Abstract
Glioblastoma (GBM) is the most malignant tumor in center nervous
system. Clinical statistics revealed that senior GBM patients had a
worse overall survival (OS) comparing with that of patients in other
ages, which is mainly related with tumor microenvironment including
tumor-associated immune cells in particular. However, the immune
heterogeneity and age-related prognosis in GBM are under studied. Here
we developed a machine learning-based method to integrate public
large-scale single-cell RNA sequencing (scRNA-seq) datasets to
establish a comprehensive atlas of immune cells infiltrating in
cross-age GBM. We found that the compositions of the immune cells are
remarkably different across ages. Brain-resident microglia constitute
the majority of glioblastoma-associated macrophages (GAMs) in patients,
whereas dramatic elevation of extracranial monocyte-derived macrophages
(MDMs) is observed in GAMs of senior patients, which contributes to the
worse prognosis of aged patients. Further analysis suggests that the
increased MDMs arisen from excessive recruitment and proliferation of
peripheral monocytes not only lead to the T cell function inhibition in
GBM, but also stimulate tumor cells proliferation via VEGFA secretion.
In summary, our work provides new cues for the correlational
relationship between the immune microenvironment of GBM and aging,
which might be insightful for precise and effective therapeutic
interventions for senior GBM patients.
Keywords: age, tumor heterogeneity, single-cell RNA sequencing
(scRNA-seq), immune microenvironment, GBM, monocyte-derived macrophage
(MDM)
1. Introduction
GBM is an aggressive brain cancer with a high incidence rate of 32 per
1,000,000 per year ([47]1). GBM is hard for radical cures surgically
and is invalid to radiotherapy and chemotherapy in clinic ([48]2,
[49]3). Patients died by rapid deterioration and shortage of effective
medicines. The median survival time of GBM patients is about 15 months
after diagnosis ([50]4–[51]6). GBM mainly occurs in the elderly and the
median age of first onset is 64 ([52]7). In general, tumors (i.e.,
leukemia or lung cancer) within younger patients tend to be more
malignant ([53]8), whereas GBM patients have worse prognosis with age.
However, how and in what extent age influences on the prognosis of GBM
are unclear.
Tumor heterogeneity exercises major influence on the prognosis of tumor
patients. The heterogeneity in GBM cancer cells has been investigated
intensively, which includes but is not limited to the divergence in
gene mutations, epigenetic modifications and cell of origins and so on
([54]9–[55]12). The great impact of heterogeneity of tumor
microenvironment and immune microenvironment in particular on the tumor
progression has been recognized in recent years, which constitutes the
theoretical basis of tumor immunotherapy ([56]13–[57]16). The studies
on tumor immunology in GBM are limited because of the existence of
brain blood barrier (BBB) that blocks the entrance of most of the
peripheral immune cells. Bulk omics whose signals summarized over
millions of cells are limited to characterize each kind of immune cell
populations in GBM tumor environment. Various GBM scRNA-seq data reveal
that resident microglia as well as infiltrated peripheral immune cells
(including MDMs) contribute to the heterogeneity of GBM immune
microenvironment ([58]17–[59]22).
To overcome the batch effects among various scRNA-seq and establish the
landscape of immune microenvironment across age, we developed a machine
learning method to integrate a variety of scRNA-seq data in public and
analyzed the immune cells infiltrating in GBM collected from
individuals of various ages. Our data suggested that the heterogeneity
of immune cells across age led to the worse OS of aged GBM patients.
MDM subpopulation in elderly suppressed the immunologic function and
promoted tumor cell growth in a paracrine manner.
2. Materials and methods
2.1. Integration of single-cell RNA sequencing datasets
We constructed an atlas of cells in GBM by integrating six public
scRNA-seq datasets generated by various studies. Because sequencing
protocols and experiment conditions are different across these studies,
batch effects shall induce an overwhelming number of technical
variations in gene expression counts, which impede the integration of
different scRNA-seq datasets and the discovery of true biological
signals. It is noticed that two kinds of information are immune to
technical variations in sequencing and experiment. First, it is the
cell-type label that indicates the cell state. Among various GBM
datasets, most cell states in tumor environments are similar. Second,
the correlation between genes can combat technical variations. Let’s
consider the example of face recognition. Even if someone wears a face
mask, human visual neurons can precisely recognize the identity of the
person. This is because the human brain neural network can utilize the
correlation structure among facial features. Similarly, the gene-gene
correlation can benefit the detection of true biological signals. The
correlation structure among genes can be characterized by relative gene
abundance, which is known as gene usage. Based on the above analyses,
we designed a factor model called scClassifier to extract gene usages
relating to cell states. scClassifier is implemented by using deep
neural networks. After training with the cell-type annotated GBM
datasets, scClassifier can automatically disentangle the factors
regarding cell states and the factors regarding batch effects. The cell
state-related factors encode true biological signals, and can be used
to predict cell type as well as integrate multiple datasets. Details
about scClassifier are given in the following.
Let’s introduce the mathematical notations used throughout the paper
first. Let y and z [1] denote a factor variable corresponding to cell
type and a factor variable accounting for technical variations, i.e.,
batch effect, respectively. The cell-type annotations y [1],···,y[N] of
N cells are given by users and are discrete variables. Thus, y is
modeled by the Categorical distribution. Besides batch effect,
technical variations of single-cell transcriptomics have multiple
resources that have been not well charted. Therefore, z [1] is modeled
with the normal distribution. For every cell, the complete hidden cell
state z[y] is determined by the factors y and z [1] with the procedure
described as follows,
[MATH:
y∼Categ
orical(α0) :MATH]
[MATH:
z1∼Normal(μ0,Σ0) :MATH]
[MATH:
zy∼Normal(μzy
,∑zy) :MATH]
where a [0], μ [0], and ∑[0] are the parameters of prior distributions.
In practice, without loss of generality, we choose
[MATH:
a0=
(1,⋯,1)︸L :MATH]
, L is the number of cell types. And we choose the mean vector
[MATH:
μ0=
(0,⋯,0)︸d :MATH]
and the diagonal covariance matrix
[MATH:
∑0=diag(1,⋯,1)︸d :MATH]
, d is the dimension of the hidden state. In the analyses of the paper,
d is set as 50. The parameters μ [z [y ]]and ∑[ z [y ]]are inferred
from y and z [1] by using a deep neural network.
Once the complete hidden cell state z[y] is determined, the generation
of single-cell gene expression follows the procedure: first, gene usage
η is sampled from a Dirichlet distribution, where the parameter is
inferred from z[y] by using a decoder neural network; second, gene
expression counts are generated from η by using the multinomial
distribution. Specifically,
[MATH:
α=Decod
er(zy) :MATH]
[MATH:
η∼Diric
hlet(α)
:MATH]
[MATH:
X∼Multi
nomial(η)
:MATH]
The parameters of the proposed model are learned by using the
stochastic variational inference introduced by the semi-supervised
variational autoencoder ([60]23), which can be scalable to large
scRNA-seq datasets. To utilize the stochastic variational inference, we
introduce the variational distribution,
[MATH: q(X,zy
msub>,y,z1)=q(X)q(zy|X
)q(y|zy
msub>)q(z1|y
,zy) :MATH]
We use the evidence lower bound (ELBO) as the loss for model training.
The deep neural network-based factor model is recently introduced in
computer vision. It has been successfully used to factorize various
kinds of factors in handwritten digit images ([61]24). In the paper, we
extend the deep neural network-based factor model to process
single-cell transcriptomics and to extract the variations of gene
expression regarding cell states.
2.2. Cell type annotation, clustering, and visualization
Besides single-cell integration, scClassifier can be used in a variety
of single-cell computational tasks, including the visualization of
cell-to-cell variability and cell type annotation. First, the variance
distribution model q(z[y] |X) yields 50-dimensional embeddings if gene
expression matrix X is given. The 50-dimensional embeddings can produce
the two-dimensional coordinates of cells through uniform manifold
approximation and projection (UMAP). Besides, given gene expression
matrix X, the model q(z[y] |X)q(y|z[y] ) can predict cell type of each
cell (cell type annotation). The two-dimensional coordinates and cell
type were used to draw the UMAP plots in the paper ([62]25).
To find the subpopulation of every cell type, we needed to perform
clustering on the two-dimensional coordinates of cells. Therefore, we
built a model that can predict the sub-clusters using a python package
Scikit-learn. This model allows for automatic estimation of the number
of subpopulations, since the main components of the model are the
Dirichlet process.
2.3. Cell communication analysis of GBM by iTALK
To explore the growth factor interactions associated with monocytes in
GBM, we performed cell communication analysis with an R package iTALK
published in 2019 ([63]26). First, we calculated differentially
expressed genes (DEGs) between adult and aged patients by using the
Wilcoxon signed-rank test. Next, we selected growth factor pairs for
our research from the L-R database. Meanwhile, a list of growth factor
pairs matching the DEGs was generated by the “FindLR” function in iTALK
package. Finally, we sorted the list using the logFC between the adults
and aged, and selected monocytes-related growth factor pairs with top
absolute value. The growth factor pairs were shown in the Circos plots
with the “LRPlot” function. The T cell-associated immune checkpoint
interactions within GBM were analyzed with the same procedure.
2.4. Differential gene analysis and violin plots
After obtaining the expression matrix corrected for batch effects, we
constructed a new Seurat object, and then calculated its differential
genes according to the FindMarkers function of the Seurat package.
Violin plots are visualized by the VlnPlot function of the Seurat
package. Besides, in order to merge different violin plots, we drew the
stacked violin plots based on our code.
2.5. Data access and survival analysis
For OS data, we used The Cancer Genome Atlas (TCGA) from 606 (Firehose
Legacy)) glioblastoma mutiforme samples (GBM_TCGA), and The
Surveillance, Epidemiology, and End Results (SEER) Program of the
National Cancer Institute (NCI) from 20878 glioblastoma multiforme.
TCGA data are accessed by the R package cgdsr (ver 1.3.0). SEER data
are downloaded from the SEERStat software (ver 8.4.0.1). To perform
survival analysis, the R packages survival (ver 3.3.1) and survminer
(ver 0.4.9) are used to calculate and plot the Kaplan-Meier curve. For
legacy data, median levels were used to segregate cancer patients
according to OS outcome. P value is adjusted with the
Benjamini-Hochberg correction method.
2.6. Gene ontology analysis
We used the R package clusterProfiler (ver 4.0.5) to perform GO pathway
enrichment analysis, where the GO annotation was given in the R package
org.Hs.eg.db (ver 3.13.0). The GO enrichment results are shown as the
bubble plots with the R package ggplot2 (ver 3.3.6). R version is
4.1.2.
2.7. Human tissue specimens and immunohistochemistry
Paraffin-embedded specimens from patients with IV GBM were obtained
surgically, from the First Affiliated Hospital of Xiamen University.
Collection of all samples was approved by the local ethical committee
and the institutional review board of the hospital. Each patient gave
written informed consent and patient data have been made anonymous.
Detailed information of patients is included in [64]Supplementary
Table 1 . For immunofluorescent staining, blocks were processed at 5
μm. After dewaxing and rehydration, heat mediated antigen retrieval was
performed using citrate buffer. And then samples were incubated with
lysozyme primary antibody (Abcam, Cat#ab108508) at 1/200 dilution
overnight at 4°C. After washing with PBS, the sections were incubated
with second antibody conjugated to Cy5 (Jackson ImmunoResearch
Laboratories, 1:200) and DAPI (Thermo Fisher Scientific, Cat#D1306) for
1 h at RT. Immunofluorescence images were taken by Leica SP8.
3. Result
3.1. Senior GBM patients have poor OS
Analyzing TCGA and SEER data of GBM patients, we discovered a negative
correlation between OS and age ([65] Figures 1A , [66]S1A ) and showed
a worse OS in senior GBM patients. Adding with the similar observations
from other studies ([67]27), it suggests that age is an independent
risk factor of poor prognosis in GBM. Therefore, patients ([68]
Figures 1A , [69]S1A ) were grouped by ages and the OS of elder group
is significantly poorer than the young group ([70] Figures 1B , [71]S1B
). To clarify that OS was not affected by other factors, we did a
survival analysis related to gender and mutation subtype in adults and
aged. No significant sexual difference was found in the aged stage
though women had a slightly better OS in the adult stage ([72]
Figure 1C ). Neither TP53 nor PTEN mutations alters OS across age ([73]
Figures 1D, E ). Similar results showed that patients with EGFR
mutation in the aged group still had a poorer OS than those in the
adult group, but this mutation could make the adult OS as poor as that
in aged group, suggesting that EGFR mutations in adults could simulate
the changes in old age, and EGFR might be involved in the regulation of
age-related OS changes ([74] Figure 1F ). Taken together, age emerges
as an independent risk factor of worse OS in GBM.
Figure 1.
[75]Figure 1
[76]Open in a new tab
Age is an independent risk factor of prognosis and overall survival.
(A) Scatter plot of OS (logarithmically transformed) correlated with
age from 606 samples in the TCGA database. (B) Kaplan-Meier curves of
survival analysis used to compare the OS of GBM patients in the SEER
database, divided into four groups according to age. (C–F) Kaplan-Meier
curves of survival analysis used to compare the differences in OS
between the two age groups [Adult (30≤age<50) and Aged (age≥64)] with
different genders (C), TP53 mutation (D), PTEN mutation (E), or EGFR
mutation (F). *(P< 0.05), **(P< 0.01), ***(P< 0.001), ****(P< 0.0001),
ns (no significance).
3.2. Integration analysis of single-cell RNA-seq data from adult and aged
patients
The heterogeneity of tumor environment and the heterogeneity of
tumor-associated immune cells in particular are unclear in GBM across
ages. Herein we focused on immune microenvironment of GBM with
scRNA-seq. We analyzed the transcriptome of 23 GBM patients across ages
from 6 public scRNA-seq datasets. GBM patients were divided into two
groups, adult (2064, n=13) ([77] Table 1
). To build a comprehensive immune landscape of primary GBM, 6 datasets
were integrated into a huge dataset for analysis ([78] Figure 2A ). The
clustering and UMAP analysis ([79] Figure S2A, C ) on the huge dataset
by traditional Seurat tools were disturbed by ineluctable batch effects
that yielded from technical variations in sequencing protocols.
Unexpectedly, the biological variations ([80] Figure S2B, D ) for
clustering were removed when batch effects were corrected by classical
harmony algorithm. Therefore, we developed a machine learning method
named scClassifier to integrate various scRNA-seq datasets to capture
the biological variations (Method1). Ultimately, 85,372 cells from the
adult tumor samples and 29,634 cells from the aged tumor samples were
integrated ([81] Table 1 , [82]Figure 2B ). Then tumor cells, normal
brain cells, and different kinds of immune cells ([83] Figure 2C ) were
clustered by their universal marker genes ([84] Figure 2D ).
Table 1.
Statistical chart shows details of all samples that we collected, such
as cell counts and number of patients.
Group Total Cell Counts Cell Counts Number of Patients Cell Type
Adult Aged Adults Aged
1 1901 1645 256 3 1 tumor,immune
2 11786 0 11786 0 6 tumor,immune
3 84969 71053 13916 5 2 tumor,immune
4 15935 12259 3676 1 4 immune
5 415 415 0 1 0 tumor,immune
6 17982 – – – – tumor
Total 132988 85372 29634 10 13
[85]Open in a new tab
Figure 2.
Figure 2
[86]Open in a new tab
Integration of GBM scRNA-seq containing immune cells and tumor cells.
(A) Workflow of scRNA-seq integration, data analysis, and validation
based on the primary tumors, which are collected from nine adult and
thirteen aged patients. (B) Corrected UMAP plots colored by the cell
types which are identified by marker genes in GBM. (C) UMAP plots
colored by the scRNA-seq source using our method in each GBM group. (D)
Heatmap showing relevant expression across different cell subsets. The
color is the same as the cell subsets in [87]Figure 2B .
3.3. More MDMs infiltrate in GBM of aged patients
Immune infiltration is essential for tumorigenesis ([88]28, [89]29).
Hereby we surveyed the entire infiltrated immune cells in both GBM and
normal brain tissues. As expected, the overall immune infiltration was
higher in tumors than normal brains ([90] Figures 3A , [91]S3 )
([92]30). Given the top incidence occurs over 65-years-old and 50% of
GBM patients are older than 64, the ratio of infiltrated immune cells
in aged patients was very close to that in general GBM patients.
Compared with aged patients, adult patients had less immune
infiltration ([93] Figure 3A ). 8 subpopulations of immune cells were
found in tumor microenvironment. In general, brain-resident microglia
composed the majority ([94]31) and the number of other immune cells
(e.g., B cells, T cells, DCs) were low no matter in adult or aged
patients ([95] Figures 3B–E ). Comparing each population of immune
cells in adult and aged patients, we found the ratio of GAMs was higher
in aged patients. GAMs were composed with microglia and MDMs. The MDM
rather than microglia population was significantly increased in aged
patients ([96] Figures 3B–E ).
Figure 3.
[97]Figure 3
[98]Open in a new tab
GAM infiltration, especially MDM infiltration, increases more
significantly in the aged group. (A) Histogram showing the proportion
of immune cells, brain somatic cells, or tumor cells in integrated GBM
scRNA seq data, grouped by adult and aged (each n=9). (B, C) UMAP plots
of immune cells in GBM of adult (B) and aged (C) patients. (D) Stacked
barplots showing cell types composition of all patients scRNA data from
adult group (left) and aged group (right). (E) Histogram calculating
and comparing all cell types proportion of immune cells in adult
group(n=10) and aged group(n=13). (F, G) Immunofluorescent staining for
lysozyme (MDM marker) in GBM sections from adult group and aged group
(F). Amplified areas of white rectangles are shown below and the ratio
of LYZ ^+ cells is quantified (G), n=24 fields from 5 GBM samples for
each group. All the quantification data are presented as mean ± SEM,
two-tailed unpaired Student’s t-test. Scale bars, 100μm. *(P< 0.05),
**(P< 0.01), ns (no significance).
To verify the conclusion above, we stained human GBM tissue samples
with LYZ, one of the MDM-specific markers ([99] Figures S4D, S4E )
yielded by feature plot and violin plot. As expected, LYZ ^+ cells were
increased in aged GBM patients. ([100] Figures 3F, G ).
We found that monocyte-associated growth factors were enriched in aged
patients which might promote their proliferation ([101] Figures 4A ,
[102]S5A ). The chemokines for monocytes were evidently up-regulated in
tumor cells and other immune cells. For example, CCL2 was highly
expressed in aged tumor cells ([103] Figures 4B , [104]S5B ) to recruit
monocyte from peripheral blood ([105]32). Highly expression of CCL2 is
regarded as an unfavorable factor for survival ([106]33). Besides, CCL2
is also considered to induce M2 polarization of MDMs ([107]34). In all,
it may result in enhanced monocytes recruitment from peripheral tissue
to GBM and promote them to differentiate into M2 MDMs.
Figure 4.
[108]Figure 4
[109]Open in a new tab
Increased proliferation and recruitment of monocytes. (A) Circos plot
showing growth factor interactions between monocytes and other cells.
The outer ring represents the cell type, and the inner ring represents
the gene. The color of the line indicates the direction of upregulation
(adult vs aged) and the width of the line indicates the degree of
upregulation, as shown in the legend. (B) Violin plot showing the
expression of CCL2 in each immune cell type, divided by adult and aged
cells.
3.4. Aged MDMs present features of M2 macrophage
To investigate how the heterogeneity of immune cells leads to the worse
OS in aged patients, we performed DEG analysis on MDMs and followed by
GO analysis for the top 100 genes. Although aged-MDMs rather than
adult-MDMs presented typical macrophage signature ([110] Figures 5A, B
, [111]S6A ), further analysis using markers of pro-inflammatory
features (M1) and anti-inflammatory features (M2) showed that the M2
type was dominant in aged MDMs ([112] Figure 5C ), which is suppressive
to tumor immune activity. In consistent, TOP 5 highly expressed genes
in aged-MDMs, i.e., FCGR2B, TGFBI, CD163 and GPNMB showed negative
correlation with the patients’ OS by association analysis of gene
expression and patient survival in TCGA ([113] Figure 5D ). Such
correlation was not found in adult MDMs ([114] Figure 5E ). Thus, the
increased immunosuppressive MDMs lead to the poor prognosis and OS in
aged patients.
Figure 5.
[115]Figure 5
[116]Open in a new tab
MDMs in the aged group are much anti-inflammatory and unfavorable to
OS. (A, B) GO (Gene ontology) analysis of top 100 specific genes of
MDMs (A) or relatively highly expressed genes in aged MDMs (B). Data
displaying the top 10 enriched GO terms ranked by p values. The color
indicates p values for GO term enrichment and the circle size indicates
the number of enriched genes for each GO term. (C) Heatmap showing the
marker gene signatures of the identified M1 and M2 subtype cells in the
adult group(left) and the aged group(right), and the ratio of each
subtype in the adult or aged were counted on the top row of columns.
(D, E) Boxplots showing the differences of OS between high or low
expressions of the top 5 genes highly expressed in MDM aged group (D)
or MDM adult group (E), respectively. Data are obtained from TCGA-GBM
database. *P<0.05, **P< 0.01. All the quantification data are presented
as mean ± SEM, two-tailed unpaired Student’s t-test. ns (no
significance).
3.5. The MDM-B subgroup is significantly increased in older age and is
critical for the regulation of age-related OS
We further characterized the MDM sub-population that responded to the
change of MDMs in both quantity and quality with age. MDMs were divided
into four sub-clusters as MDM A\B\C\D ([117] Figure 6A ). MDM-A was
dominant in adult MDMs and decreased with age; whereas the MDM-B
increased significantly with age and prevailed in most of aged MDMs
([118] Figure 6B ). In addition, MDM-B presents typical M2 features
([119] Figures 6C , [120]S7A ). Genes highly enriched in MDM-B ([121]
Figures S7B–D ) and unfavorable to OS were the key factor for poor OS
of aged patients. Therefore, a small portion of aged patients with low
expression of those genes, such as C5AR1, CD14 and SLC11A1 had very
close OS to that of adult patients ([122] Figures 6D–F ).
Figure 6.
[123]Figure 6
[124]Open in a new tab
The subgroup of MDM-B but not microglia was the key to age-related OS
reduction. (A) UMAP plot of four MDM subpopulations in primary GBM. (B)
Histogram showing the proportion of each MDM subgroup in total GBM
cells from the integrated GBM scRNA seq data, grouped by adult (n=10)
and age (n=13). ns (no significance). (C) Stacked barplot showing
macrophage subtype proportion of four MDM subpopulations. M1 and M2
means classical macrophage classification by polarization. M0 means
double-negative M1 and M2 features while M3 means double-positive M1
and M2 features. (D–G) Kaplan–Meier curves demonstrating the difference
of OS in TCGA GBM patients in adult and aged groups with high or low
expression of C5AR1 (D), CD14 (E), SLC11A1 (F), or CX3CR1 (G). All the
quantification data are presented as mean ± SEM, two-tailed unpaired
Student’s t-test. *(P< 0.05), **(P< 0.01), ***(P< 0.001), ns (no
significance).
Considering microglia are the most abundant population in all
GBM-associated immune cells, microglia from patients across age were
further clustered and showed a close proportion of M1/M2 subtypes,
excluding the major role of microglia in worse prognosis with age
([125] Figures 6G , [126]S7E, F ). These data suggest the
anti-inflammatory MDMs in aged GBM caused the poor OS with age.
3.6. Aged MDMs compromise tumor immune response and promote tumor growth in
paracrine manner
M2 macrophage inhibits T cell function ([127]35) and M2
tumor-associated macrophage restricts T cells from killing tumor cells
([128]36). To examine the immune activity of T cell in GBM, we
performed checkpoint communication analysis in both adult and aged
patients. We found the immune checkpoint genes were upregulated in T
cells from aged patients ([129] Figures 7A , [130]S8 ), which suggested
an immunosuppressive state in aged GBM. In consistent, cytotoxic CD8 ^+
T cells decreased and exhausted CD4 ^+ T cells (HAVCR2 ^+) increased in
aged GBM ([131] Figures 7B–D ).
Figure 7.
[132]Figure 7
[133]Open in a new tab
MDMs regulate age-related OS by affecting T cells and secreting growth
factors. (A) Circos plot showing immune checkpoint interactions between
T cells and other cells. The outer ring represents the cell type and
the inner ring represents the gene. The color of the line indicates the
direction of upregulation (adult vs aged) and the width of the line
indicates the degree of upregulation, as shown in the legend. (B)
Violin plot showing the expression of each T cell marker in adult or
aged T cells, which divided them into two groups by CD4 and CD8
expression. (C, D) Pie plots exhibiting the proportion of CD4 ^+ and
CD8 ^+ T cells in adult or aged T cells. (E) Kaplan–Meier curves
demonstrating the difference of OS in TCGA GBM patients in Age<64 and
Age≥64 two groups with high or low expression of TNFSF4 and VEGFA. All
the quantification data are presented as mean ± SEM, two-tailed
unpaired Student’s t-test. *(P< 0.05), ****(P< 0.0001).
MDMs promote tumor growth by secreting growth factors ([134]37). Among
all the growth factors detected in GBM, VEGFA was highly expressed in
MDMs and tumor cells ([135] Figure S5 ), suggesting that increased MDMs
could lead to upregulation of VEGFA and promote tumor growth.
TCGA data showed that aged patients with reduced VEGFA and TNFSF4 (an
immune checkpoint gene) expression had a better OS, which suggests two
oncogenic functions by MDMs were synergistic ([136] Figure 7E ).
4. Discussion
In this article, we created an atlas of immune cells infiltrating in
GBM by holistically analyzing six public scRNA-seq datasets from
patients of variant ages. To integrate the six datasets, we developed a
machine learning method named scClassifier. Besides, scClassifier shows
better performance on cell-type annotation and integration than the
standard methods ([137] Figure S9 ). Our data suggested that immune
heterogeneity by ages contributed to the worse OS in aged GBM patients
([138] Figure 8 ). The number of infiltrated immune cells was increased
with ages and GAMs had the most dramatic change. GAM infiltration has
been associated with poor prognosis and OS ([139]38, [140]39), but the
effect of MDMs and microglia may be different by age. Previous studies
have not found that MDMs is the immune cell population that changes the
most significantly with age. According to TCGA data analysis, our
research discovered targeting MDMs is probably more beneficial to
improve OS of aged patients but not adult patients.
Figure 8.
[141]Figure 8
[142]Open in a new tab
GBM immune heterogeneity across ages. (A) A model figure of this
article showing how aging influences immune microenvironment of GBM
patients.
This difference in the infiltration of MDMs with age may be useful for
classification as subtype of GBM. Our study has successfully repeated
the used MDM specific markers. Furthermore, we find FCGR2B, GPNMB etc
are probable new MDM specific markers in GBM. These genes can be used
to detect the level of MDMs, and to evaluate and predict prognosis and
OS of non IDH-mut or EGFR mutation GBM.
MDMs are derived from monocytes. Monocytes are myeloid derived
non-resident cells, which need recruitment from extracerebral
circulatory vessels. Normal brain tissue is lowly infiltrated by
monocytes/MDMs, while in GBM they infiltrate highly. This phenomenon is
observed in this article, too. But the difference is that we find a
significant increase of MDMs in aged patients. It is previously
reported that CCL2 is secreted abundantly by GBM cells ([143]35,
[144]40). Zhu et al. has found that gliomas cell lines [145]GPL261 and
U87 can secret abundant CCL2 in vitro ([146]41). Our research shows a
high expression of CCL2 in tumor cells and MDMs, and the expression is
much higher in aged group. This may induce a total up-regulation of
CCL2, and finally induce a hyper accumulation of MDMs, as well as M2
differentiation. Current therapy targeting CCL2-CCR2 axis proves it is
a promising target of GBM ([147]42). Whether targeting CCL2-CCR2 axis
is only favorable of age-related OS or highly MDM-infiltrated patients
needs further exploration.
As for downstream of MDMs, current reports show that MDMs can exert
immunosuppressive function through the regulation of T cells. Li et al.
has discovered co-culture of naive T cell with glioma-associated MDMs
can induce a differentiation into TGFBI and IL-10 secreted CD4 ^+ Treg
cell ([148]43). Meanwhile, through highly expressing PD-L1 and
PD-L2,(PD-1 ligands) CD80 and CD86(CTLA-4 ligands) and other immune
checkpoint genes, MDMs interact with CD8 ^+ T cells and then suppress
their cytotoxic function ([149]44). Our research reveals CTLA-4 and its
ligand CD86 are much more highly expressed in aged T cells and MDMs.
This verifies aged MDMs are probably more suppressive for T cells. In
addition, many other checkpoint genes are found up-regulated in aged T
cells. Among them TNFSF4 is likely to be a new therapy target to
de-repress the negative regulation of T cells from MDMs. After that,
TGFBI secreted by MDMs can also target CD8 ^+ T cells directly,
inhibiting its killing effect on tumor cells ([150]45). This is
corresponded with the largely upregulated immune checkpoint genes and
high expression of TGFBI in aged MDMs in our study. Therefore,
detecting the expression of these genes for targeted therapy may be
able to better treat of GBM.
Data availability statement
Publicly available datasets were analyzed in this study. This data can
be found here: [151]https://www.ncbi.nlm.nih.gov/geo/ Gene Expression
Omnibus via GSE numbers: [152]GSE117891 ([153]46), [154]GSE131928
([155]47), [156]GSE163120 ([157]21, [158]48), [159]GSE89567 ([160]49).
[161]GSE163120 can also be acquired by GBmap
([162]https://doi.org/10.5281/zenodo.6962901). Group3 is available at
DATE from [163]https://registry.opendata.aws/cptac-3. Group6 is
available at the Broad Institute Single-Cell Portal
([164]https://singlecell.broadinstitute.org/single_cell/study/SCP503)
([165]50). Two scRNA-seq datasets for normal brain tissue are available
at Gene Expression Omnibus via GSE numbers: [166]GSE126836 ([167]51)
and [168]GSE140231 ([169]52).
Ethics statement
The studies involving human participants were reviewed and approved by
the Scientific Ethics Committee at the First Affiliated Hospital of
Xiamen University. The ethics committee waived the requirement of
written informed consent for participation.
Author contributions
SC, FZ, WM, ZW, and SW designed experiments, performed data analyses,
and wrote the manuscript. SW, FH, XL, QC, SG, YY, YX, NX, and SC
performed experiments and data visualization. SW and XK performed
method comparison and assessment. FZ supervised the project. All
authors contributed to the article and approved the submitted version.
Acknowledgments