ArticlePDF AvailableLiterature Review

Modulation of Second Messenger Signaling in the Brain Through PDE4 and PDE5 Inhibition: Therapeutic Implications for Neurological Disorders

MDPI
Cells
Authors:

Abstract and Figures

Phosphodiesterase (PDE) enzymes regulate intracellular signaling pathways crucial for brain development and the pathophysiology of neurological disorders. Among the 11 PDE subtypes, PDE4 and PDE5 are particularly significant due to their regulation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) signaling, respectively, which are vital for learning, memory, and neuroprotection. This review synthesizes current evidence on the roles of PDE4 and PDE5 in neurological health and disease, focusing on their regulation of second messenger pathways and their implications for brain function. Elevated PDE4 activity impairs synaptic plasticity by reducing cAMP levels and protein kinase A (PKA) activity, contributing to cognitive decline, acute brain injuries, and neuropsychiatric conditions such as bipolar disorder and schizophrenia. Similarly, PDE5 dysregulation disrupts nitric oxide (NO) signaling and protein kinase G (PKG) pathways, which are involved in cerebrovascular homeostasis, recovery after ischemic events, and neurodegenerative processes in Alzheimer’s, Parkinson’s, and Huntington’s diseases. PDE4 and PDE5 are promising therapeutic targets for neurological disorders. Pharmacological modulation of these enzymes offers potential to enhance cognitive function and mitigate pathological mechanisms underlying brain injuries, neurodegenerative diseases, and psychiatric disorders. Further research into the regulation of PDE4 and PDE5 will advance therapeutic strategies for these conditions.
The diagram highlights the roles of PDE4 and PDE5 in regulating second messenger systems and their therapeutic relevance in neurological disorders [16,23]. (A) PDE4 controls cAMP levels, with its inhibition activating PKA and phosphorylating CREB, promoting neuronal survival, synaptic plasticity, cognitive function, and anti-inflammatory responses [9]. Elevated cAMP also reduces reactive oxygen species (ROS), alleviating oxidative stress [28]. (B) PDE5 inhibition prevents cGMP degradation, activating PKG, which enhances vasodilation, neuroprotection, and antioxidant defenses [34,35]. PKG also mitigates ROS production, reducing oxidative damage [34,35]. In traumatic brain injury (TBI), PDE4 and PDE5 inhibitors improve neuronal repair, reduce inflammation, and protect against oxidative stress [18,36,37]. In ischemic stroke, these inhibitors lower ROS, support neurogenesis, and facilitate recovery through CREB and PKG pathways [18,36,37,38]. In epilepsy, PDE4 inhibition regulates synaptic plasticity, while PDE5 inhibition stabilizes neuronal function by improving blood flow and reducing excitotoxicity [34,35]. In Alzheimer’s disease, combined PDE4 and PDE5 inhibition improves cognition, reduces amyloid-beta accumulation, and protects against oxidative stress [18,36,37,38]. (A,B) Together, PDE4 and PDE5 inhibitors strengthen the cAMP/CREB and cGMP/PKG pathways, offering broad neuroprotective benefits, reducing inflammation, and enhancing cognitive and synaptic function across multiple neurological conditions [2,3,9,28,39,40]. “Created with BioRender.com”.
… 
One week after epilepsy induction, brains were harvested to evaluate the effects of the PDE4 inhibitor amlexanox. NeuN staining demonstrates preserved neuronal populations in the CA1 region of the hippocampus in the amlexanox-treated group, indicating a neuroprotective effect [26]. (A) Immunofluorescence images show overexpression of PDE4B within neurons in the hippocampal CA1 region 1 week after epilepsy, detected using PDE4B immunohistochemistry with specific antibodies PDE4B [26]. PDE4 staining indirectly verifies the efficacy of the PDE4 inhibitor by detecting a decrease in the intensity of PDE4 expression (PDE4B, red) [26]. (B) Immunofluorescence images show overexpression of PDE4B within neurons in the hippocampal CA1 region 1 week after epilepsy, detected using IL-6 and TNF-α immunohistochemistry with specific antibodies IL-6, TNF-α [26]. IL-6 and TNF-α staining indirectly verifies the efficacy of the PDE4 inhibitor by detecting a decrease in the intensity of inflammation expression (TNF-α, red) (IL-6, green) [26]. (C) COX-2, the final inflammatory marker, was confirmed to have a decreased protein expression level by PDE4 inhibitors through Western blot [26]. (D) Immunofluorescence images show live neurons in the hippocampal CA1 region 1 week after epilepsy, detected using NeuN [26]. It was confirmed that more neurons survived when PDE4 inhibitors were administered after epilepsy was induced [26]. COX-2 Western blot, along with TNF-α and IL-6 immunofluorescence, reveals decreased levels of pro-inflammatory markers, suggesting anti-inflammatory effects [26]. These findings underscore the potential of amlexanox to mitigate neuronal damage and inflammation in epilepsy [26].
… 
Content may be subject to copyright.
Academic Editors: Paul M. Epstein,
Michy P. Kelly, Stefan Brocke and
Leila Gobejishvili
Received: 11 December 2024
Revised: 7 January 2025
Accepted: 7 January 2025
Published: 9 January 2025
Citation: Park, M.K.; Yang, H.W.;
Woo, S.Y.; Kim, D.Y.; Son, D.-S.; Choi,
B.Y.; Suh, S.W. Modulation of Second
Messenger Signaling in the Brain
Through PDE4 and PDE5 Inhibition:
Therapeutic Implications for
Neurological Disorders. Cells 2025,14,
86. https://doi.org/10.3390/
cells14020086
Copyright: © 2025 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license
(https://creativecommons.org/
licenses/by/4.0/).
Review
Modulation of Second Messenger Signaling in the Brain
Through PDE4 and PDE5 Inhibition: Therapeutic Implications
for Neurological Disorders
Min Kyu Park 1, Hyun Wook Yang 1, Seo Young Woo 1, Dong Yeon Kim 1, Dae-Soon Son 2, Bo Young Choi 3,4
and Sang Won Suh 1,*
1Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea;
bagmingyu50@gmail.com (M.K.P.); akqjqtj5@hallym.ac.kr (H.W.Y.); 1wsy@naver.com (S.Y.W.);
roy8596@naver.com (D.Y.K.)
2Division of Data Science, Data Science Convergence Research Center, Hallym University,
Chuncheon 24252, Republic of Korea; biostat@hallym.ac.kr
3Institute of Sport Science, Hallym University, Chuncheon 24252, Republic of Korea; bychoi@hallym.ac.kr
4Department of Physical Education, Hallym University, Chuncheon 24252, Republic of Korea
*Correspondence: swsuh@hallym.ac.kr
Abstract: Phosphodiesterase (PDE) enzymes regulate intracellular signaling pathways
crucial for brain development and the pathophysiology of neurological disorders. Among
the 11 PDE subtypes, PDE4 and PDE5 are particularly significant due to their regula-
tion of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate
(cGMP) signaling, respectively, which are vital for learning, memory, and neuroprotection.
This review synthesizes current evidence on the roles of PDE4 and PDE5 in neurologi-
cal health and disease, focusing on their regulation of second messenger pathways and
their implications for brain function. Elevated PDE4 activity impairs synaptic plasticity
by reducing cAMP levels and protein kinase A (PKA) activity, contributing to cognitive
decline, acute brain injuries, and neuropsychiatric conditions such as bipolar disorder and
schizophrenia. Similarly, PDE5 dysregulation disrupts nitric oxide (NO) signaling and
protein kinase G (PKG) pathways, which are involved in cerebrovascular homeostasis,
recovery after ischemic events, and neurodegenerative processes in Alzheimer’s, Parkin-
son’s, and Huntington’s diseases. PDE4 and PDE5 are promising therapeutic targets for
neurological disorders. Pharmacological modulation of these enzymes offers potential
to enhance cognitive function and mitigate pathological mechanisms underlying brain
injuries, neurodegenerative diseases, and psychiatric disorders. Further research into the
regulation of PDE4 and PDE5 will advance therapeutic strategies for these conditions.
Keywords: phosphodiesterase (PDE); phosphodiesterase 4 (PDE4); phosphodiesterase
5 (PDE5); protein kinase A (PKA); protein kinase G (PKG)
1. Introduction
Phosphodiesterases (PDEs) are enzymes essential for cellular signaling, responsible
for breaking down cyclic nucleotides like cyclic adenosine monophosphate (cAMP) and
cyclic guanosine monophosphate (cGMP) [
1
]. These cyclic nucleotides act as key second
messengers in numerous physiological processes, including metabolism, vascular regula-
tion, and neuronal signaling [
1
]. The PDE family is diverse, consisting of 11 subfamilies
(PDE1–PDE11), each with distinct tissue distribution, substrate specificity, and regulatory
mechanisms, reflecting their specialized roles in various biological systems [1].
Cells 2025,14, 86 https://doi.org/10.3390/cells14020086
Cells 2025,14, 86 2 of 14
Among the PDE family, PDE4 and PDE5 have garnered particular interest due to their
involvement in brain function and neurological disorders [
2
,
3
]. PDE4 primarily regulates
cAMP levels in neurons, influencing intracellular signaling pathways that govern synaptic
plasticity, neuronal survival, and memory formation [47].
Dysregulation of PDE4 and PDE5 activity has been implicated in the pathology of
several neurodegenerative and neuropsychiatric disorders, including Alzheimer’s disease,
stroke, and cognitive impairments [
3
,
8
12
]. As a result, pharmacological inhibitors tar-
geting these enzymes have emerged as promising therapeutic strategies. PDE4 inhibitors
enhance cAMP signaling, promoting neuroprotection and cognitive improvements, while
PDE5 inhibitors restore cGMP signaling, supporting cerebral blood flow, reducing oxidative
stress, and mitigating neuronal damage [3,812].
This review focuses on the roles of PDE4 and PDE5 in neurological health and disease,
highlighting their mechanisms of action, therapeutic potential, and the latest advancements
in pharmacological modulation.
2. Distribution and Functional Roles of PDE4 and PDE5 Isoforms in the
Central Nervous System and Peripheral Tissues
2.1. PDE4 Isoforms in the CNS and Peripheral Tissues
The PDE4 family includes four isoforms PDE4A, PDE4B, PDE4C, and PDE4D each
exhibiting distinct expression patterns and physiological roles [
13
,
14
]. In the CNS, PDE4
isoforms are broadly expressed in neurons and glial cells and are notably persistent in
aged and Alzheimer’s disease-affected brains, suggesting potential roles in neurodegener-
ation [
15
,
16
]. Among these, PDE4B is the most widely distributed, with high expression
in brain regions such as the cortex, hippocampus, and cerebellum [
17
19
]. PDE4A is also
abundant in the cortex but is expressed at two to four times lower levels in other areas [
17
].
PDE4D is prominent in the frontal cortex but is generally less abundant than PDE4B in
most CNS regions. Conversely, PDE4C is minimally expressed in the brain, indicating a
limited or specialized role [17] (Figure 1).
Cells 2025, 14, x FOR PEER REVIEW 2 of 15
PDE11), each with distinct tissue distribution, substrate specificity, and regulatory mecha-
nisms, reflecting their specialized roles in various biological systems [1].
Among the PDE family, PDE4 and PDE5 have garnered particular interest due to their
involvement in brain function and neurological disorders [2,3]. PDE4 primarily regulates
cAMP levels in neurons, influencing intracellular signaling pathways that govern synaptic
plasticity, neuronal survival, and memory formation [4–7].
Dysregulation of PDE4 and PDE5 activity has been implicated in the pathology of sev-
eral neurodegenerative and neuropsychiatric disorders, including Alzheimer’s disease,
stroke, and cognitive impairments [3,8–12]. As a result, pharmacological inhibitors targeting
these enzymes have emerged as promising therapeutic strategies. PDE4 inhibitors enhance
cAMP signaling, promoting neuroprotection and cognitive improvements, while PDE5 in-
hibitors restore cGMP signaling, supporting cerebral blood flow, reducing oxidative stress,
and mitigating neuronal damage [3,8–12].
This review focuses on the roles of PDE4 and PDE5 in neurological health and disease,
highlighting their mechanisms of action, therapeutic potential, and the latest advancements
in pharmacological modulation.
2. Distribution and Functional Roles of PDE4 and PDE5 Isoforms in the
Central Nervous System and Peripheral Tissues
2.1. PDE4 Isoforms in the CNS and Peripheral Tissues
The PDE4 family includes four isoforms PDE4A, PDE4B, PDE4C, and PDE4D each ex-
hibiting distinct expression patterns and physiological roles [13,14]. In the CNS, PDE4
isoforms are broadly expressed in neurons and glial cells and are notably persistent in aged
and Alzheimer’s disease-affected brains, suggesting potential roles in neurodegeneration
[15,16]. Among these, PDE4B is the most widely distributed, with high expression in brain
regions such as the cortex, hippocampus, and cerebellum [17–19]. PDE4A is also abundant
in the cortex but is expressed at two to four times lower levels in other areas [17]. PDE4D is
prominent in the frontal cortex but is generally less abundant than PDE4B in most CNS
regions. Conversely, PDE4C is minimally expressed in the brain, indicating a limited or spe-
cialized role [17] (Figure 1).
Figure 1. The chemical structures of cyclic adenosine monophosphate (cAMP) and adenosine mono-
phosphate (AMP) [20]. Phosphodiesterase type 4 (PDE4) catalyzes the hydrolysis of cAMP into AMP
[20]. In cAMP, the 3 and 5 hydroxyl groups of the adenosine ribose are connected by a phosphodiester
bond, forming its characteristic cyclic structure [20]. PDE4 cleaves the phosphodiester bond at the 3
position, disrupting the cyclic structure and resulting in the formation of a linear AMP molecule [20].
Figure 1. The chemical structures of cyclic adenosine monophosphate (cAMP) and adenosine
monophosphate (AMP) [
20
]. Phosphodiesterase type 4 (PDE4) catalyzes the hydrolysis of cAMP
into AMP [
20
]. In cAMP, the 3
and 5
hydroxyl groups of the adenosine ribose are connected by a
phosphodiester bond, forming its characteristic cyclic structure [
20
]. PDE4 cleaves the phosphodiester
bond at the 3
position, disrupting the cyclic structure and resulting in the formation of a linear AMP
molecule [
20
]. This hydrolysis reaction involves the addition of water, yielding AMP and inorganic
phosphate (Pi) as final products [20].
Cells 2025,14, 86 3 of 14
In peripheral tissues, PDE4B and PDE4D are predominant, contributing to immune
regulation, inflammation, and vascular function. Dysregulation of PDE4 activity has been
associated with psychiatric conditions, including depression and schizophrenia, due to its
influence on cAMP signaling pathways [
17
19
]. PDE4 also regulates ion channels involved
in neuronal excitability, underscoring its importance in maintaining neural communication
and preventing hyperactivity [1719].
2.2. PDE5 Isoforms in the CNS and Peripheral Tissues
Unlike PDE4, PDE5 primarily exists as a single isoform. Its expression is robust in
peripheral tissues, including vascular smooth muscle, platelets, and the gastrointesti-
nal tract, where it regulates vascular tone, platelet aggregation, and smooth muscle
contraction [3,2123]
. In the CNS, PDE5 expression is relatively limited but regionally
specific [
3
,
21
23
]. It is prominently expressed in the hippocampus, cerebellum, and cortex,
regions critical for cognitive functions and synaptic plasticity [
3
,
21
23
]. In the hippocampus,
PDE5 is involved in processes such as memory formation and long-term potentiation (LTP),
mediated by the cGMP-dependent nitric oxide (NO)-cGMP signaling pathway [
3
,
21
23
].
Moderate expression in the cerebellum suggests a role in motor coordination, while cortical
distribution varies based on localized cGMP signaling demands [3,2123] (Figure 2).
Figure 2. The chemical structures of cyclic guanosine monophosphate (cGMP) and guanosine
monophosphate (GMP) [
24
]. Phosphodiesterase type 5 (PDE5) catalyzes the hydrolysis of cGMP
into GMP. In cGMP, a phosphodiester bond links the 3
and 5
hydroxyl groups of the guanosine
ribose, creating its cyclic structure [
24
]. In cGMP, a phosphodiester bond links the 3
and 5
hydroxyl
groups of the guanosine ribose, forming a cyclic structure [
24
]. PDE5 cleaves this bond at the 3
position, breaking the cyclic structure and converting cGMP into its linear form, GMP [
24
]. This
hydrolysis reaction involves the addition of water, producing GMP and inorganic phosphate (Pi) as
final products [24].
2.3. Integration of Roles in Signaling and Pathophysiology
PDE4 and PDE5 regulate complementary but distinct pathways, with PDE4 focusing
on cAMP and PDE5 on cGMP. Both enzymes ensure the balance of second messengers
critical for neuronal signaling and plasticity [
25
,
26
]. PDE4 activity governs PKA-mediated
phosphorylation of targets like CREB, influencing gene expression essential for learning,
memory, and neuronal survival [
25
,
26
]. Similarly, PDE5 modulates PKG activation through
NO-cGMP signaling, affecting synaptic transmission and neurovascular coupling [25,26].
The dysregulation of these pathways has significant pathological implications. In the
CNS, altered PDE4 activity has been linked to neuropsychiatric disorders, while PDE5
dysfunction is implicated in neurodegenerative conditions such as Alzheimer’s disease and
Cells 2025,14, 86 4 of 14
cerebrovascular disorders like stroke [
3
,
27
,
28
]. Notably, PDE5 regulates cerebral blood flow
by modulating vascular tone, playing a crucial role in brain homeostasis and protection
against ischemic injury [28].
2.4. Therapeutic Implications
The distinct and region-specific roles of PDE4 and PDE5 in the CNS present unique
opportunities for therapeutic targeting. PDE4B and PDE4D, due to their prominence in
synaptic function, may be viable targets for treating psychiatric and neurodegenerative
disorders [
25
28
]. Similarly, PDE5’s targeted expression in cognitive-related brain regions
highlights its potential for interventions aimed at memory enhancement and neuroprotec-
tion. By selectively modulating these isoforms, therapies can address disorders linked to
dysregulated cAMP or cGMP signaling while minimizing off-target effects [2528].
3. Therapeutic Potential of PDE4 and PDE5 in Neurological Disorders
Among the diverse phosphodiesterase subtypes, PDE4 and PDE5 have garnered sig-
nificant attention due to their crucial involvement in the pathophysiology of several brain
disorders. Inhibition of these enzymes shows promising therapeutic potential, particularly
in managing traumatic brain injury (TBI), ischemic stroke, and epilepsy [
1
,
26
]. By modu-
lating intracellular cAMP and cGMP levels, targeted interventions involving PDE4 and
PDE5 may provide therapeutic advantages for both acute and progressive neurological
conditions [29].
PKA and PKG pathways, activated by cAMP and cGMP respectively, play essential
roles in cellular processes associated with brain repair and neuroprotection [
30
]. PKA
activity contributes to synaptic repair, reduces neuronal apoptosis, attenuates neuroinflam-
mation, and helps preserve the integrity of the blood-brain barrier, all of which are essential
for recovery following brain injury [
31
,
32
]. In parallel, PKG signaling mitigates oxidative
stress and combats mitochondrial dysfunction, key pathological features observed in TBI
and other neurodegenerative conditions [
33
]. This dual modulation of PKA and PKG
pathways offers a promising framework for developing therapeutic strategies targeting
PDE4 and PDE5 to mitigate brain injury, restore neural function, and protect against future
degeneration (Figure 3).
Cells 2025, 14, x FOR PEER REVIEW 5 of 15
Figure 3. The diagram highlights the roles of PDE4 and PDE5 in regulating second messenger systems
and their therapeutic relevance in neurological disorders [16,23]. (A) PDE4 controls cAMP levels, with
its inhibition activating PKA and phosphorylating CREB, promoting neuronal survival, synaptic plas-
ticity, cognitive function, and anti-inflammatory responses [9]. Elevated cAMP also reduces reactive
oxygen species (ROS), alleviating oxidative stress [28]. (B) PDE5 inhibition prevents cGMP degrada-
tion, activating PKG, which enhances vasodilation, neuroprotection, and antioxidant defenses [34,35].
PKG also mitigates ROS production, reducing oxidative damage [34,35]. In traumatic brain injury
(TBI), PDE4 and PDE5 inhibitors improve neuronal repair, reduce inflammation, and protect against
oxidative stress [18,36,37]. In ischemic stroke, these inhibitors lower ROS, support neurogenesis, and
facilitate recovery through CREB and PKG pathways [18,36–38]. In epilepsy, PDE4 inhibition regulates
synaptic plasticity, while PDE5 inhibition stabilizes neuronal function by improving blood flow and
reducing excitotoxicity [34,35]. In Alzheimer’s disease, combined PDE4 and PDE5 inhibition improves
Figure 3. Cont.
Cells 2025,14, 86 5 of 14
Cells 2025, 14, x FOR PEER REVIEW 5 of 15
Figure 3. The diagram highlights the roles of PDE4 and PDE5 in regulating second messenger systems
and their therapeutic relevance in neurological disorders [16,23]. (A) PDE4 controls cAMP levels, with
its inhibition activating PKA and phosphorylating CREB, promoting neuronal survival, synaptic plas-
ticity, cognitive function, and anti-inflammatory responses [9]. Elevated cAMP also reduces reactive
oxygen species (ROS), alleviating oxidative stress [28]. (B) PDE5 inhibition prevents cGMP degrada-
tion, activating PKG, which enhances vasodilation, neuroprotection, and antioxidant defenses [34,35].
PKG also mitigates ROS production, reducing oxidative damage [34,35]. In traumatic brain injury
(TBI), PDE4 and PDE5 inhibitors improve neuronal repair, reduce inflammation, and protect against
oxidative stress [18,36,37]. In ischemic stroke, these inhibitors lower ROS, support neurogenesis, and
facilitate recovery through CREB and PKG pathways [18,36–38]. In epilepsy, PDE4 inhibition regulates
synaptic plasticity, while PDE5 inhibition stabilizes neuronal function by improving blood flow and
reducing excitotoxicity [34,35]. In Alzheimer’s disease, combined PDE4 and PDE5 inhibition improves
Figure 3. The diagram highlights the roles of PDE4 and PDE5 in regulating second messenger
systems and their therapeutic relevance in neurological disorders [
16
,
23
]. (A) PDE4 controls cAMP
levels, with its inhibition activating PKA and phosphorylating CREB, promoting neuronal survival,
synaptic plasticity, cognitive function, and anti-inflammatory responses [
9
]. Elevated cAMP also
reduces reactive oxygen species (ROS), alleviating oxidative stress [
28
]. (B) PDE5 inhibition prevents
cGMP degradation, activating PKG, which enhances vasodilation, neuroprotection, and antioxidant
defenses [
34
,
35
]. PKG also mitigates ROS production, reducing oxidative damage [
34
,
35
]. In traumatic
brain injury (TBI), PDE4 and PDE5 inhibitors improve neuronal repair, reduce inflammation, and
protect against oxidative stress [
18
,
36
,
37
]. In ischemic stroke, these inhibitors lower ROS, support
neurogenesis, and facilitate recovery through CREB and PKG pathways [
18
,
36
38
]. In epilepsy,
PDE4 inhibition regulates synaptic plasticity, while PDE5 inhibition stabilizes neuronal function by
improving blood flow and reducing excitotoxicity [
34
,
35
]. In Alzheimer ’s disease, combined PDE4
and PDE5 inhibition improves cognition, reduces amyloid-beta accumulation, and protects against
oxidative stress [
18
,
36
38
]. (A,B) Together, PDE4 and PDE5 inhibitors strengthen the cAMP/CREB
and cGMP/PKG pathways, offering broad neuroprotective benefits, reducing inflammation, and
enhancing cognitive and synaptic function across multiple neurological conditions [
2
,
3
,
9
,
28
,
39
,
40
].
“Created with BioRender.com”.
3.1. Seizure and Epilepsy
Seizures result from abnormal electrical activity in the brain, manifesting as a spectrum
of neurological symptoms ranging from brief lapses in attention to severe convulsions [
41
].
Epilepsy, a chronic condition characterized by recurrent seizures, significantly affects
quality of life [
42
]. The PKA and PKG pathways, which regulate neuronal excitability and
synaptic function, play pivotal roles in the development and control of seizure activity and
epilepsy [34,4346] (Figure 4).
Several phosphodiesterases (PDEs), including PDE1, PDE2, PDE3, and particularly
PDE4, contribute to cAMP degradation within neurons [
47
,
48
]. PDE4 is highly expressed in
the central nervous system (CNS) and is crucial for maintaining appropriate cAMP levels,
preventing excessive neuronal excitability driven by increased PKA activation [
43
,
47
,
48
].
Dysregulated cAMP signaling can lead to abnormal neuronal firing, contributing to the on-
set and persistence of seizures. PDE4 inhibitors reduce cAMP breakdown, modulating PKA
activity to stabilize synaptic function and potentially prevent seizure
progression [43,44]
.
These inhibitors have also demonstrated protective effects in models of pilocarpine-induced
seizures, reducing neuronal cell death, oxidative stress, and inflammation while promoting
Cells 2025,14, 86 6 of 14
lysosomal function and autophagy [
26
,
43
,
44
]. The PKG pathway, regulated by cGMP, offers
neuroprotective benefits through the modulation of nitric oxide (NO) signaling, reduction
of oxidative stress, and prevention of mitochondrial dysfunction, all of which are associated
with seizures [
26
,
43
,
44
]. PKG activation also influences ion channel function, stabilizing
neuronal excitability [
34
,
35
,
45
,
46
]. PDE5 inhibitors, which enhance PKG activity, have
shown potential to reduce seizure frequency and severity by improving neuronal resilience
and mitigating oxidative damage [34,45,46].
Cells 2025, 14, x FOR PEER REVIEW 6 of 15
cognition, reduces amyloid-beta accumulation, and protects against oxidative stress [18,36–38]. (A,B)
Together, PDE4 and PDE5 inhibitors strengthen the cAMP/CREB and cGMP/PKG pathways, offering
broad neuroprotective benefits, reducing inflammation, and enhancing cognitive and synaptic func-
tion across multiple neurological conditions [2,3,9,28,39,40]. “Created with BioRender.com”.
3.1. Seizure and Epilepsy
Seizures result from abnormal electrical activity in the brain, manifesting as a spectrum
of neurological symptoms ranging from brief lapses in attention to severe convulsions [41].
Epilepsy, a chronic condition characterized by recurrent seizures, significantly affects qual-
ity of life [42]. The PKA and PKG pathways, which regulate neuronal excitability and syn-
aptic function, play pivotal roles in the development and control of seizure activity and ep-
ilepsy [34,4346] (Figure 4).
Several phosphodiesterases (PDEs), including PDE1, PDE2, PDE3, and particularly
PDE4, contribute to cAMP degradation within neurons [47,48]. PDE4 is highly expressed in
the central nervous system (CNS) and is crucial for maintaining appropriate cAMP levels,
preventing excessive neuronal excitability driven by increased PKA activation [43,47,48].
Dysregulated cAMP signaling can lead to abnormal neuronal firing, contributing to the on-
set and persistence of seizures. PDE4 inhibitors reduce cAMP breakdown, modulating PKA
activity to stabilize synaptic function and potentially prevent seizure progression [43,44].
These inhibitors have also demonstrated protective effects in models of pilocarpine-induced
seizures, reducing neuronal cell death, oxidative stress, and inflammation while promoting
lysosomal function and autophagy [26,43,44]. The PKG pathway, regulated by cGMP, offers
neuroprotective benefits through the modulation of nitric oxide (NO) signaling, reduction
of oxidative stress, and prevention of mitochondrial dysfunction, all of which are associated
with seizures [26,43,44]. PKG activation also influences ion channel function, stabilizing
neuronal excitability [34,35,45,46]. PDE5 inhibitors, which enhance PKG activity, have
shown potential to reduce seizure frequency and severity by improving neuronal resilience
and mitigating oxidative damage [34,45,46].
Figure 4. One week after epilepsy induction, brains were harvested to evaluate the effects of the PDE4
inhibitor amlexanox. NeuN staining demonstrates preserved neuronal populations in the CA1 region
of the hippocampus in the amlexanox-treated group, indicating a neuroprotective effect [26]. (A) Im-
munofluorescence images show overexpression of PDE4B within neurons in the hippocampal CA1
region 1 week after epilepsy, detected using PDE4B immunohistochemistry with specific antibodies
Figure 4. One week after epilepsy induction, brains were harvested to evaluate the effects of
the PDE4 inhibitor amlexanox. NeuN staining demonstrates preserved neuronal populations in
the CA1 region of the hippocampus in the amlexanox-treated group, indicating a neuroprotective
effect [
26
]. (A) Immunofluorescence images show overexpression of PDE4B within neurons in the
hippocampal CA1 region 1 week after epilepsy, detected using PDE4B immunohistochemistry with
specific antibodies PDE4B [
26
]. PDE4 staining indirectly verifies the efficacy of the PDE4 inhibitor by
detecting a decrease in the intensity of PDE4 expression (PDE4B, red) [
26
]. (B) Immunofluorescence
images show overexpression of PDE4B within neurons in the hippocampal CA1 region 1 week
after epilepsy, detected using IL-6 and TNF-
α
immunohistochemistry with specific antibodies IL-6,
TNF-
α
[
26
]. IL-6 and TNF-
α
staining indirectly verifies the efficacy of the PDE4 inhibitor by detecting
a decrease in the intensity of inflammation expression (TNF-
α
, red) (IL-6, green) [
26
]. (C) COX-2,
the final inflammatory marker, was confirmed to have a decreased protein expression level by PDE4
inhibitors through Western blot [
26
]. (D) Immunofluorescence images show live neurons in the
hippocampal CA1 region 1 week after epilepsy, detected using NeuN [
26
]. It was confirmed that
more neurons survived when PDE4 inhibitors were administered after epilepsy was induced [
26
].
COX-2 Western blot, along with TNF-
α
and IL-6 immunofluorescence, reveals decreased levels of
pro-inflammatory markers, suggesting anti-inflammatory effects [
26
]. These findings underscore the
potential of amlexanox to mitigate neuronal damage and inflammation in epilepsy [26].
3.2. Ischemia and Stroke
Ischemia, particularly in the context of stroke, results from reduced cerebral blood
flow and oxygen supply, causing widespread neuronal damage [
28
,
49
]. The PKA and
PKG pathways play essential roles in the brain’s response to ischemia, promoting neuronal
survival and supporting recovery processes [28,49,50].
During ischemic events, activation of the PKA pathway facilitates neural repair by
enhancing synaptic plasticity, crucial for the restoration of damaged networks [
51
,
52
].
PKA also reduces neuronal apoptosis and neuroinflammation, helping to maintain the
Cells 2025,14, 86 7 of 14
integrity of the blood-brain barrier and preventing further damage from inflammatory
infiltration [51,52]
. PDE4 inhibitors, by elevating cAMP levels and activating PKA, offer
neuroprotective effects by promoting these recovery processes and improving outcomes in
stroke [
2
]. Additionally, PKA activation triggers the Nrf-2/HO-1 pathway, reducing oxida-
tive stress and improving cell viability, which highlights its therapeutic potential in both
ischemia and neurodegenerative diseases [
53
]. Increased AMP, SIRT1, and phosphorylated
AMPK have also been linked to protection against brain edema and blood-brain barrier
dysfunction, further supporting PKA’s protective role [2].
Similarly, the PKG pathway, activated by cGMP, mitigates oxidative stress and mi-
tochondrial dysfunction—two key contributors to neuronal death in stroke [
27
,
50
]. PKG
enhances the expression of antioxidant enzymes and supports mitochondrial health, en-
suring neurons maintain energy production and resist apoptosis [
27
,
50
]. Furthermore,
PKG modulates cerebral blood flow and reduces inflammation, preserving the viability of
ischemic brain tissue [
27
,
50
]. Although PDE4 inhibitors generally have fewer side effects,
such as emesis, higher doses needed for antidepressant effects may increase the risk of ad-
verse reactions, necessitating further research into optimal dosing strategies [
27
,
50
]. PDE5
inhibitors, which prevent cGMP degradation, boost PKG activity and offer neuroprotection
by enhancing mitochondrial function and reducing reactive oxygen species (ROS) [
27
].
By stimulating both the PKG and PI3K/Akt pathways, PDE5 inhibitors present a syner-
gistic therapeutic strategy to protect neurons, minimize cell death, and improve recovery
outcomes in stroke [28].
3.3. Traumatic Brain Injury (TBI)
Following traumatic brain injury (TBI), elevated expression of PDE4B2 and PDE4D2
in the hippocampus leads to reduced cAMP levels, impacting neuronal and immune cell
function [
18
,
36
]. Inhibiting PDE4 has been shown to restore long-term potentiation (LTP) in
the hippocampus and improve basal synaptic transmission by modulating AMPA receptor
transport [
18
,
36
]. PDE4D, which is predominantly expressed in microglia and immune cells,
suggests that PDE4 inhibition also helps regulate the inflammatory response following TBI.
Collectively, PDE4 inhibitors hold potential to enhance cognitive and synaptic function
post-TBI by promoting synaptic plasticity and reducing inflammation [18,36].
PDE5 inhibitors offer additional neuroprotection in TBI by targeting the NO-cGMP
pathway, mitigating vasospasm, and preventing ischemia—two key contributors to poor
outcomes in events such as subarachnoid hemorrhage (SAH) [
37
,
38
]. Research shows that
PDE5 inhibition reduces neuronal death following SAH without increasing intracranial
pressure, confirming its safety and efficacy [
37
,
38
]. These inhibitors also improve cerebral
blood flow regulation in injured brain tissue without elevating intracranial pressure, even
under conditions of reduced mean arterial pressure, which is essential for preserving
brain function and preventing further damage [
37
,
38
]. Furthermore, PDE5 inhibition
reduces levels of endothelin-1, a potent vasoconstrictor implicated in early brain injury,
thus enhancing cerebral perfusion during the acute phase of TBI [
37
,
38
]. By improving
blood flow to damaged areas, PDE5 inhibitors limit ischemic injury, reduce neuronal
death, and support recovery. As such, PDE5 inhibition presents a promising therapeutic
strategy for TBI management by maintaining vascular function and protecting neurons
from ischemic damage and cell death [37,38].
3.4. Alzheimer’s Disease
PDE4 inhibitors hold promise for treating Alzheimer’s disease (AD) with comorbid de-
pression by targeting the PDE4B and PDE4D subtypes, which regulate mood and memory,
respectively [
9
]. By inhibiting PDE4, these compounds elevate cAMP levels, activating the
Cells 2025,14, 86 8 of 14
cAMP/CREB/BDNF signaling pathway, which supports neuroprotection, anti-apoptotic
effects, and cognitive enhancement [
9
,
39
]. Studies in APP/PS1 transgenic mice—a model
for AD—have demonstrated that PDE4 inhibitors improve cognitive function, reduce de-
pressive behaviors, and decrease neuronal apoptosis by increasing the Bcl-2/Bax ratio [
9
,
39
].
Additionally, PDE4 inhibition restores cAMP and pCREB levels, which are associated with
cognitive improvements [9,39].
PDE5 inhibitors also exhibit potential in AD treatment through multiple mechanisms.
They reduce amyloid-beta (A
β
) and phosphorylated tau levels, enhancing cognitive func-
tion in animal models of AD [
3
,
40
]. PDE5 inhibition activates the cGMP/PKG/CREB
pathway, increasing neurotrophic factors such as BDNF and NGF, preserving mitochondrial
function, and preventing apoptosis [
3
,
40
]. These inhibitors also counteract A
β
-induced
dysregulation by modulating glucocorticoid receptor activity and restoring Wnt/
β
-catenin
signaling, promoting neuroprotection [
3
,
40
]. Furthermore, PDE5 inhibitors facilitate A
β
clearance by activating the autophagy-lysosome and ubiquitin-proteasome systems, reduc-
ing toxic protein accumulation [
3
]. Their impact on reducing intracellular calcium levels in
pericytes may further enhance cerebral blood flow and improve vascular function [3,40].
3.5. Summary of Effects of PDE4 and PDE5 Inhibitors in Neurological Disorders
The regulation of PDE4 and PDE5 highlights the critical role of second messenger sys-
tems in brain health, making them promising targets for neurological disorders. Modulating
these pathways offers therapeutic potential across a range of conditions, including traumatic
brain injury, ischemic stroke, epilepsy, and neurodegenerative diseases
(Tables 1and 2).
Further research on PDE inhibitors may pave the way for innovative strategies to treat these
debilitating conditions by enhancing cognitive function, preventing neurodegeneration,
and promoting recovery [3,9,39,40].
Table 1. Observed effects of PDE4 inhibitors in neurological disorders.
PDE4 Inhibitor Neurological
Disorder Animal Model Observed Effects
References
Amlexanox Seizures and
Epilepsy
Sprague Dawley rats
(6–8 weeks old)
Neuronal cell death
Lysosomal function
Autophagy
Neuroinflammation
Reactive oxygen species (ROS)
[26]
Rolipram
FCPR03
Ischemia and
Stroke
Adult male C57BL/6 mice
(6–8 weeks old)
Sprague Dawley rats
(6–8 weeks old)
Neuronal cell death
Neuroinflammation
Blood-brain barrier integrity
ROS production
SIRT1 and p-AMPK
AKT/GSK3β/β-catenin
[2,54]
A33
Rolipram
Traumatic Brain
Injury (TBI)
Sprague Dawley rats
(6–8 weeks old)
Neuronal cell death
Cognitive function
Synaptic function
Synaptic plasticity
Neuroinflammation
[18,36]
Roflumilast Alzheimer’s
Disease (AD)
APP/PS1 double
transgenic mice
Neuronal cell death
cAMP/CREB/BDNF signaling
Bcl-2/Bax ratio
Cognitive function
Depressive-like behavior
[9]
Cells 2025,14, 86 9 of 14
Table 2. Observed effects of PDE5 inhibitors in neurological disorders.
PDE5
Inhibitor
Neurological
Disorder Animal Model Observed Effects
References
Sildenafil Seizures and
Epilepsy
Male Swiss mice
Wistar rats
Neuroprotection via NO-cGMP-PKG
pathway
ROS production
Neuronal excitability
[34,35]
Sildenafil Ischemia and
Stroke
10-day-old (P10) male
Long–Evans rat
Neuronal death
Neuroprotection via PKG pathway
ROS production
PI3K/Akt/mTOR pathway activation
Mitochondrial function
[28]
Sildenafil Traumatic Brain
Injury (TBI)
Twenty-eight
Wistar-derived albino
strain female rats
Neuronal death
Cerebral blood flow without increasing
intracranial pressure
Endothelin-1 levels
Cerebral perfusion
[37]
Mirodenafil Alzheimer’s
Disease (AD) APP-C105 AD mouse
Amyloid-beta (Aβ) and tau levels
Cognitive function
Aβclearance via autophagy-lysosome
pathway
cGMP/PKG/CREB signaling pathway
Mitochondrial function
[3]
This table highlights the therapeutic potential of various PDE4 inhibitors across a
range of neurological disorders, as demonstrated in preclinical animal models. Amlex-
anox, in a seizure and epilepsy model using Sprague Dawley rats, showed neuroprotective
effects, including reduced neuronal cell death, decreased neuroinflammation, and oxida-
tive stress, as well as enhanced lysosomal function and autophagy [
26
]. Rolipram and
FCPR03, in ischemia and stroke models using adult male C57BL/6 mice and Sprague
Dawley rats, reduced neuronal cell death, neuroinflammation, and reactive oxygen species
(ROS) production while preserving blood-brain barrier integrity and promoting SIRT1,
p-AMPK, and AKT/GSK3
β
/
β
-catenin signaling pathways [
2
,
54
]. A33 and Rolipram, in
traumatic brain injury (TBI) models with Sprague Dawley rats, demonstrated significant
neuroprotective effects, including reduced neuronal death, enhanced cognitive and synap-
tic function, and increased synaptic plasticity while mitigating neuroinflammation [
18
,
36
].
Roflumilast, in an Alzheimer’s disease (AD) model using APP/PS1 double transgenic mice,
showed reduced neuronal death, increased cAMP/CREB/BDNF signaling, and improved
Bcl-2/Bax ratios [
9
]. These effects contributed to enhanced cognitive function and alle-
viation of depressive-like behavior. These results underscore the broad neuroprotective
and therapeutic potential of PDE4 inhibitors in addressing key pathological processes in
neurological disorders.
This figure illustrates the therapeutic potential of PDE5 inhibitors across various
neurological disorders, as evidenced in preclinical animal studies. Sildenafil, in seizure
and epilepsy models using Male Swiss mice and Wistar rats, exhibited neuroprotective
effects via the NO-cGMP-PKG pathway, reducing ROS production and neuronal excitabil-
ity [
34
,
35
]. In ischemia and stroke models using 10-day-old male Long–Evans rats, Sildenafil
significantly reduced neuronal death, enhanced neuroprotection through the PKG pathway,
decreased ROS production, and activated the PI3K/Akt/mTOR pathway while improving
mitochondrial function [
28
]. In traumatic brain injury (TBI) models with twenty-eight
Wistar-derived albino strain female rats, Sildenafil reduced neuronal death, improved
Cells 2025,14, 86 10 of 14
cerebral blood flow without increasing intracranial pressure, decreased endothelin-1 lev-
els, and enhanced cerebral perfusion [
37
]. Mirodenafil, in an Alzheimer ’s disease (AD)
model using APP-C105 AD mice, decreased amyloid-beta (A
β
) and tau levels, improved
cognitive function, facilitated A
β
clearance through the autophagy-lysosome pathway,
enhanced cGMP/PKG/CREB signaling, and supported mitochondrial function [
3
]. These
findings underscore the diverse and significant neuroprotective benefits of PDE5 inhibitors,
highlighting their potential as therapeutic agents in neurological disorders.
4. Discussion
Phosphodiesterase (PDE) enzymes play an essential role in hydrolyzing phospho-
diester bonds, contributing to brain development and the pathogenesis of neurological
diseases [
1
]. Among the 11 PDE subtypes, PDE4 and PDE5 are particularly significant for
brain function due to their roles in regulating cAMP and cGMP signaling pathways [
1
,
55
]
(Table 3).
Table 3. Focused roadmap for advancing PDE inhibitor research in neurological disorders.
Focus Area Key Objectives Specific Actions Expected Outcomes
Mechanisms of
Action
1.1 Disease-Specific Pathways: Study PDE4
and PDE5 roles in Alzheimer’s, TBI, stroke,
and epilepsy.
1.2 Crosstalk Analysis: Investigate
interactions between PDE4 and PDE5 in
overlapping conditions.
- Use single-cell RNA sequencing to
identify PDE expression patterns.
- Perform proteomic studies in
disease-specific animal models.
- Analyze effects of selective PDE
inhibition in vitro and in vivo.
- Clear understanding of
PDE pathways for
specific diseases.
- Identification of potential
targets for dual PDE
modulation.
Enhancing
PDE Inhibitors
2.1 Improve Selectivity: Design
isoform-specific inhibitors (e.g.,
PDE4D, PDE5A).
2.2 Improve Bioavailability: Ensure BBB
penetration and target engagement.
2.3 Safety Testing: Examine chronic dosing
impacts (e.g., emesis, cardiovascular
side effects).
- Conduct structure-activity
relationship (SAR) studies to refine
drug candidates.
- Test nanoparticles for
enhanced delivery.
-
Perform 6–12 month chronic toxicity
studies in rodents.
- Highly selective inhibitors
with reduced
off-target effects.
- Drugs optimized for safety
and efficacy in neurological
conditions.
Preclinical to
Clinical
3.1 Validate in Human Models: Use
iPSC-derived neurons and 3D brain
organoids to test PDE inhibitors.
3.2 Initiate Clinical Trials: Prioritize stroke
and epilepsy for Phase 1 studies.
- Generate organoids to mimic
Alzheimer’s and ischemia
environments.
- Develop protocols for multicenter
trials focusing on safety and efficacy
in humans.
- Accelerated clinical
translation of
PDE inhibitors.
- Identification of optimal
dosing regimens and
therapeutic windows.
Combination
Therapies
4.1 Dual PDE Inhibition: Explore combined
PDE4 and PDE5 targeting.
4.2 Synergistic Approaches: Pair PDE
inhibitors with anti-inflammatory or
antioxidant agents.
- Conduct combinatory drug testing
in animal models of TBI
and epilepsy.
- Perform synergy analysis using
isobologram techniques.
- Enhanced efficacy through
synergistic mechanisms.
- Broader therapeutic
applications for complex
neurological disorders.
Phosphodiesterase (PDE) enzymes play a crucial role in hydrolyzing phosphodiester
bonds, influencing brain development and the pathogenesis of neurological diseases [
1
].
Among the 11 PDE subtypes, PDE4 and PDE5 are particularly important for brain function
due to their regulation of cAMP and cGMP signaling pathways [
1
]. PDE4 specifically
hydrolyzes cAMP, a second messenger critical for cognitive functions [
56
]. Increased PDE4
activity lowers cAMP levels, impairing PKA activity and disrupting synaptic plasticity,
which is essential for memory and learning [
26
,
56
]. Proper regulation of PDE4 is necessary
to prevent cognitive disorders such as Alzheimer’s disease and epilepsy [
9
]. Similarly,
PDE5 controls cGMP degradation, and its dysregulation affects vascular function and
neuronal survival, contributing to neurological diseases [1,50].
PDE4 inhibitors offer therapeutic benefits by stabilizing cAMP levels, enhancing PKA
activity, and improving synaptic plasticity [
9
,
26
]. They also reduce neuroinflammation and
Cells 2025,14, 86 11 of 14
oxidative stress, which may mitigate seizure severity and slow progression of epilepsy.
PDE5 inhibitors enhance the cGMP-PKG pathway, modulate nitric oxide (NO) signal-
ing, and reduce oxidative stress, showing potential in decreasing seizure frequency and
severity [34,35].
Both PDE4 and PDE5 inhibitors contribute to neuroprotection during ischemic strokes.
PDE4 inhibitors increase cAMP levels, activating PKA to reduce neuronal apoptosis, neu-
roinflammation, and oxidative stress, while maintaining blood-brain barrier integrity [
2
,
54
].
Activation of the Nrf-2/HO-1 pathway further reduces reactive oxygen species (ROS),
aiding recovery [
2
,
54
]. PDE5 inhibitors, through PKG activation, protect against mito-
chondrial dysfunction and promote neuronal survival by activating the PI3K/Akt/mTOR
pathway [28].
In traumatic brain injury (TBI), PDE4 inhibition improves cognitive outcomes by rais-
ing cAMP levels and reducing neuroinflammation through decreased microglia activation
and inflammatory cytokine production [
18
,
36
]. PDE5 inhibitors enhance cerebral blood
flow, reduce neuronal death, and improve perfusion in damaged brain tissue, suggesting
that PDE inhibitors could significantly enhance clinical outcomes [18,36].
In Alzheimer’s disease (AD) models, PDE4 inhibitors improve cognitive function and
alleviate depression-like behavior by activating the cAMP/CREB/BDNF pathway, promot-
ing neuroprotection and reducing apoptosis [
3
,
9
]. PDE5 inhibitors reduce amyloid-beta
(A
β
) and phosphorylated tau levels, prevent neuronal death, and support mitochondrial in-
tegrity via the cGMP/PKG/CREB signaling pathway [
3
,
9
]. They also promote A
β
clearance
and improve vascular function, making PDE5 a promising target for AD therapy [3,9].
5. Conclusions
PDE4 and PDE5 inhibitors show great promise as therapeutic agents for neurological
disorders like epilepsy, ischemia, TBI, and Alzheimer’s disease. By modulating cAMP
and cGMP signaling pathways, they enhance neuroprotection, reduce inflammation, and
improve cognitive and vascular functions. While preclinical and clinical studies are encour-
aging, further research is needed to confirm their long-term efficacy and safety, potentially
advancing treatment options for these conditions.
Author Contributions: M.K.P. performed the data analysis and reviewed and edited the manuscript.
H.W.Y., S.Y.W., D.Y.K., D.-S.S. and B.Y.C. reviewed and edited the manuscript. S.W.S. contributed
to the discussion and wrote, reviewed, and edited the manuscript. S.W.S. takes full responsibility
for the manuscript and its originality. All authors have read and agreed to the published version of
the manuscript.
Funding: Hallym University Research Fund (grant number: MHC-202402-003), National Research
Foundation of Korea (NRF) (grant number: NRF-2021R1C1C2012889), Ministry of Education and
National Research Foundation of Korea (grant number: GLOCAL-202406240001).
Data Availability Statement: Not applicable.
Acknowledgments: This research was supported by a grant received from the Hallym University
Research Fund (grant number: MHC-202402-003). This research was supported by funding from
the National Research Foundation of Korea (NRF) (NRF-2021R1C1C2012889) to Bo Young Choi.
“Created with BioRender.com”. Following are results of a study on the "Glocal University” Project
(GLOCAL-202406240001), supported by the Ministry of Education and National Research Foundation
of Korea to Dae-Soon Son.
Conflicts of Interest: The authors declare no conflict of interest in this study.
Cells 2025,14, 86 12 of 14
References
1.
Delhaye, S.; Bardoni, B. Role of phosphodiesterases in the pathophysiology of neurodevelopmental disorders. Mol. Psychiatry
2021,26, 4570–4582. [CrossRef] [PubMed]
2.
Dong, X.L.; Wang, Y.H.; Xu, J.; Zhang, N. The protective effect of the PDE-4 inhibitor rolipram on intracerebral haemorrhage is
associated with the cAMP/AMPK/SIRT1 pathway. Sci. Rep. 2021,11, 19737. [CrossRef] [PubMed]
3.
Kang, B.W.; Kim, F.; Cho, J.Y.; Kim, S.; Rhee, J.; Choung, J.J. Phosphodiesterase 5 inhibitor mirodenafil ameliorates Alzheimer-like
pathology and symptoms by multimodal actions. Alzheimers Res. Ther. 2022,14, 92. [CrossRef] [PubMed]
4.
Li, Y.F.; Cheng, Y.F.; Huang, Y.; Conti, M.; Wilson, S.P.; O’Donnell, J.M.; Zhang, H.T. Phosphodiesterase-4D knock-out and RNA
interference-mediated knock-down enhance memory and increase hippocampal neurogenesis via increased cAMP signaling. J.
Neurosci. 2011,31, 172–183. [CrossRef]
5.
Li, H.; Li, J.; Zhang, X.; Feng, C.; Fan, C.; Yang, X.; Zhang, R.; Zhu, F.; Zhou, Y.; Xu, Y.; et al. DC591017, a phosphodiesterase-4
(PDE4) inhibitor with robust anti-inflammation through regulating PKA-CREB signaling. Biochem. Pharmacol. 2020,177, 113958.
[CrossRef] [PubMed]
6.
Wang, Z.Z.; Yang, W.X.; Zhang, Y.; Zhao, N.; Zhang, Y.Z.; Liu, Y.Q.; Xu, Y.; Wilson, S.P.; O’Donnell, J.M.; Zhang, H.T.; et al.
Phosphodiesterase-4D Knock-down in the Prefrontal Cortex Alleviates Chronic Unpredictable Stress-Induced Depressive-Like
Behaviors and Memory Deficits in Mice. Sci. Rep. 2015,5, 11332. [CrossRef] [PubMed]
7. Wiescholleck, V.; Manahan-Vaughan, D. PDE4 inhibition enhances hippocampal synaptic plasticity in vivo and rescues MK801-
induced impairment of long-term potentiation and object recognition memory in an animal model of psychosis. Transl. Psychiatry
2012,2, e89. [CrossRef] [PubMed]
8.
Pierre, S.; Eschenhagen, T.; Geisslinger, G.; Scholich, K. Capturing adenylyl cyclases as potential drug targets. Nat. Rev. Drug
Discov. 2009,8, 321–335. [CrossRef]
9.
Wang, H.; Zhang, F.F.; Xu, Y.; Fu, H.R.; Wang, X.D.; Wang, L.; Chen, W.; Xu, X.Y.; Gao, Y.F.; Zhang, J.G.; et al. The
Phosphodiesterase-4 Inhibitor Roflumilast, a Potential Treatment for the Comorbidity of Memory Loss and Depression in
Alzheimer’s Disease: A Preclinical Study in APP/PS1 Transgenic Mice. Int. J. Neuropsychopharmacol. 2020,23, 700–711. [CrossRef]
[PubMed]
10.
Bollen, E.; Puzzo, D.; Rutten, K.; Privitera, L.; De Vry, J.; Vanmierlo, T.; Kenis, G.; Palmeri, A.; D’Hooge, R.; Balschun, D.; et al.
Improved long-term memory via enhancing cGMP-PKG signaling requires cAMP-PKA signaling. Neuropsychopharmacology 2014,
39, 2497–2505. [CrossRef] [PubMed]
11.
Zhao, S.; Zhang, L.; Lian, G.; Wang, X.; Zhang, H.; Yao, X.; Yang, J.; Wu, C. Sildenafil attenuates LPS-induced pro-inflammatory
responses through down-regulation of intracellular ROS-related MAPK/NF-kappaB signaling pathways in N9 microglia. Int.
Immunopharmacol. 2011,11, 468–474. [CrossRef]
12.
Puerta, E.; Hervias, I.; Barros-Minones, L.; Jordan, J.; Ricobaraza, A.; Cuadrado-Tejedor, M.; Garcia-Osta, A.; Aguirre, N. Sildenafil
protects against 3-nitropropionic acid neurotoxicity through the modulation of calpain, CREB, and BDNF. Neurobiol. Dis. 2010,38,
237–245. [CrossRef]
13.
Shelly, M.; Lim, B.K.; Cancedda, L.; Heilshorn, S.C.; Gao, H.; Poo, M.M. Local and long-range reciprocal regulation of cAMP and
cGMP in axon/dendrite formation. Science 2010,327, 547–552. [CrossRef] [PubMed]
14.
Argyrousi, E.K.; Heckman, P.R.A.; Prickaerts, J. Role of cyclic nucleotides and their downstream signaling cascades in memory
function: Being at the right time at the right spot. Neurosci. Biobehav. Rev. 2020,113, 12–38. [CrossRef] [PubMed]
15.
Zhang, H.T.; Zhao, Y.; Huang, Y.; Dorairaj, N.R.; Chandler, L.J.; O’Donnell, J.M. Inhibition of the phosphodiesterase 4 (PDE4)
enzyme reverses memory deficits produced by infusion of the MEK inhibitor U0126 into the CA1 subregion of the rat hippocampus.
Neuropsychopharmacology 2004,29, 1432–1439. [CrossRef] [PubMed]
16.
Houslay, M.D.; Adams, D.R. PDE4 cAMP phosphodiesterases: Modular enzymes that orchestrate signalling cross-talk, desensiti-
zation and compartmentalization. Biochem. J. 2003,370, 1–18. [CrossRef] [PubMed]
17.
Richter, W.; Menniti, F.S.; Zhang, H.T.; Conti, M. PDE4 as a target for cognition enhancement. Expert Opin. Ther. Targets 2013,17,
1011–1027. [CrossRef]
18.
Wilson, N.M.; Titus, D.J.; Oliva, A.A., Jr.; Furones, C.; Atkins, C.M. Traumatic Brain Injury Upregulates Phosphodiesterase
Expression in the Hippocampus. Front. Syst. Neurosci. 2016,10, 5. [CrossRef] [PubMed]
19.
Schepers, M.; Paes, D.; Tiane, A.; Rombaut, B.; Piccart, E.; van Veggel, L.; Gervois, P.; Wolfs, E.; Lambrichts, I.; Brullo, C.; et al.
Selective PDE4 subtype inhibition provides new opportunities to intervene in neuroinflammatory versus myelin damaging
hallmarks of multiple sclerosis. Brain Behav. Immun. 2023,109, 1–22. [CrossRef]
20.
Lugnier, C. The Complexity and Multiplicity of the Specific cAMP Phosphodiesterase Family: PDE4, Open New Adapted
Therapeutic Approaches. Int. J. Mol. Sci. 2022,23, 10616. [CrossRef]
21.
Hollas, M.A.; Ben Aissa, M.; Lee, S.H.; Gordon-Blake, J.M.; Thatcher, G.R.J. Pharmacological manipulation of cGMP and
NO/cGMP in CNS drug discovery. Nitric Oxide 2019,82, 59–74. [CrossRef] [PubMed]
Cells 2025,14, 86 13 of 14
22.
Garthwaite, J. NO as a multimodal transmitter in the brain: Discovery and current status. Br. J. Pharmacol. 2019,176, 197–211.
[CrossRef] [PubMed]
23.
Shimizu-Albergine, M.; Rybalkin, S.D.; Rybalkina, I.G.; Feil, R.; Wolfsgruber, W.; Hofmann, F.; Beavo, J.A. Individual cerebellar
Purkinje cells express different cGMP phosphodiesterases (PDEs):
In vivo
phosphorylation of cGMP-specific PDE (PDE5) as an
indicator of cGMP-dependent protein kinase (PKG) activation. J. Neurosci. 2003,23, 6452–6459. [CrossRef] [PubMed]
24.
Ma, Y.; Zhang, F.; Zhong, Y.; Huang, Y.; Yixizhuoma; Jia, Q.; Zhang, S. A label-free LC/MS-based enzymatic activity assay for the
detection of PDE5A inhibitors. Front. Chem. 2023,11, 1097027. [CrossRef] [PubMed]
25.
Machado Batista Sohn, J.; Cardoso, N.C.; Raymundi, A.M.; Prickaerts, J.; Stern, C.A.J. Phosphodiesterase 4 inhibition after
retrieval switches the memory fate favoring extinction instead of reconsolidation. Sci. Rep. 2023,13, 20384. [CrossRef] [PubMed]
26.
Yang, H.W.; Kho, A.R.; Lee, S.H.; Kang, B.S.; Park, M.K.; Lee, C.J.; Park, S.W.; Woo, S.Y.; Kim, D.Y.; Jung, H.H.; et al. A
phosphodiesterase 4 (PDE4) inhibitor, amlexanox, reduces neuroinflammation and neuronal death after pilocarpine-induced
seizure. Neurotherapeutics 2024,21, e00357. [CrossRef] [PubMed]
27.
Gao, F.; Sugita, M.; Nukui, H. Phosphodiesterase 5 inhibitor, zaprinast, selectively increases cerebral blood flow in the ischemic
penumbra in the rat brain. Neurol. Res. 2005,27, 638–643. [CrossRef] [PubMed]
28. Yazdani, A.; Howidi, B.; Shi, M.Z.; Tugarinov, N.; Khoja, Z.; Wintermark, P. Sildenafil improves hippocampal brain injuries and
restores neuronal development after neonatal hypoxia-ischemia in male rat pups. Sci. Rep. 2021,11, 22046. [CrossRef]
29. Baillie, G.S.; Tejeda, G.S.; Kelly, M.P. Therapeutic targeting of 3’,5’-cyclic nucleotide phosphodiesterases: Inhibition and beyond.
Nat. Rev. Drug Discov. 2019,18, 770–796. [CrossRef]
30.
Li, Z.H.; Cui, D.; Qiu, C.J.; Song, X.J. Cyclic nucleotide signaling in sensory neuron hyperexcitability and chronic pain after nerve
injury. Neurobiol. Pain 2019,6, 100028. [CrossRef] [PubMed]
31.
Li, X.; Fetter, R.; Schwabe, T.; Jung, C.; Liu, L.; Steller, H.; Gaul, U. The cAMP effector PKA mediates Moody GPCR signaling in
Drosophila blood-brain barrier formation and maturation. eLife 2021,10, e68275. [CrossRef]
32.
Liu, J.; Liu, B.; Yuan, P.; Cheng, L.; Sun, H.; Gui, J.; Pan, Y.; Huang, D.; Chen, H.; Jiang, L. Role of PKA/CREB/BDNF signaling
in PM2.5-induced neurodevelopmental damage to the hippocampal neurons of rats. Ecotoxicol. Environ. Saf. 2021,214, 112005.
[CrossRef] [PubMed]
33.
Wang, Q.; Mergia, E.; Koesling, D.; Mittmann, T. Nitric Oxide/Cyclic Guanosine Monophosphate Signaling via Guanylyl
Cyclase Isoform 1 Mediates Early Changes in Synaptic Transmission and Brain Edema Formation after Traumatic Brain Injury. J.
Neurotrauma 2021,38, 1689–1701. [CrossRef]
34.
Silva, A.; Nascimento, C.P.; Azevedo, J.E.C.; Vieira, L.R.; Hamoy, A.O.; Tiago, A.; Martins Rodrigues, J.C.; de Araujo, D.B.;
Favacho Lopes, D.C.; de Mello, V.J.; et al. Unmasking hidden risks: The surprising link between PDE5 inhibitors and seizure
susceptibility. PLoS ONE 2023,18, e0294754. [CrossRef] [PubMed]
35.
de Carvalho, M.A.J.; Chaves-Filho, A.; de Souza, A.G.; de Carvalho Lima, C.N.; de Lima, K.A.; Rios Vasconcelos, E.R.; Feitosa,
M.L.; Souza Oliveira, J.V.; de Souza, D.A.A.; Macedo, D.S.; et al. Proconvulsant effects of sildenafil citrate on pilocarpine-induced
seizures: Involvement of cholinergic, nitrergic and pro-oxidant mechanisms. Brain Res. Bull. 2019,149, 60–74. [CrossRef]
36.
Wilson, N.M.; Gurney, M.E.; Dietrich, W.D.; Atkins, C.M. Therapeutic benefits of phosphodiesterase 4B inhibition after traumatic
brain injury. PLoS ONE 2017,12, e0178013. [CrossRef]
37.
Kilicarslan, B.; Kilicarslan, E.; Kizmazoglu, C.; Aydin, H.E.; Kaya, I.; Danyeli, A.E.; Karabekir, H.S. Evaluation of the Efficacy of
Sildenafil Citrate Following Severe Head Trauma in an Experimental Rat Model. Turk. Neurosurg. 2020,30, 501–506. [CrossRef]
38.
Kalyani, P.; Lippa, S.M.; Werner, J.K.; Amyot, F.; Moore, C.B.; Kenney, K.; Diaz-Arrastia, R. Phosphodiesterase-5 (PDE-5) Inhibitors
as Therapy for Cerebrovascular Dysfunction in Chronic Traumatic Brain Injury. Neurotherapeutics 2023,20, 1629–1640. [CrossRef]
39.
Shi, Y.; Lv, J.; Chen, L.; Luo, G.; Tao, M.; Pan, J.; Hu, X.; Sheng, J.; Zhang, S.; Zhou, M.; et al. Phosphodiesterase-4D Knockdown in
the Prefrontal Cortex Alleviates Memory Deficits and Synaptic Failure in Mouse Model of Alzheimer’s Disease. Front. Aging
Neurosci. 2021,13, 722580. [CrossRef] [PubMed]
40.
Mao, F.; Wang, H.; Ni, W.; Zheng, X.; Wang, M.; Bao, K.; Ling, D.; Li, X.; Xu, Y.; Zhang, H.; et al. Design, Synthesis, and
Biological Evaluation of Orally Available First-Generation Dual-Target Selective Inhibitors of Acetylcholinesterase (AChE) and
Phosphodiesterase 5 (PDE5) for the Treatment of Alzheimer’s Disease. ACS Chem. Neurosci. 2018,9, 328–345. [CrossRef] [PubMed]
41.
Pedersen, M.; Omidvarnia, A.; Curwood, E.K.; Walz, J.M.; Rayner, G.; Jackson, G.D. The dynamics of functional connectivity in
neocortical focal epilepsy. Neuroimage Clin. 2017,15, 209–214. [CrossRef]
42. Thijs, R.D.; Surges, R.; O’Brien, T.J.; Sander, J.W. Epilepsy in adults. Lancet 2019,393, 689–701. [CrossRef]
43.
Abel, T.; Nguyen, P.V. Regulation of hippocampus-dependent memory by cyclic AMP-dependent protein kinase. Prog. Brain Res.
2008,169, 97–115. [CrossRef]
44.
Zhang, J.; Zhang, C.; Chen, X.; Wang, B.; Ma, W.; Yang, Y.; Zheng, R.; Huang, Z. PKA-RIIbeta autophosphorylation modulates
PKA activity and seizure phenotypes in mice. Commun. Biol. 2021,4, 263. [CrossRef]
45. Esplugues, J.V. NO as a signalling molecule in the nervous system. Br. J. Pharmacol. 2002,135, 1079–1095. [CrossRef]
Cells 2025,14, 86 14 of 14
46.
ElHady, A.K.; El-Gamil, D.S.; Abdel-Halim, M.; Abadi, A.H. Advancements in Phosphodiesterase 5 Inhibitors: Unveiling Present
and Future Perspectives. Pharmaceuticals 2023,16, 1266. [CrossRef] [PubMed]
47.
Syed, Y.A.; Baer, A.; Hofer, M.P.; Gonzalez, G.A.; Rundle, J.; Myrta, S.; Huang, J.K.; Zhao, C.; Rossner, M.J.; Trotter, M.W.; et al.
Inhibition of phosphodiesterase-4 promotes oligodendrocyte precursor cell differentiation and enhances CNS remyelination.
EMBO Mol. Med. 2013,5, 1918–1934. [CrossRef] [PubMed]
48.
Donders, Z.; Skorupska, I.J.; Willems, E.; Mussen, F.; Broeckhoven, J.V.; Carlier, A.; Schepers, M.; Vanmierlo, T. Beyond PDE4
inhibition: A comprehensive review on downstream cAMP signaling in the central nervous system. Biomed. Pharmacother. 2024,
177, 117009. [CrossRef] [PubMed]
49.
Olmestig, J.N.E.; Marlet, I.R.; Hainsworth, A.H.; Kruuse, C. Phosphodiesterase 5 inhibition as a therapeutic target for ischemic
stroke: A systematic review of preclinical studies. Cell. Signal. 2017,38, 39–48. [CrossRef]
50.
Xu, F.; Lv, C.; Deng, Y.; Liu, Y.; Gong, Q.; Shi, J.; Gao, J. Icariside II, a PDE5 Inhibitor, Suppresses Oxygen-Glucose
Deprivation/Reperfusion-Induced Primary Hippocampal Neuronal Death Through Activating the PKG/CREB/BDNF/TrkB
Signaling Pathway. Front. Pharmacol. 2020,11, 523. [CrossRef]
51.
Yu, S.; Doycheva, D.M.; Gamdzyk, M.; Yang, Y.; Lenahan, C.; Li, G.; Li, D.; Lian, L.; Tang, J.; Lu, J.; et al. Activation of MC1R with
BMS-470539 attenuates neuroinflammation via cAMP/PKA/Nurr1 pathway after neonatal hypoxic-ischemic brain injury in rats.
J. Neuroinflammation 2021,18, 26. [CrossRef] [PubMed]
52.
Zheng, Y.; Li, L.; Chen, B.; Fang, Y.; Lin, W.; Zhang, T.; Feng, X.; Tao, X.; Wu, Y.; Fu, X.; et al. Chlorogenic acid exerts
neuroprotective effect against hypoxia-ischemia brain injury in neonatal rats by activating Sirt1 to regulate the Nrf2-NF-kappaB
signaling pathway. Cell Commun. Signal. 2022,20, 84. [CrossRef]
53.
Xu, B.; Qin, Y.; Li, D.; Cai, N.; Wu, J.; Jiang, L.; Jie, L.; Zhou, Z.; Xu, J.; Wang, H. Inhibition of PDE4 protects neurons against
oxygen-glucose deprivation-induced endoplasmic reticulum stress through activation of the Nrf-2/HO-1 pathway. Redox Biol.
2020,28, 101342. [CrossRef]
54.
Xu, B.; Wang, T.; Xiao, J.; Dong, W.; Wen, H.Z.; Wang, X.; Qin, Y.; Cai, N.; Zhou, Z.; Xu, J.; et al. FCPR03, a novel phosphodiesterase
4 inhibitor, alleviates cerebral ischemia/reperfusion injury through activation of the AKT/GSK3beta/ beta-catenin signaling
pathway. Biochem. Pharmacol. 2019,163, 234–249. [CrossRef]
55.
Baudet, S.; Zagar, Y.; Roche, F.; Gomez-Bravo, C.; Couvet, S.; Becret, J.; Belle, M.; Vougny, J.; Uthayasuthan, S.; Ros, O.; et al.
Subcellular second messenger networks drive distinct repellent-induced axon behaviors. Nat. Commun. 2023,14, 3809. [CrossRef]
[PubMed]
56.
Itoh, T.; Abe, K.; Hong, J.; Inoue, O.; Pike, V.W.; Innis, R.B.; Fujita, M. Effects of cAMP-dependent protein kinase activator and
inhibitor on in vivo rolipram binding to phosphodiesterase 4 in conscious rats. Synapse 2010,64, 172–176. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
Article
Full-text available
Parkinson's disease (PD) is recognized as a neurological disorder with no known cure in modern medicine. While scientific research has advanced diagnostic methods and symptom management strategies, the underlying causes of PD remain elusive. From a Buddhist perspective, particularly through the teachings of Dharma Master Jun Hong Lu, PD is understood as a karmic and spiritual illness rather than merely a neurological disorder. This study explores the connection between karmic debts, spirit disturbances, and the manifestation of PD symptoms. Through case studies, we demonstrate that patients practicing Golden Buddhist Practices-such as making vows, reciting Buddhist scriptures, and performing life liberation-have experienced significant improvements and, in some cases, complete recovery. These findings suggest that PD is not inherently irreversible and that addressing its spiritual root causes can lead to effective healing. Integrating Buddhist wisdom with conventional medical approaches could provide new insights into treating PD and other intractable diseases. Based on our findings, we propose that PD be redefined as a prevalent yet curable karmic and spiritual disease, characterized by pathological changes such as neurodegeneration, including the loss of dopaminergic neurons and abnormal alpha-synuclein aggregation.
Article
Full-text available
Epilepsy, a complex neurological disorder, is characterized by recurrent seizures caused by aberrant electrical activity in the brain. Central to this study is the role of lysosomal dysfunction in epilepsy, which can lead to the accumulation of toxic substrates and impaired autophagy in neurons. Our focus is on phosphodiesterase-4 (PDE4), an enzyme that plays a crucial role in regulating intracellular cyclic adenosine monophosphate (cAMP) levels by converting it into adenosine monophosphate (AMP). In pathological states, including epilepsy, increased PDE4 activity contributes to a decrease in cAMP levels, which may exacerbate neuroinflammatory responses. We hypothesized that amlexanox, an anti-inflammatory drug and non-selective PDE4 inhibitor, could offer neuroprotection by addressing lysosomal dysfunction and mitigating neuroinflammation, ultimately preventing neuronal death in epileptic conditions. Our research utilized a pilocarpine-induced epilepsy animal model to investigate amlexanox's potential benefits. Administered intraperitoneally at a dose of 100 mg/kg daily following the onset of a seizure, we monitored its effects on lysosomal function, inflammation, neuronal death, and cognitive performance in the brain. Tissue samples from various brain regions were collected at predetermined intervals for a comprehensive analysis. The study's results were significant. Amlexanox effectively improved lysosomal function, which we attribute to the modulation of zinc's influx into the lysosomes, subsequently enhancing autophagic processes and decreasing the release of inflammatory factors. Notably, this led to the attenuation of neuronal death in the hippocampal region. Additionally, cognitive function, assessed through the modified neurological severity score (mNSS) and the Barnes maze test, showed substantial improvements after treatment with amlexanox. These promising outcomes indicate that amlexanox has potential as a therapeutic agent in the treatment of epilepsy and related brain disorders. Its ability to combat lysosomal dysfunction and neuroinflammation positions it as a potential neuroprotective intervention. While these findings are encouraging, further research and clinical trials are essential to fully explore and validate the therapeutic efficacy of amlexanox in epilepsy management.
Article
Full-text available
Background Phosphodiesterase 5 inhibitors (PDE5i) are the first line treatment for erectile dysfunction; however, several articles and case reports have shown central nervous system effects, that can cause seizures in susceptible patients. This study aims to describe the changes caused by the use of Sildenafil and Tadalafil through the analysis of abnormalities expressed in the electrocorticogram (ECoG) of rats and evaluate the seizure threshold response and treatment of seizures with anticonvulsants. Materials and methods The study used 108 rats (Wistar). Before surgery for electrode placement in dura mater, the animals were randomly separated into 3 experiments for electrocorticogram analysis. Experiment 1: ECoG response to using PD5i (Sildenafil 20mg/kg and Tadalafil 2.6mg/kg p.o.). Experiment 2: ECoG response to the use of PD5i in association with Pentylenetetrazole (PTZ—30 mg/kg i.p.), a convulsive model. Experiment 3: ECoG response to anticonvulsant treatment (Phenytoin, Phenobarbital and Diazepam) of seizures induced by association IPDE5 + PTZ. All recordings were made thirty minutes after administration of the medication and analyzed for ten minutes, only once. We considered statistical significance level of *p<0.05, **p<0.01 and ***p < 0.001. Results After administration of Sildenafil and Tadalafil, there were increases in the power of recordings in the frequency bands in oscillations in alpha (p = 0.0920) and beta (p = 0.602) when compared to the control group (p<0.001). After the use of Sildenafil and Tadalafil associated with PTZ, greater potency was observed in the recordings during seizures (p<0.001), however, the Sildenafil group showed greater potency when compared to Tadalafil (p<0.05). Phenobarbital and Diazepam showed a better response in controlling discharges triggered by the association between proconvulsant drugs. Conclusions PDE5i altered the ECoG recordings in the rats’ motor cortexes, demonstrating cerebral asynchrony and potentiating the action of PTZ. These findings demonstrate that PDE5i can lower the seizure threshold.
Article
Full-text available
Phosphodiesterase 4 (PDE4), an enzyme expressed in the dorsal hippocampus (DH), hydrolyzes the cAMP, limiting the PKA-induced CREB phosphorylation (pCREB) and BDNF expression. Depending on the brain region, PKA and pCREB mediate reconsolidation or extinction, whereas BDNF is mainly related to extinction facilitation. The mechanisms underpinning the switch between reconsolidation and extinction are relatively unknown. Here, we tested the hypothesis that PDE4 might control these processes. We showed in Wistar rats submitted to contextual fear conditioning that PDE4 inhibition with roflumilast (ROF) within the DH, after a short retrieval, did not change freezing behavior after one day (TestA1). After 10 days, the ROF-treated group significantly reduced the expression of freezing behavior. This effect depended on retrieval, Test A1 exposure, and reinstated after a remainder foot shock, suggesting an extinction facilitation. The ROF effect depended on PKA after retrieval or, protein synthesis after Test A1. After retrieval, ROF treatment did not change the pCREB/CREB ratio in the DH. It enhanced proBDNF expression without changing pre-proBDNF or mature BDNF in the DH after Test A1. The results suggest that the inhibition of PDE4 in the DH after a short retrieval changes the memory sensibility from reconsolidation to extinction via regulating proBDNF expression.
Article
Full-text available
Phosphodiesterase 5 (PDE5) inhibitors presented themselves as important players in the nitric oxide/cGMP pathway, thus exerting a profound impact on various physiological and pathological processes. Beyond their well-known efficacy in treating male erectile dysfunction (ED) and pulmonary arterial hypertension (PAH), a plethora of studies have unveiled their significance in the treatment of a myriad of other diseases, including cognitive functions, heart failure, multiple drug resistance in cancer therapy, immune diseases, systemic sclerosis and others. This comprehensive review aims to provide an updated assessment of the crucial role played by PDE5 inhibitors (PDE5-Is) as disease-modifying agents taking their limiting side effects into consideration. From a medicinal chemistry and drug discovery perspective, the published PDE5-Is over the last 10 years and their binding characteristics are systemically discussed, and advancement in properties is exposed. A persistent challenge encountered with these agents lies in their limited isozyme selectivity; considering this obstacle, this review also highlights the breakthrough development of the recently reported PDE5 allosteric inhibitors, which exhibit an unparalleled level of selectivity that was rarely achievable by competitive inhibitors. The implications and potential impact of these novel allosteric inhibitors are meticulously explored. Additionally, the concept of multi-targeted ligands is critically evaluated in relation to PDE5-Is by inspecting the broader spectrum of their molecular interactions and effects. The objective of this review is to provide insight into the design of potent, selective PDE5-Is and an overview of their biological function, limitations, challenges, therapeutic potentials, undergoing clinical trials, future prospects and emerging uses, thus guiding upcoming endeavors in both academia and industry within this domain.
Article
Full-text available
Second messengers, including cAMP, cGMP and Ca²⁺ are often placed in an integrating position to combine the extracellular cues that orient growing axons in the developing brain. This view suggests that axon repellents share the same set of cellular messenger signals and that axon attractants evoke opposite cAMP, cGMP and Ca²⁺ changes. Investigating the confinement of these second messengers in cellular nanodomains, we instead demonstrate that two repellent cues, ephrin-A5 and Slit1, induce spatially segregated signals. These guidance molecules activate subcellular-specific second messenger crosstalk, each signaling network controlling distinct axonal morphology changes in vitro and pathfinding decisions in vivo.
Article
Full-text available
Phosphodiesterase type 5 (PDE5), a cyclic nucleotide phosphodiesterase, controls the duration of the cyclic guanosine monophosphate (cGMP) signal by hydrolyzing cGMP to GMP. Inhibiting the activity of PDE5A has proven to be an effective strategy for treating pulmonary arterial hypertension and erectile dysfunction. Current enzymatic activity assay methods for PDE5A mainly use fluorescent or isotope-labeled substrates, which are expensive and inconvenient. Here, we developed an LC/MS-based enzymatic activity assay for PDE5A without labeling, which detects the enzymatic activity of PDE5A by quantifying the substrate cGMP and product GMP at a concentration of 100 nM. The accuracy of this method was verified by a fluorescently labeled substrate. Moreover, a new inhibitor of PDE5A was identified by this method and virtual screening. It inhibited PDE5A with an IC50 value of 870 nM. Overall, the proposed strategy provides a new method for screening PDE5A inhibitors.
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
Cyclic nucleotides (cAMP, cGMP) play a major role in normal and pathologic signaling. Beyond receptors, cyclic nucleotide phosphodiesterases; (PDEs) rapidly convert the cyclic nucleotide in its respective 5′-nucleotide to control intracellular cAMP and/or cGMP levels to maintain a normal physiological state. However, in many pathologies, dysregulations of various PDEs (PDE1-PDE11) contribute mainly to organs and tissue failures related to uncontrolled phosphorylation cascade. Among these, PDE4 represents the greatest family, since it is constituted by 4 genes with multiple variants differently distributed at tissue, cellular and subcellular levels, allowing different fine-tuned regulations. Since the 1980s, pharmaceutical companies have developed PDE4 inhibitors (PDE4-I) to overcome cardiovascular diseases. Since, they have encountered many undesired problems, (emesis), they focused their research on other PDEs. Today, increases in the knowledge of complex PDE4 regulations in various tissues and pathologies, and the evolution in drug design, resulted in a renewal of PDE4-I development. The present review describes the recent PDE4-I development targeting cardiovascular diseases, obesity, diabetes, ulcerative colitis, and Crohn’s disease, malignancies, fatty liver disease, osteoporosis, depression, as well as COVID-19. Today, the direct therapeutic approach of PDE4 is extended by developing allosteric inhibitors and protein/protein interactions allowing to act on the PDE interactome.
Article
Multiple phase III randomized controlled trials (RCTs) for pharmacologic interventions in traumatic brain injury (TBI) have failed despite promising results in experimental models. The heterogeneity of TBI, in terms of pathomechanisms and impacted brain structures, likely contributes to these failures. Biomarkers have been recommended to identify patients with relevant pathology (predictive biomarkers) and confirm target engagement and monitor therapy response (pharmacodynamic biomarkers). Our group focuses on traumatic cerebrovascular injury as an understudied endophenotype of TBI and is validating a predictive and pharmacodynamic imaging biomarker (cerebrovascular reactivity; CVR) in moderate-severe TBI. We aim to extend these studies to milder forms of TBI to determine the optimal dose of sildenafil for maximal improvement in CVR. We will conduct a phase II dose-finding study involving 160 chronic TBI patients (mostly mild) using three doses of sildenafil, a phosphodiesterase-5 (PDE-5) inhibitor. The study measures baseline CVR and evaluates the effect of escalating sildenafil doses on CVR improvement. A 4-week trial of thrice daily sildenafil will assess safety, tolerability, and clinical efficacy. This dual-site 4-year study, funded by the Department of Defense and registered in ClinicalTrials.gov (NCT05782244), plans to launch in June 2023. Biomarker-informed RCTs are essential for developing effective TBI interventions, relying on an understanding of underlying pathomechanisms. Traumatic microvascular injury (TMVI) is an attractive mechanism which can be targeted by vaso-active drugs such as PDE-5 inhibitors. CVR is a potential predictive and pharmacodynamic biomarker for targeted interventions aimed at TMVI. (Trial registration: NCT05782244, ClinicalTrials.gov).