www.fgks.org   »   [go: up one dir, main page]

IMR Press / FBL / Volume 29 / Issue 5 / DOI: 10.31083/j.fbl2905173
Open Access Original Research
Single-Cell Transcriptome Analysis Reveals Dynamic Populations of Vascular Cells in Neointimal Hyperplasia
Show Less
1 The Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
2 Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences (CAS), 200031 Shanghai, China
*Correspondence: liqun@sibs.ac.cn (Qun Li)
Front. Biosci. (Landmark Ed) 2024, 29(5), 173; https://doi.org/10.31083/j.fbl2905173
Submitted: 29 December 2023 | Revised: 18 February 2024 | Accepted: 13 March 2024 | Published: 6 May 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Neointimal hyperplasia (NIH) is the pathological basis of vascular injury disease. Vascular cells are the dominant cells in the process of NIH, but the extent of heterogeneity amongst them is still unclear. Methods: A mouse model of NIH was constructed by inducing carotid artery ligation. Single-cell sequencing was then used to analyze the transcriptional profile of vascular cells. Cluster features were determined by functional enrichment analysis, gene set scoring, pseudo-time analysis, and cell-cell communication analysis. Additionally, immunofluorescence staining was conducted on vascular tissues from fibroblast lineage-traced (PdgfraDreER-tdTomato) mice to validate the presence of Pecam1+Pdgfra+tdTomato+ cells. Results: The left carotid arteries (ligation) were compared to right carotid arteries (sham) from ligation-induced NIH C57BL/6 mice. Integrative analyses revealed a high level of heterogeneity amongst vascular cells, including fourteen clusters and seven cell types. We focused on three dominant cell types: endothelial cells (ECs), vascular smooth muscle cells (vSMCs), and fibroblasts. The major findings were: (1) four subpopulations of ECs, including ECs4, mesenchymal-like ECs (ECs1 and ECs2), and fibro-like ECs (ECs3); (2) four subpopulations of fibroblasts, including pro-inflammatory Fibs-1, Sca1+ Fibs-2, collagen-producing Fibs-3, and mesenchymal-like Fibs-4; (3) four subpopulations of vSMCs, including vSMCs-1, vSMCs-2, vSMCs-3, and vSMCs-3-derived vSMCs; (4) ECs3 express genes related to extracellular matrix (ECM) remodeling and cell migration, and fibro-like vSMCs showed strong chemokine secretion and relatively high levels of proteases; (5) fibro-like vSMCs that secrete Vegfa interact with ECs mainly through vascular endothelial growth factor receptor 2 (Vegfr2). Conclusions: This study presents the dynamic cellular landscape within NIH arteries and reveals potential relationships between several clusters, with a specific focus on ECs3 and fibro-like vSMCs. These two subpopulations may represent potential target cells for the treatment of NIH.

Keywords
neointimal hyperplasia
fibroblasts
endothelial cells
vascular smooth muscle cells
single-cell RNA sequencing
1. Introduction

Neointimal hyperplasia (NIH) develops after vascular damage caused by surgical operations such as balloon or stent angioplasty [1]. The formation of NIH is mediated by both cellular and humoral factors. Previous findings highlighted the role of four major cell types and associated processes in NIH: endothelial cells (ECs), adventitial fibroblasts (Fibs), vascular smooth muscle cells (vSMCs), and monocytes and/or macrophages [1]. Specific changes in these cell types during NIH include endothelial activation, monocyte accumulation, fibroblast migration, and vSMC phenotype switching [1]. However, clinical prevention and therapy of NIH remain unsatisfactory, possibly due to vascular cell heterogeneity.

There is currently only a limited understanding of the heterogeneity of different vascular cells and the roles of various subpopulations in neointimal vessels. Single-cell RNA sequencing (scRNA-seq) is a tool used to study cellular heterogeneity and to identify rare cell subtypes [2]. Recent studies have utilized scRNA-seq to profile the cellular landscape of aortic aneurysms [3], atherosclerosis [4], and endothelial denudation [5]. However, there have been no reports on the use of scRNA-seq to study NIH induced by carotid ligation in mice.

Epithelial-to-mesenchymal transition (EMT) refers to the process by which epithelial cells lose their adhesion connections and polarization, and subsequently transform into mesenchymal cells [6]. This phenomenon has been observed in blood vessels with disrupted blood flow, but has not been reported in the context of NIH [7]. Adventitial fibroblasts contribute to NIH mainly by differentiating into myofibroblasts [8], and the conversion of human fibroblasts into ECs has been reported previously [9].

In response to various stimuli, vSMCs can undergo several phenotypic modulations including de-differentiation, proliferation, and migration. This involves downregulation of the expression of vSMC marker genes such as Myh11 and Acta2, and upregulation of Ly6a expression [10, 11]. During vascular injury, vSMCs acquire characteristics of macrophages through dedifferentiation [12]. In atherosclerosis, vSMCs dedifferentiate into cells with fibrotic features, thereby maintaining stability of the fibrous cap [13]. However, this change in vSMCs has not yet been studied in NIH.

To better understand the underlying mechanism of NIH, the aim of this study was to examine the transcriptional landscape of vascular cells by using single-cell RNA sequencing coupled with contemporary analytical methods.

2. Materials and Methods
2.1 Animals

Eight-week-old C57BL/6 male mice were procured from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). The PdgfraDreER mouse line was described previously [14]. PdgfraDreER-tdTomato male mice were generated by crossing the PdgfraDreER and R26R-rox-tdTomato reporter lines. All mice were housed in specific pathogen-free conditions in an animal room with a 12/12-hour day/night cycle and with ample access to water and food. Animal experiments in this study were conducted in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals. Approval was obtained from the Ethics Committee on Animal Experiments, Shanghai Jiao Tong University School of Medicine (SYXK-2003-0026).

2.2 Carotid Artery Ligation Injury Model

To induce vascular injury, mice underwent carotid artery ligation as previously described [15]. In brief, mice were anesthetized using isoflurane, and the left carotid artery was completely ligated with 3-0 absorbable suture just proximal to the carotid bifurcation.

2.3 Immunofluorescence Staining

Tamoxifen was administered to PdgfraDreER-tdTomato mice at 5 weeks of age for 5 consecutive days, followed by a 2-week washout period and subsequent carotid ligation. The carotid arteries were then harvested, rinsed with PBS, fixed in 4% paraformaldehyde overnight, embedded in paraffin, and then sliced into 5-µm thick sections. Immunofluorescence staining was conducted as described previously [15]. The primary antibodies used were Pecam1 antibody (Santa Cruze Biotechnology, Dallas, TX, USA, sc-1506-R, 1:100 dilution), Red Fluorescent Proteins (RFP) antibody (Proteintech, Wuhan, China, 5F8, 1:200 dilution), and Pdgfra antibody (R&D, Minneapolis, MN, USA, AF1062, 10 µg/mL). The Alexa fluor-conjugated secondary antibodies employed were donkey anti-rabbit IgG (Invitrogen, Carlsbad, CA, USA, A21202 for Alexa Fluor 488), goat anti-rat IgG (Invitrogen, A11006 for Alexa Fluor 594), and donkey anti-goat IgG (Invitrogen, A21447 for Alexa Fluor 647). Images were obtained using a Zeiss fluorescence microscope (Axio Imager M2, Zeiss, Göettingen, Germany).

2.4 Single-Cell RNA Sequencing and Data Analysis

Carotid arteries from six C57BL/6 mice were harvested at 2-weeks post-ligation (Supplementary Fig. 1A). The vessels were enzymatically digested to obtain a single-cell suspension. scRNA-seq libraries were then constructed using the 10x Genomics platform. Raw, single-cell RNA-sequencing data was processed using Cell Ranger software (version 6.0, https://github.com/10XGenomics/cellranger). Briefly, the “Cellranger mkfastq” command was employed to de-multiplex raw data and generate FASTQ files. These files were further processed by “Cellranger count” to align reads to the mouse reference, count the number of barcodes and unique molecular identifiers (UMIs), and generate feature-barcode matrices. The raw gene-expression matrix was converted into a Seurat object using the Seurat package (v4.0.5) of R (v4.1.1, University of Auckland, Auckland, New Zealand) [16]. Harmony was utilized to mitigate batch effects across different groups (Supplementary Fig. 1B) [17]. For quality control, only gene features expressed in at least three cells, and cells with at least 200 detected genes, were retained. Subsequently, cells displaying <200 gene features or having >15% mitochondrial counts were filtered out. Doublet cells were identified and excluded from the remaining cells using the R package DoubletFinder (v2.0.3) [18]. A total of 17,605 cells remained after filtering. Data normalization was achieved through “LogNormalize”. The top 2000 highly variable genes were selected and scaled using the “ScaleData” function. Principle Component Analysis (PCA) was performed on these selected, highly variable genes. The first 15 principal components with a resolution of 0.4 were employed for cell clustering and visualization via t-distributed stochastic neighbor embedding (t-SNE), or uniform manifold approximation and projection (UMAP). Marker genes upregulated in each cluster were identified through “FindAllMarkers” (min. pct = 0.25, logfc. threshold = 0.25). For sub-clustering of the major cell populations, the aforementioned procedure was repeated. To ascertain similarities between different clusters, correlation matrix heatmaps for all clusters were generated by Scpub [19].

2.5 Functional Enrichment Analysis

Genes analyzed by Metascape (https://metascape.org/) were obtained by the “FindAllMarkers” method and filtered using the criteria: log2FC >0.25 and p < 0.05 (Supplementary Material) [20]. To investigate whether vSMCs-3 dedifferentiate into fibro-like vSMCs, or ECs-4 to ECs-2, related genes from different modules calculated by monocle were subjected to Gene Ontology (GO) function-annotation analysis [21].

2.6 SCENIC

The pySCENIC protocol (version 0.11.0, https://github.com/aertslab/pySCENIC) was used as part of a Python package set to default configurations in order to deduce the activity of transcription factors (TFs) and to identify their associated target genes within all sampled cells [22]. This process was segmented into three primary phases. Initially, we refined the single-cell gene expression dataset by removing genes that appeared in <10 cells. The remainder were kept for the construction of co-expression modules with the GRNBoost2 algorithm, which operates on a regression per-target basis. Subsequently, these modules were refined by eliminating indirect targets via the implementation of cis-regulatory motif analysis (cisTarget). Finally, the operational status of these regulons was determined by using the AUCell method (version 3.18, https://bioconductor.org/packages/release/bioc/html/AUCell.html) to calculate an enrichment score for the genes targeted by each regulon.

2.7 Scoring According to a Specific Gene Set

The “AddModuleScore” function built in the Seurat package was employed to quantify the activity of a specific gene set across different clusters.

2.8 Trajectory Inference

Monocle3 (version 1.0.0, https://cole-trapnell-lab.github.io/monocle3/) was used to deduce potential differentiation pathways among various EC subtypes, without any prior assumptions about the sequence or orientation of differentiation [23]. For the analysis of RNA velocity within the subsets of fibroblasts and vSMCs-3, we applied the scvelo tool (version 0.2.4, https://github.com/theislab/scvelo) as recommended by its creators [24]. This allowed investigation of the developmental trajectories from vSMCs-3 to fibro-like vSMCs using the trajectory analysis capabilities of the Monocle algorithm [25].

2.9 Analysis of Cell-Cell Communication

NicheNet (version 2.0.4, https://github.com/saeyslab/nichenetr) was used for the identification of potential ligands secreted by fibro-like vSMCs. This is an advanced approach designed to predict ligands that may cause changes in the target cell transcriptome [26]. Our analysis also incorporated CellChat (version 1.5.0, https://github.com/sqjin/CellChat) by using its comprehensive database, CellChatDB.mouse. This includes data on 1211 secreted signaling pairs, 432 extracellular matrix (ECM) receptor pairs, 378 cell-cell contact pairs, 229 families of signaling pathways (e.g., WNT, transforming growth factor-β (TGFβ), bone morphogenetic protein (BMP), epidermal growth factor (EGF), TGFα, fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), vascular endothelial growth factors (VEGF), insulin-like growth factor (IGF)), and chemokine and cytokine signaling pathways (chemoattractant cytokine ligand (CCL), C-X-C motif chemokine ligand (CXCL), interleukins (IL), interferon (IFN)). CellChat assigns a probability of communication between ligand-receptor pairs based on the principle of mass action. It considers the average expression level of ligands in one cell population, and of receptors in another. The significance of these communication probabilities was determined using permutation tests, with p-values < 0.05 deemed significant. This analytical framework facilitates the assessment of interaction strengths between various cell clusters identified from the scRNA-seq data, with the results represented graphically as either circle or dot plots.

3. Results
3.1 Heterogeneity of Vascular Cells in Arteries with Ligation-Induced NIH

To create a detailed cellular atlas of NIH at the single-cell level, we performed single-cell sequencing for both self-control and ligation-injured carotid arteries from mice. After quality control and the removal of batch effects between the two groups, 17,605 cells were further analyzed. Unsupervised clustering algorithm yielded 14 distinct clusters, including seven major cell types, and dispersion into two groups (Fig. 1A, Supplementary Fig. 1B). The 14 cell clusters were comprised of 7 cell types annotated with canonical markers: fibroblasts (3 clusters; Pdgfra, Pi16), vSMCs (4 clusters; Myh11, Tagln, Myl9, Acta2), ECs (Pecam1, Cdh5), lymphatic ECs (Lyve1, Pecam1), T cells (2 clusters; Ptprc, Cd3d, Cd3e), monocytes/macrophages (2 clusters; Lyz2, S100a8), and Schwann cells (Mbp, Sox10) (Fig. 1B). The signature genes of all clusters are shown in Fig. 1C.

Fig. 1.

The lineage and characteristics of all cells in healthy (Sham) and ligated (Ligation) carotid artery. (A) Annotated UMAP of mouse carotid artery single-cell RNA sequencing (scRNA-Seq). (B) Relative expression of marker genes in all cells from all samples projected onto a Dotplot. (C) Heatmap showing the mean expression of highest specificity gene markers of each clusters. Color represents average normalized expression level within the denoted cluster. (D) The correlation between different clusters. vSMCs, vascular Smooth Muscle Cells; ECs, Endothelial Cells; LECs, Lymphatic Endothelial Cells; SWs, Schwann Cells; UMAP, Uniform Manifold Approximation and Projection.

Tissue inflammation occurs in response to vascular injury and involves the infiltration and activation of immune cells [27]. Two subgroups of macrophages and two subgroups of T cells are present in the carotid artery, and these cells are mainly found in NIH vessels. The elevated transcription levels of Foxp3 and Satb1 in TCs1 indicate this subpopulation possessed Treg (regulatory T cell) properties (Fig. 1C, Supplementary Fig. 1C) [28]. TCs2 exhibit proliferative features due to high transcription levels of the MKI67 and TOP2A genes [29]. The increased number of lymphatic endothelial cells (LECs) suggests that lymphangiogenesis occurs in NIH induced by vascular ligation. SWs are found in both healthy and diseased carotid arteries, which may be attributed to nerve fibers on the vascular wall. Additionally, subpopulations with different annotations but belonging to the same cell type showed high correlation (Fig. 1D).

These results demonstrate the heterogeneity of fibroblasts, ECs and vSMCs, which are the predominant cell populations within the artery. Our study therefore focused on these three cell types to gain a better understanding of their role in NIH.

3.2 Features of Different Endothelial Subtypes in NIH

To further characterize the heterogeneity of ECs in NIH, we performed a re-clustering of the EC cohort (C8). This identified four distinct subpopulations (ECs1, ECs2, ECs3, and ECs4) based on their transcriptomic profiles. Interestingly, the ECs4 subpopulation was found primarily in the sham group, whereas the frequency of cells from the other subpopulations was notably higher in the ligation group (Fig. 2A). A novel finding was the exclusive presence of ECs3 in the ligation group, characterized by the expression of genes typically associated with vascular fibroblasts, including Dcn and Col3a1 (Supplementary Fig. 2A). These cells express both Pdgfra and Pecam1 and are termed fibro-like ECs (Fig. 2B). Comparative analysis revealed upregulation of mesenchymal markers (Prss23, Cd34, and Ly6a) in ECs1 and ECs2 relative to ECs4. This suggests that ECs1 and ECs2 have a mesenchymal-like phenotype, in contrast to the more endothelial-like phenotype of ECs4 (Fig. 2C).

Fig. 2.

Description of phenotype and function of ECs in NIH. (A) Re-clustering of ECs identified in carotid tissue before and after acute injury. (B) Gene expression of Pdgfra and Pecam1 for each clusters. (C) Violin plots on the expression of selected mesenchymal markers of ECs1, ECs2 and ECs4. (D) UMAP visualization showing the pseudo-time trajectory of the ECs1, ECs2 and ECs4. (E) Heatmap hierarchical clustering showing differentially expressed genes along with the pseudotime curve (left). Gene Ontology biological processes of each DEGs clusters (right). Color key from blue to red indicates relative expression levels from low to high. (F) Top-ranked enrichment analysis of feature genes of ECs3 on Metascape platform. Bars are discretely colored to encode p-values of increasing statistical significance. (G) The immunofluorescent staining of Pecam1 (green), tdTomato (red) and Pdgfra (purple, autofluorescence) in carotid artery from PdgfraDreER-tdTomato mice. Nuclei were counterstained with 4,6-diamidino-2-phenylindole (DAPI) (blue). Scale bar = 50 µm (n = 3). A, adventitia; M, media; N, neointimal hyperplasia; NIH, Neointimal Hyperplasia; DEGs, Differentially Expressed Genes; IGF, insulin like growth factor; ECM, extracellular matrix; MAPK, mitogen-activated protein kinase.

Further exploration using the Monocle3 R package revealed a positional dichotomy between ECs2 and ECs4. ECs2 was centrally located, indicating a potential differentiation pathway from ECs4 to ECs1 and ECs2, but not to ECs3 (Fig. 2D). Analysis of an RNA-seq dataset (GSE220512) revealed upregulation of feature genes for ECs1 and ECs2 in injured blood vessels at days 7 and 14 after vascular injury (Supplementary Fig. 2B). Dynamic expression of Teme252, Flt1, Nrp2 and Lrg1 showed an increasing trend along the pseudo-time trajectory (Supplementary Fig. 2C). Our analysis identified 5705 genes displaying four distinct expression patterns across the trajectory. Functional enrichment analysis found that genes associated with cell-substrate adhesion were highly expressed in ECs4, but were downregulated upon transition to ECs1 and ECs2. Conversely, genes implicated in cell proliferation and activation of the inflammasome complex were significantly upregulated in the later stages of pseudo-time (Fig. 2E).

Pathway analysis of ECs3 signature genes using Metascape provided insights into their biological functions, including ECM organization, elastic fiber formation, and cell motility. This suggests that ECs3 have ECM remodeling and cell migration activities in NIH (Fig. 2F). Previous studies have reported trans-differentiation of human fibroblasts into ECs [30]. To investigate whether ECs3 represents a novel subpopulation derived from Pdgfra+vascular fibroblasts, we utilized PdgfraDreER-tdTomato mice to trace fibroblast lineage cells within the carotid artery (Supplementary Fig. 3A) [31]. Tamoxifen-induced Dre-rox recombination facilitated effective tdTomato labeling of vascular fibroblasts (Supplementary Fig. 3B). This was followed by vessel ligation to induce NIH in these mice (Supplementary Fig. 3C). Of note, intense tdTomato fluorescence within the neointima signified fibroblast migration from the vascular adventitia to the intima (Fig. 2G), with a subset of tdTomato+ cells co-expressing Pecam1 and Pdgfra (Fig. 2G). Negative control observations are documented in Supplementary Fig. 3D.

Together, these findings revealed two subsets of ECs in carotid artery with NIH. These showed mesenchymal characteristics, markers for proliferation and the inflammasome complex, and existed alongside a subpopulation of fibro-like ECs.

3.3 Identification of a Novel Fibro-Like vSMC Subtype in NIH

Re-clustering of the fibroblasts and vSMCs shown in Fig. 1A revealed four subpopulations of fibroblasts (Fibs-1, Fibs-2, Fibs-3 and Fibs-4), three subpopulations of vSMCs (vSMCs-1, vSMCs-2 and vSMCs-3) and a special subpopulation named fibro-like vSMCs [32]. These were annotated according to their expression of Pdgfra and Myh11 (Fig. 3A,B), and by the top 10 defining genes for each cluster (Supplementary Fig. 4A). vSMCs-2 were mainly distributed in injured artery and expressed osteoblast differentiation-associated genes, including Mef2c, Tnc, Junb, and Tpm4, indicating chondrogenic features (Supplementary Fig. 4B).

Fig. 3.

Functional analysis of different subtypes of fibroblasts. (A) A t-SNE plot of all fibroblasts and vSMCs colored according to cluster (left) and group (right). (B) Relative expression of Myh11 and Pdgfra in fibroblasts and vSMCs projected onto a t-SNE plot. (C) Chemokines, Collagens and Proteinase score calculated by the AddModuleScore function. (D) Heatmap showing chemokine genes, collagen genes, and proteinase genes in eight clusters. (E) The cell‒cell junction scores between any two clusters (left) and cell matrix scores of each clusters (right).

We then analyzed the expression level of all chemokines, collagens and proteinase in all clusters. Of note, Fibs-1 and fibro-like vSMCs exhibited elevated levels of inflammatory chemokines (Cxcl14, Ccl11, Cxcl9, Ccl7 for Fibs-1 and Ccl4, Ccl22, Ccl6, Ccl25), while the latter cells also showed moderate collagen expression and elevated proteinase activity (Fig. 3C,D). Moreover, vSMCs-1 exhibited the highest scores in cell-cell and cell-ECM junctions, whereas fibro-like vSMCs had the lowest score for cell-ECM junctions, hinting at their migratory capability (Fig. 3E).

Fibs-1 emerge as pro-inflammatory, Fibs-3 as collagen-producing, and Fibs-4 as indicative of fibroblasts in homeostasis. This raises the question of the role of Fibs-2 in NIH. By conducting monocle analysis on Fibs-1, Fibs-2, Fibs-3 and Fibs-4, it was discovered that Fibs-3 likely originates from both Fibs-2 and Fibs-4, with Fibs-1 being derived from Fibs-2 (Supplementary Fig. 5A). We also examined the transcriptional level of stemness genes in different fibroblast subpopulations. Significant expression of the mesenchymal marker Gli1 was observed in Fibs-4, indicating this subpopulation was MSC-like fibroblasts (Supplementary Fig. 5B). The highest level of Sca1 transcription was seen in Fibs-2 (Supplementary Fig. 5B). The CytoTRACE algorithm revealed that Fibs-2 and Fibs-4 cells exhibited the highest level of stemness, followed by Fibs-1, and with Fibs-3 showing the least stemness (Supplementary Fig. 5C). The integration of monocle2 and CytoTRACE helps to differentiate the various fibroblast subpopulations (Supplementary Fig. 5D).

The above findings highlight the cellular heterogeneity present in NIH. The results identify fibro-like vSMCs as key players due to their robust chemokine secretion, moderate ECM production, relatively elevated proteinase levels, and diminished adhesion, thereby making them potential targets for NIH therapy. In addition, these results suggest that Fibs-2 are a minor, yet critical cell population within healthy blood vessels, and are capable of proliferation and differentiation into pro-inflammatory fibroblasts following vascular injury.

3.4 Fibro-Like vSMCs Originate from vSMCs-3 in NIH

Previous scRNA-seq analyses conducted on atherosclerotic lesions from murine and human arteries have shown that vSMCs tend to shift toward genuine fibroblasts, while remaining distinct from these [13]. We hypothesized that this transdifferentiated phenotype also exists in NIH. The transcriptional kinetics from vSMCs-3 to fibro-like vSMCs was revealed by RNA velocity, which is a high-dimensional vector that predicts the future state of individual cells over several hours (Fig. 4A) [33]. The R package Monocle was used to construct the comprehensive lineage differentiation trajectory for vSMCs-3 to fibro-like vSMCs, highlighting the overlap between the two clusters on this trajectory (Fig. 4B). Subsequent analysis of gene expression patterns during the cell state transitions along this trajectory identified 6705 dynamically expressed genes, categorizing them into four distinct patterns. Functional enrichment analysis was carried out on three of these patterns (Fig. 4C). Canonical vSMC genes, such as Myh11 and Cnn1, were down-regulated, whereas fibroblast-specific genes such as Pdgfra and Dcn were up-regulated (Fig. 4D). Furthermore, SCENIC analysis found that Dlx5, Ets2, Rxrb, Nfya and Batf were the top five most active regulons in individual cells (Fig. 4E,F).

Fig. 4.

The transdifferentiation of vSMCs-3 into fibro-like vSMCs. (A) RNA velocity overlaid on tSNE of the extracted vSMC and fibroblast subset. (B) The developmental trajectory of selected clusters, coloured-coded by the groups (upper) and associated cell subpopulations (lower). (C) Heatmap hierarchical clustering showing differentially expressed genes along with the pseudotime curve (left). Gene Ontology biological processes of each DEGs clusters (right). Color key from green to purple indicates relative expression levels from low to high. (D) Vlnplot showing the expression levels of Myh11, Cnn1, Pdgfra and Dcn in fibro-like vSMCs and vSMCs-3. (E) The regulatory activity of all clusters. (F) Scatter plot showing the specificity scores of regulons of fibro-like vSMCs. The top 6 regulons are highlighted.

Taken together, the above results suggest that the key pathogenic cell subpopulation, fibro-like vSMCs, may be derived from vSMCs-3.

3.5 Fibro-Like vSMCs are the Primary Cells Responsible for Secreting Vegfa

CellChat was employed to build a cell–cell-communication network based on known ligand-receptor pairs and their cofactors [34]. Fibro-like vSMCs had distinct effects on different cell types, and interact mainly with ECs (Fig. 5A,B). NicheNet analysis [26] revealed that Vegfa was secreted predominantly by fibro-like vSMCs (Fig. 5C) and may drive biological functions in other cell types. In NIH, Vegfa influences ECs via Vegfr2 (Fig. 5D). Additionally, Vegfa expression was significantly higher in wire-induced or ligation-induced vessels, as shown by the RNA-seq datasets GSE70410 and GSE220512 (Fig. 5E).

Fig. 5.

Cell–cell communications of fibro-like vSMCs with other cells. (A) Arrow plot showing the interaction strength between fibro-like vSMCs with other cell populations, based on the interaction scores. (B) Heatmap showing the number of interactions between different structual cells in vessel. Blue block indicate that the displayed communication is decreased in ligated carotid artery (CA), whereas red cblock indicate that the displayed communication is increased in ligated CA compared with healthy carotid artery. The darker the color, the greater the communication probability between the two cell type. (C) Genes are ligand that predicted by Nichenet, secreted by fibro-like vSMCs, ranked by prioritized liand activity. The darker the color, the higher the ranking. Dot plot showing average expression of ligand in all clusters. (D) Dot plot showing vascular endothelial growth factor (VEGF) related chemokine-receptor interaction pairs between fibro-like vSMCs and three ECs subsets or LECs, based on the communication probability. (E) Barplots showing expression level of Vegfa in different group of different database. **, p 0.05; *, p 0.05; ns = not significant. Student’s t test (left) and paired Student’s t test (right).

4. Discussion

This study established a mouse model of NIH that was induced by carotid artery ligation. We subsequently performed scRNA sequencing on ligated and self-control arteries. This analysis revealed that the NIH microenvironment is more heterogeneous and complex than previously reported [35]. Briefly, we analyzed 17,605 cells and identified 7 different cell types. These were comprised of subpopulations, thereby demonstrating their heterogeneity. The gene signatures of several of these cell subpopulations, not previously documented in earlier studies, were thoroughly elucidated.

Immune cell infiltration into vessels occurs post-injury. Our scRNA-seq data identified two distinct clusters of macrophages and T cells found predominantly in diseased vessels. Specifically, TCs1 showed significant expression of Foxp3, which is the hallmark gene of Tregs. Previous research has suggested that Tregs do not significantly affect the progression of NIH [36]. TCs2 showed increased expression of genes associated with proliferation. However, the exact role of TCs2 in NIH remains to be elucidated, highlighting the need for further investigation. SWs were present in both healthy and diseased vessels, possibly due to the presence of pre-vascular nerve. Additionally, a distinct population of LECs was identified in vessels with NIH. Previous studies have demonstrated that fibroblast-derived Vegfc promotes lymphangiogenesis in animal models of NIH induced by vascular transplantation. Moreover, the inhibition of lymphatic vessel formation has been shown to reduce neointimal thickness [37]. This suggests that lymphangiogenesis may play a significant role in NIH following ligation, warranting further investigation to elucidate its impact.

ECs have the capacity to regulate the growth and regression of vSMCs through direct interaction or via the release of mediators. Additionally, ECs regulate the thickness of the ECM by secreting enzymes or their inhibitors [38]. In the present study, a subpopulation of ECs expressing Pdgfra (fibroblast marker) and Pecam1 (endothelial cell marker) was found to play a role in ECM remodeling and cell migration in NIH. Additionally, cells co-expressing Pdgfra and Pecam1 were found in the neointima of PdgfraDreER-tdTomato mice by immunofluorescence, leading us to hypothesize that these cells originate from adventitial fibroblasts. We plan to investigate the specific role and mechanism of Pdgfra and Pecam1 co-expressing cells in NIH in future studies. Additionally, we identified two subsets of ECs with mesenchymal features in carotid arteries with ligation-induced NIH. These ECs expressed genes involved in cell proliferation and in the inflammasome complex.

A previous study reported the presence of mesenchymal-like ECs in mouse carotid artery subjected to disturbed flow [7]. In the current study, we evaluated the expression of the top 10 genes in mesenchymal-like ECs from the mouse carotid artery under disturbed flow conditions. This was carried out for three EC subsets: ECs1, ECs2, and ECs4. Our analysis revealed shared gene expression (Kit, Vcan, and Lamb1) between the two animal models, as well as genes with differential expression (Dkk2, Ngf, Col8a1, Ltbp2, Cdca1l, Dclkl, and Slc45a4) (Supplementary Fig. 6A). This suggests that ECs with mesenchymal characteristics and that arise from the two experimental methods possess distinct gene expression profiles. Furthermore, mesenchymal-like ECs under disturbed flow were characterized by the production of TGF-β, a trait observed in both ECs1 and ECs2, indicating commonality in cellular function (Supplementary Fig. 6A).

Four distinct groups of vSMCs and fibroblasts were identified. vSMCs-2 exhibited elevated expression of genes associated with osteoblast differentiation, suggesting a propensity towards an osteochondrogenic-like phenotype. This observation is significant given that calcification and bone formation are commonly observed in advanced stages of NIH patients, and implicating vSMCs-2 as a potential contributory cell type to this pathology that warrants further experimental validation. Two fibroblast subpopulations, Sca1+Fibs-2 and Gli1+Fibs-4, showed the highest stemness scores, while Fibs-3 exhibited the lowest. Fibs-3 emerged as the primary cell type for collagen production and was derived from Fibs-2 and Fibs-4. Pro-inflammatory Fibs-1 was identified as the intermediary cell during this differentiation process. Consequently, inhibiting the differentiation of Fibs-2 or Fibs-4 into other subpopulations may represent a promising therapeutic strategy.

RNA velocity and Monocle analyses showed that vSMCs-3 transitioned toward fibro-like vSMCs in NIH, accompanied by a reduction in the expression of vSMCs markers. Fibro-like vSMCs have the ability to release inflammatory chemokines, which may contribute to the accumulation of immune cells and the subsequent reduction in lumen area [39]. Moreover, fibro-like vSMCs showed minimal collagen release, but displayed high levels of protease expression and the lowest scores for cell-ECM junctions. This implies that fibro-like vSMCs are more likely to undergo cell migration [40]. In atherosclerotic plaques, vSMCs undergo dedifferentiation to a fibroblast-like phenotype. These were termed “fibromyocytes” by the authors, and are thought to play a critical role in stabilizing the plaque [13]. Fibromyocytes are a protective cell type with distinct functions compared to the fibro-like vSMCs identified in NIH. Tcf21 was identified as a key transcription factor for fibromyocytes. Fibro-like vSMCs in NIH have a relatively higher expression of Tcf21 compared to other vSMC subpopulations (Supplementary Fig. 6B). Therefore, the dedifferentiation of vSMCs-3 into fibro-like vSMCs in NIH may also be regulated by Tcf21.

Cell-communication analysis revealed that vascular injury shaped different signal-communication patterns. NicheNet analysis was used to predict ligands secreted by fibro-like vSMCs, with Vegfa notably among the top 10 active ligands. Upregulation of Vegfa has been observed consistently across different datasets, time points, and animal models of NIH. Cellchat analysis confirmed that fibro-like vSMCs are able to secrete Vegfa, which subsequently acts on different subpopulations of ECs and vascular lymphatic ECs. Previous research has indicated that inhibition of lentivirus-mediated Vegfa expression in injured vessels hinders neointimal formation [41]. Fibro-like vSMCs may therefore play a key role in disease progression and serve as a new target for therapy.

The present study has several limitations. First, although the mouse model of carotid artery ligation injury is widely used to study NIH, there is nevertheless a translational gap with human conditions. Second, the procurement of vessels with NIH from humans is challenging, and significant differences exist between mice and humans. Third, the number of cells per cluster that were analyzed was limited. Fourth, scRNA-seq provides RNA-level information only, and inferred cellular functions (e.g., cell migration) based solely on gene signature may not be accurate. Fifth, the results of the analyses may be influenced by changes in the packages and in the parameter settings. Sixth, it is difficult to accurately distinguish between fibroblasts and mesenchymal stem cells using scRNA-seq. Finally, this study lacks in vivo animal experiments, particularly those involving lineage tracing of vSMCs. We plan to conduct further research to address this latter point.

5. Conclusions

Our study investigated the heterogeneity of vascular cells in NIH. We identified a total of four subpopulations each of ECs, fibroblasts, and vSMCs. These cell clusters were characterized at the mRNA level, with a particular focus on two notable clusters: ECs3 and fibroblast-like vSMC. ECs3 is derived from fibroblasts and is involved in ECM remodeling. The fibro-like vSMC is derived from vSMCs-3 and is characterized by the secretion of chemokines and Vegfa. These findings highlight the potential of ECs3 and fibro-like vSMC as targets for NIH treatment.

Availability of Data and Materials

The data and R scripts related to the findings of this study are available upon reasonable request. The scRNA-seq and bulk RNA-seq data of this study are available at [GSE244246, GSE70410 and GSE220512, https://www.ncbi.nlm.nih.gov/geo/].

Author Contributions

GZS and QL conceived and designed the research; GZS, WHS and XRT performed the research and acquired the data; GZS, XRT, PLZ, WDC, GHJ and ZLF analyzed the data; GZS and QL wrote the paper; QL funded the research. All authors have participated sufficiently in the work to take public responsibility for appropriate portions of the content and agreed to be accountable for all aspects of the work in ensuring that questions related to its accuracy or integrity. All authors read and approved the final manuscript. All authors contributed to editorial changes in the manuscript.

Ethics Approval and Consent to Participate

Approval was obtained from the Ethics Committee on Animal Experiments, Shanghai Jiao Tong University School of Medicine (SYXK-2003-0026).

Acknowledgment

We thank Dr. Honglin Wang for his assistance in providing the PdgfraDreER-tdTomato mice.

Funding

This work is supported by grants from the National Natural Science Foundation of China (No. 82070509), and Innovative research team of high-level local universities in Shanghai (No. SHSMU-ZDCX20212700).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Ebert MLA, Schmidt VF, Pfaff L, von Thaden A, Kimm MA, Wildgruber M. Animal Models of Neointimal Hyperplasia and Restenosis: Species-Specific Differences and Implications for Translational Research. JACC. Basic to Translational Science. 2021; 6: 900–917.
[2]
Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nature Methods. 2009; 6: 377–382.
[3]
Cao G, Xuan X, Li Y, Hu J, Zhang R, Jin H, et al. Single-cell RNA sequencing reveals the vascular smooth muscle cell phenotypic landscape in aortic aneurysm. Cell Communication and Signaling: CCS. 2023; 21: 113.
[4]
Xiong J, Li Z, Tang H, Duan Y, Ban X, Xu K, et al. Bulk and single-cell characterisation of the immune heterogeneity of atherosclerosis identifies novel targets for immunotherapy. BMC Biology. 2023; 21: 46.
[5]
Jiang L, Chen T, Sun S, Wang R, Deng J, Lyu L, et al. Nonbone Marrow CD34+ Cells Are Crucial for Endothelial Repair of Injured Artery. Circulation Research. 2021; 129: e146–e165.
[6]
Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nature Reviews. Molecular Cell Biology. 2014; 15: 178–196.
[7]
Li F, Yan K, Wu L, Zheng Z, Du Y, Liu Z, et al. Single-cell RNA-seq reveals cellular heterogeneity of mouse carotid artery under disturbed flow. Cell Death Discovery. 2021; 7: 180.
[8]
Siow RCM, Churchman AT. Adventitial growth factor signalling and vascular remodelling: potential of perivascular gene transfer from the outside-in. Cardiovascular Research. 2007; 75: 659–668.
[9]
Zhang L, Jambusaria A, Hong Z, Marsboom G, Toth PT, Herbert BS, et al. SOX17 Regulates Conversion of Human Fibroblasts Into Endothelial Cells and Erythroblasts by Dedifferentiation Into CD34+ Progenitor Cells. Circulation. 2017; 135: 2505–2523.
[10]
Sorokin V, Vickneson K, Kofidis T, Woo CC, Lin XY, Foo R, et al. Role of Vascular Smooth Muscle Cell Plasticity and Interactions in Vessel Wall Inflammation. Frontiers in Immunology. 2020; 11: 599415.
[11]
Worssam MD, Lambert J, Oc S, Taylor JCK, Taylor AL, Dobnikar L, et al. Cellular mechanisms of oligoclonal vascular smooth muscle cell expansion in cardiovascular disease. Cardiovascular Research. 2023; 119: 1279–1294.
[12]
Chappell J, Harman JL, Narasimhan VM, Yu H, Foote K, Simons BD, et al. Extensive Proliferation of a Subset of Differentiated, yet Plastic, Medial Vascular Smooth Muscle Cells Contributes to Neointimal Formation in Mouse Injury and Atherosclerosis Models. Circulation Research. 2016; 119: 1313–1323.
[13]
Wirka RC, Wagh D, Paik DT, Pjanic M, Nguyen T, Miller CL, et al. Atheroprotective roles of smooth muscle cell phenotypic modulation and the TCF21 disease gene as revealed by single-cell analysis. Nature Medicine. 2019; 25: 1280–1289.
[14]
He L, Huang X, Kanisicak O, Li Y, Wang Y, Li Y, et al. Preexisting endothelial cells mediate cardiac neovascularization after injury. The Journal of Clinical Investigation. 2017; 127: 2968–2981.
[15]
Pan XX, Ruan CC, Liu XY, Kong LR, Ma Y, Wu QH, et al. Perivascular adipose tissue-derived stromal cells contribute to vascular remodeling during aging. Aging Cell. 2019; 18: e12969.
[16]
Hao Y, Hao S, Andersen-Nissen E, Mauck WM, 3rd, Zheng S, Butler A, et al. Integrated analysis of multimodal single-cell data. Cell. 2021; 184: 3573–3587.e29.
[17]
Nowotschin S, Setty M, Kuo YY, Liu V, Garg V, Sharma R, et al. The emergent landscape of the mouse gut endoderm at single-cell resolution. Nature. 2019; 569: 361–367.
[18]
McGinnis CS, Murrow LM, Gartner ZJ. DoubletFinder: Doublet Detection in Single-Cell RNA Sequencing Data Using Artificial Nearest Neighbors. Cell Systems. 2019; 8: 329–337.e4.
[19]
Blanco-Carmona E. Generating publication ready visualizations for Single Cell transcriptomics using SCpubr. bioRxiv, 2022. (preprint)
[20]
Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nature Communications. 2019; 10: 1523.
[21]
Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, et al. clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innovation (Cambridge (Mass.)). 2021; 2: 100141.
[22]
Aibar S, González-Blas CB, Moerman T, Huynh-Thu VA, Imrichova H, Hulselmans G, et al. SCENIC: single-cell regulatory network inference and clustering. Nature Methods. 2017; 14: 1083–1086.
[23]
Cao J, Spielmann M, Qiu X, Huang X, Ibrahim DM, Hill AJ, et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature. 2019; 566: 496–502.
[24]
Bergen V, Lange M, Peidli S, Wolf FA, Theis FJ. Generalizing RNA velocity to transient cell states through dynamical modeling. Nature Biotechnology. 2020; 38: 1408–1414.
[25]
Qiu X, Mao Q, Tang Y, Wang L, Chawla R, Pliner HA, et al. Reversed graph embedding resolves complex single-cell trajectories. Nature Methods. 2017; 14: 979–982.
[26]
Browaeys R, Saelens W, Saeys Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nature Methods. 2020; 17: 159–162.
[27]
Dimayuga PC, Chyu KY, Cercek B. Immune responses regulating the response to vascular injury. Current Opinion in Lipidology. 2010; 21: 416–421.
[28]
Trujillo-Ochoa JL, Kazemian M, Afzali B. The role of transcription factors in shaping regulatory T cell identity. Nature Reviews. Immunology. 2023; 23: 842–856.
[29]
Li C, Wu H, Guo L, Liu D, Yang S, Li S, et al. Single-cell transcriptomics reveals cellular heterogeneity and molecular stratification of cervical cancer. Communications Biology. 2022; 5: 1208.
[30]
Sayed N, Wong WT, Ospino F, Meng S, Lee J, Jha A, et al. Transdifferentiation of human fibroblasts to endothelial cells: role of innate immunity. Circulation. 2015; 131: 300–309.
[31]
Cai X, Han M, Lou F, Sun Y, Yin Q, Sun L, et al. Tenascin C+ papillary fibroblasts facilitate neuro-immune interaction in a mouse model of psoriasis. Nature Communications. 2023; 14: 2004.
[32]
Yap C, Mieremet A, de Vries CJM, Micha D, de Waard V. Six Shades of Vascular Smooth Muscle Cells Illuminated by KLF4 (Krüppel-Like Factor 4). Arteriosclerosis, Thrombosis, and Vascular Biology. 2021; 41: 2693–2707.
[33]
La Manno G, Soldatov R, Zeisel A, Braun E, Hochgerner H, Petukhov V, et al. RNA velocity of single cells. Nature. 2018; 560: 494–498.
[34]
Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan CH, et al. Inference and analysis of cell-cell communication using CellChat. Nature Communications. 2021; 12: 1088.
[35]
Gao XF, Chen AQ, Wang ZM, Wang F, Luo S, Chen SY, et al. Single-Cell RNA Sequencing of the Rat Carotid Arteries Uncovers Potential Cellular Targets of Neointimal Hyperplasia. Frontiers in Cardiovascular Medicine. 2021; 8: 751525.
[36]
Saxena A, Björkbacka H, Ström Å, Rattik S, Berg KE, Gomez MF, et al. Mobilization of regulatory T cells in response to carotid injury does not influence subsequent neointima formation. PloS One. 2012; 7: e51556.
[37]
Chen K, Mou R, Zhu P, Xu X, Wang H, Jiang L, et al. The Effect of Lymphangiogenesis in Transplant Arteriosclerosis. Circulation. 2023; 147: 482–497.
[38]
Tian DY, Jin XR, Zeng X, Wang Y. Notch Signaling in Endothelial Cells: Is It the Therapeutic Target for Vascular Neointimal Hyperplasia? International Journal of Molecular Sciences. 2017; 18: 1615.
[39]
Li XD, Hong MN, Chen J, Lu YY, Ye MQ, Ma Y, et al. Adventitial fibroblast-derived vascular endothelial growth factor promotes vasa vasorum-associated neointima formation and macrophage recruitment. Cardiovascular Research. 2020; 116: 708–720.
[40]
Li Y, Ren P, Dawson A, Vasquez HG, Ageedi W, Zhang C, et al. Single-Cell Transcriptome Analysis Reveals Dynamic Cell Populations and Differential Gene Expression Patterns in Control and Aneurysmal Human Aortic Tissue. Circulation. 2020; 142: 1374–1388.
[41]
Yang B, Janardhanan R, Vohra P, Greene EL, Bhattacharya S, Withers S, et al. Adventitial transduction of lentivirus-shRNA-VEGF-A in arteriovenous fistula reduces venous stenosis formation. Kidney International. 2014; 85: 289–306.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top