Abstract
Background
Diabetes mellitus is a worldwide disease with high incidence. Diabetic
peripheral neuropathy (DPN) is one of the most common but often ignored
complications of diabetes mellitus that cause numbness and pain, even
paralysis. Recent studies demonstrate that Schwann cells (SCs) in the
peripheral nervous system play an essential role in the pathogenesis of
DPN. Furthermore, various transcriptome analyses constructed by RNA-seq
or microarray have provided a comprehensive understanding of molecular
mechanisms and regulatory interaction networks involved in many
diseases. However, the detailed mechanisms and competing endogenous RNA
(ceRNA) network of SCs in DPN remain largely unknown.
Methods
Whole-transcriptome sequencing technology was applied to systematically
analyze the differentially expressed mRNAs, lncRNAs and miRNAs in SCs
from DPN rats and control rats. Gene ontology (GO) and KEGG pathway
enrichment analyses were used to investigate the potential functions of
the differentially expressed genes. Following this, lncRNA-mRNA
co-expression network and ceRNA regulatory network were constructed by
bioinformatics analysis methods.
Results
The results showed that 2925 mRNAs, 164 lncRNAs and 49 miRNAs were
significantly differently expressed in SCs from DPN rats compared with
control rats. 13 mRNAs, 7 lncRNAs and 7 miRNAs were validated by
qRT-PCR and consistent with the RNA-seq data. Functional and pathway
analyses revealed that many enriched biological processes of GO terms
and pathways were highly correlated with the function of SCs and the
pathogenesis of DPN. Furthermore, a global lncRNA–miRNA–mRNA ceRNA
regulatory network in DPN model was constructed and miR-212-5p and the
significantly correlated lncRNAs with high degree were identified as
key mediators in the pathophysiological processes of SCs in DPN. These
RNAs would contribute to the diagnosis and treatment of DPN.
Conclusion
Our study has shown that differentially expressed RNAs have complex
interactions among them. They also play critical roles in regulating
functions of SCs involved in the pathogenesis of DPN. The novel
competitive endogenous RNA network provides new insight for exploring
the underlying molecular mechanism of DPN and further investigation may
have clinical application value.
Keywords: diabetic peripheral neuropathy, Schwann cells, mRNA, lncRNA,
miRNA, RNA sequencing, competing endogenous RNA
Introduction
According to the International Diabetes Federation Diabetes Atlas,
pre-diabetes and diabetes mellitus (DM) affect 425 million people
worldwide in 2017, and this number will increase to 629 million in 2045
([51]International Diabetes Federation [IDF], 2017). Diabetic
peripheral neuropathy (DPN) is one of most common chronic complications
of both type 1 and type 2 diabetes mellitus, affecting at least half of
patients diagnosed with diabetes. DPN is characterized by pain,
tingling, paranesthesia, and hyperalgesia with a characteristic
stocking–glove pattern. Previous studies indicated that hyperglycemia,
insulin resistance, endothelial injury and microvascular dysfunctions
play critical roles in the process of DPN ([52]Gonçalves et al., 2017).
However, the particular mechanisms in the pathogenesis of DPN remain
poorly understood and the current gold standards of treatment is
lifestyle intervention and rigorous glucose control.
Schwann cells (SCs), deriving from neural crest cells, are the most
numerous glial cells of the peripheral nervous system (PNS) that form
the myelin sheath around axons ([53]Mizisin, 2014). SCs provide the
important protective effect on axons by myelinating and ensheathing all
axons in peripheral nerves, and have the potential to modulate neuronal
biology. This is done by maintaining the endoneurial microenvironment
via secreting various neurotrophic factors and the surrounding
extracellular matrix (ECM), enriching the sodium channels, insulating
the action potential velocity of axons, and promoting the regeneration
of axons after nerve injury ([54]Mizisin, 2014; [55]Gonçalves et al.,
2018). Previous investigations have demonstrated that peripheral nerve
axonal demyelination and impaired remyelination in diabetic state are
associated with Schwannopathy ([56]Mizisin et al., 1998). Further
studies conclusively have shown there are some key signaling pathways
activated in SCs during diabetic neuropathy, such as
hyperglycemia-driven high polyol pathway flux, oxidative stress,
mitochondrial dysfunctions, dyslipidaemia and inflammation ([57]Feldman
et al., 2017; [58]Gonçalves et al., 2018). However, the detailed
mechanisms underlying changes of SCs in DPN remain to be investigated.
MicroRNAs (miRNAs) are a class of non-coding RNAs consisting of 20–22
nucleotides. They negatively regulate post-transcriptional expression
levels of genes by binding to the 3′-untranslated region (UTR) to
inhibit target mRNA translation or to degrade them ([59]Bartel, 2004).
Studies have increasingly found differentially expressed miRNAs play
crucial roles in many human diseases, such as cancer, allergic
diseases, metabolic diseases. It has been proved that miR-146a and
miR-25 are involved in the pathogenesis of DPN as key regulators
([60]Liu X. S. et al., 2017; [61]Zhang et al., 2018). Long non-coding
RNAs (lncRNAs) are a group of non-coding RNAs with the length of
sequence more than 200 nucleotides. lncRNAs can interact with mRNAs and
miRNAs to construct a complicated regulatory network by imposing an
additional level of post-transcriptional regulation. Differentially
expressed lncRNAs are linked to a variety of human diseases ([62]Chen
et al., 2013). Furthermore, increasing evidence has suggested that
lncRNAs participate in the pathogenesis of DPN. For example,
upregulation of lncRNA NONRATT021972 in dorsal root ganglia results in
diabetic neuropathic pain of type 2 DM ([63]Peng et al., 2017). Growing
evidence supports that lncRNAs and circRNAs can act as competing
endogenous RNAs (ceRNAs) or miRNA sponges which could competitively
bind to miRNAs tends to regulate the expression levels of target mRNAs
([64]Salmena et al., 2011; [65]Zhang et al., 2019). However, the
competing endogenous network of lncRNA–miRNA–mRNA of SCs in DPN still
lacks systematic understanding and studying.
In our study, in order to obtain the expression profile of mRNAs,
lncRNAs and miRNAs in diabetic SCs, DPN models were established. Then
the RNA samples of SCs were collected from STZ-treated diabetic rats
and control rats to conduct RNA-seq. All the expression levels of
mRNAs, lncRNAs and miRNAs in SCs between two groups were screened and
analyzed. Bioinformatics analyses, including Gene Ontology (GO)
analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway
analysis, were performed to identify the significant biological
functions and signaling pathways of DPN. 13 mRNAs, 7 lncRNAs and 7
miRNAs were confirmed as differentially expressed by qRT-PCR. Via these
measures, ceRNA network construction was performed to reveal the
regulatory mechanism of DPN and seek potential therapeutic targets.
Materials and Methods
Animals
Male Sprague-Dawley rats (n = 20, weighing 190–210 g) were obtained
from Experimental Animal Center, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China. The animals were
maintained under standard conditions of 12-h light/dark cycle and room
temperature. Food and water were available ad libitum. The study was
performed with the approval by the Animal Ethics Committee of Huazhong
University of Science and Technology and all experimental procedures
were carried out in accordance with the National Institutes of Health
Guidelines and Regulations.
Induction of Diabetes
Animals were randomly divided into two groups: diabetic group (n = 10)
and control group (n = 10). After 12 h of fasting, rats of the diabetic
group were treated with streptozotocin (STZ, Sigma-Aldrich, United
States) injection at a dose of 65 mg/kg body weight dissolved in 0.05
mol/L citrate buffer, pH 4.5 at 4°C as described previously ([66]Tong
et al., 2012). The other group was treated with a single
intra-peritoneal injection of an equal volume of citrate buffer without
STZ as control. After STZ treatment for a week, the glucose meter
(Accu-Chek Active; Roche, Germany) was used to monitor random blood
glucose levels of all the rats from tail vein blood draws. Rats which
exhibited blood glucose levels of 16.7 mM or higher were diagnosed with
diabetes ([67]Resham and Sharma, 2019) and enrolled in this study. Rats
were kept to establish the model of DPN for 8 weeks ([68]Yu et al.,
2014). All animals survived until the end of the experiments.
Behavioral Test
Behavior was tested by blinded observers. Mechanical allodynia was
assessed by using von Frey filaments (Aesthesio, Danmic, United States)
to stimulate plantar hind paws as described previously ([69]Kroin et
al., 2010; [70]Santamaria et al., 2012). A 50% force withdrawal
threshold was determined for the plantar hind paws using the
“up-and-down” method ([71]Chaplan et al., 1994). Tail-flick test and
hot plate test were conducted to examine thermal hyperalgesia according
to the techniques described previously ([72]Sierra et al., 2015).
Briefly, for the tail-flick test, rats were restrained in a fixation
machine while tails were exposed. Approximately the distal 2/3 of the
tail was immersed in the water bath maintained at 52.0 ± 0.2°C
([73]Karna et al., 2019). The time was recorded when the tail removed
or flicks. The cut off time for tail flick test was set at 10 s to
avoid potential injury to the tail. For the hot plate test, a rat was
placed into the Perspex cylinder on the hot-plate (Model 7280, Ugo
Basile, Italy) of which the temperature was maintained 55.0 ± 0.2°C
([74]Sierra et al., 2015). Response time for discomfort action or
observed behavioral changes like licking paws, stomping or jumping was
recorded. The cut off time for hot plate test was set at 15 s in order
to avoid potential tissue injury. All the experiments were repeated
three times.
Electrophysiology
Sciatic nerve conduction velocity in two groups was tested as described
previously ([75]Ii et al., 2005; [76]Hendrix et al., 2013; [77]Baum et
al., 2016). Briefly, both the motor nerve conduction velocity (MNCV)
and sensory nerve conduction velocity (SNCV) were calculated through
dividing the distance between the recording electrodes and the
stimulation electrodes by the latency from the stimulation to the onset
of the first peak signal ([78]Ii et al., 2005). The MNCVs were recorded
by the sterilized electrodes placed into the intrinsic foot muscles,
and SNCVs were recorded by the near-nerve electrodes placed at the
sciatic notch that were used as stimulation electrodes for MNCVs
([79]Baum et al., 2016). During the experiments, the temperature of
rats was kept at 37°C while the room temperature was maintained at 20 ±
0.5°C. The experiments were repeated three times and all data was
collected and calculated by a Viking Quest Nicolet electromyography
equipment and the specific software system semi automatically.
Morphometric Analysis
After 8 weeks of STZ injection or vehicle solution injection and all
the behavioral and electrophysiology test completed, the animals (n =
10 per group) were euthanized by using sodium pentobarbital and killed
by cervical dislocation. Then all the sciatic nerves were dissected
under sterile conditions. The mid-portion of right sciatic nerve (about
1–2 cm) from two groups (n = 3) was harvested sterilely. The parts of
sciatic nerves were prefixed in 2.5% glutaraldehyde for 30 min and then
post-fixed in 1% osmium tettroxide for 1 h, then embedded in epoxy
resins. Ultra-thin (60 μm) sections were prepared and stained with
uranium acetate-lead citrate. The transmission electron microscopy
(HT7700, Hitachi, Japan) was used to image the samples of sciatic
nerves. For each sample at least three pictures were taken to assess
the development or progression of DPN ([80]Yang et al., 2018).
Subsequently, the diameter of myelinated nerve fibers and the mean of
g-ratio (the ratio of the inner axon diameter to the outer myelinated
fiber diameter) were calculated by using Image J software for further
statistical analysis.
Isolation of Primary Schwann Cells and Culture Preparations
The left sciatic nerve (between sciatic notch and the ankle) and the
remaining segment of the right sciatic nerve were isolated from each
rat sterilely. After stripping off the whole epineurium by using
microsurgical instruments under the stereomicroscope, nerves were cut
into segments of 1cm length. The segments were cultured in
pre-degeneration medium containing SCM (Schwann Cell Medium, ScienCell,
United States), 10% FBS (fetal bovine serum, ScienCell) and 1%
penicillin–streptomycin for 10 days at 37°C and 5% CO^2. After
pre-degeneration, the peripheral nerve tissue was cut into short
segments of 2–4 mm length and dissociated enzymatically for 20 h at
37°C and 5% CO^2. Dissociation medium consisted of DMEM (Thermo, United
States), 10% FBS, 0.125% collagenase type IV (Sigma-Aldrich), 1.25 U/ml
dispase II (Solaribo, China) and 1% penicillin–streptomycin
([81]Haastert et al., 2007). Then cell suspensions were centrifuged at
235 g for 5 min at +21°C, rinsed in DMEM, centrifuged again and
resuspended in SC culture medium containing SCM, 5% FBS, 1% SCGS
(Schwann Cell Growth Supplement, ScienCell) and 1%
penicillin–streptomycin. Then the cell suspensions were plated at 106
cells per well on a poly-L-ornithin (Sigma-Aldrich) and laminin
(Sigma-Aldrich) coated six well plate. The medium was changed to
serum-free SC culture medium after 24 h. SCs were enriched and purified
as described previously ([82]Haastert et al., 2007; [83]Kaewkhaw et
al., 2012). Specially, SCs dissociated from control groups and
STZ-treated groups were cultured in SC culture medium supplemented with
5.6 mM glucose (for control groups), 30 mM glucose (for STZ-treated
groups) ([84]Walsh et al., 2013).
Immunofluorescent Staining
The purity of SCs was determined by immunostaining. After being fixed
in 4% paraformaldehyde and permeabilized with 0.2% Triton X-100
(Sigma-Aldrich), SCs were incubated with primary antibody S-100β (1:
100, ProteinTech Group, Inc., United States) at 4°C overnight and then
incubated with the cy3 conjugated goat anti-rabbit IgG secondary
antibody (1: 200, ProteinTech) for 1 h in the darkness. After being
washed with PBS (phosphate-buffered saline, pH 7.4), the cells were
incubated with 4′,6-diamidino-2-phenylindole (DAPI, Boster Biological
Technology, China) to label nuclei. The purity of SCs was obtained
based on the proportion of S-100β-positive cells with DAPI-stained
nuclei.
RNA Extraction and RNA-Seq Analysis
Schwann cell samples were randomly selected from control groups (n = 3)
and diabetic groups (n = 3), rinsed with PBS twice and total RNA was
isolated by using miRNeasy Mini Kit (Qiagen, Germany) according to the
manufacturer’s instructions. The concentration and quality of RNA
samples were assessed by the NanoPhotometer spectrophotometer (IMPLEN,
USA). All RNA samples had an RNA Integrity Number (RIN) > 9.0
([85]Supplementary File S1). After total RNA from each sample was
qualified, RNA-seq was assessed to identify the differentially
expressed mRNAs, lncRNAs and miRNAs in the DPN rat models. Illumina
TruSeq RNA Sample Prep Kit was used with 1 μg of total RNA for the
construction of sequencing libraries on the Illumina HiSeq-Xten
sequencing platform (Genminix Informatics Ltd, Shanghai, China). Raw
reads were filtered and cleaned using fastp version 0.19.4 to remove
the adaptor reads and low-quality tags ([86]Yang et al., 2018). Then
the sequence reads were aligned to the reference genome using HISAT2
for transcripts and Bowtie v.1.2.2 for miRNA as described
([87]Langmead, 2010; [88]Kim et al., 2015). Subsequently, raw counts
were generated using the Stringtie v1.3.4 tool for transcripts and the
featureCounts v1.6.1 tool for miRNA ([89]Liao et al., 2014; [90]Pertea
et al., 2015). Differential expression analysis was performed using
DESeq2 v1.20.0 ([91]Love et al., 2014).
Cell RNA Extraction and Quantitative Real-Time PCR (qRT-PCR)
The remaining SCs samples in each group (n = 5 per group) were applied
to validate RNA-seq results by using quantitative real-time polymerase
chain reaction. Total cell RNA was extracted with TRIzol Reagent
(Invitrogen, United States). For miRNA, 2 μg of total RNA was reverse
transcribed to cDNA using the Mir-X miRNA First-Strand Synthesis kit
(Clontech, Japan) according to the user manual. For mRNA and lncRNA, 1
μg of RNA was reverse transcribed using Takara reverse transcription
kit (Takara, China) and qRT-PCR was performed based on the instruction
of Mir-X miRNA qRT-PCR SYBR Kit (Clontech) and ChamQ SYBR qPCR Master
Mix (Vazyme, China), respectively. The delta–delta Ct method (2^–ΔΔCt)
was used to measure the relative levels of transcripts expression and
the data were normalized to β-actin (endogenous internal controls of
mRNA and lncRNA) or U6 (endogenous internal control of miRNA). The
experiment was repeated three times. The primer sequences
([92]Supplementary Table S1) were synthesized by TSINGKE Biological
Technology Co., Ltd (China).
Functional and Pathway Enrichment
Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes
(KEGG) pathway analysis were performed to investigate biological
functions of DPN-related genes. The ontology contains three domains:
biological process, cellular components, and molecular
interaction^[93]1 and KEGG^[94]2 is a collection of online databases.
Briefly, GO analysis was applied to annotated function of
differentially expressed genes in SCs based on GO database. After
analysis for the significance level (P-value) and false discovery rate
(FDR), GO terms were selected from the enriched gene sets (FDR < 0.05).
Pathway analysis was performed to explore pathway clusters covering the
molecular interaction and reaction networks of the differentially
expressed genes according to KEGG. P-value < 0.05 was considered as
significant pathways.
Construction of lncRNA–mRNA Network and Prediction of miRNA Targets
Based on the theory of weighed gene co-expression network analysis
(WGCNA) ([95]Zhang and Horvath, 2005; [96]Horvath, 2011), the method of
lncRNA-mRNA co-expression network construction was consistent with
previous study ([97]Jiang et al., 2016). The co-expression similarity
S[ij] is defined and calculated as the absolute value of the Pearson
correlation coefficient between the expression profiles of two genes i
and j:
[MATH: Sij=|cor(i,j)| :MATH]
(1)
Then S[ij] is transformed into the adjacency a[ij] via an adjacency
function:
[MATH:
aij=<
/mo>Sijβ :MATH]
In this formula, the positive integer β represents the exponential
parameter for power law distribution and is used to characterize the
likeness to a scale-free network ([98]Langfelder and Horvath, 2008).
The connectivity of the i-th node is defined by
[MATH:
ki=
mo>∑jaij
:MATH]
In unweighted networks, the connectivity k[i] equals the number of
nodes that are directly linked to node i. In weighted networks, the
connectivity equals the sum of connection weights between node i and
the other nodes. And the distribution of connectivity (k) of genes best
fits a power law p(k) ∼ k^–γ ([99]Barabási, 2009). With 1∼30 of β value
ranges, the linear model was established by logarithms of the adjacency
degree of a node log(k) and the appearance probability of this node
log(p(k)) and ensuring that the overall average connectivity was not
too low ([100]Zhao and Fan, 2019). β = 22 was finally determined for
further analysis according to scale free topology criterion and p(k) in
our co-expressing network indicates the probability that a gene is
co-expressed with k other genes. Next, the dissimilarity between the
nodes can be calculated by the topological overlap measure (TOM) and
was used as the distance. The hybrid hierarchical clustering algorithm
was used to perform cluster analysis in order to identify gene modules
for biological significance. The module reflects the subset of tightly
connected nodes in the network. Furthermore, genes in high modules may
be important candidate hub genes which have important biological
functions ([101]Ravasz, 2002; [102]Yip and Horvath, 2006; [103]Lim et
al., 2013). The lncRNA-mRNA network was drawn by Cytoscape
3.7.2^[104]3. The targets of differentially expressed miRNAs were
predicted by miRanda and annotated with miRTarBase ([105]Joung et al.,
2007). The predicted targets involved mRNAs and lncRNAs were compared
to differentially expressed mRNAs and lncRNAs detected from RNA-Seq,
and the overlapped RNAs were collected. Then, the differentially
expressed miRNAs and selected target mRNAs and lncRNAs were combined
for RNA network analyses.
Construction of the ceRNA Regulatory Work
Some RNA molecules such as lncRNAs and mRNAs are able to communicate
with each other by sharing miRNA response elements (MREs) according to
the ceRNA hypothesis ([106]Salmena et al., 2011). To investigate the
role and interaction among mRNAs, lncRNAs, and miRNAs in DPN
pathogenesis, we constructed the ceRNA network based on miRNA-target
relationship and co-expressed relationship between mRNA and lncRNA. The
network was constructed in four steps in detail: (1) Obtention of the
differential expressed mRNAs, miRNAs and lncRNAs by RNA-seq; (2)
Prediction of the targeting relationship between differential miRNAs
and differential mRNAs (tool: miRanda). miRanda predicted the targeting
relationship between miRNA and mRNA based on the sequence alignment and
binding free energy calculation of miRNA sequences and 3′-UTR of mRNA.
(3) Prediction of the targeting relationship between differential
miRNAs and differential lncRNAs (tool: miRanda); (4) Construction of
ceRNA network: differentially expressed lncRNA-mRNA pairs were
identified based on Pearson correlation coefficients (PCC) calculated
from differentially expressed mRNAs and lncRNAs that were identified in
our RNA-seq results (PCC > 0.8 and p < 0.05). Then we used the
targeting relationship of mRNA/lncRNA regulated by microRNA to
establish a lncRNA–miRNA–mRNA interaction network. Based on the
targeting relationship between mRNA/lncRNA and microRNA, the adjacency
matrix A = [a[ij]] of mRNA/lncRNA and microRNA could be constructed,
a[ij] represents the weight of the relationship between mRNA/lncRNA i
and microRNA j. Finally, the ceRNA network was constructed using
Cytoscape v3.7.2 according to predicted shared pairs of miRNA–mRNA and
miRNA–lncRNA and the differentially expressed lncRNA-mRNA pairs
([107]Otasek et al., 2019). The significance of shared miRNAs in this
primary network was further calculated using the hypergeometric test (p
< 0.05) ([108]Le et al., 2017). The importance of RNA molecules in the
network is expressed by the degree of the network. The degree of
centrality indicates the degree of contribution of microRNA to
surrounding mRNA/lncRNA, or the degree of contribution of mRNA/lncRNA
to surrounding microRNA. The complete experiment workflow is shown in
[109]Figure 1.
FIGURE 1.
FIGURE 1
[110]Open in a new tab
The workflow of experiments and bioinformatic analysis. Male
Sprague-Dawley rats were treated with streptozotocin (STZ) or citrate
buffer and after 8 weeks Schwann cells were isolated from sciatic
nerves of rats for RNA-seq analysis. Gene ontology and KEGG pathway
enrichment analyses were used to investigate the potential functions of
the differentially expressed genes. lncRNA–mRNA co-expression network
and ceRNA regulatory network were constructed by bioinformatics
analysis methods.
Dual-Luciferase Reporter Assay
To validate whether miR-212-5p directly targets the Gucy1a3 3′-UTR,
dual-luciferase reporter assay was performed as described previously
([111]Yu et al., 2012). In brief, the wildtype (WT) and mutated (MUT)
3′-UTR regions of Gucy1a3 were cloned and inserted into the MluI and
HindIII restriction enzyme sites of the pMIR-Report vector (Obio
Technology Co., Ltd, Shanghai, China). HEK-293T cells were seeded in
96-well plates for 24 h. Then, Gucy1a3-WT, Gucy1a3-MUT reporter or pMIR
empty vector plasmid was co-transfected with Renilla luciferase (pRL)
plasmids and miR-212-5p mimic or negative control (NC) using
lipofectamine 2000 (Invitrogen). Forty eight hours after transfection,
the Dual Luciferase Reporter Assay System (Promega, United States) was
used to analyzed luciferase activity. The experiments were repeated
three times.
Statistical Analysis
Data were expressed as the mean ± SEM. The unpaired Student’s t-test
and one-way analysis of variance with Bonferroni post hoc test were
performed for comparisons between two groups. P < 0.05 was considered
statistically significant. Pearson correlation analysis was used to
assess the ceRNA network construction. Statistical analysis was
calculated with the GraphPad Prism v 8.1 software.
Results
Model Identification of Diabetic Peripheral Neuropathy
Eight weeks after STZ injection, the body weight, the blood glucose
levels, the threshold of thermal and mechanical stimuli, nervous
conduction velocity and the morphological changes of sciatic nerve were
detected to assess the signs and symptoms of DPN. The results showed
that compared with control groups, the increased non-fasting blood
glucose levels of all the rats in STZ-treated group were in accordance
with diagnostic standard of diabetes ([112]Figure 2A), whereas the body
weight decreased ([113]Figure 2B), indicating the diabetic rat models
were established successfully. The sensory functions of sciatic nerve
were examined by tail flick test, hot plate test, and von Frey hair
test. Furthermore, both the motor and sensory nerve conduction velocity
of sciatic nerve were investigated. In agreement with previous studies,
the slowing down of nerve conduction ([114]Figures 2F,G), the increase
of the mechanical threshold ([115]Figure 2E) and decrease of thermal
threshold ([116]Figures 2C,D) revealed the damage of sciatic nerve
caused by DPN ([117]Mitro et al., 2014; [118]Sierra et al., 2015;
[119]Watson and Dyck, 2015). Morphological analysis showed the g-ratio
of myelinated fibers were significantly affected by DPN ([120]Figure
2H). In addition, nerve fibers from diabetic rats showed swollen fibers
and the loss of myelinated fibers ([121]Figure 2I).
FIGURE 2.
FIGURE 2
[122]Open in a new tab
Evaluation of streptozotocin (STZ)-injected diabetic peripheral
neuropathy (DPN) rat models. The differences between streptozotocin
(STZ)-treated rats and control rats were calculated. The non-fasting
blood glucose levels (B) were significantly higher in STZ-injected rats
compared with control rats. The body weight (A), withdrawal threshold
(E) and the motor and sensory nerve conduction velocities (F,G) in
STZ-injected rats significantly decreased. The latency of hot plate
test (C), tail flick test (D), and g-ratio of myelinated nerve fibers
(H) significantly increased compared with controls (mean ± SEM, n = 10
per group, *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001). The
detail of myelinated fibers was showed in (I). Scale bars: 1.0 μm.
Characterization of SCs
Schwann cells at passage 2 were typically bipolar, tripolar or
multipolar morphology in vitro while fibroblasts were more flattened
polygonal contour than SCs under light microscope ([123]Kaewkhaw et
al., 2012) ([124]Figure 3). The immunofluorescent staining results
showed the purity of SCs reached 87.0 ± 4.9% for S-100β antibody
marker.
FIGURE 3.
FIGURE 3
[125]Open in a new tab
Identification of SCs. The morphology of Schwann cells (A) was observed
under light microscope. SCs were immunolabeled for S100β (B) and
nucleic acid was signed with DAPI (C). Image (D) was merged from (B,C).
Scale bar: 100 μm.
Differentially Expressed lncRNAs, miRNAs, mRNAs in Schwann Cells of Rats With
DPN
To identify functional mRNAs, lncRNAs, and miRNAs involved in SCs of
rats with DPN, total RNA of SCs randomly selected from three diabetic
groups and three control groups were collected for RNA-Seq. 4,622
lncRNAs, 427 miRNAs and 32,883 mRNAs were detected in total. The
results showed that 164 lncRNAs, 49 miRNAs and 2,925 mRNAs were
significantly altered with fold changes > 1.2 or <0.83 in SCs of
diabetic groups ([126]Bonfanti et al., 2015) ([127]Supplementary Tables
S2–S4, P-value < 0.05). All the differentially expressed lncRNAs,
miRNAs, and mRNAs were presented by the hierarchical clustering heat
maps and volcano plots in [128]Figure 4. Specifically, among all the
differentially expressed lncRNAs, 51 lncRNAs were significantly
upregulated whereas 113 lncRNAs were significantly downregulated in
diabetic groups. Thirty miRNAs were significantly increased and 19
miRNAs were significantly decreased. Among the differentially expressed
mRNAs, a total of 1,459 mRNAs were upregulated and 1,466 mRNAs were
downregulated in the diabetic groups compared with control groups.
FIGURE 4.
FIGURE 4
[129]Open in a new tab
Identification of mRNAs, lncRNAs, and miRNAs differentially expression
in Schwann cells from streptozotocin (STZ)-treated rats and control
rats. Hierarchical clustering analysis showed all differentially
expressed mRNAs (A), lncRNAs (C), and miRNAs (E). Red color represented
relatively high expression and green color represents relatively low
expression. Volcano plots of normalized expression levels for
differentially expressed mRNAs (B), lncRNAs (D) and miRNAs (F). The red
points in the plots represented the upregulated RNAs with statistical
significance and the blue points in the plots represented the
downregulated RNAs with statistical significance (P-value < 0.05 and
fold change > 1.2 or <0.83).
Validation of Differentially Expressed mRNAs, lncRNAs, and miRNAs in Schwann
Cells of DPN
qRT-PCR analysis was performed on RNA extracted from SCs to confirm the
expression level changes gained from the RNA-seq analysis. We randomly
selected 21 mRNAs, 13 mRNAs and 9 miRNAs with high fold change to
validate the results of RNA-seq. 13 mRNAs, 7 lncRNAs and 7 miRNAs had
been verified. The relative expression levels of the validated mRNAs,
lncRNAs and miRNAs were showed in [130]Figures 5A–C, respectively. The
qRT-PCR results were found to be consistent with the RNA-seq data.
FIGURE 5.
FIGURE 5
[131]Open in a new tab
Confirmation of RNA-seq data in Schwann cells from streptozotocin
(STZ)-treated rats and control rats by qRT-PCR. Thirteen mRNAs (A) were
consistent with the RNA-seq results. Seven lncRNAs (B) were consistent
with the RNA-seq results. Seven miRNAs (C) were consistent with the
RNA-seq results. Results were presented as mean ± SEM of five
independent experiments (*p < 0.05, **p < 0.01, ***p < 0.001, ****p <
0.0001). RNA samples used for qRT-PCR analysis were independent of
RNA-seq samples.
Functional Enrichment Analyses
To investigate the biological effects of mRNAs, lncRNAs and miRNAs in
SCs of DPN rats, GO analysis and KEGG pathway analysis were performed.
GO analysis was used to analyze the up and down-regulated
differentially expressed targeted genes individually. The GO biological
process analysis showed that 6,258 downregulated biological process of
GO terms and 6,321 upregulated biological process of GO terms were
enriched. Of which, 147 GO terms were significantly downregulated and
320 GO terms were significantly upregulated ([132]Supplementary Table
S5, FDR < 0.05). A fraction of significant enriched terms was listed in
[133]Figures 6A,B. Some significant GO terms were associated with the
changes of SCs’ function such as myelination, action potential,
axongenesis and axon development ([134]Figure 6A). Pathway enrichment
analyses were performed to explore crucial signaling pathways of DPN.
The results showed 36 upregulated and 46 downregulated remarkable
enriched pathways in [135]Supplementary Table S6. The top downregulated
signaling pathways included many pathways related to SCs, such as cell
adhesion molecules (CAMs), ECM-receptor interaction, axon guidance, and
others related to intracellular pathways, such as PI3K-Akt signaling
pathway, cAMP signaling pathway ([136]Figures 6C,D). Among the top
upregulated signaling pathway, immune response pathway and metabolism
pathway were involved like IL-17 signaling pathways, TNF signaling
pathway, p53 signaling pathway and PPAR pathway.
FIGURE 6.
FIGURE 6
[137]Open in a new tab
Gene ontology (GO) analysis and KEGG pathway analysis. GO annotation of
biological processes (BP) related to upregulated and downregulated
mRNAs (A,B) of DPN model. KEGG pathway enrichment analysis of
upregulated and downregulated mRNAs (C,D) of DPN model.
lncRNA–mRNA Network Construction and miRNA Target Prediction
Based on the expression correlation between the differentially
expressed mRNAs and lncRNAs, the lncRNA-mRNA co-expression network was
constructed firstly. The whole network consists of 161 lncRNAs and
1,511 mRNAs ([138]Supplementary Table S7 and [139]Supplementary Figure
S1). Among the network, the lncRNA ENSRNOT00000079885_AABR07064099.1
had the highest degree of 78 and the mRNAs Kcnt1, LOC691485 and P2rx2
had the highest degree of 13. Considering the figure cannot clearly
display the complex relationship in whole network, nine top lncRNAs
(degree ≥ 45) and related 147 mRNAs were selected to construct
sub-lncRNA-mRNA network ([140]Figure 7). Moreover, miRanda and
miRTarBase database were used to predict the target mRNAs and lncRNAs
([141]Supplementary Table S8). As the results, 2,634 mRNAs and 178
lncRNAs were predicted from 49 differentially expressed miRNAs
identified from RNA-seq.
FIGURE 7.
FIGURE 7
[142]Open in a new tab
Sub-lncRNA–mRNA regulatory network was established based on 147 mRNAs
and 9 lncRNAs. lncRNA and mRNA were indicated to round rectangle and
ellipse, respectively. The red nodes represented upregulated RNAs and
green nodes represented downregulated RNAs. The size of a node
indicated the degree of node in the network.
Construction of ceRNA Network
The ceRNA regulatory network was established in order to reveal the
potential molecular mechanism in SCs involved in DPN pathogenesis based
on the ceRNA hypothesis. In this study, 53 miRNAs, 59 lncRNAs and 1,229
mRNAs were found by bounding the number of miRNA response elements
(MREs) and the number of miRNAs shared by lncRNA-mRNA more than 1
([143]Supplementary Table S9 and [144]Supplementary Figure S2).
Considering the figure cannot clearly display the complex relationships
among 1,341 RNAs that constituted the ceRNA network, we selected miRNAs
which had higher degree in the network and validated by qRT-PCR to
construct the network diagram involving 38 lncRNAs, 10 miRNAs, and 702
mRNAs ([145]Figure 8A). Based on the validation results of qRT-PCR and
the connection with diabetes, miR-212-5p and predicted target Gucy1a3
was selected to further display the ceRNA network ([146]Figure 8B). The
lncRNAs such as LOC108348568, [147]AABR07030791.1, LOC102550012,
[148]AABR07054488.1, AABR07036331.2, [149]AABR07061328.1,
[150]AABR07063724.1, and [151]AABR07063724.1 associated with miR-212-5p
was also showed in [152]Figure 8B. To further confirmation,
dual-luciferase reporter assay was used to verify the relationship
between miR-212-5p and Gucy1a3. The result showed that miR-212-5p could
target the Gucy1a3 3′-UTR and decreased luciferase activity
([153]Figure 9), which was consistent with the results of RNA-seq and
qRT-PCR. However, the luciferase activities in the mutant group and
empty control group with miR-212-5p mimics also decreased ([154]Figure
9B). We analyzed that the main reason of this result was the potential
interaction between the pMIR-REPORT Luciferase vector and miR-212-5p
mimics.
FIGURE 8.
FIGURE 8
[155]Open in a new tab
Competing endogenous RNA network in DPN model. The competing endogenous
RNA network is based on miRNA-target relationship and co-expressed
interactions between mRNA and lncRNA. The whole competing endogenous
RNA network (A) in the DPN model. The sub competing endogenous RNA
network (B) consists of miR-212-5p, 7 lncRNAs and 125 mRNAs. miRNA was
indicated to V-shape, lncRNAs were indicated to round rectangle and
mRNAs were indicated to ellipse. The red nodes represented upregulated
RNAs and green nodes represented downregulated RNAs. The size of a node
indicated the degree of node in the network.
FIGURE 9.
[156]FIGURE 9
[157]Open in a new tab
The interaction between Gucy1a3 and miR-212-5p. Potential binding sites
of miR-212-5p and 3′ UTR of Gucy1a3 mRNA predicted by TargetScan (A).
Dual-Luciferase activity assay showed that miR-212-5p could bind with
the 3′ UTR of Gucy1a3 mRNA (B). (*p < 0.05, **p < 0.01, ***p < 0.001,
****p < 0.0001).
Discussion
Diabetes mellitus is a worldwide disease with high incidence, and a
growing number of DM patients are suffering from DPN ([158]Feldman et
al., 2017). A large amount of review and analysis of the literature
suggest the possible mechanism in DPN development ([159]Mizisin, 2014;
[160]Gonçalves et al., 2017, [161]2018). Nevertheless, there is a lack
of the effectual treatment for diabetic neuropathy and neuropathic
pain. SCs play a vital role in the process of DPN ([162]Gonçalves et
al., 2017). Recent experimental data provide further insight into the
complex relationship between dysfunctional SCs and neuropathy. To
better explore the new targets for early diagnosis and treatment, the
specific molecular mechanism of SCs in DPN need to be understood. In
this study, 8 weeks after streptozotocin injection, the DPN model in
rats was established ([163]Zochodne et al., 2007). Moreover, adult rat
SCs were isolated and cultured successfully. In order to elucidate the
detailed mechanism and the pathogenesis of DPN, RNA-seq technology was
applied to systematically analyze the differentially expressed lncRNAs,
miRNAs and mRNAs of SCs from DPN rats and the ceRNA regulatory network
was established.
In the present study, 2,925 mRNAs, 164 lncRNAs and 49 miRNAs were
significantly upregulated or downregulated in SCs of DPN rats. The
expression levels of mRNAs, lncRNAs, and miRNAs were confirmed in the
model of DPN via qRT-PCR validation and the results were in accordance
with RNA-seq data. Furthermore, some confirmed RNAs have been reported
associated with diabetes mellitus or DPN in the past few years. For
example, chemokine CXCL13 (C-X-C motif chemokine ligand 13) and its
receptor CXCR5 (C-X-C chemokine receptor type 5) associated with
inflammatory response have been demonstrated directly to be a pivotal
regulator in the pathogenesis of painful diabetic neuropathy via ERK
pathway ([164]Liu et al., 2019). The study shows that Ocln (occludin),
a tight junction protein involved in myelin sheath related to SCs, has
significant differences in patients with demyelinating peripheral
neuropathies ([165]Manole et al., 2015). Gene Grem1 and Slc7a9 are
associated with diabetic nephropathy ([166]McKnight et al., 2010;
[167]Guan et al., 2016). It is reported that miR-483-3p could target
vascular endothelial zinc finger 1 (VEZF1) to modulate endothelial
integrity in patients with type 2 diabetes ([168]Kuschnerus et al.,
2019). Studies also show that miR-212-5p regulated lipid metabolism
targeting fatty acid synthase (FAS) and stearoyl-CoA desaturase-1
(SCD1) ([169]Guo et al., 2017). In addition, miRNA-212-5p is related to
the neuron death in the central nervous system ([170]Sun et al., 2018;
[171]Xiao et al., 2019). Moreover, recent studies have also reported
the integral roles of lncRNAs in DPN ([172]Liu C. et al., 2017;
[173]Peng et al., 2017), and previous studies apply microarray
technology to analyze the differentially expressed lncRNAs in dorsal
root ganglia from STZ-induced diabetic rats ([174]Du et al., 2019;
[175]Guo et al., 2019). These differentially expressed RNAs may be a
potential target for diagnosis and therapy of DPN.
To further investigate in the potential biological implications of
changes in RNAs of SCs in DPN, GO analysis and KEGG pathway analysis
were carried out. In biological process fold enrichment of GO analysis,
a mass of significant enriched downregulated GO terms such as axon
development (GO:0061564), axonogenesis (GO:0007409), action potential
(GO:0001508), myelination (GO:0042552), and axon ensheathment
(GO:0008366) showed that diabetes contributed to dysfunction of SCs and
nerve degeneration which consistent with previous studies ([176]Sango
et al., 2006; [177]Mizisin, 2014). In addition, there is obvious
co-relevance between Schwannopathy and DPN. The causes for the changes
in structure and function of peripheral nerve are the
hyperglycemia-driven SC stress and neurodegeneration that leads to DPN
([178]Gonçalves et al., 2017).
According to the pathway analyses, 36 upregulated and 46 downregulated
enriched pathways were obtained to further explain the underlying
functions of differentially expressed RNAs. The remarkable
downregulated pathways involved tight junction (rno04530), cell
adhesion molecules (CAMs, rno04514), axon guidance (rno04360) and
PI3K-Akt signaling pathway (rno04151) while the upregulated pathways
involved p53 signaling pathway (rno04115), TNF signaling pathway
(rno04668), PPAR signaling pathway (rno03320) and AGE-RAGE signaling
pathway in diabetic complications (rno04933). Interestingly, there are
many signal conduction paths and molecular mechanisms that may
participate in the pathological progression of DPN which has been
reported. AGE-RAGE signaling pathway is a significantly enriched
pathway that plays roles in regulating the sustained oxidative stress
and inflammation that occurs in DPN ([179]Wang et al., 2019).
Importantly, hyperglycemia-induced excess formation of advanced
glycation end product (AGE) results in axonal demyelination and
degeneration, the structure and function changes of SCs and the
detrimental effects on axons ([180]Yagihashi et al., 1992;
[181]Sugimoto et al., 2008). In addition, the receptor for AGE (RAGE)
is expressed in endothelial and SCs and the interactions between AGEs
and RAGE induce to the Schwannopathy and microangiopathy in the
peripheral nerve under diabetic state ([182]Wada and Yagihashi, 2005).
Furthermore, gentiopicroside has been demonstrated to have protective
effect on DPN through PPAR-γ/AMPK/ACC signal pathway ([183]Lu et al.,
2018). The neuroprotective effects of Heparin-Poloxamer thermosensitive
hydrogel co-delivered with basic fibroblast growth factor (bFGF) and
nerve growth factor (NGF) on diabetic rats are mainly attributed to the
activation of phosphatidylinositol 3 kinase and protein kinase B
(PI3K-Akt) signaling pathway and mitogen-activated protein kinase
kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling
pathway ([184]Li et al., 2018). Moreover, a vast array of signaling
pathways involving axon guidance, TNF-α signaling, loss of tight
junction which accounted for the pathogenesis of DPN, have been
mentioned in many literatures ([185]Pande et al., 2011; [186]Dewanjee
et al., 2018). In this study, the enrichment of the above-mentioned
pathways suggests the possible therapeutic targets of DPN.
The most possible ceRNA network involved in the pathogenesis and
development of DPN was selected and constructed. Rno-miR-212-5p had the
high degree in the whole ceRNA network while its predicted targets
(according to the target prediction database, miRanda) included Gucy1a3
(encoding guanylate cyclase soluble subunit alpha-3). Both of them were
listed as differentially expressed RNAs and validated by qRT-PCR.
Gucy1a3, a subunit of soluble Guanylyl Cyclase, is involved in cGMP-PKG
signaling pathway ([187]Midttun et al., 2018). Furthermore, recent
research finds that sildenafil increased cGMP by blocking PDE5
(phosphodiesterase-5) which can abolish the negative effect of
hyperglycemia on SCs and exert the beneficial effects on DPN through
the cGMP-PKG signaling pathway ([188]Hendrix et al., 2013). It seems a
reasonable deduction that overexpression of miR-212-5p may downregulate
the level of Gucy1a3 in SCs contributing to DPN via cGMP-PKG signaling
pathway. To further investigate the role of miR-212-5p in SCs of DPN, a
subnetwork was extracted from the globe ceRNA network of DPN. The
subnetwork showed that some lncRNAs such as LOC102550012,
[189]AABR07061328.1, [190]AABR07063724.1, [191]AABR07054488.1, and
[192]AABR07030791.1 may participate in the regulation of interaction of
miR-212-5p and Gucy1a3. Among the mentioned lncRNAs, LOC102550012,
which had the highest degree in the downregulated lncRNAs of ceRNA
subnetwork and also had interaction with Gucy1a3, probably have the
great possibility of competitively bounding with miR-212-5p, acting as
the ceRNA and leading the expression level of Gucu1a3 changes. Above
all, LOC102550012, miR-212-5p and Gucy1a3 might play an important role
in the development of DPN related to dysfunction of SCs and need deeper
studying and probing.
In summary, our study is the first to detect and analyze mRNAs,
lncRNAs, and miRNAs changes in SCs from STZ-treated rats with DPN and
reveal the novel interactions between differentially expressed RNAs and
the pathogenesis of DPN. However, peripheral nerve system does not only
consist of axons and SCs, mesenchymal cells, endothelial cell, immune
cells like NK cells, and T cells are also abundant in nerves.
Importantly, the interactions between them under the diabetic state
still remain to be discussed. Moreover, the changes in protein
expression level need further observation and exploration. Therefore,
further studies are needed to find the complete proteomic and specific
pathways of differentially expressed RNAs, the connections among
different kinds of cells in peripheral nerve system during the process
of DPN, and novel targets to treat DPN.
Data Availability Statement
The RNA-seq data of Schwann cells from diabetic rats and control rats
can be accessed online with the accession GEO: [193]GSE142785
([194]https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?a cc=GSE142785).
Ethics Statement
The animal study was reviewed and approved by the Animal Ethics
Committee of Huazhong University of Science and Technology.
Author Contributions
CW and YC designed the conceptual idea for this study. CW, XX, and JC
performed the experiments and collected the data. YK and JG helped with
data analysis. CW and HX performed the bioinformatic analysis. SR, TJ,
and GG conducted the rats feed and DPN models. CW and XX wrote the
manuscript. DD, XY, and H-GM revised the manuscript. MY and ZC provided
study materials. All authors read and approved the final manuscript.
Conflict of Interest
The authors declare that the research was conducted in the absence of
any commercial or financial relationships that could be construed as a
potential conflict of interest.
Funding. This work was supported by the National Natural Science
Foundation of China (81772094 and 81471270), the National Natural
Science Youth Science Foundation of China (81401759), and the National
Key Research and Development Project (2016YFC1101705).
^1
[195]http://www.geneontology.org
^2
[196]http://www.genome.jp/kegg/
^3
[197]https://cytoscape.org/
Supplementary Material
The Supplementary Material for this article can be found online at:
[198]https://www.frontiersin.org/articles/10.3389/fbioe.2020.00490/full
#supplementary-material
[199]Click here for additional data file.^ (14.5KB, DOCX)
[200]Click here for additional data file.^ (667.7KB, XLSX)
[201]Click here for additional data file.^ (41.7KB, XLSX)
[202]Click here for additional data file.^ (15.5KB, XLSX)
[203]Click here for additional data file.^ (1.4MB, XLSX)
[204]Click here for additional data file.^ (73KB, XLSX)
[205]Click here for additional data file.^ (170.2KB, XLSX)
[206]Click here for additional data file.^ (558.5KB, XLSX)
[207]Click here for additional data file.^ (139.9KB, XLSX)
[208]Click here for additional data file.^ (659.4KB, DOCX)
[209]Click here for additional data file.^ (716.6KB, PDF)
[210]Click here for additional data file.^ (168.3KB, PDF)
[211]Click here for additional data file.^ (194.3KB, PDF)
References