ArticlePDF Available

N6-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis via targeting miR-144-3p/Et-1 axis

Authors:

Abstract and Figures

Kupffer cells (KCs), as residents and sentinels of the liver, are involved in the formation of hepatic fibrosis (HF). However, the biological functions of circular RNAs (circRNAs) in KCs to HF have not been determined. In this study, the expression levels of circRNAs, microRNAs, and messenger RNAs (mRNAs) in KCs from a mouse model of HF mice were investigated using microarray and circRNA-Seq analyses. circDcbld2 was identified as a candidate circRNA in HF, as evidenced by its up-regulation in KCs. Silver staining and mass spectrometry showed that Wtap and Igf2bp2 bind to cirDcbld2. The suppression of circDcbld2 expression decreased the KC inflammatory response and oxidative stress and inhibited hepatic stellate cell (HSCs) activation, attenuating mouse liver fibrogenesis. Mechanistically, Wtap mediated the N⁶-methyladenosine (m6A) methylation of circDcbld2, and Igf2bp2 recognized m6A-modified circDcbld2 and increased its stability. circDcbld2 contributes to the occurrence of HF by binding miR-144-3p/Et-1 to regulate the inflammatory response and oxidative stress. These findings indicate that circDcbld2 functions via the m6A/circDcbld2/miR-144-3p/Et-1 axis and may act as a potential biomarker for HF treatment.
Content may be subject to copyright.
ORIGINAL ARTICLE
N
6
-Methyladenosine modification of circDcbld2
in Kupffer cells promotes hepatic fibrosis via
targeting miR-144-3p/Et-1 axis
Sai Zhu
a,b,y
, Xin Chen
a,y
, Lijiao Sun
a,y
, Xiaofeng Li
a,y
, Yu Chen
c
,
Liangyun Li
a
, Xiaoguo Suo
a
, Chuanhui Xu
a
, Minglu Ji
a
,
Jianan Wang
a
, Hua Wang
a
, Lei Zhang
a
, Xiaoming Meng
a
,
Cheng Huang
a
, Jun Li
a,*
a
Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs,
School of Pharmacy, Anhui Medical University, Hefei 230032, China
b
Department of Nephropathy, the First Affiliated Hospital, Anhui Medical University, Hefei 230022, China
c
Department of Pharmacy, the Second Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
Received 1 April 2024; received in revised form 22 June 2024; accepted 26 July 2024
KEY WORDS
Kupffer cells;
circDcbld2;
miR-144-3p;
Et-1;
N
6
-methyladenosine;
Wtap;
Igf2bp2;
Hepatic fibrosis
Abstract Kupffer cells (KCs), as residents and sentinels of the liver, are involved in the formation of
hepatic fibrosis (HF). However, the biological functions of circular RNAs (circRNAs) in KCs to HF have
not been determined. In this study, the expression levels of circRNAs, microRNAs, and messenger RNAs
(mRNAs) in KCs from a mouse model of HF mice were investigated using microarray and circRNA-Seq
analyses. circDcbld2 was identified as a candidate circRNA in HF, as evidenced by its up-regulation
in KCs. Silver staining and mass spectrometry showed that Wtap and Igf2bp2 bind to cirDcbld2. The
suppression of circDcbld2 expression decreased the KC inflammatory response and oxidative stress
and inhibited hepatic stellate cell (HSCs) activation, attenuating mouse liver fibrogenesis. Mechanisti-
cally, Wtap mediated the N
6
-methyladenosine (m6A) methylation of circDcbld2, and Igf2bp2 recognized
m6A-modified circDcbld2 and increased its stability. circDcbld2 contributes to the occurrence of HF
by binding miR-144-3p/Et-1 to regulate the inflammatory response and oxidative stress. These findings
indicate that circDcbld2 functions via the m6A/circDcbld2/miR-144-3p/Et-1 axis and may act as a
potential biomarker for HF treatment.
*Corresponding author.
E-mail address: lj@ahmu.edu.cn (Jun Li).
y
These authors made equal contributions to this work.
Peer review under the responsibility of Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences.
https://doi.org/10.1016/j.apsb.2024.11.003
2211-3835 ª2025 The Authors. Published by Elsevier B.V. on behalf of Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese
Academy of Medical Sciences. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Chinese Pharmaceutical Association
Institute of Materia Medica, Chinese Academy of Medical Sciences
Acta Pharmaceutica Sinica B
www.elsevier.com/locate/apsb
www.sciencedirect.com
Acta Pharmaceutica Sinica B 2025;15(1):296e313
ª2025 The Authors. Published by Elsevier B.V. on behalf of Chinese Pharmaceutical Association and
Institute of Materia Medica, Chinese Academy of Medical Sciences. This is an open access article under
the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
1. Introduction
Hepatic fibrosis (HF), a chronic liver disease, is characterized by
excessive extracellular matrix (ECM) deposition
1,2
. HF can
progress to cirrhosis or even cancer resulting in persistent liver
injury
3-5
, and is caused by various conditions, such as alcohol
abuse and non-alcoholic steatohepatitis (NASH)
6
. Macrophage
infiltration in the liver accompanies persistent liver injury
7-9
.
Interestingly, the uncontrolled secretion of inflammatory factors
and chemokines by macrophages are important driving force for
HF formation
10
. In addition, inflammation-induced changes in the
liver microenvironment lead to dysregulation of liver function,
reducing the capacity for self-repair and accelerating HF
progression
11
.
Kupffer cells (KCs) and recruited macrophages are crucial
regulators of inflammation in the organ and are considered
potential new targets for the treatment and prevention of HF
12
.
Activated macrophages synthesize and release various inflamma-
tory cytokines and chemokines that accelerate inflammatory
infiltration and collectively drive activated hepatic stellate cells
(HSCs) to transform into myofibroblasts
12,13
, while simulta-
neously producing enormous amounts of actin alpha 2, smooth
muscle, aorta (a-SMA)
14
. Therefore, it is essential to explore the
mechanisms by which macrophage activation affects HSC acti-
vation during HF formation. Recent work has shown that liver
macrophages promote HF by enhancing HSC activation in a nu-
clear factor kappa B (NF-kB)-dependent manner
15
. However, the
precise molecular mechanisms by which macrophages are
involved in HF remain largely unclear.
N
6
-methyladenosine (m6A), a common modification of
messenger RNA (mRNA)
16,17
, has recently gained attention owing
to its potential involvement in Circular RNAs (circRNA) function.
circRNAs, are small non-coding RNAs (ncRNAs) with a cova-
lently closed single-stranded loop configuration produced by
exon skipping or direct back-splicing of precursor mRNA (pre-
mRNA)
18
, exhibit different manifestation m6A modification
patterns from those of traditional mRNA
19,20
.
Recent advances in RNA sequencing technology have revealed
that most circRNAs exhibit tissue-specific expression profiles
21
.
circRNAs act as competing endogenous RNAs (ceRNAs) and
specifically adsorb microRNAs (miRNAs) to affect downstream
target gene expression or regulate gene expression at the levels of
transcription and splicing
22-24
. Differential circRNA expression is
associated with various pathological processes, including HF
25
.
circRNAs are also dysregulated in macrophages during disease
development
26,27
. Our group has studied the roles of non-coding
RNAs in the development of HF extensively
28-32
. We have
shown that circFbxw4 (F-box and WD repeat domain containing
4) affects HF progression via the miR-18b-3p/Fbxw7 (F-box and
WD repeat domain containing 7) axis in HSCs
28
. Furthermore,
circUbe2k
33
(ubiquitin conjugating enzyme E2 K), circPsd3
29
(Pleckstrin and Sec7 domain containing 3), and circMcph1
34
(Microcephalin 1) play significant roles in HF. The functions
and mechanisms of circRNAs that are differentially expressed in
macrophages during HF formation have been a focus of our
research
35
.
In this study, the expression patterns of circRNAs, miRNAs, and
mRNAs in KCs extracted from a mouse model of HF were
analyzed to explore the potential biomarkers. We identified a novel
abnormally expressed circRNA, circDcbld2, derived from the
Discoidin, CUB and LCCL domain containing 2 (Dcbld2)gene
locus. circDcbld2 expression levels were significantly increased in
HF formation and were elevated in patients with HF. The inhibition
of circDcbld2 reduced the expression of inflammatory factors in
macrophages, attenuated oxidative stress, and affected HSC acti-
vation to mitigate liver brogenesis in mice. Mechanistically, we
discovered that circDcbld2 binds to miR-144-3p to regulate
Endothelin 1 (Et-1) expression. These findings indicate that the
circDcbld2/miR-144-3p/Et-1 axis may have important functions in
macrophages and contribute to the pathogenesis of HF. Further-
more, our investigation revealed an interaction between circDcbld2
and WT1 associated protein (Wtap), a pivotal m6A writer protein,
and the stability of circDcbld2 was increased via insulin-like growth
factor 2 mRNA binding protein 2 (Igf2bp2), a pivotal m6A reader
protein. Therefore, our results support the value of circDcbld2 as a
novel potential therapeutic biomarker for HF.
2. Materials and methods
2.1. Animals and model establishment
C57BL/6J mice (6e8 weeks of age) were obtained from the
Animal Experiment Center of Anhui Medical University (Anhui,
China). Before the experiment, mice were housed in an environ-
ment with adequate food and water for one week. Euthanasia was
performed 3 days after the last injection for modeling. A set of
mice was used for the perfusion extraction of primary cells and
detection of related indicators, and the remaining mice were used
for histopathological examination of tissue following para-
formaldehyde embedding. The animal studies were approved and
reviewed by the Animal Experimentation Ethics Committee of
Anhui Medical University.
Various methods were used for HF model were establishment.
Firstly, for carbon tetrachloride (CCl
4
)-induced HF model, as
described previously
33
. Carbon tetrachloride (CCl
4
) and olive oil
(1:4, v/v) were administered to mice by intraperitoneal injection
(1 mL/kg), biweekly for 6 weeks to trigger HF. Vehicle mice were
injected with olive oil (same volume).
Secondly, the model of cholestatic HF was established through
bile duct ligation (BDL) as described previously
36
. The cholestatic
liver fibrosis model was housed in a specific pathogen-free (SPF)-
class experimental animal room at the Experimental Animal
Center of Anhui Medical University. Mice in the BDL group were
established by common bile duct ligation, operated by double
ligation using non-resorbable surgical sutures. The sham group
underwent identical procedures without ligation. After 15 days,
mice were sacrificed to observe fibrosis.
N
6
-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis 297
Thirdly, another model of HF was established through the
injection of Thioacetamide (TAA) (200 mg/kg, diluted in saline) 3
times weekly for 8 weeks
37
. Mice were sacrificed 24 h after the
last administration, and fractional liver tissues were collected for
histopathological examination.
2.2. Generation of macrophage-specific Wtap knockout mice
Wtap-cKO (Wtap-conditional knockout) (C57BL/6J background)
mice were generated in which Wtap was specifically depleted
from macrophages (global Wtap knockout is embryonically
lethal). Wtap
F/F
(Wtap
Flox/Flox
) mice were purchased from
GemPharmatech Co., Ltd. (Nanjing, China). Wtap
F/F;Lyz-Creþ
(Wtap-cKO) mice were generated by mating Wtap
F/F
mice with
macrophage-specific promoter (Lysozyme 2)-driven Cre (Lyz-Cre)
mice.
2.3. circRNA expression analysis
2.3.1. RNA extraction and quality control
Primary macrophages were extracted from the vehicle and CCl
4
-
treated mice. Each group of samples contained primary macro-
phages from six mice for sequencing. Total RNAs of KCs were
extracted using the MIRNeasy Mini Kit (Qiagen, Hilden, Ger-
many). Then, the RNase-Free DNase Set and RNA Clean XP Kit
(Beckman Coulter, Brea, CA, USA) were used to purify the
extracted RNA. Quantitative detection was performed using the
NanoDrop 2000 Spectrophotometer (Thermo Scientific, Waltham,
MA, USA) and Agilent Bioanalyzer 2100 (Agilent Technologies,
Santa Clara, CA, USA).
2.3.2. Library preparation and high-throughput sequencing
Following the manufacturer’s instructions, RNA-seq libraries were
constructed using the TruSeqStranded Total RNA Sample
Preparation Kit (Illumina, USA). Subsequently, the Qubit2.0
Fluorometer (Life Technologies, USA) and Agilent 2100 Bio-
analyzer (Agilent Technologies, USA) were used for quantifica-
tion and validation. The library was diluted to 10 pmol/L and
sequenced using the Illumina HiSeq 2500 system (Illumina,
USA). Sequencing and library validation and construction were
performed by Origin Biotech (Shanghai, China).
2.3.3. Data analysis
Quality control for RNA-Seq reads was performed using FastQC1
(v.0.11.3). Ribosomal RNA reads, poor reads and Illumina TruSeq
adapter sequences were trimmed using seqtk. Then, BWA-MEM
(v.2.0.4) was used to map mouse reference genes and trimmed
reads. circRNA sequencing data were analyzed using CIRI, and
matched against the circBase database. The counts were normal-
ized to SRPBM. The thresholds for differential expression were
adjusted P-value <0.05 and |log
2
(fold change)| 1.0. All of the
dysregulated genes were evaluated using a heat map and KEGG
pathway enrichment analyses.
2.4. RNA sequencing and functional enrichment analysis
RNA samples were isolated and purified using TRIzol (Thermo
Fisher Scientific, USA) according to the manufacturer’s protocol.
PolyA mRNA was specifically captured through two rounds of
purification using oligo (dT) magnetic beads (Thermo Fisher,
USA). The fragmented RNA was subjected to reverse transcription
to synthesize cDNA (Invitrogen SuperScriptII Reverse
Transcriptase, Cat. 1896649, CA, USA). These compound DNA
and RNA duplexes were then converted to DNA duplexes by
double-strand synthesis using E. coli DNA polymerase I (NEB,
USA) and RNase H (NEB, USA). The two strands were digested
with UDG enzymes (NEB, cat.m0280, MA, US) to form a library
with a fragment size of 300 bp 50 bp (strand-specific library).
Then, the Illumina NovaSeq6000 platform (LC Bio Technol-
ogy Co., Ltd., Hangzhou, China) was used for paired-end
sequencing following the standard operation in PE150 mode.
Differentially expressed mRNAs were identified using adjusted
P-value <0.05 and |log
2
(fold change)| 1.0 as thresholds. Gene
functions were evaluated using Kyoto Encyclopedia of Genes and
Genomes (KEGG) and Gene Ontology (GO) functional enrich-
ment analyses.
2.5. Microarray analysis
According to the instructions provided, the Affymetrix
GeneChipmiRNA 4.0 Array (Affymetrix, USA) and FlashTag
Biotin HSR Labeling Kit (Affymetrix, USA) were used. The
GeneChip Hybridization Wash and Stain Kit (Thermo Fisher
Scientific, USA) was used to dye the miRNA array for imaging.
Differentially expressed miRNAs were identified using |log
2
(fold
change)| 1.0), P-value <0.05, and volcano plot filtering. The
targets of miRNAs were obtained using two databases TargetScan
and miRanda.
2.6. ceRNA network (ceRNET) snalysis
A ceRNET (circRNAsemiRNAsemRNAs) was generated based
on dysregulated circRNAs, miRNAs, and mRNAs. The interaction
ceRNET was established using Cytoscape.
2.7. Human samples
Human liver samples were obtained from the First Affiliated
Hospital of Anhui Medical University (Anhui, China). This study
was approved by the Biomedical Ethics Committee of Anhui
Medical University. The volunteers were undertaken with the
understanding and written consent. All protocols adhered to the
principles outlined in the Declaration of Helsinki. Characteristics
of participants are shown in Supporting Information Table S1.
2.8. circDcbld2 suppression in mice
pHBAAV-circDcbld2 (circDcbld2-KD) and control were designed
and synthesized by Hanbio Biotechnology (Hanbio Biotech-
nology, China). Mice were injected with circDcbld2-KD and the
vector at 1 10
12
vg/mL into the tail vein. After a week of
observation, the model was established by circDcbld2-KD
administration. The transfection efficiency for circDcbld2 was
measured by qRT-PCR in primary macrophages.
2.9. Flow cytometry analysis
Cells were incubated with anti-Cd11b-FITC (BD Biosciences,
USA) and anti-F4/80-PE (BD Biosciences, USA) antibodies.
Next, the CytoFLEX flow cytometer (Beckman Coulter, USA)
was used to detect compounds. Cd11b
þ
and F4/80
þ
cells were
considered macrophages for subsequent experiments, and
CytExpert software (Beckman Coulter, USA) was used to analyze
data.
298 Sai Zhu et al.
2.10. Histology and immunohistochemistry
Paraffin-embedded and 4% paraformaldehyde-fixed liver tissues
(4 mm) were used for immunofluorescence (IF) and immunohis-
tochemical (IHC) staining of Et-1 (Abcam, USA), F4/80 (Bioss,
bsm-34028M), and a-SMA (Abcam, USA). Liver pathology was
evaluated by Sirius red and H&E staining. Sections were scanned
using a digital slide scanner (Pannoramic MIDI, 3DHISTECH,
Hungary).
2.11. Examination of oxidative stress markers
Commercial assay kits for the detection of Tatalase (CAT; #A007-
1-1), glutathione (GSH; #A006-2-1), malondialdehyde (MDA;
#A003-1-3), and superoxide dismutase (SOD; #A001-3-2) were
obtained from Nanjing Jiancheng Bioengineering Institute
(Nanjing, China). Oxidative stress damage of ROS in liver tissue
for the HF model was examined by ROS assay kit (Bestbio, #BB-
470534) following the manufacturer’s protocols.
2.12. DNA sequencing
RNA was reverse-transcribed into cDNA by using
PrimeScript
TM
RT Master Mix (Takara, Japan). Polymerase chain
reaction (PCR) was performed using 2 Taq Master Mix (Takara,
Japan) following the manufacturer’s instructions. The PCR prod-
ucts were identified by using DNA sequencing (ABI3730XL,
USA).
2.13. Pull-down assay
A biotinylated circDcbld2 probe was designed for binding to the
junction site of circDcbld2. Bone marrow-derived macrophages
(BMDMs) were washed with PBS, and the cell lysate was incu-
bated with 3 mg of biotinylated probe for 4 h. Lysates were
incubated with RNase-free BSA (10 mg/mL) and Pierce
Streptavidin Magnetic Beads (Thermo Fisher Scientific, USA) at
4C (3 h) to decreasing nonspecific binding. RNA complexes
were rotated with probe-bead complexes at 4 C overnight. After
washing the beads three times, relative protein levels were
analyzed through Western blotting.
2.14. Fluorescence in situ hybridization (FISH)
In situ hybridization probes, i.e., FAM-labelled circDcbld2 and
FAM-labelled miR-144-3p probes, were established by
GenePharma (Shanghai, China). Briefly, cells were washed three
times with sterile PBS and fixed with 4% paraformaldehyde at
room temperature. A FISH Kit (GenePharma, China) was
employed for protein detection according to the manufacturer’s
instructions, with hybridization at 37 C overnight in a dark moist
chamber. DAPI was used to stain nuclei. Images were collected by
an inverted fluorescence microscope (Leica, Japan).
2.15. Isolation of bone marrow-derived macrophages, Kupffer
cells, hepatocytes, and hepatic stellate cells
Previously described methods were used to isolate KCs
38
, hepa-
tocytes, HSCs
28
, and BMDM
39
.In situ perfusion was performed
using collagenase followed by differential centrifugation accord-
ing to a density gradient. A catheter was inserted in the liver portal
vein of the mouse and dissect the inferior vena cava and perfusion.
The digested liver was passed through a sieve (200-mesh). Then,
25% and 50% Percoll were used to separate macrophages. The
isolated cells were resuspended in Dulbecco’s modified Eagle’s
medium (DMEM) with 10% fetal bovine serum (FBS). Non-
adherent cells and culture medium were discarded after the liver
macrophages adhered to the flask surface for 40 min. Macro-
phages were identified by using flow cytometry
40
. Hepatocytes
were isolated from the mouse liver by in situ collagenase perfu-
sion. The liver was perfused with HBSS (2%FBS) via the portal
vein, followed by 0.27% collagenase IV. Perfused livers were
dissected and teased through 70 mm nylon mesh cell strainers.
Hepatocytes were collected following centrifugation at 50 g
(2 min three times). Primary HSCs were isolated through two-step
collagenase (Sigma)epronase (Sigma) perfusion of mouse livers.
Then, OptiPrep (Axis Shield, Norway) was used to for density
gradient centrifugation (11.5% and 20%)
41
. Furthermore, tibias
and femurs were isolated from mice via cutting at the knee joint.
Bone marrows were collected and cultured for 7 days and termi-
nally differentiated into macrophages.
2.16. siRNA-circDcbld2 and over-expression circDcbld2
plasmid transfection
Small interfering RNAs (siRNAs) and an over-expression plasmid
for circDcbld2 were obtained from Hanheng Biotechnology
(Shanghai, China). BMDMs were transfected with siRNA-
circDcbld2 with Lipofectamine 2000 (Invitrogen, USA). The
culture medium was replaced, after 6 h, followed by incubation for
an additional 24 h. The transfection efficiency of circDcbld2 was
detected using qRT-PCR. Sequences are shown in Supporting
Information Table S2.
2.17. miR-144-3p mimics and inhibitor transfection
miR-144-3p mimics and the inhibitor were structured in Hanheng
Biotechnology (Shanghai, China). The transfection methods were
consistent with those for circDcbld2. A qRT-PCR analysis was
used to measure the transfection efficiency of the miR-144-3p
mimics and inhibitor. Related sequences are shown in Supporting
Information Table S3.
2.18. RNA immunoprecipitation (RIP)
The Magna RIP RNA-Binding Protein Immunoprecipitation Kit
(Millipore Sigma, 17-701) was used for an RIP analysis following
the manufacturer’s protocols. Magnetic beads precoated with
Igf2bp2 or IgG (Millipore) were incubated with the cell suspen-
sion at 4 C overnight. Proteinase K was used to treat
RNAeprotein complexes to remove protein impurities. Finally,
RNA was purified using TRIzol and detected using qRT-PCR.
2.19. m6A MeRIP-qRT-PCR
RNA samples were collected in Wtap-cKO or Wtap
F/F
BMDMs
under lipopolysaccharide (LPS) stimulation. The RNA was soni-
cated to obtain 100e150 nt fragments, which were incubated with
m6A antibody to analyze m6A enrichment using qRT-PCR.
Briefly, fragmented RNA was combined with the m6A antibody
in RIP immunoprecipitation buffer and incubated at 4 C over-
night, followed by incubation in proteinase K buffer at 55 C for
30 min. TRIzol was used to extract RNA, and m6A immunopur-
ification of mRNA was detected using qRT-PCR.
N
6
-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis 299
2.20. Dot blot
Extracted RNA was boiled in a metal bath at 90 C for 5 min and
cooled immediately. The RNA sample point on the nylon mem-
brane (UV lamp for 2 h) was blocked with 5% skim milk (2 h) and
incubated with the m6A antibody overnight. The nylon membrane
was washed with TBST (Tris-buffered saline containing 0.1%
Tween 20) three times on the next day. The secondary antibody
was applied, followed by detection using the Amersham Imager
600 System. Methylene blue was used to stain the membranes.
2.21. RNA extraction and qRT-PCR analysis
Total RNA was extracted with TRIzol from cells or tissues. The
NanoDrop 2000 Spectrophotometer was employed to detect the
purity and concentration of extracted RNA. PrimeScripRT
Master Mix was used to synthesize cDNA. Gapdh and b-actin
expression were used as internal controls. Primer sequences are
listed in Supporting Information Tables S4eS6.
2.22. Nuclear and cytoplasmic fractionation analysis
The Cytoplasmic & Nuclear RNA Purification Kit (#BB-36021,
BestBio) was used for a nuclear and cytoplasmic fractionation
analysis. The nuclear and cytoplasmic components were extracted
through centrifugation (1200 gfor 5 min). Then, each fraction
was collected separately.
2.23. Actinomycin D (Act-D) and RNase R treatment
Linear RNA was removed with RNase R (Epicentre Technologies,
USA). The samples (RNase Rþand RNase Re) were supple-
mented with reaction buffer (10 , 0.6 mL) and DEPC-water
(0.2 mL) for 20 min at 37 C. circRNA and mRNA exposed to
RNase R were detected with CFX96 qRT-PCR (Bio-Rad, CA).
BMDM transcription was blocked with Actinomycin D (4, 8, 16,
and 24 h) (2 mg/mL, Sigma, USA). qRT-PCR was used to evaluate
the stability of circDcbld2 and the mRNA level of linear Dcbld2.
2.24. Luciferase reporter assay
circDcbld2 sequences with miR-144-3p target sites were synthe-
sized and cloned into the pSI-Check2 reporter vector downstream
of firefly luciferase (circDcbld2-wild type and circDcbld2-mutant)
by Hanheng Biotechnology (Shanghai, China), respectively. The
reporter vector, miR-144-3p mimics, or negative control were
transfected in HEK-293T cells using Lipofectamine 3000 (Invi-
trogen, CA). Activity levels of firefly and Renilla luciferase were
measured using a Dual-Luciferase system (Promega, USA) ac-
cording to the manufacturer’s protocol and detected by GloMax
Multi Jr (Promega, USA).
2.25. Western blotting
Whole-protein samples were transferred to a PVDF membrane
(Millipore, USA). The antibodies used in the analysis were as
follows: b-actin (1:500, Sino Biological lnc., 100166-MM10),
Wtap (1:500; Cell Signaling Technology, #56501), Igf2bp2
(1:500; Abcam, ab124930). a-SMA (1:500; Abcam, ab7817),
Col1a1(1:500; Bioss, bs-0578R). Then, membranes were
incubated with secondary antibodies (HRP-coupled) (1:1000,
ZSGB-Bio, China) for 60 min at room temperature. Finally, pro-
tein signals were analyzed using an enhanced chemiluminescence
(ECL) system (Bio-Rad, USA).
2.26. Statistical analysis
Data are shown as the mean standard error of mean (SEM)
and were analyzed by one-way analysis of variance (ANOVA),
followed by NewmaneKeuls post hoc tests implemented in Prism
8.0 (GraphPad Software, USA). Correlations in expression
levels were evaluated using Pearson’s correlation coefficients.
Pvalue <0.05 were considered statistically significant.
3. Results
3.1. circDcbld2 up-regulated in mouse KCs by high-throughput
sequencing
The secretion of inflammatory factors and chemokines from KCs
is critical during HF
42
. To identify candidate circRNAs involved
in HF, primary KCs (isolated from vehicle or HF mice) were
examined by circRNA high-throughput sequencing. The progres-
sion of HF formation is shown in Supporting Information
Fig. S1A. We first successfully established BDL, CCl
4
,
and TAA-induced HF mouse models and confirmed injury and
pathological characteristics (Fig. 1A and Fig. S1BeS1D). In-
flammatory factors were increased in KCs (Fig. S1E) and tissues
(Fig. S1F) from HF mice than from control mice. The serological
indicator alanine/aspartate-transaminase (ALT/AST) in
CCl
4
-induced HF mice is summarized in Fig. S1G. These results
indicate successful model construction.
We detected differential expression of 3138 circRNAs between
KCs extracted from HF mice and control mice, of which 457
circRNAs were recorded in circBase. The circRNAs with
log
2
FC 1.0 were selected for subsequent analyses. Further
analysis indicated 99 circRNAs were expressed differently in
HF (Supporting Information Files S1 and S2), including 39
up-regulated and 60 down-regulated circRNAs (Supporting
Information File S3). These dysregulated circRNAs, after valida-
tion, may be related to the development of HF. The relative
expression levels of circRNAs are shown in Supporting
Information Fig. S2A and the circRNA gene distribution is
shown in Fig. S2B. Back-splicing from host genes is considered
the source of most circRNAs
18
. The biological characteristics
(including length, localization, host genes, and exons/introns) of
the circRNAs were evaluated. We selected 13 circRNAs for model
validation (Fig. S2C). Based on validation and expression
intensity results, we selected circDcbld2 for further analysis.
Notably, we found that circDcbld2 was up-regulated in HF mice
induced by CCl
4
, TAA, and BDL (Fig. 1B). We also detected
circDcbld2 expression in BMDMs (Fig. 1C), KCs (Fig. 1D),
HSCs, and hepatocytes, extracted from CCl
4
-induced HF mice.
Levels of circDcbld2 were significantly elevated in KCs
and BMDMs in HF, and did not differ significantly in HSCs and
hepatocytes (Fig. 1E). To explore the expression changes of
circDcbld2 (hsa_circ_0066631) in human liver fibrosis, we
extracted RNA from human liver tissues for detection, revealing
that circDcbld2 as up-regulated (Fig. 1FeG and Fig. S2D). These
findings indicate that the expression pattern of circDcbld2 corre-
sponds with the pathology of HF; accordingly, circDcbld2 is a
potential biomarker during HF formation.
300 Sai Zhu et al.
Figure 1 circDcbld2 up-regulated in mouse KCs by high-throughput sequencing. (A) Pathology observation stained with Sirius red staining
were performed in CCl
4
-induced, BDL-induced, TAA-induced and vehicle mouse liver tissues sections. Scale bar Z100 mm. (B) The up-
regulated of circDcbld2 in HF mice induced by CCl
4
, TAA and BDL (nZ6). (C) The process of BMDM extraction. (D) KCs were identi-
fied by flow cytometry. (E) The expression of circDcbld2 in KCs, BMDM, HSCs and Hepatocytes (nZ6). (F) Pathology observation of human
fibrotic liver tissues sections stained with H&E and IHC of a-SMA. (G) circDcbld2 expressed in human fibrotic liver tissues. The bar shows the
mean SEM. *P<0.05, ***P<0.001 vs vehicle group.
N
6
-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis 301
3.2. Characterization of circDcbld2
circDcbld2 (mmu_circ_0000693) is derived from the host gene
Dcbld2 located on chromosome 16 (58424673e58433576)
(366 nt). Genomic structure and back-splicing point analyses
revealed that circDcbld2 consists of two exons from the Dcbld2
(exons 2e3) (Fig. 2A). Furthermore, the qRT-PCR product of
circDcbld2 verified that the circRNA matched the circDcbld2
sequence from circBase and showed head-to-tail splicing. Using
convergent and divergent primers designed for circDcbld2, and
genomic DNA (gDNA) and complementary DNA (cDNA) as
templates, 1% agarose gel electrophoresis revealed a distinct,
single product of circDcbld2 using divergent primers from cDNA
only, while no product was obtained from gDNA (Fig. 2B). To
verify the circRNA characteristics of circDcbld2, we confirmed
resistance to RNase R digestion (Fig. 2C). Furthermore, an Acti-
nomycin D (Act D) analysis showed that circDcbld2 had greater
stability than that of the linear Dcbld2 transcript in KCs (Fig. 2D).
We also treated circDcbld2 and linear Dcbld2 with RNase R,
revealing that circDcbld2 was resistant to RNase R digestion
(Fig. 2E). FISH and cytoplasmic and nuclear fractionation assays,
with U6 and Gapdh as controls, revealed that circDcbld2 localized
to both the nucleus and cytoplasm (Fig. 2F and G).
3.3. Wtap mediates circDcbld2 m6A modification and increases
the stability via Igf2bp2
Cytoplasmic circRNAs act as miRNA sponges or as RNA-binding
proteins (RBPs), while nuclear circRNAs mainly interact with
RBPs
43
. Given the location of circDcbld2, RNA pull-down and
mass spectrometry (MS) were employed to explore its functions
(Fig. 3A). We detected m6A-related proteins with protein
molecular weights of 40e70 kDa and found two proteins, WTAP
and IGF2BP2, were consistently pulled down by the biotin-labeled
circDcbld2 antisense probe (Fig. 3B and C). The pull-down con-
centration and silver staining of the polypeptide sites of Wtap and
Igf2bp2 are shown in Fig. 3D and E, and Supporting Information
Fig. S3A and S3B. We then constructed Wtap
F/F;Lyz-Creþ
(Wtap-
cKO) mice and confirmed that the knockout efficiency through
Western blotting (Fig. 3F and Fig. S3C). Colorimetric and dot blot
assays indicated the m6A methylation level was lower in Wtap-
cKO mice than in Wtap
F/F
(Fig. S3D and S3E). We predicted a
credible methylation site of circDcbld2 using SRAMP (http://
www.cuilab.cn/sramp)(Fig. S3F). Luciferase reporter gene con-
structs with wild-type or mutant circDcbld2 further verified the
m6A modification of circDcbld2 by Wtap (Fig. S3G). The lucif-
erase reporter assay indicated that the level of wild-type
circDcbld2 decreased significantly after Wtap-cKO administra-
tion but not in response to mutant circDcbld2 (Fig. S3H).
Furthermore, circDcbld2 levels were significantly decreased in
Wtap-cKO than in Wtap
F/F
(Fig. 3G). MeRIP-qPCR results proved
that circDcbld2 was enriched for m6A modifications in the LPS-
stimulated Wtap
F/F
group, while this enrichment was decreased
in the LPS-stimulated Wtap-cKO group (Fig. 3H). These results
demonstrated that the Wtap-related m6A modification regulated
circDcbld2 expression. Recently, Igf2bp2 has been identified as an
m6A-related “reader” protein involved in the stability of down-
stream genes by recognizing m6A modifications
44
. Silver staining
and mass spectrometry revealed enrichment for the stability-
related protein Igf2bp2; therefore, we further evaluated whether
m6A modification affects circDcbld2 stability. The results showed
that Wtap-cKO significantly reduced the stability of circDcbld2, as
determined using Act D treatment (Fig. 3I). An RIP analysis
further proved that circDcbld2 and Igf2bp2 interact in LPS-
stimulated BMDMs (Fig. 3J). RNA stability analysis indicated
that Igf2bp2 inhibition reduced circDcbld2 stability following Act
D treatment (Fig. 3K), and shortened the RNA half-life of
circDcbld2 (Fig. S3I), indicating that Igf2bp2 participated in the
maintenance of circDcbld2 stability. Furthermore, RIP-qPCR
analysis showed that an Igf2bp2-specific antibody resulted in
significantly greater circDcbld2 enrichment in the Wtap
F/F
group
than in the IgG control, and this increase was obviously reduced in
the Wtap-cKO group (Fig. S3J). Therefore, the increase in
circDcbld2 expression in the model group may be due to the
modified and identified of circDcbld2 m6A site by Wtap and
Igf2bp2 to enhance the stability of circDcbld2, this clarifying the
mechanism underlying circDcbld2 up-regulation in liver fibrosis.
3.4. circDcbld2 increases inflammation and oxidative stress of
BMDMs
To analyze the function of circDcbld2 in BMDMs, we constructed
siRNA of circDcbld2 (Fig. 4A). After analyzing the efficiency of
siRNA-circDcbld2 transfection in BMDMs (Fig. 4B), we per-
formed RNA-seq (Supporting Information Fig. S4A). Genes
associated with KEGG pathways (TNF, HIF-1, and FoxO
signaling pathways), and GO terms (inflammatory response,
oxidoreductase activity) were affected by circDcbld2 silencing
(Fig. 4C and D). In addition, circDcbld2 suppression decreased
the secretion of inflammatory factors and chemokines such as
IL-1b,Tnf-a, and Mcp-1, as determined by ELISA (Fig. 4E). We
further found that circDcbld2 silencing reduced the mRNA levels
of oxidative stress markers, such as Nox1,Nox2,Nox4, and
p22
phox
(Fig. 4F). Moreover, while SOD and GSH were decreased
in LPS-stimulated BMDMs compared with controls, these de-
creases were attenuated by circDcbld2 knockdown (Fig. S4B and
S4C). In addition, the over-expression of circDcbld2 had the
opposite effects compared with those for circDcbld2 silencing
(Fig. 4G and H, Fig. S4D and S4E). Taken together, these results
show that circDcbld2 silencing effectively reduces LPS-induced
inflammation and oxidative stress in BMDM.
3.5. Pro-fibrogenic activities and pro-oxidative activities effects
of circDcbld2 in HF mice
To further explore the influence of circDcbld2 on HF, circDcbld2-
KD was injected into HF mice through the tail vein. We verified
the efficiency of circDcbld2 inhibiting in KCs (Supporting
Information Fig. S5A). Functionally, the degrees of liver
collagen deposition and parenchymal distortion were reduced,
vascular architecture was altered, and F4/80
þ
macrophagocytes
and a-SMA
þ
myofibroblasts were reduced in HF following
circDcbld2-KD treatment (Fig. 5A and Fig. S5B). We further
evaluated HF-related damage indicators, Timp-1,a-SMA, Col1a1,
and Tgf-b1, in liver tissues and the inflammation markers IL-1b,
Tnf-a, and Mcp-1 in KCs (Fig. S5C). The ALT/AST indicator in
CCl
4
-induced HF mice decreased following circDcbld2-KD
administration (Fig. S5D). circDcbld2-KD administration
decreased the levels of inflammatory factors and chemokines
(IL-1b,Tnf-a,and Mcp-1), as analyzed by ELISA (Fig. S5E).
Immunofluorescence staining showed that fibrogenic factors
(Col1a1), and macrophage factors (iNos and F4/80) were
302 Sai Zhu et al.
Figure 2 Characterization of circDcbld2. (A) The genomic structure and backsplicing point of circDcbld2. (B) Divergent primers amplified
circDcbld2 from cDNA by PCR and an agarose gel electrophoresis, rather than from gDNA, Gapdh was used as a linear control. (C) circDcbld2
from cDNA was analyzed by divergent primers even exposed to Rnase R digestion, the opposite result showed from gDNA. (D) Actinomycin D
was added to detected the circDcbld2 and linear Dcbld2 expression in KCs at the indicated time points. (E) circDcbld2 resisted to Rnase R
digestion (nZ6). (F, G) Fluorescence in situ hybridization and Cytoplasmic and nuclear fractionation assay revealed that circDcbld2 localized to
both the nucleus and cytoplasm. Scale bar Z20 mm. The bar shows the mean SEM. ns, no significance; **P<0.01, ***P<0.001 vs Linear
Dcbld2 group (D); Rnasegroup (E) and Nucleus group (F).
N
6
-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis 303
Figure 3 Wtap mediates circDcbld2 m6A modification and increases the stability via Igf2bp2. (A, B) The process of RNA pull-down and MS.
(C) Western blot analysis of WTAP and IGF2BP2. (D, E) The polypeptide sites of Wtap and Igf2bp2 in pull-down concentration and silver
staining. (F) The Western blot analysis of knockout efficiency of WTAP. (G) Relative expression of circDcbld2 decreased in Wtap-cKO group. (H)
Wtap-mediated circDcbld2 m6A modifications was detected with MeRIP-qPCR analysis. The m6A modification of circDcbld2 was decreased
following Wtap-cKO (nZ4). (I) The stability of circDcbld2 was weaken by Wtap-cKO. (J) qRT-PCR analysis of RIP in LPS-stimulated BMDMs
indicated the binding of Igf2bp2 protein and circDcbld2 (nZ4). (K) The stability of circDcbld2 was decreased following Igf2bp2 administration.
The data represent the mean SEM. *P<0.05, **P<0.01, vs Wtap
F/F
group (G), m6A group in Wtap
F/F
(H), LPS þWtap
F/F
group (I), Control
group (J) and LPS þsiRNA-NC-Igf2bp2 group (K).
304 Sai Zhu et al.
Figure 4 circDcbld2 increases inflammation and oxidative stress of BMDMs. (A) A siRNA target site of circDcbld2 was constructed. (B)
Silencing efficiency of siRNA-circDcbld2 in BMDMs following transfection. (C, D) GO and KEGG enrichment analysis in BMDMs following
circDcbld2 knock-down. (E) circDcbld2 suppression decreased the release of IL-1b,Tnf-a, and Mcp-1 by ELISA (nZ3). (F) mRNA expression
of Nox1,Nox2,Nox4, and p22
phox
were reduced by circDcbld2 administration (nZ3). (G) circDcbld2 over-expression enhanced the release of
IL-1b,Tnf-a, and Mcp-1by ELISA (nZ3). (H) mRNA expression of Nox1,Nox2,Nox4, and p22
phox
were increased following circDcbld2 over-
expression (nZ3). The data represent the mean SEM. *P<0.05, ***P<0.001 vs siRNA-NC group (B) and control group (E, F, G, H);
#
P<0.05,
##
P<0.01,
###
P<0.001 vs LPS þsiRNA-NC group (E, F) and LPS þOE-NC group (G, H).
N
6
-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis 305
increased in the HF model and decreased following circDcbld2-
KD administration (Supporting Information Fig. S6A). NAD(P)
H oxidase subunits (Nox1,Nox2,Nox4, and p22
phox
) increased in
primary KCs from HF mice, and these increases were mitigated by
circDcbld2-KD treatment (Fig. S6B). These results suggest that
circDcbld2-KD could effectively reduce inflammatory injury and
fibrosis in HF mice. Moreover, while CCl
4
induction increased
oxidative stress hallmarks significantly (MDA), CCl
4
-induced in-
creases in expression were attenuated by circDcbld2-KD treat-
ment. Levels of antioxidants, such as CAT, GSH, and SOD, were
Figure 5 Pro-fibrogenic activities and pro-oxidative activities effects of circDcbld2 in HF mice. (A) Pathology observation of H&E staining
and Sirius red staining in HF with circDcbld2-KD, IHC stain of a-SMA and F4/80. Representative images were presented, scale bar, 100 mm.
Quantification of Sirius red and a-SMA (nZ6). (B) IF staining confirmed that the level of oxidative stress activation (ROS) was highly in HF
mice and decreased following circDcbld2-KD (nZ6). Representative images were presented, scale bar Z50 mm. The data represent the
mean SEM. ***P<0.001 vs vehicle group;
###
P<0.001 vs CCl
4
-induced liver fibrosis group.
306 Sai Zhu et al.
restored in CCl
4
-induced HF mice after circDcbld2-KD adminis-
tration (Fig. S6C). Immunofluorescence staining confirmed that
ROS levels were high in CCl
4
-induced HF mice and were reduced
in circDcbld2-KD mice (Fig. 5B). These results show that
circDcbld2 affects hepatic oxidative stress and inflammatory
infiltration in HF model mice.
3.6. Silencing circDcbld2 reduces HSC activation in HF mice
HSCs, which are the primary source of mature myofibroblasts,
play a crucial role in HF formation. To explore the influence of
inflammatory factors and chemokines secreted by activated mac-
rophages on HSCs, we co-cultured primary HSCs with the
BMDM supernatant, transfected with circDcbld2-siRNA
(Fig. 6A). We detected a-SMA, Col1a1,Timp-1, and Tgf-b1
mRNA expression (Fig. 6B) and analyzed the Col1a1 and a-SMA
protein levels in HSCs (Fig. 6C). Silencing circDcbld2 in BMDMs
could effectively reduce HSC activation. Furthermore, Immuno-
fluorescence staining showed that macrophage factors (F4/80) and
HSC activation factors (a-SMA) were elevated in the HF model
but reduced following circDcbld2-KD administration in vivo
(Fig. 6D). Finally, we extracted primary HSCs from HF mice and
explored the expression levels of the HSC activation factor
a-SMA and fibrogenic factor Col1a1following circDcbld2-KD
administration. Silencing circDcbld2 through circDcbld2-KD
administration effectively reduced the expression of a-SMA and
Col1a1 in primary HSCs (Fig. 6E). Furthermore, the a-SMA,
Col1a1,Timp-1, and Tgf-b1mRNA levels were decreased
following circDcbld2-KD treatment (Fig. 6F). Taken together,
silencing circDcbld2 alleviated the secretion of inflammatory
factors and chemokines in macrophages, reduced HSC activation,
inhibited the differentiation of activated HSCs into myofibro-
blasts, decreased collagen deposition, and attenuated CCl
4
-
induced HF injury.
3.7. Microarray analysis and identification of circDcbld2/miR-
144-3p interaction
circRNAs commonly function as miRNA sponges. To identify
potential miRNAs that bind circDcbld2, we performed a miRNA
microarray analysis using primary KCs. The results revealed 138
dysregulated miRNAs in HF samples; the abnormal miRNAs were
shown in the heatmap (Supporting Information Fig. S7A,
Supporting Information Files S4 and S5). A circRNA-miRNA
network was constructed based on the dysregulated miRNAs
and circRNAs (Fig. S7B). These results were combined with
target prediction tools (TargetScan) and a microarray analysis to
evaluate potential miRNAs targets of circDcbld2 and to identify
potential miRNAs related to HF (Fig. S7C). We detected eight
miRNAs that may interact with circDcbld2 (Fig. S7D). Sequence
pairing and expression analysis further revealed that circDcbld2
potentially interacts with miR-144-3p (Fig. 7A). Subsequently,
circDcbld2 fragments with the wild-type (WT) sequence or mu-
tations in putative binding sites as well as negative control and
miRNA mimics were used to explore circDcbld2 and miR-144-3p
binding (Fig. 7A). circDcbld2-WT luciferase reporter activity was
suppressed in the presence of miR-144-3p (Fig. 7B). Furthermore,
miR-144-3p was predominantly localized in the cytoplasm in a
FISH analysis (Fig. 7C). miR-144-3p expression, as determined
by qRT-PCR, was lower in LPS-stimulated BMDMs than in
controls, consistent with the results of the microarray analysis
(Fig. 7D). Interestingly, miR-144-3p was increased in BMDMs
following siRNA-circDcbld2 administration compared with the
LPS þsiRNA-NC group (Fig. 7E). Furthermore, transfection with
miR-144-3p mimics effectively reduced the expression levels of
inflammatory factors (IL-1b,Tnf-a, and Mcp-1) and oxidative
stress markers (Nox1,Nox2,Nox4, and p22
phox
)(Supporting
Information Fig. S8A). miR-144-3p mimics could effectively
increase levels of indicators of oxidative stress (SOD and GSH)
(Fig. S8B). miR-144-3p inhibition had opposite effects on markers
of inflammation and oxidative stress compared with those of miR-
144-3p mimics (Fig. S8C and S8D). Cotransfected with siRNA-
circDcbld2 and miR-144-3p mimics resulted in further re-
ductions in the expression levels of inflammatory markers (IL-1b)
(Fig. 7F) and oxidative stress-related indicators (Nox1,Nox2, and
Nox4) than those for siRNA-circDcbld2 single transfection
(Supporting Information Fig. S9A). In addition, SOD and GSH
contents were further increased (Fig. S9B). These results indicate
that circDcbld2 may bind to and regulate the expression of miR-
144-3p in BMDMs.
3.8. circDcbld2 up-regulates Et-1 expression by sponging miR-
144-3p
miRNAs regulate gene expression by binding the 50or 30
untranslated regions of mRNAs to suppress translation. Whole-
transcriptome-seqencing was employed to explore candidate
genes regulated by circDcbld2/miR-144-3p in KCs in HF
(Supporting Information File S6). Differentially expressed
mRNAs in KCs extracted from vehicle and HF mice are shown in
scatter plots in Supporting Information Fig. S10A. These differ-
entially expressed genes were evaluated through GO classification
(Fig. S10B) and KEGG pathway enrichment analyses (Fig. S10C).
Following miRNA microarray and RNA-seq analyses (Fig. S10D),
Et-1 was identified as a candidate target mRNA of miR-144-3p.
The whole-transcriptome-seq analysis revealed that Et-1 expres-
sion is elevated in KCs (Supporting Information File S7). Et-1
binding sites in the miR-144-3p sequence were identified
(Fig. 8A). We found that Et-1 was increased in LPS-stimulated
BMDMs compared with controls (Fig. 8B). Additionally, the
expression of Et-1 decreased with the inhibition of circDcbld2 and
further decreased after the administration of miR-144-3p mimics
(Fig. 8C). In both immunohistochemical and immunofluorescent
analyses of liver tissue, Et-1 expression was decreased in the
CCl
4
-induced HF model following circDcbld2-KD suppression
(Fig. 8D). These results indicate that the circDcbld2/mir-144-3p/
Et-1 axis plays a role in HF; in particular, circDcbld2 sponges
miR-144-3p, thereby affecting Et-1 expression.
4. Discussion
4.1. Stability of circDcbld2 is regulated by m6A modification
Advances in RNA sequencing technology provide a basis for
elucidating the mechanisms underlying the effects of m6A
methylation
45
. In cancer, m6A modifications play significant roles
in RNA stability, interactions, and production
46
. Altering m6A
levels in Mettl3 and Alkbh5 affects circRNA biosynthesis
47,48
.
However, m6A modifications on circRNAs have not been
explored in KCs during HF formation. In this study, we showed
that circDcbld2 is an important promoter of HF and the presence
of methylation sites in the circDcbld2 sequence was predicted. In
N
6
-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis 307
Figure 6 Silencing circDcbld2 reduces HSC activation in HF mice. (A) Schematic representation of the co-culture of primary HSCs and
BMDMs. (B) Timp-1,a-SMA, Col1a1, and Tgf-b1mRNA level for primary HSCs, which affected by BMDM with siRNA-circDcbld2 (nZ6).
(C) The protein expression of a-SMA and Col1a1in HSCs, which affected by BMDMs with siRNA-circDcbld2 administration. (D) Immuno-
fluorescent staining indicated a-SMA and F4/80 were enhanced in the CCl
4
-induced HF model and decreased following circDcbld2-KD
administration. Representative images were presented, scale bar Z50 mm. (E) The protein expression level of a-SMA and Col1a1in pri-
mary HSCs by circDcbld2-KD administration. (F) The mRNA expression level of a-SMA, Col1a1,Timp-1 and Tgf-b1in primary HSCs by
circDcbld2-KD administration (nZ6). The data represent the mean SEM. **P<0.01, ***P<0.001 vs LPS þsiRNA-NC-circDcbld2 group
(B) and Vehicle group (F);
###
P<0.001 vs CCl
4
-induced liver fibrosis group (F).
308 Sai Zhu et al.
exploring the upstream regulation mechanism of circDcbld2, we
found a m6A methylation site in circDcbld2 and further confirmed
that circDcbld2 could bind to Wtap and Igf2bp2. The m6A
sequence motif ‘RRm6ACH’ (R ZGorA;HZA, C, or U) is the
consensus recognition sequence for Igf2bp2. We speculated that
Igf2bp2 binds circDcbld2 via the m6A motif. Furthermore, the
increase in circDcbld2 expression in the model group may be due
to the modified and identified circDcbld2 m6A site by Wtap and
Figure 7 Microarray analysis and identification of circDcbld2/miR-144-3p interaction. (A) Schematic of miR-144-3p binding site in
circDcbld2. (B) Renilla luciferase activity analysis of wild-type or mutant circDcbld2 and miRNAs mimics, respectively. (C) miR-144-3p (FAM)
predominantly localized in the cytoplasm with FISH assay. Representative images were presented, scale bar Z20 mm. (D) miR-144-3p sup-
pression was confirmed by qRT-PCR in LPS-stimulated BMDMs (nZ3). (E) Lower level of miR-144-3p in BMDMs was increased following
siRNA-circDcbld2 administration (nZ3). (F) IL-1bmRNA expression was decreased in BMDMs following siRNA-circDcbld2 administration.
The influence of siRNA-circDcbld2 was further decreased following miR-144-3p mimics (nZ3). The data represent the mean SEM. ns, no
significance; *P<0.05, **P<0.01, ***P<0.001 vs control group (D, E) and LPS group (F);
###
P<0.001 vs LPS þsiRNA-NC-circDcbld2
group (E) and LPS þsiRNA-circDcbld2 group (F).
N
6
-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis 309
Igf2bp2, which enhances the stability of circDcbld2, in turn
affecting inflammation and oxidative stress. This explained the
mechanism of circDcbld2 up-regulation in HF. These findings
show for the first time that circDcbld2 binds key m6A writer and
reader proteins, suggesting the potential for m6A modification on
circRNAs.
4.2. circDcbld2 contributes to inflammation, oxidative stress
activation, and HSC activation, and differentiation in HF
Imbalances in inflammation and oxidation play pivotal roles in HF
formation
49
. In this study, circDcbld2 levels were significantly
higher in the HF model than in controls and were elevated in
patients with HF. The differential expression of circDcbld2
prompted us to further investigate its function and mechanism of
action. We discovered that circDcbld2 suppression significantly
reduces the secretion of inflammatory cytokines in macrophages,
alleviates liver fibrogenesis injury, and inhibits chemokine
expression. The down-regulation of circDcbld2 protects against
oxidative stress in macrophages, suggesting that it contributes to
HF by regulating oxidative stress within macrophages. Addition-
ally, HSC trans-differentiation into myofibroblasts is a central
event during HF formation
13
. Therefore, investigating the impact
of inflammatory factors and chemokines secreted by macrophages
on HSC production represents an important direction for our
future research. In this study, silencing circDcbld2 in macrophages
effectively reduced HSC activation and trans-differentiation,
alleviated collagen deposition, and reduced liver injury. Collec-
tively, these results indicate that circDcbld2 exerts pro-fibrotic
effects through increased inflammatory factor and chemokine
secretion, while influencing the oxidative stress balance; further-
more, it facilitates HSC trans-differentiation into myofibroblasts,
thereby accelerating HF progression.
4.3. Identification of the circDcbld2/miR-144-3p/Et-1 axis in
KCs
Despite increasing research on circRNAs in HF, the functions
and mechanisms of circRNAs in KCs remain elusive. HF is
characterized by progressive inflammation and extracellular ma-
trix (ECM) deposition. Liver macrophages play a central role in
driving inflammation and HSC activation to trigger HF
12
. Our
previous studies have demonstrated that the aberrant expression of
circFbxw4 may inhibit HF progression by modulating HSC acti-
vation
28
. Although numerous circRNAs have been investigated in
HSCs, their functions and expression profiles within liver
Figure 8 circDcbld2 up-regulates Et-1 expression by sponging miR-144-3p. (A) Binding sites of miR-144-3p and 30UTR of Et-1.(B)Et-1 was
increased in LPS-stimulated BMDMs (nZ3). (C) Et-1 decreased with the siRNA-circDcbld2 administration and further reduced with miR-144-3p
mimics (nZ3). (D) Immunohistochemistry and immunofluorescence analysis of liver tissue, Et-1 expression was reduced in HF model following
circDcbld2-KD administration. Quantification of fibrosis based on immunohistochemistry analysis of Et-1. Representative images were presented,
scale bar = 100 mm/50 mm. The data represent the mean SEM. **P<0.01, ***P<0.001 vs Control group (B) and LPS group (C) and Vehicle
group (D);
#
P<0.05,
###
P<0.001 vs LPS þsiRNA-circDcbld2 group (C) and CCl
4
-induced liver fibrosis group (D).
310 Sai Zhu et al.
macrophages remain largely unexplored. Furthermore, circRNAs
function as miRNA sponges, transcriptional regulators, RBP-
binding molecules, and protein translation templates in cellular
physiology
50
. Mature miRNAs bind specifically to target mRNAs,
leading to translational repression or mRNA cleavage. Thus,
circRNAs can sequester miRNAs and counteract the miRNA-
mediated suppression of mRNA expression
51
. Our results
revealed that circDcbld2 functions by sponging miR-144-3p.
Notably, circDcbld2 suppression increased miR-144-3p expres-
sion in macrophages. Furthermore, miR-144-3p bound to the 30
untranslated region of Et-1 and down-regulated Et-1 expression
during liver fibrogenesis following decreased levels of circDcbld2.
The impact of circDcbld2 on Et-1 was partially reversed upon
miR-144-3p mimics administration.
5. Conclusions
In this study, we illuminated the first evidence for the pro-
fibrogenic function of circDcbld2 in KCs during the development
of HF and clarified its mechanism of action. We proved that Wtap
interacts with circDcbld2, and Igf2bp2 identifies m6A-modified
circDcbld2 and positively mediates the expression of circDcbld2
by enhancing its stability. In HF, circDcbld2 affected KC in-
flammatory factor production and HSC activation via miR-144-3p/
Et-1. Moreover, circDcbld2 increased the inflammatory response
and oxidative stress during the development of HF and facilitated
HSC trans-differentiation into myofibroblasts, thereby acceler-
ating disease progression (Fig. 9). However, several limitations of
this study need to be addressed. For example, the patient sample
size was small. Although our preliminary findings show that
circDcbld2 is up-regulated in patients with HF, the value of
circDcbld2 as a potential biomarker needs to be validated using
more samples. Moreover, the intercellular communication
between macrophages and HSCs during the experiment was of
interest; however, we only briefly explored the effect of inflam-
matory factors and chemokines secreted by macrophages on HSC
production and the underlying mechanism; these unresolved is-
sues will be a focus of our future research. Our study indicated
that circDcbld2 is a potential biomarker for HF, providing a basis
for the development of novel treatment options.
Figure 9 Wtap mediates the N
6
-methyladenosine (m6A) methylation of circDcbld2, Igf2bp2 recognized m6A-modified circDcbld2 and
increased its stability. circDcbld2 participated in the occurrence of HF by binding miR-144-3p/Et-1 to regulate inflammatory response and
oxidative stress.
N
6
-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis 311
Acknowledgments
This work was supported by the National Natural Science Foun-
dation of China (U19A2001, 82370630, and 82300722); the Anhui
Provincial Natural Science Foundation (No. 2308085QH248,
China); the Research Fund of Anhui Institute of Translational
Medicine (2021zhyx-B06 and 2022zhyx-B07, China); the China
Postdoctoral Science Foundation (No. 2022M710178); the Fund
of Traditional Chinese Medicine Institute of Anhui Dabie
Mountain (No. TCMADM-2024-02, China).
Author contributions
Sai Zhu: Writing eoriginal draft, Resources, Project adminis-
tration, Data curation. Xin Chen: Funding acquisition, Formal
analysis, Data curation. Lijiao Sun: Methodology, Data curation.
Xiaofeng Li: Writing eoriginal draft, Methodology, Formal
analysis. Yu Chen: Software. Liangyun Li: Methodology, Data
curation. Xiaoguo Suo: Methodology. Chuanhui Xu: Software,
Methodology. Minglu Ji: Methodology. Jianan Wang: Data cura-
tion. Hua Wang: Project administration. Lei Zhang: Project
administration. Xiaoming Meng: Methodology, Formal
analysis. Cheng Huang: Software, Methodology. Jun Li:
Writing ereview & editing, Funding acquisition.
Conflicts of interest
The authors declare no conflicts of interest.
Appendix A. Supporting information
Supporting information to this article can be found online at
https://doi.org/10.1016/j.apsb.2024.11.003.
References
1. Mokdad AA, Lopez AD, Shahraz S, Lozano R, Mokdad AH,
Stanaway J, et al. Liver cirrhosis mortality in 187 countries between
1980 and 2010: a systematic analysis. BMC Med 2014;12:145.
2. Luo P, Liu D, Zhang Q, Yang F, Wong YK, Xia F, etal. Celastrolinduces
ferroptosis in activated HSCs to ameliorate hepatic fibrosis via targeting
peroxiredoxins and HO-1. Acta Pharm Sin B 2022;12:2300e14.
3. Zou GL, Zuo S, Lu S, Hu RH, Lu YY, Yang J, et al. Bone morpho-
genetic protein-7 represses hepatic stellate cell activation and liver
fibrosis via regulation of TGF-beta/Smad signaling pathway. World J
Gastroenterol 2019;25:4222e34.
4. Tsochatzis EA, Bosch J, Burroughs AK. Liver cirrhosis. Lancet 2014;
383:1749e61.
5. Liu R, Li Y, Zheng Q, Ding M, Zhou H, Li X. Epigenetic modification
in liver fibrosis: promising therapeutic direction with significant
challenges ahead. Acta Pharm Sin B 2024;14:1009e29.
6. Sharma RS, Harrison DJ, Kisielewski D, Cassidy DM, McNeilly AD,
Gallagher JR, et al. Experimental nonalcoholic steatohepatitis and liver
fibrosis are ameliorated by pharmacologic activation of Nrf2 (NF-E2
p45-related factor 2). Cell Mol Gastroenterol Hepatol 2018;5:367e98.
7. Ma PF, Gao CC, Yi J, Zhao JL, Liang SQ, Zhao Y, et al. Cytotherapy
with M1-polarized macrophages ameliorates liver fibrosis by modu-
lating immune microenvironment in mice. J Hepatol 2017;67:770e9.
8. Li S, Zhou B, Xue M, Zhu J, Tong G, Fan J, et al. Macrophage-specific
FGF12 promotes liver fibrosis progression in mice. Hepatology 2023;
77:816e33.
9. Krenkel O, Tacke F. Liver macrophages in tissue homeostasis and
disease. Nat Rev Immunol 2017;17:306e21.
10. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis
and its regression. Nat Rev Gastroenterol Hepatol 2021;18:151e66.
11. Ge C, Tan J, Lou D, Zhu L, Zhong Z, Dai X, et al. Mulberrin confers
protection against hepatic fibrosis by Trim31/Nrf2 signaling. Redox
Biol 2022;51:102274.
12. Meng F, Wang K, Aoyama T, Grivennikov SI, Paik Y, Scholten D,
et al. Interleukin-17 signaling in inflammatory, Kupffer cells, and
hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenter-
ology 2012;143:765e76. e3.
13. Kong M, Dong W, Kang A, Kuai Y, Xu T, Fan Z, et al. Regulatory role
and translational potential of CCL11 in liver fibrosis. Hepatology
2023;78:120e35.
14. Kisseleva T. The origin of fibrogenic myofibroblasts in fibrotic liver.
Hepatology 2017;65:1039e43.
15. Xie X, Lv H, Liu C, Su X, Yu Z, Song S, et al. HBeAg mediates
inflammatory functions of macrophages by TLR2 contributing to he-
patic fibrosis. BMC Med 2021;19:247.
16. He L, Li H, Wu A, Peng Y, Shu G, Yin G. Functions of N
6
-methyl-
adenosine and its role in cancer. Mol Cancer 2019;18:176.
17. Bai Y, Chang D, Ren H, Ju M, Wang Y, Chen B, et al. Engagement of N
6
-
methyladenisine methylation of Gng4 mRNA in astrocyte dysfunction
regulated by CircHECW2. Acta Pharm Sin B 2024;14:1644e60.
18. Jeck WR, Sharpless NE. Detecting and characterizing circular RNAs.
Nat Biotechnol 2014;32:453e61.
19. Chen C, Yuan WT, Zhou QB, Shao B, Guo YY, Wang WW, et al. N
6
-
Methyladenosine-induced circ1662 promotes metastasis of colorectal
cancer by accelerating YAP1 nuclear localization. Theranostics 2021;
11:4298e315.
20. Chen Y, Ling Z, Cai X, Xu Y, Lv Z, Man D, et al. Activation of YAP1
by N-6-methyladenosine-modified circCPSF6 drives malignancy in
hepatocellular carcinoma. Cancer Res 2022;82:599e614.
21. Pamudurti NR, Bartok O, Jens M, Ashwal-Fluss R, Stottmeister C,
Ruhe L, et al. Translation of circRNAs. Mol Cell 2017;66:9e21.e7.
22. Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B,
Damgaard CK, et al. Natural RNA circles function as efficient
microRNA sponges. Nature 2013;495:384e8.
23. Du WW, Yang WN, Liu E, Yang ZG, Dhaliwal P, Yang BB. Foxo3
circular RNA retards cell cycle progression via forming ternary
complexes with p21 and CDK2. Nucleic Acids Res 2016;44:2846e58.
24. Yang Y, Gao X, Zhang M, Yan S, Sun C, Xiao F, et al. Novel role of
FBXW7 circular RNA in repressing glioma tumorigenesis. J Natl
Cancer Inst 2018;110:304e15.
25. Wang P, Huang Z, Peng Y, Li H, Lin T, Zhao Y, et al. Circular RNA
circBNC2 inhibits epithelial cell G2eM arrest to prevent fibrotic
maladaptive repair. Nat Commun 2022;13:6502.
26. Chen L, Yao X, Yao H, Ji Q, Ding G, Liu X. Exosomal miR-103-3p
from LPS-activated THP-1 macrophage contributes to the activation
of hepatic stellate cells. FASEB J 2020;34:5178e92.
27. Hu ZQ, Zhou SL, Li J, Zhou ZJ, Wang PC, Xin HY, et al. Circular
RNA sequencing identifies circASAP1 as a key regulator in hepato-
cellular carcinoma metastasis. Hepatology 2020;72:906e22.
28. Chen X, Li HD, Bu FT, Li XF, Chen Y, Zhu S, et al. Circular RNA
circFBXW4 suppresses hepatic fibrosis via targeting the miR-18b-
3p/FBXW7 axis. Theranostics 2020;10:4851e70.
29. Bu FT, Zhu Y, Chen X, Wang A, Zhang YF, You HM, et al. Circular
RNA circPSD3 alleviates hepatic fibrogenesis by regulating the miR-
92b-3p/Smad7 axis. Mol Ther-Nucl Acids 2021;23:847e62.
30. Lu XY, Liu YR, Xuan WT, Ye J, Yao HW, Huang C, et al. Circ_1639
induces cells inflammation responses by sponging miR-122 and
regulating TNFRSF13C expression in alcoholic liver disease. Toxicol
Lett 2019;314:89e97.
31. He Y, Feng D, Li M, Gao Y, Ramirez T, Cao H, et al. Hepatic mito-
chondrial DNA/Toll-like receptor 9/MicroRNA-223 forms a negative
feedback loop to limit neutrophil overactivation and acetaminophen
hepatotoxicity in mice. Hepatology 2017;66:220e34.
32. Yang JJ, Tao H, Liu LP, Hu W, Deng ZY, Li J. miR-200a controls
hepatic stellate cell activation and fibrosis via SIRT1/Notch1 signal
pathway. Inflamm Res 2017;66:341e52.
312 Sai Zhu et al.
33. Zhu S, Chen X, Wang JN, Xu JJ, Wang A, Li JJ, et al. Circular RNA
circUbe2k promotes hepatic fibrosis via sponging miR-149-5p/TGF-
beta2 axis. FASEB J 2021;35:e21622.
34. Xu JJ, Chen X, Zhu S, Jiang LF, Ma WX, Chen SY, et al. Myc-
mediated circular RNA circMcph1/miR-370-3p/Irak2 axis is a pro-
gressive regulator in hepatic fibrosis. Life Sci 2023;312:121182.
35. Chen YH, Yuan BY, Wu ZF, Dong YY, Zhang L, Zeng ZC. Microarray
profiling of circular RNAs and the potential regulatory role of
hsa_circ_0071410 in the activated human hepatic stellate cell induced
by irradiation. Gene 2017;629:35e42.
36. Miyoshi H, Rust C, Roberts PJ, Burgart LJ, Gores GJ. Hepatocyte
apoptosis after bile duct ligation in the mouse involves Fas. Gastro-
enterology 1999;117:669e77.
37. Kang H, Seo E, Park JM, Han NY, Lee H, Jun HS. Effects of FGF21-
secreting adipose-derived stem cells in thioacetamide-induced hepatic
fibrosis. J Cell Mol Med 2018;22:5165e9.
38. Chen X, Li XF, Chen Y, Zhu S, Li HD, Chen SY, et al. Hesperetin
derivative attenuates CCl
4
-induced hepatic fibrosis and inflammation by
Gli-1-dependent mechanisms. Int Immunopharmacol 2019;76:105838.
39. Starkey Lewis P, Campana L, Aleksieva N, Cartwright JA,
Mackinnon A, O’Duibhir E, et al. Alternatively activated macrophages
promote resolution of necrosis following acute liver injury. J Hepatol
2020;73:349e60.
40. Yang Y, Wu XQ, Li WX, Huang HM, Li HD, Pan XY, et al. PSTPIP2
connects DNA methylation to macrophage polarization in CCl
4
-induced
mouse model of hepatic fibrosis. Oncogene 2018;37:6119e35.
41. Chen Y, Chen X, Ji YR, Zhu S, Bu FT, Du XS, et al. PLK1 regulates
hepatic stellate cell activation and liver fibrosis through Wnt/beta-
catenin signalling pathway. J Cell Mol Med 2020;24:7405e16.
42. Nguyen-Lefebvre AT, Ajith A, Portik-Dobos V, Horuzsko DD,
Arbab AS, Dzutsev A, et al. The innate immune receptor TREM-1
promotes liver injury and fibrosis. J Clin Invest 2018;128:4870e83.
43. Kristensen LS, Andersen MS, Stagsted LVW, Ebbesen KK,
Hansen TB, Kjems J. The biogenesis, biology and characterization of
circular RNAs. Nat Rev Genet 2019;20:675e91.
44. Wang JN, Wang F, Ke J, Li Z, Xu CH, Yang Q, et al. Inhibition of
METTL3 attenuates renal injury and inflammation by alleviating
TAB3 m6A modifications via IGF2BP2-dependent mechanisms. Sci
Transl Med 2022;14:eabk2709.
45. Liu J, Jia G. Methylation modifications in eukaryotic messenger RNA.
J Genet Genomics 2014;41:21e33.
46. Niu Y, Zhao X, Wu YS, Li MM, Wang XJ, Yang YG. N
6
-methyl-
adenosine (m6A) in RNA: an old modification with a novel epigenetic
function. Genomics Proteomics Bioinformatics 2013;11:8e17.
47. Yi YC, Chen XY, Zhang J, Zhu JS. Novel insights into the interplay
between m(6)A modification and noncoding RNAs in cancer. Mol
Cancer 2020;19:121.
48. Wang W, Qiao SC, Wu XB,Sun B, Yang JG, Li X, et al. Circ_0008542 in
osteoblast exosomes promotes osteoclast-induced bone resorption
through m6A methylation. Cell Death Dis 2021;12:628.
49. Ramirez T, Li YM, Yin S, Xu MJ, Feng D, Zhou Z, et al. Aging
aggravates alcoholic liver injury and fibrosis in mice by down-
regulating sirtuin 1 expression. J Hepatol 2017;66:601e9.
50. Zhou WY, Cai ZR, Liu J, Wang DS, Ju HQ, Nxu RH. Circular RNA:
metabolism, functions and interactions with proteins. Mol Cancer
2020;19:172.
51. Chen LL. The expanding regulatory mechanisms and cellular func-
tions of circular RNAs. Nat Rev Mol Cell Biol 2020;21:475e90.
N
6
-Methyladenosine modification of circDcbld2 in Kupffer cells promotes hepatic fibrosis 313
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The N⁶-methyladenosine (m⁶A) modification is the most prevalent modification of eukaryotic mRNAs and plays a crucial role in various physiological processes by regulating the stability or function of target mRNAs. Accumulating evidence has suggested that m⁶A methylation may be involved in the pathological process of major depressive disorder (MDD), a common neuropsychiatric disorder with an unclear aetiology. Here, we found that the levels of the circular RNA HECW2 (circHECW2) were significantly increased in the plasma of both MDD patients and the chronic unpredictable stress (CUS) mouse model. Notably, the downregulation of circHECW2 attenuated astrocyte dysfunction and depression-like behaviors induced by CUS. Furthermore, we demonstrated that the downregulation of circHECW2 increased the expression of the methylase WTAP, leading to an increase in Gng4 expression via m⁶A modifications. Our findings provide functional insight into the correlation between circHECW2 and m⁶A methylation, suggesting that circHECW2 may represent a potential target for MDD treatment.
Article
Full-text available
Liver fibrosis, characterized by scar tissue formation, can ultimately result in liver failure. It's a major cause of morbidity and mortality globally, often associated with chronic liver diseases like hepatitis or alcoholic and non-alcoholic fatty liver diseases. However, current treatment options are limited, highlighting the urgent need for the development of new therapies. As a reversible regulatory mechanism, epigenetic modification is implicated in many biological processes, including liver fibrosis. Exploring the epigenetic mechanisms involved in liver fibrosis could provide valuable insights into developing new treatments for chronic liver diseases, although the current evidence is still controversial. This review provides a comprehensive summary of the regulatory mechanisms and critical targets of epigenetic modifications, including DNA methylation, histone modification, and RNA modification, in liver fibrotic diseases. The potential cooperation of different epigenetic modifications in promoting fibrogenesis was also highlighted. Finally, available agonists or inhibitors regulating these epigenetic mechanisms and their potential application in preventing liver fibrosis were discussed. In summary, elucidating specific druggable epigenetic targets and developing more selective and specific candidate medicines may represent a promising approach with bright prospects for the treatment of chronic liver diseases.
Article
Full-text available
Aims Treating hepatic fibrosis (HF) is a major challenge worldwide. However, the biological functions and regulatory mechanisms of circular RNAs (circRNAs) remain unclear in HF. The present study aimed to elucidate the novel role of circMcph1 in HF. Main methods HF mouse model was established by injecting CCl4 intraperitoneally and validated using hematoxylin and eosin staining, immunohistochemistry, and serological tests in vivo. RAW264.7 cells were treated with lipopolysaccharide (LPS) and interferon-γ (IFN-γ) in vitro inflammatory damage model. Gel electrophoresis, DNA sequencing, RNase R and actinomycin D treatment, random 6 primers and oligo dT primers assay, nuclear and cytoplasmic fractionation assays, and fluorescence in situ hybridization were performed to identify the characteristics of circMcph1. Functional assays such as ELISA, flow cytometry, and adeno-associated virus administration in vivo and liposome delivery gene therapy in vitro were used to determine the functional effects of circMcph1/miR-370-3p/interleukin-1 receptor-associated kinase 2 (Irak2) axis. Mechanistic assays such as luciferase reporter analysis, and chromatin immunoprecipitation revealed the molecular mechanism of the Myc/circMcph1/miR-370-3p/Irak2 axis in HF. Key findings CircMcph1 expression was upregulated in liver tissues and primary Kupffer cells of CCl4-induced HF mice, as well as in LPS- and IFN-γ-treated RAW264.7 cells. Knockdown of circMcph1 ameliorated liver fibrogenesis and inflammatory damage in HF mice and reduced the inflammatory response in LPS and IFN-γ-treated RAW264.7 cells. Mechanically, circMcph1 mediated by Myc regulated the expression of Irak2 by sponging miR-370-3p in HF. Significance The study findings suggested that the Myc/circMcph1/miR-370-3p axis might be a novel identifier and therapeutic target for HF.
Article
Full-text available
The mechanisms underlying fibrogenic responses after injury are not well understood. Epithelial cell cycle arrest in G2/M after injury is a key checkpoint for determining wound-healing leading to either normal cell proliferation or fibrosis. Here, we identify a kidney- and liver-enriched circular RNA, circBNC2, which is abundantly expressed in normal renal tubular cells and hepatocytes but significantly downregulated after acute ischemic or toxic insult. Loss of circBNC2 is at least partially mediated by upregulation of DHX9. Gain- and loss-of-function studies, both in vitro and in vivo, demonstrate that circBNC2 acts as a negative regulator of cell G2/M arrest by encoding a protein that promotes formation of CDK1/cyclin B1 complexes. Restoring circBNC2 in experimentally-induced male mouse models of fibrotic kidney and liver, decreases G2/M arrested cell numbers with secretion of fibrotic factors, thereby mitigating extracellular matrix deposition and fibrosis. Decreased expression of circBNC2 and increased G2/M arrest of epithelial cells are recapitulated in human ischemic reperfusion injury (IRI)-induced chronic kidney disease and inflammation-induced liver fibrosis, highlighting the clinical relevance. These findings suggest that restoring circBNC2 might represent a potential strategy for therapeutic intervention in epithelial organ fibrosis. G2/M arrest of epithelial cells leads to fibrosis with unclear mechanisms. This study identifies a protein-encoding circRNA, circBNC2, which inhibits epithelial cells G2/M arrest to prevent fibrotic maladaptive repair in damaged kidney and liver, revealing a potential intervention target for fibrosis.
Article
Full-text available
Mulberrin (Mul) is a key component of the traditional Chinese medicine Romulus Mori with various biological functions. However, the effects of Mul on liver fibrosis have not been addressed, and thus were investigated in our present study, as well as the underlying mechanisms. Here, we found that Mul administration significantly ameliorated carbon tetrachloride (CCl4)-induced liver injury and dysfunction in mice. Furthermore, CCl4-triggerd collagen deposition and liver fibrosis were remarkably attenuated in mice with Mul supplementation through suppressing transforming growth factor β1 (TGF-β1)/SMAD2/3 signaling pathway. Additionally, Mul treatments strongly restrained the hepatic inflammation in CCl4-challenged mice via blocking nuclear factor-κB (NF-κB) signaling. Importantly, we found that Mul markedly increased liver TRIM31 expression in CCl4-treated mice, accompanied with the inactivation of NOD-like receptor protein 3 (NLRP3) inflammasome. CCl4-triggered hepatic oxidative stress was also efficiently mitigated by Mul consumption via improving nuclear factor E2-related factor 2 (Nrf2) activation. Our in vitro studies confirmed that Mul reduced the activation of human and mouse primary hepatic stellate cells (HSCs) stimulated by TGF-β1. Consistently, Mul remarkably retarded the inflammatory response and reactive oxygen species (ROS) accumulation both in human and murine hepatocytes. More importantly, by using hepatocyte-specific TRIM31 knockout mice (TRIM31Hep-cKO) and mouse primary hepatocytes with Nrf2-knockout (Nrf2KO), we identified that the anti-fibrotic and hepatic protective effects of Mul were TRIM31/Nrf2 signaling-dependent, relieving HSCs activation and liver fibrosis. Therefore, Mul-ameliorated hepatocyte injury contributed to the suppression of HSCs activation by improving TRIM31/Nrf2 axis, thus providing a novel therapeutic strategy for hepatic fibrosis treatment.
Article
Full-text available
Ferroptosis is a form of regulated cell death, characterized by excessive membrane lipid peroxidation in an iron- and ROS-dependent manner. Celastrol, a natural bioactive triterpenoid extracted from Tripterygium wilfordii, shows effective anti-fibrotic and anti-inflammatory activities in multiple hepatic diseases. However, the exact molecular mechanisms of action and the direct protein targets of celastrol in the treatment of liver fibrosis remain largely elusive. Here, we discover that celastrol exerts anti-fibrotic effects via promoting the production of reactive oxygen species (ROS) and inducing ferroptosis in activated hepatic stellate cells (HSCs). By using activity-based protein profiling (ABPP) in combination with bio-orthogonal click chemistry reaction and cellular thermal shift assay (CETSA), we show that celastrol directly binds to peroxiredoxins (PRDXs), including PRDX1, PRDX2, PRDX4 and PRDX6, through the active cysteine sites, and inhibits their anti-oxidant activities. Celastrol also targets to heme oxygenase 1 (HO-1) and upregulates its expression in activated-HSCs. Knockdown of PRDX1, PRDX2, PRDX4, PRDX6 or HO-1 in HSCs, to varying extent, elevated cellular ROS levels and induced ferroptosis. Taken together, our findings reveal the direct protein targets and molecular mechanisms via which celastrol ameliorates hepatic fibrosis, thus supporting the further development of celastrol as a promising therapeutic agent for liver fibrosis.
Article
Background and aims: Myofibroblasts are considered the major effector cell type of liver fibrosis and primarily derived from hepatic stellate cells (HSCs). In the present study, we investigated the contribution of C-C motif chemokine (CCL11) to HSC-myofibroblast trans -differentiation and its implication in liver fibrosis. Approach and results: We report that CCL11 levels were elevated in HSCs, but not in hepatocytes or Kupffer cells, isolated from mice with liver fibrosis compared with the control mice. CCL11 levels were also up-regulated by 2 pro-fibrogenic growth factors TGF-β and platelet derived growth factor in cultured HSCs. Mechanistically, zinc finger factor 281 bound to the CCL11 promoter and mediated CCL11 trans -activation in HSCs. Depletion of CCL11 attenuated whereas treatment with recombinant CCL11 promoted HSC activation. Further, global CCL11 deletion ( CCL11-/- ) or HSC/myofibroblast-specific CCL11 knockdown mitigated fibrogenesis in mice. RNA-sequencing revealed that CCL11 might regulate HSC activation by stimulating the transcription of Jagged 1. Reconstitution of Jagged 1 restored the fibrogenic response in CCL11-/- mice. Finally, several targeting strategies that aimed at blockading CCL11 signaling, either by administration of an antagonist to its receptor C-C motif chemokine receptor 3 or neutralizing antibodies against CCL11/C-C motif chemokine receptor 3, ameliorated liver fibrosis in mice. Conclusions: Our data unveil a previously unrecognized role for CCL11 in liver fibrosis and provide proof-of-concept evidence that targeting CCL11 can be considered as an effective therapeutic approach.
Article
Background & aims: Chronic liver diseases are associated with the development of liver fibrosis. Without treatment, liver fibrosis commonly leads to cirrhosis and hepatocellular carcinoma. Fibroblast growth factor 12 (FGF12) is an intracrine factor belonging to the FGF superfamily, but its role in liver homeostasis is largely unknown. This study aimed to investigate the role of FGF12 in the regulation of liver fibrosis. Approach & results: FGF12 was upregulated in bile duct ligation (BDL)- and carbon tetrachloride (CCL4)-induced liver fibrosis mouse models. The expression of FGF12 was specifically upregulated in non-parenchymal liver cells, especially in hepatic macrophages. By constructing myeloid-specific FGF12 knockout mice, we found that deletion of FGF12 in macrophages protected against BDL- and CCL4-induced liver fibrosis. Further results revealed that FGF12 deletion dramatically decreased the population of lymphocyte antigen 6 complex locus C (Ly6C) high macrophages in mouse fibrotic liver tissue, and reduced the expression of proinflammatory cytokines and chemokines. Meanwhile, loss- and gain-of-function approaches revealed that FGF12 promoted the proinflammatory activation of macrophages, thus inducing hepatic stellate cell activation mainly through the MCP-1/CCR2 axis. Further experiments indicated that the regulation of macrophage activation by FGF12 was mainly mediated through the Janus kinase-signal transducer of activators of transcription pathway. Finally, results revealed that FGF12 expression correlates with the severity of fibrosis across the spectrum of fibrogenesis in human liver samples. Conclusions: FGF12 is a novel factor to promote liver fibrosis progression. Therapeutic approaches to inhibit macrophage FGF12 may be used to combat liver fibrosis in the future.
Article
The role of N ⁶ -methyladenosine (m6A) modifications in renal diseases is largely unknown. Here, we characterized the role of N ⁶ -adenosine-methyltransferase-like 3 (METTL3), whose expression is elevated in renal tubules in different acute kidney injury (AKI) models as well as in human biopsies and cultured tubular epithelial cells (TECs). METTL3 silencing alleviated renal inflammation and programmed cell death in TECs in response to stimulation by tumor necrosis factor–α (TNF-α), cisplatin, and lipopolysaccharide (LPS), whereas METTL3 overexpression had the opposite effects. Conditional knockout of METTL3 from mouse kidneys attenuated cisplatin- and ischemic/reperfusion (I/R)–induced renal dysfunction, injury, and inflammation. Moreover, TAB3 [TGF-β–activated kinase 1 (MAP3K7) binding protein 3] was identified as a target of METTL3 by m6A methylated RNA immunoprecipitation sequencing and RNA sequencing. The stability of TAB3 was increased through binding of IGF2BP2 (insulin-like growth factor 2 binding protein 2) to its m6A-modified stop codon regions. The proinflammatory effects of TAB3 were then explored both in vitro and in vivo. Adeno-associated virus 9 (AAV9)–mediated METTL3 silencing attenuated renal injury and inflammation in cisplatin- and LPS-induced AKI mouse models. We further identified Cpd-564 as a METTL3 inhibitor that had better protective effects against cisplatin- and ischemia/reperfusion-induced renal injury and inflammation than S -adenosyl- l -homocysteine, a previously identified METTL3 inhibitor. Collectively, METTL3 promoted m6A modifications of TAB3 and enhanced its stability via IGF2BP2-dependent mechanisms. Both genetic and pharmacological inhibition of METTL3 attenuated renal injury and inflammation, suggesting that the METTL3/TAB3 axis is a potential target for treatment of AKI.
Article
Circular RNAs (circRNA) and N6-methyladenosine (m6A) modification are extensively involved in the progression of diverse tumors, including hepatocellular carcinoma (HCC). However, the cross-talk between circRNAs and m6A remains elusive in the pathogenesis of HCC. Here we investigated m6A-mediated regulation of circRNAs in HCC. m6A-related circRNAs were identified by integrating information from two published studies, revealing circular cleavage and polyadenylation specific factor 6 (circCPSF6) as a novel m6A-modified circRNA. circCPSF6 was dominated by ALKBH5-mediated demethylation, followed by the recognization and destabilization by YTHDF2. Meanwhile, circCPSF6 was upregulated in HCC specimens, and elevated circCPSF6 expression served as an independent prognostic factor for worse survival of patients with HCC. Loss-of-function assays demonstrated that circCPSF6 maintained cell proliferation and tumorigenicity and reinforced cell motility and tumor metastasis. circCPSF6 triggered expression of YAP1, further activating its downstream cascade. Mechanistically, circCPSF6 competitively bound PCBP2, blunting its binding to YAP1 mRNA, thereby sustaining the stability of YAP1. Functionally, removal of YAP1 reversed the effects of circCPSF6 in vitro and in vivo. Aberrant activation of the circCPSF6-YAP1 axis promoted HCC malignancy. These findings offer novel insights into the regulation of circRNAs by m6A modifications and the role of this epigenetic reprogramming in HCC. Significance: This study advances the understanding of the interplay between m6A methylation and circRNAs in hepatocellular carcinoma, highlighting the potential of circCPSF6 as a therapeutic target.