ArticlePDF Available

Circular RNA circPPM1F modulates M1 macrophage activation and pancreatic islet inflammation in type 1 diabetes mellitus

Authors:

Abstract and Figures

Rationale: Macrophages play critical roles in the pathogenesis of type 1 diabetes mellitus (T1DM). Circular RNAs (circRNAs) are a novel class of endogenous RNAs with covalently closed loop structures, implicated in various disease processes. However, their impact on macrophage activation and T1DM pathogenesis remains elusive. Methods: circRNA expression profiles of peripheral blood mononuclear cells (PBMCs) from T1DM children were determined by whole transcriptome microarray. Bioinformatics, quantitative real-time PCR, Western blot, RNA immunoprecipitation (RIP), cell co-culture, cell proliferation, and cell apoptosis assays were performed to investigate the expression, function, and regulatory mechanisms of circPPM1F in vitro. The regulatory role of circPPM1F in vivo was evaluated in the streptozocin-induced diabetic mouse model. Results: We identified 27 upregulated and 31 downregulated differentially expressed circRNAs in T1DM patients. circPPM1F, a circRNA with unknown function, was dominantly expressed in monocytes and significantly upregulated in T1DM patients. Functionally, circPPM1F promoted lipopolysaccharide (LPS)-induced M1 macrophage activation via enhancement of the NF-κB signaling pathway. Mechanistically, circPPM1F competitively interacted with HuR to impair the translation of protein phosphatase, Mg²⁺/Mn²⁺ dependent 1F (PPM1F), thus alleviating the inhibitory effect of PPM1F on the NF-κB pathway. Moreover, eukaryotic initiation factor 4A-III (EIF4A3) and fused in sarcoma (FUS) coordinately regulated circPPM1F expression during M1 macrophage activation. In addition, circPPM1F could exacerbate pancreas injury in the streptozocin-induced diabetic mice by activation of M1 macrophages in vivo. Conclusions: circPPM1F is a novel positive regulator of M1 macrophage activation through the circPPM1F-HuR-PPM1F-NF-κB axis. Overexpression of circPPM1F could promote pancreatic islet injury by enhancing M1 macrophage activation and circPPM1F may serve as a novel potential therapeutic target for T1DM in children.
circPPM1F promotes M1 macrophage activation by enhancing NF-κB signaling. A. The location of circPPM1F in the PPM1F transcript was validated by Sanger sequencing. B. Total RNA was digested with or without RNase R, followed by quantitative real-time PCR (qRT-PCR) measurements of circPPM1F, PPM1F, MALAT1, and GAPDH. C. The stability of circPPM1F was detected by qRT-PCR in THP1 macrophages after actinomycin D (Act D) treatment. D. qRT-PCR analysis of circPPM1F expression levels in THP1 macrophages following circPPM1F knockdown by two distinct siRNAs. E. qRT-PCR analyses of IL-1β, TNF-α, and CXCL10 in THP1 macrophages with conditional treatment. Mock-, untransfected, and unstimulated cells; mock+, LPS stimulated alone cells; si-scramble, LPS-stimulated cells following transfection with si-scramble; si-circPPM1F-2, LPS-stimulated cells following transfection with si-circPPM1F-2. F. ELISA analyses of secreted cytokine levels in THP1 macrophages with circPPM1F knockdown, followed by LPS treatment. G. qRT-PCR analyses of circPPM1F in THP1 macrophages with 3D5-circPPM1F or pZW1 transfection (left); M1-associated gene expressions in LPS stimulated THP1 macrophages overexpressing circPPM1F were quantified by qRT-PCR analysis (right). H. ELISA analyses of secreted cytokine levels in circPPM1F-overexpressed THP1 macrophages, followed by LPS treatment. I. Western blot showing total p65, p38, ERK1/2, JNK and their phosphorylation levels in THP1 macrophages with or without circPPM1F knockdown after LPS treatment. J. Western blotting analysis to evaluate levels of total p65, phosphorylated p65 in circPPM1F-overexpressed THP1 macrophages. The levels of p-p65 were normalized to that of β-tubulin and quantified using Image J software. Data are presented as mean ± SEM from three independent experiments. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ns indicates no significance.
… 
circPPM1F competitively binds to HuR to impair PPM1F translation. A. Western blotting analysis to evaluate levels of PPM1F in circPPM1F knocked down-THP1 macrophages. B. qRT-PCR analyses of PPM1F expression in THP1 macrophages with circPPM1F knockdown. C. Analysis of PPM1F expression in THP1 macrophages transfected with PPM1F siRNA (200 nM) or control siRNA by qRT-PCR. D. Western blot analysis of PPM1F, p65, and p-p65 in THP1 macrophages with PPM1F knockdown, followed by LPS stimulation. E. Quantitative real-time PCR (qRT-PCR) analysis of M1-associated gene expression in THP1 macrophages with PPM1F knockdown, followed by LPS stimulation. F. qRT-PCR results showing the distribution of circPPM1F in the cytoplasmic and nuclear fractions of THP1 macrophages. GAPDH as cytoplasm control transcript, and U1 as nuclear control transcript. G. Putative HuR binding sites within circPPM1F full-length sequence. H. The enrichment levels of circPPM1F and PPM1F in the products of the RNA immunoprecipitation (RIP) assay (HuR IP compared with IgG IP) as detected by qRT-PCR. I. qRT-PCR analysis of HuR in THP1 macrophages transfected with HuR siRNA (200 nM) or control siRNA. J. Protein levels of PPM1F and HuR were detected by western blotting in THP1 macrophages with HuR knockdown. K. qRT-PCR analysis of circPPM1F and PPM1F in THP1 macrophages with HuR knockdown. The levels of PPM1F, p-p65 and HuR were normalized to those of β-tubulin and quantified using Image J software. Data are presented as mean ± SEM from three independent experiments. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ns indicates no significance.
… 
Content may be subject to copyright.
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10908
Theranostics
2020; 10(24): 10908-10924. doi: 10.7150/thno.48264
Research Paper
Circular RNA circPPM1F modulates M1 macrophage
activation and pancreatic islet inflammation in type 1
diabetes mellitus
Caiyan Zhang1,2#, Xiao Han1,2#, Lan Yang1,2#, Jinrong Fu1#, Chengjun Sun3, Saihua Huang1,2, Wenfeng
Xiao1,2, Yajing Gao1,2, Qiuyan Liang1,2, Xiang Wang1,2, Feihong Luo3, Wei Lu3, Yufeng Zhou1,2
1. Institute of Pediatrics, Children’s Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of
Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
2. National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China.
3. Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children’s Hospital of Fudan University, Shanghai, 201102, China.
#These authors contributed equally to this work.
Corresponding author: Yufeng Zhou, M.D., Ph.D., 399 Wanyuan Rd, Minhang, Shanghai 201102, China. Telephone: 86-021-64932907; E-mail: yfzhou1@fudan.edu.cn.
© The author(s). This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/).
See http://ivyspring.com/terms for full terms and conditions.
Received: 2020.05.16; Accepted: 2020.08.21; Published: 2020.08.29
Abstract
Rationale: Macrophages play critical roles in the pathogenesis of type 1 diabetes mellitus (T1DM). Circular
RNAs (circRNAs) are a novel class of endogenous RNAs with covalently closed loop structures, implicated in
various disease processes. However, their impact on macrophage activation and T1DM pathogenesis remains
elusive.
Methods: circRNA expression profiles of peripheral blood mononuclear cells (PBMCs) from T1DM children
were determined by whole transcriptome microarray. Bioinformatics, quantitative real-time PCR, Western
blot, RNA immunoprecipitation (RIP), cell co-culture, cell proliferation, and cell apoptosis assays were
performed to investigate the expression, function, and regulatory mechanisms of circPPM1F in vitro. The
regulatory role of circPPM1F in vivo was evaluated in the streptozocin-induced diabetic mouse model.
Results: We identified 27 upregulated and 31 downregulated differentially expressed circRNAs in T1DM
patients. circPPM1F, a circRNA with unknown function, was dominantly expressed in monocytes and
significantly upregulated in T1DM patients. Functionally, circPPM1F promoted lipopolysaccharide (LPS)-induced
M1 macrophage activation via enhancement of the NF-κB signaling pathway. Mechanistically, circPPM1F
competitively interacted with HuR to impair the translation of protein phosphatase, Mg2+/Mn2+ dependent 1F
(PPM1F), thus alleviating the inhibitory effect of PPM1F on the NF-κB pathway. Moreover, eukaryotic initiation
factor 4A-III (EIF4A3) and fused in sarcoma (FUS) coordinately regulated circPPM1F expression during M1
macrophage activation. In addition, circPPM1F could exacerbate pancreas injury in the streptozocin-induced
diabetic mice by activation of M1 macrophages in vivo.
Conclusions: circPPM1F is a novel positive regulator of M1 macrophage activation through the
circPPM1F-HuR-PPM1F-NF-κB axis. Overexpression of circPPM1F could promote pancreatic islet injury by
enhancing M1 macrophage activation and circPPM1F may serve as a novel potential therapeutic target for T1DM
in children.
Key words: Circular RNA; Type 1 diabetes mellitus; Macrophage activation; RNA-binding protein; Islet injury
Introduction
Type 1 diabetes mellitus (T1DM, also known as
insulin-dependent diabetes) is a chronic autoimmune
disease, driven by the interplay between individual
genetics and environmental triggers [1, 2]. T1DM is
diagnosed at all ages [3, 4], and is characterized by
aberrant autoimmune destruction of pancreatic islet β
cells, resulting in a complete lack of insulin synthesis
and necessitating lifelong hormone replacement
therapy [5]. Recent studies have revealed that a spike
in the incidence of T1DM was found at ages 1014
Ivyspring
International Publisher
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10909
years, and worldwide estimates of numbers of
children and adolescents with T1DM continue to
increase [6].
Emerging evidence has suggested that,
throughout the immune system, macrophages play a
critical role in insulitis, and support autoimmune T
cells to aggravate the infiltration of inflammatory cells
during T1DM [7]. Macrophages have been classified
as two extreme examples on the activation spectrum:
classically activated macrophages (M1) with a pro-
inflammatory phenotype and alternatively activated
macrophages (M2) with an anti-inflammatory
phenotype. Generally, lipopolysaccharide (LPS) and
interferon (IFN)-γ are the main stimulating factors for
M1 macrophage activation, while interleukin (IL)-4
and IL-13 can activate macrophages to M2 [8, 9].
Notably, M1 macrophage activation drives
pathogenesis and progression of diabetes through
exacerbation of inflammatory responses via secretion
of inflammatory cytokines [10, 11]. For example,
Arnush et al. found M1 macrophages promoted
destruction of β cells in T1DM mice through excessive
production of IL- [12]. In addition, Tim-3
exacerbated podocyte injury via M1 macrophage
activation in streptozocin (STZ)-induced diabetic
nephropathy [13]. Importantly, blocking macrophage
infiltration into the pancreas or restraining
macrophage activation in diabetic mice maintained
pancreas function and prevented T1DM initiation [14,
15]. Therefore, elucidating the underlying mechanism
of M1 macrophage activation is likely to lead to a
better understanding of the pathogenesis and therapy
of T1DM.
Circular RNA (circRNA) is a covalently closed
loop molecule ligated by a 3′5′ phosphodiester bond
at the junction site, which is generated through back-
splicing and often considered to be a byproduct of
aberrant splicing [16, 17]. Recently, high-throughput
sequencing has been used to identify thousands of
endogenous circRNA species in mammalian cells,
which are stable, conserved, and abundant [18, 19]. By
modulation of gene expression, interaction with RNA
binding proteins (RBPs), circRNAs directly or
indirectly participate in autoimmune diseases and
cancers. For instance, circ-RasGEF1B promotes the
antigen presentation process by positively regulating
the stability of intercellular adhesion molecule 1
(ICAM-1) mRNA in macrophages exposed to LPS [20].
Additionally, circZKSCAN1 negatively regulates
cancer stem cells by physically binding fragile X
mental retardation protein (FMRP) against cell cycle
and apoptosis regulator 1 (CCAR1) complex in
hepatocellular carcinoma [21]. However, the roles of
circRNAs in regulating M1 macrophage activation
and T1DM are currently undefined.
In this study, we conducted a genome-wide
analysis of circRNA expression profiles in peripheral
blood mononuclear cells (PBMCs) from T1DM
patients and healthy controls and identified a novel
circRNA, circPPM1F, which was significantly
upregulated in the PBMCs of T1DM patients.
Functionally, circPPM1F could promote LPS-induced
M1 macrophage activation via enhancement of NF-κB
signaling. Mechanistically, circPPM1F competitively
interacted with HuR to impair the translation of
PPM1F, thus alleviating the inhibitory effect of
PPM1F on the NF-κB pathway. Moreover, we found
that EIF4A3 and FUS participated in the maintenance
of high levels of circPPM1F expression. In addition,
circPPM1F could exacerbate pancreas injury in STZ-
induced diabetic mice by activating M1 macrophages.
Overall, our study indicates that circPPM1F plays an
important role in the development of T1DM and
suggests a potential therapeutic target for T1DM.
Materials and Methods
Human study subjects
T1DM patients were diagnosed according to the
criteria of American Diabetes Association (ADA) [22].
Human peripheral blood samples were collected from
T1DM patients (8.5 ± 0.6 y) and age-matched healthy
controls (8.0 ± 1.2 y), following informed consent from
all patients, at the Children′s Hospital of Fudan
University, Shanghai, China. Detailed characteristics
of the study subjects are presented in Table S1. The
study was approved by the Research Ethics Board of
the Children′s Hospital of Fudan University [No.
(2016) 96].
PBMCs were isolated using Ficoll-Hypaque (GE
Healthcare, USA). Briefly, peripheral blood was
mixed with phosphate-buffered saline (PBS) and
overlaid on the Ficoll-Hypaque solution (density:
1.077 g/mL). Following centrifugation at 400 ×g for 30
min, PBMCs were aspirated from the interface. The
cell pellet was washed twice with PBS and
resuspended in TRIzol for subsequent RNA extraction
and quantitative real-time PCR (qRT-PCR). In
addition, CD14+, CD3+, and CD19+ cells were sorted
from PBMCs with a magnetic cell sorting system
(Miltenyi Biotec, Germany). Sorted cells were
subjected to RNA extraction and qRT-PCR.
RNA extraction and quantitative real-time
PCR
The nuclear and cytoplasmic fractions or total
RNA were extracted using TRIzol (Invitrogen, USA)
followed by reverse transcription of mRNAs and
circRNAs using PrimeScript II 1st Strand cDNA
Synthesis Kit (Takara, Japan) according to the
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10910
standard manufacturer′s instructions. A qRT-PCR
assay was performed to measure mRNAs and
circRNAs expression with SYBR® Premix Ex Taq™ II
(Takara, Japan) using the Roche 480 Real Time PCR
System. β-actin served as internal control for mRNAs
and circRNAs. Relative quantification (2ΔΔCT) was
used for results analysis. The primers sequences in
qRT-PCR are listed in Table S2.
Microarray
The circRNA microarray analysis was performed
at the Shanghai Biotechnology Corporation. In brief,
Total RNA of PBMCs from T1DM patients (n = 4) and
age-matched healthy controls (n = 4) were extracted
and purified followed by amplification and labeling
with a Low Input Quick Amp WT Labeling Kit.
Labeled cRNAs were purified using the RNeasy mini
kit. Each slide was hybridized with 1.65 μg
Cy3-labeled cRNA for 17 h using a gene expression
hybridization kit, and then scanned with an Agilent
microarray scanner using default settings. Data were
extracted with the Feature Extraction software 10.7
(Agilent Technologies, Santa Clara, CA, US). Raw
data were normalized by the Quantile algorithm and
limma package in R. Overall, 88750 circRNAs were
tested.
Cell culture and transfection
The human THP1 cell line was obtained from the
American Type Culture Collection (ATCC, Manassas,
VA, USA), and MIN6 and Raw264.7 cells were
obtained from the Fudan IBS cell resource center
(FDCC, Shanghai, China). THP1 and MIN6 cells were
maintained in RPMI-1640 medium (Gibco,
Gaithersburg, MD, USA) with 10% fetal bovine serum
(FBS, Gibco) and 1% penicillin/streptomycin (Gibco),
and the MIN6 cell culture medium was supplemented
with 50 μM 2-mercaptoethanol. Raw264.7 cells were
maintained in Dulbecco′s modified Eagle′s medium
with high glucose (DMEM, Gibco) supplemented
with 10% FBS. All cells were kept in a humidified cell
incubator with 5% CO2 at 37 °C.
Cells were transiently transfected with
Lipofectamine RNAiMAX reagent (Invitrogen, USA)
and chemically synthesized si-circPPM1F, siPPM1F,
siHuR, siEIF4A3, or siFUS (GenePharma, Shanghai,
China) according to standard protocols. Cells were
transiently transfected with 500 ng/mL ectopic
expression vector of circPPM1F with Lipofectamine
2000 reagent (Invitrogen, USA), according to the
manufacturer’s instructions. qRT-PCR assay was used
to assess RNA expression levels 48 h after
transfection. Relative sequences of siRNA are listed in
Table S3.
For the activation of THP1-derived macrophage,
THP1 cells were treated as previously described [23].
Briefly, THP1 were treated with 500 ng/mL phorbol
12-myristate 13-acetate (PMA) for 48 h to induce the
differentiation of THP1 into macrophages (THP1
macrophages), and then 200 ng/mL LPS was used to
induce M1 macrophage activation.
Western blot
Total protein was extracted using Radio
Immunoprecipitation Assay (RIPA) lysis buffer
(Thermo Scientific, USA). Lysates were resolved by
electrophoresis, transferred to polyvinylidene
fluoride (PVDF) membranes, and probed with
antibodies directed against PPM1F (Abcam); HuR,
phosphorylated NF-κB p65 (Ser536) (p-p65), p65,
phosphorylated p38 (T180/Y182) (p-p38), p38,
phosphorylated JNK (T183/Y185) (p-JNK), JNK,
phosphorylated ERK1/2 (T202/Y204) (p-ERK1/2),
ERK1/2, phosphorylated m-TOR (Ser2448)
(p-mTOR), mTOR, phosphorylated Stat3 (Y705)
(p-Stat3), Stat3, Bcl2 (Cell Signaling Technology,
USA); Bax (EMD Millipore, USA), and β-tubulin
(Abcam, USA). The bands were detected by
developing with chemiluminescent HRP substrate
(Thermo Scientific, USA), and intensity of bands was
determined by imaging with a Molecular Imager®
(Bio-RAD, ChemiDocTM XRS+ Imaging System,
USA). All results were normalized to those of
β-tubulin, which was used as a loading control.
Detailed characteristics of the antibodies are
presented in Table S4.
RNase R digestion
Four micrograms total RNA from THP1 cells
was either untreated (control) or treated with 20 units
of RNase R (Epicenter; USA, RNR07250) in the
presence of 1 × Reaction Buffer, and incubated for 30
min at 37 °C. The digested RNA was isolated using
acid phenol-chloroform (5:1). Then reverse
transcription and qRT-PCR were performed, as
described in the RNA extraction and qRT-PCR
section.
RNA stability
THP1 cells (1 × 105) were placed in 24-well plates
and treated with 250 ng/mL actinomycin D (Act D,
Sigma) added to the cell culture medium. The levels
of circPPM1F and PPM1F were detected at 0, 3, 6, 12,
and 24 h.
Subcellular fractionation and localization
Nuclear and cytosolic fractions were separated
using the Nuclear and Cytoplasmic Extraction Kit
(Cwbio, China). A total of 1 × 107 cells were harvested,
re-suspended in 1 mL of Nc-buffer A and 55 µL
Nc-Buffer B, and incubated for 20 min on ice. Cells
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10911
were then centrifuged for 15 min at 12, 000 ×g; the
resulting supernatants (containing the cytoplasmic
component) and nuclear pellets were used for RNA
extraction.
RNA immunoprecipitation
The RNA immunoprecipitation (RIP) assay was
performed using the Magna RIP RNA-Binding
Protein Immunoprecipitation Kit (EMD Millipore
Corp., Billerica MA, USA). Two 10-cm culture dishes
of THP1 macrophages (1 × 107/dish) were harvested,
centrifuged and re-suspended using 100 μL RIP lysis
buffer supplemented with protease and RNase
inhibitors, and 5 g IgG or HuR antibody-coated
beads were incubated with the cell lysates under
rotary agitation at 4 °C overnight. Following
proteinase K treatment, the immunoprecipitated
RNAs were extracted and reversely transcribed as
described in the RNA extraction and qRT-PCR
section. Levels of circPPM1F and PPM1F were
detected by qRT-PCR assay.
circPPM1F vector construction
3D5 is a modified plasmid based on the pZW1
vector. Two reverse complementary sequences helped
to form a circular structure derived from the POLR2A
gene and were inserted upstream of the restriction site
Xho I and downstream of Pac I. Primer 1 or primer 2
(Table S2) was used to amplify the sequence of
circPPM1F upstream (~340 bp) or downstream (~3800
bp) of cDNA or gDNA. Two PCR segments were
inserted into the 3D5 vector following digestion with
restriction enzymes Xho I and Pac I using a seamless
cloning assay (Figure S1), and the product was
transformed into STBL3 competent cells. The
recombinant vector sequences were validated by
Sanger sequencing.
Conditional media collection and cell
treatment
Raw264.7 cells were seeded in six-well plates,
and transfected with 3D5-circPPM1F, or
corresponding negative control plasmids according to
the cell transfection method used. Next, 48 h post
transfection, the transfected cells were stimulated
with 200 ng/mL LPS for 20 h. Culture media were
gathered and centrifuged at 3000 rpm at 4 °C for 30
min, after which the supernatants (conditional media)
were collected. The conditional media were used for
treatment of MIN6 cells, at a ratio of 3:1 with complete
medium.
Enzyme-linked immunosorbent assay (ELISA)
The protein levels of IL-6 and TNF-α in the
conditional media were measured with the mouse
IL-6 and TNF-α DuoSet ELISA kit (eBioscience)
according to the manufacturer’s instructions. A
microplate reader (Synergy4; BioTek, Winooski, VT,
USA) was used to read the absorbance at 450/570 nm.
Cell proliferation assay
The CCK8 assay was performed to assess the
proliferative ability of MIN6 cells according to the
manufacturer’s instructions (DOJINDO Molecular
Technologies, Inc., Kumamoto, Japan). Briefly, MIN6
cells (1.8 × 103) were placed in 96-well plates and
cultured in conditional medium. Each sample was
assayed in triplicate. Cell viability was determined at
0, 24, 48, and 72 h using 10 μL CCK8 solution
treatment for 2 h. The optical density of each well was
assessed using a Microplate reader (Synergy4; BioTek,
Winooski, VT, USA) at 450 nm.
Cell apoptosis assay
MIN6 cells were cultured in six-well plates (1 ×
105 cells/well) for 48 h using conditional media,
followed by H2O2 (500 μΜ in FBS-free medium)
treatment for 15 h, and then pooled together after
trypsin without EDTA digestion. Cell apoptosis was
analyzed using the Annexin V-FITC/Propidium
Iodide (PI) Apoptosis Detection Kit (BD Pharmingen,
New York, USA, #556547) according to the
manufacturer’s instructions. MIN6 cells were stained
with FITC and PI and then analyzed by
fluorescence-activated cell sorting using FACS Canto
II (BD Biosciences, San Jose, CA, USA). The cell
apoptosis data were analyzed by FlowJo V10 software
(Tree Star, San Francisco, CA, USA). Cells of quadrant
4 that were considered viable were FITC Annexin V
and PI negative; cells of quadrant 3 that were in early
apoptosis were FITC Annexin V positive and PI
negative; cells of quadrant 2 that were in late
apoptosis were both FITC Annexin V and PI positive;
and cells of quadrant 1 that were necrotic were FITC
Annexin V negative and PI positive. The percentages
of cell apoptosis were the sum of that from early
apoptosis and late apoptosis.
Animal studies in the STZ-induced diabetic
mouse model
C57BL/6 mice (8-week-old, male) were
purchased from Shanghai Slac Laboratory Animal Co.
Ltd. All mice were housed at the Experiment Animal
Center of Childrens Hospital of Fudan University at
room temperature 22 °C under a 12:12 h light/dark
cycle, and were provided with rodent chow and tap
water. The mice were randomly divided into four
experimental groups: Control, STZ, STZ+pZW1, and
STZ+circPPM1F group. On day 0, STZ+circPPM1F
and STZ+pZW1 group mice were intraperitoneally
(i.p.) injected with circPPM1F plasmid (8 μg/mouse,
150 μL) and an equal amount of pZW1 using the
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10912
Entranster™-in vivo Transfection Reagent (Engreen,
China), respectively; control and STZ group mice
were injected with PBS. From day one, STZ,
STZ+pZW1, and STZ+circPPM1F group mice were
i.p. injected with repeated low doses of STZ (50
mg/kg body weight/day, 300 μL/mouse for five
consecutive days); control group mice were injected
with sodium citrate buffer (Fankew, FK4006). On day
seven, mice were injected circPPM1F, control
plasmids, or PBS again, and then received two
injections per week. The animal experiments were
repeated independently 3 times.
Body weight was measured weekly from day
one. Blood glucose was also detected weekly from the
tail vein blood using a Roche glucose reader (Roche
Diagnostics GmbH, Germany). All mice were fasted
for 12 h before glucose detection. On day 29, mice
were sacrificed and pancreas tissues dissected and
analyzed for pathology.
This project complied with the institutional
guidelines and laws for the care and use of laboratory
animals. The study was approved and overseen by
the Animal Studies Committee of the Childrens
Hospital of Fudan University [No. (2016) 96].
Immunohistochemistry and
Immunofluorescence studies
Pancreas tissues were fixed with 4%
paraformaldehyde and then embedded in paraffin
and cut into slices. For immunohistochemical
analysis, a portion of paraffin sections were routinely
stained with hematoxylin and eosin (H&E), while
others were incubated with antibodies against Ki-67
(Signalway Antibody, USA), Insulin (Proteintech,
USA), and F4/80 (Cell Signaling Technology, USA).
Islets from consecutive tissue cross-sections were
photographed at identical exposure conditions and
magnification (400×) using a microscope (Leica). For
immunofluorescence analysis, paraffin slides of 3-4
μm were prepared, and then incubated with rabbit
anti-mouse F4/80 and rabbit anti-mouse iNOS
(Servicebio, China). FITC (green) and Cy3
(red)-conjugated goat anti-rabbit IgG (Servicebio)
were used to visualize F4/80 and iNOS, respectively.
DAPI (4′, 6-diamidino-2-phenylindole) was used to
stain the cell nuclei (blue). Images were captured with
a fluorescence microscope (Leica). Detailed
characteristics of the antibodies used are presented in
Table S4.
Preparation of pancreas single cell suspensions
and flow cytometry
Pancreas tissues were collected into the
gentleMACS C Tubes (#130-093-237; Miltenyi Biotec,
Germany) containing the enzyme mix of the Multi
Tissue Dissociation Kit 1 (#130-110-201, Miltenyi
Biotec,Germany) in serum-free RPM-1640 and cut into
2 × 4-mm pieces. Tissues were subjected to the run
program Multi_37C_m (30 min) on the gentleMACS
Octo Dissociator with Heaters (#130-096-427, Miltenyi
Biotec, Germany). The cell suspensions were passed
through a 70-μm MACS SmartStrainer (#130-098-462,
Miltenyi Biotec, Germany), followed by centrifuging
at 300 ×g for 7 min at 4 °C. All isolated cells were
suspended in 100 μL iced PBS supplemented with 2%
FBS. Cells were then counted with trypan blue and
processed for flow cytometry in as indicated below.
For surface marker analysis, cells were stained
with anti-mouse F4/80 (eBioscience, USA) for 30 min
at 4 °C. For intracellular cytokine staining, cells were
fixed and permeabilized and labeled with anti-mouse
iNOS (eBioscience, USA) after anti-F4/80 staining.
The concentration of each antibody was used
according to the recommended product protocol.
Cells were examined by flow cytometry using a BD
FACSCanto II instrument (BD Biosciences, San Jose,
CA, USA) and analyzed with FlowJo V10 software
(Tree Star, San Francisco, CA, USA). Detailed
characteristics of the antibodies used are presented in
Table S4.
Statistical analysis
Results from three independent experiments are
expressed as mean ± standard error of the mean
(SEM). The two-tailed Student’s t-tests were used for
comparisons between two groups, and one-way
analysis of variance (ANOVA) was used for
multifactorial comparisons. A p-value of < 0.05 was
considered statistically significant. The relationship
between circPPM1F and IL-6, IL-1β, TNF-α, EIF4A3 or
FUS was tested using Pearson’s correlation and linear
regression. Statistical analyses were performed with
SPSS v.19.0 software or GraphPad Prism 7.0 software.
Results
circPPM1F was upregulated in PBMCs from
children with type 1 diabetes mellitus
To identify differentially expressed circRNAs in
T1DM, we first analyzed circRNA transcript profiles
of PBMCs from T1DM children (n = 4) and
age-matched healthy controls (n = 4) by circRNA
microarray. Using a two-fold change and p < 0.05 as
the threshold to define up- or down-regulated
circRNAs, we identified 27 upregulated and 31
downregulated differentially expressed circRNAs in
T1DM patients compared with healthy controls
(Figure 1A). We analyzed the composition of the
differentially expressed circRNAs in light of the
positions of circRNAs in the transcripts; the profile
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10913
consisted of 27 exonic circRNAs (47%), three intronic
circRNAs (5%), two exonic-intronic circRNAs (3%), 21
exonic-UTR circRNA (36%), and others (9%) (Figure
1B). Next, we evaluated the expression of 27
upregulated circRNAs in PBMCs from children with
T1DM and healthy control subjects in an expanded
cohort. The results showed that only hsa_circ_0062444,
hsa_circ_0009718 and hsa_circ_0060450 were detectable
and significantly upregulated in PBMCs from T1DM
patients compared with healthy controls (Figure 1C).
Furthermore, higher expression levels of IL-6, IL-1β,
and TNF-α were also observed in T1DM patients
(Figure 1D). Importantly, the positive correlation
between hsa_circ_0062444 and IL-6, IL-or TNF-α was
validated in human patients with T1DM, while no
correlations between the hsa_circ_0009718 or
hsa_circ_0060450 and IL-6, IL- or TNF-α were
identified (Figure 1E and Figure S2A). Furthermore,
the ability of hsa_circ_0062444 to differentiate T1DM
patients from healthy subjects was assessed by
receiver operating curve (ROC) analysis, which
yielded an area under the curve of 0.839 (Figure 1F).
Together, these findings implied that hsa_circ_0062444
might have important role in T1DM pathogenesis.
Next, bioinformatics analysis showed that
hsa_circ_0062444 was 4291 nt long and was the
back-spliced circular product of the last three exons of
the PPM1F transcript; thus, as an exonic circRNA, it
was named as circPPM1F (Figure 1G). To assess the
role of circPPM1F in the regulation of subsets of
PBMCs, we separated human PBMCs into T cells, B
cells, and monocytes to detect the expression levels of
circPPM1F. Surprisingly, circPPM1F was mainly
expressed in monocytes rather than T and B cells
(Figure S2B). Considering monocytes can transform
into macrophages in inflammatory tissues and
circPPM1F was closely associated with inflammatory
cytokines in T1DM patients, we subsequently focused
on the regulatory role of circPPM1F in LPS-induced
M1 macrophage activation.
circPPM1F promoted M1 macrophage
activation through enhancement of NF-κB
signaling
To verify whether circPPM1F was a truly
circRNA and not a linear RNA, we performed
RT-PCR and Sanger sequencing assays. A strict
concordance between the sequencing results and the
public circPPM1F sequence in circBase (http://www.
circbase.org/cgi-bin/simplesearch.cgi) was observed
(Figure 2A). Furthermore, we found the endogenous
expression of circPPM1F was resistant to excessive
ribonuclease R (RNase R) digestion while linear
mRNAs were severely degraded (Figure 2B).
Additionally, the stability of circPPM1F after Act D
treatment in THP1 macrophages was examined. We
found that circPPM1F was highly stable, with a
half-life > 24 h, whereas PPM1F mRNA was readily
degraded and had a half-life < 6 h (Figure 2C).
Collectively, this evidence suggested that endogenous
circPPM1F was truly a circular RNA.
To determine the functional influence of
circPPM1F on M1 macrophage activation, we first
knocked down the expression of circPPM1F in
THP1-derived macrophages with two small
interfering RNAs (siRNAs) targeting the circPPM1F
junction site; the knockdown efficiency was
confirmed by qRT-PCR and si-circPPM1F-2 was
selected for the later study (Figure 2D). We found the
expression of M1 macrophage associated genes (IL-1β,
TNF-α and CXCL10) was downregulated in the cells
with circPPM1F knockdown after LPS stimulation
(Figure 2E). Meanwhile, the IL-6 and TNF-α protein
expression were suppressed in the supernatant
(Figure 2F). Further, in the “gain-of-function” studies
with ectopically expressing circPPM1F, we found the
expression of IL-1β, TNF-α, and CXCL10 were
upregulated following LPS stimulation (Figure 2G),
concomitant with significantly enhanced IL-6 and
TNF-α protein levels in the supernatant (Figure 2H).
Taken together, these findings suggested that
circPPM1F can promote M1 macrophage activation.
Upon activation, LPS-TLR4 augments
macrophage activity through the production of
inflammatory cytokines, and activation of NF-κB and
MAPK pathways [24]. To explore signaling pathways
involved in circPPM1F regulation of M1 macrophage
activation, we investigated the effects of circPPM1F on
NF-κB and MAPK pathways. Intriguingly, western
blotting showed that circPPM1F knockdown did
indeed result in markedly reduced levels of
phosphorylated p65 in THP1 macrophages, whereas
ectopic expression of circPPM1F significantly
increased the levels of phosphorylated p65. However,
THP1 macrophages with circPPM1F knockdown
exhibited no differences in phosphorylation levels of
JNK, p38, and ERK (Figure 2I-J). Overall, these
findings implied that circPPM1F can promote M1
macrophage activation by enhancing NF-κB signaling.
circPPM1F inhibited PPM1F translation
through competitively binding to HuR
PPM1F, a member of the PP2C family of Ser/Thr
protein phosphatases, is a negative regulator of the
IKK-NF-κB pathway through its effects on the
dephosphorylation of TAK1 [25]. As circPPM1F is the
back-spliced circular product of the coding gene,
PPM1F, and circRNAs have been reported to exert
regulatory effects on host genes at both transcriptional
and post-transcriptional levels [26, 27], we questioned
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10914
whether the effects of circPPM1F on NF-κB signaling
activation were due to its influence on PPM1F
expression. To verify our hypothesis, the levels of
PPM1F protein and mRNA expression were
determined in THP1 macrophages following
circPPM1F knockdown. Surprisingly, knockdown of
circPPM1F resulted in a significant increase of PPM1F
protein expression, with no changes in its mRNA
expression (Figure 3A-B), indicating that circPPM1F
negatively regulated the expression of PPM1F at the
translational stage. Furthermore, we knocked down
the expression of PPM1F in macrophages and found
phosphorylation of p65 and the expression of IL-6,
and CXCL10 were increased following LPS
stimulation (Figure 3C-E). Taken together, these data
implied that circPPM1F suppressed PPM1F
translation, thereby facilitating NF-κB pathway and
M1 macrophage activation.
Figure 1. circPPM1F is upregulated and is associated with inflammatory cytokines in peripheral blood mononuclear cells from type 1 diabetes mellitus
patients. A. Heatmap showing 27 upregulated and 31 downregulated differentially expressed circRNAs in peripheral blood mononuclear cell (PBMC)s of type 1 diabetes
mellitus (T1DM) patients (n = 4) and age-matched healthy controls (n = 4) (fold change > 2.0, p < 0.05). B. Composition of the circRNAs according to the position of the gene
in the transcript. C, D. Analyses of the expression levels of hsa_circ_0062444, hsa_circ_0009718, hsa_circ_0060450, IL-6, IL-1β, and TNF-α (normalized to β-actin) in PBMCs from
43 T1DM patients and 45 healthy controls, as determined by qRT-PCR. E. Correlation analysis of the expression of hsa_circ_0062444 and IL-6, IL-1β or TNF-α in 43 T1DM
patients (Pearson’s correlation). F. Receiver operating curve (ROC) analysis of circPPM1F levels in the study population. G. Schematic illustration showing the location of
hsa_circ_0062444 in host gene PPM1F. Data are presented as mean ± SEM. *p ≤ 0.05, ***p ≤ 0.001.
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10915
Figure 2. circPPM1F promotes M1 macrophage activation by enhancing NF-κB signaling. A. The location of circPPM1F in the PPM1F transcript was validated by
Sanger sequencing. B. Total RNA was digested with or without RNase R, followed by quantitative real-time PCR (qRT-PCR) measurements of circPPM1F, PPM1F, MALAT1, and
GAPDH. C. The stability of circPPM1F was detected by qRT-PCR in THP1 macrophages after actinomycin D (Act D) treatment. D. qRT-PCR analysis of circPPM1F expression
levels in THP1 macrophages following circPPM1F knockdown by two distinct siRNAs. E. qRT-PCR analyses of IL
1β, TNF
‐α
, and CXCL10 in THP1 macrophages with conditional
treatment. Mock-, untransfected, and unstimulated cells; mock+, LPS stimulated alone cells; si-scramble, LPS-stimulated cells following transfection with si-scramble;
si-circPPM1F-2, LPS-stimulated cells following transfection with si-circPPM1F-2. F. ELISA analyses of secreted cytokine levels in THP1 macrophages with circPPM1F knockdown,
followed by LPS treatment. G. qRT-PCR analyses of circPPM1F in THP1 macrophages with 3D5-circPPM1F or pZW1 transfection (left); M1-associated gene expressions in LPS
stimulated THP1 macrophages overexpressing circPPM1F were quantified by qRT-PCR analysis (right). H. ELISA analyses of secreted cytokine levels in circPPM1F-overexpressed
THP1 macrophages, followed by LPS treatment. I. Western blot showing total p65, p38, ERK1/2, JNK and their phosphorylation levels in THP1 macrophages with or without
circPPM1F knockdown after LPS treatment. J. Western blotting analysis to evaluate levels of total p65, phosphorylated p65 in circPPM1F-overexpressed THP1 macrophages. The
levels of p-p65 were normalized to that of β-tubulin and quantified using Image J software. Data are presented as mean ± SEM from three independent experiments. *p 0.05,
**p 0.01, ***p 0.001, ns indicates no significance.
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10916
Figure 3. circPPM1F competitively binds to HuR to impair PPM1F translation. A. Western blotting analysis to evaluate levels of PPM1F in circPPM1F knocked
down-THP1 macrophages. B. qRT-PCR analyses of PPM1F expression in THP1 macrophages with circPPM1F knockdown. C. Analysis of PPM1F expression in THP1 macrophages
transfected with PPM1F siRNA (200 nM) or control siRNA by qRT-PCR. D. Western blot analysis of PPM1F, p65, and p-p65 in THP1 macrophages with PPM1F knockdown,
followed by LPS stimulation. E. Quantitative real-time PCR (qRT-PCR) analysis of M1-associated gene expression in THP1 macrophages with PPM1F knockdown, followed by LPS
stimulation. F. qRT-PCR results showing the distribution of circPPM1F in the cytoplasmic and nuclear fractions of THP1 macrophages. GAPDH as cytoplasm control transcript, and
U1 as nuclear control transcript. G. Putative HuR binding sites within circPPM1F full-length sequence. H. The enrichment levels of circPPM1F and PPM1F in the products of the
RNA immunoprecipitation (RIP) assay (HuR IP compared with IgG IP) as detected by qRT-PCR. I. qRT-PCR analysis of HuR in THP1 macrophages transfected with HuR siRNA
(200 nM) or control siRNA. J. Protein levels of PPM1F and HuR were detected by western blotting in THP1 macrophages with HuR knockdown. K. qRT-PCR analysis of
circPPM1F and PPM1F in THP1 macrophages with HuR knockdown. The levels of PPM1F, p-p65 and HuR were normalized to those of β-tubulin and quantified using Image J
software. Data are presented as mean ± SEM from three independent experiments. *p ≤ 0.05, **p 0.01, ***p ≤ 0.001, ns indicates no significance.
Subcellular localization contributes to the
modulatory mechanism of circRNAs on their targets.
To further explore the mechanism underlying
circPPM1F-mediated effects on PPM1F, we performed
subcellular fractionation and localization assays.
Surprisingly, in contrast to the cytoplasmic
localization observed for a large number of verified
exonic circRNAs, circPPM1F was primarily localized
to the nucleus of THP1 macrophages (Figure 3F).
circRNAs in the nucleus are reported to regulate gene
translation via interaction with RNA-binding proteins
(RBPs). We therefore searched for all putative RBPs
binding to circPPM1F using the circRNA interactome
database (https://circinteractome.nia.nih.gov/
RNA_Binding_Protein/rna_binding_protein.html).
Notably, the analysis revealed that the RBP containing
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10917
the most potential binding sites with the circPPM1F
was HuR (Figure 3G). HuR, a member of the ELAVL
family of RBPs and selectively bind to AU-rich
elements (AREs) within the 3′-untranslated regions
(UTRs) of mRNA, stabilizing ARE-containing mRNA
to regulate gene translation [28]. To determine
whether the interaction between circPPM1F and HuR
was responsible for circPPM1F-mediated translational
inhibition of PPM1F, we conducted RIP using an
antibody specific for HuR and found a 5-fold
enrichment of circPPM1F and 30-fold enrichment of
PPM1F when the anti-HuR antibody was used,
relative to use the IgG control (Figure 3H). These
results thus implied a direct interaction of HuR with
circPPM1F or PPM1F.
Next, we explored whether HuR is involved in
the effects of circPPM1F on PPM1F translation; thus,
siRNA directed at HuR was transfected to knock
down HuR expression in THP1 macrophages. The
knockdown of HuR resulted in a significant decrease
in protein expression of PPM1F in THP1 macrophages
(Figure 3I-J), whereas no changes in circPPM1F and
PPM1F mRNA expression were detected (Figure 3K).
Collectively, these studies strongly suggested that
circPPM1F could suppress PPM1F translation through
competitive interaction with HuR, which
subsequently promotes activation of M1
macrophages.
EIF4A3 and FUS coordinately regulated
circPPM1F expression
Next, we explored the biogenesis of circPPM1F.
Previous studies have shown that the biogenesis of
circRNA depends on the spliceosomal machinery
containing with cis- and trans-regulatory elements.
Reverse intronic complementary sequences (ICS)
induced “head-to-tail” splicing by bringing the 5′- and
3′-termini of an exon or of consecutive exons into
spatial proximity [29, 30]. However, there were no ICS
in either of the intronic flanking regions of the splice
Figure 4. EIF4A3 and FUS cooperatively regulate circPPM1F expression. A, B. Bioinformatic prediction of binding sites of EIF4A3 (A) and FUS (B) on circPPM1F. C.
Expression levels of circPPM1F, EIF4A3, and FUS were detected by quantitative real-time PCR (qRT-PCR) in THP1 macrophages treated with LPS. D. qRT-PCR analyses of the
expression levels of EIF4A3 and FUS in PBMCs from 43 T1DM patients and 45 healthy controls. E. Correlation analysis of the expression of circPPM1F and EIF4A3, or FUS levels
in patients with type 1 diabetes mellitus (T1DM) (Pearson’s correlation). F. qRT-PCR was used to measure the expression levels of EIF4A3 and FUS in THP1 macrophages with
EIF4A3 or FUS knockdown. G. qRT-PCR analyses of the expression levels of circPPM1F in EIF4A3 or FUS
knocked down THP1 macrophages with or without lipopolysaccharide
(LPS) treatment. Data are presented as mean ± SEM from three independent experiments. **p ≤ 0.01, ***p ≤ 0.001, ns indicates no significance.
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10918
junction sites of circPPM1F. Therefore, RBPs might be
responsible for expression of circPPM1F. Using a
bioinformatics method (https://circinteractome.nia.
nih.gov/index.html), we queried all RBPs that were
reported to be involved in circRNA generation. The
result revealed that a binding site for EIF4A3 is
present in the downstream region of the circPPM1F
transcript, and FUS-binding sites were identified in
the mature circPPM1F (Figure 4A-B). We then
measured the levels of circPPM1F, EIF4A3, and FUS
expression over time in THP1 macrophages
stimulated by LPS. Interestingly, FUS exhibited a
time-dependent increased expression following LPS
treatment, while EIF4A3 was upregulated at 3 h after
stimulation, dropped to a basic level at 6 h, and
thereafter was gradually upregulated. circPPM1F was
downregulated at 3 h, but afterward its expression
appeared to be gradually upregulated (Figure 4C).
Furthermore, we analyzed the levels of EIF4A3 and
FUS expression in PBMCs from T1DM patients and
healthy controls. The results indicated that the
expression level of EIF4A3 was significantly
decreased, whereas that of FUS was increased in
T1DM patients compared with healthy controls
(Figure 4D). Importantly, there was a negative
correlation between EIF4A3 and circPPM1F, while a
positive correlation was observed between FUS and
circPPM1F (Figure 4E). The above results implied that
EIF4A3 and FUS may play important roles in
circPPM1F expression.
To further investigate the roles of EIF4A3 and
FUS in circPPM1F expression, we knocked down
EIF4A3 and FUS expression in THP1 macrophages by
transfection with their individual siRNAs (Figure 4F).
Surprisingly, knockdown of EIF4A3 or FUS resulted
in no differences in the expression level of circPPM1F
in THP1 macrophages without LPS treatment.
However, knockdown of EIF4A3 significantly
elevated the expression level of circPPM1F, while
knockdown of FUS decreased its level in THP1
macrophages treated with LPS (Figure 4G). Overall,
these results implied that EIF4A3 and FUS
coordinately regulate circPPM1F expression during
M1 macrophage activation.
circPPM1F induced pancreatic β-cell apoptosis
by promoting M1 macrophage activation in
vitro
To assess the pathological effects of circPPM1F-
induced M1 macrophage activation on pancreatic
β-cells, we performed a cell co-culture assay with
Raw264.7 cells overexpressing circPPM1F and murine
pancreatic β-cells MIN6. Consistent with the mRNA
levels, the IL-6 and TNF-α protein levels were
enhanced in conditional media from Raw264.7 cells
overexpressing circPPM1F (Figure 5A). Notably, we
found that the proliferation rate of MIN6 cells was
significantly inhibited by the conditional media
(Figure 5B). In addition, the media markedly
enhanced the rate of apoptosis in MIN6 cells (Figure
5C). Previous studies have indicated that mTOR and
MAPK pathways are crucial for pancreatic β-cell
Figure 5. circPPM1F induces pancreatic β cell apoptosis through M1 macrophage activation. A.
from circPPM1F-overexpressed Raw264.7 cells, followed by LPS treatment. B.
CCK-8 assay. C. After the incubation with conditional media, MIN6 cells were stained with Annexin V-FITC and propidium iodide (PI) before fluorescence analys
cytometry. The percentage of cells in the four different quadrants was calculated and the results presented in different hist
Annexin V+/PI- and Annexin V+/PI+. D. Western blotting analysis to detect expression levels of Bcl2, Bax, p38, p-p38, JNK, p-JNK, ERK1/2, p-ERK1/2, mTOR, and p-
MIN6 cells cultured with conditional media. Data are presented as means ± SEM from three independent experiments. **p ≤ 0.01, ***p ≤ 0.001.
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10919
apoptosis [31, 32]. To investigate the potential
molecular mechanisms inducing MIN6 cell apoptosis,
we confirmed the effects of the conditional media on
mTOR and MAPK pathways in MIN6 cells via
western blotting. Decreased expression levels of Bcl2
and elevated Bax were observed in MIN6 cells
cultured with media from circPPM1F overexpressing
Raw264.7, and this was accompanied by increased
levels of p38 and JNK phosphorylation (Figure 5D).
However, there were no apparent changes in the
phosphorylation levels of mTOR and ERK (Figure
5D). Collectively, these data suggested that
circPPM1F-mediated M1 macrophage activation
could induce pancreatic β cell apoptosis through the
MAPK pathway in vitro.
circPPM1F exacerbated pancreas injury in
STZ-induced diabetic mice through M1
macrophage activation
To assess the role of circPPM1F in pancreas
injury of diabetic mice, we generated a diabetic mouse
model using intraperitoneal (i.p.) injection of STZ
(Figure 6A). We first investigated the dynamic
expression profiles of circPPM1F in the pancreas,
liver, spleen, and PBMCs of mice injected with
circPPM1F or pZW1 plasmid for 3, 5, and 7 days. The
results showed that the level of circPPM1F expression
was highest in pancreas compared with that in the
liver, spleen and PBMCs, and peaked on day 5 (Figure
S3A). Next, successful induction of the diabetic mouse
model was evidenced by significant weight loss one
week after initial injection of STZ, and hyperglycemia
two weeks after the injection. Importantly, the
STZ+circPPM1F group mice achieved more severe
weight reduction and hyperglycemia than STZ or
STZ+pZW1 treated mice groups (Figure 6B). Further,
the levels of circPPM1F and insulin expression in
pancreas tissues were measured. Mice in the
STZ+circPPM1F group displayed significantly
enhanced circPPM1F and decreased insulin expression
in the pancreas compared with the other three groups
(Figure 6C). Furthermore, we assessed the effects of
circPPM1F on pancreatic injury. The expression levels
of inflammatory cytokines, oxidative stress indicators,
indicators related to insulin secretion and cell
apoptosis were detected. Specifically, STZ+circPPM1F
group mice exhibited significantly higher levels of
IL-6, IL-1β, iNOS, and Bax expression, accompanied by
lower levels of Sod, Cat, GSH-Px, Glut2, Gck, and Bcl2
expression, relative to control, STZ, and STZ+pZW1
groups (Figure S3B). Similar to the results in Bcl2 and
Bax at the mRNA level, decreased levels of Bcl2 and
enhanced Bax protein were detected in mice of the
STZ+circPPM1F group compared with those of the
STZ+pZW1 group (Figure S3C).
Pancreatic dysfunction usually represents
pathological changes in the diabetic pancreas.
Notably, Histological analysis showed that STZ and
STZ+pZW1 groups displayed distinctly abnormal
islets structure as compared with the control group,
appearing as small islets, inhomogeneous islet cells,
and cytoplasmic vacuolation. Specifically,
STZ+circPPM1F group mice exerted more severe
damage of islets. Furthermore, Ki-67 staining
indicated decreased proliferation of pancreas cells in
the STZ and STZ+pZW1 groups, and a greater
decrease in the STZ+circPPM1F group, compared
with the control group. In addition, similar to the
mRNA expression patterns observed for insulin in the
experimental mice, protein levels of insulin were
significantly lower in mice overexpressing circPPM1F
(Figure 6D). Altogether, these findings indicated that
circPPM1F could aggravate the pancreas injury of
STZ-induced diabetic mice.
Subsequently, we wanted to examine the
molecular mechanism of pancreas injury. Considering
that the Stat3, MAPK, and Akt-mTOR pathways play
a prominent role in oxidative stress, inflammatory
response, and cell apoptosis [33, 34], we measured the
levels of Stat3, p38/JNK, mTOR and their
corresponding phosphorylated protein expression.
Importantly, the results showed that the mice of the
STZ+circPPM1F group exhibited significantly
enhanced levels of phosphorylated Stat3, p38, and
JNK expression compared with control, STZ and
STZ+pZW1 groups. However, there were no
differences in the phosphorylation levels of mTOR
(Figure 6E). Therefore, these data suggested that
circPPM1F could increase pancreas injury by
activating MAPK and Stat3 pathways in vivo.
In order to further evaluate effects of circPPM1F
on macrophage activation in pancreas islets of
diabetic mice, we assessed the infiltration of
macrophages into islets cells. Compared with control
mice, increased levels of F4/80+ cells were observed in
pancreas islets in STZ and STZ+pZW1 group mice,
and the highest levels of F4/80+ cells were detected in
the STZ+circPPM1F group (Figure 6F). Moreover, to
demonstrate whether circPPM1F promoted
infiltration of macrophages into the islet cells due to
M1 macrophage activation, we detected levels of
F4/80+/iNOS+ cells by immunofluorescence staining.
Notably, elevated levels of F4/80+/iNOS+ cells were
observed in pancreas islets of mice from the
STZ+circPPM1F group compared with that from mice
with or without STZ treatment (Figure 6G). In
addition, the levels of M1 macrophage activation in
the pancreas were validated by flow cytometry.
Similar to the positive patterns of F4/80 and iNOS
observed in immunohistochemistry (IHC) and
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10920
immunofluorescence assays, the STZ+circPPM1F
group mice displayed higher frequencies of
macrophages (F4/80+) and M1 macrophages
(F4/80+/iNOS+) in comparison to the other groups
(Figure 6H). Taken together, these findings implied
that circPPM1F could facilitate injury of pancreatic
islets in diabetic mice by promoting M1 macrophage
activation.
Figure 6. circPPM1F facilitates pancreatic islet injury in diabetic mice through M1 macrophage activation. A. Treatment of circPPM1F in the STZ-induced diabetic
mouse model (15 mice per group). B. Mean weekly body weight (left) and fasting blood glucose (right) change in four-group mice. C. Levels of circPPM1F and insulin in pancreas
tissues were detected by quantitative real-time PCR (qRT-PCR). D. Representative hematoxylin and eosin (H&E)-stained pancreas tissues, immunohistochemistry (IHC) images
of Ki-67 and insulin expression in pancreatic islets from experimental mice. Scale bar indicates 50 µm. Semi-quantification of Ki-67 and insulin staining of per islet were done by
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10921
using Image J software. E. The levels of total p38, JNK, Stat3, mTOR, and their corresponding phosphorylated forms in pancreas tissues from experimental models were
quantified by western blot. F. Representative IHC images of F4/80 expression in pancreatic islets from experimental mice. Scale bar indicates 50 µm. Semi-quantification of F4/80
staining of per islet were done by using Image J software. G. Immunofluorescence staining of infiltrated F4/80+/iNOS+ M1 macrophages in mice pancreatic islets. Green represents
anti-F4/80 Ab; red represents anti-iNOS Ab; yellow represents F4/80 and iNOS merged; blue represents DAPI. Scale bar indicates 50 µm. H. The percentages of M1
macrophages in pancreas tissue cells from STZ-treated mice with or without circPPM1F overexpression and control mice were determined by flow cytometry (left).
Quantification analyses of macrophages (F4/80+) and M1 macrophages (F4/80+/iNOS+) in pancreas tissue cells (right). *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001.
Discussion
In this study, we showed for the first time that
circPPM1F was overexpressed in PBMCs from T1DM
patients, and it increased M1 macrophage activation
through the circPPM1F-HuR-PPM1F-NF-κB axis.
Moreover, EIF4A3 and FUS were critical for
coordinately controlling circPPM1F expression. In
addition, circPPM1F could accelerate pancreatic islet
cell apoptosis in diabetic mice by promoting the
activation of M1 macrophages. Thus, we speculate
that circPPM1F might potential represent a new
therapeutic target for T1DM, adding a new dimension
to the functional importance of circRNA regulation in
diabetes mellitus.
Exonic circRNAs are usually abundant in the
cytoplasm and function mainly through the
microRNA “sponge” mechanism [35]. In contrast,
intron-containing circRNAs (intronic circRNAs and
exon-intron circRNAs), in general, are enriched in the
nucleus and are involved in regulation of host gene
expression [36]. However, Errichelli et al. found that
three completely spliced exonic circRNAs were
almost exclusively located to the nucleus [37].
Consistent with this finding, in our study, as an
exonic circRNA, circPPM1F was also constitutively
expressed in the nucleus rather than in the cytoplasm
of THP1 macrophages. Among the predicted RBPs of
circPPM1F, we found 14 binding sites for fragile X
mental retardation 1 (FMR1, also known as FMRP)
were present in circPPM1F. Interestingly, FMRP
participated in RNA trafficking from the nucleus to
the cytoplasm, displaying different subcellular
distribution due to alternative splicing [38, 39]. We
speculated that FMRP might be responsible for the
nuclear and cytoplasmic localization of circPPM1F. It
would be helpful to investigate the interaction of
FMRP with circPPM1F to better understand the
nuclear location of circPPM1F.
Recently, circRNAs have attracted increasing
attention for their potential roles in regulating
parental gene expression [26, 27]. Meanwhile,
parental genes may also be involved in circRNA
biosynthesis [40]. PPM1F has been reported to
regulate cancer cell growth and metastasis [41],
whereas its roles in the pathogenesis of T1DM and M1
macrophage activation remain unclear. Notably, our
findings provide evidence supporting the hypothesis
that circPPM1F-mediated M1 macrophage activation
may be attributable to reduced protein levels of the
PPM1F gene. Recently, HuR is a well-studied RBP
that positively augments stability of a number of
linear mRNAs and ncRNAs, but also binds to introns
of pre-mRNAs to modulate splicing [28]. It has been
revealed that the interaction of HuR and circPABPN1
impaired the normal interaction of HuR’s with linear
mRNAs, especially parental pre-mRNA of
circPABPN1, which consequently suppressed the
production of PABPN1 protein [42]. Importantly, our
data provide further evidence that HuR is a key
contributor to circPPM1F and PPM1F interactions.
Instead of impacting on circPPM1F transcription, the
interaction with HuR and circPPM1F prevented HuR
binding to PPM1F mRNA, resulting in a reduction of
PPM1F translation. Such a relationship between
circRNA and host mRNA is conceptually intriguing,
and it might be generalizable to other RBPs.
Additionally, our study was the first to reveal the role
of PPM1F in M1 macrophage activation, implying that
the circPPM1F-HuR-PPM1F axis may represent a
novel potential therapeutic target in T1DM.
RBPs are required for regulation of the
biogenesis specific circRNAs in a positive or negative
way. In this study, our data showed that EIF4A3 and
FUS oppositely regulated circPPM1F expression in
response to external stimuli. EIF4A3, a member of the
DEAD box protein family, has been implicated in
nuclear and mitochondrial splicing, ribosome and
spliceosome assembly, and translation initiation.
Recent studies have revealed that EIF4A3 promoted
circMMP9 and circSEPT9 expression via binding to
parental pre-mRNAs [43, 44]. However, in our study
we found that EIF4A3 could inhibit circPPM1F
expression. Unexpectedly, we identified a binding site
for EIF4A3 in the downstream region (i.e., at intron 6
of PPM1F transcript) of circPPM1F. Therefore, we
assume that this unusual binding site might be
responsible for the suppression of EIF4A3 on
circPPM1F biogenesis. In addition, Errichelli et al.
found FUS either increased or repressed circRNA
biogenesis by binding to the introns flanking the
back-splicing junctions, and the interaction machinery
could control a complex interplay between linear and
back-splicing [37]. In contrast to back-splicing
regulation, our data indicated that FUS might also
bind to mature circPPM1F at the post-transcriptional
level and positively mediate its expression upon
stimulation. Overall, these findings demonstrated that
diverse RBPs participate in circRNA biogenesis, and
that some may contribute to diverse regulatory
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10922
mechanisms, even antagonistically. Hopefully, our
study will prompt future work exploring RBP-based
diverse regulation of circRNA biogenesis.
STZ is a broad-spectrum antibiotic possessing
antitumor, oncogenic, and diabetogenic properties,
and multiple small dose injections of STZ in mice
produce pancreatic insulitis, with progression to
nearly complete β-cell destruction and diabetes
mellitus [45]. Although the timing and appearance of
the inflammatory islet lesions do not demonstrate that
STZ stimulation acts by initiating a cell-mediated
immune reaction, multiple low doses of STZ have
been shown to selectively destruct β-cells, which in
turn induces immune reactions against pancreatic
islets, leading to β-cell apoptosis and subsequently
diabetes mellitus. This model resembles the key
features of T1DM patients with a loss of β-cell
function and the development of hyperglycemia
[46-48]. At present, the STZ-induced mouse model is
one of the most widely used animal models of human
autoimmune diabetes in T1DM studies [46, 49-51]. In
our study, we focused the effect of circPPM1F on
development of STZ-induced diabetes mellitus.
Consistent with in vitro studies of increased pancreatic
β-cell apoptosis, we found that circPPM1F-mediated
M1 macrophage activation could also facilitate
pancreas injury in diabetic mice. However, it remains
to be clarified whether macrophage-specific
expression of circPPM1F facilitates the development
of T1DM. Treatment of circPPM1F with a
macrophage-specific promoter in the STZ-treated
NOD mouse model, or generating a chimeric,
macrophage-specific circPPM1F knocked-in T1DM
mouse model would be helpful to address the issue.
In summary, our studies demonstrate the
positive role of circPPM1F in LPS-induced M1
macrophage activation through the circPPM1F-HuR-
PPM1F-NF-κB axis. In vivo, circPPM1F facilitated
pancreas injury in STZ-induced diabetic mice by
promoting M1 macrophage activation. Additionally,
EIF4A3 and FUS might be required for the
maintenance of circPPM1F expression during the
progression of T1DM. Taken together, our work
provides new insights into the pathogenesis of T1DM
and suggests a potential novel biomarker or
therapeutic target for T1DM.
Abbreviations
Act D: actinomycin D; circRNAs: circular RNAs;
EIF4A3: eukaryotic initiation factor 4A-III; FMR1:
fragile X mental retardation 1 (also known as FMRP);
FUS: fused in sarcoma; GEO: Gene Expression
Omnibus; HuR: human antigen R; MAPK: mitogen-
activated protein kinase; NF-κB: nuclear factor kappa
B; mTOR: mammalian target of rapamycin; PPM1F:
protein phosphatase, Mg2+/Mn2+ dependent 1F;
PBMCs: peripheral blood mononuclear cells; qRT-
PCR: quantitative real-time PCR; RBP: RNA-binding
protein; RIP: RNA immunoprecipitation; ROC:
receiver operating curve; siRNA: small interfering
RNA; Stat3: signal transducers and activators of
transcription 3; STZ: streptozocin; T1DM: type 1
diabetes mellitus.
Supplementary Material
Supplementary figures and tables.
http://www.thno.org/v10p10908s1.pdf
Acknowledgments
Availability of data and material
The datasets and computer code produced in
this study are available in the following databases:
circBase (http://www.circbase.org/cgi-bin/
simplesearch.cgi), and circRNA interactome database
(https://circinteractome.nia.nih.gov/RNA_Binding_
Protein/rna_binding_protein.html).
Accession numbers
Microarray data have been deposited in the Gene
Expression Omnibus (GEO) under accession numbers
GSE133225.
Ethics approval and consent to participate
T1DM patients and age-matched healthy
controls were recruited from the Children’s Hospital
of Fudan University following informed consent from
their parents was obtained. The study was approved
by the Research Ethics Board of the Children′s
Hospital of Fudan University [No. (2016) 96].
Funding
This work was supported by grants from the
National Key R&D Program of China
(2016YFC1305102 to YZ), National Natural Science
Foundation of China (81671561, 81974248 to YZ,
81900751 to HX), the International Joint Laboratory
Program of National Children’s Medical Center
(EK1125180109 to YZ), Program for Outstanding
Medical Academic Leader (2019LJ19 to YZ) and
Shanghai Municipal Planning Commission of Science
and Research Fund (201740065 to YZ and 20174Y0079
to HX). Shanghai Pujiang Program (16PJ1401600 to
FJ). Shanghai Committee of Science and Technology
(19ZR1406400 to FJ).
Authors' contributions
CZ, XH, LY, JR, SH, WX, YG, QL and XW
designed and carried out experiments, and analyzed
data. CS, FL and WL recruited and characterized the
human participants. CZ and YZ wrote the
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10923
manuscript. YZ planned, designed, supervised, and
coordinated the overall research efforts.
Competing Interests
The authors have declared that no competing
interest exists.
References
1. Patterson CC, Dahlquist GG, Gyurus E, Green A, Soltesz G, Group ES.
Incidence trends for childhood type 1 diabetes in Europe during
1989-2003 and predicted new cases 2005-20: a multicentre prospective
registration study. Lancet. 2009; 373: 2027-33.
2. Rewers M, Ludvigsson J. Environmental risk factors for type 1 diabetes.
Lancet. 2016; 387: 2340-8.
3. Diaz-Valencia PA, Bougneres P, Valleron AJ. Global epidemiology of
type 1 diabetes in young adults and adults: a systematic review. BMC
Public Health. 2015; 15: 255.
4. Thomas NJ, Jones SE, Weedon MN, Shields BM, Oram RA, Hattersley
AT. Frequency and phenotype of type 1 diabetes in the first six decades
of life: a cross-sectional, genetically stratified survival analysis from UK
Biobank. Lancet Diabetes Endocrinol. 2018; 6: 122-9.
5. Hughes JW, Bao YK, Salam M, Joshi P, Kilpatrick CR, Juneja K, et al.
Late-onset T1DM and older age predict risk of additional autoimmune
disease. Diabetes Care. 2019; 42: 32-8.
6. Patterson CC, Karuranga S, Salpea P, Saeedi P, Dahlquist G, Soltesz G, et
al. Worldwide estimates of incidence, prevalence and mortality of type 1
diabetes in children and adolescents: results from the International
Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res Clin Pract.
2019; 157: 107842.
7. Cnop M, Welsh N, Jonas JC, Jorns A, Lenzen S, Eizirik DL. Mechanisms
of pancreatic beta-cell death in type 1 and type 2 diabetes: many
differences, few similarities. Diabetes. 2005; 54 Suppl 2: S97-107.
8. Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and
polarization. Front Biosci. 2008; 13: 453-61.
9. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, et al.
Macrophage activation and polarization: nomenclature and
experimental guidelines. Immunity. 2014; 41: 14-20.
10. Calderon B, Suri A, Unanue ER. In CD4+ T-cell-induced diabetes,
macrophages are the final effector cells that mediate islet beta-cell killing:
studies from an acute model. Am J Pathol. 2006; 169: 2137-47.
11. Wang F, Sun F, Luo J, Yue T, Chen L, Zhou H, et al. Loss of
ubiquitin-conjugating enzyme E2 (Ubc9) in macrophages exacerbates
multiple low-dose streptozotocin-induced diabetes by attenuating M2
macrophage polarization. Cell Death Dis. 2019; 10: 892.
12. Arnush M, Scarim AL, Heitmeier MR, Kelly CB, Corbett JA. Potential
role of resident islet macrophage activation in the initiation of
autoimmune diabetes. J Immunol. 1998; 160: 2684-91.
13. Yang H, Xie T, Li D, Du X, Wang T, Li C, et al. Tim-3 aggravates
podocyte injury in diabetic nephropathy by promoting macrophage
activation via the NF-kappaB/TNF-alpha pathway. Mol Metab. 2019; 23:
24-36.
14. Jun HS, Yoon CS, Zbytnuik L, van Rooijen N, Yoon JW. The role of
macrophages in T cell-mediated autoimmune diabetes in nonobese
diabetic mice. J Exp Med. 1999; 189: 347-58.
15. Thayer TC, Delano M, Liu C, Chen J, Padgett LE, Tse HM, et al.
Superoxide production by macrophages and T cells is critical for the
induction of autoreactivity and type 1 diabetes. Diabetes. 2011; 60:
2144-51.
16. Pasman Z, Been MD, Garcia-Blanco MA. Exon circularization in
mammalian nuclear extracts. RNA. 1996; 2: 603-10.
17. Wilusz JE. A 360 degrees view of circular RNAs: From biogenesis to
functions. Wiley Interdiscip Rev RNA. 2018; 9: e1478.
18. Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, et al.
Circular RNAs are abundant, conserved, and associated with ALU
repeats. RNA. 2013; 19: 141-57.
19. Rybak-Wolf A, Stottmeister C, Glazar P, Jens M, Pino N, Giusti S, et al.
Circular RNAs in the mammalian brain are highly abundant, conserved,
and dynamically expressed. Mol Cell. 2015; 58: 870-85.
20. Ng WL, Marinov GK, Liau ES, Lam YL, Lim YY, Ea CK. Inducible
RasGEF1B circular RNA is a positive regulator of ICAM-1 in the
TLR4/LPS pathway. RNA Biol. 2016; 13: 861-71.
21. Zhu YJ, Zheng B, Luo GJ, Ma XK, Lu XY, Lin XM, et al. Circular RNAs
negatively regulate cancer stem cells by physically binding FMRP
against CCAR1 complex in hepatocellular carcinoma. Theranostics. 2019;
9: 3526-40.
22. American Diabetes A. (2) Classification and diagnosis of diabetes.
Diabetes Care. 2015; 38 Suppl: S8-S16.
23. Han X, Huang S, Xue P, Fu J, Liu L, Zhang C, et al. LncRNA PTPRE-AS1
modulates M2 macrophage activation and inflammatory diseases by
epigenetic promotion of PTPRE. Sci Adv. 2019; 5: eaax9230.
24. Nyati KK, Masuda K, Zaman MM, Dubey PK, Millrine D, Chalise JP, et
al. TLR4-induced NF-kappaB and MAPK signaling regulate the IL-6
mRNA stabilizing protein Arid5a. Nucleic Acids Res. 2017; 45: 2687-703.
25. Weng T, Koh CG. POPX2 phosphatase regulates apoptosis through the
TAK1-IKK-NF-kappaB pathway. Cell Death Dis. 2017; 8: e3051.
26. Yang F, Hu A, Li D, Wang J, Guo Y, Liu Y, et al. Circ-HuR suppresses
HuR expression and gastric cancer progression by inhibiting CNBP
transactivation. Mol Cancer. 2019; 18: 158.
27. Zhou J, Zhang S, Chen Z, He Z, Xu Y, Li Z. CircRNA-ENO1 promoted
glycolysis and tumor progression in lung adenocarcinoma through
upregulating its host gene ENO1. Cell Death Dis. 2019; 10: 885.
28. Lebedeva S, Jens M, Theil K, Schwanhausser B, Selbach M, Landthaler
M, et al. Transcriptome-wide analysis of regulatory interactions of the
RNA-binding protein HuR. Mol Cell. 2011; 43: 340-52.
29. Chen LL, Yang L. Regulation of circRNA biogenesis. RNA Biol. 2015; 12:
381-8.
30. Zhang XO, Wang HB, Zhang Y, Lu X, Chen LL, Yang L. Complementary
sequence-mediated exon circularization. Cell. 2014; 159: 134-47.
31. Liu Y, Han J, Zhou Z, Li D. Paeoniflorin protects pancreatic beta cells
from STZ-induced damage through inhibition of the p38 MAPK and
JNK signaling pathways. Eur J Pharmacol. 2019; 853: 18-24.
32. Yang Z, Liu F, Qu H, Wang H, Xiao X, Deng H. 1, 25(OH)2D3 protects
beta cell against high glucose-induced apoptosis through mTOR
suppressing. Mol Cell Endocrinol. 2015; 414: 111-9.
33. Li J, He J, Zhang X, Li J, Zhao P, Fei P. TSP1 ameliorates age-related
macular degeneration by regulating the STAT3-iNOS signaling
pathway. Exp Cell Res. 2020; 388: 111811.
34. Wu J, Yang C, Liu J, Chen J, Huang C, Wang J, et al. Betulinic acid
attenuates T-2-Toxin-induced testis oxidative damage through
regulation of the JAK2/STAT3 signaling pathway in mice. Biomolecules.
2019; 9.
35. 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: 384-8.
36. Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, et al. Circular
intronic long noncoding RNAs. Mol Cell. 2013; 51: 792-806.
37. Errichelli L, Dini Modigliani S, Laneve P, Colantoni A, Legnini I,
Capauto D, et al. FUS affects circular RNA expression in murine
embryonic stem cell-derived motor neurons. Nat Commun. 2017; 8:
14741.
38. Bagni C, Greenough WT. From mRNP trafficking to spine
dysmorphogenesis: the roots of fragile X syndrome. Nat Rev Neurosci.
2005; 6: 376-87.
39. Fu XG, Yan AZ, Xu YJ, Liao J, Guo XY, Zhang D, et al. Splicing of exon 9a
in FMR1 transcripts results in a truncated FMRP with altered subcellular
distribution. Gene. 2020; 731: 144359.
40. Ashwal-Fluss R, Meyer M, Pamudurti NR, Ivanov A, Bartok O, Hanan
M, et al. circRNA biogenesis competes with pre-mRNA splicing. Mol
Cell. 2014; 56: 55-66.
41. Wang H, Chen W, Jin M, Hou L, Chen X, Zhang R, et al. CircSLC3A2
functions as an oncogenic factor in hepatocellular carcinoma by
sponging miR-490-3p and regulating PPM1F expression. Mol Cancer.
2018; 17: 165.
42. Abdelmohsen K, Panda AC, Munk R, Grammatikakis I, Dudekula DB,
De S, et al. Identification of HuR target circular RNAs uncovers
suppression of PABPN1 translation by CircPABPN1. RNA Biol. 2017; 14:
361-9.
43. Wang R, Zhang S, Chen X, Li N, Li J, Jia R, et al. EIF4A3-induced circular
RNA MMP9 (circMMP9) acts as a sponge of miR-124 and promotes
glioblastoma multiforme cell tumorigenesis. Mol Cancer. 2018; 17: 166.
44. Zheng X, Huang M, Xing L, Yang R, Wang X, Jiang R, et al. The circRNA
circSEPT9 mediated by E2F1 and EIF4A3 facilitates the carcinogenesis
and development of triple-negative breast cancer. Mol Cancer. 2020; 19:
73.
45. Like AA, Rossini AA. Streptozotocin-induced pancreatic insulitis: new
model of diabetes mellitus. Science. 1976; 193: 415-7.
46. Fidler TP, Marti A, Gerth K, Middleton EA, Campbell RA, Rondina MT,
et al. Glucose metabolism is required for platelet hyperactivation in a
murine model of type 1 diabetes. Diabetes. 2019; 68: 932-8.
47. Lin Y, Sun Z. Antiaging gene klotho attenuates pancreatic beta-cell
apoptosis in type 1 diabetes. Diabetes. 2015; 64: 4298-311.
48. O'Brien BA, Harmon BV, Cameron DP, Allan DJ. Beta-cell apoptosis is
responsible for the development of IDDM in the multiple low-dose
streptozotocin model. J Pathol. 1996; 178: 176-81.
Theranostics 2020, Vol. 10, Issue 24
http://www.thno.org
10924
49. Tekula S, Khurana A, Anchi P, Godugu C. Withaferin-A attenuates
multiple low doses of Streptozotocin (MLD-STZ) induced type 1
diabetes. Biomed Pharmacother. 2018; 106: 1428-40.
50. Wu M, Chen W, Zhang S, Huang S, Zhang A, Zhang Y, et al. Rotenone
protects against beta-cell apoptosis and attenuates type 1 diabetes
mellitus. Apoptosis. 2019; 24: 879-91.
51. Cheng RX, Feng Y, Liu D, Wang ZH, Zhang JT, Chen LH, et al. The role
of Nav1.7 and methylglyoxal-mediated activation of TRPA1 in itch and
hypoalgesia in a murine model of type 1 diabetes. Theranostics. 2019; 9:
4287-307.
... Although it has been demonstrated by global interaction assays that ~8.1% of proteins binding to nucleic acids have a dual function (119) (that is, binding to dNA and RNA), the interaction of circRNA_ci-Ins2/ci-INS with TAR dNA-binding protein 43 kda in T2d reported by Stoll et al (118) is one of the few reports between circRNAs and proteins with a dual function found in the literature. On the other hand, in type 1 dM, circRNA_PPM1F modulates M1 macrophage activation and inflammation of pancreatic β-cells through the circRNA_ PPM1F/ELAV-like protein 1/protein phosphatase 1F/nuclear factor NF-κB (NF-κB) axis (120). ...
Article
Full-text available
Circular RNAs (circRNAs) are non-coding single-stranded covalently closed RNA molecules that are considered important as regulators of gene expression at the transcriptional and post-transcriptional levels. These molecules have been implicated in the initiation and progression of multiple human diseases, ranging from cancer to inflammatory and metabolic diseases, including diabetes mellitus and its vascular complications. The present article aimed to review the current knowledge on the biogenesis and functions of circRNAs, as well as their role in cell processes associated with diabetic nephropathy. In addition, novel potential interactions between circRNAs expressed in renal cells exposed to high-glucose concentrations and the transcription factors c-Jun and c-Fos are reported.
... Notably, circRNA Cdyl has been found to promote M1 polarization by inhibiting the nuclear translocation of interferon regulatory factor 4 (IRF4) (Song et al., 2022). In a similar vein, circRNA PPM1F is known to enhance the NF-κB signaling pathway following LPS stimulation, promoting M1 polarization (Zhang et al., 2020). This highlights the growing importance of understanding ncRNA-mediated regulation in macrophages, particularly in the context of their role in tissue healing and adhesion mechanisms. ...
Article
Full-text available
Tendon injuries, a common musculoskeletal issue, usually result in adhesions to the surrounding tissue, that will impact functional recovery. Macrophages, particularly through their M1 and M2 polarizations, play a pivotal role in the inflammatory and healing phases of tendon repair. In this review, we explore the role of macrophage polarization in tendon healing, focusing on insights from animal models. The review delves into the complex interplay of macrophages in tendon pathology, detailing how various macrophage phenotypes contribute to both healing and adhesion formation. It also explores the potential of modulating macrophage activity to enhance tendon repair and minimize adhesions. With advancements in understanding macrophage behavior and the development of innovative biomaterials, this review highlights promising therapeutic strategies for tendon injuries.
... In addition to directly damaging beta cells, pro-inflammatory cytokines also contribute to the development and progression of insulitis [6]. Proinflammatory cytokines are mainly secreted from inflammatory T helper cells and M1 macrophages [7,8]. Understanding the role of PBMCs in the disease process is a key area of investigation. ...
Article
Background: Type 1 diabetes mellitus (T1DM) is an autoimmune disease characterized by immune response mediated islet beta cells destruction. However, the mechanisms that cause immune response in TIDM are still under investigation. Therefore, the goal of this study was to investigate the role of advanced glycation end products (AGEs) in the regulation of the immune response in peripheral blood mononuclear cells (PBMCs) from patients with T1DM. Methods: PBMCs isolated from T1DM patients and control subjects were used in the current study. Cytokines, AGEs related to glyoxalase 1 (GLO1), methylglyoxal (MG)-derived AGEs were assessed longitudinally. Results: The results of published T1DM PBMC microarray datasets using random-effects meta-analysis models revealed immune responses in the PBMCs of patients with T1DM compared with control subjects. Moreover, the activity of GLO1, which is the key MG-metabolizing enzyme, was significantly reduced in PBMCs from T1DM patients. We confirmed that, compared to the control subjects, GLO1 expression and activity were markedly decreased and MG-derived AGEs were significantly accumulated in the PBMCs from T1DM patients. In addition, phytohemagglutinin stimulated the secretion of tumor necrosis factor alpha (TNF-α), and interferon gamma (IFN-γ) was positively correlated with the accumulation of cellular AGEs. Therefore, the exposure of PBMCs from control subjects to MG and a GLO1 inhibitor enhanced the accumulation of cellular MG-derived AGEs and the secretion of TNF-α and IFN-γ. Conclusions: The results of this study showed that the accumulation of cellular AGEs causes a decline in the immune response of patients with T1DM.
... hsa_circ_0060450, circPPM1F, and hsa_ circ_0002202 are seen to be upregulated in peripheral blood mononuclear cells (PBMCs) in case of T1DM patients, that further help on the sponging of miRNA-199a −5p that inhibits the JAK-STAT signaling involved in the macrophage mediated inflammation. 141 Various studies documented the histone modification of H3K9Ac, H4K16Ac, H3K4me3, H3K9me2, and 3, H3K27me3 genes in the diabetic pathway, further closely located to the DQB1 and DRB1 genes. This leads to a surge in the transcription process in the monocytic cell line, thereby triggering the immunogenic pathway. ...
Article
Full-text available
Background Autoimmune diseases (AD) are severe pathophysiological ailments that are stimulated by an exaggerated immunogenic response towards self‐antigens, which can cause systemic or site‐specific organ damage. An array of complex genetic and epigenetic facets majorly contributes to the progression of AD, thus providing significant insight into the regulatory mechanism of microRNA (miRNA). miRNAs are short, non‐coding RNAs that have been identified as essential contributors to the post‐transcriptional regulation of host genome expression and as crucial regulators of a myriad of biological processes such as immune homeostasis, T helper cell differentiation, central and peripheral tolerance, and immune cell development. Aims This article tends to deliberate and conceptualize the brief pathogenesis and pertinent epigenetic regulatory mechanism as well as miRNA networks majorly affecting five different ADs namely rheumatoid arthritis (RA), type 1 diabetes, multiple sclerosis (MS), systemic lupus erythematosus (SLE) and inflammatory bowel disorder (IBD) thereby providing novel miRNA‐based theranostic interventions. Results & Discussion Pertaining to the differential expression of miRNA attributed in target tissues and cellular bodies of innate and adaptive immunity, a paradigm of scientific expeditions suggests an optimistic correlation between immunogenic dysfunction and miRNA alterations. Conclusion Therefore, it is not astonishing that dysregulations in miRNA expression patterns are now recognized in a wide spectrum of disorders, establishing themselves as potential biomarkers and therapeutic targets. Owing to its theranostic potencies, miRNA targets have been widely utilized in the development of biosensors and other therapeutic molecules originating from the same.
Article
Purpose The role of circular RNAs (circRNAs) in type 1 diabetes (T1D) is largely unknown. We aimed to identify some circRNAs as differential diagnostic biomarkers for T1D to discriminate patients with latent autoimmune diabetes in adults (LADA), and type 2 diabetes (T2D). Methods The circRNA expression profiles were determined by Arraystar human circRNA microarray in T1D compared to controls (n = 6 each). The differentially expressed circRNAs were validated by RT-qPCR using a validation cohort with 20 T1D and 20 controls. The diagnostic performances of the candidate circRNAs and the clinical parameters were assessed using the logistic least absolute shrinkage and selection operator (LASSO) regression model in a larger cohort with 457 subjects including patients with T1D, T2D and LADA, and controls. Results We identified 110 differentially expressed circular transcripts (53 upregulated and 57 downregulated) in T1D patients compared with controls. Further analysis showed that the levels of hsa_circRNA_405498 and hsa_circRNA_100033 were significantly downregulated in T1D compared to controls (both P < 0.05). Moreover, the expression levels of these two circRNAs showed sequential downregulation from controls, patients with T2D, LADA, to T1D (P < 0.05). The area under the curve (AUC) of receiver operating characteristic (ROC) plots in logistic LASSO regression model showed high diagnostic accuracy for combination model with the two circRNAs and some clinical parameters in discriminating T1D from LADA (AUC = 0.915), T2D (AUC = 0.993), and controls (AUC = 0.992). Conclusions Our study demonstrated that hsa_circRNA_405498 and hsa_circRNA_100033 are promising novel differential diagnostic biomarkers for T1D.
Article
With a growing body of evidence establishing circular RNAs (circRNAs) are widely exploited in eukaryotic cells and have a significant contribution in the occurrence and development of many complex human diseases. Disease-associated circRNAs can serve as clinical diagnostic biomarkers and therapeutic targets, providing novel ideas for biopharmaceutical research. However, available computation methods for predicting circRNA-disease associations (CDAs) do not sufficiently consider the contextual information of biological network nodes, making their performance limited. In this work, we propose a multi-hop attention graph neural network-based approach MAGCDA to infer potential CDAs. Specifically, we first construct a multi-source attribute heterogeneous network of circRNAs and diseases, then use a multi-hop strategy of graph nodes to deeply aggregate node context information through attention diffusion, thus enhancing topological structure information and mining data hidden features, and finally use random forest to accurately infer potential CDAs. In the four gold standard data sets, MAGCDA achieved prediction accuracy of 92.58%, 91.42%, 83.46% and 91.12%, respectively. MAGCDA has also presented prominent achievements in ablation experiments and in comparisons with other models. Additionally, 18 and 17 potential circRNAs in top 20 predicted scores for MAGCDA prediction scores were confirmed in case studies of the complex diseases breast cancer and Almozheimer's disease, respectively. These results suggest that MAGCDA can be a practical tool to explore potential disease-associated circRNAs and provide a theoretical basis for disease diagnosis and treatment.
Article
Cisplatin‐based chemotherapy is often used in advanced gastric cancer (GC) treatment, yet resistance to cisplatin may lead to treatment failure. Mechanisms underlying cisplatin resistance remain unclear. Recent evidence highlighted the role of macrophages in cancer chemoresistance. Macrophage‐derived exosomes were shown to facilitate intercellular communication. Here, we investigated the cisplatin resistance mechanism based on macrophage‐derived exosomes in gastric cancer. Cell growth and apoptosis detection experiments revealed that M2‐polarized macrophages increased the resistance of GC cells to cisplatin. qRT‐PCR, RNAase R assay, actinomycin D assay and cell nucleo‐cytoplasmic separation experiments confirmed the existence of circTEX2 in macrophage cytoplasm, with a higher expression level in M2 macrophages than that in M1 macrophages. Further experiments showed that circTEX2 acted as microRNA sponges for miR‐145 and regulated the expression of ATP Binding Cassette Subfamily C Member 1 (ABCC1). Inhibition of the circTEX2/miR‐145/ABCC1 axis blocked the cisplatin resistance of gastric cancer induced by M2 macrophages, as evidenced by in vitro and in vivo experiments. In conclusion, our research suggests that the exosomal transfer of M2 macrophage‐derived circTEX2 enhances cisplatin resistance in gastric cancer through miR‐145/ABCC1. Additionally, communication between macrophages and cancer cells via exosomes may be a promising therapeutic target for the treatment of cisplatin‐resistant gastric cancer.
Article
Full-text available
Background: Increasing studies have shown that circRNA is closely related to the carcinogenesis and development of many cancers. However, biological functions and the underlying molecular mechanism of circRNAs in triple-negative breast cancer (TNBC) remain largely unclear so far. Methods: Here, we investigated the expression pattern of circRNAs in four pairs of TNBC tissues and paracancerous normal tissues using RNA-sequencing. The expression and prognostic significance of circSEPT9 were evaluated with qRT-PCR and in situ hybridization in two TNBC cohorts. The survival curves were drawn by the Kaplan-Meier method, and statistical significance was estimated with the log-rank test. A series of in vitro and in vivo functional experiments were executed to investigate the role of circSEPT9 in the carcinogenesis and development of TNBC. Mechanistically, we explored the potential regulatory effects of E2F1 and EIF4A3 on biogenesis of circSEPT9 with chromatin immunoprecipitation (ChIP), luciferase reporter and RNA immunoprecipitation (RIP) assays. Furthermore, fluorescent in situ hybridization (FISH), luciferase reporter and biotin-coupled RNA pull-down assays were implemented to verify the relationship between the circSEPT9 and miR-637 in TNBC. Results: Increased expression of circSEPT9 was found in TNBC tissues, which was positively correlated with advanced clinical stage and poor prognosis. Knockdown of circSEPT9 significantly suppressed the proliferation, migration and invasion of TNBC cells, induced apoptosis and autophagy in TNBC cells as well as inhibited tumor growth and metastasis in vivo. Whereas up-regulation of circSEPT9 exerted opposite effects. Further mechanism research demonstrated that circSEPT9 could regulate the expression of Leukemia Inhibitory Factor (LIF) via sponging miR-637 and activate LIF/Stat3 signaling pathway involved in progression of TNBC. More importantly, we discovered that E2F1 and EIF4A3 might promote the biogenesis of circSEPT9. Conclusions: Our data reveal that the circSEPT9 mediated by E2F1 and EIF4A3 facilitates the carcinogenesis and development of triple-negative breast cancer through circSEPT9/miR-637/LIF axis. Therefore, circSEPT9 could be used as a potential prognostic marker and therapeutical target for TNBC.
Article
Full-text available
Long noncoding RNAs (lncRNAs) are important regulators of diverse biological processes; however, their function in macrophage activation is undefined. We describe a new regulatory mechanism, where an unreported lncRNA, PTPRE-AS1, targets receptor-type tyrosine protein phosphatase ε (PTPRE) to regulate macrophage activation. PTPRE-AS1 was selectively expressed in IL-4-stimulated macrophages, and its knockdown promoted M2 macrophage activation via MAPK/ERK 1/2 pathway. In vivo, PTPRE-AS1 deficiency enhanced IL-4-mediated M2 macrophage activation and accelerated pulmonary allergic inflammation while reducing chemical-induced colitis. Mechanistically, PTPRE-AS1 bound WDR5 directly, modulating H3K4me3 of the PTPRE promoter to regulate PTPRE-dependent signaling during M2 macrophage activation. Further, the expression of PTPRE-AS1 and PTPRE was significantly lower in peripheral blood mononuclear cells from patients with allergic asthma. These results provide evidence supporting the importance of PTPRE-AS1 in controlling macrophage function and the potential utility of PTPRE-AS1 as a target for controlling inflammatory diseases.
Article
Full-text available
T-2 toxin is one of the most toxic type A trichothecene mycotoxins in nature, and it exhibits reproductive toxicity. Betulinic acid (BA) is a natural pentacyclic triterpene compound found in species of Betula, and it has been reported to have antioxidant activity. The aim of the present study was to investigate the protective effect of BA on T-2-toxin-induced testicular injury in mice and explore its molecular mechanism. Sixty adult male mice were randomly divided into groups. The mice were pretreated orally with BA (0.25, 0.5, and 1.0 mg/kg) daily for 14 days, and the T-2 toxin (4 mg/kg body weight) was administered via intraperitoneal injection to induce oxidative stress after the last administration of BA. BA pretreatment significantly increased the secreted levels of testosterone and sperm motility. Moreover, BA pretreatment significantly increased the total antioxidant capacity (T-AOC), the activity of SOD and CAT, and the content of GSH, and it reduced the content of MDA. Furthermore, BA relieved testicular injury and reduced the number of apoptotic cells, and it significantly decreased the protein expression of Janus kinase 2 (JAK2), signal transducers and activators of transcription 3 (STAT3), caspsae-3, and Bcl-2-associated X protein (Bax). BA also increased the expression of B-cell lymphoma-2 (Bcl-2). We suggest that BA reduced the oxidative damage induced by T-2 toxin, and that these protective effects may be partially mediated by the JAK2/STAT3 signaling pathway.
Article
Full-text available
Type 1 diabetes (T1D) is characterized by the selective autoimmune destruction of the islet β cells, and macrophages play a significant role in this process. Small ubiquitin-like modification (SUMOylation) is an important posttranslational modification involved in T1D pathogenesis, but its function in macrophages remains unexplored. We presently developed and used macrophage-specific ubiquitin-conjugating enzyme E2 (Ubc9) knockout (LyzM-Cre-Ubc9fl/fl, KO) mice to address the impact of SUMOylation on macrophage function in a T1D model. We observed that blocking Ubc9 in macrophages exacerbated multiple-low dose streptozotocin (MLD-STZ)-induced diabetes. Specifically, after STZ treatment, blood glucose levels were consistently elevated in the KO mice. The KO mice exhibited a higher diabetes incidence than WT controls (85% vs. 55%, P < 0.01) along with a higher insulitis severity. The loss of Ubc9 impaired macrophage energy metabolism and attenuated macrophage M2 program, thereby enhancing T cell activation. Pancreas-resident macrophages, rather than migrant macrophages, played a predominant role in MLD-STZ-induced diabetes. Mechanistically, Ubc9-mediated SUMOylation of interferon regulator factor 4 (IRF4) enhanced its nuclear localization and stability, thereby transcribing IL-4 and arginase 1 (Arg1) to promote the macrophage M2 program. Ubc9-mediated SUMOylation modulates T1D risk at least in part by regulating macrophage function. Modulation of disturbed SUMOylation process in macrophages, either through cell adoptive transfer or targeted drug-delivery, could help to establish a tolerant pancreatic microenvironment and promote inflammation resolution in early insulitis stage, thus hindering T1D progression.
Article
Full-text available
Lung adenocarcinoma (LUAD) has long been one of the predominant reasons for the global cancer-linked mortality. The tumor progression is shown by several studies to be promoted by increased glycolysis. Enolase 1 (ENO1), as a glycolysis enzyme, performs pivotal role in glucose metabolism and contributes to tumor progression of numerous cancers. Circular RNAs (circRNAs) are catching increasing attentions for their surging roles in regulating gene expression in cancers. Our work is to uncover the regulatory mechanism circ-ENO1 on its host gene ENO1 and its function in glycolysis and tumor progression. Circ-ENO1 and its host gene ENO1 were identified to be upregulated in LUAD cells. Functionally, silencing circ-ENO1 retarded glycolysis, inhibited proliferation, migration and EMT, induced apoptosis. The cytoplasmic localization of circ-ENO1 was determined by FISH and subcellular fractionation. Mechanistically, circ-ENO1 acted as a ceRNA to interact with miR-22-3p and upregulate ENO1 expression. In vivo experiments certified that circ-ENO1 drove tumor growth and metastasis in vivo. In summary, current study elucidated that circ-ENO1 promoted glycolysis and tumor progression in LUAD by miR-22-3p/ENO1 axis, indicating circ-ENO1 as a promising treatment target for LUAD patients.
Article
Full-text available
Background: Circular RNAs (circRNAs), a subclass of non-coding RNAs, play essential roles in tumorigenesis and aggressiveness. Our previous study has identified that circAGO2 drives gastric cancer progression through activating human antigen R (HuR), a protein stabilizing AU-rich element-containing mRNAs. However, the functions and underlying mechanisms of circRNAs derived from HuR in gastric cancer progression remain elusive. Methods: CircRNAs derived from HuR were detected by real-time quantitative RT-PCR and validated by Sanger sequencing. Biotin-labeled RNA pull-down, mass spectrometry, RNA immunoprecipitation, RNA electrophoretic mobility shift, and in vitro binding assays were applied to identify proteins interacting with circRNA. Gene expression regulation was observed by chromatin immunoprecipitation, dual-luciferase assay, real-time quantitative RT-PCR, and western blot assays. Gain- and loss-of-function studies were performed to observe the impacts of circRNA and its protein partner on the growth, invasion, and metastasis of gastric cancer cells in vitro and in vivo. Results: Circ-HuR (hsa_circ_0049027) was predominantly detected in the nucleus, and was down-regulated in gastric cancer tissues and cell lines. Ectopic expression of circ-HuR suppressed the growth, invasion, and metastasis of gastric cancer cells in vitro and in vivo. Mechanistically, circ-HuR interacted with CCHC-type zinc finger nucleic acid binding protein (CNBP), and subsequently restrained its binding to HuR promoter, resulting in down-regulation of HuR and repression of tumor progression. Conclusions: Circ-HuR serves as a tumor suppressor to inhibit CNBP-facilitated HuR expression and gastric cancer progression, indicating a potential therapeutic target for gastric cancer.
Article
Full-text available
Type 1 diabetes mellitus (T1DM) is caused by pancreatic β-cell dysfunction and apoptosis, with consequent severe insulin deficiency. Thus, β-cell protection may be a primary target in the treatment of T1DM. Evidence has demonstrated that defective mitochondrial function plays an important role in pancreatic β-cell dysfunction and apoptosis; however, the fundamental effect of mitochondrial complex I action on β-cells and T1DM remains unclear. In the current study, the pancreas protective effect of complex I inhibitor rotenone (ROT) and its potential mechanism were assessed in a streptozotocin (STZ)-induced mouse model of T1DM and in cultured mouse pancreatic β-cell line, Min6. ROT treatment exerted a hypoglycemic effect, restored the insulin level, and decreased inflammation and cell apoptosis in the pancreas. In vitro experiments also showed that ROT decreased STZ- and inflammatory cytokines-induced β-cell apoptosis. These protective effects were accompanied by attenuation of reactive oxygen species, increased mitochondrial membrane potential, and upregulation of transcriptional coactivator PPARα coactivator 1α (PGC-1α)-controlled mitochondrial biogenesis. These findings suggest that mitochondrial complex I inhibition may represent a promising strategy for β-cell protection in T1DM.
Article
FMRP is an RNA-binding protein, loss of which causes fragile X syndrome (FXS). FMRP has several isoforms resulted from alternative splicing (AS) of fragile X mental retardation 1 (FMR1) gene, but their biological functions are still poorly understood. In the analysis of alternatively spliced FMR1 transcripts in the blood cells from a patient with FXS-like phenotypes (normal CGG repeats and no mutation in coding sequence of FMR1), we identified three novel FMR1 transcripts that include a previously unidentified microexon (46 bp), terming the exon 9a. This microexon exists widely in unaffected individuals, inclusion of which introduces an in-frame termination codon. To address whether these exon 9a-containing transcripts could produce protein by evading nonsense-mediated decay (NMD), Western blot was used to analysis blood cell lysate from unaffected individuals and a 34 kDa protein that consistent in size with the molecular weight of the predicted truncated protein produced from mRNA with this microexon was found. Meanwhile, treatment of peripheral blood mononuclear cells with an inhibitor of NMD (Cycloheximide) did not result in significant increase in exon 9a-containing transcripts. Using confocal immunofluorescence, we found the truncated protein displayed both nuclear and cytoplasmic localization in HEK293T and HeLa cells due to lacking C-terminal domains including KH2, NES, and RGG, while the full-length FMRP protein mainly localized in the cytoplasm. Therefore, we hypothesize that the inclusion of this microexon to generate exon 9a-containing transcripts may regulate the normal functionality of FMRP, and the dysregulation of normal FMRP due to increased exon 9a-containing alternatively spliced transcripts in that patient may be associated with the manifestation of FXS phenotype.
Article
Age-related macular degeneration is a progressive ocular disease that is the leading cause of vision loss among elderly. AMD usually is divided into two types: wet and dry AMD, which is linked with inflammation. Choroidal Neovascularization (CNV) formation or wet AMD is also associated with oxidative stress. Previously, TSP1 has been shown to have a significant alleviating effect on CNV in TSP1 knockout (TSP1-/-) mice. However, the mechanism by which TSP1 ameliorates CNV remains unclear. Here we report that TSP1 reduces nitric oxide production to prevent cells from forming tubes formation and reduced the levels of vascular endothelial growth factor (VEGF) and lipid peroxides (LPO) during oxidative stress. We measured RF/6A cell viability by CCK-8 assay and apoptosis by flow cytometry. RF/6A cell were transfected with TSP1 and STAT3 overexpression, and then the mRNA and protein levels of TSP1 and also the signal pathways were detected by qRT-PCR and Western blot analysis. Migration assays were performed using a transwell system. Co-Immunoprecipitation was used to analyze the binding relationship between CD47 and SHP-2. The results show that overexpression of TSP1 alleviated the damage of oxidative stress to RF/6A cells including increased cell activity and migration, decreased apoptosis and reduced migration compared to the control group. SHP-2 was activated by TSP1 through its receptor CD47 and STAT3 phosphorylation was reduced by activation of SHP-2, thereby blocking STAT3-iNOS pathway and reducing NO concentration in RF/6A cells ultimately protecting them from oxidative stress. Finally, the CNV mice model confirmed that TSP1 overexpression could protect the mice against CNV in vivo, modified the antioxidants levels and decreased the expression of TNF-α and IL-6 under laser irradiation. These results indicate a potential mechanism of TSP1 to slow down formation of CNV in wet AMD, which may bring hope for new treatment strategies.
Article
Aims: This article describes the methods, results and limitations of the International Diabetes Federation (IDF) Diabetes Atlas 9th edition estimates of worldwide numbers of cases of type 1 diabetes in children and adolescents. Methods: Most information in the published literature is in the form of incidence rates derived from registers of newly-diagnosed cases. After systematic review of the published literature and recent conference abstracts, identified studies were quality graded. If no study was available, extrapolation was used to assign a country the rate from an adjacent country with similar characteristics. Estimates of incident cases were obtained by applying incidence rates to United Nations 2019 population estimates. Estimates of prevalent cases were derived from incidence rates after making allowance for higher mortality rates in less-developed countries. Results: Incidence rates were available for 45% of countries (ranging from 6% in the sub-Saharan Africa region to 77% in the European region). Worldwide annual incidence estimates were 98,200 (128,900) new cases in the under 15 year (under 20 year) age-groups. Corresponding prevalence estimates were 600,900 (1,110,100) existing cases. Compared with estimates in earlier Atlas editions, numbers have increased in most IDF regions, reflecting incidence rate increases, but prevalence estimates have decreased in sub-Saharan Africa because allowance has been made for increased mortality in those with diabetes. Conclusions: Worldwide estimates of numbers of children and adolescents with type 1 diabetes continue to increase.