Bone is a metabolically active organ that undergoes continuous remodeling throughout life (1). Bone remodeling involves the bone resorption by osteoclasts followed by the formation of bone matrix through the osteoblasts. Osteoblasts are derived from bone marrow mesenchymal stem cells (BMMSCs). In addition to osteoblasts, BMMSCs can differentiate into adipocytes and chondrocytes. The adipogenic and osteoblastogenic differentiation are competitively balanced (2, 3). Osteoblastogenic development is a complex process that requires the tight regulation of specific transcription factors activation or suppression in response to local signaling pathways (4, 5). For instance, Wnt signaling is implicated in cell fate determination, proliferation, differentiation and apoptosis of osteoblasts (4). The combination of the Wnt and the seven-pass transmembrane receptor Frizzled (FZD) or single-pass transmembrane co-receptor complex LRP5/6 activates the canonical Wnt signaling pathway to promote downstream osteoblast target gene expression (5).
Cell heterogeneity is an important contributor to biological function. During the entire osteoblast development process from BMMSCs to mature osteoblasts, several cell subtypes have been identified. Two subpopulations of BMMSCs were found: a skeletal stem cell subpopulation that differentiated into bone, cartilage and fat, and another subpopulation that modulated immune function and inflammation (6). Our previous studies performed single-cell sequencing of human BMMSCs and osteoblasts. In BMMSCs population, we identified osteoblast precursors, adipocyte precursors, chondrocyte precursors and terminal-stage cells subpopulations in LEPRhighCD45low BMMSCs (7). In osteoblasts, we identified two known cell subtypes and a novel undetermined osteoblasts subtype. Meanwhile, our studies revealed the functional characteristics of each osteoblast subtype in bone metabolism, angiogenesis modulation and hematopoietic stem cells (HSC) niche regulation (8). The osteoblast development from BMMSCs to mature osteoblast was a continuous process. But in our previous studies, BMMSCs and osteoblasts were analyzed separately, which neglected the continuity of differentiation and the cell to cell communication of different cell subtypes. Therefore, due to the lack of comprehensive combined analysis of BMMSCs and osteoblasts, the regulation mechanisms involved in osteoblast development process and cell to cell communication between BMMSCs and osteoblasts at single cell level remained limited.
In this study, we conducted an integrated analysis of BMMSCs and osteoblasts at the single-cell level, reconstructed pseudotime trajectories and analyzed cell to cell communication between BMMSCs and osteoblasts. We suggested that SFRP4 and WIF1 which highly expressed in mature osteoblasts, inhibited the binding of FZD1 to Wnt ligand in BMMSCs, thereby further inhibiting the regulation of osteogenic differentiation.
The study subject of osteoblast included one 31-year-old male with osteoarthritis and osteopenia (BMD T-score: 0.6 at lumbar spine, −1.1 at the total hip). And study subjects of bone marrow mesenchymal stem cells consisted of two subjects: one 67-year-old female with osteoporosis (T-score: −3.3 at lumbar spine, −3.7 at the total hip), the other 84-year-old male with osteoarthritis (T-score: −0.9 at lumbar spine, 2.7 at the total hip).
All subjects were screened with a detailed questionnaire, medical history, physical examination, and bone mineral density (BMD) testing before surgery. Subjects were excluded from this study if they had conditions that affect bone metabolism, including diseases of the kidney, liver, parathyroid, and thyroid, or any of the following conditions: diabetes mellitus, hyperprolactinemia, oophorectomy, ankylosing spondylitis, malabsorption syndromes, malignant tumors, hematologic diseases, or previous pathologic fractures.
We isolated osteoblasts from the human femoral head according to a previously widely used protocol with only minor modifications (9). Briefly, bone tissue was divided into bone fragments with bone forceps and washed with phosphate-buffered saline (PBS). The fragments were placed into the digestive solution containing a highly purified, endotoxin-free type II collagenase (1 mg/ml, Cat: A004174-0001, Sangon Biotech, Shanghai, China) and digested for two rounds at 37℃, the first round for 30 min and the second round for 60 min. The solution was then collected and filtered through a 70 μm filter and incubated with red blood cell lysis buffer for five minutes. The collected cells were washed twice with PBS and incubated with human CD31/34/45-PE (Cat:303106, Cat:343606, Cat:304008, BioLegend, San Diego, CA, USA), and human ALPL-APC (Cat: FAB1448A, R&D Systems, Minneapolis, MN, USA) antibodies. After incubation, ALP+/CD45/34/31− cells were collected as osteoblasts by fluorescence-activated cell sorting (FACS).
BMMSCs were isolated from human bone marrow cells extracted from the bone marrow cavity of the femoral diaphysis according to the widely used isolation protocol (10). Briefly, the bone marrow was diluted with twice the volume of PBS and mixed gently. The mixture was then evenly layered onto an equivalent volume of Ficoll (GE Healthcare, Chicago, Illinois, USA) and the buffy coat was separated by centrifugation (440 g, 35 min, 4℃). The separated buffy coat was transferred to a new 15 ml centrifuge tube and washed with PBS. Then, red blood cells were lysed with RBC Lysis Buffer (Thermo Fisher, Waltham, MA, USA). After washing twice with PBS, the remaining cells were further purified with CD271 magnetic MicroBeads for positive selection (11). BMMSCs were incubated for 10 min at 4℃∼8℃ with monoclonal antibody (mAb) against CD271. After washing, the cells were incubated with anti-IgG1 immunomagnetic beads for 15 min at 4℃. The cell suspension was placed on a column in a cell separator (Miltenyi Biotec), and the positive fraction was subjected to a second separation step. The cells were then counted and assessed for viability with a CountstarⓇ Rigel S3 fluorescence cell analyzer (ALIT Life Science Co., Ltd, Shanghai, China).
The RNA library was constructed using Single Cell 3'Library Gel Bead Kit V3 on the 10x platform. The sequencing principle was based on the droplet method. The single cell suspension was added to the 10x Genomics Chromium Controller for single cell capture and sequencing tag addition. After PCR amplification of the library, the two types of single-cell RNA-seq libraries were sequenced separately on the Illumina Novaseq6000 platform.
For separate preprocessing of BMMSCs and osteoblast data sets, Seurat (12) package V3 (https://satijalab.org/seurat/) was used for filtering, variable gene selection, dimensionality reduction analysis, and clustering. From the dataset of osteoblast, we obtained 7,506
After the first pre-screening, we got 3,756 BMMSCs. For details, please refer to our previous research (7) and GSE147287. Among them, cluster B5 highly expressed hematopoietic marker gene
To analyze the datasets from different cell types, we also used Seurat V3 for integrated analysis. To unify quality control criteria, we removed cells with a high fraction of mitochondrial transcripts (20% for BMMSCs and osteoblasts, respectively), a low unique molecular identifier counts >40,000, and a low number of detected genes (<200&>5,000) for analysis. Finally, we retained 8,841 cells (including 2,865 BMMSCs and 5,976 osteoblasts) for further analyses. First, the first 2,000 genes with the most obvious cell-to-cell differences were selected by the “FindVariableFeatures” function in the Seurat package. Subsequently, principal component analysis (PCA) analysis was performed on the submatrix consisting of the highly variable genes. The top 20 principal components were used for clustering analysis and visualization.
To assess cell cycle effects on the dataset, we used the “CellCycleScoring” function in the Seurat package to score each cell. Each cell was divided into G1, G2M, and S phases based on Seurat’s built-in cell cycle marker gene dataset (https://satijalab.org/seurat/v3.2/cell_cycle_vignette.html).
To delineate the developmental progression of bone marrow mesenchymal cells to mature osteoblasts in pseudotime, we performed the diffusion map analysis (14) as implemen-ted in the R package destiny (bioconductor.org/packages/destiny). The method inferred the low-dimensional manifolds by estimating the eigenvalues and eigenvectors for the diffusion operator related to the data. We used the variable genes identified by Seurat as the basis to construct a single cell differentiation trajectory. Then, the diffusion map was constructed using the “DiffusionMap” function of R package destiny.
Gene Ontology (GO) term enrichment analysis was performed using the standalone clusterProfiler package (https://github.com/YuLab-SMU/clusterProfiler). When the adjusted p-value was less than 0.05, the term we obtained was considered to be significantly enriched.
To infer the hypothetical intercellular communication, we used iTALK (15) R packages (https://github.com/Coolgenome/iTALK) to visualize the ligand-receptor networks. We used the built-in database of ligand-receptor interaction for iTALK, which collected a total of 2,648 nonredundant and known interacting ligand-receptor pairs. The ligand receptor pairs were classified into 4 categories based on the primary function: cytokines/chemokines, immune checkpoint genes, growth factors, and others. iTALK incorporates commonly used algorithms for batch effects corrections and differential gene expression analysis.
We constructed the Protein-Protein Interaction (PPI) network based on STRING v10 (https://string-db.org/) database and Cytoscape (16) software. The first 30 genes of each subcluster were selected, and then the genes of the two subclusters were input into the STRING v10 database to obtain the basic protein-protein interaction network. We imported the network data into Cytoscape software, and used the built-in program cytoHubba (17) to identify hub genes of the PPI network.
The scRNA-seq data of human osteoblasts (8) and BMMSCs (7) from two subjects used in this study were detailed in our previous publication. After quality control and excluding contaminant cell types, 8,841 cells (5,976 osteoblasts and 2,865 BMMSCs) were used in the integration analysis. We performed unsupervised detection using principal component analysis in Seurat (12) and generated a two-dimensional (2D) embedding plot for visualization using the uniform manifold approximation and projection (UMAP) algorithm (18). 8 clusters (C1-C8) were identified based on highly variable genes (Fig. 1A). C1-C3 were BMMSCs (BMMSC1, BMMSC2, BMMSC3). C4-C6 were preosteoblasts1-3 clusters (PreOB1, PreOB2, PreOB3). C7 and C8 were immature osteoblasts cluster (ImmatOB) and mature osteoblasts cluster (MatOB) respectively. The gene expression profiles between the two subjects were strongly correlated (R2=0.96, Fig. 1B), so we combined the data from the two subjects for subsequent analysis.
Next, we identified highly expressed genes in each cluster (Fig. 1C) and used Gene Ontology (GO) enrichment analysis to characterize the most significant gene expression signatures from each cell cluster (Fig. 1D). Most of clusters were enriched for GO terms related to “ossification”, “bone development”, “osteoblast differentiation”, “bone mineralization”. Most of clusters expressed osteoblasts related marker genes (eg,
To assess if clustering was critically influenced by the cell cycle, we calculated the cell cycle score to determine the stages of the cells, in which cell cycle scores were based on its expression of G2/M and S phase markers (12). We found different cell cycle stages were present at a roughly similar proportion in each subpopulation (Supplementary Fig. S3B and S3C), therefore, the cluster results were not affected by cell cycle heterogeneity.
Next, we inferred osteogenesis differentiation trajectory using unsupervised ordering of the scRNA-seq for osteogenesis cells by diffusion mapping (Fig. 2A) (14). By comparing the distribution of the pseudotime, we found that cluster BMMSC1was enriched in the early stage of pseudotime (Fig. 2B). The clusters ImmatOB and MatOB, which highly expressed specific genes of late osteoblasts stage like
Based on the functional enrichment analysis of the highly expressed genes, we found that the GO terms related to the Wnt/β-catenin pathway, such as “regulation of Wnt signaling pathway”, “positive regulation of Wnt signaling pathway”, “positive regulation of canonical Wnt signaling pathway”, “non-canonical Wnt signaling pathway” were mainly enriched in BMMSC1 (Fig. 3A). Comparing the BMMSCs subclusters related to osteogenesis (BMMSC1) with adipogenesis (BMMSC2), we found that the expression of
Preosteoblasts, which were in the transitional stage from skeletal progenitors to osteoblasts, not only expressed BMMSCs specific genes, such as leptin receptor (
Interestingly, we noticed that two preosteoblasts subclusters with opposite functions were novelly identified in our studies. PreOB2 highly expressed nuclear receptor subfamily 4 group A member 1 and 2 (
GO enrichment analysis showed that terms which were related to bone development in PreOB2 included “positive regulation of osteoblast differentiation”, “regulation of bone mineralization”, “extracellular structure organization”, “extracellular matrix organization”
Next, we checked the expression of mature osteoblasts signature genes in the integrated data. Except the expression of
To investigate the biological interaction between BMMSCs and osteoblasts, we performed intercellular communication analysis using an R Package “iTALK” (15), which can identify ligands and receptors in scRNA-seq data (Supplementary Table S1) and analyze mediated cross-signal of cell communication (Fig. 4A). The top 30 pairs of ligand-receptor related to intercellular communication in the osteogenic lineage were visualized (Fig. 4B). We observed strong interaction between mature osteoblasts and BMMSCs. The results showed integrin subunit β1 (
Discoidin domain receptor 2 (DDR2) and COL1A1 were another significant ligand-receptor pair. COL1A1 is a common marker of osteoblasts, and the expression of COL1A1 is up-regulated during the differentiation of osteoblasts from preosteoblasts to mature osteoblasts (42). Previous studies have shown that DDR2 is a potential cellular marker for osteogenic progenitors and principally binds to collagens type I, II, III and X (43, 44). After being activated by collagen ligands, the downstream signals propagated by DDR2 stimulated the ERK1/2 and p38 MAP kinase pathways to increase osteoblast differentiation (44, 45). In our study, DDR2 was mostly expressed in BMMSC1, and COL1A1 was highly expressed in MatOB (Fig. 4C). Therefore, COL1A1 specifically expressed by mature osteoblasts could mediate the differentiation of BMMSCs towards osteogenic direction.
In order to identify significant genes that play a critical important role in mature osteoblasts and BMMSCs interactions. We mapped top 30 genes of the two clusters to a protein-protein interaction network based on the STRING dataset, and then used Cytoscape (16) to identify biologically meaningful genes and display their relationships. We found some interesting protein-protein interactions in mature osteoblasts (MatOB) and BMMSCs (BMMSC1). A total of 41 nodes and 120 edges were constructed in the PPI network (Supplementary Fig. S4A and S4B). This included a ribosomal protein (RP) family network that played an important role in protein biosynthesis (Supplementary Fig. S4B). Two sub-networks were identified by using MCODE (Fig. 5A and 5B).
FZD1 was a transmembrane receptor that could bind to Wnt ligand and further mediate Wnt canonical and non-canonical pathways (46). As a member of FZD protein family, the specific mechanism of how FZD1 regulated cell differentiation had not yet been elucidated. Some studies showed that transcription factor activating protein 2 (
BMMSCs are the major source of osteoblasts and adipocytes in bone (56). Various factors participate in the process of osteogenesis via complex biochemical reactions and cellular communication. Our study integrated scRNA-seq transcriptomes of BMMSCs and osteoblasts in humans, and provided important insights into the differentiation fate of BMMSCs and osteoblasts. In addition, we also analyzed the interaction between BMMSCs and osteoblasts in terms of the ligand receptor pairs.
Previous studies have shown that during osteogenic development at least three stages were recognized: preosteoblasts, immature osteoblasts and mature osteoblasts (34, 57). Some
We described two subclusters of osteoblasts (PreOB2 and PreOB3), both of which expressed the marker genes of preosteoblast, such as
Each cell in a multicellular organism was not independent, but connected and communicated with other cells in a variety of ways. BMMSCs and osteoblasts play a crucial role in bone metabolism, and they communicate with others through some of cell signal molecules. Under the condition of co-culture with osteoblasts, it was found that the expression levels of
There were two limitations in the present study. At first, all cells were collected from diseased individuals. Comparing with healthy individuals, the types or proportions of osteoblast subclusters we got from the data might have some deviations. We don’t know if the conclusion (feedback mechanism between WIF1/SFRP4 and FZD1/Wnt) derived from the data of the patient’s osteoblast and BMMSCs was applied to osteoblast and BMMSCs from a normal person. Therefore, normal subjects are needed in future studies to reveal osteoblast and BMMSCs subclusters and the molecular mechanism between WIF1/SFRP4 and FZD1/Wnt. On the other hand, age was the main factor affecting the transcription pattern of BMMSCs (7). In this study, the subjects of BMMSCs consisted of a 67-year-old postmenopausal female with osteoporosis and an 85-year-old male with osteoarthritis. The osteoblasts were collected from the femur head of one 31-year-old male patient with osteoarthritis and osteopenia. Although there were certain limitations, we attempted to reveal a continuum of the dynamic gene expression pattern of BMMSCs to osteoblasts, and our dataset and analysis will serve as a resource for future studies investigating osteogenesis differentiation.
In summary, we have sorted out the development trajectory from BMMSCs to mature osteoblasts
This research was benefited by grants from the National Institutes of Health (R01AR069055, U19AG055373, P20GM109036, R01AG061917), National Natural Science Foundation of China (NSFC) (Grant No. 81902277 and 81570807), National Key R&D Program of China (Grant No. 2017YFC1001100), Natural Science Foundation of Hunan Province (S2019JJQNJJ2093), Changsha Science and technology project (kq1907153), Central South University (Grant Nos. 164990007, 2018zzts886), and Xiangya Clinical Big Data Project (xyyydsj9).
The study was approved by the Medical Ethics Committee of Xiangya Hospital of Central South University, and the IRB approval number is No. 201912315.
All authors have no conflicts of interest to declare.
HWD and LJT conceived the study and designed the experiments. YG provided technical support. XHL, HXZ, CZ, YC and YL performed most of the experiments. YL and YC analyzed the data, wrote the manuscript and designed the figures. LJT, HS, HMX and HWD corrected the language, managed editing of the manuscript, supervised writing and approved the final draft. All authors have read and agreed to the published version of the manuscript.
The scRNA-seq datasets of human BMMSCs and human osteoblasts can be accessed from Gene Expression Omnibus (GEO) database under the accession numbers of GSE147287 and GSE147390, respectively.
Supplementary data including one table and four figures can be found with this article online at https://doi.org/10.15283/ijsc22101.
CrossRef (6) |