326
of mouse aggrecan
Feline spinal
cord fixed
gray matter
Hyaluronate-
binding
region of
human
versican
PBS-soluble
CSPGs from
rat brain
Mouse myeloma cell
line NS0 derived
recombinant human
Brevican Asp23
Pro911
mouse myloma cell line
NS0-derived
recombinant human
tenascin-R isoform 1,
Glu34-Phe1358
mouse myeloma cell
line NS0-derived
recombinant human
HAPLN1, Asp16-
Asn354
Dilution 1:100 1:500 1:500 1:100 1:100 1:100 1:100 1:300 1:300
a
Biotinylated Hyaluronan Binding Protein.
b
Biotinylated Wisteria floribunda agglutinin.
c
Anti-chondroitin sulfate proteoglycan Clone CAT-301.
d
Hyaluronan and Proteoglycan Link Protein 1.
180 D. Szarvas et al. / Neuroscience 394 (2018) 177–188
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
camera and processed by Photoshop CS4 v11.0 (Adobe
Systems Inc., San Jose, CA, USA), with minimal
adjustments of contrast and background.
RESULTS
All the ECM reactions studied were positive in the
pararubral area. The staining was detected
predominantly in the neuropil, whereas the presence of
characteristic forms of condensed ECM was variable
with the different reactions.
The HA appeared mostly in diffuse staining in the
neuropil, showing weak intensity. The PNN was
sporadically observed, the other forms of condensed
ECM were not recognizable (Fig. 1A, B). The WFA
staining revealed that some neurons, mostly medium
sized, were surrounded by PNN. WFA-positive PNNs
were rarely shown around the small-sized neurons
(Fig. 1C–F). In case of PNN bearing neurons, the
labeling was also detected around the dendrites as
shown with the combination of WFA and MAP2
reactions (Fig. 1F). The small, ring-like forms of the
condensed ECM, the axonal coats, were also
recognizable. The staining intensity of the neuropil was
the strongest with the WFA among the ECM reactions
studied. The aggrecan reaction was similar to the WFA
staining, regarding the condensed forms of ECM. On
the basis of morphology, both the WFA and aggrecan
reactions revealed the three types of PNNs (Wegner
et al., 2003; Jager et al., 2013). One part of neurons
was surrounded by 1–2 mm clearly contoured PNNs
described as classical or robust type (Fig. 2F), the other
parts were enwrapped by a thin, faintly stained sheath
of ECM (Fig. 2E). The third type of PNNs were not clearly
contoured, the thickness of the lattice-like structure was
about 3–5 mm(Fig. 2C, D). When the aggrecan reaction
labeled the PNN, the labeling was followed into the peri-
dendritic area (Fig. 2B). The staining of neuropil was less
intense than with WFA reaction. Using the versican anti-
body resulted in the characteristic dot like appearance
of versican (Bekku et al., 2009; Bekku and Oohashi,
2010; Racz et al., 2014; Gaal et al., 2015b; Kecskes
et al., 2015). A partial overlap was seen with the combina-
tion of versican and neurofilament reactions indicating the
presence of versican-positive dots around the nodes of
Ranvier (Fig. 2I). Other forms of ECM organization were
not recognizable with versican staining (Fig. 2G–I). The
brevican staining did not reveal PNNs, whereas the other
forms of condensed ECM, the dot like and small, ring like
structures were frequently recognizable indicating the
presence of this molecule at the nodes of Ranvier and
axonal coats. Labelings were shown around the den-
drites, as demonstrated by the MAP2 staining (Fig. 3B,
C). Among the dot and ring like forms of ECM, a weak dif-
fuse staining was observed in the neuropil with brevican
antibody (Fig. 3A–E). Similarly to brevican reaction, the
neurocan labeling indicated its absence in PNNs. In con-
trast, the neuropil was more intensely labeled, whereas
the structures representing the nodal ECM and axonal
coats were rarely observed (Fig. 3F, G). The overall inten-
sity of the tenascin-R immunostaining was very weak,
none of the characteristic forms of ECM were detected
in the lightly stained neuropil (Fig. 4A, B). The HAPLN1
reaction showed patch-like, irregular appearance in the
perisomatic and peridendritic area, however the charac-
teristic form of PNN was rarely identified (Fig. 4D, E).
Axonal coats were also occasionally visible. In the neu-
ropil, darker and lighter HAPLN1-positive irregular areas
were recognizable without showing any regional distribu-
tion in the pararubral area (Fig. 4C–H).
DISCUSSION
Using histochemistry and immunohistochemistry, we
have described for the first time the presence of major
ECM components, the hyaluronan, aggrecan, versican,
Table 2. Primary antibodies used for double labeling with ECM molecules
Anti-NeuN
a
Anti-NeuN
a
Anti-neurofilament Anti-MAP2
b
Supplier, cat. No. Merck Millipore;
ABN78
Merck Millipore;
MAB377
Sigma-Aldrich;
N4142
Merck Millipore;
AB5622
Species of origin,
type
Rabbit;
polyclonal, IgG
Mouse;
monoclonal, IgG
Rabbit;
polyclonal, IgG
Rabbit;
polyclonal, IgG
Immunogen GST-tagged recombinant protein
corresponding to mouse NeuN
Purified cell nuclei
from mouse brain
Purified neurofilament 200
from bovine spinal cord
Purified Microtubule-
associated protein from rat
brain
Dilution 1:1000 1:100 1:80 1:500
a
Neuronal nuclei.
b
Microtubule-associated protein 2.
Table 3. Semiquantitative assessment of the staining intensity of
perineuronal nets (PNN) and neuropil, and presence of axonal coats
(ACs) in the pararubral area of the mesencephalon
PNN Neuropil ACs
Hyaluronan + ++
WFA +++ ++++ +
Aggrecan ++++ +++
*
+
Versican #
Brevican +++ +
Neurocan +# +
Tenascin-R +
HAPLN1 +/++
*
+
The staining intensity of perineuronal net and neuropil was scored as :no
staining, +: weak staining, ++: moderate staining, +++: strong staining, ++
++: very strong staining. #: Presence of heavily stained dots in the less intense
neuropil.
ACs: +: presence of ACs; : no ACs.
*
Regional differences in the intensity of reaction.
D. Szarvas et al. / Neuroscience 394 (2018) 177–188 181
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
neurocan, brevican, TN-R, and the HAPLN1 link protein in
the mesencephalic precerebellar nucleus, the pararubral
area. The most common form of the aggrecan based
ECM was the diffuse network in the neuropil, but each
type of the condensed forms was also recognizable.
The presence of the individual ECM molecules and the
staining intensity of reactions were different in these
compartments of ECM.
Fig. 2. (A–F) Distribution of aggrecan immunoreactivity (red) in the pararubral nucleus. NeuN reaction (green) in (A) and (C–F) detects the neurons.
MAP2 immunostaining (green) in (B) labels dendrites (white arrowheads). Purple arrow in (F) shows robust type of perineuronal net (PNN). The
white arrows in (C, D) indicate lattice-like PNNs, the yellow arrow in (E) shows faintly stained PNN, respectively. (G–I) Distribution of versican
immunoreactivity in the pararubral nucleus. NeuN reaction (green) in (G, H) detects the neurons. Neurofilament immunoreactivity (green) in (I)
represents the axons. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)
182 D. Szarvas et al. / Neuroscience 394 (2018) 177–188
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
Expression pattern of ECM molecules in the
pararubral area
Characteristic PNNs were only recognizable with WFA
and aggrecan staining around some of the medium-
sized neurons, whereas the small cells were rarely
surrounded by a weakly stained PNN. Besides the WFA
and aggrecan-positive PNNs, the HA and HAPLN1
antibody showed occasionally patch-like staining around
Fig. 3. (A–E) Distribution of brevican immunoreactivity in the pararubral nucleus. NeuN reaction (green) in (A, D and E) detects the neurons. MAP2
immunostaining (green) in (B, C) labels dendrites (white arrowheads). Yellow arrowheads in D show axonal coats. (F-G) Distribution of neurocan
immunoreactivity in the pararubral nucleus. NeuN reaction (green) detects the neurons. (For interpretation of the references to color in this figure
legend, the reader is referred to the web version of this article.)
D. Szarvas et al. / Neuroscience 394 (2018) 177–188 183
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
the neuronal cell bodies, but no one characteristic form of
PNN was identifiable. These findings indicate the
moderate expression of key molecules of PNN, the HA
and HAPLN1 (Carulli et al., 2010; Kwok et al., 2011), in
the pararubral area. One of the important functions of
the PNN is to restrict the neural plasticity and instead sup-
port the synaptic stability (Happel and Frischknecht,
2016; Bozzelli et al., 2018). The reduced PNN has been
linked to improved cognitive flexibility (Morellini et al.,
2010) and the lack of link protein Crtl1 results in persistent
plasticity in the visual system (Carulli et al., 2010).
Consistent with these results we assume that the lesser
stability of ECM assembly around the pararubral PNNs
may allow quicker response to the modified neuronal
Fig. 4. (A, B) Distribution of TN-R immunoreactivity in the pararubral nucleus. NeuN reaction (green) detects the neurons. (C–H) Distribution of
HAPLN1 immunoreactivity in the pararubral nucleus. NeuN reaction in (C, F–H) (green) detects the neurons. MAP2 immunostaining (green) in (D,
E) labels dendrites (white arrowheads). Yellow arrowheads in (E, H) show axonal coats. (For interpretation of the references to color in this figure
legend, the reader is referred to the web version of this article.)
184 D. Szarvas et al. / Neuroscience 394 (2018) 177–188
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
activity and contributes to the high level of plasticity in the
vestibular system.
The presence and molecular composition of PNNs is
highly variable in different regions of CNS, as well as
species and age differences have also been reported
(Wang and Fawcett, 2012; Sorg et al., 2016; Bozzelli
et al., 2018). PNNs were most frequently recognizable
around fast-spiking, parvalbumin-positive GABA-ergic
interneurons in different parts of the brain which are asso-
ciated with low plasticity (Hartig et al., 1992; Bruckner
et al., 1999; Schuppel et al., 2002; Dityatev et al., 2007;
Kwok et al., 2011). Although the neurons of the pararubral
area were also positive for the parvalbumin and GAD (Liu
et al., 2002) and physiological studies also suggested
their GABAergic nature of the pararubral neurons, the
PNNs are only present around some of the pararubral
neurons. This finding is similar to the neuron specific
expression pattern of PNNs in the vestibular nuclei and
related structures in the rat brainstem (Racz et al.,
2014; Gaal et al., 2015a; Racz et al., 2016) and in the
parts of the spinal cord related to the motor function
(Jager et al., 2013). Here, the PNNs are dominant around
the large, excitatory projection neurons, whereas the
majority of small, inhibitory interneurons were not sur-
rounded by ECM. The rare occurrence of PNN around
the inhibitory interneurons seems to be a common feature
of CNS structures which are engaged in the motor coordi-
nation and execution. Accordingly, the majority of small,
inhibitory interneurons are not surrounded by ECM in
the vestibular nuclei and related structures in the rat brain-
stem (Racz et al., 2014; Gaal et al., 2015a; Racz et al.,
2016) and in the parts of the spinal cord related to the
motor function (Jager et al., 2013). On the other hand,
the large, excitatory projection neurons are surrounded
by the PNN in these areas in contrast to the weak or miss-
ing PNNs around the majority of pyramidal cells in the
secondary motor cortex (Alpa
´r et al., 2006). On the basis
of these findings it is tempting to speculate that this
unique organization of the PNN is related to the motor
function of CNS, which needs a continuous balance con-
trol during the body displacement.
The axonal coats stained with aggrecan, brevican and
HAPLN1 antibodies as it was shown in other parts of the
CNS including the vestibular areas of brainstem
(Bruckner et al., 2008; Frischknecht and Seidenbecher,
2012; Lendvai et al., 2012; Blosa et al., 2013; Jager
et al., 2013; Lendvai et al., 2013; Morawski et al., 2014;
Racz et al., 2014; Gaal et al., 2015a; Racz et al., 2016).
The nodal ECM was observed most frequently with the
versican and brevican reactions indicating the presence
of essential molecules of node of Ranvier in the pararu-
bral area (Bekku et al., 2009; Bekku and Oohashi, 2010).
The neuropil was stained with all ECM reactions
studied. The most intense staining was revealed with the
WFA reaction, whereas it was faintly labeled with TN-R.
Comparison between the organization of ECM in the
pararubral area and parvicellular part of red nucleus
(pRN)
Using histochemical and immunohistochemical reactions,
all the ECM molecules studied were present both in the
pararubral area and in the previously studied pRN (Racz
et al., 2016). The expression pattern of ECM was mostly
similar with some differences in the two areas.
Contrary to what its name suggest, the pRN contains
large-sized neurons besides the dominant small- and
medium-sized cells. Since the large neurons are not
present in the pararubral area, in the following part of
discussion the comparison refers to the small- and
medium-sized neurons. Characteristic PNNs were
observed with WFA and aggrecan stainings in both
areas, whereas the HA, brevican, neurocan, TN-R, and
HAPLN1 reactions showed pericellular positivity only
around the small- and medium-sized cells in the pRN
(Racz et al., 2016). The similarity in the ECM staining pat-
tern may be related to their common afferent connections
from the somatosensory cortex and cerebellum and effer-
ent projection to the rubrospinal neurons. The neurons of
sensorimotor cortex and cerebellum excite the GABAer-
gic neurons of pararubral area and pRN (Toyama et al.,
1968; Brown, 1974; Gwyn and Flumerfelt, 1974;
Bernays et al., 1988; Oka, 1988; Billard et al., 1991;
Ruigrok and de Zeeuw, 1993) which inhibit the reticu-
lospinal neurons in the magnocellular part of the red
nucleus. In case of the cerebellum, significant overlap
with some differences were revealed in the origin of pro-
jection fibers by the application of Phaseolus vulgaris leu-
coagglutinin or biotinylated dextran amine into small
areas cerebellar nuclei in the rat (Tenue et al., 2000).
Thus, the pararubral area received fibers only from the
medial cerebellar nucleus (MCN) and all subdivisions of
the lateral cerebellar nucleus (LCN), whereas the labeled
terminals detected in the pRN arrived from all cerebellar
nuclei including their subdivisions. In addition, the density
of labeled cerebellar fibers and terminals were different in
the pararubral area and pRN. Accordingly, the projection
of MCN was stronger in the pararubral area compared
to the density of terminals in the pRN. In case of LCN,
stronger projection was observed from its caudal part to
the pararubral area, whereas the pRN was more abun-
dantly supplied by the rostral part of the LCN. Although
the experiments revealed significant overlap in the termi-
nation areas, the functional differences of the cerebellar
nuclei indicate that the pararubral area is involved mostly
in the visuomotor and vestibular activity due to its stronger
connection with the MCN. The visuomotor function of the
pararubral area was suggested by its polysynaptic projec-
tions to orbicularis oculi muscle (Morcuende et al., 2002).
Moreover, the pararubral nucleus receives substantial
projection form the retina (Cooper et al., 1990) and by
its indirect connection with the suprachiasmatic nucleus
it plays a role in the circadian rhythm regulation
(Horowitz et al., 2004). To the best of our knowledge, nei-
ther the visuomotor of the pRN nor its participation in the
regulation of circadian rhythm was reported.
CONCLUSIONS
Similarly to our previous findings on the mesencephalic
vestibular structure, the parvicellular part of the red
nucleus, we have demonstrated the presence of
aggrecan based ECM in the adjacent, functionally
D. Szarvas et al. / Neuroscience 394 (2018) 177–188 185
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
related pararubral area. In spite of their common neuronal
architecture, we have found some differences in the ECM
expression pattern related probably to the unique
connection and function of the pararubral area in the
visuomotor system. Our findings support the generally
accepted view that the structural and molecular
organization of the ECM shows regional variation in the
CNS.
ACKNOWLEDGMENT
The authors thank Ms. Timea Horvath for skillful technical
assistance. Grant Sponsors from Hungarian Scientific
Research Fund (OTKA K115471), Hungarian Academy
of Sciences Fund (MTA-TKI 11008).
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
ROLE OF AUTHORS
All authors had full access to all the data in the study and
take responsibility for the integrity of the data and the
accuracy of the data analysis.
Study concept and design: E.R., D.Sz.
Acquisition of data: D.Sz; B.G., E.R.
Analysis and interpretation of data: D.Sz, B.G., E.R.,
K.M.
Drafting of the manuscript: E.R.
Critical revision of the manuscript for important
intellectual content: K.M., B.G.
Statistical analysis: -
Obtained funding: E.R., K.M.
Administrative, technical, and material support: E.R.,
B.G., K.M.
Study supervision: E.R.
REFERENCES
Alpa
´rA,Ga
¨rtner U, Ha
¨rtig W, Bru
¨ckner G (2006) Distribution of
pyramidal cells associated with perineuronal nets in the neocortex
of rat. Brain Res 1120:13–22.
Bekku Y, Oohashi T (2010) Neurocan contributes to the molecular
heterogeneity of the perinodal ECM. Arch Histol Cytol 73
(2):95–102.
Bekku Y, Rauch U, Ninomiya Y, Oohashi T (2009) Brevican distinctively
assembles extracellular components at the large diameter nodes of
Ranvier in the CNS. J Neurochem 108(5):1266–1276. https://doi.
org/10.1111/j.1471-4159.2009.05873.x.
Bernays RL, Heeb L, Cuenod M, Streit P (1988) Afferents to the rat
red nucleus studied by means of
D
-[3H]aspartate, [3H]choline and
non-selective tracers. Neuroscience 26(2):601–619.
Billard JM, Daniel H, Pumain R (1991) Sensitivity of rubrospinal
neurons to excitatory amino acids in the rat red nucleus in vivo.
Neurosci Lett 134(1):49–52.
Blosa M, Sonntag M, Bruckner G, Jager C, Seeger G, Matthews RT,
Rubsamen R, Arendt T, Morawski M (2013) Unique features of
extracellular matrix in the mouse medial nucleus of trapezoid
body–implications for physiological functions. Neuroscience
228:215–234. https://doi.org/10.1016/j.
neuroscience.2012.10.003.
Bozzelli PL, Alaiyed S, Kim E, Villapol S, Conant K (2018) Proteolytic
remodeling of perineuronal nets: effects on synaptic plasticity and
neuronal population dynamics. Neural Plasticity. https://doi.org/
10.1155/2018/5735789. 5735789.
Bradbury EJ, Moon LD, Popat RJ, King VR, Bennett GS, Patel PN,
Fawcett JW, McMahon SB (2002) Chondroitinase ABC promotes
functional recovery after spinal cord injury. Nature 416
(6881):636–640. https://doi.org/10.1038/416636a. 416636a [pii].
Brown LT (1974) Rubrospinal projections in the rat. J Comp Neurol
154(2):169–187. https://doi.org/10.1002/cne.901540205.
Bruckner G, Hausen D, Hartig W, Drlicek M, Arendt T, Brauer K
(1999) Cortical areas abundant in extracellular matrix chondroitin
sulphate proteoglycans are less affected by cytoskeletal changes
in Alzheimer’s disease. Neuroscience 92(3):791–805.
Bruckner G, Morawski M, Arendt T (2008) Aggrecan-based
extracellular matrix is an integral part of the human basal
ganglia circuit. Neuroscience 151(2):489–504. https://doi.org/
10.1016/j.neuroscience.2007.10.033.
Busch SA, Silver J (2007) The role of extracellular matrix in CNS
regeneration. Curr Opin Neurobiol 17(1):120–127. https://doi.org/
10.1016/j.conb.2006.09.004.
Carulli D, Pizzorusso T, Kwok JC, Putignano E, Poli A, Forostyak S,
Andrews MR, Deepa SS, Glant TT, Fawcett JW (2010) Animals
lacking link protein have attenuated perineuronal nets and
persistent plasticity. Brain 133(Pt 8):2331–2347. https://doi.org/
10.1093/brain/awq145.
Carulli D, Rhodes KE, Brown DJ, Bonnert TP, Pollack SJ, Oliver K,
Strata P, Fawcett JW (2006) Composition of perineuronal nets in
the adult rat cerebellum and the cellular origin of their
components. J Comp Neurol 494(4):559–577. https://doi.org/
10.1002/cne.20822.
Celio MR, Spreafico R, De Biasi S, Vitellaro-Zuccarello L (1998)
Perineuronal nets: past and present. Trends Neurosci 21
(12):510–515. doi: S0166-2236(98)01298-3 [pii].
Chelini G, Pantazopoulos H, Durning P, Berretta S (2018) The
tetrapartite synapse: a key concept in the pathophysiology of
schizophrenia. European Psychiatry 50:60–69. https://doi.org/
10.1016/j.eurpsy.2018.02.003.
Cooper EJ, Millar TJ, Anderton PJ (1990) Distribution of [H-3] Mk-801
binding in the turtle retina an autoradiographical study. Neurosci
Lett 112(2–3):127–132. https://doi.org/10.1016/0304-3940(90)
90190-K.
Deak A, Bacskai T, Gaal B, Racz E, Matesz K (2012) Effect of
unilateral labyrinthectomy on the molecular composition of
perineuronal nets in the lateral vestibular nucleus of the rat.
Neurosci Lett 513(1):1–5. https://doi.org/10.1016/j.
neulet.2012.01.076.
Dityatev A (2010) Remodeling of extracellular matrix and
epileptogenesis. Epilepsia 51(Suppl 3):61–65. https://doi.org/
10.1111/j.1528-1167.2010.02612.x.
Dityatev A, Bruckner G, Dityateva G, Grosche J, Kleene R,
Schachner M (2007) Activity-dependent formation and functions
of chondroitin sulfate-rich extracellular matrix of perineuronal
nets. Dev Neurobiol 67(5):570–588. https://doi.org/10.1002/
dneu.20361.
Dityatev A, Fellin T (2008) Extracellular matrix in plasticity and
epileptogenesis. Neuron Glia Biol 4(3):235–247. https://doi.org/
10.1017/S1740925X09000118.
Dityatev A, Frischknecht R, Seidenbecher CI (2006) Extracellular
matrix and synaptic functions. Results Probl Cell Differ 43:69–97.
Dityatev A, Rusakov DA (2011) Molecular signals of plasticity at the
tetrapartite synapse. Curr Opin Neurobiol 21(2):353–359. https://
doi.org/10.1016/j.conb.2010.12.006.
Dityatev A, Schachner M (2003) Extracellular matrix molecules and
synaptic plasticity. Nat Rev Neurosci 4(6):456–468. https://doi.
org/10.1038/nrn1115 nrn1115.
Dityatev A, Schachner M (2006) The extracellular matrix and
synapses. Cell Tissue Res 326(2):647–654. https://doi.org/
10.1007/s00441-006-0217-1.
Faissner A, Pyka M, Geissler M, Sobik T, Frischknecht R,
Gundelfinger ED, Seidenbecher C (2010) Contributions of
astrocytes to synapse formation and maturation - Potential
functions of the perisynaptic extracellular matrix. Brain Res Rev
63(1–2):26–38. https://doi.org/10.1016/j.
brainresrev.2010.01.001.
186 D. Szarvas et al. / Neuroscience 394 (2018) 177–188
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
Faralli A, Dagna F, Albera A, Bekku Y, Oohashi T, Albera R, Rossi F,
Carulli D (2016) Modifications of perineuronal nets and
remodelling of excitatory and inhibitory afferents during
vestibular compensation in the adult mouse. Brain Struct Funct
221(6):3193–3209. https://doi.org/10.1007/s00429-015-1095-7.
Ferrer-Ferrer M, Dityatev A (2018) Shaping Synapses by the Neural
Extracellular Matrix. Front Neuroanat 12. https://doi.org/10.3389/
Fnana.2018.00040.
Frischknecht R, Seidenbecher CI (2012) Brevican: a key
proteoglycan in the perisynaptic extracellular matrix of the brain.
Int J Biochem Cell Biol 44(7):1051–1054. https://doi.org/10.1016/
j.biocel.2012.03.022.
Gaal B, Johannesson EO, Dattani A, Magyar A, Weber I, Matesz C
(2015a) Modification of tenascin-R expression following unilateral
labyrinthectomy in rats indicates its possible role in neural
plasticity of the vestibular neural circuit. Neural Regener Res 10
(9):1463–1470. https://doi.org/10.4103/1673-5374.165517.
Gaal B, Kecskes S, Matesz C, Birinyi A, Hunyadi A, Racz E (2015b)
Molecular composition and expression pattern of the extracellular
matrix in a mossy fiber-generating precerebellar nucleus of rat,
the prepositus hypoglossi. Neurosci Lett 594:122–126. https://doi.
org/10.1016/j.neulet.2015.03.056.
Gaal B, Racz E, Juhasz T, Hollo K, Matesz C (2014) Distribution of
Extracellular Matrix Macromolecules in the Vestibular Nuclei and
Cerebellum of the Frog, Rana Esculenta. Neuroscience
258:162–173. https://doi.org/10.1016/j.
neuroscience.2013.10.080.
Gaati G, Lendvai D, Hokfelt T, Harkany T, Alpar A (2014) Revival of
Calcium-Binding Proteins for Neuromorphology: Secretagogin
Typifies Distinct Cell Populations in the Avian Brain. Brain
Behav Evolut 83(2):82–92. https://doi.org/10.1159/000357834.
Gati G, Morawski M, Lendvai D, Matthews RT, Jager C, Zachar G,
Arendt T, Alpar A (2010) Chondroitin sulphate proteoglycan-
based perineuronal net establishment is largely activity-
independent in chick visual system. J Chem Neuroanat 40
(3):243–247. https://doi.org/10.1016/j.jchemneu.2010.07.002.
Gwyn DG, Flumerfelt BA (1974) A comparison of the distribution of
cortical and cerebellar afferents in the red nucleus of the rat. Brain
Res 69(1):130–135.
Hartig W, Brauer K, Bruckner G (1992) Wisteria floribunda agglutinin-
labelled nets surround parvalbumin-containing neurons.
NeuroReport 3(10):869–872.
Happel MF, Frischknecht R (2016) Neuronal plasticity in the juvenile
and adult brain regulated by extracellular matrix. In: Travascio F,
editor. Composition and function of the extracellular matrix in the
human body. Rijeka, Croatia: INTECH.
Horowitz SS, Blanchard JH, Morin LP (2004) Intergeniculate leaflet
and ventral lateral geniculate nucleus afferent connections: an
anatomical substrate for functional input from the vestibulo-
visuomotor system. J Comp Neurol 474(2):227–245. https://doi.
org/10.1002/cne.20125.
Jager C, Lendvai D, Seeger G, Bruckner G, Matthews RT, Arendt T,
Alpar A, Morawski M (2013) Perineuronal and perisynaptic
extracellular matrix in the human spinal cord. Neuroscience
238:168–184. https://doi.org/10.1016/j.
neuroscience.2013.02.014.
Kecskes S, Gaal B, Racz E, Birinyi A, Hunyadi A, Matesz C (2015)
Extracellular matrix molecules exhibit unique expression pattern
in the climbing fiber-generating precerebellar nucleus, the inferior
olive. Neuroscience 284:412–421. https://doi.org/10.1016/j.
neuroscience.2014.09.080.
Kwok JC, Afshari F, Garcia-Alias G, Fawcett JW (2008)
Proteoglycans in the central nervous system: plasticity,
regeneration and their stimulation with chondroitinase ABC.
Restor Neurol Neurosci 26(2–3):131–145.
Kwok JCF, Dick G, Wang DF, Fawcett JW (2011) Extracellular matrix
and perineuronal nets in CNS repair. Developmental
Neurobiology 71(11):1073–1089. https://doi.org/10.1002/
Dneu.20974.
Lendvai D, Morawski M, Bruckner G, Negyessy L, Baksa G, Glasz T,
Patonay L, Matthews RT, Arendt T, Alpar A (2012) Perisynaptic
aggrecan-based extracellular matrix coats in the human lateral
geniculate body devoid of perineuronal nets. J Neurosci Res 90
(2):376–387. https://doi.org/10.1002/jnr.22761.
Lendvai D, Morawski M, Negyessy L, Gati G, Jager C, Baksa G,
Glasz T, Attems J, Tanila H, Arendt T, Harkany T, Alpar A (2013)
Neurochemical mapping of the human hippocampus reveals
perisynaptic matrix around functional synapses in Alzheimer’s
disease. Acta Neuropathol 125(2):215–229. https://doi.org/
10.1007/s00401-012-1042-0.
Liu CL, Wang YJ, Chen JR, Tseng GF (2002) Parvalbumin-containing
neurons mediate the feedforward inhibition of rat rubrospinal
neurons. Anat Embryol (Berl) 205(3):245–254. https://doi.org/
10.1007/s00429-002-0250-0.
Matesz C, Modis L, Halasi G, Szigeti ZM, Felszeghy S, Bacskai T,
Szekely G (2005) Extracellular matrix molecules and their
possible roles in the regeneration of frog nervous system. Brain
Res Bull 66(4–6):526–531. https://doi.org/10.1016/j.
brainresbull.2005.06.014. S0361-9230(05)00245-5 [pii].
Meszar Z, Felszeghy S, Veress G, Matesz K, Szekely G, Modis L
(2008) Hyaluronan accumulates around differentiating neurons in
spinal cord of chicken embryos. Brain Res Bull 75(2–4):414–418.
https://doi.org/10.1016/j.brainresbull.2007.10.052. S0361-9230
(07)00336-X [pii].
Morawski M, Bruckner G, Jager C, Seeger G, Matthews RT, Arendt T
(2012) Involvement of perineuronal and perisynaptic extracellular
matrix in Alzheimer’s disease neuropathology. Brain Pathol 22
(4):547–561. https://doi.org/10.1111/j.1750-3639.2011.00557.x.
Morawski M, Dityatev A, Hartlage-Rubsamen M, Blosa M, Holzer M,
Flach K, Pavlica S, Dityateva G, Grosche J, Bruckner G,
Schachner M (2014) Tenascin-R promotes assembly of the
extracellular matrix of perineuronal nets via clustering of
aggrecan. Philos Trans R Soc Lond B Biol Sci 369
(1654):20140046. https://doi.org/10.1098/rstb.2014.0046.
Morawski M, Reinert T, Meyer-Klaucke W, Wagner FE, Troger W,
Bruckner G, Fiedler A, Butz T, Arendt T (2009) Aggrecan-based
extracellular matrix provides cationic binding. J Neurochem
110:72–73.
Morcuende S, Delgado-Garcia JM, Ugolini G (2002) Neuronal
premotor networks involved in eyelid responses: retrograde
transneuronal tracing with rabies virus from the orbicularis oculi
muscle in the rat. J Neurosci 22(20):8808–8818.
Morellini F, Sivukhina E, Stoenica L, Oulianova E, Bukalo O,
Jakovcevski I, Dityatev A, Irintchev A, Schachner M (2010)
Improved reversal learning and working memory and enhanced
reactivity to novelty in mice with enhanced GABAergic innervation
in the dentate gyrus. Cereb Cortex 20(11):2712–2727.
Morita S, Oohira A, Miyata S (2010) Activity-dependent remodeling of
chondroitin sulfate proteoglycans extracellular matrix in the
hypothalamo-neurohypophysial system. Neuroscience 166
(4):1068–1082. https://doi.org/10.1016/j.
neuroscience.2010.01.041.
Oka H (1988) Functional organization of the parvocellular red nucleus
in the cat. Behav Brain Res 28(1–2):233–240.
Paxinos G, Watson Ch (1998) The rat brain In stereotaxic
coordinates. San Diego, California, USA: Academic Press.
Pizzorusso T, Medini P, Berardi N, Chierzi S, Fawcett JW, Maffei L
(2002) Reactivation of ocular dominance plasticity in the adult
visual cortex. Science 298(5596):1248–1251. https://doi.org/
10.1126/science.1072699 298/5596/1248.
Racz E, Gaal B, Kecskes S, Matesz C (2014) Molecular composition
of extracellular matrix in the vestibular nuclei of the rat. Brain
Struct Funct 219(4):1385–1403. https://doi.org/10.1007/s00429-
013-0575-x.
Racz E, Gaal B, Matesz C (2016) Heterogeneous expression of
extracellular matrix molecules in the red nucleus of the rat.
Neuroscience 322:1–17. https://doi.org/10.1016/j.
neuroscience.2016.02.005.
Ruigrok TJ, de Zeeuw CI (1993) Electron microscopy of in vivo
recorded and intracellularly injected inferior olivary neurons and
their GABAergic innervation in the cat. Microsci Res Technol 24
(1):85–102. https://doi.org/10.1002/jemt.1070240108.
D. Szarvas et al. / Neuroscience 394 (2018) 177–188 187
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
Schuppel K, Brauer K, Hartig W, Grosche J, Earley B, Leonard
BE, Bruckner G (2002) Perineuronal nets of extracellular
matrix around hippocampal interneurons resist destruction by
activated microglia in trimethyltin-treated rats. Brain Res 958
(2):448–453.
Sorg BA, Berretta S, Blacktop JM, Fawcett JW, Kitagawa H, Kwok
JC, Miquel M (2016) Casting a wide net: role of perineuronal nets
in neural plasticity. J Neurosci 36(45):11459–11468. https://doi.
org/10.1523/JNEUROSCI.2351-16.2016.
Suttkus A, Morawski M, Arendt T (2016) Protective properties of
neural extracellular matrix. Mol Neurobiol 53(1):73–82. https://doi.
org/10.1007/s12035-014-8990-4.
Szigeti ZM, Matesz C, Szekely G, Felszeghy S, Bacskai T, Halasi G,
Meszar Z, Modis L (2006) Distribution of hyaluronan in the central
nervous system of the frog. J Comp Neurol 496(6):819–831.
https://doi.org/10.1002/cne.20960.
Toyama K, Tsukahara N, Udo M (1968) Nature of the cerebellar
influences upon the red nucleus neurones. Exp Brain Res 4
(4):292–309.
Wang D, Fawcett J (2012) The perineuronal net and the control of
CNS plasticity. Cell Tissue Res 349(1):147–160. https://doi.org/
10.1007/s00441-012-1375-y.
Wegner F, Hartig W, Bringmann A, Grosche J, Wohlfarth K,
Zuschratter W, Bruckner G (2003) Diffuse perineuronal nets and
modified pyramidal cells immunoreactive for glutamate and the
GABA(A) receptor alpha1 subunit form a unique entity in rat
cerebral cortex. Exp Neurol 184(2):705–714. https://doi.org/
10.1016/S0014-4886(03)00313-3 S0014488603003133.
(Received 11 July 2018, Accepted 15 October 2018)
(Available online 24 October 2018)
188 D. Szarvas et al. / Neuroscience 394 (2018) 177–188
Downloaded for Anonymous User (n/a) at HUNGARY - Debrecen University from ClinicalKey.com by Elsevier on March 05, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
... 34 HAPLN1 was also reported to be associated with the ECM function in the neuron system. 35 Considering the close linkage of ECM mineralization to bone loss and the aforementioned results, we determined the specific effects of ASPN and HAPLN1 on the osteogenic differentiation of BMSCs and ECM mineralization of OBs. OPN is a secreted phosphoprotein involved in the progression of bone diseases. ...
Article
Full-text available
Objective: Bone marrow mesenchymal stromal cells (BMSCs) are major sources of osteogenic precursor cells in bone remodeling, which directly participate in osteoporosis (OP) progression. However, the involved specific mechanisms of BMSCs in OP warrant mass investigations. Initially, our bioinformatics analysis uncovered the prominent up-regulation of Asporin (ASPN) and proteoglycan link protein 1 (HAPLN1) in osteoblasts (OBs) of OP patients and their possible protein interaction. Hence, this study aimed to explore the effects of ASPN and HAPLN1 on osteogenic differentiation of BMSCs, extracellular matrix (ECM) mineralization of OBs, and osteoclastogenesis, hoping to offer research basis for OP treatment. Methods: GSE156508 dataset was used for analysis and screening to acquire the differentially expressed genes in OBs of OP patients, followed by the predicative analysis via STRING. OP mouse models were induced by ovariectomy (OVX), and ASPN and HAPLN1 expression was determined. BMSCs and bone marrow macrophages (BMMs) were isolated from OVX mice and induced for osteogenic differentiation and osteoclastogenesis, respectively. After knockdown experiments, we assessed adipogenic differentiation and osteogenic differentiation in BMSCs. Osteogenic (OPN, OCN, and COL1A1) and osteoclast (Nfatc1 and c-Fos) marker protein expression was determined. The binding of ASPN to HAPLN1 was analyzed. Results: High expression of ASPN and HAPLN1 and their protein interaction were observed in OBs of OP patients via bioinformatics and in bone tissues of OVX mice. ASPN interacted with HAPLN1 in BMSCs of OVX mice. ASPN/HAPLN1 knockdown increased ALP, OPN, OCN, and COL1A1 protein expression and ECM mineralization in BMSCs while decreasing Nfatc1 and c-Fos expression in BMMs. These effects were aggravated by the simultaneous knockdown of ASPN and HAPLN1. Conclusion: Our results indicate that ASPN synergises with HAPLN1 to suppress the osteogenic differentiation of BMSCs and ECM mineralization of OBs and promote the osteoclastogenesis in OP.
... HAPLN1, discovered 50 years ago, has a wide range of physiological effects with an important contribution to cartilage formation and homeostasis as well as to the regulation of the development of the central nervous system (41). Besides HAPLN1, the HAPLN family includes paralogs of HAPLN2, HAPLN3, and HAPLN4, all of which related pathways are Phospholipase-C Pathway and Integrin Pathway. ...
Article
Full-text available
HAPLN1 maintains aggregation and the binding activity of extracellular matrix (ECM) molecules (such as hyaluronic acid and proteoglycan) to stabilize the macromolecular structure of the ECM. An increase in HAPLN1 expression is observed in a few types of musculoskeletal diseases including rheumatoid arthritis (RA); however, its functions are obscure. This study examined the role of HAPLN1 in determining the viability, proliferation, mobility, and pro-inflammatory phenotype of RA- fibroblast-like synoviocytes (RA-FLSs) by using small interfering RNA (siHAPLN1), over-expression vector (HAPLN1OE), and a recombinant HAPLN1 (rHAPLN1) protein. HAPLN1 was found to promote proliferation but inhibit RA-FLS migration. Metformin, an AMPK activator, was previously found by us to be able to inhibit FLS activation but promote HAPLN1 secretion. In this study, we confirmed the up-regulation of HAPLN1 in RA patients, and found the positive relationship between HAPLN1 expression and the AMPK level. Treatment with either si-HAPLN1 or HAPLN1OE down-regulated the expression of AMPK-ɑ gene, although up-regulation of the level of p-AMPK-ɑ was observed in RA-FLSs. si-HAPLN1 down-regulated the expression of proinflammatory factors like TNF-ɑ, MMPs, and IL-6, while HAPLN1OE up-regulated their levels. qPCR assay indicated that the levels of TGF-β, ACAN, fibronectin, collagen II, and Ki-67 were down-regulated upon si-HAPLN1 treatment, while HAPLN1OE treatment led to up-regulation of ACAN and Ki-67 and down-regulation of cyclin-D1. Proteomics of si-HAPLN1, rHAPLN1, and mRNA-Seq analysis of rHAPLN1 confirmed the functions of HAPLN1 in the activation of inflammation, proliferation, cell adhesion, and strengthening of ECM functions. Our results for the first time demonstrate the function of HAPLN1 in promoting the proliferation and pro-inflammatory phenotype of RA-FLSs, thereby contributing to RA pathogenesis. Future in-depth studies are required for better understanding the role of HAPLN1 in RA.
Article
Full-text available
Fine control of extraocular muscle fibers derives from two subpopulations of cholinergic motoneurons in the oculomotor-, trochlear- and abducens nuclei. Singly- (SIF) and multiply innervated muscle fibers (MIF) are supplied by the SIF- and MIF motoneurons, respectively, representing different physiological properties and afferentation. SIF motoneurons, as seen in earlier studies, are coated with chondroitin sulfate proteoglycan rich perineuronal nets (PNN), whereas MIF motoneurons lack those. Fine distribution of individual lecticans in the composition of PNNs and adjacent neuropil, as well as the pace of their postnatal accumulation is, however, still unknown. Therefore, the present study aims, by using double immunofluorescent identification and subsequent morphometry, to describe local deposition of lecticans in the perineuronal nets and neuropil of the three eye movement nuclei. In each nucleus PNNs were consequently positive only with WFA and aggrecan reactions, suggesting the dominating role of aggrecan is PNN establishment. Brevican, neurocan and versican however, did not accumulate at all in PNNs but were evenly and moderately present throughout the neuropils. The proportion of PNN bearing motoneurons appeared 76% in oculomotor-, 72.2% in trochlear- and 78.3% in the abducens nucleus. We also identified two morphological subsets of PNNs, the focal and diffuse nets of SIF motoneurons. The process of CSPG accumulation begins just after birth, although considerable PNNs occur at week 1 age around less than half of the motoneurons, which ratio doubles until 2-month age. These findings may be related to the postnatal establishment of the oculokinetic network, performing different repertoires of voluntary eye movements in functionally afoveolate and foveolate animals.
Preprint
Full-text available
Background To investigate HAPLN1 contribution to the viability of RA-FLSs and identify its potential role in RA pathogenesis. Methods Plasma levels and synovial expression of HAPLN1 were compared between healthy controls, and osteoarthritis (OA) and RA patients. Proliferation and migration of RA-FLSs transfected with siHAPLN1, HAPLN1OE (over-expression vector) and respective controls or treated with rHAPLN1 were measured by MTT and CCK8 assays as well as wound healing and transwell assays. RT-qPCR and automated WB analysis were used to compare the expression of AMPK-ɑ, TNF-ɑ, TGF-β, ACAN, MMPs, Cyclin-D1 and Ki-67 after siHAPLN1 or HAPLN1OE transfection. Proteomics and mRNA-seq analysis was done to study the differentially expressed proteins/genes after siHAPLN1 or rHAPLN1 treatment. Results Expression of HAPLN1 was increased in the plasma samples and synovium tissues of RA patients. HAPLN1OE transfected or rHAPLN1 treated RA-FLSs showed an increased proliferation capacity. However, Si-HAPLN1 has failed to affect the viability of RA-FLSs, though it decreased the migration ability of these cells. On the other hand, HAPLN1OE or rHAPLN1 had inhibitory effect on migration. Both si-HAPLN1 and HAPLN1OE treated RA-FLSs had down-regulated AMPK-ɑ gene expression, while protein level was found to be up-regulated. Furthermore, si-HAPLN1 has down-regulated TNF-ɑ, MMPs, IL-6, TGF-β, fibronectin and ACAN levels, while HAPLN1OE has up-regulated the levels of TNF-ɑ, MMPs, IL-6 and ACAN. Proteomics and mRNA-Seq analysis demonstrated HAPLN1 function in the activation of inflammation, proliferation, increased cell adhesion and strengthening of ECM function. Conclusons HAPLN1 accelerated the proliferation of RA-FLSs but inhibited its migration ability. HAPLN1 network was found to be mainly involved in the activation of inflammation, cell proliferation and increased cell adhesion.
Article
Full-text available
Accumulating data support the importance of interactions between pre- and postsynaptic neuronal elements with astroglial processes and extracellular matrix (ECM) for formation and plasticity of chemical synapses, and thus validate the concept of a tetrapartite synapse. Here we outline the major mechanisms driving: (i) synaptogenesis by secreted extracellular scaffolding molecules, like thrombospondins (TSPs), neuronal pentraxins (NPs) and cerebellins, which respectively promote presynaptic, postsynaptic differentiation or both; (ii) maturation of synapses via reelin and integrin ligands-mediated signaling; and (iii) regulation of synaptic plasticity by ECM-dependent control of induction and consolidation of new synaptic configurations. Particularly, we focused on potential importance of activity-dependent concerted activation of multiple extracellular proteases, such as ADAMTS4/5/15, MMP9 and neurotrypsin, for permissive and instructive events in synaptic remodeling through localized degradation of perisynaptic ECM and generation of proteolytic fragments as inducers of synaptic plasticity.
Article
Full-text available
Growing evidence points to synaptic pathology as a core component of the pathophysiology of schizophrenia (SZ). Significant reductions of dendritic spine density and altered expression of their structural and molecular components have been reported in several brain regions, suggesting a deficit of synaptic plasticity. Regulation of synaptic plasticity is a complex process, one that requires not only interactions between pre- and post-synaptic terminals, but also glial cells and the extracellular matrix (ECM). Together, these elements are referred to as the 'tetrapartite synapse', an emerging concept supported by accumulating evidence for a role of glial cells and the extracellular matrix in regulating structural and functional aspects of synaptic plasticity. In particular, chondroitin sulfate proteoglycans (CSPGs), one of the main components of the ECM, have been shown to be synthesized predominantly by glial cells, to form organized perisynaptic aggregates known as perineuronal nets (PNNs), and to modulate synaptic signaling and plasticity during postnatal development and adulthood. Notably, recent findings from our group and others have shown marked CSPG abnormalities in several brain regions of people with SZ. These abnormalities were found to affect specialized ECM structures, including PNNs, as well as glial cells expressing the corresponding CSPGs. The purpose of this review is to bring forth the hypothesis that synaptic pathology in SZ arises from a disruption of the interactions between elements of the tetrapartite synapse.
Article
Full-text available
The perineuronal net (PNN) represents a lattice like structure that is prominently expressed along the soma and proximal dendrites of parvalbumin (PV) positive interneurons in varied brain regions including cortex and hippocampus. It is thus apposed to sites at which PV neurons receive synaptic input. Emerging evidence suggests that changes in PNN integrity may affect glutamatergic input to PV interneurons, a population that is critical to the expression of synchronous neuronal population discharges that occur with gamma oscillations and sharp wave ripples. The present review is focused on the composition of PNNs, post-translation modulation of PNN components by sulfation and proteolysis, PNN alterations in disease, and potential effects of PNN remodeling on neuronal plasticity at the single cell and population level.
Article
Full-text available
Perineuronal nets (PNNs) are unique extracellular matrix (ECM) structures that wrap around certain neurons in the central nervous system (CNS) during development and control plasticity in the adult CNS. They appear to contribute to a wide range of diseases/disorders of the brain, are involved in recovery from spinal cord injury, and are altered during ageing, learning and memory, and after exposure to drugs of abuse. Here the focus is on how a major component of PNNs, chondroitin sulfate proteoglycans (CSPGs), control plasticity, and on the role of PNNs in memory in normal ageing, in a tauopathy model of Alzheimer’s disease, and in drug addiction. Also discussed is how altered ECM/PNN formation during development may produce synaptic pathology associated with schizophrenia, bipolar disorder, major depression, and autism spectrum disorders. Understanding the molecular underpinnings of how PNNs are altered in normal physiology and disease will offer insights into new treatment approaches for these diseases.
Chapter
Full-text available
In brains of higher vertebrates, the delicate balance of structural remodeling and stabilization of neuronal networks changes over the lifespan. While the juvenile brain is characterized by high structural plasticity, it is more restricted in the adult. During brain maturation, the occurrence of the extracellular matrix (ECM) is a critical step to restrict the potential for neuronal remodeling and regeneration, but providing structural tenacity. How this putative limitation of adult neuronal plasticity might impact on learning-related plasticity, lifelong memory reformation, and higher cognitive functions is subject of current research. Here, we summarize recent evidence that recognizes the ECM and its activity-dependent modulation as a key regulator of learning-related plasticity in the adult brain. We will first outline molecular concepts of enzymatic ECM modulation and its impact on synaptic plasticity mechanisms. Thereafter, the ECM's role in converting juvenile to adult plasticity will be explained by several key studies in wild-type and genetic knockout animals. Finally, current research evidences the impact of ECM dynamics in different brain areas including neocortex on learning-related plasticity in the adult brain impacting on lifelong learning and memory. Experimental modulation of the ECM in local neuronal circuits further opens short-term windows of activity-dependent reorganization. Malfunctions of the ECM might contribute to a variety of neurological disorders. Therefore, experimental ECM modulation might not only promote complex forms of learning and cognitive flexible adaptation of valuable behavioral options, but has further implications for guided neuroplasticity with regenerative and therapeutic potential.
Article
Full-text available
We have previously found that unilateral labyrinthectomy is accompanied by modification of hyaluronan and chondroitin sulfate proteoglycan staining in the lateral vestibular nucleus of rats and the time course of subsequent reorganization of extracellular matrix assembly correlates to the restoration of impaired vestibular function. The tenascin-R has repelling effect on pathfinding during axonal growth/regrowth, and thus inhibits neural circuit repair. By using immunohistochemical method, we studied the modification of tenascin-R expression in the superior, medial, lateral, and descending vestibular nuclei of the rat following unilateral labyrinthectomy. On postoperative day 1, tenascin-R reaction in the perineuronal nets disappeared on the side of labyrinthectomy in the superior, lateral, medial, and rostral part of the descending vestibular nuclei. On survival day 3, the staining intensity of tenascin-R reaction in perineuronal nets recovered on the operated side of the medial vestibular nucleus, whereas it was restored by the time of postoperative day 7 in the superior, lateral and rostral part of the descending vestibular nuclei. The staining intensity of tenascin-R reaction remained unchanged in the caudal part of the descending vestibular nucleus bilaterally. Regional differences in the modification of tenascin-R expression presented here may be associated with different roles of individual vestibular nuclei in the compensatory processes. The decreased expression of the tenascin-R may suggest the extracellular facilitation of plastic modifications in the vestibular neural circuit after lesion of the labyrinthine receptors. © 2015, Editorial Board of Neural Regeneration Research. All rights reserved.
Article
Full-text available
Perineuronal nets (PNNs) are aggregates of extracellular matrix molecules surrounding several types of neurons in the adult CNS, which contribute to stabilising neuronal connections. Interestingly, a reduction of PNN number and staining intensity has been observed in conditions associated with plasticity in the adult brain. However, it is not known whether spontaneous PNN changes are functional to plasticity and repair after injury. To address this issue, we investigated PNN expression in the vestibular nuclei of the adult mouse during vestibular compensation, namely the resolution of motor deficits resulting from a unilateral peripheral vestibular lesion. After unilateral labyrinthectomy, we found that PNN number and staining intensity were strongly attenuated in the lateral vestibular nucleus on both sides, in parallel with remodelling of excitatory and inhibitory afferents. Moreover, PNNs were completely restored when vestibular deficits of the mice were abated. Interestingly, in mice with genetically reduced PNNs, vestibular compensation was accelerated. Overall, these results strongly suggest that temporal tuning of PNN expression may be crucial for vestibular compensation.
Article
Full-text available
Extracellular matrix (ECM) accumulates around different neuronal compartments of the central nervous system (CNS) or appears in diffuse reticular form throughout the neuropil. In the adult CNS, the perineuronal net (PNN) surrounds the perikarya and dendrites of various neuron types, whereas the axonal coats are aggregations of ECM around the individual synapses, and the nodal ECM is localized at the nodes of Ranvier. Previous studies in our laboratory demonstrated on rats that the heterogeneous distribution and molecular composition of ECM is associated with the variable cytoarchitecture and hodological organization of the vestibular nuclei and may also be related to their specific functions in gaze and posture control as well as in the compensatory mechanisms following vestibular lesion. Here, we investigated the ECM expression pattern in the climbing fiber-generating inferior olive (IO), which is functionally related to the vestibular nuclei. By using histochemical and immunohistochemical methods, the most characteristic finding was the lack of PNNs, presumably due to the absence of synapses on the perikarya and proximal dendrites of IO neurons. On the other hand, the darkly stained dots or ring-like structures in the neuropil might represent the periaxonal coats around the axon terminals of olivary synaptic glomeruli. We have observed positive ECM reaction for the hyaluronan, tenascin-R, hyaluronan and proteoglycan link protein 1 (HAPLN1) and various chondroitin sulfate proteoglycans. The staining intensity and distribution of ECM molecules revealed a number of differences between the functionally different subnuclei of IO. We hypothesized that the different molecular composition and intensity differences of ECM reaction is associated with different control mechanisms of gaze and posture control executed by the visuomotor-vestibular, somatosensory and integrative subnuclei of the IO.
Article
Previous studies in our laboratory showed that the organization and heterogeneous molecular composition of extracellular matrix is associated with the variable cytoarchitecture, connections and specific functions of the vestibular nuclei and two related areas of the vestibular neural circuits, the inferior olive and prepositus hypoglossi nucleus. The aim of the present study is to reveal the organization and distribution of various molecular components of extracellular matrix in the red nucleus, a midbrain premotor center. Morphologically and functionally the red nucleus is comprised of the magno- and parvocellular parts, with overlapping neuronal population. By using histochemical and immunohistochemical methods, the extracellular matrix appeared as perineuronal net, axonal coat, perisynaptic matrix or diffuse network in the neuropil. In both parts of the red nucleus we have observed positive hyaluronan, tenascin-R, link protein, and lectican (aggrecan, brevican, versican, neurocan) reactions. Perineuronal nets were detected with each of the reactions and the aggrecan showed the most intense staining in the pericellular area. The two parts were clearly distinguished on the basis of neurocan and HAPLN1 expression as they have lower intensity in the perineuronal nets of large cells and in the neuropil of the magnocellular part. Additionally, in contrast to this pattern, the aggrecan was heavily labeled in the magnocellular region sharply delineating from the faintly stained parvocellular area. The most characteristic finding was that the appearance of perineuronal nets was related with the neuronal size independently from its position within the two subdivisions of red nucleus. In line with these statements none of the extracellular matrix molecules were restricted exclusively to the magno- or parvocellular division. The chemical heterogeneity of the perineuronal nets may support the recently accepted view that the red nucleus comprises more different population of neurons than previously reported.
Article
The prepositus hypoglossi nucleus (PHN) is a mossy fiber-generating precerebellar nucleus of the brainstem, regarded as one of the neural integrators of the vestibulo-ocular reflex. The aim of the present work is to reveal the distribution of various molecular components of the extracellular matrix (ECM) in the prepositus hypoglossi nucleus by using histochemical and immunohistochemical methods. Our most characteristic finding was the accumulation of the ECM as perineuronal net (PNN) and axonal coat and we detected conspicuous differences between the magnocellular (PHNm) and parvocellular (PHNp) divisions of the PHN. PNNs were well developed in the PHNm, whereas the pericellular positivity was almost absent in the PHNp, here a diffuse ECM was observed. In the PHNm the perineuronal net explored the most intense staining with the aggrecan, and tenascin-R antibodies followed by the hyaluronan, then least with reactions for chondroitin sulfate-based proteoglycan components and HAPLN1 link protein reactions, but PNNs were not observed with the versican, neurocan, and brevican staining. We hypothesized that the difference in the ECM organization of the two subnuclei is associated with their different connections, cytoarchitecture, physiological properties and with their different functions in the vestibular system. Copyright © 2015. Published by Elsevier Ireland Ltd.