Invest Clin 66(2): 191 - 204, 2025 https://doi.org/10.54817/IC.v66n2a06
Corresponding author: Wei Huang, Neurology Department, The Third people’s Hospital of Yunnan Province, 292#
Beijing Road, Guandu District, Kunming City, Yunnan Province 650000, China. Email: huangweidr@yeah.net
The p300-NF-κB pathway induces the
activation of the NLRP3 inflammasome
and the pyroptosis of neurons in an in vitro
model of Alzheimer’s disease.
Fengqin Sun and Wei Huang
Neurology Department, The Third People’s Hospital of Yunnan Province, China.
Keywords: Alzheimer’s disease; p300; pyroptosis; NLRP3 inflammasome.
Abstract. Inflammation-induced neuronal death is the primary cause of
Alzheimer’s disease (AD). p300 plays an important role in brain disorders. How-
ever, the role of p300 in AD remains unclear. This study aimed to investigate the
potential of p300 in an in vitro model of AD. Protein expression was detected
using western blotting. The mRNA levels were determined by reverse transcrip-
tion-quantitative polymerase chain reaction. Cytokine release was detected us-
ing an enzyme-linked immunosorbent assay. Cellular function was determined
using the cell counting kit-8, lactate dehydrogenase, and flow cytometry assays.
Chromatin immunoprecipitation and luciferase assays verified the interaction
between nuclear factor kappa B (NF-κB) and the NLR family pyrin domain con-
taining 3 (NLRP3). E1A binding protein p300 (p300) was overexpressed in the
Aβ1-42 induced AD model in vitro. However, treatment with the p300 inhibitor
(GNE-049) alleviated inflammation and Aβ1-42-induced pyroptosis in the neu-
rons. p300 activates NF-κB, which antagonizes the effects of GNE-049 and pro-
motes neuronal pyroptosis. Moreover, NF-κB epigenetically activates the NLRP3
inflammasome. The p300/NF-κB pathway promotes neuronal pyroptosis in an in
vitro AD model by activating the NLRP3 inflammasome. Therefore, the p300/
NF-κB/NLRP3 signalling pathway may be a potential therapeutic target for AD.
192 Sun and Huang
Investigación Clínica 66(2): 2025
En el modelo in vitro de la enfermedad de Alzheimer, la vía
p300 NF - Kappa B induce la activación del inflamasoma NLRP3
y la piroptosis neuronal en un modelo in vitro de la enfermedad
de Alzheimer.
Invest Clin 2025; 66 (2): 191 – 204
Palabras clave: enfermedad de Alzheimer; P300; muerte por quemadura celular; cuerpo
inflamatorio nlrp3.
Resumen. La muerte neuronal inducida por la inflamación es la principal
causa de la enfermedad de Alzheimer (AD). El p300 juega un papel impor-
tante en las enfermedades cerebrales. Sin embargo, se desconoce el papel del
p300 en la AD. El objetivo de este estudio es explorar el potencial del p300 en
modelos in vitro de AD. Se utilizó Western blot para detectar la expresión de
proteínas. Los niveles de ARNm se determinaron mediante la reacción cuanti-
tativa en cadena de la polimerasa de transcripción inversa. Se utilizó la prueba
de inmunoabsorción enzimática para detectar la liberación de citocinas. La
función celular se determinó mediante el contador celular Kit - 8, la lactato
deshidrogenasa y la medición con citometría de flujo. La interacción entre el
factor nuclear Kappa b (nf - Kappa b) y el dominio Pirin 3 (nlrp3), que contiene
la familia NLR, fue verificada por inmunoprecipitación de cromatina y detec-
ción de luciferasa. La proteína de unión a E1A p300 (p300) está sobreexpresada
en un modelo de AD inducido por Aβ1-42. Sin embargo, el tratamiento con un
inhibidor del p300 (GNE - 049) redujo la inflamación y redujo la muerte por
piroptosis neuronal inducida por Aβ1-42. El p300 activa NF - Kappa b, que inhibe
el efecto del GNE - 049 y promueve la muerte por piroptosis neuronal. Además,
NF-κB epigeneticamentelly activa el NLRP3 inflamasoma Epigenética NF-Kappa
B activa los cuerpos inflamatorios nlrp3. La vía p300 / NF - Kappa B promueve
la muerte focal neuronal en modelos in vitro de AD activando el inflamasoma
NLRP3. Por lo tanto, la transmisión de la señal p300/NF-Kappa B/NLRP3 puede
ser un objetivo terapéutico potencial para la AD.
Received: 08-03-2025 Accepted: 10-05-2025
INTRODUCTION
Alzheimer’s disease (AD) is a common
neurodegenerative disorder 1. The symptoms
include apraxia, agnosia, aphasia, and emo-
tional disturbance 2. AD is the leading cause
of dementia and accounts for > 60% of all
cases 3. Currently, nearly 50,000,000 people
suffer from AD worldwide 4. In China, among
the 15.07 million people (≥60 years old)
with dementia, 9.83 million (65.2%) were di-
agnosed with AD 5. Moreover, the incidence
of AD is increasing with an aging popula-
tion6. However, the pathogenesis of AD is
complex, and there are no effective prognos-
tic biomarkers 7. Therefore, identifying the
potential molecular mechanisms underlying
AD may provide novel therapeutic strategies.
Pyroptosis is a form of programmed
cell death characterized by inflammasomes8.
Effect of the p300/NF-kB pathway in an in vitro model of Alzheimer’s disease 193
Vol. 66(2): 191 - 204, 2025
The NLR family pyrin domain containing 3
(NLRP3) is the main inflammasome expressed
in brain tissues 9. Stimuli-induced activation
of NLRP3 inflammasomes cleaves caspase1 10.
The cleaved caspase-1 then cleaves gasdermin
D (GSDMD) and induces the accumulation of
the N-terminus of GSDMD (GSDMD-N), which
drives GSDMD to move to the cell membranes
11. The enrichment of GSDMD-N in the cell
membranes contributes to pore formation
and subsequent relapse, releasing interleukin
(IL)-1β and IL-18 12,13. The activation of the
NLRP3 inflammasome is frequently observed
in patients with AD. The NLRP3 inflammasome
mediates neuroinflammation, cell senescence,
and loss of neurons, which are key causes of AD
14-16. However, inhibition of the NLRP3 inflam-
masome restores neuronal function and allevi-
ates AD development 17.
Nuclear phosphoprotein E1A binding
protein p300 (p300) is an acetyl transfer-
ase 18. p300 regulates numerous biological
processes such as proliferation, autophagy,
apoptosis, and pyroptosis 19,20. Increasing
evidence has suggested that p300 is abnor-
mally expressed in patients with brain disor-
ders. For instance, p300 is downregulated in
ischemia/reperfusion injury, whereas over-
expressed p300 enhances the anti-apoptotic
effects of myocardin-related transcription
factor A 21. However, p300 deficiency in-
hibits neuroepithelial cell proliferation in
diabetes-induced tube defects 22. Moreover,
high levels of p300 in human brains with AD
contribute to neuronal loss 23. However, the
role of p300 in AD has not yet been com-
pletely elucidated. In the present study, we
investigated the potential role of p300 in an
in vitro model of AD. We hypothesized that
p300-mediated neuronal pyroptosis exacer-
bates the progression of AD.
MATERIALS AND METHODS
Cell culture
The mouse neuronal cell lines HT22 and
HEK293T were obtained from ATCC (Manas-
sas, VA, USA). The cells were cultured in
Dulbecco’s Modified Eagle’s Medium supple-
mented with 10% fetal bovine serum (FBS).
The cells were incubated at 37°C in a 5% CO2.
HT22 cells were exposed to Aβ1-42 (20
μM) and a p300 inhibitor (GNE-049, 500
nM) for 24 h. Cells in the control groups
were cultured with FBS.
HT22 cells were transfected with small
hairpin RNA shRNA) of nuclear factor kap-
pa B (NF-κB) and overexpression plasmids
or the control/vector using Lipofectamine
3000 (Invitrogen) according to the manu-
facturer’s instructions.
Enzyme-linked immunosorbent assay
(ELISA) assay
Cytokine levels were measured using
ELISA kits (Abcam, Cambridge, USA), includ-
ing IL-6, IL-1β, IL-18, tumor necrosis factor
(TNF)-α, interferon (IFN)-γ, and IL-10.
Lactate dehydrogenase (LDH) assay
The release of LDH was determined
using the corresponding LDH kit (Abcam,
Cambridge, USA).
Reverse transcription-quantitative
polymerase chain reaction (RT-qPCR)
Total RNA was extracted from the HT22
cells. A HiScript II 1st Strand cDNA Synthe-
sis Kit (Vazyme, China) was used to synthe-
size the cDNA. PCR was performed using the
HiScript II One-Step RT-PCR Kit (Vazyme,
China) on an ABI 7900 system. Glyceralde-
hyde-3-phosphate dehydrogenase served as
the loading control. The mRNA levels were
calculated using the 2ΔΔCT method.
Western blot
HT22 cells were harvested, and total
protein was extracted. After centrifugation at
12000 × g, a BCA assay was performed to de-
termine the protein concentration. Forty mi-
crograms of protein were isolated using 10%
Sodium dodecyl sulphate-polyacrylamide gel
electrophoresis. Proteins were transferred
to polyvinylidene fluoride membranes. After
sealing with 5% skim milk, the membranes
194 Sun and Huang
Investigación Clínica 66(2): 2025
were incubated with primary antibodies, in-
cluding anti-p300 (ab275378; 1: 1000, Ab-
cam, UK), anti-p-p65 (ab32536; 1: 2000,
Abcam, UK), and anti-GAPDH (ab9485; 1:
2500, Abcam, UK) and then with goat anti-
rabbit IgG H&L (ab205718; 1: 10000, Ab-
cam, UK). Subsequently, the bands were im-
aged using an enhanced chemiluminescence
kit (6104-58-1; Sigma-Aldrich, Germany).
Luciferase assay
JASPAR (https://jaspar.elixir.no/) was
used to predict the binding sites between
NF-κB and the promoter of NLRP3. Bind-
ing was amplified and inserted into the
pMIR-GLOTM luciferase vector (Promega).
HEK293T cells were transfected with wild-
type (WT)/mutant type (MUT) of NLRP3
and NF-κB shRNA/overexpression plasmids.
After 48 h, luciferase activity was detected
using a kit (Promega).
Chromatin immunoprecipitation (ChIP)
assay
A ChIP assay was conducted on HT22 cells
using a ChIP kit (Sigma-Aldrich). Briefly, cells
were crosslinked with 1% formalin, afterwards,
cells were lysed and sonicated. The sonicated
chromatin was incubated with antibodies, in-
cluding anti-NF-κB (ab32536; 1: 30, Abcam,
UK) and anti-IgG (ab172730; 1: 50, Abcam,
UK) using Protein G magnetic beads. Finally,
the DNA fragments were analyzed by RT-qPCR.
Cell counting kit-8 (CCK-8) assay
The cells were seeded in a 6-well plate
(4000 cells/well) and cultured for 0, 24, 48,
and 72 h. The cells were then supplemented
with CCK-8 reagent. Finally, cell viability was
determined using a microplate reader at an
absorbance of 450 nm.
Flow cytometry
Neuronal pyroptosis was detected using
flow cytometry with propidium iodide (PI)
and caspase-1 staining. Briefly, neurons were
digested with ethylenediamine tetraacetic
acid-free trypsin. Then, the cells were har-
vested by centrifugation at 1000 rpm for 5
min. Afterwards, cells were resuspended and
washed with PBS twice. The cells were in-
cubated in the dark with FAM FLICA™ Cas-
pase-1 Kit (ICT098; Bio-Rad, USA) and PI (4
μL). The results were analyzed using a flow
cytometer (Biosciences, USA).
Statistical analysis
Each independent experiment was per-
formed in triplicate. Graphpad v.8. software was
used to analyze the data. Data are presented
as the mean±SD. Student’s t-test and ANOVA
were used to analyze differences. p<0.05 was
considered statistically significant.
RESULTS
p300 is upregulated in an in vitro model
of AD
p300 is frequently upregulated in pa-
tients. Therefore, we determined the p300
expression in an AD model in vitro. We found
that p300 mRNA expression in HT-22 cells
exposed to Aβ1-42 was markedly increased
compared with that in the control group
(Fig. 1A). This finding was consistent with
the Western blot results. Aβ1-42 treatment
markedly increased the protein expression
of p300 (Fig. 1B).
p300 deficiency inhibits
neuroinflammation
p300 is a key regulator of the inflam-
matory response and mediates cerebral
injury by activating inflammation-related
signalling. Therefore, we hypothesized that
p300 promotes AD pathogenesis by inducing
neuroinflammation. As shown in Fig. 2A-E,
the release of proinflammatory cytokines,
such as IL-6, IL-1β, IL-18, tumor necrosis
factor (TNF)-α, and interferon (IFN)-γ, was
significantly increased after Aβ1-42 exposure,
whereas IL-10 was markedly decreased (Fig.
2F). However, GNE-049 treatment signifi-
cantly alleviated the effects of Aβ1-42, inhib-
ited the release of IL-6, IL-1β, IL-18, TNF-α,
and IFN-γ, and increased the release of IL-
Effect of the p300/NF-kB pathway in an in vitro model of Alzheimer’s disease 195
Vol. 66(2): 191 - 204, 2025
Fig. 1. The expression of p300 in in vitro model of AD.
(A) RT-qPCR was conducted to detect p300 mRNA expression in HT-22 cells exposed to Aβ1-42. (B)
Western blot was conducted to detect p300 protein expression in HT-22 cells exposed to Aβ1-42. The
difference in comparison was analyzed using the Student t-test. AD: Alzheimer’s disease; p300:
E1A binding protein p300; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; RT-qPCR: reverse
transcription-quantitative polymerase chain reaction. **p<0.01.
Fig. 2. p300 deficiency inhibits neuroinflammation.
(A-F) ELISA was used to detect the release of cytokines in HT-22 cells. (G-L) RT-qPCR was conducted to detect
cytokine mRNA expression in HT-22 cells. Comparison difference was analyzed using one-way ANOVA. p300:
E1A binding protein p300; IL-6: interleukin 6; IL-1β: interleukin 1β; IL-18: interleukin 18, TNF-α: tumor
necrosis factor α; IFN-γ: interferon γ; IL-10: interleukin 10. ELISA: enzyme-linked immunosorbent assay;
RT-qPCR: reverse transcription-quantitative polymerase chain reaction. **p<0.01, ***p<0.001.
196 Sun and Huang
Investigación Clínica 66(2): 2025
10. This finding is consistent with the RT-qP-
CR results. GNE-049 treatment suppressed
the mRNA expression of IL-6, IL-1β, IL-18,
TNF-α, and IFN-γ (Fig. 2 G-K), while increas-
ing IL-10 mRNA expression (Fig. 2 L). These
findings suggest that the inhibition of p300
expression suppresses neuroinflammation.
p300 deficiency inhibits the pyroptosis
of neurons
Inflammation-induced pyroptosis,
which is characterized by the activation of
the inflammasome and an increase in cy-
totoxicity and death, is a key cause of AD.
p300 protein expression was markedly re-
duced by GNE-049 treatment (Fig. 3A). Aβ1-
42 exposure markedly increased LDH release
(Fig. 3B), which was antagonized by GNE-
049 treatment. Moreover, GNE-049 treat-
ment promoted neuronal viability (Fig. 3C).
GNE-049 treatment markedly alleviated the
pyroptosis in the neurons induced by Aβ1-42
exposure (Fig. 3D). Additionally, GNE-049
treatment suppressed the mRNA expression
of NLRP3, PYD and CARD domain contain-
ing (ASC), and caspase-1 (Fig. 3E). These
findings suggested that p300 inhibition al-
leviates neuronal pyroptosis in AD.
Fig. 3. p300 deficiency inhibits the pyroptosis of neurons.
(A) p300 protein expression was detected using Western blot. (B) LDH assay was conducted to detect
cytotoxicity of HT-22 cells. (C) CCK-8 was performed to determine the cell viability of HT-22 cells.
(D) Flow cytometry was used to detect the pyroptosis of HT-22 cells. (E) RT-qPCR was conducted to
detect mRNA expression in HT-22 cells. Comparison difference was analyzed using one-way ANOVA.
p300: E1A binding protein p300; NLRP3: NLR family pyrin domain containing 3; ASC: PYD and CARD
domain containing; LDH: lactate dehydrogenase; RT-qPCR: reverse transcription-quantitative polyme-
rase chain reaction. **p<0.01, ***p<0.001.
Effect of the p300/NF-kB pathway in an in vitro model of Alzheimer’s disease 197
Vol. 66(2): 191 - 204, 2025
p300 activates NF-κB signaling
p300 participates in the inflammatory
response by activating inflammatory signal-
ling. Therefore, we hypothesized that p300
mediates neuroinflammation by activating
NF-κB signaling. Aβ1-42 exposure significantly
increased the protein expression of p-p65
(Fig. 4), which GNE-049 alleviated.
p300 induces neuroinflammation via
activating NF-κB signaling
To verify the role of NF-κB in AD,
neurons were transfected with an NF-κB-
overexpression plasmid. We found that over-
expression of NF-κB alleviated the effects of
GNE-049 and promoted the release of IL-6,
IL-1β, IL-18, TNF-α, and IFN-γ, as well as
decreased IL-10 (Fig.5A-F). Moreover, over-
expression of NF-κB markedly increased
the mRNA expression of IL-6, IL-1β, IL-18,
TNF-α, and IFN-γ (Fig. 5G-K), but decreased
IL-10 mRNA expression (Fig. 5L).
p300 induces pyroptosis via activating NF-
κB signaling
A rescue assay was conducted to con-
firm further the role of p300/NF-κB signal-
ing in AD. We found that overexpression
of NF-κB markedly alleviated the effects of
GNE-049 and promoted neuronal cytotox-
icity (Fig. 6A). However, NF-κB overexpres-
sion suppressed neuronal viability (Fig. 6B).
Overexpression of NF-κB alleviated the ef-
fects of GNE-049 and promoted pyroptosis
in neurons (Fig. 6C). Moreover, overexpres-
sion of NF-κB markedly increased the mRNA
expression of NLRP3, ASC, and caspase-1
(Fig. 6D). These findings suggest that p300
regulates neuronal pyroptosis by activating
NF-κB signaling.
p300-dependent activation of NF-κB
epigenetically activates NLRP3.
NF-κB, a key transcription factor in
inflammatory signalling, participates in
biological processes via its downstream reg-
ulation. We found that NLRP3 mRNA expres-
sion was markedly increased by p300 overex-
pression and returned to normal levels after
transfection with NF-κB shRNA (Fig. 7A).
NF-κB regulates its downstream activity by
binding to the promoters of its target genes.
Therefore, we hypothesized that NF-κB binds
to the NLRP3 promoter (Fig. 7 B). Fig. 7C
shows the binding motif for NF-κB. Four
binding sites were identified in the promoter
of NLRP3 (Fig. 7c). Overexpression of p300
and NF-κB markedly enhanced the transcrip-
tion of NLRP3 (Fig. 7D). To identify the site
that binds NF-κB, the 3’-UTR of the binding
Fig. 4. p300 activates NF-κB signaling.
Western blot was performed to detect the p-p65 protein expression in HT-22 cells. Comparison diffe-
rence was analyzed using one-way ANOVA. p300: E1A binding protein p300; GAPDH: glyceraldehyde-
3-phosphate dehydrogenase. **p<0.01, ***p<0.001.
198 Sun and Huang
Investigación Clínica 66(2): 2025
sites was mutated and inserted into lucifer-
ase reporters. Co-transfection with p300 and
NF-κB significantly increased luciferase ac-
tivity (Fig. 7E). Moreover, luciferase activity
was markedly increased in MUT1/3/4 cells
after Aβ1-42 exposure, which was antagonized
by GNE-049 treatment (Fig. 7F), whereas
there was no significant alteration in MUT2.
Additionally, p300 deficiency markedly
suppressed the co-occupancies of site2 in
HEK293T cells (Fig. 7G-H). These findings
suggest that the p300-mediated activation
of NF-κB epigenetically upregulates NLRP3
expression.
Fig. 5. p300 induces neuroinflammation via activating NF-κB signalling.
(A-F) ELISA was used to detect the release of cytokines in HT-22 cells. (G-L) RT-qPCR was performed
to detect the p300 mRNA expression in HT-22 cells. Comparison difference was analyzed using one-
way ANOVA. p300: E1A binding protein p300; IL-6: interleukin 6; IL-1β: interleukin 1β; IL-18: inter-
leukin 18, TNF-α: tumor necrosis factor α; IFN-γ: interferon γ; IL-10: interleukin 10. ELISA: enzyme-
linked immunosorbent assay; RT-qPCR: reverse transcription-quantitative polymerase chain reaction.
*p<0.05, **p<0.01, ***p<0.001.
Effect of the p300/NF-kB pathway in an in vitro model of Alzheimer’s disease 199
Vol. 66(2): 191 - 204, 2025
DISCUSSION
In this study, p300 was upregulated in
an in vitro model of AD. Interestingly, p300
deficiency inhibits neuroinflammation and
suppresses pyroptosis in the neurons. More-
over, p300 activates NF-κB, and its overex-
pression promotes pyroptosis in neurons.
Additionally, the p300-mediated activation
of NF-κB epigenetically upregulates NLRP3,
which induces pyroptosis in neurons. There-
fore, the p300/NF-κB/NLRP3 pathway may
be a potential target in AD.
p300 is aberrantly expressed in several
brain disorders. Chatterjee et al.24 revealed
that CBP/p300 activation enhances neuro-
Fig. 6. p300 induces pyroptosis via activating NF-κB signaling.
(A) The LDH assay detected cytotoxicity in HT-22 cells. (B) The CCK-8 assay was performed to deter-
mine the viability of HT-22 cells. (C) Flow cytometry was used to detect pyroptosis in the HT-22 cells.
(D) RT-qPCR was performed to detect the mRNA expression in HT-22 cells. Comparison difference was
analyzed using one-way ANOVA. p300: E1A binding protein p300; NLRP3: NLR family pyrin domain
containing 3; ASC: PYD and CARD domain containing; LDH: lactate dehydrogenase; RT-qPCR: reverse
transcription-quantitative polymerase chain reaction. **p<0.01, ***p<0.001.
200 Sun and Huang
Investigación Clínica 66(2): 2025
genesis and prolongs memory duration, and
maturation, and differentiation of adult neu-
ronal progenitors. However, hyperactivation
of p300 contributes to tauopathy pathogen-
esis 25. Therefore, p300 may play protective
and passive roles in brain disorders. This may
be because the roles of p300 vary with dis-
ease subtype and signalling. Therefore, it is
crucial to identify the role of p300 in AD,
which is very important. CBP/p300 activa-
tion-mediated acetylation of tau exacerbates
traumatic brain injury, which is the most
significant non-genetic, non-aging-related
risk factor for AD 26. Moreover, p300-medi-
ated autophagy promoted neuronal damage
and inflammation in AD 27. Therefore, p300
may promote the pathogenesis of AD. In this
study, p300 was upregulated in an in vitro
AD model. Targeting p300 may be an effec-
tive strategy for alleviating AD 25,26. In this
study, p300 deficiency induced by a specific
inhibitor suppressed neuroinflammation and
pyroptosis in the neurons. These findings
suggest that p300 inhibition may alleviate
neuronal loss during AD pathogenesis, con-
sistent with previous studies.
p300, an acetyl transferase, has no
DNA-binding domain 28. p300 regulates gene
expression by interacting with transcription
factors. For instance, Sox8 induces the acti-
vation of the LIF interleukin 6 family cyto-
kine downstream transcription factor signal
transducer and activator of transcription 3
via p300 to promote astrocytic differentia-
Fig. 7. p300-depedent activation of NF-κB epigenetically activates NLRP3.
(A) RT-qPCR was conducted to detect mRNA expression in HT-22 cells. (B) A hypothesis of NF-κB/
NLRP3 signalling. (C) JASPAR was used to analyze the binding motif of NF-κB. (D) JASPAR was used
to analyze the binding sites between NF-κB and the promoter of NLRP3. (E) Luciferase assay was
conducted to confirm that the p300/NF-κB promoted the transcription of NLRP3 in HEK293T cells.
(F) Luciferase assay was performed to verify the exact binding site between NF-κB and the promoter
of NLRP3 in HEK293T cells. (G-H) ChIP assay was performed to verify the binding sites in HEK293T
cells. The difference in comparison was analyzed using one-way or two-way ANOVA. p300: E1A binding
protein p300; NLRP3: NLR family pyrin domain containing 3; WT: wild type; MUT: mutant type; RT-
qPCR: reverse transcription-quantitative polymerase chain reaction. ChIP: chromatin immunopreci-
pitation. **p<0.01, ***p<0.001.
Effect of the p300/NF-kB pathway in an in vitro model of Alzheimer’s disease 201
Vol. 66(2): 191 - 204, 2025
tion 29. Moreover, REST recruits CBP/p300
to the EAAT2 promoter to alleviate manga-
nese-induced excitotoxicity 30. Geong et al.
31 reported that p300/NF-κB promotes mi-
croglial activation and neuroinflammation.
In the present study, p300 activated NF-κB.
NF-κB is a key regulator of inflammatory
signalling. NF-κB is a key mediator of brain
inflammation in AD 32. Activating NF-κB sig-
naling stimulates the innate immune system
and induces neurodegeneration and neuro-
nal loss 33-35. In this study, overexpression of
NF-κB antagonized the effects of GNE-049
and promoted neuroinflammation and neu-
ronal death.
Pyroptosis is a type of inflammation-re-
lated cell death process. Inflammation also
induces necroptosis 36. Although pyropto-
sis and necroptosis share some properties,
such as lytic and inflammatory types of pro-
grammed cell death and releasing damage-
associated molecular patterns, pyroptosis
is differentiated from necroptosis, a backup
cell death defense mechanism. In contrast,
pyroptosis is a primary cellular response af-
ter sensing potentially damaging insults37.
Necroptosis is characterized by the activa-
tion of receptor-interacting serine/threonine
kinase 3/mixed lineage kinase domain-like
pseudokinase signalling, whereas pyropto-
sis is characterized by the activation of in-
flammasomes and executed by GSDMD38. In
this study, p300-dependent upregulation of
NF-κB activated the NLRP3 inflammasome
and increased the release of IL-1β and IL-18
after Aβ1-42 exposure. Therefore, the p300/
NF-κB/NLRP3 pathway-mediated neuronal
death occurred via pyroptosis.
In conclusion, p300 was upregulated in
an in vitro model of AD. p300-mediated up-
regulation of NF-κB epigenetically activates
the NLRP3 inflammasome and pyroptosis in
neurons. However, p300 inhibition alleviated
neuroinflammation and neuronal pyroptosis.
Therefore, targeting p300/NF-κB/NLRP3 may
be a promising strategy for alleviating AD.
Ethics approval and consent to participate
This study was approved by the Ethics
Committee of the Third People’s Hospital of
Yunnan Province. All experimental animal
procedures followed the Guidelines for the
Care and Use of Laboratory Animals for-
mulated by China’s Ministry of Science and
Technology.
Funding
Not applicable.
Consent for publication
Not applicable.
Availability of data and material
The datasets used and/or analyzed dur-
ing the current study are available from the
corresponding author upon reasonable re-
quest.
Competing interests
Not applicable.
Number ORCID of authors
Fengqin Sun (FQS):
0009-0009-0865-8011
Wei Huang (WH):
0009-0008-9840-1633.
Author contributions
FQS performed data analysis and draft-
ed the manuscript. FQS and WH conceived
and supervised the study and reviewed the
manuscript. FQS and WH ran the software
and modified the code. FQS and WH were
involved in the study design and contributed
to the data collection procedure and inter-
pretation.
ACKNOWLEDGEMENTS
Not applicable.
202 Sun and Huang
Investigación Clínica 66(2): 2025
Conflicts of interest
The authors declared that they have no
conflicts of interest regarding this work.
REFERENCES
1. Scheltens P, De Strooper B, Kivipelto
M, Holstege H, Chetelat G, Teunissen
CE, et al. Alzheimer’s disease. Lancet.
2021;397(10284):1577-90. doi: 10.1016/
S0140-6736(20)32205-4.
2. Porsteinsson AP, Isaacson RS, Knox
S, Sabbagh MN, Rubino I. Diagno-
sis of Early Alzheimer’s Disease: Clini-
cal Practice in 2021. J Prev Alzheimers
Dis. 2021;8(3):371-86. doi: 10.14283/
jpad.2021.23.
3. 2023 Alzheimer’s disease facts and figu-
res. Alzheimers Dement. 2023;19(4):1598-
695. doi: 10.1002/alz.13016.
4. Graff-Radford J, Yong KXX, Apostolova
LG, Bouwman FH, Carrillo M, Dicker-
son BC, et al. New insights into atypical
Alzheimer’s disease in the era of biomar-
kers. Lancet Neurol. 2021;20(3):222-34.
doi: 10.1016/S1474-4422(20)30440-3.
5. Wang Q, Gao F, Dai LN, Zhang J, Bi D,
Shen Y. Clinical Research Investigating
Alzheimer’s Disease in China: Current Sta-
tus and Future Perspectives Toward Preven-
tion. J Prev Alzheimers Dis. 2022;9(3):532-
41. doi: 10.14283/jpad.2022.46.
6. Fan R, Peng X, Xie L, Dong K, Ma D, Xu W,
et al. Importance of Bmal1 in Alzheimer’s
disease and associated aging-related disea-
ses: Mechanisms and interventions. Aging
Cell. 2022;21(10):e13704. doi: 10.1111/
acel.13704.
7. Chen P, Guo Z, Zhou B. Insight into the
role of adult hippocampal neurogenesis
in aging and Alzheimer’s disease. Ageing
Res Rev. 2023;84:101828. doi: 10.1016/j.
arr.2022.101828.
8. Yang F, Bettadapura SN, Smeltzer MS,
Zhu H, Wang S. Pyroptosis and pyroptosis-
inducing cancer drugs. Acta Pharmacol
Sin. 2022;43(10):2462-73. doi: 10.1038/
s41401-022-00887-6.
9. Moonen S, Koper MJ, Van Schoor E,
Schaeverbeke JM, Vandenberghe R, von
Arnim CAF, et al. Pyroptosis in Alzheimer’s
disease: cell type-specific activation in
microglia, astrocytes and neurons. Acta
Neuropathol. 2023;145(2):175-95. doi:
10.1007/s00401-022-02528-y.
10. Zhou J, Qiu J, Song Y, Liang T, Liu S, Ren
C, et al. Pyroptosis and degenerative diseases
of the elderly. Cell Death Dis. 2023;14(2):94.
doi: 10.1038/s41419-023-05634-1.
11. Elias EE, Lyons B, Muruve DA. Gasder-
mins and pyroptosis in the kidney. Nat
Rev Nephrol. 2023;19(5):337-50. doi:
10.1038/s41581-022-00662-0.
12. Li Z, Ji S, Jiang ML, Xu Y, Zhang CJ.
The Regulation and Modification of GSD-
MD Signaling in Diseases. Front Immu-
nol. 2022;13:893912. doi: 10.3389/
fimmu.2022.893912.
13. Huang Y, Xu W, Zhou R. NLRP3 inflam-
masome activation and cell death. Cell
Mol Immunol. 2021;18(9):2114-27. doi:
10.1038/s41423-021-00740-6.
14. Yao H, Zhang D, Yu H, Yuan H, Shen H, Lan
X, et al. Gut microbiota regulates chronic
ethanol exposure-induced depressive-like
behavior through hippocampal NLRP3-me-
diated neuroinflammation. Mol Psychiatry.
2023;28(2):919-30. doi: 10.1038/s41380-
022-01841-y.
15. Hou Y, Wei Y, Lautrup S, Yang B, Wang Y,
Cordonnier S, et al. NAD(+) supplemen-
tation reduces neuroinflammation and cell
senescence in a transgenic mouse model
of Alzheimer’s disease via cGAS-STING.
Proc Natl Acad Sci U S A. 2021;118(37).
doi: 10.1073/pnas.2011226118.
16. Han YH, Liu XD, Jin MH, Sun HN, Kwon
T. Role of NLRP3 inflammasome-media-
ted neuronal pyroptosis and neuroinflam-
mation in neurodegenerative diseases.
Inflamm Res. 2023;72(9):1839-59. doi:
10.1007/s00011-023-01790-4.
17. Cai Y, Chai Y, Fu Y, Wang Y, Zhang Y,
Zhang X, et al. Salidroside Ameliorates
Alzheimer’s Disease by Targeting NLRP3
Inflammasome-Mediated Pyroptosis. Front
Aging Neurosci. 2021;13:809433. doi:
10.3389/fnagi.2021.809433.
Effect of the p300/NF-kB pathway in an in vitro model of Alzheimer’s disease 203
Vol. 66(2): 191 - 204, 2025
18. Chakraborty R, Ostriker AC, Xie Y,
Dave JM, Gamez-Mendez A, Chatter-
jee P, et al. Histone Acetyltransferases
p300 and CBP Coordinate Distinct Chro-
matin Remodeling Programs in Vascular
Smooth Muscle Plasticity. Circulation.
2022;145(23):1720-37. doi: 10.1161/CIR-
CULATIONAHA.121.057599.
19. Xu Y, Wan W. Acetylation in the regulation of
autophagy. Autophagy. 2023;19(2):379-87.
doi: 10.1080/15548627.2022.2062112.
20. Chen Q, Yang B, Liu X, Zhang XD, Zhang
L, Liu T. Histone acetyltransferases CBP/
p300 in tumorigenesis and CBP/p300 in-
hibitors as promising novel anticancer
agents. Theranostics. 2022;12(11):4935-
48. doi: 10.7150/thno.73223.
21. Cao W, Feng Z, Zhu D, Li S, Du M, Ye S,
et al. The Role of PGK1 in Promoting Is-
chemia/Reperfusion Injury-Induced Mi-
croglial M1 Polarization and Inflammation
by Regulating Glycolysis. Neuromolecular
Med. 2023;25(2):301-11. doi: 10.1007/
s12017-023-08736-3.
22. Bai B, Zhang Q, Wan C, Li D, Zhang T, Li
H. CBP/p300 inhibitor C646 prevents high
glucose exposure induced neuroepithe-
lial cell proliferation. Birth Defects Res.
2018;110(14):1118-28. doi: 10.1002/
bdr2.1360.
23. Cintra MTG, Avila RT, Soares TO, Cun-
ha LCM, Silveira KD, de Moraes EN, et
al. Increased N200 and P300 latencies in
cognitively impaired elderly carrying ApoE
epsilon-4 allele. Int J Geriatr Psychia-
try. 2018;33(2):e221-e7. doi: 10.1002/
gps.4773.
24. Chatterjee S, Mizar P, Cassel R, Neidl R,
Selvi BR, Mohankrishna DV, et al. A novel
activator of CBP/p300 acetyltransferases
promotes neurogenesis and extends me-
mory duration in adult mice. J Neurosci.
2013;33(26):10698-712. doi: 10.1523/
JNEUROSCI.5772-12.2013.
25. Chen X, Li Y, Wang C, Tang Y, Mok SA,
Tsai RM, et al. Promoting tau secretion
and propagation by hyperactive p300/CBP
via autophagy-lysosomal pathway in tauo-
pathy. Mol Neurodegener. 2020;15(1):2.
doi: 10.1186/s13024-019-0354-0.
26. Shin MK, Vazquez-Rosa E, Koh Y, Dhar M,
Chaubey K, Cintron-Perez CJ, et al. Re-
ducing acetylated tau is neuroprotective in
brain injury. Cell. 2021;184(10):2715-32
e23. doi: 10.1016/j.cell.2021.03.032.
27. Wu J, Han Y, Xu H, Sun H, Wang R, Ren
H, et al. Deficient chaperone-mediated
autophagy facilitates LPS-induced mi-
croglial activation via regulation of the
p300/NF-kappaB/NLRP3 pathway. Sci
Adv. 2023;9(40):eadi8343. doi: 10.1126/
sciadv.adi8343.
28. Kikuchi M, Morita S, Wakamori M, Sato
S, Uchikubo-Kamo T, Suzuki T, et al. Epi-
genetic mechanisms to propagate histone
acetylation by p300/CBP. Nat Commun.
2023;14(1):4103. doi: 10.1038/s41467-
023-39735-4.
29. Takouda J, Katada S, Imamura T, Sanosa-
ka T, Nakashima K. SoxE group transcrip-
tion factor Sox8 promotes astrocytic diffe-
rentiation of neural stem/precursor cells
downstream of Nfia. Pharmacol Res Pers-
pect. 2021;9(6):e00749. doi: 10.1002/
prp2.749.
30. Pajarillo E, Digman A, Nyarko-Dan-
quah I, Son DS, Soliman KFA, Aschner
M, et al. Astrocytic transcription factor
REST upregulates glutamate transporter
EAAT2, protecting dopaminergic neurons
from manganese-induced excitotoxicity.
J Biol Chem. 2021;297(6):101372. doi:
10.1016/j.jbc.2021.101372.
31. Jeong GW, Lee HH, Lee-Kwon W, Kwon
HM. Microglial TonEBP mediates LPS-
induced inflammation and memory loss
as transcriptional cofactor for NF-ka-
ppaB and AP-1. J Neuroinflammation.
2020;17(1):372. doi: 10.1186/s12974-
020-02007-9.
32. Chen S, Liu H, Wang S, Jiang H, Gao L,
Wang L, et al. The Neuroprotection of Ver-
bascoside in Alzheimer’s Disease Mediated
through Mitigation of Neuroinflammation
via Blocking NF-kappaB-p65 Signaling.
Nutrients. 2022;14(7). doi: 10.3390/
nu14071417.
33. Zhou L, Kong G, Palmisano I, Cencioni
MT, Danzi M, De Virgiliis F, et al. Rever-
sible CD8 T cell-neuron cross-talk causes
204 Sun and Huang
Investigación Clínica 66(2): 2025
aging-dependent neuronal regenerative de-
cline. Science. 2022;376(6594):eabd5926.
doi: 10.1126/science.abd5926.
34. Yu CH, Davidson S, Harapas CR, Hilton
JB, Mlodzianoski MJ, Laohamonthonkul
P, et al. TDP-43 Triggers Mitochondrial
DNA Release via mPTP to Activate cGAS/
STING in ALS. Cell. 2020;183(3):636-49
e18. doi: 10.1016/j.cell.2020.09.020.
35. Jung BK, Park Y, Yoon B, Bae JS, Han
SW, Heo JE, et al. Reduced secretion
of LCN2 (lipocalin 2) from reactive as-
trocytes through autophagic and pro-
teasomal regulation alleviates inflam-
matory stress and neuronal damage.
Autophagy. 2023;19(8):2296-317. doi:
10.1080/15548627.2023.2180202.
36. Chen S, Guan S, Yan Z, Ouyang F, Li S,
Liu L, et al. Role of RIPK3CaMKIImPTP
signaling pathwaymediated necroptosis
in cardiovascular diseases (Review). Int
J Mol Med. 2023;52(4). doi: 10.3892/
ijmm.2023.5301.
37. Frank D, Vince JE. Pyroptosis versus ne-
croptosis: similarities, differences, and
crosstalk. Cell Death Differ. 2019;26(1):99-
114. doi: 10.1038/s41418-018-0212-6.
38. Gao W, Wang X, Zhou Y, Wang X, Yu Y.
Autophagy, ferroptosis, pyroptosis, and ne-
croptosis in tumor immunotherapy. Signal
Transduct Target Ther. 2022;7(1):196. doi:
10.1038/s41392-022-01046-3.