ArticlePDF Available

Cullin 4A/protein arginine methyltransferase 5 (CUL4A/PRMT5) promotes cell malignant phenotypes and tumor growth in nasopharyngeal carcinoma

Taylor & Francis
Bioengineered
Authors:

Abstract and Figures

Targeted therapy is an important therapeutic strategy currently, however, the development of targeted therapy for nasopharyngeal carcinoma (NPC) is relatively lagging. Cullin 4A (CUL4A) was reported to be overexpressed in NPC; nevertheless, the specific role of CUL4A remains unrevealed. NPC cells and tumor-bearing mice were cultivated to explore the role and mechanism of CUL4A in NPC. After evaluating CUL4A levels in NPC cells, functional experiments were carried out to investigate the effects of CUL4A knockdown and overexpression on cell proliferative, invasive and migratory aptitude as well as NF-κB signaling. Following the GeneMANIA database predicted that protein arginine methyltransferase 5 (PRMT5) was downstream of CUL4A, the mediated role of PRMT5 in the regulation of CUL4A on cells was then determined. Moreover, the tumor volumes and weights of tumor-bearing mice were recorded, and the levels of proliferation-, migration-, and NF-κB signaling-related proteins in the tumor were determined. Herein, CUL4A was enhanced in NPC cells, and its knockdown and overexpression separately suppressed and promoted cell proliferative, invasive, and migratory aptitude as well as NF-κB signal activation. Novelty, PRMT5 knockdown reversed the influences of CUL4A overexpression on these aspects. In addition, its knockdown likewise reversed the facilitating impact of CUL4A expression on tumor growth and declined the expression levels of proliferation-, migration-, and NF-κB signaling-related protein in the tumor. Together, this paper indicated that CUL4A promoted the proliferative, invasive, and migratory aptitude of NPC cells as well as tumor growth by promoting PRMT5 to activate NF-κB signaling.
This content is subject to copyright. Terms and conditions apply.
RESEARCH PAPER
Cullin 4A/protein arginine methyltransferase 5 (CUL4A/PRMT5) promotes cell
malignant phenotypes and tumor growth in nasopharyngeal carcinoma
Xiuying Sun, Jinhui Zhou, and Zhicun Zhang
Department of Otolaryngology, The Affiliated Huai’an No. 1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu, China
ABSTRACT
Targeted therapy is an important therapeutic strategy currently, however, the development
of targeted therapy for nasopharyngeal carcinoma (NPC) is relatively lagging. Cullin 4A
(CUL4A) was reported to be overexpressed in NPC; nevertheless, the specific role of CUL4A
remains unrevealed. NPC cells and tumor-bearing mice were cultivated to explore the role
and mechanism of CUL4A in NPC. After evaluating CUL4A levels in NPC cells, functional
experiments were carried out to investigate the effects of CUL4A knockdown and over-
expression on cell proliferative, invasive and migratory aptitude as well as NF-κB signaling.
Following the GeneMANIA database predicted that protein arginine methyltransferase 5
(PRMT5) was downstream of CUL4A, the mediated role of PRMT5 in the regulation of
CUL4A on cells was then determined. Moreover, the tumor volumes and weights of tumor-
bearing mice were recorded, and the levels of proliferation-, migration-, and NF-κB signaling-
related proteins in the tumor were determined. Herein, CUL4A was enhanced in NPC cells,
and its knockdown and overexpression separately suppressed and promoted cell prolifera-
tive, invasive, and migratory aptitude as well as NF-κB signal activation. Novelty, PRMT5
knockdown reversed the influences of CUL4A overexpression on these aspects. In addition,
its knockdown likewise reversed the facilitating impact of CUL4A expression on tumor
growth and declined the expression levels of proliferation-, migration-, and NF-κB signaling-
related protein in the tumor. Together, this paper indicated that CUL4A promoted the
proliferative, invasive, and migratory aptitude of NPC cells as well as tumor growth by
promoting PRMT5 to activate NF-κB signaling.
ARTICLE HISTORY
Received 29 January 2022
Revised 9 March 2022
Accepted 14 March 2022
KEYWORDS
Cullin 4A; protein arginine
methyltransferase 5; p65;
nasopharyngeal carcinoma;
C666-1 cell
CONTACT Xiuying Sun sunxiuy2107@163.com Department of Otolaryngology, The Affiliated Huai’an No. 1 People’s Hospital of Nanjing Medical
University, 1 Huanghe West Road, Huai’an, Jiangsu 223300, China
BIOENGINEERED
2022, VOL. 13, NO. 4, 8712–8723
https://doi.org/10.1080/21655979.2022.2054756
© 2022 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted
use, distribution, and reproduction in any medium, provided the original work is properly cited.
Introduction
Nasopharyngeal carcinoma (NPC) is an epithelial
malignant tumor that occurs in the nasopharynx.
It originates from the pharyngeal recesses and
easily invades adjacent tissues and lymph nodes
[1]. The degree of malignancy is relatively high
and distant metastasis can occur [2]. NPC was
divided into keratinizing, non-keratinizing, and
basaloid squamous cell carcinoma by World
Health Organization according to its histology
[3]. Numerous epidemiological surveys indicate
that Epstein-Barr virus (EBV) infection is the
most universal cause, meanwhile, environmental
and genetic factors may also be involved [4].
EBV infection is common in the non-keratinizing
type, taking up over 90% of cases; EBV is rare in
keratinizing type with a relatively favorable prog-
nosis [5]. Because NPC has an insidious onset and
is prone to regional lymph nodes and distant
hematogenous metastasis, the most patients are
in the middle and late stages when the disease is
clinically diagnosed [6]. Intensity-modulated
radiotherapy (IMRT) can significantly improve
the local control rate of NPC [7], however, distant
metastasis, especially to the lung, bone, and liver,
is still an important reason for failed treatment [2].
Molecular targeted therapy of tumors has become
a hot spot in the field of tumor research, and it is
also an important therapeutic strategy currently.
Compared with other tumors, the development of
targeted therapy of NPC is relatively lagging.
Being a bridging protein in the ubiquitin ligase
complex, Cullin 4A (CUL4A) is involved in the
ubiquitination degradation and ubiquitination mod-
ification of proteins in vivo and determines the
specificity of substrate proteins [8]. Previous studies
have revealed that CUL4A is involved in various
physiological activities of cells, including the regula-
tion of cell cycle, the transduction of signal, the
repair of damaged DNA, the methylation of histone,
the regulation of transcription as well as the activa-
tion of an oncogene [9]. Therefore, abnormal
expression of CUL4A protein may affect the phy-
siological activities of cells and lead to tumorigenesis
[10]. Overexpression of CUL4A protein was initially
found in primary breast cancer, suggesting that
CUL4A overexpression has closed relation to the
development of breast cancer [11]. Subsequent
studies found that CUL4A overexpression also
occurred in the prostate, colon, liver, as well as
lung cancer [
12–15
]. Of note, it was evidenced that
CUL4A was overexpressed in NPC [16], nonethe-
less, the role of CUL4A, as well as its mechanism,
has not been revealed.
Therefore, this study selected NPC cells and
constructed tumor-bearing mouse model to
explore the role and mechanism of CUL4A in
NPC. Exploring molecular therapeutic targets is
of great significance for enriching the comprehen-
sive treatment strategies for patients with NPC and
improving the therapeutic efficacy.
Materials and methods
Cell culture
Human nasopharyngeal epithelial NP69 cells
(Biobw, Beijing, China) and NPC cells C666-1,
5–8 F, 6–10B (Meisen, Hangzhou, China), and
SUNE-1 (EK-Bioscience, Shanghai, China) were
cultured in RPMI-1640 (Gibco; Thermo Fisher
Scientific) supplemented with 10% FBS (Gibco)
and 1% P/S in an incubator set at 37°C and 5%
CO
2
[17].
Cell transfection
CUL4A knockdown was achieved by transfection
with small interfering (si)RNA targeting CUL4A
(siRNA-CUL4A-1/2), and a non-targeted siRNA
was used as negative control (siRNA-NC).
CUL4A overexpression was achieved through the
transfection with pcDNA3.1 vector containing
CUL4A (Ov-CUL4A), and the empty vector was
used as Ov-NC. PRMT5 knockdown was achieved
by transfection with short hairpin RNA (shRNA)
targeting PRMT5 (shRNA-PRMT5-1/2), and
a scrambled shRNA was used as shRNA-NC. All
these plasmids were provided by GenePharma
(Shanghai, China). The transfection [18] was car-
ried out with the aid of Lipofectamine® 2000
(Invitrogen; Thermo Fisher Scientific).
Preparation of tumor transplants
All procedures followed the ethical guidelines of The
Affiliated Huai’an No. 1 People’s Hospital of Nanjing
Medical University and were approved by The
BIOENGINEERED 8713
Research and Education Animal Use and Care
Committee. Great efforts have been made to relieve
animal suffering. The experiments were carried out
using 18 adult male Balb/c nude mice [19] (age, 4–
6 weeks; weight, 16–18 g; Cyagen Biosciences, Jiangsu,
China) which were housed at a constant ambient
temperature of 21 ± 2°C and relative humidity of
55 ± 5%, with a 12-h light/dark cycle, and free access
to food and water. For each group (Ov-NC, Ov-
CUL4A, and Ov-CUL4A + shRNA-PRMT5), the
resuspension of cells (2 × 10
6
) was separately con-
ducted in a 0.2-ml volume and the cell suspension
was injected using a syringe. The weights of the mice
along with the size of the tumors were recorded every
3 days from the fourth day. A total of 28 days later,
after the nude mice were euthanized using an over-
dose of sodium pentobarbital (100 mg/kg, ip) followed
by cervical dislocation, the tumor tissue was taken out
for the subsequent assays.
Western blotting
Extracted proteins from tumor tissue or cells by
RIPA lysis buffer (Solarbio, Beijing, China) were
quantified with the adoption of a BCA assay
(Solarbio). After the separation with 10% polyacry-
lamide gel, the transfer of proteins (25 μg) was car-
ried out into PVDF membranes (Millipore). Prior to
the incubation with primary antibodies at 4°C, the
blocking of membranes with skimmed milk was
operated. Thereafter, membranes were cut in strips,
HRP-conjugated secondary antibody was applied to
incubate the strips. Taking advantage of an ECL
detection reagent (Millipore), the blots were tracked
and ImageJ version 1.52 software was utilized to
analyze the obtained data [20]. The details of the
antibodies are presented in Table 1.
Reverse transcription-quantitative PCR (RT-qPCR)
The synthesization of RNA isolated by TRIzol®
reagent (Invitrogen) into cDNA was operated with
PrimeScript™ RT Master Mix (Takara, Beijing,
China). qPCR was carried out with the application
of the QuantiTect SYBR-Green PCR kit (Qiagen).
The data were quantified with the ∆∆Cq method
[21] following normalization against GAPDH. The
following were the applied primers: CUL4A forward,
5’-AGGCACAGATCCTTCCGTTT-3’ and reverse,
5’-TCCTGCCAGCACGTGTTAAT-3’; PRMT5 for-
ward, 5’-CTGTCTTCCATCCGCGTTTCA-3’ and
reverse, 5’-GCAGTAGGTCTGATCGTGTCTG-3’;
and GAPDH forward, 5’- GACTCATGACCA
CAGTCCATGC-3’ and reverse, 5’-AGAGGCA
GGGATGATGTTCTG-3’.
Cell Counting Kit-8 (CCK-8) assay
Cells (5 × 10
3
cells/well) that plated into a 96-well
plate were cultured for 24, 48, and 72 h. For each
time point, the CCK-8 solution (Dojindo
Molecular Technologies) was added to each well
[22]. Absorbance was then tracked under the
application of a microplate reader = 450 nm,
Bio-Rad Laboratories).
Co-Immunoprecipitation (Co-IP)
The lysis of cells was operated on ice for 10 min,
following which was the centrifugation at 13,000 x g
for 10 min and the collection of supernatants.
Subsequently, the lysate (500 µg/ IP) was supplemen-
ted with the 2.5 µg CUL4A or PRMT5 antibody and
10 µl protein A + G magnetic beads (Beyotime,
Shanghai, China) followed by gentle rotation at
room temperature for 2 h. The supernatant was
removed by magnetic force and the magnetic beads
with the added 1X SDS sample buffer were boiled for
5 min for western blotting [23].
Wound healing assay
Cells (5 × 10
5
cells/well) in six-well plates were
cultured until ~90% confluence. A 200-μl sterile
pipette tip was applied to create a wound in the
Table 1. Antibodies used for western blotting.
Antibody
Catalog
number Host
Dilution
ratio Company
CUL4A GTX33129 Rabbit 1:1,000 GeneTex
PRMT5 GTX116004 Rabbit 1:1,000 GeneTex
MMP12 ab52897 Rabbit 1:5,000 Abcam
MMP9 ab283575 Rabbit 1:1,000 Abcam
Ki67 ab16667 Rabbit 1:1,000 Abcam
PCNA ab29 Rabbit 1:1,000 Abcam
Phospho-p65 AF5875 Rabbit 1:1,000 Beyotime
p65 AF0246 Rabbit 1:1,000 Beyotime
IKBα ab97783 Rabbit 1:5,000 Abcam
Histone3 Gtx122148 Rabbit 1:5,000 GeneTex
HRP anti-rabbit lgG A0208 goat 1:1,000 Beyotime
8714 X. SUN ET AL.
cell monolayer. Following the rinse with PBS, the
cells were starved for 24 h [24]. Under an inverted
microscope (x100; Olympus Corporation), the
images were visualized at 0 and 24 h.
Transwell assay
Cells (3 × 10
4
cells) were suspended in 1 ml
serum-free medium. The upper chamber was pre-
coated with Matrigel (Sigma-Aldrich) and cells
were then cultivated in the upper chamber with
0.1 ml cell suspension in each well. RPMI-1640
which contains 20% FBS was applied to perfuse
the lower chamber. Following 24 h of incubation,
the fixation and staining with 4% paraformalde-
hyde (Biobw) and 0.5% crystal violet solution
(Yeasen, Shanghai, China) were operated [25].
Finally, the stained cells were totaled under an
inverted microscope (x100).
Colony formation assay
Cells were inoculated into culture dishes at a density
of 500 cells/dish. After discarding the supernatants,
the cells were fixed with 4% paraformaldehyde
(Biobw). The fixative solution was then replaced
with crystal violet (Yeasen) to stain the cells.
A cluster of >50 cells was considered a colony [26].
Bioinformatics and statistical analysis
GeneMANIA (http://genemania.org/) online data-
bases were used to search for the association
between known proteins [27]. Data that demon-
strated as the mean ± SD were analyzed with
Prism 8.0 software (GraphPad Software). To
show differences among multiple groups, one-
way ANOVA followed by Tukey’s post hoc test
was employed. P less than 0.05 is judged to be of
statistical significance.
Results
This study used NPC cells and tumor-bearing
mouse models to explore the role and mechanism
of CUL4A in NPC. CUL4A expression was
enhanced in NPC cells, and its knockdown and
overexpression separately suppressed and pro-
moted cell proliferative, invasive, and migratory
aptitude as well as NF-κB signal activation.
Novelty, PRMT5 knockdown reversed the influ-
ences of CUL4A overexpression on these aspects.
Moreover, its knockdown also reversed the facil-
itating impact of CUL4A expression on tumor
growth and declined the expression levels of pro-
liferation-, migration-, and NF-κB signaling-
related protein in the tumor. Together, these
results indicated that CUL4A promoted the pro-
liferative, invasive, and migratory aptitude of NPC
cells as well as tumor growth by promoting
PRMT5 to activate NF-κB signaling.
CUL4A expression level impacts NPC cell
proliferation
The levels of CUL4A in NP69, C666-1, 5–8 F, 6–
10B, and SUNE-1 cells were assessed by RT-
qPCR along with western blotting (Figure 1a-b).
Higher expression levels were found in NPC cells
compared with NP69 cells. To highlight the prob-
able effects of CUL4A level on NPC cells, we
adopted C666-1 cells for ensuing experiments.
The C666-1 cells were transfected with siRNAs
or plasmids to knock down or elevate the expres-
sion of CUL4A, respectively. The transfection
efficacy was assessed using RT-qPCR and western
blotting (Figure 1c-f). The level of CUL4A was
effectively down-regulated in the siRNA-CUL4A
-1/2 groups, and the siRNA-CUL4A-1 group was
selected for the following assays due to the rela-
tively lower level. In addition, CUL4A was effec-
tively overexpressed in the Ov-CUL4A group.
Cell proliferation in the knockdown and overex-
pression groups was determined using the CCK-8
assay (Figure 1g). During the period of 24, 48,
96 h, the proliferative ability of cells in the
siRNA-CUL4A group slowed down, whereas the
proliferation in the Ov-CUL4A group accelerated.
Moreover, the levels of Ki67 and PCNA were
evaluated by western blotting (Figure 1h). Their
levels were declined in the siRNA-CUL4A group
and elevated in the Ov-CUL4A group. Colony-
forming efficiency was likewise assessed by plate
colony formation assay (Figure 1i). The colony-
forming efficiency in the siRNA-CUL4A group
was obviously decreased, while that in the Ov-
CUL4A group was improved.
BIOENGINEERED 8715
Figure 1. CUL4A expression level impacts NPC cell proliferation (a) The expression levels of CUL4A in NP69, C666-1, 5–8 F,
6–10B, and SUNE-1 cells were determined using RT-qPCR and (b) western blotting. *P < 0.05, **P < 0.01, ***P < 0.001 vs.
NP69. (c) The efficacy of CUL4A knockdown was assessed using RT-qPCR and (d) western blotting. ***P < 0.001 vs. siRNA-NC.
(e) The efficacy of CUL4A overexpression was assessed using RT-qPCR and (f) western blotting. ***P < 0.001 vs. Ov-NC. (g) Cell
proliferation in each group was determined using the CCK-8 assay. (h) The expression level of Ki67 and PCNA was determined
using western blotting. (i) Colony-forming efficiency was assessed by colony formation assay. **P < 0.01, ***P < 0.001 vs.
siRNA-NC;
##
P < 0.01,
###
P < 0.001 vs. Ov-NC.
8716 X. SUN ET AL.
CUL4A expression level impacts NPC cell
migration and invasion, and NF-κB signaling
The cell migratory and invasive aptitude were
assessed employing wound healing along with
Transwell assay (Figure 2a-b). The migration rate
of cells in the siRNA-CUL4A group was slightly
decreased and significantly increased in the Ov-
CUL4A group. The invasion rate of cells was sig-
nificantly decreased in the siRNA-CUL4A group
and increased in the Ov-CUL4A group, respec-
tively. In addition, with the adoption of western
blotting, the MMP12 and MMP9 levels were
detected (Figure 2c). Their levels were markedly
declined in the siRNA-CUL4A group and
increased in the Ov-CUL4A group. Furthermore,
as Figure 2d depicted, the levels of nuclear p65 and
IκBα were conspicuously descended in the siRNA-
CUL4A group and elevated in the Ov-CUL4A
group.
CUL4A regulates PRMT5 to impact NF-κB
signaling
To further figure out the mechanism of CUL4A,
the GeneMANIA database was applied to search
proteins associated with CUL4A (Figure 3a).
Figure 2. CUL4A expression level impacts NPC cell migration and invasion, and NF-κB signaling (a) Cell migration was
determined using wound healing assay. (b) Cell invasion was determined using Transwell assay. (c) The expression levels of MMP12
and MMP9 were determined using western blotting. (d) The expression level of nuclear p65, total p65, and IκBα were determined
using western blotting. **P < 0.01, ***P < 0.001 vs. siRNA-NC;
#
P < 0.05,
##
P < 0.01,
###
P < 0.001 vs. Ov-NC.
BIOENGINEERED 8717
PRMT5 was found to be co-expressed with
CUL4A. Hence, the levels of PRMT5 in NP69,
C666-1, 5–8 F, 6–10B, and SUNE-1 cells were
assessed with RT-qPCR and western blotting
(Figure 3b-c). The level of PRMT5 was enhanced
in the NPC cells. Moreover, the PRMT5 level was
markedly declined in the siRNA-CUL4A group
and elevated in the Ov-CUL4A group (Figure 3d-
e). Afterward, the association between CUL4A and
PRMT5 was verified using the Co-IP assay (figure
3f). The results indicated that CUL4A and PRMT5
could bind to each other and co-precipitate.
Figure 3. CUL4A regulates PRMT5 to impact NF-κB signaling (a) The GeneMANIA database was applied to search proteins
associated with CUL4A. (b) The expression level of PRMT5 in NP69, C666-1, 5–8 F, 6–10B, and SUNE-1 cells were determined using
RT-qPCR and (c) western blotting. ***P < 0.001 vs. NP69. (d) The expression level of PRMT5 in the transfected cells was determined
using RT-qPCR and (e) western blotting. ***P < 0.001 vs. siRNA-NC;
###
P < 0.001 vs. Ov-NC. (f) The association between CUL4A and
PRMT5 was verified using the Co-IP assay. (g) The efficacy of PRMT5 knockdown was confirmed with RT-qPCR and (h) western
blotting. ***P < 0.001 vs. shRNA-NC. (i) The expression level of nuclear p65, total p65, and IκBα were determined using western
blotting. ***P < 0.001 vs. Ov-NC;
#
P < 0.05,
##
P < 0.01 vs. Ov-CUL4A + shRNA-NC.
8718 X. SUN ET AL.
Following confirming the efficacy of shRNA-
PRMT5 with RT-qPCR along with western blot-
ting (Figure 3g-h), shRNA-PRMT5-1 and Ov-
CUL4A were co-transfected into C666-1 cells.
The levels of nuclear p65, total p65, and IκBα
were evaluated with the application of western
blotting (Figure 3i). PRMT5 knockdown reversed
the elevated expression levels of nuclear p65 and
IκBα caused by CUL4A overexpression.
CUL4A regulates PRMT5 to impact NPC cell
malignant phenotypes
The influences of PRMT5 knockdown on the reg-
ulatory roles of CUL4A in cell malignant pheno-
types were evaluated. Cell proliferative ability was
firstly tracked by CCK-8 assay (Figure 4a), western
blotting (Figure 4b), as well as colony formation
assay (Figure 4c). PRMT5 knockdown reversed the
promotion of cell proliferation resulting from
CUL4A overexpression and simultaneously
reduced the expression levels of Ki67 and PCNA.
Moreover, the colony-forming efficiency in the
Ov-CUL4A + shRNA-PRMT5 group was declined
compared with the Ov-CUL4A + shRNA-NC
group. Next, the capacity of cells to migrate and
invade was determined using wound healing
(Figure 4d), Transwell assay (Figure 4e), and wes-
tern blotting (figure 4f). PRMT5 knockdown like-
wise reversed the promoted migration and
invasion caused by CUL4A overexpression, mean-
while, it reduced the expression levels of MMP12
and MMP9.
CUL4A regulates PRMT5 to promote tumor
growth
Based on cellular experiments, we performed
in vivo study using tumor-bearing mouse models.
The body weights of mice (Figure 5a-b) and the
volumes of tumors (Figure 5c-d) were recorded
over 28 days. Overall, the body weights presented
fluctuating upward trends. Compared with the
Ov-NC group, the weight of the mice in the Ov-
CUL4A group was lower for the first 13 days and
surpassed in the following days. Compared with
the Ov-CUL4A group, the weight of mice in the
Ov-CUL4A + shRNA-PRMT5 group was higher in
the first 20 days, and lower in the remaining days.
The tumor volume in the Ov-CUL4A group
increased sharply and was significantly larger
than that in the Ov-NC group on the day of strip-
ping. The tumor volume in the Ov-CUL4A +
shRNA-PRMT5 group increased slowly and was
tremendously smaller than that in the Ov-CUL4A
group on the day of stripping. The tumor weight
in the Ov-CUL4A group was also greatly larger
than that in the Ov-NC group, and PRMT5 knock-
down greatly inhibited the growth of tumor weight
(Figure 5e). The levels of Ki67, PCNA, MMP12,
and MMP9 in the tumor were assessed by western
blotting (figure 5f). Their levels in the tumor of the
Ov-CUL4A group were elevated and partly
reversed in the Ov-CUL4A + shRNA-PRMT5
group. Furthermore, the levels of CUL4A,
PRMT5, nuclear p65, total p65, and IκBα in the
tumor were assessed by western blotting
(Figure 5g). CUL4A overexpression promoted
intratumoral PRMT5 levels, as well as nuclear
p65 and IκBα levels, whereas PRMT5 knockdown
reduced their levels.
Discussion
The incidence of NPC has unique geographical
distribution characteristics, with the highest inci-
dence in China and Southeast Asian countries,
followed by North Africa, and the least in
Europe, America, and Oceania [28]. This distribu-
tion is thought to be relevant to racial genetics,
consumption of pickled foods, high levels of nickel
in soil and water, and climate suitable for EBV
survival. EBV is best suited to survive in hot and
humid conditions [4,29]. As aforementioned, its
infection is intimately associated with the occur-
rence and advancement of NPC. EBV DNA level
has been identified as markers of tumor burden,
recurrence monitoring, and prognosis in NPC
[30]. The C666-1 cells, majorly studied in this
paper, are EBV-positive cells [31], and the expres-
sion level of CUL4A is higher in them compared
with other cells. As NPC is prone to metastasis,
this study evaluated the effects of low and high
CUL4A expression on C666-1 cell proliferative,
invasive and migratory aptitude, respectively. The
results indicated that down-regulated and up-
regulated CUL4A expression separately inhibited
and promoted cell proliferation, invasion, and
BIOENGINEERED 8719
Figure 4. CUL4A regulates PRMT5 to impact NPC cell malignant phenotypes (a) Cell proliferation level was determined with the
CCK-8 assay. (b) The expression level of Ki67 and PCNA was determined using western blotting. (c) Colony-forming efficiency was
assessed by colony formation assay. (d) Cell migration was determined using a wound healing assay. (e) Cell invasion was
determined using Transwell assay. (f) The expression levels of MMP12 and MMP9 were determined using western blotting.
***P < 0.001 vs. Ov-NC;
#
P < 0.05,
##
P < 0.01,
###
P < 0.001 vs. Ov-CUL4A + shRNA-NC.
8720 X. SUN ET AL.
migration. Although we found that CUL4A has
a regulatory effect on cells as above-mentioned,
the pathway mediated by it remains unknown,
hence, we continued to investigate the
downstream.
EBV can establish latent or lytic infection in
host cells, both of which are involved in the carci-
nogenesis of NPC. Among them, EBV infection of
NPC is dominantly based on latent infection [32].
EBV-encoded latent membrane protein 1 is the
primary oncogenic protein of NPC, and many of
its downstream events are mediated through acti-
vation of NF-κB [33]. Moreover, ubiquitination
plays a very important role in the activation of
the NF-κB signaling pathway and participates in
the degradation of IκB protein [34]. Hence, we
Figure 5. CUL4A regulates PRMT5 to promote tumor growth (a) Photo of tumor-bearing mouse models. (b) The body weights of
mice and (c) the volumes of tumors were recorded over 28 days. (d) Photo of the dissected tumor. (e) The weights of dissected
tumors were recorded. (f) The expression levels of Ki67, PCNA, MMP12, and MMP9 in the tumor were determined using western
blotting. (g) The expression level of CUL4A, PRMT5, nuclear p65, total p65, and IκBα in the tumor was determined using western
blotting. **P < 0.01, ***P < 0.001 vs. Ov-NC;
#
P < 0.05,
##
P < 0.01,
###
P < 0.001 vs. Ov-CUL4A.
BIOENGINEERED 8721
speculated that CUL4A could regulate NPC cells
through the activation of the NF-κB signaling
pathway. The results of this study display that
CUL4A can promote the activation of NF-κB sig-
naling in NPC cells, and inhibit its activity when
its expression is knocked down. Activated NF-κB
is an integral part of inducing tumor formation,
and it has been suggested that NF-κB signaling is
involved in the malignant behavior of NPC
cells [35].
Afterward, we screened the GeneMANIA data-
base and found that CUL4A can co-express with
PRMT5. PRMT5 has been testified to be enhanced
in NPC tissues and cells, and its knockdown can
enhance the radiosensitivity [36]. We co-
transfected CUL4A overexpression plasmid and
PRMT5 interference plasmid into C666-1 cells
and injected the cells subcutaneously into nude
mice. We found that PRMT5 knockdown inhib-
ited cell migration and invasion and slowed
tumor growth in nude mice. PRMT5 is an emer-
ging epigenetic enzyme that regulates the tran-
scription of target genes through di-methylation
of histone residues [37]. At present, there are
highly selective PRMT5 inhibitors developed
with PRMT5 as the target, which are used for
the treatment of malignant solid tumors and
hematological tumors, and have been approved
for clinical trials [38]. This suggests that the use
of PRMT5 as a target for the treatment of NPC is
of great practical significance.
Conclusion
Taken together, this paper indicates that CUL4A
promotes the proliferation, migration, invasion of
NPC cells, and tumor growth by promoting
PRMT5 to activate NF-κB signaling. This finding
revealed the mechanism of CUL4A/PRMT5 in
NPC and laid a theoretical foundation for the
selection of therapeutic targets. It is hoped that
this research will advance the development of tar-
geted drugs for the treatment of NPC.
Disclosure statement
No potential conflict of interest was reported by the author(s).
Funding
Nanjing Medical University Science and Technology
Development Fund NMUB2020149
Ethics approval
All procedures followed the ethical guidelines of The
Affiliated Huai’an No. 1 People’s Hospital of Nanjing
Medical University and were approved by The Research and
Education Animal Use and Care Committee of our hospital.
Competing interests
The authors declare that they have no competing interests.
References
[1] Li H, Huang C, Chen Q, et al. Lymph-node
Epstein-Barr virus concentration in diagnosing cervical
lymph-node metastasis in nasopharyngeal carcinoma.
Eur Arch Otorhinolaryngol. . 2020Sep; 2774:
2513–2520
[2] Sun XS, Li XY, Chen QY, et al. Future of Radiotherapy
in Nasopharyngeal Carcinoma. Br J Radiol. 2019
Oct;92(1102):20190209.
[3] Holmes BJ, Wenig BM. Virus-associated carcinomas of
the head & neck: update from the 2017 WHO classifi-
cation. Ann Diagn Pathol. 2019 Feb;38:29–42.
[4] Lee HM, Okuda KS, González FE, et al. Current
Perspectives on Nasopharyngeal Carcinoma. Adv Exp
Med Biol. 2019;1164:11–34.
[5] Guan S, Wei J, Huang L, et al. Chemotherapy and
chemo-resistance in nasopharyngeal carcinoma. Eur
J Med Chem. 2020 Dec 1;207:112758.
[6] Hong M, Tang K, Qian J, et al. Immunotherapy for
EBV-Associated Nasopharyngeal Carcinoma. Crit Rev
Oncog. 2018;23(3–4):219–234.
[7] Lee AWM, Ng WT, Chan JYW, et al. Management of
locally recurrent nasopharyngeal carcinoma. Cancer
Treat Rev. 2019 Sep;79:101890.
[8] Wang X, Chen T. CUL4A regulates endometrial cancer
cell proliferation, invasion and migration by interacting
with CSN6. Mol Med Rep. 2021 Jan;23(1):23.
[9] Ashok C, Owais S, Srijyothi L, et al. A feedback regula-
tion of CREB activation through the CUL4A and ERK
signaling. Med Oncol. 2019 Jan 21 36(2):20.
[10] Cui H, Wang Q, Lei Z, et al. DTL promotes cancer
progression by PDCD4 ubiquitin-dependent
degradation. J Exp Clin Cancer Res. 2019 Aug 13 38
(1):350.
[11] Mei Q, Ye LJ, Lin H, et al. CUL4A promotes the
invasion of cervical cancer cells by regulating NF-κB
signaling pathway. Eur Rev Med Pharmacol Sci. 2020
Oct;24(20):10403–10409.
[12] Ni W, Zhang Y, Zhan Z, et al. A novel lncRNA uc.134
represses hepatocellular carcinoma progression by
8722 X. SUN ET AL.
inhibiting CUL4A-mediated ubiquitination of LATS1.
J Hematol Oncol. 2017 Apr 19 10(1):91.
[13] Hung MS, Chen YC, Lin P, et al. Cul4A Modulates
Invasion and Metastasis of Lung Cancer Through
Regulation of ANXA10. Cancers (Basel). 2019 May 2
11(5):618.
[14] Yi LJ, Yi LJ, Ding N, et al. CUL4A promotes prolifera-
tion and inhibits apoptosis of colon cancer cells via
regulating Hippo pathway. Eur Rev Med Pharmacol
Sci. 2020 Oct;24(20):10518–10525.
[15] Chen HH, Fan P, Chang SW, et al. NRIP/DCAF6
stabilizes the androgen receptor protein by displacing
DDB2 from the CUL4A-DDB1 E3 ligase complex in
prostate cancer. Oncotarget. 2017 Mar 28 8
(13):21501–21515.
[16] Jin X, Ma YC, Zhu WY, et al. CUL4A expression is
associated with tumor stage and prognosis in naso-
pharyngeal carcinoma. Medicine (Baltimore). 2019
Dec;98(51):e18036.
[17] Li M, Huang H, Cheng F, et al. miR-141-3p promotes
proliferation and metastasis of nasopharyngeal carci-
noma by targeting NME1. Adv Med Sci. 2020 Sep;65
(2):252–258.
[18] Chan KC, Ting CM, Chan PS, et al. A novel Hsp90
inhibitor AT13387 induces senescence in EBV-positive
nasopharyngeal carcinoma cells and suppresses tumor
formation. Mol Cancer. 2013 Oct 24 12(1):128.
[19] Yu X, Lin XJ, Wang S, et al. Antitumor Efficacy of
Huqizhengxiao (HQZX) Decoction Based on
Inhibition of Telomerase Activity in Nude Mice of
Hepatocarcinoma Xenograft. Integr Cancer Ther.
2018 Dec;17(4):1216–1224.
[20] Gallo-Oller G, Ordoñez R, Dotor J. A new background
subtraction method for Western blot densitometry
band quantification through image analysis software.
J Immunol Methods. 2018 Jun;457:1–5.
[21] Livak KJ, Schmittgen TD. Analysis of relative gene
expression data using real-time quantitative PCR and
the 2(-Delta Delta C(T)) Method. Methods (San Diego.
Calif). 2001 Dec;25(4):402–408.
[22] Pan S, Deng Y, Fu J, et al. TRIM52 promotes colorectal
cancer cell proliferation through the STAT3 signaling.
Cancer Cell Int. 2019;19(1):57.
[23] Sun S, Gao T, Pang B, et al. RNA binding protein NKAP
protects glioblastoma cells from ferroptosis by promoting
SLC7A11 mRNA splicing in an m(6)A-dependent
manner. Cell Death Dis. [2022 Jan 21];13(1):73.
[24] He L, Zhu C, Jia J, et al. ADSC-Exos containing
MALAT1 promotes wound healing by targeting miR-
124 through activating Wnt/β-catenin pathway. Biosci
Rep. 2020 May 29;40(5). 10.1042/BSR20192549.
[25] Wu D, Kang L, Tian J, et al. Exosomes Derived
from Bone Mesenchymal Stem Cells with the
Stimulation of Fe(3)O(4) Nanoparticles and Static
Magnetic Field Enhance Wound Healing Through
Upregulated miR-21-5p. Int J Nanomedicine.
2020;15:7979–7993.
[26] Wang C, Cheng Y, Liu H, et al. Pectolinarigenin
Suppresses the Tumor Growth in Nasopharyngeal
Carcinoma. Cell Physiol Biochem. 2016;39
(5):1795–1803.
[27] Franz M, Rodriguez H, Lopes C, et al. GeneMANIA
update 2018. Nucleic Acids Res. 2018 Jul 2 46(W1):
W60–w64.
[28] Guo R, Mao YP, Tang LL, et al. The evolution of
nasopharyngeal carcinoma staging. Br J Radiol. 2019
Oct;92(1102):20190244.
[29] Chen YP, Chan ATC, Le QT, et al. Nasopharyngeal
carcinoma. Lancet (London. England). [2019 Jul 6];394
(10192):64–80.
[30] Song Y, Xiao H, Yang Z, et al. The predictive value of
pre- and post-induction chemotherapy plasma EBV
DNA level and tumor volume for the radiosensitivity
of locally advanced nasopharyngeal carcinoma. EXCLI
J. 2017;16:1268–1275.
[31] Ji Y, Li H, Wang F, et al. PPARβ/δ Agonist GW501516
Inhibits Tumorigenicity of Undifferentiated
Nasopharyngeal Carcinoma in C666-1 Cells by
Promoting Apoptosis. Front Pharmacol. 2018;9:648.
[32] Wakae K, Kondo S, Pham HT, et al. EBV-LMP1
induces APOBEC3s and mitochondrial DNA hyper-
mutation in nasopharyngeal cancer. Cancer Med.
2020 Oct;9(20):7663–7671.
[33] Tsao SW, Tsang CM, Lo KW. Epstein-Barr virus infec-
tion and nasopharyngeal carcinoma. Philos Trans
R Soc London, Ser B. 2017 Oct 19;372:20160270.
[34] Schnappauf O, Aksentijevich I. Mendelian diseases of
dysregulated canonical NF-κB signaling: from immu-
nodeficiency to inflammation. J Leukoc Biol. 2020
Aug;108(2):573–589.
[35] Liang Y, Feng G, Wu L, et al. Caffeic acid phenethyl
ester suppressed growth and metastasis of nasopharyn-
geal carcinoma cells by inactivating the NF-κB path-
way. Drug Des Devel Ther. 2019;13:1335–1345.
[36] Yang D, Liang T, Gu Y, et al. Protein N-arginine
methyltransferase 5 promotes the tumor progression
and radioresistance of nasopharyngeal carcinoma.
Oncol Rep. 2016 Mar;35(3):1703–1710.
[37] Sloan SL, Renaldo KA, Long M, et al. Validation of
protein arginine methyltransferase 5 (PRMT5) as
a candidate therapeutic target in the spontaneous
canine model of non-Hodgkin lymphoma. PloS one.
2021;16(5):e0250839.
[38] Yuan Y, Nie H. Protein arginine methyltransferase 5:
a potential cancer therapeutic target. Cell Oncol
(Dordr). 2021 Feb;44(1):33–44.
BIOENGINEERED 8723
Article
Full-text available
Nasopharyngeal carcinoma (NPC) is one of the most common squamous cell carcinomas of the head and neck, and Epstein-Barr virus (EBV) infection is one of the pathogenic factors involved in the oncogenetic development and progression of NPC. E3 ligases, which are key members of the ubiquitin proteasome system (UPS), specifically recognize various oncogenic factors and tumor suppressors and contribute to determining their fate through ubiquitination. Several studies have demonstrated that E3 ligases are aberrantly expressed and mutated in NPC and that these changes are closely associated with the occurrence and progression of NPC. Herein, we aim to thoroughly review the specific action mechanisms by which E3 ligases participate in NPC signaling pathways and discuss their functional relationship with EBV. Moreover, we describe the current progress in and limitations for targeted therapies against E3 ligases in NPC. Key messages • E3 ubiquitin ligases, as members of the UPS system, determine the fate of their substrates and may act either as oncogenic or anti-tumorigenic factors in NPC. • Mutations or dysregulated expression of E3 ubiquitin ligases is closely related to the occurrence, development, and therapeutic sensitivity of NPC, as they play important roles in several signaling pathways affected by EBV infection. • As promising therapeutic targets, E3 ligases may open new avenues for treatment and for improving the prognosis of NPC patients.
Article
Full-text available
Ferroptosis is a form of cell death characterized by lipid peroxidation. Previous studies have reported that knockout of NF-κB activating protein (NKAP), an RNA-binding protein, increased lipid peroxidation level in naive T cells and induced cell death in colon cancer cells. However, there was no literature reported the relationship between NKAP and ferroptosis in glioblastoma cells. Notably, the mechanism of NKAP modulating ferroptosis is still unknown. Here, we found NKAP knockdown induced cell death in glioblastoma cells. Silencing NKAP increased the cell sensitivity to ferroptosis inducers both in vitro and in vivo. Exogenous overexpression of NKAP promoted cell resistance to ferroptosis inducers by positively regulating a ferroptosis defense protein, namely cystine/glutamate antiporter (SLC7A11). The regulation of SLC7A11 by NKAP can be weakened by the m ⁶ A methylation inhibitor cycloleucine and knockdown of the m ⁶ A writer METTL3. NKAP combined the “RGAC” motif which was exactly in line with the m ⁶ A motif “RGACH” (R = A/G, H = A/U/C) uncovered by the m ⁶ A-sequence. RNA Immunoprecipitation (RIP) and Co-Immunoprecipitation (Co-IP) proved the interaction between NKAP and m ⁶ A on SLC7A11 transcript. Following its binding to m ⁶ A, NKAP recruited the splicing factor proline and glutamine-rich (SFPQ) to recognize the splice site and then conducted transcription termination site (TTS) splicing event on SLC7A11 transcript and the retention of the last exon, screened by RNA-sequence and Mass Spectrometry (MS). In conclusion, NKAP acted as a new ferroptosis suppressor by binding to m ⁶ A and then promoting SLC7A11 mRNA splicing and maturation.
Article
Full-text available
Non-Hodgkin lymphoma (NHL) is a heterogeneous group of blood cancers arising in lymphoid tissues that commonly effects both humans and dogs. Protein arginine methyltransferase 5 (PRMT5), an enzyme that catalyzes the symmetric di-methylation of arginine residues, is frequently overexpressed and dysregulated in both human solid and hematologic malignancies. In human lymphoma, PRMT5 is a known driver of malignant transformation and oncogenesis, however, the expression and role of PRMT5 in canine lymphoma has not been explored. To explore canine lymphoma as a useful comparison to human lymphoma while validating PRMT5 as a rational therapeutic target in both, we characterized expression patterns of PRMT5 in canine lymphoma tissue microarrays, primary lymphoid biopsies, and canine lymphoma-derived cell lines. The inhibition of PRMT5 led to growth suppression and induction of apoptosis, while selectively decreasing global marks of symmetric dimethylarginine (SDMA) and histone H4 arginine 3 symmetric dimethylation. We performed ATAC-sequencing and gene expression microarrays with pathway enrichment analysis to characterize genome-wide changes in chromatin accessibility and whole-transcriptome changes in canine lymphoma cells lines upon PRMT5 inhibition. This work validates PRMT5 as a promising therapeutic target for canine lymphoma and supports the continued use of the spontaneously occurring canine lymphoma model for the preclinical development of PRMT5 inhibitors for the treatment of human NHL.
Article
Full-text available
Objective: The aim of this study was to investigate the effects of cullin 4A (CUL4A) on promoting the proliferation and inhibiting the apoptosis of cervical cancer (CC) cells by regulating the nuclear factor-kappa B (NF-κB) signaling pathway. Patients and methods: The protein expressions of CUL4A and NF-κB in 75 CC tissues were detected through immunohistochemistry. The correlation between the expressions of the two proteins in CC tissues was analyzed via Spearman's correlation test. Meanwhile, the prognostic significance of CUL4A expression for CC patients was analyzed by Kaplan-Meier curve. CUL4A small interfering ribonucleic acid (siRNA) was transfected into CC cells (HeLa) to downregulate the expression level of CUL4A. Subsequently, the effects of CUL4A on the proliferation and apoptosis of HeLa cells were detected by methyl thiazolyl tetrazolium (MTT) assay and flow cytometry, respectively. Finally, the effect of CUL4A on the activity of the NF-κB signaling pathway was analyzed through quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). Results: The protein expressions of CUL4A and NF-κB in CC tissues were significantly higher than those in normal tissues (p<0.01). The results of the survival curve showed that the prognosis of CC patients with highly expressed CUL4A is poor (p<0.001). Meanwhile, lowly expressed CUL4A protein significantly inhibited the proliferation and promoted the apoptosis of HeLa cells (p<0.01). QRT-PCR results indicated that the relative messenger RNA (mRNA) expression levels of downstream genes of the NF-κB signaling pathway were significantly lower in CC cells than those in the control group (p<0.001). In addition, CUL4A expression was positively correlated with NF-κB expression in CC (p<0.001). Conclusions: CUL4A promotes the invasion of CC cells through the NF-κB signaling pathway.
Article
Full-text available
Objective: The aim of this study was to investigate the effect of cullin 4A (CUL4A) on the proliferation and apoptosis of colon cancer (CC) cells, and to elucidate its regulatory relationship with the Hippo pathway. Patients and methods: Paired CC tissues and adjacent normal tissues were obtained from patients. CC cells were isolated and cultured in vitro. CUL4A was interfered by small interfering ribonucleic acid (siRNA) (siR-CUL4A group) or overexpressed by overexpression vector (CUL4A-Vector group), with negative control (NC)-CUL4A or CUL4A -NC as the control group. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the expression level of CUL4A in CC tissues and cells. The proliferative ability of cells was detected by cell counting kit-8 (CCK-8) assay. Flow cytometry was applied to measure the apoptosis of cells in each group. Western blotting (WB) was conducted to determine the protein expression of CUL4A. In addition, the proliferative ability was examined in vivo through subcutaneous injection of cells into nude mice. Results: QRT-PCR showed that CUL4A was highly expressed in 66.67% of CC samples (p<0.01). In vivo and in vitro proliferative ability was significantly reduced in siR-CUL4A group (p<0.01), whereas the apoptosis rate was promoted (p<0.01). However, in vivo and in vitro proliferative ability increased significantly in CUL4A-Vector group (p<0.01), while the apoptosis rate was reduced (p<0.01). The protein expressions of MST1, LATS1 and p-YAP were significantly up-regulated in siR-CUL4A group (p<0.01), while they were remarkably down-regulated in CUL4A-Vector group (p<0.05, p<0.01). Conclusions: CUL4A is highly expressed in CC and promotes the proliferation and inhibits the apoptosis of CC cells by regulating the Hippo pathway.
Article
Full-text available
Endometrial cancer (EC) is a common malignant gynecological tumor arising from the endometrium, with an annually increasing morbidity and mortality. The present study aimed to investigate the functions of cullin 4A (CUL4A) in EC, as well as the underlying mechanisms. CUL4A expression was assessed in several human EC cells and normal human endometrial epithelial cells (hEECs) via reverse transcription‑quantitative polymerase chain reaction and western blotting. Subsequently, short hairpin (sh)RNA‑CULA4 was transfected into cells, and cell proliferation, invasion and migration were detected using Cell Counting kit‑8, Transwell and wound healing assays, respectively. The STRING database identified that CSN6 interacted with CULA4, and immunoprecipitation was performed to verify the interaction. Subsequently, following CUL4A knockdown, pcDNA3.1‑CSN6 was transfected into cells and its effects on cell proliferation, invasion and migration were assessed. The expression levels of matrix metallopeptidase (MMP)2, MMP9 and p53 were evaluated via western blotting. The results indicated that CUL4A was highly expressed in EC cells, compared with hEECs. CULA4‑knockdown notably inhibited EC cell proliferation, invasion and migration. The expression levels of MMP2 and MMP9 were significantly decreased, while p53 expression was enhanced following CUL4A‑knockdown. The immunoprecipitation assay verified that COP9 signalosome subunit 6 (CSN6) interacted with CULA4. Furthermore, CSN6‑overexpression alleviated the inhibitory effects of CUL4A‑knockdown on EC cell proliferation, invasion and migration. Similarly, CSN6 overexpression reversed CUL4A‑knockdown‑mediated effects on the expression of MMP2, MMP9 and p53. In summary, the results demonstrated that CUL4A regulated EC cell proliferation, invasion and migration by interacting with CSN6.
Article
Full-text available
Background Both magnetic nanoparticles (MNPs) and exosomes derived from bone mesenchymal stem cells (BMSC-Exos) have been reported to improve wound healing. In this study, novel exosomes (mag-BMSC-Exos) would be fabricated from BMSCs with the stimulation of MNPs and a static magnetic field (SMF) to further enhance wound repair. Methods Mag-BMSC-Exos, namely, exosomes derived from BMSCs preconditioned with Fe3O4 nanoparticles and a SMF, together with BMSC-Exos were both first isolated by ultracentrifugation, respectively. Afterwards, we conducted in vitro experiments, including scratch wound assays, transwell assays, and tube formation assays, and established an in vivo wound healing model. The miRNA expression profiles were compared between BMSC-Exos and mag-BMSC-Exos to detect the potential mechanism of improving wound healing. At last, the function of exosomal miR-21-5p during wound healing was confirmed by utilizing a series of gain- and loss-of-function experiments in vitro. Results The optimal working magnetic condition was 50 µg/mL Fe3O4 nanoparticles combined with 100 mT SMF. In vitro, mag-BMSC-Exo administration promoted proliferation, migration and angiogenesis to a greater extent than BMSC-Exo administration. Local transplantation of mag-BMSC-Exos into rat skin wounds resulted in accelerated wound closure, narrower scar widths and enhanced angiogenesis compared with BMSC-Exo transplantation. Notably, miR-21-5p was found to be highly enriched in mag-BMSC-Exos and served as a critical mediator in mag-BMSC-Exo-induced regulatory effects through inhibition of SPRY2 and activation of the PI3K/AKT and ERK1/2 signaling pathways. Conclusion Mag-BMSC-Exos can further enhance wound healing than BMSC-Exos by improving angiogenesis and fibroblast function, and miR-21-5p upregulation in mag-BMSC-Exos might be the potential mechanism. This work offers an effective and promising protocol to improve wound healing in clinic.
Article
Full-text available
An Epstein‐Barr virus (EBV)—encoded latent membrane protein 1 (LMP1) is a principal oncogene that plays a pivotal role in EBV‐associated malignant tumors including nasopharyngeal cancer (NPC). Recent genomic landscape studies revealed that NPC also contained many genomic mutations, suggesting the role of LMP1 as a driver gene for the induction of these genomic mutations. Nonetheless, its exact mechanism has not been investigated. In this study, we report that LMP1 alters the expression profile of APOBEC3s(A3s), host deaminases that introduce consecutive C‐to‐U mutations (hypermutation). In vitro, LMP1 induces APOBEC3B (A3B) and 3F(A3F), in a nasopharyngeal cell line, AdAH. Overexpression of LMP1, A3B, or A3F induces mtDNA hypermutation, which is also detectable from NPC specimens. Expression of LMP1 and A3B in NPC was correlated with neck metastasis. These results provide evidence as to which LMP1 induces A3s and mtDNA hypermutation, and how LMP1 facilitates metastasis is also discussed. EBV LMP1 induces host cytidine deaminases, APOBECs, in nasopharyngeal cells. TES2 domain is important for LMP1‐induced LMP1 hypermutates host mitochondrial genome.
Article
BackgroundPRMT5 is a type II protein arginine methyltransferase that methylates histone or non-histone proteins. Arginine methylation by PRMT5 has been implicated in gene transcription, ribosome biogenesis, RNA transport, pre-mRNA splicing and signal transduction. High expression of PRMT5 has been observed in various cancers and PRMT5 overexpression has been reported to improve cancer cell survival, proliferation, migration and metabolism and to inhibit cancer cell apoptosis. In addition, PRMT5 has been found to be required for cancer stem cell survival, self-renewal and differentiation. Several microRNAs have been shown to regulate PRMT5 expression. As PRMT5 has oncogene-like properties, several PRMT5 inhibitors have been used to explore their efficacy as potential drugs for different types of cancer, and three of them are now being tested in clinical trials.Conclusions In this review, we summarize current knowledge on the role of PRMT5 in cancer development and progression, including its functions and underlying mechanisms. In addition, we highlight the rapid development of PRMT5 inhibitors and summarize ongoing clinical trials for cancer therapy. By affecting both tumor cells and the tumor microenvironment, PRMT5 inhibitors may serve as effective anti-cancer agents, especially when combined with immune therapies.
Article
Nasopharyngeal carcinoma (NPC) is closely associated with Epstein-Barr virus (EBV) and occurs frequently in the south of China and Southeast Asian countries. Concurrent chemo-radiotherapy is one of the main treatments for NPC. Although, the combined treatment of chemo-radiotherapy produces a satisfying survival rate, the chemo-resistance arises as a big obstacle in curing recurrent NPC patients. The acquirement of chemo-resistance is usually along with a poor prognosis. So far, the mechanism of chemo-resistance in NPC has not been fully elucidated and there have not been a comprehensive review on this issue. Thus, it is of great significance to summarize the mechanisms involved in NPC chemo-resistance. In this review, the importance and limitations of chemotherapy and the mechanisms of chemo-resistances in NPC were discussed.
Article
NF-κB is a master transcription factor that activates the expression of target genes in response to various stimulatory signals. Activated NF-κB mediates a plethora of diverse functions including innate and adaptive immune responses, inflammation, cell proliferation, and NF-κB is regulated through interactions with IκB inhibitory proteins, which are in turn regulated by the inhibitor of κB kinase (IKK) complex. Together, these 3 components form the core of the NF-κB signalosomes that have cell-specific functions which are dependent on the interactions with other signaling molecules and pathways. The activity of NF-κB pathway is also regulated by a variety of post-translational modifications including phosphorylation and ubiquitination by Lys63, Met1, and Lys48 ubiquitin chains. The physiologic role of NF-κB is best studied in the immune system due to discovery of many human diseases caused by pathogenic variants in various proteins that constitute the NF-κB pathway. These disease-causing variants can act either as gain-of-function (GoF) or loss-of-function (LoF) and depending on the function of mutated protein, can cause either immunodeficiency or systemic inflammation. Typically, pathogenic missense variants act as GoF and they lead to increased activity in the pathway. LoF variants can be inherited as recessive or dominant alleles and can cause either a decrease or an increase in pathway activity. Dominantly inherited LoF variants often result in haploinsufficiency of inhibitory proteins. Here, we review human Mendelian immunologic diseases, which results from mutations in different molecules in the canonical NF-κB pathway and surprisingly present with a continuum of clinical features including immunodeficiency, atopy, autoimmunity, and autoinflammation.