Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) exhibit
multi-lineage differentiation potential and robust proliferative
capacity. The late stage of differentiation signifies the functional
maturation and characterization of specific cell lineages, which is
crucial for studying lineage-specific differentiation mechanisms.
However, the molecular processes governing late-stage BMSC
differentiation remain poorly understood. This study aimed to elucidate
the key biological processes involved in late-stage BMSC
differentiation. Publicly available transcriptomic data from human
BMSCs were analyzed after approximately 14 days of osteogenic,
adipogenic, and chondrogenic differentiation. Thirty-one differentially
expressed genes (DEGs) associated with differentiation were identified.
Pathway enrichment analysis indicated that the DEGs were involved in
extracellular matrix (ECM)-receptor interactions, focal adhesion, and
glycolipid biosynthesis, a ganglion series process. Subsequently, the
target genes were validated using publicly available single-cell
RNA-seq data from mouse BMSCs. Lamc1 exhibited predominant distribution
in adipocytes and osteoblasts, primarily during the G2/M phase. Tln2
and Hexb were expressed in chondroblasts, osteoblasts, and adipocytes,
while St3gal5 was abundantly distributed in stem cells. Cell
communication analysis identified two receptors that interact with
LAMCI. q-PCR results confirmed the upregulation of Lamc1, Tln2, Hexb,
and St3gal5 during osteogenic differentiation and their downregulation
during adipogenic differentiation. Knockdown of Lamc1 inhibited
adipogenic and osteogenic differentiation. In conclusion, this study
identified four genes, Lamc1, Tln2, Hexb, and St3gal5, that may play
important roles in the late-stage differentiation of BMSCs. It
elucidated their interactions and the pathways they influence,
providing a foundation for further research on BMSC differentiation.
Keywords: Mesenchymal stem cell, Osteogenesis, Adipogenesis, Laminin,
Single-cell RNA-seq
Subject terms: Cell biology, Computational biology and bioinformatics,
Developmental biology, Molecular biology, Stem cells
Introduction
Mesenchymal stem cells (MSCs) are pluripotent that can be obtained from
various tissues, umbilical cord, amniotic fluid, and
placenta^[30]1–[31]4. It is capable of expanding and differentiating in
vitro into a variety of mesodermal-type lineages, including bone,
adipose, cartilage, muscle, tendon, and stroma, supporting
hematopoiesis and the vascular system, and undergoes migration and
paracrine signaling, which is not only crucial in embryonic development
but also plays an important role in tissue homeostasis and repair
throughout the life of an organism^[32]5,[33]6. MSCs from different
sources exhibit different differentiation tendencies, and studies have
shown that BMSCs exhibit greater APL activity, calcium deposition, and
transformation capacity at an earlier age,compared with adipose-derived
MSCs^[34]7. Therefore, BMSCs were chosen for studying the molecular
mechanisms of osteogenic differentiation.
Key transcription factors for osteoblast differentiation are required
for the expression of osteoblast-specific genes, including
Runt-associated transcription factor 2 (Runx2) and osteocalcin (Ocn),
alkaline phosphatase (ALP), type I collagen, bone bridging proteins,
and bone sialic acid protein^[35]8–[36]13. Bone morphogenetic proteins
(BMPs), especially BMP-2, BMP-4, and BMP-7, activate the Smad signaling
pathway, which in turn leads to the expression of Runx2 and Ocn and
inhibits adipogenic differentiation^[37]14–[38]16. In addition to BMP,
the Wnt/
[MATH: β :MATH]
-catenin signaling pathway plays a key role in osteoblast
differentiation^[39]17. Activation of this pathway leads to the
accumulation of
[MATH: β :MATH]
-catenin in the cytoplasm, which then translocates to the nucleus,
activates the transcription of osteoblast-specific genes, and inhibits
adipogenesis. On the other hand, adipocyte differentiation is regulated
by peroxisome proliferator-activated receptor
[MATH: γ :MATH]
(PPAR
[MATH: γ :MATH]
)^[40]18. PPAR
[MATH: γ :MATH]
is considered a major regulator of adipogenesis because it controls the
expression of many adipocyte-specific genes. PPAR
[MATH: γ :MATH]
activation not only promotes adipogenesis but also inhibits
osteogenesis^[41]19. The balance between osteoblast and adipocyte
differentiation is a tightly regulated process^[42]20. Therefore,
understanding the mechanisms that regulate this balance is essential
for developing therapeutic approaches for diseases such as osteoporosis
and obesity, which are characterized by an imbalance between bone and
fat formation.
In mesenchymal stem cells, cellular matrix components play a key
regulatory role in the directed differentiation of mesoderm into
osteoblasts or adipogenic cells^[43]21. The extracellular matrix is a
complex network of many molecules, including collagens, glycoproteins
(GPs), and ECM-associated proteins^[44]22. Specific components of the
ECM can influence cell fate decisions; for instance, collagen promotes
osteoblast differentiation, whereas fibronectin may promote adipogenic
cell differentiation^[45]23,[46]24. Cell adhesion molecules and cell
signaling molecules in the cell matrix are important regulators of the
differentiation process, transmitting signals and modulating cell
behavior by interacting with receptors on the cell
surface^[47]25,[48]26. For example, integrins increase the ability of
cells to differentiate osteoblastically, maintain bone homeostasis, and
regulate bone mass while inhibiting the adipogenic differentiation of
BMSCs^[49]27–[50]29. Taken together, the study of cellular matrix
components contributes to our understanding of the mechanisms of germ
layer-directed differentiation.
BMSCs exhibit the most pronounced difference between days 14 and 17 of
differentiation, and the difference between days 17 and 21 of
differentiation is not significant^[51]7. Therefore, our study combined
tri-lineage differentiation with bulk RNA-seq analysis and single-cell
RNA-seq (scRNA-seq) analysis to determine which factors simultaneously
play key roles in osteogenic, adipogenic, and chondrogenic terminal
phase differentiation, and cell staining and RT-qPCR were used to
verify the changes in target genes during differentiation, aiming to
provide a new basis for MSC therapies.
Results
hBMSCs osteogenic, adipogenic, and chondrogenic differentiation 14-day
transcriptome data cross-identify 31 DEGs
A preliminary comparison of the six datasets ([52]GSE36923,
[53]GSE44303, [54]GSE109503, [55]GSE140861, [56]GSE28205, [57]GSE37558)
was performed before the differential gene analysis. Through the
box-and-line plot of the distribution of the 6 datasets, we concluded
that there was no batch effect within the datasets (Fig. [58]1a–c).
According to the PCA plot of the distribution of the 6 datasets, the
differences between different types of data were significant, and the
analysis was feasible (Fig. [59]1d–i). Differential gene analysis of
different types of data was performed separately, and DEGs coexpressed
in similar datasets were selected to obtain 347 upregulated genes and
263 downregulated genes related to osteogenic differentiation, 658
upregulated genes and 704 downregulated genes related to adipogenic
differentiation, and 106 upregulated genes and 48 downregulated genes
related to cartilaginous differentiation (Fig. [60]2a–i). There was an
antagonistic relationship between the three classifications. To
identify the genes that were coexpressed in each category based on the
Venn diagram, the genes coexpressed in osteogenic, adipogenic, and
chondrogenic differentiation were compared two by two with opposite
differentiation trends, and a total of 31 DEGs were obtained (Fig.
[61]2j–o).
Figure 1.
[62]Figure 1
[63]Open in a new tab
Quality of RNA-seq data of bone marrow-derived MSCs at the late stage
of differentiation. Box plot: (a) osteogenic differentiation dataset
([64]GSE28205, [65]GSE37558); (b) adipogenic differentiation dataset
([66]GSE36923, [67]GSE44303); (c) chondrogenic differentiation dataset
([68]GSE109503, [69]GSE140861); PCA plot; (d) [70]GSE37558; (e)
[71]GSE36923; (f) [72]GSE140861; (g) [73]GSE28205; (h) [74]GSE44303;
(i) [75]GSE109503.
Figure 2.
[76]Figure 2
[77]Open in a new tab
Target gene identification and enrichment analysis of RNA-seq data from
bone marrow-derived MSCs at the late stage of differentiation (a,b)
volcano plots of differential gene expression in osteogenic
differentiation datasets ([78]GSE28205 and [79]GSE37558); (c,d) volcano
plots of differential gene expression in adipogenic differentiation
datasets ([80]GSE36923 and [81]GSE44303); (e,f) volcano plots of
differential gene expression in cartilageogenic differentiation
datasets ([82]GSE109503 and [83]GSE140861) volcano plots of
differential gene expression; (g) heatmap of differential gene
expression of osteogenic differentiation datasets ([84]GSE28205 and
[85]GSE37558) after integration by the RRA algorithm; (h) heatmap of
differential gene expression of adipogenic differentiation datasets
([86]GSE36923 and [87]GSE44303) after integration of by the RRA
algorithm; (i) heatmap of differential gene expression of chondrogenic
differentiation dataset ([88]GSE109503 and [89]GSE140861) heatmap of
differential gene expression after integration by the RRA algorithm.
Venn diagrams of DEGs with opposite differentiation trends in the three
differentiation directions. A total of 31 DEGs were obtained. (j)
Intersection of osteogenic upregulated differential genes and
adipogenic downregulated differential genes; (k) intersection of
osteogenic upregulated differential genes and chondrogenic
downregulated differential genes; (l) intersection of chondrogenic
upregulated differential genes and adipogenic downregulated
differential genes; (m) intersection of adipogenic upregulated
differential genes and chondrogenic downregulated differential genes;
(n) intersection of adipogenic upregulated differential genes and
chondrogenic downregulated differential genes; (o) intersection of
adipogenic upregulated differential genes and osteogenic downregulated
differential genes; (p) differential gene enrichment analysis (GO) bar
graph; (q) differential gene enrichment analysis (KEGG) network graph.
AD adipogenesis, OS osteogenesis, CH chondrogenesis.
Enrichment analysis revealed that DEGs were concentrated in the extracellular
matrix and related to ECM-receptor interactions
Gene Ontology (GO) analysis revealed that the differentially expressed
genes (DEGs) associated with an adjusted p-value (adj. P) < 0.05 were
primarily enriched in biological processes and molecular functions
related to collagen-containing extracellular matrix, hexosaminidase
activity, binding to laminin, structural components of the
extracellular matrix that confer tensile strength, and integrin binding
(Fig. [90]2p). At KEGG pathway analysis revealed that DEGs with adj.P <
0.05 were associated with the ECM-receptor interaction, local adhesion,
and glycolipid biosynthesis-ganglio series pathways (Fig. [91]2q).
ceRNA network construction
Using the String website, six target genes enriched in extracellular
mesenchymal tissues were identified, namely, Hapln1, Col4a1, Lamc1,
Itga10, Col10a14, and Tln2, and two genes enriched in the target genes
involved in the biosynthesis of glycosphingolipid-ganglio series
pathways were identified, namely, Hexb and St3gal5 (Fig. [92]3a,b). The
gene symbols, abbreviations and functions are shown in Table [93]1. The
two PPI networks of the target genes (60 nodes, 115 edges; 19 nodes, 20
edges) were obtained through the MCODE module of Cytoscape, with
retention greater than or equal to 2, K-Core taken as 2, and Max. depth
taken as 100 nodes. mRNAs are pink, miRNAs are purple, and lncRNAs are
orange; the greater the connectivity of the nodes is, the greater the
number of nodes. Lamc1 and Col4a1 are the core of the mRNA-miRNA-lncRNA
interaction network and are closely related to noncoding RNAs that have
been shown to play important roles in osteogenesis or adipogenesis,
such as MALAT1, H19, XIST, and NEAT1.
Figure 3.
[94]Figure 3
[95]Open in a new tab
Expression of target genes in the database (a) mRNA–miRNA–lncRNA
interaction network map composed of Hapln1, Col4a1, Lamc1, Itga10,
Col10a1, and Tln2; (b) mRNA–miRNA–lncRNA interaction network map
composed of Hexb and St3gal5. Expression in the HPA database; (c)
histogram of differential gene tissue expression levels; (d) histogram
of differential gene gene expression levels; expression in the Bgee
database; (e) histogram of differential gene tissue expression levels.
Table 1.
Eight hub genes and their functions.
Gene symbol Describe Function
Col4a1 Collagen Type IV Alpha 1 Chain Type IV collagen proteins are
integral components of basement membranes
Lamc1 Laminin Subunit Gamma 1 Laminins, a family of extracellular
matrix glycoproteins, are the major noncollagenous constituent of
basement membranes. They have been implicated in a wide variety of
biological processes including cell adhesion, differentiation,
migration, signaling, neurite outgrowth, and metastasis
Itga10 Integrin Alpha-10 They participate in cell adhesion as well as
cell-surface-mediated signaling
Hapln1 Hyaluronan And Proteoglycan Link Protein 1 It is predicted to
enable hyaluronic acid binding activity, to be an extracellular matrix
structural constituent conferring compression resistance. And it is
predicted to be involved in skeletal system development
Col10a1 Collagen Type X Alpha 1 Chain This gene is a short chain
collagen expressed by hypertrophic chondrocytes during endochondral
ossification
Tln2 Talin 2 It is thought to associate with unique transmembrane
receptors to form novel linkages between extracellular matrices and the
actin cytoskeleton
Hexb Hexosaminidase Subunit Beta Related to hydrolase activity
St3gal5 ST3 Beta-Galactoside AlphA-2,3-Sialyltransferase 5 The protein
encoded by this gene is a type II membrane protein
[96]Open in a new tab
Target gene expression was higher at both the adipose gene level and tissue
level than at bone tissue
Analysis of the Human Protein Atlas (HPA) database revealed that the
expression levels of Col4a1 and Lamc1 were significantly higher in
adipose tissue compared to bone marrow tissue, both at the gene and
tissue levels. Similarly, Tln2 exhibited slightly elevated gene
expression in adipose tissue relative to bone marrow (Fig. [97]3c,d).
Interestingly, while Hexb and St3gal5 displayed moderately higher gene
expression in adipose tissue than in bone marrow, their tissue-level
expression patterns were reversed, with higher levels observed in bone
marrow compared to adipose tissue. The gene expression scores of the
hub genes in abdominal adipose tissue, omental fat pads, subcutaneous
adipose tissue, synovial joints, cartilage tissue, trabecular bone
tissue, tibia, bone marrow, and myeloid cells were compared using the
data obtained from the BGEE database. Col4a1 and Lamc1 had the highest
expression in abdominal adipose tissue, omental fat pads, and
subcutaneous adipose tissue, and Hexb had the highest expression in
synovial joints, trabecular bone tissues, and bone marrow cells. Hapln1
was most highly expressed in cartilage tissues, and only Tln2 and
Itga10 were expressed in bone marrow (Fig. [98]3e).
BMSCs are categorized into 11 clusters at single-cell resolution
We used t-distributed stochastic neighbor embedding (tSNE) to
investigate cellular heterogeneity within clusters, and after the
integration of the two datasets ([99]GSE128423, [100]GSE156635) (n =
44053), we identified 21 cell clusters and 11 cell types-osteoblasts (n
= 2411), chondrocytes (n = 8251), adipogenic cells (n = 5396),
endothelial cells (n = 7682), fibroblasts (n= 6949), neutrophils (n =
912), lymphocytes (n = 3957), megakaryocytes (n= 682), erythroblasts
(n= 735), stem cells (n = 4908) and blastocytes (n = 2170) -and the
biomarker expression of each subgroup is given (Fig. [101]4a,b).
Erythrocytes were not removed since our focus was on the directed
differentiation of stem cells to adipocytes, osteoblasts, and
chondrocytes. The distribution of the target genes is shown in Fig.
[102]4c.
Figure 4.
[103]Figure 4
[104]Open in a new tab
Subcluster identification of bone marrow-derived MSC single-cell
sequencing data. (a) The TSNE algorithm was applied to the first 17
PCAs for dimensionality reduction, and 22 cell clusters were
successfully classified; (b) cell clusters were manually annotated with
CellMarker 2.0 according to the composition of marker genes and were
successfully annotated into 11 subpopulations; (c) the expression
levels of the target genes
(Col4a1,Lamc1,Itga10,Hapln1,Hexb,Col10a1,Tln2,St3gal5)in the
subpopulations; (d) the marker gene bubble map. Reliability of
subcluster annotation of single-cell data.
Scoring of scRNA-seq annotation reliability proves cluster annotation
reliability
We chose three methods to evaluate the reliability of the single-cell
annotations. First, we used the WNT pathway and the ADIPOGENESIS
pathway to sort the cells (Fig. [105]5a,b). The WNT pathway was mainly
distributed in osteoblasts, adipocytes, stromal cells, and
chondroblasts, and the ADIPOGENESIS pathway was mainly distributed in
adipocytes, stromal cells, and megakaryocytes, which was in line with
our biological a priori experience. Then using the local similarity
between cells, the stemness and differentiation potential of cells were
assessed according to CytoTrace, and the degree of differentiation was
in the following order: stem cells < chondrocytes < osteoblasts <
adipoblasts. Col4a1 and Lamc1 were mainly distributed in adipocytes and
osteoblasts; Tln2 and Hexb were expressed in chondrocytes, osteoblasts,
and adipocytes; Col10a1 was distributed in a small number of
chondrocytes, Itga10 and Hapln1 were distributed in a large number of
chondrocytes, and St3gal5 was distributed in a large number of stem
cells (Fig. [106]5c). Using Monocle2 to perform a time-matching
analysis of the four types of cells, it was concluded that the BMSCs
can be divided into two different types of sources, both of which can
be differentiated into chondroblasts, and that the later stage is
accompanied by the differentiation of the adipoblasts with the
osteoblasts with a large number of adipoblasts (Fig. [107]5d). Finally,
cell cycle maps for all the subpopulations were generated and we
concluded that stem cells, lymphocytes, and neutrophils were in the
early stage of differentiation, while the remaining subpopulations were
in the late stage of differentiation (Fig. [108]5e).
Figure 5.
[109]Figure 5
[110]Open in a new tab
Proof of subgroup annotation reliability (a) the WNT pathway and the
adult ADIPOGENESIS pathway were selected to score the cell subclusters;
(b) the differentiation of the four classes of cells was assessed
according to Cytotrace; (c) the expression levels of the target genes
(Col4a1,Lamc1,Itga10,Hapln1,Hexb,Col10a1,Tln2,St3gal5)according to
Cytotrace; (d) the differentiation of the four classes of cells was
assessed based on Monocle2; (e)the differentiation of the 11 classes of
cells was assessed based on Tricycle.
Cell cycle analysis of target genes
Based on the scatter plots, Col4a1, Lamc1, Hapln1, Itga10, Tln2, Hexb,
were expressed at all times in osteoblasts, Col10a1 and St3gal5 were
expressed to a lesser extent, and Itga10 with Lamc1 became more
abundantly expressed at mitotic phase (M). Scatter plots of Runx2 and
Bglap expression cycles are given as reference (Fig. [111]6). In
adipocytes, Col4a1 and Lamc1 were significantly more expressed than in
osteoblasts, being abundantly expressed during mitosis (M), and Itga10,
St3gal5, and Col10a1 were expressed during DNA synthesis (S), which is
consistent with our previous prediction. Scatter plots of Ppar
[MATH: γ :MATH]
and Fabp4 are given as reference (Fig. [112]7). In chondrocytes, Hapln1
and Itga10 expression is reduced and Tln2 expression is elevated in
mitosis (M). Scatter plots of Sox9 and Col2a1 are given as reference
(Fig. [113]8).
Figure 6.
[114]Figure 6
[115]Open in a new tab
Expression period of target genes of Lamc1 interaction in osteoblasts
(a) Col4a1; (b) Lamc1; (c) Itga10; (d) Hapln1; (e) Hexb; (f) Col10a1;
(g) Tln2; (h) St3gal5;(i) Bglap; (j) Runx2.
Figure 7.
[116]Figure 7
[117]Open in a new tab
Expression period of target genes of Lamc1 interaction in in adipocytes
(a) Col4a1; (b) Lamc1; (c) Itga10; (d) Hapln1; (e) Hexb; (f) Col10a1;
(g) Tln2; (h) St3gal5;(i) Adipoq; (j) Fabp4.
Figure 8.
[118]Figure 8
[119]Open in a new tab
Expression period of target genes of Lamc1 interaction in chondrocy (a)
Col4a1; (b) Lamc1; (c) Itga10; (d) Hapln1; (e) Hexb; (f) Col10a1; (g)
Tln2; (h) St3gal5; (i) Col2a1; (j) Sox9.
Intercellular interactions in adipogenic, osteogenic differentiation target
two pairs of ligands that interact with Lamc1
In BMSCs, osteoblasts communicate with adipogenic cells when adipogenic
cells are used as receptors, and all three types of cells communicate
with stem cells when stem cells are used as receptors (Fig. [120]9a).
Considering that the pathway enriched for target genes is the
ECM-receptor interaction pathway, to determine the mechanism of
interactions in the stroma, we investigated the most significant
ligand–receptor interactions in the LAMININ pathway (Fig. [121]9b).
Lamc1-CD44 and Lamc1-Dag1 exhibited extreme differentiation, followed
by interactions at the level of individual ligands, and the ligands
that interact with Lamc1 were identified. LAMC1-CD44 interaction may
affect stem cell differentiation to bone or cartilage and adipose, and
LAMC1-DAG1 interaction may affect osteogenic, adipogenic, and
chondrogenic differentiation relationships (Fig. [122]9c–e).
Figure 9.
[123]Figure 9
[124]Open in a new tab
The molecular mechanism of Lamc1 interaction (a) hierarchical plot of
cellular communication; (b) the most important ligand-receptor
interactions in the laminin pathway (top 20); (c) LAMC1-Dag1
interaction chord diagram; (d) LAMC1-CD44 interaction chord diagram;
(e) Demonstrate the specific ligand–receptor interaction mechanisms of
stem cells, osteoblasts, and adipogenic and chondrogenic cells in the
Laminin pathway.
Target genes promote osteogenic differentiation and inhibit adipogenic
differentiation
The target genes Col10a1 and Hapln are predicted to play important
roles in chondrogenic differentiation, and we have confirmed this in
past reports. Similarly, Col4a1 and Itga10 have been shown to play
important roles in osteogenic differentiation, and therefore, no
subsequent validation will be performed. During osteogenic
differentiation, the alizarin red staining results revealed gradual
reddening with increasing days of induction, with MC3T3-E1 cells
reaching the reddest at 7 days (Fig. [125]10a,b). We detected the
expression of target genes and marker genes at 3 days, 5 days, and 7
days. Lamc1, Tln2, Hexb, and St3gal5 tended to be upregulated, and the
expression of the marker gene Runx2 gradually increased to a maximum at
7 days (Fig. [126]10c–e). During adipogenic differentiation, as the
duration of induction increased, the red intensity of the 3T3-L1 cells
gradually decreased, and on day 8, the red intensity of the 3T3-L1
cells decreased (Fig. [127]10f,g). We detected the expression of target
genes and marker genes after 4 days, 6 days, and 8 days. Lamc1, Tln2,
Hexb, and St3gal5 tended to be downregulated, and the expression of the
marker gene Ppar
[MATH: γ :MATH]
gradually increased to a maximum on 8 days (Fig. [128]10h–j). Laminin
can increase the expression of calcium and increase the concentration
of osteocalcin. After Lamc1 was knocked down in MC3T3-E1 and 3T3-L1
cells, the gene expression levels of Runx2 and Ocn were significantly
downregulated, and those of Ppar
[MATH: γ :MATH]
and Fabp4 were also downregulated (Fig. [129]11c–e,h–j).Similarly, the
red color of the knocked-down cells was significantly lighter than that
of the controls (Fig. [130]11a,b,f,g). Similarly, the previous
experiment was repeated with the housekeeping gene GAPDH and yielded
similar results (Supplementary Fig. [131]S1).
Figure 10.
[132]Figure 10
[133]Open in a new tab
Results of target gene expression experiments. (a) Alizarin red
staining of osteoblasts induced for 3, 5 and 7 days; (b) capture images
with camera,100
[MATH: × :MATH]
microscope,scale bar
[MATH: =500μm :MATH]
; qPCR results of target gene expression in osteoblasts: (c) 3 days;
(d) 5 days; (e) 7 days. (f) Oil Red O staining of adipocytes induced
for 4, 6 and 8 days; (g) capture images with camera, 100
[MATH: × :MATH]
microscope, scale bar
[MATH: = :MATH]
[MATH: 500μm :MATH]
; qPCR results of target gene expression in adipocyte: (h) 4 days; (i)
6 days; (j) 8 days.
Figure 11.
[134]Figure 11
[135]Open in a new tab
Results of target gene expression experiments after knockdown of Lamc1.
(a) Alizarin red staining of osteoblasts induced for 3, 5 and 7
daysafter knockdown of Lamc1; (b) capture images with camera, 100
[MATH: × :MATH]
microscope,scale bar
[MATH: = :MATH]
[MATH: 500μm :MATH]
; qPCR results of target gene expression after knockdown of Lamc1 in
osteoblasts: (c) 3 days; (d) 5 days; (e) 7 days. (f) Oil Red O staining
of adipocytes induced for 4, 6 and 8 days after knockdown of Lamc1; (g)
capture images with camera, 100
[MATH: × :MATH]
microscope, scale bar
[MATH: = :MATH]
[MATH: 500μm :MATH]
; qPCR results of target gene expression after knockdown of Lamc1 in
adipocyte: (h) 4 days; (i) 6 days; (j) 8 days.
Methods
Data collection and processing
Gene expression profiling data ([136]GSE36923, [137]GSE44303,
[138]GSE109503, [139]GSE140861, [140]GSE28205 and [141]GSE37558) were
obtained from the Gene Expression Omnibus (GEO). [142]GSE109503
contained RNA-seq data, and the remaining five gene sets were obtained
from bulk RNA-seq data. [143]GSE109503 and [144]GSE140861 are
chondrogenic differentiation datasets; [145]GSE109503 has four subsets
(two controls and two experimental groups); [146]GSE140861 has six
subsets (three controls and three experimental groups); [147]GSE28205
and [148]GSE37558 are osteogenic differentiation datasets;
[149]GSE28205 has six subsets (three control groups and three
experimental groups); [150]GSE37558 has six subsets (three control
groups and three experimental groups); [151]GSE36923 and [152]GSE44303
are adipogenic differentiation datasets; [153]GSE36923 has seven
subsets (four control groups and three experimental groups); and
[154]GSE44303 has seven subsets (four controls and three experimental
groups).
Single-cell RNA-seq data ([155]GSE128423 and [156]GSE156635) were
obtained from the Gene Expression Omnibus (GEO) based on a 10
[MATH: × :MATH]
Genomics assay with eight normal BMSC tissue samples. The summary
results of the gene expression profiling dataset information are
provided in Table [157]2.
Table 2.
Summary information on gene expression profiling datasets.
DataSet Samples Organism Type of cell Differentiation time Group DOI
[158]GSE36923
[159]GSM906367,
[160]GSM906368,
[161]GSM906369.
Homo sapiens BMSCs 0 Day Control 10.1016/j.biocel.2013.11.010
[162]GSM906370,
[163]GSM906371,
[164]GSM906372.
15 Days Adipogenic
[165]GSE44303
[166]GSM1082817,
[167]GSM1082818,
[168]GSM1082819.
Homo sapiens BMSCs 0 Day Control 10.1093/nar/gku567
[169]GSM1082844,
[170]GSM1082845,
[171]GSM1082846.
14 Days Adipogenic
[172]GSE109503
[173]GSM2944707,
[174]GSM2944708,
[175]GSM2944709.
Homo sapiens BMSCs 0 Day Control 10.1096/fj.201800534R
[176]GSM1082844,
[177]GSM1082845,
[178]GSM1082846.
14 Days Chondrocyte
[179]GSE140861
[180]GSM4188963,
[181]GSM4188970.
Homo sapiens BMSCs 0 Day Control 10.1089/ten.TEA.2016.0559
[182]GSM4188967,
[183]GSM4188974.
14 Days Chondrocyte
[184]GSE28205
[185]GSM698427,
[186]GSM698433,
[187]GSM698439.
Homo sapiens BMSCs 0 Day Control 10.1002/jbmr.1578
[188]GSM698429,
[189]GSM698431,
[190]GSM698436.
14 Days Osteoblast
[191]GSE37558
[192]GSM921574,
[193]GSM921575,
[194]GSM921576,
[195]GSM921578.
Homo sapiens BMSCs 0 Day Control 10.1186/1471-2164-15-965
[196]GSM921584,
[197]GSM921585,
[198]GSM921586.
12 Days Osteoblast
[199]GSE128423
[200]GSM3674224,
[201]GSM3674225,
[202]GSM3674226,
[203]GSM3674227,
[204]GSM3674228,
[205]GSM3674229.
Mus musculus BMSCs Undifferentiation – 10.1016/j.cell.2019.04.040
[206]GSE156635
[207]GSM4735397,
[208]GSM4735398.
Mus musculus BMSCs Undifferentiation – 10.1016/j.celrep.2021.109352
[209]Open in a new tab
Microarray data were normalized ,and variance was analyzed using the
Limma package in the R/Bioconductor software for matrix data from each
GEO dataset, and RNA-seq data were normalized ,as was variance analyzed
using the edgeR package. DEGs of the same cell type identified from
each of the six datasets were integrated based on different
differentiation categories using the RobustRankAggreg package. |log2FC|
[MATH: ≥ :MATH]
1 and P
[MATH: ≤ :MATH]
0.05 were considered to indicate statistical significance.
PCA plots for each dataset were plotted using the FactoMineR package
and the factoextra package in the R/Bioconductor software. pheatmap
package was used to plot heat maps of the genes after data merging.
ggplot2 package was used to plot volcano plots after data merging. Venn
plots were generated using TBtools software (version x64 v1.09876). GO
function enrichment and KEGG pathway enrichment were performed for 31
differential genes (DEGs) using the clusterProfiler package in R
software. adj. P
[MATH: ≤ :MATH]
0.05 was considered statistically significant. Bar and bubble plots
were drawn using the ggplot2 package, and network plots were drawn
using the enrichplot package.
scRNA-seq data were preprocessed using the Seurat package in R software
for matrix data from each GEO dataset, the PercentageFeatureset
function was used to determine the proportion of mitochondrial genes,
and correlation analyses were used to investigate the relationships
between sequencing depth and mitochondrial gene sequences and total
intracellular sequences. Each gene was expressed in at least 3 cells.
The gene expression in each cell was greater than 500 and less than
6000, the mitochondrial content was less than 30%, and the UMI in each
cell was at least greater than 200. The scRNA-seq data were normalized
by the NormalizeData method after data filtering, and highly variable
genes were identified by the FindVariableFeatures method to compute the
mean expression and dispersion and converted to logarithmic format.
Next, the data from the two scRNA-seq datasets were integrated using
the Harmony package, and then we performed principal component analysis
(PCA) and reduced the data to the first 17 PCA components (the number
of components selected based on the standard deviation of the principal
components—in the platform area of the Elbow Graph). To visualize the
data, we used the Seurat package for t-distribution random neighbor
embedding (tSNE) to project the cells in two-dimensional space. We
visualized clusters on 2D maps generated using t-distributed random
neighbor embedding (t-SNE). Clusters were annotated according to
CellMarker 2.0 ([210]http://yikedaxue.slwshop.cn/), and the annotated
clusters were scored using the AUCell package, selecting the WNT
pathway-a key pathway for osteogenesis-and the ADIPOGENESIS pathway-a
key pathway for adipogenesis. The cell cycle of the annotated clusters
was inferred using the tricycle package, and the cell cycle in which
the target genes are located was determined. The Cytotrace package was
used to determine the stemness and differentiation potential of stem
cells, chondrocytes, osteoblasts, and adipocytes, and based on the
results of Cytotrace, Monocle2 was used to determine the direction of
differentiation of the four types of cells and to determine the site of
expression of the target gene.
The CellChat package was used to query the role of intercellular
communication, and the ECM-Receptor database was selected for
subsequent analysis based on the KEGG enrichment results. The identify
OverExpressedGenes function of the CellChat package was used to
identify highly expressed genes, the identify OverExpressedInteractions
function was used to identify highly expressed pathways, the project
Data function was used to project the relationship between the highly
expressed genes and the pathways to the PPI network, the
computeCommunProb function was used to calculate the probability of
cell-to-cell communication, data with the minimum number of cells less
than 10 were removed, the computeCommunProbPathway function was used to
calculate the cell-to-cell communication at the level of signaling
pathways, the aggregateNet function was used to calculate the
aggregated cell-to-cell communication network, and the “LAMININ”
signaling pathway was extracted.
The mechanism map was drawn via Figdraw
([211]https://www.figdraw.com/static/index.html).
PPI network construction
The String website ([212]https://cn.string-db.org/) analyzed protein
functional interactions and selected interactions with a composite
score>0.4. The RNAInter database ([213]http://www.rnainter.org/) is a
complete source of RNA interaction data for studying the relationships
between mRNAs, lncRNAs, and microRNAs. The RNAInter database was used
to identify miRNAs that interact with mRNAs, as well as lncRNAs that
interact with miRNAs, and interactions with a score > 0.5 was selected.
Cytoscape (version v3.9.1) is an open-source bioinformatics software
platform for visualizing molecular interaction networks. The
relationship between target genes and non-coding RNAs was plotted using
Cytoscape.
Tissue distribution and gene expression identification
Protein and gene expression levels in bone marrow and adipose tissue
were compared with those in the HPA database using The Human Protein
Atlas ([214]https://www.proteinatlas.org/) as a validation tool to
determine whether there are differences in the expression of these
genes in osteogenic and adipogenic differentiation. Expression scores
in abdominal adipose tissue, omental fat pads, subcutaneous adipose
tissue, tibia, synovial tissue, synovial joints, cartilage tissue,
trabecular bone tissue, and bone marrow tissue were used to validate
whether there are differences in the expression of these genes in
osteogenic and adipogenic as well as cartilaginous differentiation
using the BGEE database ([215]https://bgee.org/). Bar graphs were drawn
using GraphPad Prism 9.
Cell culture and differentiation
MC3T3-E1 cells (Procell Life, Science & Technology, China) were
cultured in MEM
[MATH: α :MATH]
medium supplemented with 10% fetal bovine serum (FBS), 100 units/ml
penicillin and streptomycin at
[MATH: 37∘
C :MATH]
in a 5%
[MATH: CO2 :MATH]
incubator. The medium was changed every 2–3 days and the cells were
used for experiments in which the cells were grown to the logarithmic
growth phase. For osteogenic differentiation, MC3T3-E1 cells were
cultured in osteogenic medium supplemented with 10 mM
[MATH: β :MATH]
-glycerophosphate and 50
[MATH: μ :MATH]
g/mL L ascorbic acid osteogenic inducer. The cells were induced for 3,
5, or 7 days. 3T3-L1 cells (Procell Life, Science & Technology, China)
were cultured in DMEM medium supplemented with 10% fetal bovine serum
(FBS), 100 units/ml penicillin and streptomycin at
[MATH: 37∘
C :MATH]
in a 5%
[MATH: CO2 :MATH]
incubator.The medium was changed every 2-3 days, and the cells were
used for experiments in which the cells were grown to the logarithmic
growth phase. For adipogenic differentiation, 3T3-L1 cells were
cultured in adipogenic induction medium supplemented with 0.5
[MATH: μ :MATH]
M 3-isobutyl-1-methyl xanthine , 1
[MATH: μ :MATH]
M dexamethasone and 10
[MATH: μ :MATH]
g/mL insulin adipogenic inducer for 2 days, and the medium was replaced
with adipogenic maintenance medium supplemented with 10
[MATH: μ :MATH]
g/mL insulin for 2 days. The medium was finally replaced with DMEM, and
the culture was continued for 2–4 days.
Alizarin red staining method and Oil Red O staining
After fixation with 80% ethanol for 30 min, cells were incubated with
fresh alizarin red solution (Cyagen) for 10 min. After fixation with 4%
neutral formaldehyde for 30 min, cells were incubated with saturated
Oil Red O staining solution (Beyotime Biotechnology; Shanghai; China)
for 10 min.
Quantitative reverse transcription PCR
Total RNA was extracted using triazole lysate lysis (Sangon Biotech;
Shanghai; China), isopropanol, chloroform, and 75% ethanol solutions.
Total RNA reverse transcription was induced using the HiScirpt Reverse
Transcriptase kit (Novozymes Bio; Nanjing; China) The genes and primers
used in the osteogenesis and adipogenesis process are listed in Table
[216]3. Performed with a QuantStudio 5 real-time fluorescent
quantitative PCR system (Applied Biosystems; Shanghai; China). Each
sample was performed in triplicate. The sequences of the mRNAs were
calculated according to the
[MATH: ΔΔCT :MATH]
relative stacking method. mRNAs were sequenced as Table [217]3.
Table 3.
qPCR primer sequences.
Gene Forward primers Reverse primers
Lamc1 TGCCGGAGTTTGTTAATGCC TGGTTGTTGTAGTCGGTCAGG
Tln2 AGGAAAGGGATTTGGCTAGAAGC CCAACATTCGGATTTTCTGAGGC
Hexb CTGGTGTCGCTAGTGTCGC CAGGGCCATGATGTCTCTTGT
St3gal5 AAAAGAATGCACTATGTGGACCC ATAGCCGTCTTCGCGTACCT
Runx2 CGGTCTCCTTCCAGGATGGT GCTTCCGTCAGCGTCAACA
Ocn GAACAGACAAGTCCCACACAGC TCAGCAGAGTGAGCAGAAAGAT
Ppar
[MATH: γ :MATH]
GGAAGACCACTCGCATTCCTT GTAATCAGCAACCATTGGGTCA
Fabp4 ATCAGCGTAAATGGGGATTTGG GTCTGCGGTGATTTCATCGAA
[MATH: β :MATH]
-actin GTGACGTTGACATCCGTAAAGA GCCGGACTCATCGTACTCC
[218]Open in a new tab
Lamc1 siRNA interference
Lamc1 siRNA was obtained from GenePharma Corporation. siRNA was
transfected into MC3T3-El cells using the gp transfection mate
(GenePharma Corporation) according to the manufacturer’s instructions.
24 h later, induction was initiated, and cells were collected 3–7 days
later for alizarin red staining, and real-time quantitative RT PCR
analysis. The siRNA was transfected into 3T3-L1 cells using gp
transfection mate (GenePharma Corporation) according to the
manufacturer’s instructions. After 90% confluence, induction was
initiated, and the cells were collected 4–8 days later for oil red o
staining, and real-time quantitative qPCR analysis experiments. The
sequence of the siRNA was as follows. NC:5
[MATH:
′
:MATH]
-UUCUCCGAACGUGUCACGUTT-3
[MATH:
′
:MATH]
and 5
[MATH:
′
:MATH]
-ACGUGACACGUUCGGAGAATT-3
[MATH:
′
:MATH]
; Lamc1-siRNA: 5
[MATH:
′
:MATH]
-GCCGUAAUCUCAGACAGUUTT-3
[MATH:
′
:MATH]
and 5
[MATH:
′
:MATH]
- AACUGUCUGAGAUUACGGCTT-3
[MATH:
′
:MATH]
.
Molecular docking analysis
The Hdock website ([219]http://hdock.phys.hust.edu.cn/) was used to
perform simulated docking studies of Laminin-111 (Entry ID:5mc9)with
DAG1 (Entry ID: 5llk) or CD44 (Entry ID:4pz3), the PymoL software
(version v3.0.0) was used to perform pre-docking treatments (water
molecules from the structure and co-crystallized ligands were removed,
H atoms were added) and Pymol was used for the visual presentation of
the docking complexes, with the results with the highest docking scores
being selected.
Discussion
To investigate the molecular mechanisms of the multidirectional
differentiation of BMSCs, especially osteogenesis, adipogenesis and
chondrogenesis, BMSCs exhibited significant cellular heterogeneity, and
an integrated analysis of the multiscale transcriptome was performed in
the present study. These DEGs were analyzed by bulk RNA-seq and
scRNA-seq, and it was found that the late stage of differentiation was
the most significant in terms of interactions with the ECM receptor. It
is well known that the ECM enhances cell recruitment through cell
surface receptors, which determine cellular interactions with the ECM
and trigger specific cellular functions such as adhesion, migration,
proliferation, and differentiation^[220]30. Several studies have
reported the critical role of the matrix in regulating the
differentiation of MSCs toward the bone or adipose
lineage^[221]30,[222]31.Recent studies have shown that stem cells can
mechanically sense the hardness of their microenvironment, and the
direction of differentiation of MSCs may change with changes in the
matrix composition^[223]32,[224]33. For example, the extracellular
matrix supplemented with glucan sulfate promotes the osteogenic
differentiation of mesenchymal stem cells^[225]34, and type I collagen
inhibits adipogenesis and differentiation by activating yes-associated
proteins^[226]35. The latest research by Yangzi Jiang et al. showed
that ECM produced by adult stem cells from different tissues has
different cartilage induction abilities^[227]36. Osteoblasts are the
main bone-forming cells, and they produce extracellular proteins that
constitute the major components of bone. During bone formation, BMSCs
differentiate into osteoblasts, which produce bone by synthesizing an
extracellular matrix composed of various proteins. The initial
deposition of the extracellular matrix is called the osteoid, which is
then mineralized into bone tissue by the accumulation of hydroxyapatite
(
[MATH:
Ca10(PO4)6(OH)2 :MATH]
) as calcium phosphate^[228]37. As osteoblasts mature, they transform
into osteocytes, and revealing the detailed mechanisms that regulate
the later stages of osteoblast differentiation and function is
important for clinical applications^[229]38. However, studies on the
mechanisms underlying the differentiation of bone marrow-derived MSCs
in the late stages of differentiation still need to be performed. We
therefore selected an mRNA microarray dataset at the end of
differentiation to identify genes that showed significant heterogeneity
in the differentiation of BMSCs into adipocytes, osteoblasts, and
chondrocytes. lamc1 tended to increase osteoblastic differentiation and
decrease adipocytes, and Col4a1 had the opposite effect on
differentiation. Highly expressed Lamc1 and Col4a1 play key roles in
the protein interaction network, and their interactions with the key
molecules involved in osteogenic, adipogenic, and chondrogenic
differentiation, H19, XIST, NEAT1, and MALAT1, have been identified.
According to our results, Hui-jian Chen et al. reported that Lama4 and
Col4a1 are expressed in white adipose tissue at different sites and are
upregulated during adipocyte differentiation^[230]39. Interestingly,
the mRNA expression of Col4a1 was significantly increased after
adipogenic induction of MSCs in a recent study, which is consistent
with our expectations^[231]40. More importantly, laminin in the cell
matrix contains three subunits:
[MATH: α :MATH]
,
[MATH: β :MATH]
, and
[MATH: γ :MATH]
. Lamc1 is the most commonly used
[MATH: γ :MATH]
chain and is present in most laminin molecules^[232]41. During the
process of osteogenesis, Laminin activates downstream signaling
pathways such as Rho GTPase and MAPK by interacting with Integrin,
which in turn regulates the processes of cellular bone morphogenesis
and bone matrix deposition, and participates in the directional
differentiation of stem cells to osteoblasts^[233]42,[234]43.Lamc1 is
present in LN-111 and LN-511, it has been shown to promote osteogenic
differentiation by binding to integrins^[235]41,which are key factors
for the development of osteoblasts. The HPA database, Bgee database,
and single-cell transcriptome data analysis revealed that Lamc1
expression was significantly greater in adipocytes than in osteoblasts,
but Lamc1 tended to promote bone formation and inhibit adipogenic, and
this tendency became more obvious with increasing induction time.
Further studies showed that after Lamc1 knockdown, the expression of
osteogenic and adipogenic markers was significantly decreased, and
differentiation was inhibited. It is therefore reasonable to assume
that Lamc1 plays an important role in regulating the balance of bone
adipogenesis^[236]44.
The distribution of target genes in bone marrow-derived MSCs, the cell
cycle, and ligand–receptor interactions were determined by single-cell
transcriptome analysis. Among these genes, Lamc1, which is expressed
mainly during the G2/M phase in adipogenic cells and may affect
intracellular signaling through interactions with Dag1, was used as a
focus for determining the balance between osteogenic and adipogenic
differentiation. Dag1 (also known as
[MATH: α :MATH]
-glycoprotein) is involved in the adhesion between the extracellular
matrix and cells, helps to maintain cell aggregation and structural
stability, and participates in the assembly process of laminin and the
basement membrane^[237]45. Recent studies have shown that Dag1 plays a
role in cartilage formation and osteoblast differentiation in
vivo^[238]46,[239]47.Another finding is that the interaction of Dag1
with laminin has been reported^[240]48.To prove our prediction,
molecular docking experiments were performed on laminin and Dag1. The
experimental results showed that laminin and Dag1 formed a total of 11
hydrogen bonds (Fig. [241]12a). Our data support the conclusion that
the interaction of Dag1 and Lamc1 may influence the balance between the
adipogenic and osteogenic differentiation of MSCs through cell
adhesion, signaling, and gene regulation (Fig. [242]12c). Similarly, a
total of four hydrogen bonds were formed after molecular docking
experiments were performed on laminin and CD44, initially validating
the interaction between Lamc1 and CD44 (Fig. [243]12b).
Figure 12.
[244]Figure 12
[245]Open in a new tab
Molecular docking and mechanism maps (a) Results of molecular docking
of laminin to DAG1; (b) Results of molecular docking of laminin to
CD44; (c) mechanism maps.
In conclusion, this study aimed to identify DEGs that may be involved
in osteogenic differentiation, adipogenic differentiation, and the
onset and progression of chondrogenic differentiation of bone marrow
mesenchymal stem cells (BMSCs). A total of eight hub genes were
identified, four of which were discovered and proposed for the first
time in this study. Although these findings are preliminary, they
suggest the existence of previously unknown regulatory relationships
governing the differentiation process of BMSCs. Unfortunately, our
experiments did not reveal new regulatory relationships between
chondrogenesis and adipogenesis or osteogenesis, which may be related
to our choice of a more conservative data merging approach.However,
this conservative strategy ensured the accuracy of bioinformatic
predictions. Despite these limitations, the identified hub genes and
their potential interactions provide valuable insights and lay the
foundation for further investigation into the molecular mechanisms
underlying the multi-lineage differentiation of BMSCs.
Conclusion
This study aimed to identify the key biological processes involved in
the late stages of differentiation of BMSCs. The following conclusions
were demonstrated: eight target genes were identified in the context of
trilineage differentiation antagonism, which is concentrated in the
extracellular matrix and is associated with ECM-receptor
interactions.Notably, Lamc1 expression was higher in adipose tissue
than in bone tissue. Furthermore, Lamc1 exhibited maximal expression
during the mitotic phase of the cell cycle. Bioinformatic analysis
suggested that Lamc1 interacts with CD44 and may influence the
differentiation of stem cells towards osteoblastic, chondrogenic, or
adipogenic lineages. Additionally, the interaction between Lamc1 and
Dag1 may affect the balance of trilineage differentiation in BMSCs.
Experimental validation confirmed that Lamc1 was upregulated during
osteogenic differentiation and downregulated during adipogenic
differentiation, indicating its essential role in regulating the
differentiation process.
Supplementary Information
[246]Supplementary Information.^ (1MB, pdf)
Author contributions
L.Z. and S.L. wrote the main manuscript text and Y.P. typesetted and
checked the manuscripts. J.Z. led and supervised the planning and
execution of the experiments. All authors reviewed the manuscript.
Funding
This research was supported by the National Natural Science Foundation
of China (Grant numbers 81960171 and 82360167) and Guizhou Provincial
Science and Technology Program (Guizhou Science and Technology
Foundation-ZK[2022] General 620).
Data availibility
The datasets covered in this reasearch are all publicly available
transcriptomic data from the Gene Expression Omnibus (GEO). Ullah M,
Sittinger M, Ringe J. Transdifferentiation of adipogenically
differentiated cells into osteogenically or chondrogenically
differentiated cells: phenotype switching via dedifferentiation. Int J
Biochem Cell Biol. 2014 Jan;46:124-37. doi:
10.1016/j.biocel.2013.11.010. Epub 2013 Nov 22. [247]GSE36923. Tomaru
Y, Hasegawa R, Suzuki T, Sato T, Kubosaki A, Suzuki M, Kawaji H,
Forrest AR, Hayashizaki Y; FANTOM Consortium; Shin JW, Suzuki H. A
transient disruption of fibroblastic transcriptional regulatory network
facilitates trans-differentiation. Nucleic Acids Res. 2014
Aug;42(14):8905-13. doi: 10.1093/nar/gku567. Epub 2014 Jul 10.
[248]GSE44303. Huynh NPT, Zhang B, Guilak F. High-depth transcriptomic
profiling reveals the temporal gene signature of human mesenchymal stem
cells during chondrogenesis. FASEB J. 2019 Jan;33(1):358-372. doi:
10.1096/fj.201800534R. Epub 2018 Jul 9. [249]GSE109503. Somoza RA,
Correa D, Labat I, Sternberg H, Forrest ME, Khalil AM, West MD, Tesar
P, Caplan AI. Transcriptome-Wide Analyses of Human Neonatal Articular
Cartilage and Human Mesenchymal Stem Cell-Derived Cartilage Provide a
New Molecular Target for Evaluating Engineered Cartilage. Tissue Eng
Part A. 2018 Feb;24(3-4):335-350. doi: 10.1089/ten.TEA.2016.0559. Epub
2017 Jul 28.[250]GSE140861. Dani N, Olivero M, Mareschi K, van Duist
MM, Miretti S, Cuvertino S, Patané S, Calogero R, Ferracini R,
Scotlandi K, Fagioli F, Di Renzo MF. The MET oncogene transforms human
primary bone-derived cells into osteosarcomas by targeting committed
osteo-progenitors. J Bone Miner Res. 2012 Jun;27(6):1322-34. doi:
10.1002/jbmr.1578. [251]GSE28205. Alves RD, Eijken M, van de Peppel J,
van Leeuwen JP. Calcifying vascular smooth muscle cells and
osteoblasts: independent cell types exhibiting extracellular matrix and
biomineralization-related mimicries. BMC Genomics. 2014 Nov
7;15(1):965. doi: 10.1186/1471-2164-15-965. [252]GSE37558. Baryawno N,
Przybylski D, Kowalczyk MS, Kfoury Y, Severe N, Gustafsson K,
Kokkaliaris KD, Mercier F, Tabaka M, Hofree M, Dionne D, Papazian A,
Lee D, Ashenberg O, Subramanian A, Vaishnav ED, Rozenblatt-Rosen O,
Regev A, Scadden DT. A Cellular Taxonomy of the Bone Marrow Stroma in
Homeostasis and Leukemia. Cell. 2019 Jun 13;177(7):1915-1932.e16. doi:
10.1016/j.cell.2019.04.040. Epub 2019 May 23. [253]GSE128423. Sivaraj
KK, Jeong HW, Dharmalingam B, Zeuschner D, Adams S, Potente M, Adams
RH. Regional specialization and fate specification of bone stromal
cells in skeletal development. Cell Rep. 2021 Jul 13;36(2):109352. doi:
10.1016/j.celrep.2021.109352.[254]GSE156635.
Competing interests
The authors declare no competing interests.
Footnotes
Publisher's note
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
These authors contributed equally: Lixia Zhao and Shuai Liu.
Supplementary Information
The online version contains supplementary material available at
10.1038/s41598-024-69629-4.
References