ArticlePDF Available

The fatty acid binding protein FABP7 is required for optimal oligodendrocyte differentiation during myelination but not during remyelination

Authors:
  • Institute for Health and Biomedical Research of Alicante (ISABIAL) and Institute of Neuroscience CSIC-UMH

Abstract

The major constituents of the myelin sheath are lipids, which are made up of fatty acids (FAs). The hydrophilic environment inside the cells requires FAs to be bound to proteins, preventing their aggregation. Fatty acid binding proteins (FABPs) are one class of proteins known to bind FAs in a cell. Given the crucial role of FAs for myelin sheath formation we investigated the role of FABP7, the major isoform expressed in oligodendrocyte progenitor cells (OPCs), in developmental myelination and remyelination. Here, we show that the knockdown of Fabp7 resulted in a reduction of OPC differentiation in vitro. Consistent with this result, a delay in developmental myelination was observed in Fabp7 knockout animals. This delay was transient with full myelination being established before adulthood. FABP7 was dispensable for remyelination, as the knockout of Fapb7 did not alter remyelination efficiency in a focal demyelination model. In summary, while FABP7 is important in OPC differentiation in vitro, its function is not crucial for myelination and remyelination in vivo.
RESEARCH ARTICLE
The fatty acid binding protein FABP7 is required for optimal
oligodendrocyte differentiation during myelination but not
during remyelination
Sarah Foerster
1
| Alerie Guzman de la Fuente
1
| Yoshiteru Kagawa
2
|
Theresa Bartels
1
| Yuji Owada
2
| Robin J. M. Franklin
1
1
Wellcome-Medical Research Council
Cambridge Stem Cell Institute, Jeffrey Cheah
Biomedical Centre, University of Cambridge,
Cambridge, UK
2
Department of Organ Anatomy, Tohoku
University Graduate School of Medicine,
Sendai, Japan
Correspondence
Robin J. M. Franklin, Wellcome-Medical
Research Council Cambridge Stem Cell
Institute, Jeffrey Cheah Biomedical Centre,
University of Cambridge, Cambridge, UK.
Email: rjf1000@cam.ac.uk
Present Address
Alerie Guzman de la Fuente, Wellcome-
Wolfson Institute for Experimental Medicine,
Queen's University Belfast, 97 Lisburn Road,
Belfast, BT9 7JL, UK.
Funding information
Dr. Miriam and Sheldon G. Adelson Medical
Research Foundation; European Committee for
Treatment and Research in Multiple Sclerosis;
Japan Society for the Promotion of Science;
Medical Research Council; Multiple Sclerosis
Society; Wellcome
Abstract
The major constituents of the myelin sheath are lipids, which are made up of fatty
acids (FAs). The hydrophilic environment inside the cells requires FAs to be bound to
proteins, preventing their aggregation. Fatty acid binding proteins (FABPs) are one
class of proteins known to bind FAs in a cell. Given the crucial role of FAs for myelin
sheath formation we investigated the role of FABP7, the major isoform expressed in
oligodendrocyte progenitor cells (OPCs), in developmental myelination and
remyelination. Here, we show that the knockdown of Fabp7 resulted in a reduction
of OPC differentiation in vitro. Consistent with this result, a delay in developmental
myelination was observed in Fabp7 knockout animals. This delay was transient with
full myelination being established before adulthood. FABP7 was dispensable for
remyelination, as the knockout of Fapb7 did not alter remyelination efficiency in a
focal demyelination model. In summary, while FABP7 is important in OPC differentia-
tion in vitro, its function is not crucial for myelination and remyelination in vivo.
KEYWORDS
fatty acid binding protein, myelination, remyelination, OPC
1|INTRODUCTION
The myelin sheath of the central nervous system (CNS) has a unique
molecular composition that distinguishes it from other cell mem-
branes: it comprises 7181% of lipids, which include cholesterols
(26%), galactolipids (31%), and phospholipids (44%) (Norton &
Poduslo, 1973). The specific lipid composition allows close compac-
tion of the myelin sheath, creating a highly ordered, hydrophobic
barrier that enables myelin to function as an electric insulator
(Simons & Nave, 2015).
Fatty acids (FAs) are the building blocks of the galactolipids and
phospholipids found in myelin. To maintain water solubility and prevent
aggregation, FAs are bound to proteins in the cytoplasm (Cistola, Hamil-
ton, Jackson, & Small, 1988). There are ten FA binding proteins (FABPs),
of which FABP3, FABP5, and FABP7 are expressed in the CNS (Owada,
Yoshimoto, & Kondo, 1996). FABP7 is mainly expressed in astrocytes
(Owada et al., 1996) and oligodendrocyte progenitor cells (OPCs; Sharifi
et al., 2013), a progenitor cell giving rise to mature oligodendrocytes
Sarah Foerster and Alerie Guzman de la Fuente are joint first authors.
Received: 11 October 2019 Revised: 10 January 2020 Accepted: 23 January 2020
DOI: 10.1002/glia.23789
This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2020 The Authors. Glia published by Wiley Periodicals, Inc.
Glia. 2020;111. wileyonlinelibrary.com/journal/glia 1
(OLs) in development and in response to injury (Franklin & ffrench-Con-
stant, 2017). OPCs isolated from Fabp7 knockout (Fabp7KO) mice
have impaired differentiation in vitro (Sharifi et al., 2013). In animal
models of primary demyelination, Fabp7 expression is increased in
CNS resident cells (Bannerman, Hahn, Soulika, Gallo, & Pleasure,
2006; Huang et al., 2011; Kipp et al., 2011). However, the impor-
tance of FABP7 in the OPCs biology in vivo is not well understood.
In this study, we found that the pattern of FABP7 expression
closely follows the timeline of myelination during postnatal develop-
ment. This suggested a role in developmental myelination which was
confirmed by a delay in developmental myelination in Fabp7 knockout
mice. From early adulthood onward, the expression of FABP7 was
dramatically reduced and the protein was only re-expressed following
a demyelinating insult. However, Fabp7KO mice showed no effect on
the remyelination capacity of OPCs.
2|MATERIALS AND METHODS
2.1 |Animal husbandry
Animal experiments conformed to the UK Animals (Scientific Pro-
cedures) Act 1986 and were approved by the Cambridge University
local ethical committees before licensing by the UK Home Office.
The animals were housed under standard laboratory conditions
on a 12 hr light/dark cycle with constant access to food and
water. Fabp7KO mice were obtained from the Owada laboratory,
Tohoku University, Japan (Owada et al., 2006). Demyelination
experiments in the Fabp7KO animals were performed at Tohoku
University, Japan. The experimental protocol for performing
demyelinating lesions was reviewed by the ethics committee for
Animal Experimentation of Tohoku University Graduate School of
Medicine and carried out according to the guidelines for animal
experimentation of the Tohoku University Graduate School of
Medicine and under the law and notification requirements of the
Japanese government.
2.2 |Isolation of primary OPCs
OPCs were isolated from neonatal (P7P30),2months,3months,
9 months, 12 months, or 24 months old Sprague Dawley rats as publi-
shed previously (Neumann et al., 2019; Segel et al., 2019). Briefly, brains
were digested in a papain solution (34U/ml, Worthington) containing
DNAseTypeIV(20μg/ml, Gibco) for 3040 min at 37C.Thetissuewas
then triturated into a single cell suspension in Half (made in house) sup-
plemented with B27 (1×, Gibco) and sodium pyruvate (2 mM, Gibco).
After trituration, the single cell suspension was filtered through a 70 μm
strainer and separated from debris by gradient density centrifugation
(800 g, 20 min, RT) using 22.5% Percoll
®
(GE Healthcare). The cell pellet
was then subjected to red blood cell lysis using the red blood cell lysis
buffer(Sigma)for2min.Finally,OPCs were purified using the anti-A2B5
microbeads MACS
®
cell separation system according to the
manufacturer's protocol (Miltenyi Biotech). If used for Western blot anal-
ysis, obtained OPCs were resuspended in IP lysis/wash buffer (25 mM
Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% NP-40, and 5% glyc-
erol; Thermo-Fisher Scientific). If used for qRT-PCR analysis, obtained
OPCs were resuspended in TRIzol Reagent (Thermo-Fisher Scientific).
2.3 |Isolation of OPCs using mixed glia culture
OPCs were obtained from P0P3 old Sprague Dawley rats following
the protocol of McCarthy and de Vellis (McCarthy & de Vellis, 1980).
Briefly, the cortex was digested in a papain solution (34U/ml,
Worthington) for 1 hr at 37C. Digestion was stopped by adding
DMEM (Gibco) supplemented with 10% FBS (Biosera) and the tissue
was spun at 300 g for 5 min. The tissue was resuspended in DMEM
with 10% FBS and 1% penicillin/streptomycin and cells from two brains
were plated in a poly-D-lysine (Sigma) coated T75 flask. Mix glial cells
were cultured at 37Cand5%CO
2
for 10 days with media changes
every 3 days. After 10 days in vitro (DIV), the flasks were subjected to a
shake off protocol to separate OPCs from the rest of the glial cells
(McCarthy & de Vellis, 1980). OPCs were cultured in OPC medium
(DMEM F12 (Gibco), 2 mM sodium pyruvate (Gibco), 60 μgN-acetyl-
cysteine (Sigma-Aldrich), 5 μg/ml insulin (Gibco), 21 mM D-glucose
(Sigma-Aldrich), 50 μg/ml apo-transferrin (Sigma-Aldrich), 16.1 μg/ml
putrescine (Sigma-Aldrich), 40 ng/ml sodiumselenite (Sigma-Aldrich),
and 60 ng/ml progesterone (Sigma-Aldrich) with daily addition of
10 ng/ml PDGF-AA and 10ng/ml bFGF (PeproTech) at 37Cand5%
CO
2
. Cells were plated on poly-D-lysine coated coverslips at a density
of 20,000 cells/13 mm coverslip. Each n-number represents an OPC
isolation from independent mix glial cell culture preparations on differ-
ent days, each preparation performed from four individual P0P3 old
Sprague Dawley rats.
2.4 |Fabp7 siRNA knockdown
OPCs isolated from mixed glial cell cultures were cultured for 2 DIV with
growth factors, then the medium was changed to OPC medium without
penicillin/streptomycin overnight. Cells were transfected with 50 nM
FABP7 siRNA or equivalent non-targeting control (GE Healthcare) using
1% Lipofectamine siRNAMAX (Invitrogen) diluted in Opti-MEM (Gibco)
according to the manufacturer's protocol. Six hours after transfection, the
medium was replaced by OPC medium without growth factors, thyroxine
and triiodothyronine. After 48 hr of transfection, cells were fixed with 4%
(w/v) PFA for 10 min if used for immunocytochemistry staining, lysed in
IP lysis/wash buffer (Thermo-Fisher Scientific) if used for Western blot
analysis or TRIzol Reagent (Thermo-Fisher Scientific) if used for qPCR.
2.5 |Immunocytochemistry
After siRNA treatment, cells were blocked with 5% normal donkey
serum (NDS) (Sigma-Aldrich) supplemented with 0.1% Triton X-100
2FOERSTER ET AL.
(Sigma-Aldrich) in PBS for 1 hr at RT. Then cells were incubated for
1 hr at RT with primary antibodies (Mouse anti-CNPase antibody,
1:500 (C5922, Sigma-Aldrich); Rabbit anti-Olig2 antibody, 1:500
(AB9610, Millipore); or rat anti-MBP antibody, 1:500 (MCA4095,
Serotec)) in 5% NDS with 0.1% Triton X-100. After three washes, the
cells were incubated with the appropriate Alexa Flour secondary anti-
bodies (1:500, Life-technologies) diluted in 5% NDS with 0.1% Triton
X-100 for 1 hr at RT. Nuclei were stained with Hoechst (2μg/ml,
Sigma-Aldrich) for 10 min at RT, before the coverslips were mounted
using Fluoromount G (Southern Biotech).
2.6 |Western blot
Protein lysates were mixed with NuPage loading buffer and NuPage
reducing agent (DTT) (Life-technologies) according to manufacturer's
protocol and boiled for 10 min at 95C. After denaturation, 5 μgof
each sample were loaded on 412% Bolts Gels (Life Technologies)
and the gels were subjected to electrophoresis at 120 V for 2 hr. In
the sumoylation and ubiquitination experiments, 1 μg ubiquitin (U-
100H, R&D Systems) and Sumo 1, 2, and 3 (K700, R&D Systems)
were loaded as control. The gels were then transferred onto a metha-
nol preactivated PVDF membrane (Millipore) using a wet transfer tank
(Bio-Rad) and 1×NuPage transfer buffer (Life-technologies) with 20%
ethanol for 90 min at 100 V. The PVDF membrane was then blocked
in blocking buffer (TBS blocking agent 1:1 (Li-Cor) with TBS (Fischer
scientific)-0.1% Tween (Sigma-Aldrich) or 5% skimmed milk in TBST
(TBS with 0.1% Tween)) for 1 hr at RT. PVDF membranes were incu-
bated overnight at 4C with the primary antibodies (Rabbit anti-BLBP,
1:500 (ab32423, Abcam); Mouse anti-actin, 1:5000 (A5441, Sigma);
Mouse anti-actin (sc-47778, SantaCruz Biotechnology); Mouse anti-
ubiquitin, 1:200 (sc8017, Santa Cruz); or mouse anti-sumo1 antibody,
1:500 (sc5308, Santa Cruz). Subsequently the membranes were incu-
bated with secondary antibodies (Donkey anti-rabbit 680, 1:10,000
(926_38073, Li-Cor); donkey anti-mouse 800, 1:10,000 (926_32212,
Li-Cor); goat anti-mouse IgG-HRP conjugated (AP307P, Millipore) or
goat anti-mouse IgG-HRP conjugated (AP124P, Millipore)). If appro-
priate, membranes were then incubated with an actin-peroxidase anti-
body, 1:25,000 (A3854, Sigma) for 20 min at RT. Fluorescent and
HRP signals were detected using the Odyssey apparatus (Li-Cor) with
an exposure time of 2 min.
2.7 |Phosphorylation and glycosylation
experiments
Cells were lysed in IP lysis/wash buffer (Thermo-scientific) without
the addition of protease and phosphatase inhibitors. For glycosylation
analysis, 5 μg OPC lysates were then subjected to digestion for 1 hr at
37C with EndoH enzyme or EndoH glycobuffer (NEB) alone as con-
trol. For phosphorylation analysis, 5 μg of OPC lysate were subjected
to digestion for 30 min at 30C with Lambda protein phosphatase or
10×NeBuffer Protein MetalloPhosphatases (PMP) and MnCl
2
only as
control. The OPC lysates were then boiled for 10 min at 95C and fur-
ther processed for Western Blot as described above.
2.8 |Real-time qPCR
RNA was isolated from cultured OPCs according to the RNAeasy Mini
Kit (74104, Qiagen). All RNA samples were stored at 80C prior to
further processing. cDNA was generated using the QuantiTect
Reverse Transcription Kit according to the instructions of the manu-
facturer (205310, Qiagen). For RT-qPCR, Fabp7 primers (forward
(50à30): AAGATGGTCGTGACTCTTAC; reverse (50à30): GGAA
ACCAAGTTGTCAAAAG) were used at a concentration of 400 μM.
The efficiency of the primer was greater than ~95% as determined by
serial dilutions of OPC cDNA. cDNA, primers, and the SYBR Green
Master Mix (204141, Qiagen) were mixed as instructed by the manu-
facturer, and RT-qPCR and melting curve analysis were performed on
Life Technologies' QuantStudio 6 Flex Real-Time PCR System. Fold
changes in gene expression were calculated using the ΔΔCt method
in Microsoft Excel.
2.9 |Toxin induced (lysolecithin) demyelination
model in the spinal cord
For spinal cord lysolecithin lesions 23 months old wild type and
homozygous Fabp7 knockout mice (Owada et al., 2006) were used.
Demyelination was induced in the caudal thoracic ventral funiculus of
the spinal cord by injection of 1% (v/v) lysolecithin as previously
described (Fancy et al., 2009).
2.10 |Immunohistochemistry
Mice were terminally anaesthetized and fixed by intracardiac perfu-
sion at 5, 10, or 21 days post lesion (dpl) induction using 4% (w/v)
PFA. Spinal cords were removed, postfixed in 4% (w/v) PFA overnight
at 4C, cryoprotected with 20% (w/v) sucrose for 2448 hr, embed-
ded, frozen in OCT medium and stored at 80C. Tissues were sec-
tioned at 12 μm and collected onto poly-L-lysine-coated glass slides.
12 μm cryo-sections were dried at RT and then rehydrated in PBS.
After rehydration, slides were postfixed for 10 min with 4% (w/v) PFA
and then washed 3×10 min with PBS. Sections were blocked with
5% NDS and 0.1% Triton X-100 for 1 hr at RT. In case the mouse-anti
APC antibody was used, the slides were blocked using the MOM Kit
according to the manufacturer's instructions (BMK-2202, Vector Lab-
oratories). After blocking, slides were incubated with primary anti-
bodies in blocking solutions overnight at 4C (Rabbit anti-OLIG2,
1:500 (AB9610, Millipore); Goat anti-SOX10, 1:100 (sc365692, Santa
Cruz); Rabbit anti-Ki67, 1:500 (ab16667, Abcam); or mouse anti-APC,
1:300 (OP80, Millipore). Slides were then incubated with the
appropriate Alexa Fluor
®
secondary antibodies 1:500 (Life Technolo-
gies) for 2 hr at RT. Nuclei were stained with Hoechst (2 μg/ml,
FOERSTER ET AL.3
Sigma-Aldrich) for 10 min at RT, before the coverslips were mounted
using Fluoromount G (Southern Biotech).
2.11 |Toluidine blue staining
For toluidine blue and electron microscopy experiments, mice were
terminally anaesthetized and fixed by intracardiac perfusion at 14 and
21 days post lesion (dpl) using 4% (w/v) glutaraldehyde. Spinal cords
were removed and postfixed in 4% (w/v) glutaraldehyde overnight at
4C. The tissue was dehydrated in a series of ethanol washes (1×70%
EtOH for 15 min, 1×95% EtOH for 15 min, and 3×100% EtOH for
10 min (Sigma)), washed twice in propylene oxide for 15 min and incu-
bated in a one-to-one mix of propylene oxide and resin (50% resin,
34% dodecenyl succinic anhydride (DDSA), 16% methyl nadic anhy-
dride (MNA), 2% 2,4,6-Tris(dimethylaminomethyl)phenol (DMP-30),
all (v/v), TAAB Laboratories) for at least 3 hr at RT. The tissue was
then incubated twice in pure resin for 12 hr at RT, before samples
were embedded in plastic containers in fresh resin and hardened for
2 days at 60C. Samples were cut into 0.75 μm resin sections and sec-
tions were then stained for 30 s with toluidine blue at 65C on a heat
plate. The toluidine blue images were blindly ranked by two indepen-
dent assessors, who ranked them according to their level of demyelin-
ation and remyelination. In resin sections, remyelinated axons can be
readily distinguished from normally myelinated axons outside the
lesion by the thinness of the myelin sheath. Within the lesion,
remyelinated axons can be distinguished from demyelinated axons
because the former have myelin sheaths recognizable as a dark
staining rim around the axon. The highest rank was given to the ani-
mal exhibiting the highest proportion of remyelinated axons. If it was
not possible to differentiate two animals using this method then they
were given the same rank. In this method, no attempt is made to
assign a value to the proportion of remyelination, but simply to estab-
lish how a section from an individual animal ranks relative to others.
FIGURE 1 FABP7 expression in OPCs. (a) Western blot of FABP7 expression in acutely isolated OPCs at different developmental time points
(P = postnatal day). (b) Quantification of FABP7 protein expression normalized to actin (one-way ANOVA, p= .0001, Bonferroni post hoc test;
(1) P7 versus P14, p= .0010; (2) P7 versus P21, p= .0003, (3) P7 versus P30, p= .0003; n= 3, mean ± SEM). (c) Western blot of FABP7
expression in ageing OPCs: note the higher molecular weight band appearing in adult OPCs. (d) Quantification of FAPB7 protein expression of
the lower and higher molecular weight bands normalized to actin (low molecular weight band: one-way ANOVA, p< .0001, Bonferroni posthoc
test; Neo (P7) vs 3, 9, and 24 months, p<0.0001; High molecular weight band: one-way ANOVA, p= .0366, Bonferroni posthoc test; Neo
(P7) verssus 3 months, p= .0300; n= 3, mean ± SEM). (e) qPCR quantification of the relative expression of Fabp7 mRNA in ageing OPCs
normalized to Pop4 (housekeeping gene) (one-way ANOVA; p< .0001, Bonferroni post hoc test; (1) Neo (P7) versus 2 and 12 months, p< .0001;
(2) 2 versus 12 months, p= .0030; n= 3, mean ± SEM)
4FOERSTER ET AL.
2.12 |Quantification
To quantify in vitro proliferation and differentiation assays, five ran-
domly chosen areas of the coverslip were imaged per condition with
the 20×objective of the SP5 Leica confocal with a 512 ×512 resolu-
tion and 2 μm stacks. Counting was performed manually using the cell
counter plugin in ImageJ (Version 2.0.0-rc-68/1.52 hr). To quantify
remyelination efficiency, three lesions per animal were imaged with
the 20×objective (SPR Leica Confocal) at a 512 ×512 resolution and
2μm stacks. Using ImageJ software (Version 2.0.0-rc-68/1.52 hr), the
lesion area was delineated and measured, and the number of different
cell types within the lesion was counted manually with the cell coun-
ter plugin. To quantify OPC differentiation during developmental
myelination, white matter of three sections per animal were imaged
using the 20×objective (Leica SP5 confocal) at 512 ×512 resolution
with 2 μm stacks. The area of interest was measured using Image the
FIGURE 2 Fabp7 siRNA knockdown affects OPC differentiation. (a) Western blot of FABP7 expression in OPCs in vitro after siRNA
knockdown compared to scrambled siRNA control. NT = non-targeting siRNA. DHA = docosahexaenoic acid. (b,c) Quantification of FABP7
protein (b) or mRNA (c) expression after siRNA treatment compared to scrambled siRNA control (unpaired Student's t-test, p= .0135 (b); unpaired
Student's t-test, p= .0239 (c); n= 3, mean ± SEM). (d) Immunocytochemistry staining for OLIG2, CNPase, and MBP (marker of differentiated
oligodendrocytes) in mixed glia derived OPCs after Fabp7 siRNA knockdown. (Scale bar: 100 μm) (eg) Quantification of the percentage of DAPI
+
cells expressing OLIG2 (e), OLIG2
+
cells expressing CNPase (f) or MBP (g) (unpaired Student's t-test, p= .0701 (f ), p= .0291 (g); n=3,
mean ± SEM)
FOERSTER ET AL.5
FIGURE 3 Developmental myelination in the spinal cord is delayed in Fabp7 knockout mice. (a) Immunohistochemistry staining for OLIG2
and APC (marker of differentiated oligodendrocytes) in the ventral white matter of the spinal cord of WT and Fabp7KO mice at P7 and P14 (Scale
bar: 100 μm). (b) Quantification of OLIG2
+
cells per area in the whole spinal cord of WT and Fabp7KO mice at P7 and P14 (unpaired Student's
t-test, p= .3430 (P7) and p= .1089 (P14); n=56, mean ± SEM). (c) Quantification of the percentage of OLIG2
+
APC
+
in the white matter of the
spinal cord of WT and Fabp7KO mice at P7 and P14 (unpaired Student's t-test, p= .0435 (P7) and p= .1439(P14); n=56, mean ± SEM).
(d) Toluidine blue staining of the ventral spinal cord in WT and Fabp7KO mice at P7 and P14 (Scale bar: 50 μm). White squares highlight areas
shown in the higher magnification insets. (e) Quantification of the number of myelinated axons per area in the ventral white matter at P7 and P14
(unpaired Student's t-test, p= .009 (P7); n= 3 (P7), n= 2 (P14), mean ± SEM)
6FOERSTER ET AL.
J software and the number of cells positive for the indicated marker
proteins within the area of interest was counted manually. Toluidine
blue staining was imaged using 63×objective of the Zeiss Apotome
with a 2048 ×2048 resolution. White matter of three sections per
animal were imaged and the number of myelinated axons in the white
matter were manually counted using the cell counter plugin in ImageJ
(Version 2.0.0-rc-68/1.52 hr). Western blots were quantified by mea-
suring the integrated density of each of the bands by the quantifica-
tion tool in the Image studio software (Version 4.0, Li-Cor).
2.13 |Statistics
Statistical analysis was performed using the Prism 7.0 and 8.0
(GraphPad Software) and SPSS Statistics 20.0 (IBM). Mean ± SEM are
shown in all the graphs. The data were analyzed for normal distribu-
tion using D'Agostino-Pearson omnibus and ShapiroWilk normality
test. If the number of biological replicates was low (n < 4), normality
was calculated using normality via residuals. A two-tailed unpaired
Student's t-test was performed to assess the statistical significance
between two groups. In case the data were not normally distributed, a
U-MannWhitney test was performed. When comparing more than
two groups, a one-way Anova test was used followed by a Tukey's
posthoc test. If the sample was not normally distributed, a Kruskal
Wallis test combined with a Dunn's posthoc test was carried out.
Remyelination ranking was evaluated using a U-MannWhitney test.
Western-blot data were analyzed using a one-way ANOVA and the
corresponding Bonferroni posthoc test. qPCR data were analyzed
using a one sample t-test. In case the data were shown in percentages,
adequate arcsin conversion was done prior to the unpaired Student's
t-test.
3|RESULTS
FABP7 was highly expressed in OPCs isolated from rat brain at post-
natal day 7 (P7; Figure 1a,b). However, with postnatal development,
FABP7 protein expression steadily declined, reaching significance as
early as P14 (P7 vs. P14, p= .001, Bonferroni posthoc test; Figure 1a,
b). From P30, a higher molecular weight of FABP7 is detected at
40 kDa (Figure 1a,c). This is likely attributable to posttranslational
changes as the adult form of FABP7 is more glycosylated and phos-
phorylated compared to its neonatal counterpart (Figure S1a).
Ubiquitination and sumoylation do not contribute to the high molecu-
lar weight band in young adult OPCs (Figure S1b,c). With ageing,
Fabp7 mRNA and protein expression continue to decrease until it is
undetectable in the aged rat (Figure 1ce). The antibody used for the
Western Blot analysis was specific to FABP7 protein as we did not
detect FAPB7 protein in Fabp7KO animals, as it has been reported in
a previous publication (Figure S1d; Driessen et al., 2018).
As FABP7 is highly expressed in neonatal OPCs (Figure 1), we
investigated its role in OPC proliferation and differentiation by
FIGURE 4 Developmental myelination in the brain is also delayed in Fabp7 knockout mice. (a) Immunohistochemistry staining for OLIG2 and
APC in the corpus callosum of WT and Fabp7KO mice at P7 and P14 (Scale bar: 100 μm). (b) Quantification of OLIG2
+
cells per area in the corpus
callosum of WT and Fabp7KO mice at P7 and P14 (unpaired Student's t-test, p= .9307 (P7) and p= .3601 (P14); n=45, mean ± SEM).
(c) Quantification of the percentage of OLIG2
+
APC
+
cells in the white matter of the corpus callosum of WT and Fabp7KO mice at P7 and P14
(unpaired Student's t-test, p= .0001 (P7) and p= .0045 (P14); n=45, mean ± SEM)
FOERSTER ET AL.7
knocking down Fabp7 in vitro using siRNA. siRNA knockdown
(KD) efficiency was >75% both on protein (Figure 2a,b) and RNA
(Figure 2c) level. Mixed glial culture-derived neonatal OPC cultures
contained 90% Olig2
+
cells (Figure 2d,e), minimizing any possible
indirect effect on OPCs from Fabp7KD in other CNS cell types
in vitro. Knockdown of Fabp7 in these mixed glial culture-derived neo-
natal OPCs resulted in a 2.2-fold reduction of OPC differentiation into
CNP
+
/OLIG2
+
mature oligodendrocytes in differentiation medium
FIGURE 5 FABP7 is not essential for remyelination. (a) Schematic drawing of lysolecithin induced demyelination in the ventral white matter
of the spinal cord. (b) Immunohistochemistry staining for OLIG2 and APC in the lesion in the ventral white matter of WT and Fabp7KO mice at
14 days post lesion (dpl) (Scale bar: 100 μm). (c) Quantification of OLIG2
+
cells per area in the lesion of WT and Fabp7KO mice at 14dpl (unpaired
Student's t-test, p= .4371; n= 5, mean ± SEM) (d) Quantification of the percentage of OLIG2
+
APC
+
cells in the lesion of WT and Fabp7KO mice
at 14dpl (unpaired Student's t-test, p= .3154; n= 5, mean ± SEM) (e,g) Toluidine blue staining of the lesion in WT and Fabp7KO mice at 14dpl
(e) and 21dpl (g) (Scale bar: 25 μm). (f,h) Ranking analysis of remyelination efficiency in WT and Fabp7KO mice at 14dpl (f ) and 21dpl
(h) (U-MannWhitney test, p= .8571 (f), p= .2000 (h); n= 3-4, mean ± SEM)
8FOERSTER ET AL.
from which the growth factors PDGF and FGF-2 were removed (WT:
73% CNP
+
/OLIG2
+
cells, Fabp7KD: 33% CNP
+
/OLIG2
+
cells, p= .07,
unpaired Student's t-test; Figure 2d,f). Similarly, the proportion of
OPCs differentiating into MBP
+
/OLIG2
+
myelin-sheath forming oligo-
dendrocytes is also significantly reduced (WT: 27% MBP
+
/OLIG2
+
cells, Fabp7KD: 5% MBP
+
/OLIG2
+
cells, p= .03, unpaired Student's t-
test) (Figure 2d,g). These findings concur with a study by Sharifi and
colleagues, whose data was also consistent with a role for FABP7 in
OPC differentiation (Sharifi et al., 2013). In the same publication it
was also shown that the knockout of Fabp7 (Fabp7KO) affected the
OPC proliferation capacity in vitro (Sharifi et al., 2013). In agreement
with this study, we found that, while the number of SOX10
+
cells
stays constant (Figure S2a,b), Fabp7KD led to a reduction of the pro-
portion of SOX10
+
oligodendrocyte lineage cells that expressed KI67
(WT: 72% SOX10
+
/KI67
+
cells, Fabp7KD: 61% SOX10
+
/KI67
+
cells,
p= .03, unpaired Student's t-test; Figure S2a,c).
Given that the expression pattern of FABP7 is reminiscent of the
timeline of developmental myelination (Figure 1) and FABP7 contrib-
utes to OPC differentiation in vitro (Figure 2), we investigated
whether the knockout of Fabp7 altered developmental myelination.
The global homozygous knockout of Fabp7 did not affect the density
of oligodendrocyte lineage cells, identified by the expression of OLIG2
(Figure 3a,b). However, Fabp7KO mice showed a significantly reduced
percentage of APC
+
/OLIG2
+
mature oligodendrocytes in the spinal
cord at P7 (WT: 40% APC
+
OLIG2
+
,Fabp7KO: 32% APC
+
OLIG2
+
,
p= .04, unpaired Student's t-test; Figure 3a,c). This corresponded with
a decreased number of myelinated axons in the spinal cord at P7 (WT:
78977 myelinated axons/mm
2
,Fabp7KO: 67110 myelinated axons/
mm
2
,p= .009, unpaired Student's t-test; Figure 3d,e). However, this
hypomyelination was transient, as the percentage of mature APC
+
/
OLIG2
+
oligodendrocytes, as well as the number of myelinated axons,
was not significantly different in the spinal cord of WT or Fabp7KO
mice at P14 (Figure 3ae). As in the spinal cord, the percentage of
APC
+
/OLIG2
+
mature oligodendrocytes was significantly decreased in
the corpus callosum of Fabp7KO animals at P7 (WT: 47% APC
+
/
OLIG2
+
,Fabp7KO: 31% APC
+
/OLIG2
+
,p= .0001, unpaired Student's
t-test) and P14 (WT: 81% APC
+
/OLIG2
+
,Fabp7KO: 66% APC
+
/
OLIG2
+
,p= .0045, unpaired Student's t-test; Figure 4a,c), while no
differences were detected in the density of OLIG2
+
oligodendrocyte
lineage cells (Figure 4a,b). The extended period of delayed myelination
in the corpus callosum compared to the spinal cord is possibly due to
a later onset of myelination in the corpus callosum. Unlike in the
already published data (Sharifi et al., 2013) and our own in vitro
results (Figure S2a,c), OPC proliferation was not altered by the
absence of FABP7 in the spinal cord at P7 and P14 (Figure S2d,e).
These data indicate that FABP7 plays a role in OPC differentiation,
but not proliferation, during development.
OPCs generate oligodendrocytes not only during developmental
myelination, but also for remyelination in response to demyelinating
injury. Given the involvement of FABP7 in OPC differentiation during
developmental myelination (Figures 3 and 4) and that FABP7 expres-
sion is increased at 14 days post lesion (dpl) during remyelination
(Huang et al., 2011), we next asked whether FABP7 also plays a role
in response to a toxin-induced demyelination. To assess the
remyelination capacity of Fabp7KO OPCs, we created a demyelinating
lesion in the ventral spinal cord white matter of young adult mice by
direct injection of lysolecithin (Figure 5a). There was no difference in
the density of OLIG2
+
oligodendrocyte lineage cells in the lesion
(Figure 5b,c), neither in the percentage of APC
+
/OLIG2
+
mature oligo-
dendrocytes at 14 dpl (Figure 5b,d). Similarly, unbiased ranking of the
proportion of remyelinated axons in the lesion did not show any sig-
nificant difference between WT and Fabp7KO animals at 14 and
21 dpl (Figure 5eh). Additionally, we also did not find a difference in
the proliferation capacity of OPCs in WT and Fabp7KO after a demye-
linating insult (Figure S3ac), indicating that the loss of FABP7 in oli-
godendrocyte lineage cells does not impede their remyelination
capacity.
4|DISCUSSION
4.1 |FABP7 is involved in OPC differentiation
in vitro
Here we confirm that FABP7 plays a role in OPCs differentiation
in vitro (Figure 2), which agrees with an earlier study using a different
experimental approach (Sharifi et al., 2013). The mechanism by which
FABP7 modulates OPC differentiation are not known and require fur-
ther exploration. FABP7 has high binding affinities to doco-
sahexaenoic acid (DHA, 22:6(n3)), α-linolenic acid (LA, 18:2(n6)),
and eicosapentaenoic acid (EPA, 20:5(n3); Balendiran et al., 2000),
thereby playing a central role in the intracellular transport of these
FAs to various cellular organelles. In astrocytes, FABP7 can bind to
PPAR-γ(Tripathi et al., 2017) and modulate ERK phosphorylation
(Yasumoto et al., 2018), both pathways involved in OPC maturation
and differentiation (Fyffe-Maricich, Karlo, Landreth, & Miller, 2011;
Saluja, Granneman, & Skoff, 2001). However, whether the same path-
ways are employed in oligodendrocyte lineage cells to regulate their
differentiation potential remains to be addressed.
4.2 |FABP7 is dispensable for OPC differentiation
in vivo
In Fabp7KO animals, developmental myelination was delayed at P7,
but oligodendrocyte numbers recovered to physiological levels at P14
in the spinal cord (Figure 3). This delay in developmental myelination
might be caused by a direct effect on the OPCs as suggested by the
in vitro data (Figure 2). However, as the major FABP7 expressing cell
type in the CNS are astrocytes it is also feasible that an indirect effect
of the Fabp7KO in astrocytes attenuates OPC differentiation. Indeed,
mice lacking connexin 47 and 30, preventing the coupling of astrocytes
to oligodendrocytes, leads to a transient reduction in the number of
oligodendrocytes and thinner myelin sheaths (Tress et al., 2012).
In the demyelination model, however, we did not observe a simi-
lar delay of OPC differentiation in Fabp7KO animals (Figure 5). While
FOERSTER ET AL.9
it is possible that a potential delay in OPCs differentiation in response
to demyelination was not detected due to the time point chosen for
analysis (14 and 21 days post lesion, Figure 5), there might also be
differences in the FA transport between developmental myelination
and remyelination. For example, changes in the lipid composition in
the myelin sheath formed in remyelination have been reported
(Wilson & Tocher, 1991), that might render FABP7 dispensable for
remyelination.
Nevertheless, regardless of whether there is an undetected delay
in OPC differentiation after demyelination, the absence of FABP7
does not have a long-term effect on either developmental myelination
or remyelination in vivo. A reason for its dispensability could be a
compensatory mechanism in which other FABP isoforms, also physio-
logically expressed in the brain, are upregulated. However, no increase
in FABP3/5 expression in response to Fabp7 knockout has been
observed in development and early adulthood (Owada et al., 2006),
rendering the compensation by other FABP isoforms unlikely. As long
chain FAs would aggregate in the cytoplasm, an alternative FA trans-
port pathway must exist in oligodendrocytes. Elucidating these FA
transport pathways in oligodendrocytes could provide new therapeu-
tic targets to enhance OPC differentiation as FAs are crucial for the
production of many myelin sheath components.
ACKNOWLEDGMENTS
This work was supported by grants from the UK Multiple Sclerosis
Society, the Adelson Medical Research Foundation, the Japan Society
for the Promotion of Science (JSPS) KAKENHI Grant and a core sup-
port grant from the Wellcome Trust and M.R.C. to the Wellcome-
Medical Research Council Cambridge Stem Cell Institute. A.G.F. was
also supported by an ECTRIMS postdoctoral fellowship from July
2018. S.F. and T.B. were also supported by a Wellcome-Trust PhD
studentship.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
DATA AVAILABILITY STATEMENT
The data supporting the findings of this study are available from the
corresponding author upon request.
ORCID
Sarah Foerster https://orcid.org/0000-0002-2585-0621
Robin J. M. Franklin https://orcid.org/0000-0001-6522-2104
REFERENCES
Balendiran, G. K., Schnütgen, F., Scapin, G., Borchers, T., Xhong, N.,
Lim, K., Sacchettini J. C. (2000). Crystal structure and thermody-
namic analysis of human brain fatty acid-binding protein. The Journal
of Biological Chemistry,275(35), 2704527054 http://doi.org/10.
1074/jbc.M003001200
Bannerman, P., Hahn, A., Soulika, A., Gallo, V., & Pleasure, D. (2006).
Astrogliosis in EAE spinal cord: Derivation from radial glia, and rela-
tionships to oligodendroglia. Glia,55(1), 5764 http://doi.org/10.
1002/glia.20437
Cistola, D. P., Hamilton, J. A., Jackson, D., & Small, D. M. (1988). Ionization
and phase behavior of fatty acids in water: Application of the Gibbs
phase rule. Biochemistry,27(6), 18811888.
Driessen, T. M., Zhao, C., Saenz, M., Stevenson, S. A., Owada, Y., &
Gammie, S. C. (2018). Down-regulation of fatty acid binding protein
7 (Fabp7) is a hallmark of the postpartum brain. Journal of Chemical
Neuroanatomy,92,92101 http://doi.org/10.1016/j.jchemneu.2018.
07.003
Fancy, S. P. J., Baranzini, S. E., Zhao, C., Yuk, D.-I., Irvine, K.-A., Kaing, S.,
Rowitch D. H. (2009). Dysregulation of the Wnt pathway inhibits
timely myelination and remyelination in the mammalian CNS. Genes &
Development,23(13), 15711585 http://doi.org/10.1101/gad.
1806309
Franklin, R. J. M., & ffrench-Constant, C. (2017). Regenerating CNS
myelinFrom mechanisms to experimental medicines. Nature
Reviews. Neuroscience,18(12), 753769 http://doi.org/10.1038/nrn.
2017.136
Fyffe-Maricich, S. L., Karlo, J. C., Landreth, G. E., & Miller, R. H. (2011). The
ERK2 mitogen-activated protein kinase regulates the timing of oligo-
dendrocyte differentiation. The Journal of Neuroscience: The Official
Journal of the Society for Neuroscience,31(3), 843850 http://doi.org/
10.1523/JNEUROSCI.3239-10.2011
Huang, J. K., Jarjour, A. A., Oumesmar, B. N., Kerninon, C., Williams, A.,
Krezel, W., Franklin R. J. M. (2011). Retinoid X receptor gamma sig-
naling accelerates CNS remyelination. Nature Neuroscience,14(1),
4553 http://doi.org/10.1038/nn.2702
Kipp, M., Clarner, T., Gingele, S., Pott, F., Amor, S., van der Valk, P., &
Beyer, C. (2011). Brain lipid binding protein (FABP7) as modulator of
astrocyte function. Physiological Research,60(Suppl 1), S49S60.
McCarthy, K. D., & de Vellis, J. (1980). Preparation of separate astroglial
and oligodendroglial cell cultures from rat cerebral tissue. The Journal
of Cell Biology,85(3), 890902 http://doi.org/10.1083/jcb.85.3.890
Neumann, B., Baror, R., Zhao, C., Segel, M., Dietmann, S., Rawji, K. S.,
Franklin R. J. M. (2019). Metformin Restores CNS Remyelination
Capacity by Rejuvenating Aged Stem Cells. Cell Stem Cell,25(4),
473485.e8 http://doi.org/10.1016/j.stem.2019.08.015
Norton, W. T., & Poduslo, S. E. (1973). Myelination in rat brain: Changes
in myelin composition during brain maturation. Journal of Neurochemistry,
21(4), 759773 http://doi.org/10.1111/j.1471-4159.1973.tb07520.x
Owada, Y., Abdelwahab, S. A., Kitanaka, N., Sakagami, H., Takano, H.,
Sugitani, Y., Kondo, H. (2006). Altered emotional behavioral
responses in mice lacking brain-type fatty acid-binding protein gene.
The European Journal of Neuroscience,24(1), 175187 http://doi.org/
10.1111/j.1460-9568.2006.04855.x
Owada, Y., Yoshimoto, T., & Kondo, H. (1996). Spatio-temporally differen-
tial expression of genes for three members of fatty acid binding pro-
teins in developing and mature rat brains. Journal of Chemical
Neuroanatomy,12(2), 113122.
Saluja, I., Granneman, J. G., & Skoff, R. P. (2001). PPAR delta agonists stim-
ulate oligodendrocyte differentiation in tissue culture. Glia,33(3),
191204.
Segel, M., Neumann, B., Hill, M. F. E., Weber, I. P., Viscomi, C., Zhao, C.,
Chalut, K. J. (2019). Niche stiffness underlies the ageing of central ner-
vous system progenitor cells. Nature,573(7772), 130134 http://doi.
org/10.1038/s41586-019-1484-9
Sharifi, K., Ebrahimi, M., Kagawa, Y., Islam, A., Tuerxun, T., Yasumoto, Y.,
Owada, Y. (2013). Differential expression and regulatory roles of FABP5
and FABP7 in oligodendrocyte lineage cells. Cell and Tissue Research,
354(3), 683695 http://doi.org/10.1007/s00441-013-1730-7
Simons, M., & Nave, K.-A. (2015). Oligodendrocytes: Myelination and Axo-
nal Support. Cold Spring Harbor Perspectives in Biology,8(1), a020479
http://doi.org/10.1101/cshperspect.a020479
Tress, O., Maglione, M., May, D., Pivneva, T., Richter, N., Seyfarth, J.,
Willecke, K. (2012). Panglial gap junctional communication is essential
for maintenance of myelin in the CNS. The Journal of Neuroscience: The
10 FOERSTER ET AL.
Official Journal of the Society for Neuroscience,32(22), 74997518
http://doi.org/10.1523/JNEUROSCI.0392-12.2012
Tripathi, S., Kushwaha, R., Mishra, J., Gupta, M. K., Kumar, H., Sanyal, S.,
Bandyopadhyay, S. (2017). Docosahexaenoic acid up-regulates both
PI3K/AKT-dependent FABP7-PPARγinteraction and MKP3 that
enhance GFAP in developing rat brain astrocytes. Journal of Neuro-
chemistry,140(1), 96113 http://doi.org/10.1111/jnc.13879
Wilson, R., & Tocher, D. R. (1991). Lipid and fatty acid composition is
altered in plaque tissue from multiple sclerosis brain compared with
normal brain white matter. Lipids,26(1), 915.
Yasumoto, Y., Miyazaki, H., Ogata, M., Kagawa, Y., Yamamoto, Y., Islam, A.,
Owada, Y. (2018). Glial fatty acid-binding protein 7 (FABP7) regu-
lates neuronal leptin sensitivity in the hypothalamic arcuate nucleus.
Molecular Neurobiology,491(7424), 357313 http://doi.org/10.1007/
s12035-018-1033-9
SUPPORTING INFORMATION
Additional supporting information may be found online in the
Supporting Information section at the end of this article.
How to cite this article: Foerster S, Guzman de la Fuente A,
Kagawa Y, Bartels T, Owada Y, Franklin RJM. The fatty acid
binding protein FABP7 is required for optimal oligodendrocyte
differentiation during myelination but not during
remyelination. Glia. 2020;111. https://doi.org/10.1002/glia.
23789
FOERSTER ET AL.11
... FABP7 has been implicated in neurogenesis and neuroprotection (Foerster et al., 2020;Islam et al., 2019). We investigated its potential involvement in the protective role of SCFAs against hypoxia-reperfusion injury. ...
Preprint
Full-text available
Gut microbe-derived short-chain fatty acids (SCFAs) are known to have a profound impact on various brain functions, including cognition, mood, and overall neurological health. However, their role, if any, in protecting against hypoxic injury and ischemic stroke has not been extensively studied. In this study, we investigated the effects of two major SCFAs abundant in the gut, propionate (P) and butyrate (B), on hypoxia-reperfusion injury using a neuronal cell line and a zebrafish model. Neuro 2a (N2a) cells treated with P and B exhibited reduced levels of mitochondrial and cytosolic reactive oxygen species (ROS), diminished loss of mitochondrial membrane potential, suppressed caspase activation, and lower rates of cell death when exposed to CoCl 2 -induced hypoxia, compared to the control group. Furthermore, adult zebrafish fed with SCFAs-supplemented feeds showed less susceptibility to hypoxic conditions compared to the control group, as indicated by multiple behavioral measures. Histological analysis of TTC-stained brain sections revealed lesser damage in the SCFAs-fed group. We also found that FABP7 (also known as BLBP), a neuroprotective fatty acid binding protein, was upregulated in the brains of the SCFAs-fed group. Additionally, when FABP7 was overexpressed in N2a cells, it protected the cells from hypoxia-reperfusion injury. Overall, our data clearly demonstrates a neuroprotective role of P and B against hypoxic brain injury and suggests the potential of dietary supplementation with SCFAs to mitigate stroke-induced brain damage. Highlights Short-chain fatty acid (SCFA) Propionate (P) and Butyrate (B) protect N2a cells from hypoxia-reperfusion. Zebrafish, when fed an SCFA-supplemented diet, are more resilient to hypoxia-reperfusion. SCFAs in the diet boost brain expression of FABP7 (fatty acid binding protein). FABP7 overexpression in N2a cells provides protection against hypoxia-reperfusion. SCFAs reduce reactive oxygen species (ROS) levels and increase FABP7, contributing to neuroprotection.
... Lipids, specifically fatty acids (FAs), are the main components involved in the formation of the myelin sheath. Fatty acid-binding proteins (FABPs) are a class of 14-15 kDa proteins that prevent FA aggregation and facilitate cellular transportation of long-chain FAs, allowing them to perform their functions within different cell compartments [28]. Astrocyte-expressed FABP7 controls FA uptake and transportation, signal transduction, gene transcription, and plays a crucial role in neurogenesis involving the formation of a radial fiber scaffold in the developing brain [29]. ...
Article
Full-text available
Embolization is the preferred method for treating intracranial aneurysms due to its less invasive nature. However, recent findings suggest that even uncomplicated embolization may cause structural damage to the brain through ischemic or inflammatory mechanisms. This study aimed to find possible biomarkers of brain injury and inflammation in patients suffering from intracranial aneurysms who underwent endovascular treatment by measuring serological markers indicating brain damage. The study involved 26 patients who underwent uncomplicated intravascular stenting for unruptured intracranial aneurysms between January 2020 and December 2021. Blood samples were collected before the procedure, at 6–12 h, and at 24 h after the procedure. The following protein biomarkers levels were tested with ELISA: S100B, hNSE, TNF, hsCRP, FABP7, NFL, and GP39. Statistical analysis of the results revealed significant increases in serum levels for the four biomarkers: FABP7—before 0.25 (ng/mL) vs. 6–12 h 0.26 (p = 0.012) and vs. 24 h 0.27 (p < 0.001); GP39—before 0.03 (pg/mL) vs. 6–12 h 0.64 (p = 0.011) and vs. 24 h 0.57 (p = 0.001); hsCRP—before 1.65 (μg/mL) vs. 24 h 4.17 (p = 0.037); NFL—before 0.01 (pg/mL) vs. 6–12 h 3.99 (p = 0.004) and vs. 24 h 1.86 (p = 0.033). These biomarkers are recognized as potential indicators of neurovascular damage and should be monitored in clinical settings. Consequently, serum levels of NFL, GP39, hsCRP, and FABP7 measured before and 24 h after endovascular procedures can serve as important markers for assessing brain damage and indicate avenues for further research on biomarkers of neurovascular injury.
... In both the neocortex and the hippocampus and at both time points examined, FABP7, a protein involved in radial glial cell function, and GFAP, a cytoskeletal protein in astrocytes, were found to be highly upregulated in the Bmal1 CKO. GFAP is a well-established astrocyte marker (Middeldorp & Hol, 2011), while FABP7 in addition to oligodendrocyte precursor cells is also expressed in astrocytes of the adult rodent brain (Foerster et al., 2020;Schmid et al., 2006;Sharifi et al., 2011;Sharifi et al., 2013), thus suggesting that astrocytes may be heavily affected by the knockout of the circadian clock. On the other hand, mitochondrial proteins (MTHFD1L and ACAA2) were generally downregulated in the neocortex and hippocampus of the Bmal1 CKO. ...
Article
Circadian oscillators, defined by cellular 24 h clock gene rhythms, are found throughout the brain. Cerebral cortex-specific conditional knockout of the clock gene Bmal1 (Bmal1 CKO) leads to depressive-like behavior, but the molecular link from clock gene to altered behavior is unknown. Further, diurnal proteomic data on the cerebral cortex are currently unavailable. With the aim of determining the diurnal proteome profile and downstream targets of the cortical circadian clock, we here performed a proteomic analysis of the mouse cerebral cortex. Proteomics identified approximately 2700 proteins in both the neocortex and the hippocampus. In the neocortex, 15 proteins were differentially expressed (>2-fold) between day and night, mainly mitochondrial and neuronal plasticity proteins. Only three hippocampal proteins were differentially expressed, suggesting that daily protein oscillations are more prominent in the neocortex. The number of differentially expressed proteins was reduced in the Bmal1 CKO, suggesting that daily rhythms in the cerebral cortex are primarily driven by local clocks. The proteome of the Bmal1 CKO cerebral cortex was dominated by upregulated proteins expressed in astrocytes, including GFAP (4-fold) and FABP7 (>20-fold), in both the neocortex and hippocampus. These findings were confirmed at the transcript level. Cellular analyses of astrocyte components revealed an increased number of GFAP-positive cells in the Bmal1 CKO cerebral cortex. Further, BMAL1 was found to be expressed in both GFAP- and FABP7-positive astrocytes of control animals. Our data show that Bmal1 is required for proper cellular composition of the cerebral cortex, suggesting that increased cortical astrocyte activity may induce behavioral changes.
... To better understand how OPCs may be affected, we further analyzed omni-ATAC-seq analyses in wild type and linc-mipep;linc-wrb mutant brains. These analyses revealed differentially accessible regions downstream of olig2 (a transcription factor that activates the expression of myelin-associated genes), within a large intronic span of sgms2b (which synthesizes a component of myelin sheath), and upstream of fabp7a (which is important for OPC differentiation in vitro in mouse) (Foerster et al., 2020;Figure 4-figure supplement 6A-C). To validate that OPCs are affected in vivo, we found a significant 13% decrease (p=0.0053) in olig2 + oligodendrocyte progenitor cells' signal in mutant brains compared to WT brains, with most of the loss coming from the optic tectum and the cerebellum of Tg(olig2:eGFP); linc-mipep -/-; linc-wrb -/compared to control larvae ( Figure 4G; Figure 4-figure supplement 4A). ...
Article
Full-text available
Thousands of long intergenic non-coding RNAs (lincRNAs) are transcribed throughout the vertebrate genome. A subset of lincRNAs enriched in developing brains have recently been found to contain cryptic open-reading frames and are speculated to encode micropeptides. However, systematic identification and functional assessment of these transcripts have been hindered by technical challenges caused by their small size. Here, we show that two putative lincRNAs (linc-mipep, also called lnc-rps25, and linc-wrb) encode micropeptides with homology to the vertebrate-specific chromatin architectural protein, Hmgn1, and demonstrate that they are required for development of vertebrate-specific brain cell types. Specifically, we show that NMDA receptor-mediated pathways are dysregulated in zebrafish lacking these micropeptides and that their loss preferentially alters the gene regulatory networks that establish cerebellar cells and oligodendrocytes - evolutionarily newer cell types that develop postnatally in humans. These findings reveal a key missing link in the evolution of vertebrate brain cell development and illustrate a genetic basis for how some neural cell types are more susceptible to chromatin disruptions, with implications for neurodevelopmental disorders and disease.
... On the one hand, some molecules are more involved in initial myelination than in remyelination. For example, a recent study shows that fatty acid-binding protein 7 (FABP7) is important in OPCs differentiation during development but not in remyelination (Foerster et al., 2020). On the other hand, some mechanisms are, at least for now, remyelination-specific. ...
Article
Full-text available
Myelination contributes not only to the rapid nerve conduction but also to axonal insulation and protection. In the central nervous system (CNS), the initial myelination features a multistep process where oligodendrocyte precursor cells undergo proliferation and migration before differentiating into mature oligodendrocytes. Mature oligodendrocytes then extend processes and wrap around axons to form the multilayered myelin sheath. These steps are tightly regulated by various cellular and molecular mechanisms, such as transcription factors (Olig family, Sox family), growth factors (PDGF, BDNF, FGF-2, IGF), chemokines/cytokines (TGF-β, IL-1β, TNFα, IL-6, IFN-γ), hormones (T3), axonal signals (PSA-NCAM, L1-CAM, LINGO-1, neural activity), and intracellular signaling pathways (Wnt/β-catenin, PI3 K/AKT/mTOR, ERK/MAPK). However, the fundamental mechanisms for initial myelination are yet to be fully elucidated. Identifying pivotal mechanisms for myelination onset, development, and repair will become the focus of future studies. This review focuses on the current understanding of how CNS myelination is initiated and also the regulatory mechanisms underlying the process.
Article
Polyunsaturated fatty acids (PUFAs) are essential for brain development and function, and an imbalance of brain PUFAs is linked to mental disorders like autism and schizophrenia. However, the cellular and molecular mechanisms underlying the effects of PUFAs on the brain remain largely unknown. Since they are insoluble in water, specific transporters like fatty acid binding proteins (FABPs), are required for transport and function of PUFAs within cells. We focused on the relationship between FABP-mediated homeostasis of brain PUFAs and neural plasticity. We found that FABP3, with a high affinity for n-6 PUFAs, is predominantly expressed in the GABAergic inhibitory interneurons of the anterior cingulate cortex (ACC) in the adult mouse brain. FABP3 knockout (KO) mice show increased GABA synthesis and inhibitory synaptic transmission in the ACC. We also found that FABP7 controls lipid raft function in astrocytes, and astrocytes lacking FABP7 exhibit changes in response to external stimuli. Furthermore, in FABP7 KO mice, dendritic protrusion formation in pyramidal neurons becomes abnormal, and we have reported a decrease in spine density and excitatory synaptic transmission. Here, we introduced recent advances in the understanding of the functions of PUFAs and FABPs in the brain, focusing especially on FABP3 and FABP7, in relation to human mental disorders.
Article
Background: Farnesol (FOL) prevents the onset of experimental autoimmune encephalitis (EAE), a murine model of multiple sclerosis (MS). Objective: We examined the transcriptomic profile of the brains of EAE mice treated with daily oral FOL using next-generation sequencing (RNA-seq). Methods: Transcriptomics from whole brains of treated and untreated EAE mice at the peak of EAE was performed. Results: EAE-induced mice, compared to naïve, healthy mice, overall showed increased expression in pathways for immune response, as well as an increased cytokine signaling pathway, with downregulation of cellular stress proteins. FOL downregulates pro-inflammatory pathways and attenuates the immune response in EAE. FOL downregulated the expression of genes involved in misfolded protein response, MAPK activation/signaling, and pro-inflammatory response. Conclusion: This study provides insight into the molecular impact of FOL in the brain and identifies potential therapeutic targets of the isoprenoid pathway in MS patients.
Article
Fatty acid binding proteins (FABPs) are key proteins in lipid transport, and 12 family members have been documented in the literature. In recent years, new insights have been gained into the structure and function of FABPs, which are important regulators of lipid metabolic processes in the body and play a central role in coordinating lipid transport and metabolism in various tissues and organs across species. This paper provides a brief overview of the structure and biological functions of FABPs and reviews related studies on lipid metabolism in livestock and poultry to lay the foundation for research on the mechanism underlying the regulatory effect of FABPs on lipid metabolism in livestock and poultry and for the genetic improvement of livestock and poultry.
Article
Full-text available
In the developing central nervous system, oligodendrocyte precursor cells (OPCs) differentiate into oligodendrocytes, which form myelin around axons. Oligodendrocytes and myelin are essential for the function of the central nervous system, as evidenced by the severe neurological symptoms that arise in demyelinating diseases such as multiple sclerosis and leukodystrophy. Although many cell-intrinsic mechanisms that regulate oligodendrocyte development and myelination have been reported, it remains unclear whether interactions among oligodendrocyte-lineage cells (OPCs and oligodendrocytes) affect oligodendrocyte development and myelination. Here, we show that blocking vesicle-associated membrane protein (VAMP) 1/2/3-dependent exocytosis from oligodendrocyte-lineage cells impairs oligodendrocyte development, myelination, and motor behavior in mice. Adding oligodendrocyte-lineage cell-secreted molecules to secretion-deficient OPC cultures partially restores the morphological maturation of oligodendrocytes. Moreover, we identified L-type prostaglandin D synthase as an oligodendrocyte-lineage cell-secreted protein that promotes oligodendrocyte development and myelination in vivo. These findings reveal a novel autocrine/paracrine loop model for the regulation of oligodendrocyte and myelin development.
Article
Full-text available
Expression of brain fatty acid-binding protein (B-FABP) is spatially and temporally correlated with neuronal differentiation during brain development. Isothermal titration calorimetry demonstrates that recombinant human B-FABP clearly exhibits high affinity for the polyunsaturated n-3 fatty acids α-linolenic acid, eicosapentaenoic acid, docosahexaenoic acid, and for monounsaturated n-9 oleic acid (K d from 28 to 53 nm) over polyunsaturated n-6 fatty acids, linoleic acid, and arachidonic acid (K d from 115 to 206 nm). B-FABP has low binding affinity for saturated long chain fatty acids. The three-dimensional structure of recombinant human B-FABP in complex with oleic acid shows that the oleic acid hydrocarbon tail assumes a “U-shaped” conformation, whereas in the complex with docosahexaenoic acid the hydrocarbon tail adopts a helical conformation. A comparison of the three-dimensional structures and binding properties of human B-FABP with other homologous FABPs, indicates that the binding specificity is in part the result of nonconserved amino acid Phe¹⁰⁴, which interacts with double bonds present in the lipid hydrocarbon tail. In this context, analysis of the primary and tertiary structures of human B-FABP provides a rationale for its high affinity and specificity for polyunsaturated fatty acids. The expression of B-FABP in glial cells and its high affinity for docosahexaenoic acid, which is known to be an important component of neuronal membranes, points toward a role for B-FABP in supplying brain abundant fatty acids to the developing neuron.
Article
Full-text available
These guidelines are a consensus work of a considerable number of members of the immunology and flow cytometry community. They provide the theory and key practical aspects of flow cytometry enabling immunologists to avoid the common errors that often undermine immunological data. Notably, there are comprehensive sections of all major immune cell types with helpful Tables detailing phenotypes in murine and human cells. The latest flow cytometry techniques and applications are also described, featuring examples of the data that can be generated and, importantly, how the data can be analysed. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid, all written and peer‐reviewed by leading experts in the field, making this an essential research companion.
Article
Full-text available
The age-related failure to produce oligodendrocytes from oligodendrocyte progenitor cells (OPCs) is associated with irreversible neurodegeneration in multiple sclerosis (MS). Consequently, regenerative approaches have significant potential for treating chronic demyelinating diseases. Here, we show that the differentiation potential of adult rodent OPCs decreases with age. Aged OPCs become unresponsive to pro-differentiation signals, suggesting intrinsic constraints on therapeutic approaches aimed at enhancing OPC differentiation. This decline in functional capacity is associated with hallmarks of cellular aging, including decreased metabolic function and increased DNA damage. Fasting or treatment with metformin can reverse these changes and restore the regenerative capacity of aged OPCs, improving remyelination in aged animals following focal demyelination. Aged OPCs treated with metformin regain responsiveness to pro-differentiation signals, suggesting synergistic effects of rejuvenation and pro-differentiation therapies. These findings provide insight into aging-associated remyelination failure and suggest therapeutic interventions for reversing such declines in chronic disease.
Article
Full-text available
Ageing causes a decline in tissue regeneration owing to a loss of function of adult stem cell and progenitor cell populations¹. One example is the deterioration of the regenerative capacity of the widespread and abundant population of central nervous system (CNS) multipotent stem cells known as oligodendrocyte progenitor cells (OPCs)². A relatively overlooked potential source of this loss of function is the stem cell ‘niche’—a set of cell-extrinsic cues that include chemical and mechanical signals3,4. Here we show that the OPC microenvironment stiffens with age, and that this mechanical change is sufficient to cause age-related loss of function of OPCs. Using biological and synthetic scaffolds to mimic the stiffness of young brains, we find that isolated aged OPCs cultured on these scaffolds are molecularly and functionally rejuvenated. When we disrupt mechanical signalling, the proliferation and differentiation rates of OPCs are increased. We identify the mechanoresponsive ion channel PIEZO1 as a key mediator of OPC mechanical signalling. Inhibiting PIEZO1 overrides mechanical signals in vivo and allows OPCs to maintain activity in the ageing CNS. We also show that PIEZO1 is important in regulating cell number during CNS development. Thus we show that tissue stiffness is a crucial regulator of ageing in OPCs, and provide insights into how the function of adult stem and progenitor cells changes with age. Our findings could be important not only for the development of regenerative therapies, but also for understanding the ageing process itself.
Article
Full-text available
The hypothalamus is involved in the regulation of food intake and energy homeostasis. The arcuate nucleus (ARC) and median eminence (ME) are the primary hypothalamic sites that sense leptin and nutrients in the blood, thereby mediating food intake. Recently, studies demonstrating a role for non-neuronal cell types, including astrocytes and tanycytes, in these regulatory processes have begun to emerge. However, the molecular mechanisms involved in these activities remain largely unknown. In this study, we examined in detail the localization of fatty acid-binding protein 7 (FABP7) in the hypothalamic ARC and sought to determine its role in the hypothalamus. We performed a phenotypic analysis of diet-induced FABP7 knockout (KO) obese mice and of FABP7 KO mice treated with a single leptin injection. Immunohistochemistry revealed that FABP7⁺ cells are NG2⁺ or GFAP⁺ in the ARC and ME. In mice fed a high-fat diet, weight gain and food intake were lower in FABP7 KO mice than in wild-type (WT) mice. FABP7 KO mice also had lower food intake and weight gain after a single injection of leptin, and we consistently confirmed that the number of pSTAT3⁺ cells in the ARC indicated that the leptin-induced activation of neurons was significantly more frequent in FABP7 KO mice than in WT mice. In FABP7 KO mice-derived primary astrocyte cultures, the level of ERK phosphorylation was lower after leptin treatment. Collectively, these results indicate that in hypothalamic astrocytes, FABP7 might be involved in sensing neuronal leptin via glia-mediated mechanisms and plays a pivotal role in controlling systemic energy homeostasis.
Article
Full-text available
Myelinated nerve fibers have evolved to enable fast and efficient transduction of electrical signals in the nervous system. To act as an electric insulator, the myelin sheath is formed as a multilamellar membrane structure by the spiral wrapping and subsequent compaction of the oligodendroglial plasma membrane around central nervous system (CNS) axons. Current evidence indicates that the myelin sheath is more than an inert insulating membrane structure. Oligodendrocytes are metabolically active and functionally connected to the subjacent axon via cytoplasmic-rich myelinic channels for movement of macromolecules to and from the internodal periaxonal space under the myelin sheath. This review summarizes our current understanding of how myelin is generated and also the role of oligodendrocytes in supporting the long-term integrity of myelinated axons. Copyright © 2015 Cold Spring Harbor Laboratory Press; all rights reserved.
Article
Fatty acid binding protein 7 (Fabp7) is a versatile protein that is linked to glial differentiation and proliferation, neurogenesis, and multiple mental health disorders. Recent microarray studies identified a robust decrease in Fabp7 expression in key brain regions of the postpartum rodents. Given its diverse functions, Fabp7 could play a critical role in sculpting the maternal brain and promoting the maternal phenotype. The present study aimed at investigating the expression profile of Fabp7 across the postpartum CNS. Quantitative real-time PCR (qPCR) analysis showed that Fabp7 mRNA was consistently down-regulated across the postpartum brain. Of the 9 maternal care-related regions tested, seven exhibited significant decreases in Fabp7 in postpartum (relative to virgin) females, including medial prefrontal cortex (mPFC), nucleus accumbens (NA), lateral septum (LS), bed nucleus of stria terminalis dorsal (BnSTd), paraventricular nucleus (PVN), lateral hypothalamus (LH), and basolateral and central amygdala (BLA/CeA). For both ventral tegmental area (VTA) and medial preoptic area (MPOA) levels of Fabp7 were lower in mothers, but levels of changes did not reach significance. Confocal microscopy revealed that protein expression of Fabp7 in the LS paralleled mRNA findings. Specifically, the caudal LS exhibited a significant reduction in Fabp7 immunoreactivity, while decreases in medial LS were just above significance. Double fluorescent immunolabeling confirmed the astrocytic phenotype of Fabp7-expressing cells. Collectively, this research demonstrates a broad and marked reduction in Fabp7 expression in the postpartum brain, suggesting that down-regulation of Fabp7 may serve as a hallmark of the postpartum brain and contribute to the maternal phenotype.
Article
Although the core concept of remyelination — based on the activation, migration, proliferation and differentiation of CNS progenitors — has not changed over the past 20 years, our understanding of the detailed mechanisms that underlie this process has developed considerably. We can now decorate the central events of remyelination with a host of pathways, molecules, mediators and cells, revealing a complex and precisely orchestrated process. These advances have led to recent drug-based and cell-based clinical trials for myelin diseases and have opened up hitherto unrecognized opportunities for drug-based approaches to therapeutically enhance remyelination.
Article
The astrocyte marker, glial fibrillary acidic protein (GFAP), has essential functions in the brain, but may trigger astroglial scarring when expressed in excess. Docosahexaenoic acid (DHA) is an n-3 fatty acid that is protective during brain development. However, the effect of DHA on GFAP levels of developing brain remains unexplored. Here we detected that treating developing rats with DHA-enriched fish-oil caused dose-dependent GFAP augmentation. We investigated the mechanism promoting GFAP, hypothesizing the participation of fatty acid-binding protein-7 (FABP7), known to bind DHA. We identified that DHA stimulated FABP7 expression in astrocytes, and FABP7-silencing suppressed DHA-induced GFAP, indicating FABP7-mediated GFAP increase. Further investigation proved FABP7 expression to be phosphatidylinositide 3-kinases (PI3K)/AKT and nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARγ)-dependent. We found that PI3K/AKT activated PPARγ that triggered F ABP7 expression via PPARγ-responsive elements within its gene. Towards identifying FABP7-downstream pathways, we considered our previous report that demonstrated cyclin-dependent kinase-5 (CDK5)-PPARγ-protein/protein complex to suppress GFAP. We found that the DHA-induced FABP7 underwent protein/protein interaction with PPARγ, which impeded CDK5-PPARγ formation. Hence, it appeared that enhanced FABP7-PPARγ in lieu of CDK5-PPARγ resulted in increased GFAP.PI3K/AKT not only stimulated formation of FABP7-PPARγ protein/protein-complex, but also up-regulated a FABP7-independent MAP-kinase-phosphatase-3 (MKP3) pathway that inactivated CDK5 and hence attenuated CDK5-PPARγ. Overall, our data reveal that via the proximal PI3K/AKT, DHA induces FABP7-PPARγ, through genomic and non-genomic mechanisms, and MKP3 that converged at attenuated CDK5-PPARγ and therefore, enhanced GFAP. Accordingly, our study demonstrates a DHA-mediated astroglial hyperactivation, pointing towards a probable injurious role of DHA in brain development. This article is protected by copyright. All rights reserved.