ArticlePDF Available

Amelioration of functional and histopathological consequences after spinal cord injury through phosphodiesterase 4D (PDE4D) inhibition

Authors:

Abstract

Spinal cord injury (SCI) is a life-changing event that severely impacts the patient's quality of life. Modulating neuroinflammation, which exacerbates the primary injury, and stimulating neuro-regenerative repair mechanisms are key strategies to improve functional recovery. Cyclic adenosine monophosphate (cAMP) is a second messenger crucially involved in both processes. Following SCI, intracellular levels of cAMP are known to decrease over time. Therefore, preventing cAMP degradation represents a promising strategy to suppress inflammation while stimulating regeneration. Intracellular cAMP levels are controlled by its hydrolyzing enzymes phosphodiesterases (PDEs). The PDE4 family is most abundantly expressed in the central nervous system (CNS) and its inhibition has been shown to be therapeutically relevant for managing SCI pathology. Unfortunately, the use of full PDE4 inhibitors at therapeutic doses is associated with severe emetic side effects, hampering their translation toward clinical applications. Therefore, in this study, we evaluated the effect of inhibiting specific PDE4 subtypes (PDE4B and PDE4D) on inflammatory and regenerative processes following SCI, as inhibitors selective for these subtypes have been demonstrated to be well-tolerated. We reveal that administration of the PDE4D inhibitor Gebr32a, even when starting 2 dpi, but not the PDE4B inhibitor A33, improved functional as well as histopathological outcomes after SCI, comparable to results obtained with the full PDE4 inhibitor roflumilast. Furthermore, using a luminescent human iPSC-derived neurospheroid model, we show that PDE4D inhibition stabilizes neural viability by preventing apoptosis and stimulating neuronal differentiation. These findings strongly suggest that specific PDE4D inhibition offers a novel therapeutic approach for SCI.
Original Article
Amelioration of functional and histopathological consequences after spinal
cord injury through phosphodiesterase 4D (PDE4D) inhibition
Melissa Schepers
a
,
b
,
c
,
1
, Sven Hendrix
d
,
1
, Femke Mussen
a
,
b
,
e
,
1
, Elise van Breedam
f
,
Peter Ponsaerts
f
, Stefanie Lemmens
e
, Niels Hellings
c
,
e
, Roberta Ricciarelli
g
,
h
, Ernesto Fedele
g
,
i
,
Olga Bruno
j
, Chiara Brullo
j
, Jos Prickaerts
k
, Jana Van Broeckhoven
c
,
e
,
2
, Tim Vanmierlo
a
,
b
,
c
,
*
,
2
a
Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
b
Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands
c
University MS Centre (UMSC) Hasselt Pelt, Belgium
d
Institute for Translational Medicine, Medical School Hamburg, 20457 Hamburg, Germany
e
Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
f
Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610 Wilrijk, Belgium
g
IRCCS Ospedale Policlinico San Martino, 16100 Genoa, Italy
h
Department of Experimental Medicine, Section of General Pathology, University of Genova, 16100 Genoa, Italy
i
Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, 16100 Genoa, Italy
j
Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, 16100 Genoa, Italy
k
Peitho Translational, 6229ER Maastricht, the Netherlands
ARTICLE INFO
Keywords:
Spinal cord injury
Phosphodiesterase 4
cAMP
Neuroinammation
Regeneration
ABSTRACT
Spinal cord injury (SCI) is a life-changing event that severely impacts the patient's quality of life. Modulating
neuroinammation, which exacerbates the primary injury, and stimulating neuro-regenerative repair mechanisms
are key strategies to improve functional recovery. Cyclic adenosine monophosphate (cAMP) is a second messenger
crucially involved in both processes. Following SCI, intracellular levels of cAMP are known to decrease over time.
Therefore, preventing cAMP degradation represents a promising strategy to suppress inammation while stim-
ulating regeneration. Intracellular cAMP levels are controlled by its hydrolyzing enzymes phosphodiesterases
(PDEs). The PDE4 family is most abundantly expressed in the central nervous system (CNS) and its inhibition has
been shown to be therapeutically relevant for managing SCI pathology. Unfortunately, the use of full PDE4 in-
hibitors at therapeutic doses is associated with severe emetic side effects, hampering their translation toward
clinical applications. Therefore, in this study, we evaluated the effect of inhibiting specic PDE4 subtypes (PDE4B
and PDE4D) on inammatory and regenerative processes following SCI, as inhibitors selective for these subtypes
have been demonstrated to be well-tolerated. We reveal that administration of the PDE4D inhibitor Gebr32a, even
when starting 2 dpi, but not the PDE4B inhibitor A33, improved functional as well as histopathological outcomes
after SCI, comparable to results obtained with the full PDE4 inhibitor roumilast. Furthermore, using a lumi-
nescent human iPSC-derived neurospheroid model, we show that PDE4D inhibition stabilizes neural viability by
preventing apoptosis and stimulating neuronal differentiation. These ndings strongly suggest that specic
PDE4D inhibition offers a novel therapeutic approach for SCI.
Introduction
Spinal cord injury (SCI) is characterized by a complex secondary
injury phase that drives further permanent damage and causes neuro-
logical dysfunction [1,2]. To date, regeneration and recovery of function
remain limited after SCI [3]. The provoked neuroinammation and the
limited endogenous regeneration potential of neural tissue are the critical
bottlenecks. Despite multiple efforts, current treatments suppress in-
ammatory processes (e.g. corticosteroids) but remain ineffective in
promoting repair. Therefore, there is an urgent need to develop new
* Corresponding author.
E-mail address: t.vanmierlo@maastrichtuniversity.nl (T. Vanmierlo).
1
Equally contributing rst authors.
2
Equally contributing last authors.
Contents lists available at ScienceDirect
Neurotherapeutics
journal homepage: www.sciencedirect.com/journal/neurotherapeutics
https://doi.org/10.1016/j.neurot.2024.e00372
Received 15 December 2023; Received in revised form 15 April 2024; Accepted 1 May 2024
1878-7479/©2024 The Author(s). Published by Elsevier Inc. on behalf of American Society for Experimental NeuroTherapeutics. This is an open access article under
the CC BY license (http://creativecommons.org/licenses/by/4.0/).
Neurotherapeutics 21 (2024) e00372
therapeutic strategies that tackle both neuroinammatory and regener-
ative processes.
Cyclic adenosine monophosphate (cAMP) is a crucial molecule
involved as a second messenger in multiple signaling pathways and ex-
erts broad modulatory effects in various cell types [4,5]. In the context of
central nervous system (CNS) injury, cAMP has been shown to exhibit
both anti-inammatory and neuroregenerative functions. Upon SCI,
cAMP levels in both neurons and glial cells decrease dramatically [6].
Therefore, maintaining or elevating the intracellular cAMP levels to
regulate the immune responses or to stimulate neuroregeneration can be
considered a valuable approach to temper SCI pathogenesis. Phospho-
diesterases (PDEs) are a class of enzymes responsible for the degradation
of cyclic nucleotides, such as cAMP. In the CNS, PDE4 is primarily
responsible for the breakdown of cAMP [4,7]. As such, PDE4 inhibition,
initiated after SCI induction, demonstrated a wide range of benecial
actions in a SCI mouse model [812]. The golden standard,
rst-generation PDE4 inhibitor rolipram was found to act as
anti-inammatory, neuroprotective, and regenerative agent [911].
Continuous and controlled mini-osmotic pump-mediated release of roli-
pram (0.4
μ
mol/kg/h) attenuated astrogliosis and enhanced axonal
outgrowth following hemisection SCI, while administration of a single
dose of 0.5 or 1 mg/kg rolipram per day appeared to be sufcient to
enhance neuronal survival and simultaneously protect oligodendrocytes,
thus preserving CNS myelination following contusion SCI [911]. Addi-
tionally, the PDE4 inhibitor IC486051 further conrmed the
anti-inammatory properties observed upon rolipram administration as
bolus doses of 0.5 or 1.0 mg/kg IC486051 decreased oxidative stress
markers and leukocyte inltration into the lesion size, thereby reducing
the resulting tissue damage following compression SCI in Wistar rats [8].
However, despite these promising ndings, the clinical translation of pan
PDE4 inhibitors has been hampered due to tolerability problems (e.g.,
emesis). Therefore, to mitigate the side effects and increase tolerability,
PDE4 subtype-selective inhibitors have been developed. The PDE4 family
consists of four genes yielding different PDE4 subtypes (PDE4A-PDE4D).
PDE4A, B, and D are widely expressed in the CNS of rodents and humans,
while PDE4C has limited CNS expression. In particular, the PDE4B and
PDE4D subtypes recently gained more interest in neurological disorders
because of their anti-inammatory and neuroplastic effects, respectively
[1315]. More specically, the inhibition of PDE4B suppresses the neu-
roinammatory responses of macrophages and microglia, while PDE4D
blockage has been shown to successfully boost myelin regeneration and
enhance neuroplasticity [1418]. Targeting these individual subtypes
circumvented the emetic side-effects accompanied by pan PDE4 in-
hibitors such as roumilast and rolipram, and is predicted as a valuable
strategy to target neuroinammation [19].
In this study, we aimed to disentangle the effect of PDE4B and PDE4D
inhibition on SCI pathology. Using the SCI hemisection model, we show
in mice that, in contrast to the PDE4B inhibitor A33, the PDE4D inhibitor
Gebr32a improved functional and histopathological outcomes after SCI
to a similar extent as the pan PDE4 inhibitor roumilast. Gebr32a could
still improve SCI outcomes even when administered 2 dpi. In vitro,
neuronal apoptosis was prevented by inhibiting PDE4D, as demonstrated
with primary neuronal mouse cultures, human iPSC-derived neuronal
precursor cultures, and luminescent iPSC-derived neurospheroids.
In addition, increased neuronal differentiation was observed in these
iPSC-derived neurospheroids. Overall, these ndings underline the
therapeutic potential of specic PDE4D inhibition to act neuroprotective
and consequently improve neural plasticity, leading to functional re-
covery after SCI.
Methods
Animals
In vivo experiments were performed with 10- to 12-week-old female
WT C57BL/6j mice (Janvier Labs). Mice were housed in groups at the
conventional animal facility of Hasselt University under stable conditions
(temperature-controlled room, 12 h light/dark cycle, food, and water ad
libitum). Experiments were approved by the local ethical committee of
Hasselt University (ethical ID 202060) and were performed according to
the guidelines of Directive 2010/63/EU on the protection of animals
used for scientic purposes.
Spinal cord injury model
A standardized T-cut spinal cord hemisection injury was performed as
previously described [2022]. In brief, mice were anesthetized with
23% isourane and a partial laminectomy was performed at thoracic
level 8. A bilateral hemisection injury was done using iridectomy scissors
to transect the dorsomedial and ventral corticospinal tract. Afterward,
the back muscles were sutured, and the skin was closed with wound clips
(Bioconnect). Post-operative treatment included blood-loss compensa-
tion by glucose solution (20%, intraperitoneal [i.p.]) and pain relief by
buprenorphine (0.1 mg/kg body weight, Temgesic, subcutaneous [s.c.]).
In addition, mice were placed in a heated recovery chamber (33 C) until
they regained consciousness. Bladders were voided daily until the
micturition reex was restored spontaneously. The in vivo experiments
were conducted in two independent cohorts. Table 1 provides a sample
size overview for each experimental animal group for both cohorts.
Animal treatments
Starting 1 h after SCI, mice were injected twice daily s.c. for 28 days
with either (1) vehicle (0.1% DMSO (VWR prolabo) þ0.5% methylcel-
lulose þ2% Tween80), (2) the pan PDE4 inhibitor roumilast (3 mg/kg,
Xi'an leader biochemical engineering co., LTD), (3) the PDE4B inhibitor
A33 (3 mg/kg, Sigma-Aldrich), or (4) the PDE4D inhibitor Gebr32a (0.3
mg/kg, University of Genoa [23]). We included (5) a sequential treat-
ment group who rst received A33 (3 mg/kg) until 10 days post injury
(dpi), followed by Gebr32a (0.3 mg/kg) until the end of the experiment
(injection volume: 10
μ
l/g body weight). This treatment was used to rst
diminish the acute inammatory responses with PDE4B inhibition in the
early phase, followed by PDE4D inhibition later to boost endogenous
repair mechanisms and hence, promote SCI recovery by tackling both the
inammatory and repair processes. Lastly, we elucidated the therapeutic
potential of Gebr32a when administered in a more clinically relevant
therapeutic window. To this purpose, mice were injected twice daily s.c.
for 28 days with (1) vehicle (0.1% DMSO) or Gebr32a (0.3 mg/kg)
starting from (2) 2 dpi or (3) 10 dpi onwards.
Locomotion test
Following SCI, functional recovery was assessed using the standard-
ized Basso Mouse Scale (BMS) score for locomotion [24]. This 10-point
scale ranges from 0, indicating complete hind limb paralysis, to 9, rep-
resenting normal motor function. These scores are based on hind limb
movements in an open eld during a 4 min testing window. The
Table 1
Overview of the number of animals receiving a hemisection SCI which were
included in cohort 1 and cohort 2 for functional and histopathological analysis
based on locomotor function. The A33 and Gebr32a sequential treatment group
was not included in cohort 2 due to the lack of additional efcacy compared to
continuous PDE4D inhibition.
Treatment group Number of animals
Included cohort 1 Included cohort 2 Total
Vehicle 6 9 15
Roumilast (3 mg/kg) 7 7 14
A33 (3 mg/kg) 6 10 16
Gebr32a (0.3 mg/kg) 8 6 14
A33 followed by Gebr32a 10 / 10
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
2
evaluation was done by an investigator blinded to treatment groups and
was performed daily from 1 until 7 dpi, followed by a scoring every 23
days until the end of the experiment (28 dpi). The mean BMS score of the
left and right hind limb was used per animal. Mice were excluded from
the analysis if 1) they had a BMS score higher than 1 at 1 dpi or 2) they
did not show an increase in BMS score of at least 1 point at 28 dpi.
Mouse primary neuronal cultures
Fetal mice brains (E16-19) were used to obtain primary cortical
neuron cultures [25]. Meninges-free cerebral cortices were chemically
dissociated for 15 min using trypsin. Next, chemical dissociation was
stopped by washing cortices with minimal essential medium (MEM)
supplemented with 1% heat-inactivated horse serum (Thermo Fisher),
0.6% glucose (Sigma-Aldrich) and 100 U/ml penicillin/streptomycin
(Life Technologies). The cortical tissue was subsequentially mechanically
dissociated with a P1000 pipette to generate single-cell suspensions.
Primary mouse neurons were seeded (8 10
4
cells/well) in a poly--
L-lysine (PLL, Sigma) coated 96-well plate (at bottom, Greiner) in MEM
supplemented medium. After allowing attachment for 4 h, plating MEM
medium was replaced by neurobasal medium supplemented with 1B27
supplement (Thermo Fisher), 2 mM L-glutamine (Thermo Fisher), and
100 U/ml penicillin/streptomycin (Life Technologies). Cells were
maintained at 37 C with 5% CO
2
culture conditions. Treatment (vehicle:
0.1% DMSO; roumilast: 1
μ
M; A33: 1
μ
M; Gebr32a: 1
μ
M) was started
24 h after isolation, under growth factor B27 deprivation to induce
neuronal cell death (an additional 48 h).
Propidium iodide (PI) viability assay
The viability of mouse primary neurons was assessed using a propi-
dium iodide (PI) viability assay as described previously [26,27]. Briey,
48 h after B27 growth factor deprivation and PDE inhibitor treatment,
culture medium was replaced with Lysis buffer A100 (ChemoMetec) and
lysis reaction was then halted by adding equal amounts of stabilization
buffer B (ChemoMetec), supplemented with PI (10
μ
g/ml, Sigma). After
15 min incubation in the dark, uorescent emission was measured using
the FluoStart OPTIMA plate reader (Bottom-up, excitation: 540 nm;
emission: 612 nm).
Luminescent iPSC-derived neurospheroid cultures
Neurospheroids were formed as described previously [28]. Briey,
eGFP/Luc human iPSC-NSCs were seeded at equal densities of 1.6 10
4
cells per well (ULA 96-well plate (Corning)) in neural expansion medium
(NEM, Gibco). Neurospheroids were maintained at 37 C, 5% CO
2
culture
conditions under constant orbital shaking (85 rpm). Two days after
plating, fresh NEM was added. A 50% medium change was conducted
every other day. Additionally, the luminescent signal was measured
weekly by adding 1.5 mg/ml Beetle luciferin (E1601, Promega) for 48 h
to the neurospheroid cultures. The luminescent signal was measured
using the Clariostar Plus plate reader, after which a complete medium
change was performed to eliminate remaining luciferin. On the same day
of bioluminescence evaluation, phase contrast pictures (4magnica-
tion) of every neurospheroid were taken using the Incucyte system to
evaluate overall neurospheroid size. The size of each neurospheroid was
determined using Image J by manually delineating the spheroids. PDE
inhibition treatment (vehicle: 0.1% DMSO; roumilast: 1
μ
M; A33: 1
μ
M;
Gebr32a: 1
μ
M) was initiated following 1 week of culturing (after the rst
luminescent signal measurement). At the end of the experiment (6 weeks
of culturing), neurospheroids were xed with 4% paraformaldehyde
(PFA) for 150 min at room temperature (RT), incubated overnight in 20%
sucrose (w/v in 1phosphate-buffered saline [PBS]) and consecutively
used for cryosectioning and immunocytochemical analysis.
Immunouorescence
Post-mortem spinal cord tissue
At 28 dpi, mice received an overdose of i.p. dolethal (200 mg/kg)
(Vetiquinol B.V.) and were transcardially perfused with Ringer solution
containing heparin (50 units/l), followed by a 4% PFA in 1xPBS perfu-
sion [29]. Longitudinal spinal cord cryosections of 10
μ
m thickness were
obtained. Immunouorescent staining was performed as described pre-
viously [2,3]. In brief, sections were blocked using 10% protein block
(Dako) in PBS containing 0.5% Triton-X-100 for 1 h at RT. For evaluating
oligodendrocyte differentiation using Olig2 and CC1, an additional an-
tigen retrieval step using a sodium citrate buffer (10 mM Sodium citrate,
0.05% Tween20, pH 6.0) was conducted. Primary antibodies were
diluted in PBS with 1% protein block and 0.5% Triton-X-100 and were
incubated overnight at 4 C(Table 2). Following washing, secondary
antibody incubation was done for 1 h at RT. Antibodies used were: goat
anti-rat Alexa uor 488 (1:250, A11006, ThermoFisher Scientic), goat
anti-mouse Alexa uor 568 (1:250, A11004, ThermoFisher Scientic),
goat anti-rat Alexa uor 568 (1:250, A11077, Invitrogen), and goat
anti-rabbit Alexa 488 (1:250, A11008, Invitrogen). Specicity of sec-
ondary antibodies was tested by omitting the primary antibody. Coun-
terstaining with DAPI (1:1000, Sigma-Aldrich) was performed for 10
min. Pictures were taken using a LEICA DM4000 B LED microscope and
LAS X software (Leica).
Neurospheroids
After xation, neurospheroids were processed as described previously
to allow high-throughput staining [28]. Briey, a silicone mold with 66
wells corresponding to the size of the neurospheroids was lled with
Tissue-Tek-OCT (VWR). Single neurospheroids were loaded into separate
wells of the silicone mold. Next, the mold was snap-frozen in isopentane
at a xed temperature (50 C), after which the resulting OCT-block was
removed from the silicon mold, turned upside down and covered with
additional OCT before freezing a second time. Cryosections of 10
μ
m
were obtained on PLL-coated glass slides. For immunocytochemical
Table 2
List of primary antibodies used in immunouorescence experiments.
Immunouorescence Antibody Host Source Dilution factor
Immunohistochemistry GFAP Mouse Sigma Aldrich (G3893) 1/500
MBP Rat Merck Millipore (MAB386) 1/250
CD4 Rat BD Biosciences (553043) 1/250
Cleaved caspase 3 Rabbit Cell signaling (9661) 1/100
NeuN Mouse Merck
Millipore (MAB377)
1/1000
Olig2 Goat Biotechne R&D(AF2418) 1/50
APC (CC1) Mouse Calbiochem (OP80) 1/50
5HT Rabbit Immunostar (20080) 1/1000
Immunocytochemistry NeuN Guinea-pig Merck
Millipore (ABN90P)
1/100
Cleaved caspase 3 Rabbit Cell signaling (9661) 1/400
Doublecortin (DCX) Rabbit Abcam (ab18723) 1/500
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
3
analysis, neurospheroid sections were permeabilized for 30 min (10%
milk solution [Sigma] in tris-buffered saline [TBS]). Primary antibodies
were diluted in 10% milk solution (Sigma) in TBS and were incubated
overnight at 4 C(Table 2). Following washing, secondary antibody in-
cubation was done for 1 h at RT. Antibodies used were: donkey
anti-rabbit Alexa uor 488 (1:600, A11006, ThermoFisher Scientic),
donkey anti-rabbit Alexa uor 555 (1:600, A11004, ThermoFisher Sci-
entic), and goat anti-guinea pig Alexa uor 555 (1:600, A11077, Invi-
trogen). DAPI was used to counterstain cellular nuclei. Pictures were
taken using an Axioscan 7 microscope slide scanner (Zeiss).
Fluorescence quantication
The original, unedited pictures were used for quantication. Repre-
sentative images were digitally enhanced to improve readability
(contrast and brightness). Quantication of histopathological parameters
was performed as described previously by investigators blinded to
experimental groups [2,3[. To quantify lesion size (GFAP
area) and
demyelinated area (MBP
area), 5710
μ
m thick sections per animal
were obtained, whereby the lesion center and consecutive rostral and
caudal area were analyzed. An intensity analysis was performed to
determine astrogliosis (GFAP expression) using ImageJ [2]. To assess
neuronal cell death at the lesion site, we quantied the number of cells
positive for both cleaved caspase 3 and NeuN markers. This counting was
performed in both the rostral and caudal regions relative to the lesion
epicenter. The obtained values were normalized by the total number of
NeuN
þ
cells. Similarly, to quantify oligodendrocyte differentiation, cells
positive for Olig2 and CC1 were counted using ImageJ in both rostral and
caudal regions relative to the injury. The results were normalized by the
total number of Olig2
þ
cells. For evaluating serotonergic 5-HT regrowth,
the rostral and caudal white matter regions of the ventral funiculus were
analyzed for the amount of descending 5-HT
þ
bers. T helper cells,
identied as CD4
þ
Iba-1
-
, were quantied by counting their number in
one microscope eld both rostral and caudal of the lesion site [30].
Differences in cleaved caspase, NeuN, and DCX positive cells within the
neurospheroids were determined by intensity analysis using ImageJ,
after which the positive area for each marker was corrected for the
number of nuclei (based on the DAPI count) present in the pictures.
IncuCyte live-cell imaging of cleaved caspase 3/7
eGFP/Luc human iPSC-NSCs were seeded at a density of 1 10
4
cells
per well in a Geltrex (Life Technologies) coated 96-well plate (at bottom,
Greiner). After allowing attachment for 24 h, cells were treated with the
PDE4 inhibitors (vehicle: 0.1% DMSO; roumilast: 1
μ
M; A33: 1
μ
M;
Gebr32a: 1
μ
M), and apoptosis was induced by oxygen deprivation using a
hypoxic chamber (1% O
2
for 4 h). Simultaneously, the IncuCyte Caspase-
3/7 Red apoptosis reagent (1.5
μ
M; #4704, Sartorius) was supplemented
to the cultures. Cell plates were placed into the IncuCyte live-cell analysis
system, and 5 images were taken per well. End-point apoptosis was
measured 6 h after oxygen deprivation. The IncuCyte integrated analysis
software was used to quantify the total level of apoptosis.
Statistics
Data were analyzed using GraphPad Prism version 9 (GraphPad
Software). All data were checked for normality using the Shapiro-Wilk
test. The BMS scores, GFAP intensity and neurospheroid biolumines-
cent results were analyzed using a two-way ANOVA for repeated mea-
surements with a Bonferroni post hoc test. Normally distributed data
were subsequently analyzed with a one-way ANOVA with Dunnett's
multiple comparisons (compared to vehicle). Not normally distributed
data were tested using the non-parametric Kruskal-Wallis test with
Dunn's multiple comparisons (compared to vehicle). Data are presented
as mean standard error of the mean (SEM). Differences with P values <
0.05 were considered signicant.
Results
Pan PDE4 and selective PDE4D, but not PDE4B, inhibition improve
functional recovery and histopathological outcomes after SCI
Initially, we examined whether selective PDE4B or PDE4D inhibitors
could improve functional recovery in a hemisection model of SCI. As a
positive control, we included roumilast, a second-generation pan PDE4
inhibitor. Starting 1 h post SCI injury, mice received either vehicle
(DMSO) or the different PDE4 inhibitors: the pan PDE4 inhibitor rou-
milast, the selective PDE4B inhibitor A33, the selective PDE4D inhibitor
Gebr32a, or a sequential administration of A33 (19 dpi) followed by
Gebr32a (1028 dpi) to evaluate possible additive effects. Functional
recovery was measured for 4 weeks using the BMS score. Both roumilast
and Gebr32a signicantly improved functional recovery compared to
vehicle-treated mice, whereas A33 treatment did not show any signi-
cant effect (Fig. 1A and B). Noteworthy, Gebr32a had a similar recovery
prole as roumilast (Fig. 1A and B). The sequential treatment with A33,
followed by Gebr32a showed the same trend in functional recovery as
Gebr32a treatment alone, namely starting from day 10, functional re-
covery of these mice mimicked the curve of the continuous Gebr32a
treatment (Fig. 1A and B).
To further elucidate the clinical translatability of PDE4D inhibition,
we administered Gebr32a starting 2 dpi which is considered a more
clinically relevant treatment window. In addition, as the sequential
treatment (A33 followed by Gebr32a after 10 dpi) resembled the results
of Gebr32a treatment alone (Fig. 1A), we included a new treatment group
where Gebr32a was administered alone starting 10 dpi to elucidate
whether therapeutic efcacy is maintained without the preceding A33
treatment. Inhibition of PDE4D with Gebr32a, starting 2 dpi signicantly
improved functional recovery compared to vehicle-treated mice. How-
ever, starting Gebr32a treatment 10 dpi had no signicant therapeutic
benet compared to vehicle treatment observed in the BMS scores but
showed a signicantly higher area under the curve compared to vehicle
(Fig. 2A and B). Notably, the administration of Gebr32a 1 h after injury
appeared to have a more pronounced improvement in the BMS score
compared to Gebr32a treatment starting 2 dpi (Gebr32a start 1 h pi 3.93
0.43 vs Gebr32a start 2 dpi 3.13 0.29).
Histological GFAP and MBP analyses indicated signicantly reduced
lesion size and demyelinated areas, respectively, in roumilast- and
Gebr32a-treated mice compared to the vehicle group whereas the
treatment with A33 was ineffective (Fig. 3AJ). Next, the level of
astrogliosis was determined by analyzing GFAP intensity at varying dis-
tances to the lesion center. Whereas roumilast and Geb32a treatment
did not alter GFAP intensities, A33 exacerbated astrogliosis, especially at
the rostral lesion site (Fig. 4AE). As an additional neuroinammatory
outcome measurement, we determined the number of inltrated CD4
þ
T
lymphocytes in the perilesional area. However, no differences between
vehicle and treatment groups were observed (Supplementary Fig. S1).
Pan PDE4 and selective PDE4D inhibition increase the number of
differentiated oligodendrocytes at the (peri)lesion site following SCI, unlike
PDE4B inhibition
Due to the reduced demyelinated area observed at the lesion site upon
both PDE4 and PDE4D inhibition, we next investigated the presence of
differentiated oligodendrocytes (CC1
þ
) at the lesion site. The total
number of oligodendrocyte lineage cells was rst determined based on
sole Olig2 positivity at the lesion site, which was unaltered upon either
pan PDE4 or PDE4 subtype-selective inhibition compared to vehicle-
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
4
treated animals (Supplementary Fig. S2). However, mice treated with
roumilast or Gebr32a did display a signicant higher number of mature
oligodendrocytes (CC1
þ
Olig2
þ
) compared to vehicle- or A33-treated
animals (Fig. 5AE).
Pan PDE4 and selective PDE4D inhibition is neuroprotective and stimulates
5-HT serotonergic regrowth after SCI, unlike PDE4B inhibition
Next, we investigated whether the abovementioned decreased lesion
size was accompanied by neuroprotection and neuroregeneration. First,
the neuroprotective effects of pan PDE4 or PDE4 subtype-selective inhi-
bition were determined based on Cleaved Caspase 3 and NeuN double
positivity at the lesion site to evaluate the number of apoptotic neurons.
Mice treated with the pan PDE4 inhibitor roumilast or the selective
PDE4D inhibitor Gebr32a displayed a reduced number of Cleaved Caspase
3
þ
NeuN
þ
double positive cells compared to vehicle-treated animals at the
(peri-)lesion (Fig. 6AE). Furthermore, A33-treated animals displayed a
trend (p ¼0.08) toward reduced neuronal cell death (Fig. 6AE).
Next, to determine the spinal dendritic plasticity of serotoninergic
ber projections, the number of descending 5-HT positive tracts was
determined. In comparison with A33, both roumilast- and Gebr32a-
treated animals showed signicantly increased mean number of
descending 5-HT dendrites, indicating serotonergic neuroregeneration or
protection (Fig. 7A and B).
Pan PDE4 and selective PDE4D inhibition prevents apoptosis of primary
murine neurons and human iPSC-derived neural stem cells, unlike PDE4B
inhibition
To evaluate whether the neuroprotection by roumilast and Gebr32a
observed in vivocould be attributed to direct neural protection, we assessed
the effects of these PDE4 inhibitors on neural apoptosis in both primary
mouse-derived neurons and human iPSC-derived NSCs. Fig. 8A shows the
level of neuronal apoptosis of mouse neurons following 48 h of B27 growth
factor deprivation. Both roumilast-mediated PDE4 inhibition and
Gebr32a-mediated PDE4D inhibition partly prevented the stress-induced
neuronal apoptosis as observed by a higher PI signal compared to
vehicle-treated cultures (Fig. 8A). Similarly, human iPSC-derived neural
stem cell cultures subjected to oxygen deprivation for 4 h and treated with
either roumilast or Gebr32a displayed a signicant reduction of Cleaved
Caspase 3/7 positivity 6 h post stress-induced neural apoptosis (Fig. 8B).
Real-time bioluminescence monitoring of human neurospheroids
demonstrates the neuroprotective feature of both pan PDE4 and selective
PDE4D inhibition, which is accompanied by increased neuronal
differentiation
To enable real-time read-out of neurospheroid viability, we used an
eGFP/Luc human iPSC-derived neural stem cells stably expressing the
Fig. 2. Treatment with the PDE4D inhibitor
Gebr32a starting 2 dpi, but not from 10 dpi, im-
proves functional recovery after a spinal cord
injury.(AB) Starting directly after injury, mice were
treated with vehicle. The Gebr32a treated animals
changed from vehicle to Gebr32a (0.3 mg/kg) at 2 dpi
or 10 dpi, while the vehicle croup continued DMSO
(0.1%) treatment until 28 dpi. (A) Starting Gebr32a
treatment 2 dpi, but not 10 dpi, signicantly
improved functional outcomes, measured by the BMS
score. The black, and gray arrows indicate the start of
Gebr32a treatment 2 dpi and 10 dpi, respectively. (B)
Starting at 2 dpi and 10 dpi with Gebr32a signi-
cantly improved the area under the curve, compared
to vehicle-treated mice over time. Data were analyzed
using a two-way ANOVA with Bonferroni multiple
comparison test (compared to vehicle). Data are dis-
played as mean SEM. n¼59 mice/group. (A) *p
0.05 Gebr32a start 2 dpi vs vehicle; (B) *p <0.05,
**p <0.01 compared to vehicle.
Fig. 1. Treatment with the PDE4 inhibitor
roumilast or the PDE4D inhibitor
Gebr32a improve functional recovery
after spinal cord injury, whereas the
PDE4B inhibitor A33 has no effect.(AB)
Starting 1 h after injury, mice were treated
with vehicle, a general PDE4 inhibitor
roumilast (3 mg/kg), or gene-specic PDE4
inhibitors, A33 (3 mg/kg) and Gebr32a (0.3
mg/kg). In contrast to A33, roumilast and
Gebr32a signicantly improved (A) func-
tional outcomes, measured by the BMS score
and (B) area under the curve, compared to
vehicle-treated mice over time. Data were
analyzed using a two-way ANOVA with
Bonferroni multiple comparison test
(compared to vehicle). Data are displayed as
mean SEM. n¼1016 mice/group. #p
0.05 vehicle versus roumilast; **p <0.01
vehicle versus Gebr32a.
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
5
rey luciferase reporter. Over time, decreased viability was observed in
the neurospheroids due to oxygen/nutrient deprivation in the core
(Fig. 9A). In contrast to the selective PDE4B inhibitor A33, treatment
with either the pan PDE4 inhibitor roumilast or the selective PDE4D
inhibitor Gebr32a stabilized neurospheroid viability when stress-induced
core cell loss occurred (Fig. 9A). Of note, the overall size of the neuro-
spheroids was not different between treatment groups (Fig. 9B). At the
end of the 6-week culture period, neurospheroids were characterized for
the level of apoptosis (Cleaved Caspase 3), neuronal differentiation
(NeuN), and neurogenesis (DCX). Quantication of the Cleaved Caspase
3
þ
area revealed a signicant reduction in apoptosis when inhibiting
PDE4 or PDE4D (Fig. 9CF). Furthermore, both roumilast and Gebr32a
treatment signicantly increased neuronal differentiation as more
NeuN
þ
cells were present at the end of the experiment (Fig. 9DF). At end
Fig. 3. Roumilast or Gebr32a treatments
reduce the lesion size and demyelinated
area after spinal cord injury, whereas A33
has no effect.(AJ) Starting 1 h after injury,
mice were treated with vehicle, the pan
PDE4 inhibitor roumilast (3 mg/kg), or the
subtype-selective PDE4 inhibitors, A33 (3
mg/kg) and Gebr32a (0.3 mg/kg). (A)
Quantication of lesion size, determined by
the GFAP negative area, showed that this
was reduced in mice treated with roumilast
or Gebr32a compared to the vehicle group.
No difference between the vehicle and A33
groups was observed. Data were normalized
to vehicle and are shown as mean SEM. n
¼78 mice/group. (BE) Representative
images from the spinal cord sections are
shown. Lesion size (GFAP
area) was deter-
mined as depicted by the dotted white line.
Scale bar ¼250
μ
m. (F) Quantication of the
demyelinated area, determined by the MBP
negative area, showed that this was reduced
in mice treated with roumilast or Gebr32a
compared to the vehicle group. No difference
between vehicle and A33 groups was
observed. Data were normalized to vehicle
and are shown as mean SEM. n¼78
mice/group. (GJ) Representative images
from the spinal cord sections are shown.
Demyelinated area (MBP
area) was deter-
mined as depicted by the dotted white line.
Scale bar ¼250
μ
m. Demyelination area and
lesion size were analyzed using a one-way
ANOVA with Dunnett's multiple comparison
test (compared to vehicle), *p <0.05, ***p <
0.005. Data are displayed as mean SEM.
Fig. 4. Roumilast and Gebr32a treat-
ment do not affect astrogliosis after spinal
cord injury, whereas A33 administration
exacerbates astrocyte reactivity.(AE)
Starting 1 h after injury, mice were treated
with vehicle, a pan PDE4 inhibitor roumi-
last (3 mg/kg), or subtype-selective PDE4
inhibitors, A33 (3 mg/kg) and Gebr32a (0.3
mg/kg). (A) Quantication of astrogliosis by
GFAP intensity analysis showed that, in
contrast to other treatment groups, A33
application exacerbated astrogliosis
compared to vehicle-treated mice. Data are
shown as mean SEM. n¼46 mice/group.
GFAP intensity was analyzed using a two-
way ANOVA with a Bonferroni post hoc
test. *p <0.05 A33 versus vehicle, **p <
0.01 A33 versus vehicle.(BE) Representa-
tive images from the spinal cord sections are
shown. All analyses were quantied within
square areas of 100
μ
m100
μ
m perile-
sional placed as indicated in the gure,
extending 600
μ
m rostral to 600
μ
m caudal
from the lesion center (white line). Scale bar
¼500
μ
m.
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
6
stage, no signicant differences were found in DCX
þ
area, nor for A33
treatment within any marker evaluated (Fig. 9E and F).
Discussion
In SCI research, PDE4 inhibition has yielded promising results due to
its broad effects on different secondary injury-related outcomes, such as
immune cell inltration, inammation, and axonal sprouting [811,31].
However, the clinical translation of pan-PDE4 inhibitors remains limited
due to their poor tolerability in patients at doses required for clinical
effectiveness [32]. In order to circumvent this pitfall, we investigated
the potential of specic non-emetic PDE4B and PDE4D inhibitors,
respectively A33 and Gebr32a. We now show that the PDE4D inhibitor
Gebr32a signicantly improved functional recovery after SCI similarly to
the pan PDE4 inhibitor roumilast, whereas the PDE4B inhibitor A33 did
not. Moreover, starting Gebr32a at a more clinically relevant therapeutic
window (2 dpi) still promoted functional recovery. In addition, both
roumilast and Gebr32a reduced the lesion size, demyelinated area, and
Fig. 5. Roumilast and Gebr32a treat-
ments increase the amount of mature ol-
igodendrocytes at the peri-lesion site
after spinal cord injury.(AE) Starting 1 h
after injury, mice were treated with vehicle,
the pan PDE4 inhibitor roumilast (3 mg/
kg), or the subtype-selective PDE4 inhibitors,
A33 (3 mg/kg) and Gebr32a (0.3 mg/kg).
(A) Using a double staining for Olig2 (oli-
godendrolineage marker) and CC1 (mature
oligodendrocyte marker), we showed a
signicantly increased percentage of mature
oligodendrocytes at the lesion site following
PDE4 (roumilast) and PDE4D (Gebr32a)
inhibition. n¼78 mice/group. Data were
normalized to vehicle and are shown as
mean SEM. Results were analyzed using a
one-way ANOVA with Dunnett's multiple
comparison test (compared to vehicle), **p
<0.01, ****p <0.001. (BE) Representative
images of the Olig2-CC1 double staining at
the lesion site. Single stainings are shown
above the merged image. Scale bar ¼100
μ
m.
Fig. 6. Roumilast and Gebr32a treat-
ment act neuroprotective at the lesion
site after spinal cord injury.(AE) Starting
1 h after injury, mice were treated with
vehicle, the pan PDE4 inhibitor roumilast
(3 mg/kg), or the subtype-selective PDE4
inhibitors, A33 (3 mg/kg) and Gebr32a (0.3
mg/kg). (A) Quantication of the number of
Cleaved Caspase 3
þ
NeuN
þ
neurons at the
(peri)lesion site indicated a neuroprotective
effect of both PDE4 and PDE4D inhibition as
reduced neuronal apoptosis was observed. n
¼78 mice/group. Data are normalized to
vehicle and are shown as mean SEM. Re-
sults were analyzed using a one-way ANOVA
with Dunnett's multiple comparison test
(compared to vehicle), ***p <0.005. (BE)
Representative images of the Cleaved Cas-
pase 3 NeuN staining at the lesion site. Single
stainings are shown above the merged
image. The white boxed regions are shown at
higher magnication (40) underneath the
merged image. Scale bar ¼100
μ
m.
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
7
neuronal apoptosis while increasing the number of mature oligoden-
drocytes, and 5-HT
þ
serotonergic bers. The neuroprotective feature of
both pan PDE4 inhibition and PDE4D subtype inhibition can be partially
attributed to a direct neur(on)al effect as we showed a decreased
neur(on)al apoptosis in vitro using murine neurons and iPSC-derived 3D
neurospheroid cultures, which is in line with the previously described
neuroplasticity enhancing properties of PDE4D inhibition [33]. These
results support the use of the PDE4D inhibitor Gebr32a for SCI therapy.
We conrm that the second-generation PDE4 inhibitor roumilast
promoted SCI recovery, as demonstrated previously [34]. This
second-generation PDE4 inhibitor is accompanied with less emetic side
effects compared to rst-generation pan-PDE4 inhibitors (e.g., rolipram)
[35,36]. Noteworthy, the second-generation PDE3, PDE4, and PDE5 in-
hibitor Ibudilast is currently being evaluated for degenerative cervical
myelopathy, a non-traumatic SCI [37]. However, translation of the
roumilast dose to promote CNS repair from rodents to humans, results
in a dose approximately 500 times higher than the approved dose for
roumilast in chronic obstructive pulmonary disease. Hence, the CNS
repair-inducing dose is still emetic [38]. Therefore, research to safer and
more targeted options are crucial to inuence discrete cell processes and
promote SCI outcomes. Previous studies showed that PDE4B orchestrates
the inammatory immune response, while PDE4D contributes to adult
neurogenesis, neuroplasticity, and myelin regeneration [39]. As SCI is
characterized by a robust neuroinammatory response and limited
Fig. 7. Roumilast and Gebr32a induce 5-HT serotonergic regrowth following SCI as indicated by the increased number of descending 5-HT tracts over the
lesion site.(AB) Starting 1 h after injury, mice were treated with vehicle, the pan PDE4 inhibitor roumilast (3 mg/kg), or the subtype-specic PDE4 inhibitors, A33
(3 mg/kg) and Gebr32a (0.3 mg/kg). (A) Quantication of the 5-HT serotonergic staining showed an increase in the number (A) of descending 5-HT tracts over the SCI
lesion site upon PDE4 (roumilast) and PDE4D (Gebr32a) inhibition. Data were normalized to vehicle and are shown as mean SEM. n¼49 mice/group. (B)
Representative images of the 5-HT staining at the lesion site. The white arrows indicate examples of 5-HT descending tracts. Results were analyzed using a one-way
ANOVA with Dunnett's multiple comparison test (compared to vehicle), ***p <0.005.
Fig. 8. Apoptosis of primary mouse neu-
rons and human iPSC-derived NSCs was
prevented by both roumilast and
Gebr32a treatment.(A) Primary mouse
neurons deprived of the growth factor B27
for 48 h showed a decreased neuronal
viability at the end of the experiment, which
was partly prevented by inhibiting PDE4
(roumilast, 1
μ
M) or PDE4D (Gebr32a, 1
μ
M). Data were normalized to vehicle and
are shown as mean SEM. n¼67/group
with an nrepresentative for one well. PI
measurements of primary mouse neurons
were analyzed using a one-way ANOVA with
Dunnett's multiple comparison test
(compared to vehicle). (B) Human iPSC-
derived neural stem cells showed increased
levels of Cleaved Caspase 3/7 upon oxygen
deprivation, which was signicantly reduced
upon PDE4 (roumilast, 1
μ
M) and PDE4D
(Gebr32a, 1
μ
M) inhibition. n¼89/group
with an nrepresentative for one well.
Cleaved Caspase 3/7 signal measurements
were analyzed using a non-parametric Krus-
kal-Wallis test with Dunn's multiple com-
parison test (compared to vehicle), *p <
0.05, **p <0.01, ****p <0.001. Data are
displayed as mean SEM.
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
8
axonal regeneration, we focused on PDE4B and PDE4D inhibition.
Important to note is that the inhibitors used in this study (A33 and
Gebr32a) both lack an emetic response up to 100 mg/kg in animals,
highlighting the clinical relevance of the evaluated compounds [38].
After SCI, PDE4B is acutely upregulated in the damaged spinal cord,
especially in phagocytes [40]. The PDE4B subfamily is an important
modulator of the intracellular cAMP levels in inammatory cells,
including macrophages, microglia, and astrocytes [5,14]. In a mouse
model of multiple sclerosis, the PDE4B expression in antigen-presenting
cells, such as phagocytes, was correlated with the disease severity [41].
Pharmacological inhibition of PDE4B by 0.3 mg/kg A33 i.p. administered
30 min after traumatic brain injury induction, induced anti-inammatory
markers (e.g., Arginase-1) in phagocytes and limited lesion size [4244].
Complete PDE4B knockdown had benecial effects on recovery in a
contusion SCI model [40]. Based on these data, it was somewhat sur-
prising that PDE4B inhibition by 3 mg/kg A33 (s.c) did not improve
functional or histopathological recovery in our study. Moreover, PDE4B
inhibition did exaggerate astrogliosis, which is considered detrimental
for SCI outcomes since astrocytes contribute to the formation of the
regenerative-limiting glial scar 4548. Therefore, the absence of func-
tional outcomes upon A33 treatment in our study could be attributed to
the excessive astrogliosis. Importantly, it has been previously shown that
treatment protocol and dose are important determinants for benecial
effects [49]. For example, intravenous (i.v.) or s.c. administration of the
PDE4 inhibitor rolipram has been shown to be more effective to treat SCI
compared to oral administration. Additionally, netuning the PDE4
concentration showed to be crucial. More specically, a low rolipram
dose of 0.5 mg/kg/day had no benecial effect for SCI treatment, while a
higher dose of 1 mg/kg with the same treatment protocol could signi-
cantly improve SCI recovery [9]. However, an even higher dose of 0.8
mmol/kg/day demonstrated to not improve SCI recovery, postulated
through off-target effects [10]. Therefore, performing a dose response
study, testing another administration route, or different treatment tim-
ings can potentially result in ameliorated SCI outcomes upon A33
treatment. Hence, we cannot exclude that A33 could still provide
long-term benets after SCI. It is additionally crucial to highlight that the
hemisection SCI model is highly effective for studying neuronal regen-
eration, but does not fully represent the human SCI pathology due to the
lack of the complex neuroinammatory responses. Conversely, the
contusion SCI model provides a more faithful representation of neuro-
inammation and axonal preservation evident in human SCI [50].
Consequently, it remains essential to consider the prospect that the
anti-inammatory attributes associated with the inhibition of PDE4B
could potentially confer an indirect neuroprotective inuence within the
contusion SCI model. This warrants a thorough exploration when
assessing the therapeutic viability of PDE4B inhibition within the
Fig. 9. Roumilast and Gebr32a treatment protected human iPSC-derived neurospheroids from neural apoptosis and stimulated neuronal differentiation,
while not affecting neurogenesis.(A) Weekly luminescence measurement of neurospheroids showed a stabilized viability over time, which decreased at 6 weeks of
culture. However, this decrease was counteracted by treatment with roumilast (1
μ
M) or Gebr32a (1
μ
M). (B) The size of the neurospheroids was not different
between groups. (A, B) Data are shown as mean SEM. n¼24 spheroids/group. Data were analyzed using a two-way ANOVA with Dunnets multiple comparison test
(compared to vehicle), *p <0.05, ***p <0.005. (CF) At the end of the culture experiment, the 6-week-old neurospheroids were stained and quantied for (C)
Cleaved Caspase 3 (apoptosis), (D) NeuN (neuronal differentiation), or (E) DCX (neurogenesis) positive cells with respect to the total number of nuclei. Data were
normalized to vehicle and are shown as mean SEM. n¼68 spheroids/group. The amount of cleaved caspase positive cells was analyzed using a non-parametric
Kruskal-Wallis test with Dunn's multiple comparisons (compared to vehicle). The amount of NeuN or DCX positive cells was analyzed using a one-way ANOVA with
Dunnett's multiple comparison test (compared to vehicle), **p <0.01. (F) Representative immunouorescent images of the human iPSC-derived neurospheroids. Scale
bar ¼500
μ
m.
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
9
framework of the contusion SCI model.
Pan PDE4 inhibition using rolipram treatment after SCI has previ-
ously been shown to attenuate oligodendrocyte apoptosis and promote
axonal growth and plasticity [4,49]. We have found that selective PDE4D
inhibition by Gebr32a boosted oligodendrocyte precursor cell differen-
tiation in vitro and stimulated remyelination in an ex vivo model [38]. In
the current study, we show that Gebr32a administration, even when
started 2 dpi, improved functional recovery after SCI. Previous research
showed that earlier intervention results in better SCI outcomes [51,52].
Noteworthy, Gebr32a improved SCI outcomes signicantly when
administered in a clinically relevant therapeutic window but indeed, our
results suggest that starting earlier with Gebr32a leads to better SCI
outcomes (Gebr32a start 1 h pi 3.93 0.43 vs Gebr32a start 2 dpi 3.13
0.29). In mice treated 1 h post injury, we observed reduced lesion size
and decreased demyelinated area, which was accompanied by increased
numbers of mature oligodendrocytes, pointing toward reduced demye-
lination, mature oligodendrocyte protection or increased remyelination.
Moreover, Gebr32a acts neuroprotective following SCI as demonstrated
by the decreased number of apoptotic neurons. Previously, Gebr32a was
shown to regulate neuronal morphology as demonstrated by the
increased neurite outgrowth in both N2a and HT22 cells [13]. Due to this
neuroregenerative feature of Gebr32a, combined with the observation
that the loss of locomotor function following SCI correlates to the damage
of 5-HT serotonergic projections in the spinal cord, we aimed to evaluate
in vivo neuroregeneration by quantifying the descending 5-HT tracts over
the lesion site [53]. Treatment of Gebr32a increased the number of
descending 5-HT bers, indicating either axonal sparing or neuro-
regenerative features of PDE4D inhibition. The effects observed after
Gebr32a treatment were comparable to roumilast. Although we cannot
exclude any indirect neuroprotective effects of Gebr32a so far, we
demonstrated here, at least partially, that the observed decrease in
neuronal apoptosis can be attributed to direct neuronal protection. In
both murine and human iPSC-derived neural stem cells, Gebr32a treat-
ment diminished neur (on)al cell death. Similarly, Gebr32a stabilized the
human neurospheroid viability, accompanied by decreased apoptosis
and increased neuronal differentiation. Spinal cord neurons differ in
calcium load, protection against reactive oxygen species, gene expres-
sion, and mitochondrial metabolism compared to brain-derived neurons,
which suggests that spinal cord-derived neurons are more prone to
trauma-induced injury [5456]. Hence, validation of the neuroprotective
and differentiation-stimulating effects of Gebr32a on spinal cord-derived
neurons could be interesting for future research. Notably, previous
research already demonstrated the neuroprotective effects of
cAMP-elevating agents in spinal cord and brain-derived neurons [13,57,
58]. Therefore, the neuroprotective effects and enhanced differentiation
capacity upon Gebr32a administration in brain-derived cultures are
proposed to translate toward spinal cord neurons.
Due to the hypothesized anti-inammatory properties of PDE4B in-
hibition, and the previously observed regenerative properties of PDE4D
inhibition, we evaluated whether a sequential treatment regimen could
further improve SCI outcomes compared to monotreatment strategies.
Thus, the PDE4B inhibitor A33 was administered during the initial phase
of SCI and subsequently substituted by the PDE4D inhibitor Gebr32a
from day 10 onwards. While inammatory processes are essential for
removing pathogens and cell debris, their benets are overshadowed by
the accumulation of inammatory cytokines in the CNS upon inam-
matory immune cell inltration and activation [59,60]. These secondary
inammatory-mediated damage processes severely impair regenerative
processes and glial functioning [61]. Therefore, by diminishing the
neuroinammatory response with PDE4B inhibition, we aimed to create
a favorable micro-environment thereby allowing regeneration to occur
more efciently. By inhibiting PDE4D in a later phase, we hypothesized
that the regenerative process could be further enhanced, and functional
outcome would be improved even more compared to continuous PDE4B
or PDE4D subtype inhibition throughout the disease course. However, in
our model, inhibiting PDE4B by means of A33 during the early phase of
the disease did not provide any additional benet on functional outcome
following hemisection SCI compared to continuous PDE4D inhibition by
Gebr32a. However, when PDE4D inhibition was started 10 dpi, without
preceding A33 administration, Gebr32a did not signicantly ameliorate
functional recovery, observed in the BMS scores. We propose that
Gebr32a provides a therapeutic benet when started 10 dpi, upon A33
treatment, due to the priming of A33 in the SCI environment. A33 can
potentially create a more favorable CNS environment for neuronal repair,
resulting in the therapeutic efcacy of Gebr32a when administered 10
dpi. Based on post-mortem analysis, A33 alone lacked efcacy which
could be due to the dose, timing, route of administration or the harsh
neurodegenerative conditions accompanied with a hemisection SCI
lesion. Therefore, it cannot be excluded that sequential PDE4
subtype-specic treatment can be even more efcient and clinically
relevant compared to only PDE4D inhibition to treat SCI.
Despite the promising preclinical ndings on PDE4 inhibitors, the
accompanied severe side effects at the therapeutic dose have hindered
their clinical translation so far. In this study, we analyzed the impact of
the PDE4B inhibitor A33 and the PDE4D inhibitor Gebr32a in a mouse
model of SCI. In contrast to A33, Gebr32a improved functional recovery,
even when treatment was initiated 2 dpi. In addition Gebr32a amelio-
rated histopathological outcomes to a comparable level as the pan PDE4
inhibitor roumilast. Whereas roumilast is associated with emetic-like
side effects at its repair-inducing dose, Gebr32a is not. These data
strongly support the notion that the selective PDE4D inhibitor Gebr32a
holds great potential as a novel therapeutic approach for SCI treatment.
Funding
This study was supported by grants from Fonds Wetenschappelijk
Onderzoek (FWO-Vlaanderen) to TVM, MS, SH, JVB, and FM
(12G0817N, 1S57521N, G041421N, 1272324N, 1209123N, and
1SH2E24N). We also acknowledge partial funding from the University of
Antwerp IOF-SBO brain organoid project granted to PP.
Author contributions
MS, SH, SL, NH, PP, JVB, and TV participated in the conceptualiza-
tion, data interpretation and supervised the project. MS, FM, EvB, JVB
carried out the investigation, and participated in data collection. MS, FM,
and JVB wrote the manuscript. SH, EvB, PP, SL, NH, RR, EF, OB, CB, and
JP reviewed the manuscript. All authors read and approved the nal
manuscript.
Declaration of competing interest
The authors declare the following nancial interests/personal re-
lationships which may be considered as potential competing interests:
MS, JP, and TV have a proprietary interest in selective PDE4D inhibitors
for the treatment of demyelinating disorders and neurodegenerative
disorders. RR, EF, OB, CB, and JP have a proprietary interest in the use of
Gebr32a. If there are other authors, they declare that they have no known
competing nancial interests or personal relationships that could have
appeared to inuence the work reported in this paper.
Acknowledgments
The authors would like to thank Dr. Leen Timmermans for her
excellent technical assistance with the in vivo experiments.
Appendix A. Supplementary data
Supplementary data to this article can be found online at https
://doi.org/10.1016/j.neurot.2024.e00372.
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
10
References
[1] Ren Y, Young W. Managing inammation after spinal cord injury through
manipulation of macrophage function. Neural Plast 2013;2013:945034.
[2] Van Broeckhoven J, Sommer D, Dooley D, Hendrix S, Franssen A. Macrophage
phagocytosis after spinal cord injury: when friends become foes. Brain 2021;
144(10):293345.
[3] Tran AP, Warren PM, Silver J. The biology of regeneration failure and success after
spinal cord injury. Physiol Rev 2018;98(2):881917.
[4] Hannila SS, Filbin MT. The role of cyclic AMP signaling in promoting axonal
regeneration after spinal cord injury. Exp Neurol 2008;209(2):32132.
[5] Schepers M, Tiane A, Paes D, Sanchez S, Rombaut B, Piccart E, et al. Targeting
phosphodiesterases-towards a tailor-made approach in multiple sclerosis treatment.
Front Immunol 2019;10:1727.
[6] Zhou G, Wang Z, Han S, Chen X, Li Z, Hu X, et al. Multifaceted roles of cAMP
signaling in the repair process of spinal cord injury and related combination
treatments. Front Mol Neurosci 2022;15:808510.
[7] Paes D, Schepers M, Rombaut B, van den Hove D, Vanmierlo T, Prickaerts J. The
molecular biology of phosphodiesterase 4 enzymes as pharmacological targets: an
interplay of isoforms, conformational states, and inhibitors. Pharmacol Rev 2021;
73(3):101649.
[8] Bao F, Fleming JC, Golshani R, Pearse DD, Kasabov L, Brown A, et al. A selective
phosphodiesterase-4 inhibitor reduces leukocyte inltration, oxidative processes,
and tissue damage after spinal cord injury. J Neurotrauma 2011;28(6):103549.
[9] Schaal SM, Garg MS, Ghosh M, Lovera L, Lopez M, Patel M, et al. The therapeutic
prole of rolipram, PDE target and mechanism of action as a neuroprotectant
following spinal cord injury. PLoS One 2012;7(9):e43634.
[10] Nikulina E, Tidwell JL, Dai HN, Bregman BS, Filbin MT. The phosphodiesterase
inhibitor rolipram delivered after a spinal cord lesion promotes axonal regeneration
and functional recovery. Proc Natl Acad Sci U S A 2004;101(23):878690.
[11] Whitaker CM, Beaumont E, Wells MJ, Magnuson DS, Hetman M, Onifer SM.
Rolipram attenuates acute oligodendrocyte death in the adult rat ventrolateral
funiculus following contusive cervical spinal cord injury. Neurosci Lett 2008;
438(2):2004.
[12] Mussen F, Broeckhoven JV, Hellings N, Schepers M, Vanmierlo T. Unleashing spinal
cord repair: the role of cAMP-specic PDE inhibition in attenuating
neuroinammation and boosting regeneration after traumatic spinal cord injury. Int
J Mol Sci 2023;24(9).
[13] Paes D, Schepers M, Willems E, Rombaut B, Tiane A, Solomina Y, et al. Ablation of
specic long PDE4D isoforms increases neurite elongation and conveys protection
against amyloid-beta pathology. Cell Mol Life Sci 2023;80(7):178.
[14] Pearse DD, Hughes ZA. PDE4B as a microglia target to reduce neuroinammation.
Glia 2016;64(10):1698709.
[15] Avila DV, Myers SA, Zhang J, Kharebava G, McClain CJ, Kim HY, et al.
Phosphodiesterase 4b expression plays a major role in alcohol-induced neuro-
inammation. Neuropharmacology 2017;125:37685.
[16] Jin SL, Lan L, Zoudilova M, Conti M. Specic role of phosphodiesterase 4B in
lipopolysaccharide-induced signaling in mouse macrophages. J Immunol 2005;
175(3):152331.
[17] Shi Y, Lv J, Chen L, Luo G, Tao M, Pan J, et al. Phosphodiesterase-4D knockdown in
the prefrontal cortex alleviates memory decits and synaptic failure in mouse
model of alzheimer's disease. Front Aging Neurosci 2021;13:722580.
[18] Sierksma AS, van den Hove DL, Pfau F, Philippens M, Bruno O, Fedele E, et al.
Improvement of spatial memory function in APPswe/PS1dE9 mice after chronic
inhibition of phosphodiesterase type 4D. Neuropharmacology 2014;77:12030.
[19] Schepers M, Vanmierlo T. Novel insights in phosphodiesterase 4 subtype inhibition
to target neuroinammation and stimulate remyelination July 20. Neural
Regeneration Research 2023;19(3):4934.
[20] Van Broeckhoven J, Erens C, Sommer D, Scheijen E, Sanchez S, Vidal PM, et al.
Macrophage-based delivery of interleukin-13 improves functional and
histopathological outcomes following spinal cord injury. J Neuroinammation
2022;19(1):102.
[21] Sommer D, Corstjens I, Sanchez S, Dooley D, Lemmens S, Van Broeckhoven J, et al.
ADAM17-deciency on microglia but not on macrophages promotes phagocytosis
and functional recovery after spinal cord injury. Brain Behav Immun 2019;80:
12945.
[22] Erens C, Van Broeckhoven J, Hoeks C, Schabbauer G, Cheng PN, Chen L, et al. L-
arginine depletion improves spinal cord injury via immunomodulation and nitric
oxide reduction. Biomedicines 2022;10(2).
[23] Ricciarelli R, Brullo C, Prickaerts J, Arancio O, Villa C, Rebosio C, et al. Memory-
enhancing effects of GEBR-32a, a new PDE4D inhibitor holding promise for the
treatment of Alzheimer's disease. Sci Rep 2017;7:46320.
[24] Basso DM, Fisher LC, Anderson AJ, Jakeman LB, McTigue DM, Popovich PG. Basso
Mouse Scale for locomotion detects differences in recovery after spinal cord injury
in ve common mouse strains. J Neurotrauma 2006;23(5):63559.
[25] Sciarretta C, Minichiello L. The preparation of primary cortical neuron cultures and
a practical application using immunouorescent cytochemistry. Methods Mol Biol
2010;633:22131.
[26] Lo Monaco M, Gervois P, Beaumont J, Clegg P, Bronckaers A, Vandeweerd JM, et al.
Therapeutic potential of dental pulp stem cells and leukocyte- and platelet-rich
brin for osteoarthritis. Cells 2020;9(4).
[27] Evens L, Heeren E, Rummens JL, Bronckaers A, Hendrikx M, Deluyker D, et al.
Advanced glycation end products impair cardiac atrial appendage stem cells
properties. J Clin Med 2021;10(13).
[28] Van Breedam E, Nijak A, Buyle-Huybrecht T, Di Stefano J, Boeren M, Govaerts J,
et al. Luminescent human iPSC-derived neurospheroids enable modeling of
neurotoxicity after oxygen-glucose deprivation. Neurotherapeutics 2022;19(2):
55069.
[29] Nelissen S, Vangansewinkel T, Geurts N, Geboes L, Lemmens E, Vidal PM, et al.
Mast cells protect from post-traumatic spinal cord damage in mice by degrading
inammation-associated cytokines via mouse mast cell protease 4. Neurobiol Dis
2014;62:26072.
[30] Dooley D, Lemmens E, Vangansewinkel T, Le Blon D, Hoornaert C, Ponsaerts P,
et al. Cell-based delivery of interleukin-13 directs alternative activation of
macrophages resulting in improved functional outcome after spinal cord injury.
Stem Cell Rep 2016;7(6):1099115.
[31] Butler MB, Vellaiyappan SK, Bhatti F, Syed FE, Rafati Fard A, Teh JQ, et al. The
impact of phosphodiesterase inhibition on neurobehavioral outcomes in preclinical
models of traumatic and non-traumatic spinal cord injury: a systematic review.
Front Med 2023;10:1237219.
[32] Knott EP, Assi M, Rao SN, Ghosh M, Pearse DD. Phosphodiesterase inhibitors as a
therapeutic approach to neuroprotection and repair. Int J Mol Sci 2017;18(4).
[33] Paes D, Lardenoije R, Carollo RM, Roubroeks JAY, Schepers M, Coleman P, et al.
Increased isoform-specic phosphodiesterase 4D expression is associated with
pathology and cognitive impairment in Alzheimer's disease. Neurobiol Aging 2021;
97:5664.
[34] Moradi K, Golba khsh M, Haghighi F, Afshar i K, Nikbakhsh R, Khavandi MM ,
et al. Inhibitio n of phosphodiesterase IV en zyme improves locomotor a nd
sensory compl ications of spinal cord in jury via altering microgl ial activity:
introduction of Roumilast as an alter native therapy. Int Immun opharm 2020;
86:106743.
[35] Vanmierlo T, Creemers P, Akkerman S, van Duinen M, Sambeth A, De Vry J, et al.
The PDE4 inhibitor roumilast improves memory in rodents at non-emetic doses.
Behav Brain Res 2016;303:2633.
[36] Richter W, Menniti FS, Zhang HT, Conti M. PDE4 as a target for cognition
enhancement. Expert Opin Ther Targets 2013;17(9):101127.
[37] Davies B, Mowforth OD, Yordanov S, Alvarez-Berdugo D, Bond S, Nodale M, et al.
Targeting patient recovery priorities in degenerative cervical myelopathy: design
and rationale for the RECEDE-Myelopathy trial-study protocol. BMJ Open 2023;
13(3):e061294.
[38] Schepers M, Paes D, Tiane A, Rombaut B, Piccart E, van Veggel L, et al. Selective
PDE4 subtype inhibition provides new opportunities to intervene in
neuroinammatory versus myelin damaging hallmarks of multiple sclerosis. Brain
Behav Immun 2023;109:122.
[39] Titus DJ, Wilson NM, Freund JE, Carballosa MM, Sikah KE, Furones C, et al. Chronic
cognitive dysfunction after traumatic brain injury is improved with a
phosphodiesterase 4B inhibitor. J Neurosci 2016;36(27):7095108.
[40] Myers SA, Gobejishvili L, Saraswat Ohri S, Garrett Wilson C, Andres KR, Riegler AS,
et al. Following spinal cord injury, PDE4B drives an acute, local inammatory
response and a chronic, systemic response exacerbated by gut dysbiosis and
endotoxemia. Neurobiol Dis 2019;124:35363.
[41] Sanabra C, Johansson EM, Mengod G. Critical role for PDE4 subfamilies in the
development of experimental autoimmune encephalomyelitis. J Chem Neuroanat
2013;47:96105.
[42] Wilson NM, Gurney ME, Dietrich WD, Atkins CM. Therapeutic benets of
phosphodiesterase 4B inhibition after traumatic brain injury. PLoS One 2017;12(5):
e0178013.
[43] Ghosh M, Xu Y, Pearse DD. Cyclic AMP is a key regulator of M1 to M2a phenotypic
conversion of microglia in the presence of Th2 cytokines. J Neuroinammation
2016;13:9.
[44] Erdely A, Kepka-Lenhart D, Clark M, Zeidler-Erdely P, Poljakovic M, Calhoun WJ,
et al. Inhibition of phosphodiesterase 4 amplies cytokine-dependent induction of
arginase in macrophages. Am J Physiol Lung Cell Mol Physiol 2006;290(3):L5349.
[45] Silver J, Miller JH. Regeneration beyond the glial scar. Nat Rev Neurosci 2004;5(2):
14656.
[46] Kanemaru K, Kubota J, Sekiya H, Hirose K, Okubo Y, Iino M. Calcium-dependent N-
cadherin up-regulation mediates reactive astrogliosis and neuroprotection after
brain injury. Proc Natl Acad Sci USA 2013;110(28):116127.
[47] Fawcett JW, Asher RA. The glial scar and central nervous system repair. Brain Res
Bull 1999;49(6):37791.
[48] Sofroniew MV. Molecular dissection of reactive astrogliosis and glial scar formation.
Trends Neurosci 2009;32(12):63847.
[49] Costa LM, Pereira JE, Filipe VM, Magalhaes LG, Couto PA, Gonzalo-Orden JM, et al.
Rolipram promotes functional recovery after contusive thoracic spinal cord injury
in rats. Behav Brain Res 2013;243:6673.
[50] Sharif-Alhoseini M, Khormali M, Rezaei M, Safdarian M, Hajighadery A,
Khalatbari MM, et al. Animal models of spinal cord injury: a systematic review.
Spinal Cord 2017;55(8):71421.
[51] Pedro KM, Fehlings MG. Time is spine: what's over the horizon. J Clin Orthop
Trauma 2022;35:102043.
[52] Badhiwala JH, Ahuja CS, Fehlings MG. Time is spine: a review of translational
advances in spinal cord injury. J Neurosurg Spine 2018;30(1):118.
[53] Perrin FE, Noristani HN. Serotonergic mechanisms in spinal cord injury. Exp Neurol
2019;318:17491.
[54] Panov AV, Kubalik N, Zinchenko N, Ridings DM, Radoff DA, Hemendinger R, et al.
Metabolic and functional differences between brain and spinal cord mitochondria
underlie different predisposition to pathology. Am J Physiol Regul Integr Comp
Physiol 2011;300(4):R84454.
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
11
[55] Psterer U, Khodosevich K. Neuronal survival in the brain: neuron type-specic
mechanisms. Cell Death Dis 2017;8(3):e2643.
[56] Liu Y, Zhou LJ, Wang J, Li D, Ren WJ, Peng J, et al. TNF-Alpha differentially regulates
synaptic plasticity in the Hippocampus and spinal cord by microglia-dependent
mechanisms after peripheral nerve injury. J Neurosci 2017;37(4):87181.
[57] Montoya GJ, Sutachan JJ, Chan WS, Sideris A, Blanck TJ, Recio-Pinto E. Muscle-
conditioned media and cAMP promote survival and neurite outgrowth of adult
spinal cord motor neurons. Exp Neurol 2009;220(2):30315.
[58] Aglah C, Gordon T, Posse de Chaves EI. cAMP promotes neurite outgrowth and
extension through protein kinase A but independently of Erk activation in cultured
rat motoneurons. Neuropharmacology 2008;55(1):817.
[59] Freyermuth-Trujillo X, Segura-Uribe JJ, Salgado-Ceballos H, Orozco-Barrios CE,
Coyoy-Salgado A. Inammation: a target for treatment in spinal cord injury. Cells
2022;11(17).
[60] Orr MB, Gensel JC . Spinal cord injury scarring and inammation: therapies
targeting glial a nd inammatory responses. Ne urotherapeutics 2018;15 (3):
54153.
[61] Lin ZH, Wang SY, Chen LL, Zhuang JY, Ke QF, Xiao DR, et al. Methylene blue
mitigates acute neuroinammation after spinal cord injury through inhibiting
NLRP3 inammasome activation in microglia. Front Cell Neurosci 2017;11:
391.
M. Schepers et al. Neurotherapeutics 21 (2024) e00372
12
... Apremilast was approved by the FDA in March 2014 for patients with psoriatic arthritis [33][34][35]. Spinal cord injury (SCI) is a life-altering event that significantly impacts the patient's quality of life [41]. Modulating neuroinflammation, which worsens the initial injury, and promoting neuroregenerative repair mechanisms are crucial strategies for enhancing functional recovery. ...
... Decreased levels of cAMP are observed following SCI, highlighting the importance of preventing its degradation to reduce inflammation and promote regeneration. Inhibiting specific PDE4 subtypes, such as PDE4B and PDE4D, has shown therapeutic potential for managing SCI pathology [41]. A recent study investigated the effects of inhibiting PDE4B and PDE4D on inflammatory and regenerative processes post-SCI [41]. ...
... Inhibiting specific PDE4 subtypes, such as PDE4B and PDE4D, has shown therapeutic potential for managing SCI pathology [41]. A recent study investigated the effects of inhibiting PDE4B and PDE4D on inflammatory and regenerative processes post-SCI [41]. Results indicate that treatment with the PDE4D inhibitor Gebr32a, initiated even at 2 days post-injury, improves functional and histopathological outcomes after SCI, similar to the effects of the full PDE4 inhibitor roflumilast [41]. ...
Article
The selective phosphodiesterase 4 D enzymes play a crucial role in regulating signaling through the cyclic adenosinmonophosphate second messenger system by hydrolyzing cyclic nucleotides. The PDE4 gene family includes four subtypes, PDE4A-D, distinguished by the presence of conserved regions called upstream conserved regions UCR1 and UCR2. These enzymes can exist as dimers or monomers, with UCR1 facilitating dimerization and UCR2 controlling enzyme activity by modulating access to cAMP. Dimeric isoforms exhibit increased activity in response to cAMP signaling through PKA-mediated phosphorylation of UCR1. Mutations in the PDE4D gene have been linked to the rare neurodevelopmental disorder acrodysostosis-2 (ACRDYS2), characterized by intellectual disability and brachydactyly. These mutations, predominantly missense mutations on the protein surface, disrupt protein kinase A phosphorylation sites or alter interactions between UCR2 and the catalytic domain, affecting enzyme activity. Some mutations at the dimerization site increase basal enzyme activity. Genetic variations in PDE4D also influence human cognitive abilities, as evidenced by GWAS studies linking allelic variation in the gene's 5' exons encoding dimeric forms to cognitive function. This highlights the significance of dimeric PDE4D isoforms in normal brain function, both in rare disorders like ACRDYS2 and in common genetic variants associated with cognitive abilities. Blocking PDE4D can boost signaling through the cAMP-PKA-SIRT1-Akt-Bcl-2/Bax pathway, potentially offering therapeutic advantages in neurocognitive disorders. We focus on the role of selective phosphodiesterase 4D (PDE4D) allosteric inhibitors for the treatment of fragile X mental retardation protein (FMR-1) gene defects and other brain disorders in childhood with focus on spinal cord injury in childhood, Down syndrome, Angelman syndrome, Rett syndrome and Prader Willi syndrome.
Article
Full-text available
Study design Systematic review. Objective The objective of this study was to evaluate the impact of phosphodiesterase (PDE) inhibitors on neurobehavioral outcomes in preclinical models of traumatic and non-traumatic spinal cord injury (SCI). Methods A systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines and was registered with PROSPERO (CRD42019150639). Searches were performed in MEDLINE and Embase. Studies were included if they evaluated the impact of PDE inhibitors on neurobehavioral outcomes in preclinical models of traumatic or non-traumatic SCI. Data were extracted from relevant studies, including sample characteristics, injury model, and neurobehavioral assessment and outcomes. Risk of bias was assessed using the SYRCLE checklist. Results The search yielded a total of 1,679 studies, of which 22 met inclusion criteria. Sample sizes ranged from 11 to 144 animals. PDE inhibitors used include rolipram ( n = 16), cilostazol ( n = 4), roflumilast ( n = 1), and PDE4-I ( n = 1). The injury models used were traumatic SCI ( n = 18), spinal cord ischemia ( n = 3), and degenerative cervical myelopathy ( n = 1). The most commonly assessed outcome measures were Basso, Beattie, Bresnahan (BBB) locomotor score ( n = 13), and grid walking ( n = 7). Of the 22 papers that met the final inclusion criteria, 12 showed a significant improvement in neurobehavioral outcomes following the use of PDE inhibitors, four papers had mixed findings and six found PDE inhibitors to be ineffective in improving neurobehavioral recovery following an SCI. Notably, these findings were broadly consistent across different PDE inhibitors and spinal cord injury models. Conclusion In preclinical models of traumatic and non-traumatic SCI, the administration of PDE inhibitors appeared to be associated with statistically significant improvements in neurobehavioral outcomes in a majority of included studies. However, the evidence was inconsistent with a high risk of bias. This review provides a foundation to aid the interpretation of subsequent clinical trials of PDE inhibitors in spinal cord injury. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=150639 , identifier: CRD42019150639.
Article
Full-text available
Inhibition of phosphodiesterase 4D (PDE4D) enzymes has been investigated as therapeutic strategy to treat memory problems in Alzheimer’s disease (AD). Although PDE4D inhibitors are effective in enhancing memory processes in rodents and humans, severe side effects may hamper their clinical use. PDE4D enzymes comprise different isoforms, which, when targeted specifically, can increase treatment efficacy and safety. The function of PDE4D isoforms in AD and in molecular memory processes per se has remained unresolved. Here, we report the upregulation of specific PDE4D isoforms in transgenic AD mice and hippocampal neurons exposed to amyloid-β. Furthermore, by means of pharmacological inhibition and CRISPR-Cas9 knockdown, we show that the long-form PDE4D3, -D5, -D7, and -D9 isoforms regulate neuronal plasticity and convey resilience against amyloid-β in vitro. These results indicate that isoform-specific, next to non-selective, PDE4D inhibition is efficient in promoting neuroplasticity in an AD context. Therapeutic effects of non-selective PDE4D inhibitors are likely achieved through actions on long isoforms. Future research should identify which long PDE4D isoforms should be specifically targeted in vivo to both improve treatment efficacy and reduce side effects. Supplementary Information The online version contains supplementary material available at 10.1007/s00018-023-04804-w.
Article
Full-text available
Traumatic spinal cord injury (SCI) is characterized by severe neuroinflammation and hampered neuroregeneration, which often leads to permanent neurological deficits. Current therapies include decompression surgery, rehabilitation, and in some instances, the use of corticosteroids. However, the golden standard of corticosteroids still achieves minimal improvements in functional outcomes. Therefore, new strategies tackling the initial inflammatory reactions and stimulating endogenous repair in later stages are crucial to achieving functional repair in SCI patients. Cyclic adenosine monophosphate (cAMP) is an important second messenger in the central nervous system (CNS) that modulates these processes. A sustained drop in cAMP levels is observed during SCI, and elevating cAMP is associated with improved functional outcomes in experimental models. cAMP is regulated in a spatiotemporal manner by its hydrolyzing enzyme phosphodiesterase (PDE). Growing evidence suggests that inhibition of cAMP-specific PDEs (PDE4, PDE7, and PDE8) is an important strategy to orchestrate neuroinflammation and regeneration in the CNS. Therefore, this review focuses on the current evidence related to the immunomodulatory and neuroregenerative role of cAMP-specific PDE inhibition in the SCI pathophysiology.
Article
Full-text available
Introduction Degenerative cervical myelopathy (DCM) is a common and disabling condition of symptomatic cervical spinal cord compression secondary to degenerative changes in spinal structures leading to a mechanical stress injury of the spinal cord. RECEDE-Myelopathy aims to test the disease-modulating activity of the phosphodiesterase 3/phosphodiesterase 4 inhibitor Ibudilast as an adjuvant to surgical decompression in DCM. Methods and analysis RECEDE-Myelopathy is a multicentre, double-blind, randomised, placebo-controlled trial. Participants will be randomised to receive either 60–100 mg Ibudilast or placebo starting within 10 weeks prior to surgery and continuing for 24 weeks after surgery for a maximum of 34 weeks. Adults with DCM, who have a modified Japanese Orthopaedic Association (mJOA) score 8–14 inclusive and are scheduled for their first decompressive surgery are eligible for inclusion. The coprimary endpoints are pain measured on a visual analogue scale and physical function measured by the mJOA score at 6 months after surgery. Clinical assessments will be undertaken preoperatively, postoperatively and 3, 6 and 12 months after surgery. We hypothesise that adjuvant therapy with Ibudilast leads to a meaningful and additional improvement in either pain or function, as compared with standard routine care. Study design Clinical trial protocol V.2.2 October 2020. Ethics and dissemination Ethical approval has been obtained from HRA—Wales.The results will be presented at an international and national scientific conferences and in a peer-reviewed journals. Trial registration number ISRCTN Number: ISRCTN16682024 .
Article
Full-text available
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterized by focal inflammatory lesions and prominent demyelination. Even though the currently available therapies are effective in treating the initial stages of disease, they are unable to halt or reverse disease progression into the chronic progressive stage. Thus far, no repair-inducing treatments are available for progressive MS patients. Hence, there is an urgent need for the development of new therapeutic strategies either targeting the destructive immunological demyelination or boosting endogenous repair mechanisms. Using in vitro, ex vivo, and in vivo models, we demonstrate that selective inhibition of phosphodiesterase 4 (PDE4), a family of enzymes that hydrolyzes and inactivates cyclic adenosine monophosphate (cAMP), reduces inflammation and promotes myelin repair. More specifically, we segregated the myelination-promoting and anti-inflammatory effects into a PDE4D- and PDE4B-dependent process respectively. We show that inhibition of PDE4D boosts oligodendrocyte progenitor cells (OPC) differentiation and enhances (re)myelination of both murine OPCs and human iPSC-derived OPCs. In addition, PDE4D inhibition promotes in vivo remyelination in the cuprizone model, which is accompanied by improved spatial memory and reduced visual evoked potential latency times. We further identified that PDE4B-specific inhibition exerts anti-inflammatory effects since it lowers in vitro monocytic nitric oxide (NO) production and improves in vivo neurological scores during the early phase of experimental autoimmune encephalomyelitis (EAE). In contrast to the pan PDE4 inhibitor roflumilast, the therapeutic dose of both the PDE4B-specific inhibitor A33 and the PDE4D-specific inhibitor Gebr32a did not trigger emesis-like side effects in rodents. Finally, we report distinct pde4d isoform expression patterns in human area postrema neurons and human oligodendroglia lineage cells. Using the CRISPR-Cas9 system, we confirmed that PDE4D1/2 and PDE4D6 are the key targets to induce OPC differentiation. Collectively, these data demonstrate that gene specific PDE4 inhibitors have potential as novel therapeutic agents for targeting the distinct disease processes of MS.
Article
Full-text available
Spinal cord injury (SCI) is a significant cause of disability, and treatment alternatives that generate beneficial outcomes and have no side effects are urgently needed. SCI may be treatable if intervention is initiated promptly. Therefore, several treatment proposals are currently being evaluated. Inflammation is part of a complex physiological response to injury or harmful stimuli induced by mechanical, chemical, or immunological agents. Neuroinflammation is one of the principal secondary changes following SCI and plays a crucial role in modulating the pathological progression of acute and chronic SCI. This review describes the main inflammatory events occurring after SCI and discusses recently proposed potential treatments and therapeutic agents that regulate inflammation after insult in animal models.
Article
Full-text available
Background Spinal cord injury (SCI) elicits a robust neuroinflammatory reaction which, in turn, exacerbates the initial mechanical damage. Pivotal players orchestrating this response are macrophages (Mφs) and microglia. After SCI, the inflammatory environment is dominated by pro-inflammatory Mφs/microglia, which contribute to secondary cell death and prevent regeneration. Therefore, reprogramming Mφ/microglia towards a more anti-inflammatory and potentially neuroprotective phenotype has gained substantial therapeutic interest in recent years. Interleukin-13 (IL-13) is a potent inducer of such an anti-inflammatory phenotype. In this study, we used genetically modified Mφs as carriers to continuously secrete IL-13 (IL-13 Mφs) at the lesion site. Methods Mφs were genetically modified to secrete IL-13 (IL-13 Mφs) and were phenotypically characterized using qPCR, western blot, and ELISA. To analyze the therapeutic potential, the IL-13 Mφs were intraspinally injected at the perilesional area after hemisection SCI in female mice. Functional recovery and histopathological improvements were evaluated using the Basso Mouse Scale score and immunohistochemistry. Neuroprotective effects of IL-13 were investigated using different cell viability assays in murine and human neuroblastoma cell lines, human neurospheroids, as well as murine organotypic brain slice cultures. Results In contrast to Mφs prestimulated with recombinant IL-13, perilesional transplantation of IL-13 Mφs promoted functional recovery following SCI in mice. This improvement was accompanied by reduced lesion size and demyelinated area. The local anti-inflammatory shift induced by IL-13 Mφs resulted in reduced neuronal death and fewer contacts between dystrophic axons and Mφs/microglia, suggesting suppression of axonal dieback. Using IL-4Rα-deficient mice, we show that IL-13 signaling is required for these beneficial effects. Whereas direct neuroprotective effects of IL-13 on murine and human neuroblastoma cell lines or human neurospheroid cultures were absent, IL-13 rescued murine organotypic brain slices from cell death, probably by indirectly modulating the Mφ/microglia responses. Conclusions Collectively, our data suggest that the IL-13-induced anti-inflammatory Mφ/microglia phenotype can preserve neuronal tissue and ameliorate axonal dieback, thereby promoting recovery after SCI.
Article
The overarching theme in the early treatment of acute spinal cord injury (SCI) is to reduce the extent of secondary damage to facilitate early neurological and functional recovery. Although multiple studies have brought us innovative and potential new therapies to treat SCI, ameliorating neural damage remains a formidable challenge. Knowledge translation of clinical and basic research studies has shown that surgical intervention is a valuable modality of treatment; however, the role, timing and optimal technique in surgery remains a topic of great controversy. While evidence to support the concept of ultra-early surgery for acute SCI continues to emerge, current protocols and international guidelines that encourage reducing time from trauma to surgery support the concept of “Time is Spine”. The present article provides a critical narrative review of the current best practice, with a particular focus on the timing of surgical intervention, that shapes our understanding of how time is of the essence in the management of acute SCI.
Article
Despite the considerable impact of stroke on both the individual and on society, a neuroprotective therapy for stroke patients is missing. This is partially due to the current lack of a physiologically relevant human in vitro stroke model. To address this problem, we have developed a luminescent human iPSC-derived neurospheroid model that enables real-time read-out of neural viability after ischemia-like conditions. We subjected 1- and 4-week-old neurospheroids, generated from iPSC-derived neural stem cells, to 6 h of oxygen–glucose deprivation (OGD) and measured neurospheroid luminescence. For both, we detected a decrease in luminescent signal due to ensuing neurotoxicity, as confirmed by conventional LDH assay and flow cytometric viability analysis. Remarkably, 1-week-old, but not 4-week-old neurospheroids recovered from OGD-induced injury, as evidenced by their reduced but overall increasing luminescence over time. This underscores the need for more mature neurospheroids, more faithfully recapitulating the in vivo situation. Furthermore, treatment of oxygen- and glucose-deprived neurospheroids with the pan-caspase inhibitor Z-VAD-FMK did not increase overall neural survival, despite its successful attenuation of apoptosis, in a human-based 3D environment. Nevertheless, owing to its three-dimensional organization and real-time viability reporting potential, the luminescent neurospheroids may become readily adopted in high-throughput screens aimed at identification of new therapeutic agents to treat acute ischemic stroke patients.