ArticlePDF Available

Integrative analysis of single-cell and bulk RNA sequencing unveils the senescence landscape in ischemic stroke

Authors:

Abstract and Figures

Ischemic stroke (IS) is a fatal neurological disease that occurs when the blood flow to the brain is disrupted, leading to brain tissue damage and functional impairment. Cellular senescence, a vital characteristic of aging, is associated with a poor prognosis for IS. This study explores the potential role of cellular senescence in the pathological process following IS by analyzing transcriptome data from multiple datasets (GSE163654, GSE16561, GSE119121, and GSE174574). By using bioinformatics methods, we identified hub-senescence-related genes such as ANGPTL4, CCL3, CCL7, CXCL16, and TNF and verified them using quantitative reverse transcription polymerase chain reaction. Further analysis of single-cell RNA sequencing data suggests that MG4 microglial is highly correlated with cellular senescence in MCAO, and might play a crucial role in the pathological process after IS. Additionally, we identified retinoic acid as a potential drug for improving the prognosis of IS. This comprehensive investigation of cellular senescence in various brain tissues and peripheral blood cell types provides valuable insights into the underlying mechanisms of the pathology of IS and identifies potential therapeutic targets for improving patient outcomes.
Content may be subject to copyright.
www.aging-us.com 5497 AGING
INTRODUCTION
Stroke is among the most fatal neurological diseases, and
it is the second leading cause of death in those aged >60
years and the fifth leading cause of death in those aged
<15 years [13]. Strokes are clinically classified into
ischemic stroke (IS), hemorrhagic stroke, and transient
ischemic attack, with IS accounting for 80% of all stroke
cases [3, 4]. IS is not only a major cause of death but is
also responsible for a significant number of disability-
adjusted life years, which increased by 138.6% from
1990 to 2019 [5]. Therefore, improving the prognosis of
IS is crucial for alleviating the disease burden.
Senescence is a fundamental biological process
characterized by a general decline in tissue function,
increased susceptibility to neurological diseases, and
decreased resistance to inflammation and infection [6].
Typically, IS is accompanied by accelerated sensory-
motor and neurocognitive decline, which are signs of
senescence [7, 8]. Accordingly, advanced age is a known
risk factor for IS [9]. Furthermore, IS is strongly
www.aging-us.com AGING 2023, Vol. 15, No. 12
Research Paper
Integrative analysis of single-cell and bulk RNA sequencing unveils
the senescence landscape in ischemic stroke
Longhui Fu1,2,*, Beibei Yu 1,2,*, Yongfeng Zhang1,2, Shuai Cao4, Boqiang Lv1,2, Yunze Tian1,2,3,
Huangtao Chen1,2, Shijie Yang1,2, Yutian Hu1,2, Jinghua Hua1,2, Pengyu Ren1,2, Jianzhong Li1,3,
Shouping Gong1,2,5
1Xi’an Jiaotong University, Xi’an, China
2Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
3Department of Thoracic Surgery, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
4Department of Orthopedics, Civil Aviation General Hospital, Chaoyang, Beijing, China
5Xi’an Medical University, Xi’an, China
*Co-first author
Correspondence to: Pengyu Ren, Jianzhong Li, Shouping Gong; email: renpengyu.xjut@mail.xjtu.edu.cn; jianzhong-
0520@163.com, https://orcid.org/0000-0002-7846-2921; shpingg@126.com, https://orcid.org/0000-0002-1723-938X
Keywords: ischemic stroke, aging, cellular senescence, single-cell RNA-seq, bioinformatics
Received: April 6, 2023 Accepted: May 27, 2023 Published: June 28, 2023
Copyright: © 2023 Fu et al. This is an open access article distributed under the terms of the Creative Commons Attribution
License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original
author and source are credited.
ABSTRACT
Ischemic stroke (IS) is a fatal neurological disease that occurs when the blood flow to the brain is disrupted,
leading to brain tissue damage and functional impairment. Cellular senescence, a vital characteristic of aging, is
associated with a poor prognosis for IS. This study explores the potential role of cellular senescence in the
pathological process following IS by analyzing transcriptome data from multiple datasets (GSE163654,
GSE16561, GSE119121, and GSE174574). By using bioinformatics methods, we identified hub-senescence-
related genes such as ANGPTL4, CCL3, CCL7, CXCL16, and TNF and verified them using quantitative reverse
transcription polymerase chain reaction. Further analysis of single-cell RNA sequencing data suggests that MG4
microglial is highly correlated with cellular senescence in MCAO, and might play a crucial role in the
pathological process after IS. Additionally, we identified retinoic acid as a potential drug for improving the
prognosis of IS. This comprehensive investigation of cellular senescence in various brain tissues and peripheral
blood cell types provides valuable insights into the underlying mechanisms of the pathology of IS and identifies
potential therapeutic targets for improving patient outcomes.
www.aging-us.com 5498 AGING
associated with cellular senescence, a major cause of
aging [10]. Cellular senescence refers to the permanent
state of cell cycle arrest, which is a defense mechanism
that prevents unwanted damage to cells [11]. The
inability of cells to re-enter the cell cycle in response to
irreversible growth arrest, resistance to apoptosis,
production of the senescence-associated secretory
phenotype (SASP), mitochondrial dysfunction, and
changes in DNA and chromatin levels are common
pathophysiological processes of cellular senescence [12].
High levels of inflammatory cytokines and SASP have
been detected in the IS-pedunculated region [13]. Various
studies have shown that cellular senescence intervention
improves the prognosis of patients with IS and is a
promising therapeutic approach [14, 15]. There are good
reasons to believe that cellular senescence plays an
important role in the pathophysiological process of IS,
and there are solid grounds for the assertion that cellular
senescence is crucial to the pathophysiology of IS.
Identifying senescent cells in vivo remains challenging,
although cellular senescence can drive a variety of age-
related disease manifestations through aging-related
secretory phenotypes. Recently published gene sets
related to senescent cells can aid in identifying in vivo
cellular senescence [16]. Moreover, senescence can vary
significantly in different cell types. The senescence of
endothelial, smooth muscle and immune cells is believed
to participate in the senescence of blood vessels, and the
senescence of immune cells is believed to promote the
aging of other cell types [17, 18]. Additionally, the
senescence of neurons and glial cells is widespread in
neurodegenerative diseases [19]. However, few studies
have examined cellular senescence after IS, and there is
a lack of research on the relationship between cellular
senescence and a wide range of cell types in the brain. In
this study, we identified hub genes for cellular
senescence in IS using bioinformatics and experimental
validation and explored their biological pathways. Using
single-cell RNA sequencing (scRNA-seq), we evaluated
the hub senescence-related gene (HSRG) expressions in
various cell types and mapped the developmental
trajectories of microglia and cellular communication
networks. Finally, we predicted potential therapeutic
drugs based on the HSRGs. The approach used in this
study is depicted in the flow diagram (Figure 1).
MATERIALS AND METHODS
Microarray datasets
SenMayo is a recently published gene set that
includes 125 and 118 unrepeatable genes in humans
and mice, respectively [16]. The gene set was
downloaded from the supplementary information of
the original article. The Gene Expression Omnibus
(http://www.ncbi.nlm.nih.gov/geo/”) is an open-
source database that provides gene expression profiles
for our study. Four datasets including GSE163654,
GSE16561, GSE119121, and GSE174574 were used
(Table 1). Bulk RNA-sequencing (bulk RNA-seq) of
brain tissue from three sham-operated rats and three
middle cerebral artery occlusion (MCAO) rats in
GSE163654 was used for differential expression
analysis. GSE16561 contains bulk RNA-seq data of
peripheral blood from 39 patients with IS and 24
patients with normal groups, which were used for
expression and immune cell infiltration analyses.
Figure 1. The flowchart of data preparation and analysis.
www.aging-us.com 5499 AGING
Table 1. Detailed information of the gene expression matrixes and platform.
GEO dataset
Platform
Species
Tissue
Country
GSE163654
GPL17117
Rat
Penumbras tissue of brains
Canada
GSE16561
GPL6883
Human
Peripheral blood
USA
GSE119121
GPL6247
Rat
Blood
Belgium
GSE174574
GPL21103
C57BL/6
Brain
China
GSE119121 contains bulk RNA-seq data from rat
peripheral blood from the MCAO and sham groups
used for the temporal analysis of gene expression.
Finally, scRNA-seq data of the brain tissue from
GSE174574 with three sham group mice and three
MCAO group mice were processed and used for cell
communication analysis.
Differential expression analysis
The R software (v4.2.1, R Foundation, Vienna, Austria)
was used for all analyses and visualizations in this study.
To create the analysis matrix, all original bulk RNA-seq
matrices were normalized and coupled with the
associated RNA probes. Data with non-mRNA
expression loss and no corresponding gene names were
excluded. The differentially expressed genes (DEGs)
were screened using the criteria |log2 (fold change) | >0.5
and a p-value of <0.05. Heatmaps and volcano plots were
generated using the “heatmap” and “ggplot2” packages,
respectively. Finally, a Venn diagram was created using
the website http://www.bioinformatics.com.cn/ for the
Venn analysis.
Pathway enrichment analysis and protein-protein
interaction network
Gene ontology (GO) and Kyoto Encyclopedia of Genes
and Genomes (KEGG) pathway analysis of DEGs were
performed and visualized using the “org.Mm.eg.db” and
“clusterProfiler” packages. The free website, STRING
(https://www.string-db.org/), was used to analyze
functional protein association networks of DEGs. A
minimum interaction score of ≥0.150 was defined as the
cut-off value, and the resulting protein-protein
interaction network was visualized using Cytoscape
software. Finally, the hub genes were ascertained by
visualizing the bulk RNA-seq of DEGs in GSE16561
using the “reshape2” and “ggpubr” packages.
Animal and establishment of the MCAO model
The Medical Experimental Animal Center (Xi’an
Jiaotong University) provided 12 pathogen-free male
Sprague-Dawley rats (weight: 280300 g). A modified
Zea-Longa model, in which the coil occlusion was
permanently placed in the middle cerebral artery, was
used to create a rat permanent MCAO model [20]. The
rats (n = 8) were randomly allocated to either the sham
group or the MCAO group, with four rats in each group.
The Longa scale was used to assess the neurobehavioral
scores of rats in each group two hours after MCAO.
Animals with no neurological impairment following
surgery were excluded from the study. The rats were
euthanized three days after the operation via
intraperitoneal injection. The brains were removed and
sliced before being put in 2% triphenyl tetrazolium
chloride (TTC) (Solarbio Life Science, Beijing, China)
and incubated at 37° C for 30 minutes.
Quantitative reverse transcription polymerase chain
reaction (RT-qPCR)
Three rats from each group were anesthetized 48 hours
after surgery, and tissue samples were collected from the
ischemic penumbra. The samples were immediately
stored in liquid nitrogen, and total RNA was extracted
from each sample using the TRIzol reagent (Sinopharm
Chemical Reagents Co., Ltd., China). The extracted RNA
was reverse-transcribed into complementary DNA using
SweScript All-in-One RT SuperMix (Wuhan Saiwei
Biotechnology Co., Ltd., China). Table 2 shows the
primer sequences used in the study. The 2-Ct method was
used to calculate the relative mRNA expression, which
was then compared to that of the normal group
(glyceraldehyde 3-phosphate dehydrogenase mRNA
expression). A student’s t-test was used for statistical
comparisons, and differences with a p-value of <0.05
were considered statistically significant.
Construction of a prediction model
The nomogram model, calibration, decision, and
clinical impact curves were based on the expression
data of HSRGs in GSE16561, implemented by the
“rms” and “rmda” packages. The receiver operating
characteristic (ROC) curve was also plotted through the
“ROCR” package, and the calibration, decision curve
analysis (DCA), and clinical impact curves were drawn.
Temporal analysis of expression
GSE119121 contains the bulk RNA-seq of MCAO
rats at different time points, and the DEGs expression
www.aging-us.com 5500 AGING
Table 2. Specific primers used for quantitative real-time PCR.
Gene
Forward
Reverse
Angptl4
CATGGCTGCCTGCGGTAACG
AGTTGCTGGATCTTGCTGTTCTGAG
Ccl3
CACCGCTGCCCTTGCTGTTC
GGAATTTGCCGTCCATAGGAGAAGC
Ccl7
GATCTCTGCCGCGCTTCTGTG
TGGATGAATTGGTCCCATCTGGTTG
Cxcl16
CAGTTTCAGAGCACCCAGCAGTC
GCCTAGCCTCCAGACCATAGCC
Tnf
CACCACGCTCTTCTGTCTACTGAAC
TGGGCTACGGGCTTGTCACTC
was described by a heatmap and a violin plot using
the “heatmap,” “reshape2,” and “ggpubr” packages.
Simultaneously, the mean value of hub gene expression
at different time points was calculated, and a line graph
was drawn.
Immune cell infiltration analysis
To analyze immune cell infiltrations in GSE16561 and
calculate merged expression data, we used the
CIBERSORT method, which is a technique for
analyzing different immune cell types in tissues [21].
The samples were filtered using a p-value of <0.05, and
a bar plot was generated to show the percentage of each
immune cell type in each sample. The “pheatmap”
package was used to generate a heatmap of the 22
immune cells and a heatmap describing the hub gene
expression in immune cells as well. The package
“vioplot” was used to compare and visualize the levels
of 22 immune cells in IS and normal samples. Using
the “corrplot” package, a correlation heatmap was
generated that revealed the correlation of 22 different
types of infiltrating immune cells.
ScRNA-seq data processing and cell communication
analysis
GSE174574 contains the scRNA-seq of three sham group
mice and three MCAO group mice. ScRNA-seq data
were processed using the “Seurat package for
unsupervised graph-based clustering before analysis [22].
The following were the screening criteria for the cells
examined: Cells with 5006,000 unique molecular
identifiers and 35% of mitochondrial genes judged to be
of high quality were eliminated from further research.
The normalized data function was used to normalize the
quality-controlled data, and then the find variable
features tool was used to select 2000 highly variable
genes. The mutual principal component analysis tool
“Seurat” was used to integrate the data. The proportion of
cells was determined by selecting the top 20 main
components for the visualization of dimensionality
reduction using uniform manifold approximation and
projection (UMAP). The “SingleR” package was used for
cell type identification, in which “MouseRNAseqData”
was used as a reference. Additionally, the “cellcall
package was used to infer intercellular communication
[23]. To determine differentiation trajectories for major
clusters with large cell numbers, the “monocle3” package
was used for cell trajectory analysis [24].
Drug screening and molecular docking
The DSigDB database contains the Food and Drug
Administration-approved drugs and experimental
compounds (http://tanlab.ucdenver.edu/DSigDB) and
is a free website with the DSigDB interface
(https://maayanlab.cloud/Enrichr/). Drugs and compounds
were predicted using Enrichr, based on hub genes. The
screening criterion was adj. p <0.05, and the ranking
was based on the comprehensive score. The protein was
converted to the PDBQT file format so that AutoDock 4
software could recognize and read the modified protein.
To prepare the ligands for docking, charges were added
and optimized. Three PDBQT files were identified:
rigid DEG proteins, flexible proteins, and drug ligands.
Finally, we used AutoDock 4 software to perform
molecular docking.
RESULTS
Identification of the senescence-related genes (SRGs)
The DEGs between the six-hour rat MCAO groups and
sham groups in GSE163654 were discovered and are
shown in a volcano plot (Figure 2A). Among them, 326
genes were upregulated and 199 genes were
downregulated. Subsequently, according to the Venn
plot, 14 upregulated DEGs (CCL3, Jun, CCL4, Il1a,
VGF, VEGFA, IL1B, ANGPTL4, TNF, CCL2, CXCL16,
GEM, ICAM1, CCL7) and two downregulated SRGs
(CXCL12, SELPLG) were involved in the senescence-
related SenMayo dataset (Figure 2B). Specifically, 92
edges were linked between 16 corresponding proteins in
the protein-protein interaction network (Figure 2C).
A heatmap shows the expression of these 16 genes
(Figure 2D). Based on these 16 genes, GO/KEGG
functional enrichment analysis was performed. The
subsequent GO/KEGG functional enrichment analysis
showed that these genes were highly enriched in
leukocyte migration, positive regulation of the ERK1
and ERK2 cascades, cytokine activity, and cytokine
www.aging-us.com 5501 AGING
receptor binding (Figure 2E). Finally, to identify the
conservation of these genes between species, we
compared the expression of these 16 genes in human
peripheral blood between the IS and normal groups
(Figure 2F). Thirteen of these 16 genes were expressed
in both mice and humans, and five (ANGPTL4, CCL3,
CCL7, CXCL16, and TNF) showed statistically
significant differences.
SRGs expression in the peripheral blood of the rat
By analyzing the GSE119121 expression matrix, we
visualized the expression of these 16 SRGs in the
peripheral blood of the MCAO and sham groups
(Figure 3A). We further compared the expression trends
of these SRGs at five different time points (1, 2, 3, 6,
and 24 hours) (Figure 3B). Notably, CXCL16 expression
and GEM decreased in the MCAO group but tended to
recover after 24 hours. The expression levels of VGF,
CCL3, and CCL4 decreased at later time points.
Meanwhile, the expression levels of the other 11 genes
increased at different time points within 24 hours in the
MCAO group, and most of them recovered 24 hours
after the operation. Additionally, the expression of five
SRGs was significantly different at some points after the
MACO operation compared with that before the
operation. Finally, a line graph was drawn to describe
the variation trends of the five hub genes (Figure 3C).
Thus, ANGPTL4, CCL3, CCL7, CXCL16, and TNF have
better species conservation in the peripheral blood of rats
and humans and were identified as HSRGs for further
research.
Figure 2. Discovery of SRGs in rat MCAO model and human peripheral blood. (A) The volcano plot for DEGs of brain tissue in
GSE163654. The genes related to cellular senescence were labeled. Red represents high gene expression and blue represents low expression.
(B) The Venn plot for the distribution of DEGs. (C) The protein-protein interaction network for SRGs. (D) The heatmap for SRGs in GSE163654.
(E) GO/KEGG pathway analysis and protein interaction network of SRGs. The color of the proteins corresponds to the pathway and the
number shows the count of genes. (F) The violin plot for SRGs of human peripheral blood in GSE16561. *p < 0.05, **p < 0.01, ***p < 0.001.
www.aging-us.com 5502 AGING
Validation of HSRGs by RT-qPCR in the MCAO
model
We performed RT-qPCR in the MCAO model to
demonstrate the critical role of HSRGs in IS. To verify
the success of our MCAO model, TTC staining was
performed on the brain tissue of MCAO rats (Figure
4A). TTC staining stains the normal brain tissue red,
while the infarct lesions appear white, allowing a good
evaluation of the infarct in the brain. According to the
RT-qPCR results, the expression levels of all five genes
differed significantly (p <0.05) (Figure 4B). Gene
expression levels were higher in the brain tissues of rats
in the MCAO model than in the sham group, and the
expression trend was consistent with that shown in
Figure 2D between the MCAO and sham groups in
GSE163654.
Construction of a clinical prediction model
Based on the expression levels of HSRGs in GSE16561,
we constructed a nomogram prediction model (Figure
4C). To verify the effectiveness of the model, ROC,
calibration, DCA, and clinical impact curves were plotted
(Figure 4D4G). The area under the curve (AUC) of the
prediction model was approximately 0.956, and the
calibration curve showed good calibration. The DCA
curve showed that this predictive model could yield
significantly greater net benefits for making clinical
decisions. In terms of the clinical impact curve, the
prediction model determined that the population at risk
for IS was strongly matched to the actual population
when the threshold probability was >65% of the
predicted score probability value, confirming the good
clinical efficiency of the prediction model.
Immune cell infiltration analysis
The CIBERSORT algorithm was used to predict
immune cell infiltration in the IS and normal groups.
The bar plot and heatmap displayed the percentage of
each of the 22 types of immune cells in each human
blood sample from GSE16561 (Figure 5A, 5B).
Correlation analysis of immune cells revealed that
resting mast cells and activated mast cells had the most
significant negative correlation, while naïve B cells and
CD8 T cells, follicular helper cells, resting mast cells
and activated CD4+ memory T cells, M2 macrophages
and monocytes, resting dendritic cells and M1
macrophages, neutrophils and activated mast cells had a
significant positive correlation (Figure 5C). The violin
plot of the immune cell infiltration difference showed
that, in comparison to the normal group, patients with
IS had lower levels of CD8+ T cells and activated
NK cells (Figure 5D). Finally, we analyzed HSRG
expression in 22 types of immune cells (Figure 5E).
Figure 3. Expression of SRGs in the rat peripheral blood and identification of HSRGs. (A) The heatmap for SRGs in GSE119121 at
different time points. (B) The violin plot for SRGs in GSE119121. (C) The line graph describes the variation trend of HSRGs expression at
different time points.
www.aging-us.com 5503 AGING
ScRNA-seq reveals the cellular senescence pattern
after IS
Cell clusters were identified by UMAP analysis in
MCAO and sham-operated mice (Figure 6A). We
further annotated the cell clusters through the “SingleR”
package and mapped them to the UMAP (Figure 6B).
Nine cell clusters were identified: astrocytes,
endothelial cells, epithelial cells, fibroblasts,
granulocytes, microglia, monocytes, natural killer cells,
and oligodendrocytes. Subsequently, HSRG expression
in each cell cluster was mapped onto UMAP diagrams
and quantified (Figure 6C, 6D). Finally, we used the
“AUCell” package to evaluate the total hub gene
expressions in all types of cells and cells with an AUC
value greater than 0.078 were adopted (Figure 6E).
Significantly, HSRGs were highly expressed in
microglia and monocytes (Figure 6F). Based on the
UMAP and violin plots, microglia and monocytes had
the highest senescence scores, as shown in Figure 6G.
Intercellular communication and internal signaling
based on scRNA-seq
The intercellular communication in the MCAO and
sham groups is shown, respectively (Figure 7A).
Monocytes, granulocytes, and microglia were more
involved in cellular communication as receptors in the
sham group. Subsequently, we identified microglia and
monocytes as receivers and assessed their cellular
interactions with astrocytes and monocytes (Figure 7B,
7C). Further analysis of transcription factors (TFs)
involved in cellular communication revealed that Mef2c
and Myc were activated when microglia served as
recipients, whereas Fos, Nfkb1, and Stat1 were activated
when monocytes served as recipients. Finally, we
presented TF activities in receiver cells using a TF
enrichment plot, and all TFs were activated in
monocytes, microglia, and granulocytes (Figure 7D).
Cell trajectory analysis of microglia
The cell trajectories of the microglia are presented as
3D images in Figure 8A. Individual clustering and
UMAP mapping showed that microglia were divided
into four clusters (Figure 8B). To annotate these
microglial cell sub-clusters, we identified the top five
cell marker genes in each cluster (Figure 8D). The
marker genes include “P2RY12”, “SIGLECH”,
“GPR34”, mt-ATP8”, “SELPLGof MG1, “CCL12”,
“TNF”, “ADAMTS1”, “SOCS3”, “CCL2” of MG2,
“SPP1”, “LGALS3”, “LPL”, “LILRB4A”, “LILR4B”
of MG3, and “CTLA2A”, “IGFBP7”, “CLDN5”,
“PGLYRP1”, “SLC2A1” of MG4. Analysis of the
differences in the number of cell clusters showed that
MG1 was the main microglia in the sham group,
Figure 4. Validation of HSRGs by rat MCAO model and construction of prediction model. (A) TTC staining verification of rat MCAO
model. (B) Validation of quantitative real-time PCR analysis. (C) Nomogram of HSRGs for predicting IS. A calibration curve (D), Clinical decision
analysis (E, F), and ROC curve (G) of the nomogram.
www.aging-us.com 5504 AGING
whereas MG2, MG3, and MG4 were the main microglia
in the MCAO group (Figure 8E). Additionally,
ANGPTL4 showed specificity for MG4, and its
expression was higher in the MCAO group. In MG2-4,
CCL7, CXCL16, and TNF were highly expressed in the
MCAO group. Although CCL3 was expressed in
different subgroups, its expression level was higher in
the MCAO group (Figure 8F). Finally, the cell
trajectory of microglia was determined to explore their
divergent trajectory.
Figure 5. Immune cell infiltration analysis in human peripheral blood. (A, B) The landscape of immune infiltration between IS and
normal groups in GSE16561. (C) Correlation matrix of all 22 immune cell subtype compositions. Higher, lower, and the same correlation levels
are displayed in red, blue, and white. (D) Comparison of 22 immune cell subtypes between patients in IS and normal groups. (E) The heatmap
for HSRGs in 22 immune cell subtype compositions.
www.aging-us.com 5505 AGING
Drug screening and molecular docking
Small-molecule compounds that may bind to
ANGPTL4, CCL3, CCL7, CXCL16, and TNF were
identified using the DSigDB database; the top 10
compounds are listed in Table 3. Among these, retinoic
acid had the highest combined score (608812). We then
drew a structural diagram of retinoic acid, which can
bind to ANGPTL4, CCL3, CCL7, CXCL16, and TNF
(Figure 9A9F).
DISCUSSION
Senescence has long been a significant issue for
researchers and has accelerated since the occurrence of
IS [6]. Cellular senescence is one of the significant
causes of senescence and has recently attracted
considerable attention [11]. SenMayo is a set of genes
that accurately describes and assesses cellular
senescence [16]. In this study, we aimed to identify the
HSRGs involved in IS and cellular senescence in brain
tissue by the SenMayo gene set. A nomogram model
was constructed based on HSRGs and was evaluated
preliminarily to predict cellular senescence in patients
with IS. Immune activity plays a vital role in cellular
senescence after IS, which has been discussed in detail
using immune cell infiltration analysis. To further
elucidate the mechanism, scRNA-seq analysis was
performed to determine the cellular localization of
HSRGs, intercellular communication, and cellular
Figure 6. The scRNA-seq reveals the expression of HSRGs in mouse brains. (A) Cluster analysis of scRNA-seq in GSE174574 dataset.
Red represents the cells in the MCAO group and blue represents the cells in the Sham group. (B) Cell cluster identification was obtained in
(A). Different colors represent different cell clusters, with a total of 9 identified. (C) Distribution of HSRGs expression in different cell clusters.
Compared with the Sham group, red represents the high expression of genes in the IS group. (D) Quantified expression of HSRGs in different
cell clusters. (E) The distribution of cell AUC value, an AUC value greater than 0.078 were adopted. (F) Distribution of HSRGs expression in
different cell clusters based on AUC value. (G) Quantified AUC value of HSRGs in different cell clusters.
www.aging-us.com 5506 AGING
trajectory. Finally, small molecules that can bind to hub
gene expression proteins are considered potential drugs
for alleviating cellular senescence after IS. Overall, this
study combined multiple bioinformatic analysis
methods and experimental verification to conduct a
rigorous discussion of cellular senescence after IS at
different transcriptome levels, providing a reference for
further research in this field.
Functional enrichment analysis revealed that SRGs
were primarily related to leukocyte migration and
cytokine-cytokine receptor interactions. This suggests
that the immune response plays a significant role in
cellular senescence after an IS. However, while the
immune response can be protective, the invasion of
innate immune cells into the brain and meninges during
the acute phase can exacerbate ischemic damage [25].
Additionally, peripheral organs can become a second
“battlefield” for the immune response after IS. Danger
signals are released from damaged brain cells into the
circulatory system, which then activates systemic
immunity, causing severe immunosuppression, life-
threatening infections, and a poor prognosis [26]. In the
chronic phase, antigen presentation initiates an adaptive
immune response against the brain, which may underlie
the neuropsychiatric sequelae [25]. Studies have also
shown that microglia, astrocytes, foam cells, and
lymphocytes are activated in IS, forming glial scars that
persist for ten years later and are associated with
cognitive decline [27]. During the acute phase of IS, a
significant number of injured immune cells secrete
various cytokines, while some cells exhibit a SASP
pattern, which is an important indicator of cellular
senescence after IS [28].
Figure 7. Intercellular communication analysis based on scRNA-seq. (A) The intercellular communication in the MCAO and sham
groups. Microglia (B) and monocytes (C) as receivers assessed the cellular interactions with astrocytes and monocytes. (D) The TF enrichment
plot in monocytes, microglia, and granulocytes.
www.aging-us.com 5507 AGING
After validating with rat and human blood samples and
RT-qPCR of rat brain tissue, ANGPTL4, CCL3, CCL7,
CXCL16, and TNF were identified as HSRGs.
ANGPTL4 is a protein associated with endothelial cell
integrity, inflammation, oxidative stress, and lipid
metabolism and may be involved in the pathogenesis
of IS [29]. CCL3 and CCL7, as chemokines, and
CXCL16, as chemokine ligands, are associated with
the recruitment and activation of inflammatory cells,
neuronal survival, and neoangiogenesis, and are
important mediators of IS [30]. Previous studies have
reported that CCL3 may play an important role in
neutrophil recruitment and the development of
atherosclerosis [31]. Moreover, Waśkiel-Burnat et al.
recently reported that CCL7 may be a significant
biomarker of atherosclerosis [32]. Additionally, CXCL16
is implicated in the immune inflammatory response to
atherosclerotic plaques, from antigen recognition to the
migration and infiltration of immune cells into areas of
inflammation [33, 34]. At the same time, TNF is not
only associated with neuroinflammation after IS but also
promotes SASP-stimulated lysosomal extravasation,
Figure 8. Cell trajectory analysis and identification of microglia. (A) 3D images of cell trajectories and the microglia part are amplified.
(B) Individual clustering and UMAP mapping for microglia. (C) Cell trajectories of the microglia. (D) The bubble pattern of the top five cell
marker genes in four microglia clusters. (E) Distribution of four cell clusters in the IS and Sham groups. (F) Quantified expression of HSRGs in
four cell clusters.
www.aging-us.com 5508 AGING
Table 3. The top 10 compounds bind to HSRGs.
Term
Adjusted P-value
Combined score
Genes
Retinoic acid
0.012148
608812
CCL7, CCL3, ANGPTL4, TNF, CXCL16
Roflumilast
0.001246
15085.69
CCL3, TNF
indinavir
0.001246
13656.83
CCL3, TNF
PCI-24781
0.001246
12459.14
CCL7, CCL3
Lopinavir
0.001609
8557.58
CCL3, TNF
Antimycin A
0.002311
5832.819
CCL3, TNF
isoproterenol
5.20E-04
5550.691
CCL7, CCL3, TNF
Honokiol
0.002577
5105.231
CCL3, TNF
15-Acetyldeoxynivalenol
5.20E-04
4787.728
CCL3, ANGPTL4, TNF
palmitic acid
0.003584
3921.078
CCL3, TNF
leading to cellular senescence [35]. Sequencing data
from the brain tissue in the MCAO model demonstrated
good agreement with our PCR validation results, but a
degree of variability was observed in the peripheral
blood. For the lack of data from human brain tissue, we
combined data from human peripheral blood to
determine HSRGs. In human peripheral blood, CCL3
and TNF showed decreased expression after IS, which
may be related to the blood-brain barrier. However, it
still indicates that the expression changes of HSRGs
after IS are more sensitive than other SRGs. Moreover,
we noticed that the expression of Cxcl16 and Tnf in rat
peripheral blood was different from that in humans, and
the Temporal analysis showed that their expression trend
changed again 6 hours after MCAO. We supposed that
the difference is due to different stages of disease
development at different points in time. Overall, the
HSRGs we identified were species-conserved and
showed some efficacy in predicting the onset of IS.
Our study also suggests that HSRGs expression may
change over time and that predictive models may need
to be adjusted over time. After IS, damaged neuronal
cells release large amounts of senescence-associated
cytokines that affect immune cell function [13].
Therefore, we focused on the different immune cell type
expressions in the peripheral blood of patients with IS.
Notably, the IS group had lower numbers of CD8+
Figure 9. Molecular docking of proteins corresponding to HSRGs and retinoic acid. (A) The structural diagram of retinoic acid. (BF)
Docking simulation of proteins and small molecule compounds.
www.aging-us.com 5509 AGING
T cells and activated NK cells. Additionally, HSRGs
were significantly differentially expressed in neutrophils,
naïve B cells, CD8+ T cells, and T-cell follicular helper
cells, particularly in neutrophils and CD8+ T cells, where
all hub genes were differentially expressed. CD8+ T cells
in the peripheral blood migrate to the brain parenchyma
after IS [36]. Ritzel et al. recently reported that CD8+ T
cells enhance inflammation and leukocyte recruitment
and act as a marker of senescence of the central nervous
system [37]. Moreover, the majority of research
suggested that the dynamics of NK cells in IS are
characterized by an increase in the brain and a decrease
in the peripheral blood, which was consistent with our
results [38, 39]. Meanwhile, brain ischemia weakens NK
cell-mediated immune defenses by interfering with
neurogenic and intracellular pathways [40].
Among the various cell types in IS, microglia and
monocytes are prone to show a cellular senescence
phenotype in the brain tissue (Figure 6). Microglia play
a key role in IS as resident central nervous system
immune cells and are a double-edged sword for neural
healing [41, 42]. Raffaele et al. have shown that
microglia release microcytes that enhance the prognosis
of IS by limiting the senescence of immune cells and
promoting the formation of oligodendrocytes [43].
Furthermore, senescence-associated microglia can
substantially affect brain homeostasis, particularly iron
storage and metabolism, leading to senescence-related
susceptibility and poor functional recovery after IS
[44, 45]. Several studies have also indicated that the
cellular senescence of monocytes is an important
feature of immune-senescence that can delay or
accelerate the establishment of atherosclerotic plaques
[46, 47]. The present study further resolved the issue of
communication between these cells and glial cells. We
found that the intensity of cellular communication
between granulocytes, microglia, and monocytes, which
act as receivers in the MCAO group, was significantly
increased compared to other cells, further illustrating
the important role of the immune response after IS
(Figure 7A). Taken together, we suggest that
intervention in the cellular senescence phenotype of
immune cells, especially microglia and monocytes, may
be the key to reducing senescence and improving the
prognosis of IS.
Microglia are intrinsic brain cells that are important for
senescence. After subpopulation analysis of microglia,
MG4 was found to be closely related to cellular
senescence owing to higher levels of hub gene
expression in the MCAO group. Based on marker
genes, MG4 cells were identified as vessel-associated
microglia, maintaining blood-brain barrier integrity via
Claudin-5 expression, a tight-junction protein. Vessel-
associated microglia maintain BBB integrity at first by
expressing the tight-junction protein Claudin-5 and
making physical contact with endothelial cells, while
microglia phagocytose astrocytic end-feet and disrupt
BBB function during chronic inflammation [48].
Furthermore, retinoic acid was identified as a small
compound that could bind to HSRGs. The positive
effect of retinoic acid in improving the prognosis of IS
possibly relies on improving blood-brain barrier
disruption and reducing apoptosis and neuronal damage,
which has been demonstrated in animal studies but is
still lacking in clinical studies [4951]. The reversal
effects of retinoic acid on cellular senescence
phenotypes have also been documented [52, 53]. We
believe that retinoic acid could be used as a possible
medication to ameliorate the cellular senescence
phenotype and improve the prognosis of IS.
Even though we rigorously discussed the senescence of
various cell types after IS, this study has some
limitations. First, although a clinical prediction model
constructed based on HSRGs was verified, further
verification using external data is lacking. Additionally,
vessel-associated microglia have been identified to play
an important role in cellular senescence, and further
flow cytometry to verify their function. Moreover,
owing to the lack of data from neuronal cells, the
cellular senescence of neurons after IS was not
discussed in this study. Finally, retinoic acid has been
identified as a potential drug for improving the cellular
senescence phenotype and prognosis of IS; however,
further experimental validation is required.
Abbreviations
IS: ischemia stroke; SASP: senescence-associated
secretory phenotype; scRNA-seq: single-cell RNA
sequencing; HSRG: hub senescence-related gene; bulk
RNA-seq: bulk RNA-sequencing; MCAO: middle
cerebral artery occlusion; DEGs: differentially
expressed genes; GO: gene ontology; KEGG: Kyoto
Encyclopedia of Genes and Genomes; TTC: triphenyl
tetrazolium chloride; RT-qPCR: reverse transcription
polymerase chain reaction; ROC: receiver operating
characteristic; DCA: decision curve analysis; UMAP:
uniform manifold approximation and projection; SRGs:
senescence-related genes; AUC: area under the curve;
TFs: transcription factors.
AUTHOR CONTRIBUTIONS
S.G. and P.R. designed the experiments; L.F. and B.Y.
performed the bulk RNA-seq data analysis; L.F., Y.Z.,
and S.C. designed and performed scRNA-seq data
analysis; Y.T. and B.L. performed the establishment of
the MCAO model; H.C. and S.Y. performed the qPCR
experiments; Y.H. and H.H. bred the mice; L.F., B.Y.,
www.aging-us.com 5510 AGING
P.R., J.L., and S.G. prepared the manuscript; All the
authors edited the manuscript.
ACKNOWLEDGMENTS
We thank Mr. Xiaofei Wang at Biomedical
Experimental Center of Xi’an Jiaotong University for
their assistance with the experiment and data analysis.
CONFLICTS 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.
ETHICAL STATEMENT
The animal study was reviewed and approved by Xian
Jiaotong University.
FUNDING
This work was supported by Xi‘an Science and
Technology Plan (21YXYJ0116), the Key Research and
Development Project of Shaanxi Province (Grant
No.2022ZDLSF04-01, and No.2019KW-071), The
National Natural Science Foundation of China (Grant
No. 81971766, and Grant No. 81903268), and China
Postdoctoral Science Foundation (No.2021M692577).
REFERENCES
1. Katan M, Luft A. Global Burden of Stroke. Semin
Neurol. 2018; 38:20811.
https://doi.org/10.1055/s-0038-1649503
PMID:29791947
2. Feigin VL, Vos T, Nichols E, Owolabi MO, Carroll WM,
Dichgans M, Deuschl G, Parmar P, Brainin M, Murray C.
The global burden of neurological disorders:
translating evidence into policy. Lancet Neurol. 2020;
19:25565.
https://doi.org/10.1016/S1474-4422(19)30411-9
PMID:31813850
3. Campbell BCV, Khatri P. Stroke. Lancet. 2020;
396:12942.
https://doi.org/10.1016/S0140-6736(20)31179-X
PMID:32653056
4. Feske SK. Ischemic Stroke. Am J Med. 2021;
134:145764.
https://doi.org/10.1016/j.amjmed.2021.07.027
PMID:34454905
5. Ma Q, Li R, Wang L, Yin P, Wang Y, Yan C, Ren Y, Qian Z,
Vaughn MG, McMillin SE, Hay SI, Naghavi M, Cai M,
et al. Temporal trend and attributable risk factors of
stroke burden in China, 1990-2019: an analysis for the
Global Burden of Disease Study 2019. Lancet Public
Health. 2021; 6:e897906.
https://doi.org/10.1016/S2468-2667(21)00228-0
PMID:34838196
6. Roy AL, Sierra F, Howcroft K, Singer DS, Sharpless N,
Hodes RJ, Wilder EL, Anderson JM. A Blueprint
for Characterizing Senescence. Cell. 2020;
183:11436.
https://doi.org/10.1016/j.cell.2020.10.032
PMID:33128870
7. Weaver NA, Kuijf HJ, Aben HP, Abrigo J, Bae HJ,
Barbay M, Best JG, Bordet R, Chappell FM, Chen CPLH,
Dondaine T, van der Giessen RS, Godefroy O, et al.
Strategic infarct locations for post-stroke cognitive
impairment: a pooled analysis of individual patient
data from 12 acute ischaemic stroke cohorts. Lancet
Neurol. 2021; 20:44859.
https://doi.org/10.1016/S1474-4422(21)00060-0
PMID:33901427
8. Regenhardt RW, Das AS, Lo EH, Caplan LR. Advances in
Understanding the Pathophysiology of Lacunar Stroke:
A Review. JAMA Neurol. 2018; 75:127381.
https://doi.org/10.1001/jamaneurol.2018.1073
PMID:30167649
9. Roy-O’Reilly M, McCullough LD. Age and Sex Are
Critical Factors in Ischemic Stroke Pathology.
Endocrinology. 2018; 159:312031.
https://doi.org/10.1210/en.2018-00465
PMID:30010821
10. López-Otín C, Blasco MA, Partridge L, Serrano M,
Kroemer G. Hallmarks of aging: An expanding universe.
Cell. 2023; 186:24378.
https://doi.org/10.1016/j.cell.2022.11.001
PMID:36599349
11. Calcinotto A, Kohli J, Zagato E, Pellegrini L, Demaria M,
Alimonti A. Cellular Senescence: Aging, Cancer, and
Injury. Physiol Rev. 2019; 99:104778.
https://doi.org/10.1152/physrev.00020.2018
PMID:30648461
12. Hernandez-Segura A, Nehme J, Demaria M. Hallmarks
of Cellular Senescence. Trends Cell Biol. 2018;
28:43653.
https://doi.org/10.1016/j.tcb.2018.02.001
PMID:29477613
13. Torres-Querol C, Torres P, Vidal N, Portero-Otín M,
Arque G, Purroy F. Acute ischemic stroke triggers a
cellular senescence-associated secretory phenotype.
Sci Rep. 2021; 11:15752.
https://doi.org/10.1038/s41598-021-95344-5
PMID:34344977
www.aging-us.com 5511 AGING
14. Baixauli-Martín J, Aliena-Valero A, Castelló-Ruiz M,
Burguete MC, López-Morales MA, Muñoz-Espín D,
Torregrosa G, Salom JB. Brain Cell Senescence: A New
Therapeutic Target for the Acute Treatment of
Ischemic Stroke. J Neuropathol Exp Neurol. 2022;
81:61420.
https://doi.org/10.1093/jnen/nlac048 PMID:35763058
15. Zhang H, Lin S, McElroy CL, Wang B, Jin D, Uteshev
VV, Jin K. Circulating Pro-Inflammatory Exosomes
Worsen Stroke Outcomes in Aging. Circ Res. 2021;
129:e12140.
https://doi.org/10.1161/CIRCRESAHA.121.318897
PMID:34399581
16. Saul D, Kosinsky RL, Atkinson EJ, Doolittle ML, Zhang X,
LeBrasseur NK, Pignolo RJ, Robbins PD, Niedernhofer
LJ, Ikeno Y, Jurk D, Passos JF, Hickson LJ, et al. A new
gene set identifies senescent cells and predicts
senescence-associated pathways across tissues. Nat
Commun. 2022; 13:4827.
https://doi.org/10.1038/s41467-022-32552-1
PMID:35974106
17. Yousefzadeh MJ, Flores RR, Zhu Y, Schmiechen ZC,
Brooks RW, Trussoni CE, Cui Y, Angelini L, Lee KA,
McGowan SJ, Burrack AL, Wang D, Dong Q, et al. An
aged immune system drives senescence and ageing of
solid organs. Nature. 2021; 594:1005.
https://doi.org/10.1038/s41586-021-03547-7
PMID:33981041
18. Ding YN, Wang HY, Chen HZ, Liu DP. Targeting
senescent cells for vascular aging and related diseases.
J Mol Cell Cardiol. 2022; 162:4352.
https://doi.org/10.1016/j.yjmcc.2021.08.009
PMID:34437878
19. Kritsilis M, V Rizou S, Koutsoudaki PN, Evangelou K,
Gorgoulis VG, Papadopoulos D. Ageing, Cellular
Senescence and Neurodegenerative Disease. Int J Mol
Sci. 2018; 19:2937.
https://doi.org/10.3390/ijms19102937 PMID:30261683
20. Longa EZ, Weinstein PR, Carlson S, Cummins R.
Reversible middle cerebral artery occlusion without
craniectomy in rats. Stroke. 1989; 20:8491.
https://doi.org/10.1161/01.str.20.1.84 PMID:2643202
21. Newman AM, Steen CB, Liu CL, Gentles AJ, Chaudhuri
AA, Scherer F, Khodadoust MS, Esfahani MS, Luca BA,
Steiner D, Diehn M, Alizadeh AA. Determining cell type
abundance and expression from bulk tissues with
digital cytometry. Nat Biotechnol. 2019; 37:77382.
https://doi.org/10.1038/s41587-019-0114-2
PMID:31061481
22. Satija R, Farrell JA, Gennert D, Schier AF, Regev A.
Spatial reconstruction of single-cell gene expression
data. Nat Biotechnol. 2015; 33:495502.
https://doi.org/10.1038/nbt.3192
PMID:25867923
23. Zhang Y, Liu T, Hu X, Wang M, Wang J, Zou B, Tan P, Cui
T, Dou Y, Ning L, Huang Y, Rao S, Wang D, Zhao X.
CellCall: integrating paired ligand-receptor and
transcription factor activities for cell-cell
communication. Nucleic Acids Res. 2021; 49:852034.
https://doi.org/10.1093/nar/gkab638 PMID:34331449
24. Cao J, Spielmann M, Qiu X, Huang X, Ibrahim DM, Hill
AJ, Zhang F, Mundlos S, Christiansen L, Steemers FJ,
Trapnell C, Shendure J. The single-cell transcriptional
landscape of mammalian organogenesis. Nature. 2019;
566:496502.
https://doi.org/10.1038/s41586-019-0969-x
PMID:30787437
25. Iadecola C, Buckwalter MS, Anrather J. Immune
responses to stroke: mechanisms, modulation, and
therapeutic potential. J Clin Invest. 2020; 130:277788.
https://doi.org/10.1172/JCI135530 PMID:32391806
26. Wu F, Liu Z, Zhou L, Ye D, Zhu Y, Huang K, Weng Y,
Xiong X, Zhan R, Shen J. Systemic immune responses
after ischemic stroke: From the center to the
periphery. Front Immunol. 2022; 13:911661.
https://doi.org/10.3389/fimmu.2022.911661
PMID:36211352
27. Doyle KP, Quach LN, Solé M, Axtell RC, Nguyen TVV,
Soler-Llavina GJ, Jurado S, Han J, Steinman L, Longo
FM, Schneider JA, Malenka RC, Buckwalter MS. B-
lymphocyte-mediated delayed cognitive impairment
following stroke. J Neurosci. 2015; 35:213345.
https://doi.org/10.1523/JNEUROSCI.4098-14.2015
PMID:25653369
28. Zafar A, Farooqui M, Ikram A, Suriya S, Kempuraj D,
Khan M, Tasneem N, Qaryouti D, Quadri S, Adams HP,
Ortega-Gutierrez S, Leira E, Zaheer A. Cytokines, brain
proteins, and growth factors in acute stroke patients: A
pilot study. Surg Neurol Int. 2021; 12:366.
https://doi.org/10.25259/SNI_569_2021
PMID:34513133
29. Xu L, Guo ZN, Yang Y, Xu J, Burchell SR, Tang J, Zhang J,
Xu J, Zhang JH. Angiopoietin-like 4: A double-edged
sword in atherosclerosis and ischemic stroke? Exp
Neurol. 2015; 272:616.
https://doi.org/10.1016/j.expneurol.2015.05.020
PMID:26033474
30. Mirabelli-Badenier M, Braunersreuther V, Viviani GL,
Dallegri F, Quercioli A, Veneselli E, Mach F,
Montecucco F. CC and CXC chemokines are pivotal
mediators of cerebral injury in ischaemic stroke.
Thromb Haemost. 2011; 105:40920.
https://doi.org/10.1160/TH10-10-0662
PMID:21174009
www.aging-us.com 5512 AGING
31. de Jager SCA, Bot I, Kraaijeveld AO, Korporaal SJA, Bot
M, van Santbrink PJ, van Berkel TJC, Kuiper J, Biessen
EAL. Leukocyte-specific CCL3 deficiency inhibits
atherosclerotic lesion development by affecting
neutrophil accumulation. Arterioscler Thromb Vasc
Biol. 2013; 33:e7583.
https://doi.org/10.1161/ATVBAHA.112.300857
PMID:23288165
32. Waśkiel-Burnat A, Niemczyk A, Blicharz L, Chmielińska
P, Zaremba M, Gąsecka A, Filipiak KJ, Olszewska M,
Rudnicka L. Chemokine C-C Motif Ligand 7 (CCL7), a
Biomarker of Atherosclerosis, Is Associated with the
Severity of Alopecia Areata: A Preliminary Study. J Clin
Med. 2021; 10:5418.
https://doi.org/10.3390/jcm10225418 PMID:34830700
33. Gencer S, Evans BR, van der Vorst EPC, Döring Y,
Weber C. Inflammatory Chemokines in Atherosclerosis.
Cells. 2021; 10:226.
https://doi.org/10.3390/cells10020226
PMID:33503867
34. Yin R, Ma A, Pan X, Yang S. Biomarkers of cerebral
microembolic signals. Clin Chim Acta. 2017; 475:1648.
https://doi.org/10.1016/j.cca.2017.10.028
PMID:29111093
35. Bae EJ, Choi M, Kim JT, Kim DK, Jung MK, Kim C, Kim
TK, Lee JS, Jung BC, Shin SJ, Rhee KH, Lee SJ. TNF-α
promotes α-synuclein propagation through stimulation
of senescence-associated lysosomal exocytosis. Exp
Mol Med. 2022; 54:788800.
https://doi.org/10.1038/s12276-022-00789-x
PMID:35790884
36. Lee J, McCullough LD. Revisiting regulatory T cells for
stroke therapy. J Clin Invest. 2022; 132:e161703.
https://doi.org/10.1172/JCI161703
PMID:35912860
37. Ritzel RM, Crapser J, Patel AR, Verma R, Grenier JM,
Chauhan A, Jellison ER, McCullough LD. Age-Associated
Resident Memory CD8 T Cells in the Central Nervous
System Are Primed To Potentiate Inflammation after
Ischemic Brain Injury. J Immunol. 2016; 196:331830.
https://doi.org/10.4049/jimmunol.1502021
PMID:26962232
38. Chen C, Ai QD, Chu SF, Zhang Z, Chen NH. NK cells in
cerebral ischemia. Biomed Pharmacother. 2019;
109:54754.
https://doi.org/10.1016/j.biopha.2018.10.103
PMID:30399590
39. Liu Q, Jin WN, Liu Y, Shi K, Sun H, Zhang F, Zhang C,
Gonzales RJ, Sheth KN, La Cava A, Shi FD. Brain
Ischemia Suppresses Immunity in the Periphery and
Brain via Different Neurogenic Innervations. Immunity.
2017; 46:47487.
https://doi.org/10.1016/j.immuni.2017.02.015
PMID:28314594
40. Feng Y, Li Y, Zhang Y, Zhang BH, Zhao H, Zhao X, Shi FD,
Jin WN, Zhang XA. miR-1224 contributes to ischemic
stroke-mediated natural killer cell dysfunction by
targeting Sp1 signaling. J Neuroinflammation. 2021;
18:133.
https://doi.org/10.1186/s12974-021-02181-4
PMID:34118948
41. Jia J, Yang L, Chen Y, Zheng L, Chen Y, Xu Y, Zhang M.
The Role of Microglial Phagocytosis in Ischemic Stroke.
Front Immunol. 2022; 12:790201.
https://doi.org/10.3389/fimmu.2021.790201
PMID:35082781
42. Wang Y, Leak RK, Cao G. Microglia-mediated
neuroinflammation and neuroplasticity after stroke.
Front Cell Neurosci. 2022; 16:980722.
https://doi.org/10.3389/fncel.2022.980722
PMID:36052339
43. Raffaele S, Gelosa P, Bonfanti E, Lombardi M,
Castiglioni L, Cimino M, Sironi L, Abbracchio MP,
Verderio C, Fumagalli M. Microglial vesicles improve
post-stroke recovery by preventing immune cell
senescence and favoring oligodendrogenesis. Mol
Ther. 2021; 29:143958.
https://doi.org/10.1016/j.ymthe.2020.12.009
PMID:33309882
44. Shi L, Rocha M, Zhang W, Jiang M, Li S, Ye Q, Hassan
SH, Liu L, Adair MN, Xu J, Luo J, Hu X, Wechsler LR,
et al. Genome-wide transcriptomic analysis of
microglia reveals impaired responses in aged mice
after cerebral ischemia. J Cereb Blood Flow Metab.
2020; 40:S4966.
https://doi.org/10.1177/0271678X20925655
PMID:32438860
45. Greenwood EK, Brown DR. Senescent Microglia: The
Key to the Ageing Brain? Int J Mol Sci. 2021;
22:4402.
https://doi.org/10.3390/ijms22094402
PMID:33922383
46. Wu CM, Zheng L, Wang Q, Hu YW. The emerging role
of cell senescence in atherosclerosis. Clin Chem Lab
Med. 2020; 59:2738.
https://doi.org/10.1515/cclm-2020-0601
PMID:32692694
47. Vellasamy DM, Lee SJ, Goh KW, Goh BH, Tang YQ, Ming
LC, Yap WH. Targeting Immune Senescence in
Atherosclerosis. Int J Mol Sci. 2022; 23:13059.
https://doi.org/10.3390/ijms232113059
PMID:36361845
48. Haruwaka K, Ikegami A, Tachibana Y, Ohno N,
Konishi H, Hashimoto A, Matsumoto M, Kato D, Ono R,
www.aging-us.com 5513 AGING
Kiyama H, Moorhouse AJ, Nabekura J, Wake H. Dual
microglia effects on blood brain barrier permeability
induced by systemic inflammation. Nat Commun.
2019; 10:5816.
https://doi.org/10.1038/s41467-019-13812-z
PMID:31862977
49. Kong L, Wang Y, Wang XJ, Wang XT, Zhao Y, Wang LM,
Chen ZY. Retinoic acid ameliorates blood-brain barrier
disruption following ischemic stroke in rats. Pharmacol
Res. 2015; 99:12536.
https://doi.org/10.1016/j.phrs.2015.05.014
PMID:26066585
50. Kang JB, Park DJ, Shah MA, Koh PO. Retinoic acid exerts
neuroprotective effects against focal cerebral ischemia
by preventing apoptotic cell death. Neurosci Lett.
2021; 757:135979.
https://doi.org/10.1016/j.neulet.2021.135979
PMID:34023410
51. Kang JB, Park DJ, Koh PO. Retinoic Acid Prevents the
Neuronal Damage Through the Regulation of
Parvalbumin in an Ischemic Stroke Model. Neurochem
Res. 2023; 48:487501.
https://doi.org/10.1007/s11064-022-03769-9
PMID:36245066
52. Zhao J, Jiang X, Yan L, Lin J, Guo H, Yu S, Ye B, Zhu J,
Zhang W. Retinoic acid inducible gene-I slows down
cellular senescence through negatively regulating the
integrin β3/p38 MAPK pathway. Cell Cycle. 2019;
18:337892.
https://doi.org/10.1080/15384101.2019.1677074
PMID:31595820
53. Park SH, Lim JS, Jang KL. All-trans retinoic acid induces
cellular senescence via upregulation of p16, p21, and
p27. Cancer Lett. 2011; 310:2329.
https://doi.org/10.1016/j.canlet.2011.07.009
PMID:21803488
... In contrast to previous study by Torres-Querol et al. [33], Baixauli-Martin et al. [79] proposed lipofuscin as a superior marker of senescence over SA-β-gal. In silico analysis of bulk RNA sequence data from public databases showed a differential expression of senescence-related genes, such as angiopoietin-like protein (ANGPTL4), Ccl3, Ccl7, Cxcl16, and Tnf, after stroke in a species-conserved manner [81]. ANGPTL4 is an endothelial protein associated with the pathogenesis of ischemic stroke and inflammation [82]. ...
... The expression of these genes was further confirmed by qPCR analysis in rat brain tissues after MCAO. A Further single RNA sequence demonstrated that senescence-related genes are highly expressed in microglia and monocytes, more precisely in MG4 microglia, a sub-population of microglia involved in blood-brain barrier integrity [81]. A summary of studies on the role of senescence and SASP in ischemic stroke is listed in Table 1. ...
Article
Full-text available
Aging is a known co-morbidity of ischemic stroke with its risk and severity increasing every year past 55+. While many of the current stroke therapies have shown success in reducing mortality, post-stroke morbidity has not seen the same substantial reduction. Recently, the involvement of cellular senescence and SASP in brain injury and neurological degeneration has been recognized. Ischemic injury causes oxidative stress and mitochondrial damage that induces senescence through the activation of p21 and p16 pathways, ultimately leading to synthesis and release of senescence-associated secretory phenotype (SASP). This ischemic event causes stress-induced premature senescence (SIPS), aging the brain decades beyond the standard biological age due to an increase in senescent cells in the ischemic core and ipsilateral hemisphere. Therefore, therapies that target the senescent cells and SASP, including senolytics, senomorphic drugs, stem cell therapies, and other cell-specific interventions, may be a new path for stroke treatment.
... However, excessive and prolonged inflammatory stimulation occurring at chronic stages of IS and SCI has been demonstrated to exhaust the pro-regenerative properties of microglial cells, which instead become dystrophic, acquire senescence-like traits, and contribute to damage progression and worsening of the functional outcome (Brennan et al., 2022;Raffaele et al., 2021). Indeed, at late injury stages, phagocytic microglia almost disappear and are replaced by cells displaying upregulation of proinflammatory and iron processing pathways, which are no longer able to sustain tissue repair (Brennan et al., 2022;Fu et al., 2023;Jin et al., 2023). On this basis, gaining a better understanding of the mechanisms that drive and maintain protective microglial responses to CNS injury may uncover novel therapeutic targets to promote tissue repair, by preventing the transition of microglia towards a dysfunctional and damaging state. ...
Article
Full-text available
Microglia, the resident immune cells of the central nervous system (CNS), play a major role in damage progression and tissue remodeling after acute CNS injury, including ischemic stroke (IS) and spinal cord injury (SCI). Understanding the molecular mechanisms regulating microglial responses to injury may thus reveal novel therapeutic targets to promote CNS repair. Here, we investigated the role of microglial tumor necrosis factor receptor 2 (TNFR2), a transmembrane receptor previously associated with pro-survival and neuroprotective responses, in shaping the neuroinflammatory environment after CNS injury. By inducing experimental IS and SCI in Cx3cr1CreER:Tnfrsf1bfl/fl mice, selectively lacking TNFR2 in microglia, and corresponding tnfrsf1bfl/fl littermate controls, we found that ablation of microglial TNFR2 significantly reduces lesion size and pro-inflammatory cytokine levels, and favors infiltration of leukocytes after injury. Interestingly, these effects were paralleled by opposite sex-specific modifications of microglial reactivity, which was found to be limited in female TNFR2-ablated mice compared to controls, whereas it was enhanced in males. In addition, we show that TNFR2 protein levels in the cerebrospinal fluid (CSF) of human subjects affected by IS and SCI, as well as healthy donors, significantly correlate with disease stage and severity, representing a valuable tool to monitor the inflammatory response after acute CNS injury. Hence, these results advance our understanding of the mechanisms regulating microglia reactivity after acute CNS injury, aiding the development of sex- and microglia-specific, personalized neuroregenerative strategies.
... Taken together, single-cell sequencing technology plays a key role in solving AIS research and can deeply reveal the molecular mechanism of stroke, treatment targets, and the development of personalized treatment strategies. 50,145,178,187,188 Although single-cell sequencing technology is now widely used, it is undeniable that single-cell sequencing technology currently has certain limitations. Single-cell sequencing technology also presents a number of technical challenges when dealing with single cells. ...
Article
Full-text available
Background Acute ischemic stroke (AIS) is a common cerebrovascular event associated with high incidence, disability, and poor prognosis. Studies have shown that various cell types, including microglia, astrocytes, oligodendrocytes, neurons, and neutrophils, play complex roles in the early stages of AIS and significantly affect its prognosis. Thus, a comprehensive understanding of the mechanisms of action of these cells will be beneficial for improving stroke prognosis. With the rapid development of single‐cell sequencing technology, researchers have explored the pathophysiological mechanisms underlying AIS at the single‐cell level. Method We systematically summarize the latest research on single‐cell sequencing in AIS. Result In this review, we summarize the phenotypes and functions of microglia, astrocytes, oligodendrocytes, neurons, neutrophils, monocytes, and lymphocytes, as well as their respective subtypes, at different time points following AIS. In particular, we focused on the crosstalk between microglia and astrocytes, oligodendrocytes, and neurons. Our findings reveal diverse and sometimes opposing roles within the same cell type, with the possibility of interconversion between different subclusters. Conclusion This review offers a pioneering exploration of the functions of various glial cells and cell subclusters after AIS, shedding light on their regulatory mechanisms that facilitate the transformation of detrimental cell subclusters towards those that are beneficial for improving the prognosis of AIS. This approach has the potential to advance the discovery of new specific targets and the development of drugs, thus representing a significant breakthrough in addressing the challenges in AIS treatment.
Article
Full-text available
Atherosclerosis is one of the main underlying causes of cardiovascular diseases (CVD). It is associated with chronic inflammation and intimal thickening as well as the involvement of multiple cell types including immune cells. The engagement of innate or adaptive immune response has either athero-protective or atherogenic properties in exacerbating or alleviating atherosclerosis. In atherosclerosis, the mechanism of action of immune cells, particularly monocytes, macrophages, dendritic cells, and B- and T-lymphocytes have been discussed. Immuno-senescence is associated with aging, viral infections, genetic predispositions, and hyperlipidemia, which contribute to atherosclerosis. Immune senescent cells secrete SASP that delays or accelerates atherosclerosis plaque growth and associated pathologies such as aneurysms and coronary artery disease. Senescent cells undergo cell cycle arrest, morphological changes, and phenotypic changes in terms of their abundances and secretome profile including cytokines, chemokines, matrix metalloproteases (MMPs) and Toll-like receptors (TLRs) expressions. The senescence markers are used in therapeutics and currently, senolytics represent one of the emerging treatments where specific targets and clearance of senescent cells are being considered as therapy targets for the prevention or treatment of atherosclerosis.
Article
Full-text available
Ischemic stroke is a neurological disease that causes brain damage by increasing oxidative stress and ion imbalance. Retinoic acid is a major metabolite of vitamin A and regulates oxidative stress, calcium homeostasis, and cell death. Intracellular calcium is involved in neuronal growth and synaptic plasticity. Parvalbumin is a calcium-binding protein that is mainly expressed in brain. In this study, we investigated whether retinoic acid has neuroprotective effects by controlling intracellular calcium concentration and parvalbumin expression in ischemic brain damage. Middle cerebral artery occlusion (MCAO) was performed to induce cerebral ischemia. Retinoic acid (5 mg/kg) or vehicle was injected into the abdominal cavity for four days before surgery and cerebral cortices were collected 24 h after MCAO for further studies. MCAO damage induced neurological deficits and histopathological changes and decreased parvalbumin expression. However, retinoic acid treatment alleviated these changes. In cultured neurons, glutamate (5 mM) exposure induced neuronal cell death, increased intracellular calcium concentration, and decreased parvalbumin expression. Retinoic acid treatment attenuated these changes against glutamate toxicity in a dose-dependent manner. It also regulates glutamate induced change in bcl-2 and bax expression. The mitigation effects of retinoic acid were greater under non-transfection conditions than under parvalbumin siRNA transfection conditions. Our findings showed that retinoic acid modulates intracellular calcium concentration and parvalbumin expression and prevents apoptosis in ischemic brain injury. In conclusion, retinoic acid contributes to the preservation of neurons from ischemic stroke by controlling parvalbumin expression and apoptosis-related proteins.
Article
Full-text available
Ischemic stroke is a leading cause of disability and death. It imposes a heavy economic burden on individuals, families and society. The mortality rate of ischemic stroke has decreased with the help of thrombolytic drug therapy and intravascular intervention. However, the nerve damage caused by ischemia-reperfusion is long-lasting and followed by multiple organ dysfunction. In this process, the immune responses manifested by systemic inflammatory responses play an important role. It begins with neuroinflammation following ischemic stroke. The large number of inflammatory cells released after activation of immune cells in the lesion area, along with the deactivated neuroendocrine and autonomic nervous systems, link the center with the periphery. With the activation of systemic immunity and the emergence of immunosuppression, peripheral organs become the second “battlefield” of the immune response after ischemic stroke and gradually become dysfunctional and lead to an adverse prognosis. The purpose of this review was to describe the systemic immune responses after ischemic stroke. We hope to provide new ideas for future research and clinical treatments to improve patient outcomes and quality of life.
Article
Full-text available
Although cellular senescence drives multiple age-related co-morbidities through the senescence-associated secretory phenotype, in vivo senescent cell identification remains challenging. Here, we generate a gene set (SenMayo) and validate its enrichment in bone biopsies from two aged human cohorts. We further demonstrate reductions in SenMayo in bone following genetic clearance of senescent cells in mice and in adipose tissue from humans following pharmacological senescent cell clearance. We next use SenMayo to identify senescent hematopoietic or mesenchymal cells at the single cell level from human and murine bone marrow/bone scRNA-seq data. Thus, SenMayo identifies senescent cells across tissues and species with high fidelity. Using this senescence panel, we are able to characterize senescent cells at the single cell level and identify key intercellular signaling pathways. SenMayo also represents a potentially clinically applicable panel for monitoring senescent cell burden with aging and other conditions as well as in studies of senolytic drugs.
Article
Full-text available
Stroke remains a major cause of long-term disability and mortality worldwide. The immune system plays an important role in determining the condition of the brain following stroke. As the resident innate immune cells of the central nervous system, microglia are the primary responders in a defense network covering the entire brain parenchyma, and exert various functions depending on dynamic communications with neurons, astrocytes, and other neighboring cells under both physiological or pathological conditions. Microglia activation and polarization is crucial for brain damage and repair following ischemic stroke, and is considered a double-edged sword for neurological recovery. Microglia can exist in pro-inflammatory states and promote secondary brain damage, but they can also secrete anti-inflammatory cytokines and neurotrophic factors and facilitate recovery following stroke. In this review, we focus on the role and mechanisms of microglia-mediated neuroinflammation and neuroplasticity after ischemia and relevant potential microglia-based interventions for stroke therapy.
Article
Full-text available
Stroke is a leading cause of death and long-term disability. T cells have been extensively studied for their dual role in regulating immunity and inflammation following stroke. In this issue of the JCI, Cai, Shi, et al. demonstrated that CD8+ regulatory-like T cells (CD8+ TRLs) are one of the earliest lymphocyte subtypes to enter the brain after experimental ischemic stroke. Using a mouse model of stroke and comprehensive experimental approaches, the authors found that CD8+ TRLs reduced both brain damage and functional deficits in both young and aged mice. These unique early responding regulatory T cells may also play a role in a wide array of other T cell-mediated neurological disorders.
Article
Full-text available
Cell-to-cell propagation of α-synuclein is thought to be the underlying mechanism of Parkinson’s disease progression. Recent evidence suggests that inflammation plays an important role in the propagation of protein aggregates. However, the mechanism by which inflammation regulates the propagation of aggregates remains unknown. Here, using in vitro cultures, we found that soluble factors secreted from activated microglia promote cell-to-cell propagation of α-synuclein and further showed that among these soluble factors, TNF-α had the most robust stimulatory activity. Treatment of neurons with TNF-α triggered cellular senescence, as shown by transcriptomic analyses demonstrating induction of senescence-associated genes and immunoanalysis of senescence phenotype marker proteins. Interestingly, secretion of α-synuclein was increased in senescent neurons, reflecting acquisition of a senescence-associated secretory phenotype (SASP). Using vacuolin-1, an inhibitor of lysosomal exocytosis, and RNAi against rab27a , we demonstrated that the SASP was mediated by lysosomal exocytosis. Correlative light and electron microscopy and immunoelectron microscopy confirmed that propagating α-synuclein aggregates were present in electron-dense lysosome-like compartments. TNF-α promoted the SASP through stimulation of lysosomal exocytosis, thereby increasing the secretion of α-synuclein. Collectively, these results suggest that TNF-α is the major inflammatory factor that drives cell-to-cell propagation of α-synuclein by promoting the SASP and subsequent secretion of α-synuclein.
Article
Full-text available
Microglia are the resident immune cells of the central nervous system that exert diverse roles in the pathogenesis of ischemic stroke. During the past decades, microglial polarization and chemotactic properties have been well-studied, whereas less attention has been paid to phagocytic phenotypes of microglia in stroke. Generally, whether phagocytosis mediated by microglia plays a beneficial or detrimental role in stroke remains controversial, which calls for further investigations. Most researchers are in favor of the former proposal currently since efficient clearance of tissue debris promotes tissue reconstruction and neuronal network reorganization in part. Other scholars propose that excessively activated microglia engulf live or stressed neuronal cells, which results in neurological deficits and brain atrophy. Upon ischemia challenge, the microglia infiltrate injured brain tissue and engulf live/dead neurons, myelin debris, apoptotic cell debris, endothelial cells, and leukocytes. Cell phagocytosis is provoked by the exposure of “eat-me” signals or the loss of “don’t eat-me” signals. We supposed that microglial phagocytosis could be initiated by the specific “eat-me” signal and its corresponding receptor on the specific cell type under pathological circumstances. In this review, we will summarize phagocytic characterizations of microglia after stroke and the potential receptors responsible for this programmed biological progress. Understanding these questions precisely may help to develop appropriate phagocytic regulatory molecules, which are promoting self-limiting inflammation without damaging functional cells.
Article
Aging is driven by hallmarks fulfilling the following three premises: (1) their age-associated manifestation, (2) the acceleration of aging by experimentally accentuating them, and (3) the opportunity to decelerate, stop, or reverse aging by therapeutic interventions on them. We propose the following twelve hallmarks of aging: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. These hallmarks are interconnected among each other, as well as to the recently proposed hallmarks of health, which include organizational features of spatial compartmentalization, maintenance of homeostasis, and adequate responses to stress.
Article
Aging is a major risk factor for cerebral infarction. Since cellular senescence is intrinsic to aging, we postulated that stroke-induced cellular senescence might contribute to neural dysfunction. Adult male Wistar rats underwent 60-minute middle cerebral artery occlusion and were grouped according to 3 reperfusion times: 24 hours, 3, and 7 days. The major biomarkers of senescence: 1) accumulation of the lysosomal pigment, lipofuscin; 2) expression of the cell cycle arrest markers p21, p53, and p16INK4a; and 3) expression of the senescence-associated secretory phenotype cytokines interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), and interleukin-1β (IL-1β) were investigated in brain samples. Lipofuscin accumulation was scarce at the initial stage of brain damage (24 hours), but progressively increased until it reached massive distribution at 7 days post-ischemia. Lipofuscin granules (aggresomes) were mainly confined to the infarcted areas, that is parietal cortex and adjacent caudate-putamen, which were equally affected. The expression of p21, p53, and p16INK4a, and that of IL-6, TNF-α, and IL-1β, was significantly higher in the ischemic hemisphere than in the non-ischemic hemisphere. These data indicate that brain cell senescence develops during acute ischemic infarction and suggest that the acute treatment of ischemic stroke might be enhanced using senolytic drugs.