- Research
- Open access
- Published:
Assessing personalized molecular portraits underlying endothelial-to-mesenchymal transition within pulmonary arterial hypertension
Molecular Medicine volume 30, Article number: 189 (2024)
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and rapidly fatal disease with an intricate etiology. Identifying biomarkers for early PAH lesions based on the exploration of subtle biological processes is significant for timely diagnosis and treatment. In the present study, nine distinct cell populations identified based on gene expression profiles revealed high heterogeneity in cell composition ratio, biological function, distribution preference, and communication patterns in PAH. Notably, compared to other cells, endothelial cells (ECs) showed prominent variation in multiple perspectives. Further analysis demonstrated the endothelial-to-mesenchymal transition (EndMT) in ECs and identified a subgroup exhibiting a contrasting phenotype. Based on these findings, a machine-learning integrated program consisting of nine learners was developed to create a PAH Endothelial-to-mesenchymal transition Signature (PETS). This study identified cell populations underlying EndMT and furnished a potential tool that might be valuable for PAH diagnosis and new precise therapies.
Introduction
Pulmonary arterial hypertension (PAH) is a relatively uncommon yet severe vascular disorder marked by heightened blood pressure within the pulmonary artery (mean > 20 mm Hg) (Simonneau, et al. 2019; Mocumbi, et al. 2024), resulting in a progressive condition characterized by heightened strain on the right ventricle that may culminate in right heart failure and mortality (Humbert, et al. 2022). Timely diagnoses of PAH patients remain a research focus, with the aim of improving outcomes and providing more treatment options (Ruopp and Cockrill 2022).
The etiology of PAH is intricate, encompassing pulmonary vascular remodeling, augmented vasoconstriction, and inflammation in both arterial and microvascular structures (Dai, et al. 2018; Zhong and Yu 2022; Mocumbi, et al. 2024). Endothelial cells (ECs) play a crucial role in the initial vascular alterations that precipitate PAH, which involves in dysfunctional signaling, injury, and apoptosis (Evans, et al. 2021). Endothelial-to-mesenchymal transition (EndMT), in which ECs obtain characteristics of mesenchymal cells, is deemed to be involved in this pathogenic process (Ranchoux, et al. 2015; Xue, et al. 2020; Evans, et al. 2021). Following EndMT, there is an increase in EC proliferation and migration, leading to pulmonary vascular remodeling and the progression of PAH (Li, et al. 2018; Alvandi and Bischoff 2021).
Recently, high-throughput sequencing technology has advanced rapidly and is now commonly used in clinical settings, such as disease mechanism exploration and infection pathogen identification (Daniloski, et al. 2021; Zhang, et al. 2022; Li, et al. 2023; Ren, et al. 2024). It is also a novel and effective way to guide individualized treatment based on multi-omics information integration (Dave, et al. 2023). While numerous studies have been carried out using bulk RNA sequencing (RNA-seq) data to explore PAH in detail, limitations in resolution and cellular heterogeneity have hindered further exploration. However, single-cell RNA sequencing (scRNA-seq) technology has emerged as a robust platform for investigating alterations in cellular function, cell–cell communication, and cellular development at a more refined level (Aldridge and Teichmann 2020; Zhang et al. 2023a, b). Thus, single-cell technology is regarded as an ideal method for exploring cell heterogeneity and identifying specific pathogenic cell populations (Fernandez and Giannarelli 2022). For instance, Rodor and colleagues discovered endothelial cell function alterations in mice with PAH through the scRNA-seq platform (Rodor, et al. 2022). Besides, Ferrian et al. constructed the PAH single cell architectural map, identified unique inflammation cell subsets, and characterized them (Ferrian, et al. 2024). Nevertheless, at single-cell resolution, specific research remains largely deficient in PAH. Therefore, it is essential to undertake further scRNA-seq research to elucidate mechanisms and identify novel biomarkers in PAH.
Here, we systematically integrated both scRNA-seq and bulk RNA-seq data to depict biological characteristics of PAH patients. Further trajectory analyses were performed on ECs because of its apparent change in function and role in PAH. Based on the observed EndMT process and a unique EC subset, we obtained EndMT pattern genes (ETPGs). Afterward, we implemented a machine learning (ML) program integrating nine learners into our research to develop a PAH Endothelial-mesenchymal Transition Signature (PETS), which exhibited favorable performance in discriminating between PAH patients and healthy people.
Materials and methods
scRNA-seq data collecting, processing and analysis
The 10X single-cell transcriptome data from GSE169471 (including 31,444 cells) were downloaded from the Gene Expression Omnibus (GEO) (http://www.ncbi.nlm.nih.gov/geo/) database (Saygin, et al. 2020). Quality control of single-cell RNA sequencing data was executed with the following criteria: > 200 genes/cell, < 4000 genes/cell, > 3 cells/gene, and < 10% mitochondrial genes. Then, the integration work was conducted as follows: (i) For individual objects, perform Log-Normalization and identify 2000 highly variable genes (HVGs) using the “vst” method. (ii) Divide the combined object into a list based on the “SplitObject” function. (iii) Use the “FindIntegrationAnchors” function to identify the “anchors” in the different objects to construct a reference. (iv) Eliminate the batch effect of nine samples through the “IntegrateData” function. Finally, a Seurat object with a batch-corrected expression matrix for all cells was outputted (Butler, et al. 2018; Hao, et al. 2021). Depending on the expression of the top 2000 HVGs, the top 20 Principal Component Analysis (PCA) components were determined. Afterward, the first 20 significant PCs determined by the Elbow plot were incorporated to conduct cell clustering using the “FindNeighbors” function and “FindClusters” method (resolution = 0.5), which implements the Louvain network-based clustering algorithm. Subsequently, Uniform Manifold Approximation and Projection (UMAP) analysis was applied for further dimensional reduction and clustering visualization (Becht, et al. 2018). To annotate cell types, highly expressed genes of all cell subclusters were used as potential references and combined with canonical cell-type-specific surface markers derived from CellMarker (http://yikedaxue.slwshop.cn/index.php).
Bulk data processing and analysis
The raw data were obtained from the GEO database, providing detailed information on the platform, samples, and GSE records. Probes were converted into gene symbols based on the annotation information from the platform. Subsequently, differential expression analysis was conducted using the limma package to identify differentially expressed genes (DEGs). Afterward, gene set enrichment analysis (GSEA) was performed using the clusterProfiler package.
Tissue preference of distinct cell types
We calculated the observed-to-predicted cell number ratio (Ro/e) for each cell type in different groups to quantify the preference of each cell type (Zhang, et al. 2018). The chi-square test was used to determine the predicted cell numbers for each combination of cell types and groups.
Assessing the contribution score of different cell types
In our study, the contribution score was calculated to assess the contribution of each cell cluster to PAH. The “FindAllMarkers” function was used to list the top 100 differentially expressed genes (DEGs) between two groups. Subsequently, we calculated the fold change in gene expression (FCexp) and the proportion of gene expression (FCprop) of PAH relative to the control group across different cell types. The square root of FCexp and FCprop was regarded as the contribution score.
Functional enrichment analysis at single-cell resolution
Gene sets of immune, metabolism, signaling, and proliferation pathways were collected from MSigDB (Liberzon, et al. 2011). Then, the AUCell algorithm was utilized to assess the activity of various biological pathways in both the PAH and control groups (Aibar, et al. 2017). Pathway activity scores were compared between the two groups using empirical Bayes’ statistics.
Cell–cell communication analysis
We applied Cellchat to depict cell–cell interactions among cell populations in both PAH and control groups (Jin, et al. 2021). Based on CellchatDB, a signaling molecule interaction database, over-expressed ligands and receptors were identified. Afterward, Cellchat quantifies intercellular communications by associating each interaction with a probability value, which is recalculated based on the average expression values of a ligand by one cell group and that of a receptor by another cell group, as well as their cofactors. We adopted the default settings of recommended pipelines to conduct a quantitative analysis of intercellular communications in an unsupervised manner.
Pseudotime trajectory inference
After further dimensionality reduction and clustering, we utilized the Slingshot algorithm to reconstruct the differentiation trajectories of endothelial cells (Street, et al. 2018). The input for the Slingshot calculation and cell lineage trajectory formation consisted of UMAP embedding and subclustering assignments for 3165 endothelial cells from both PAH and control groups. Additionally, we applied the Monocle2 pseudotime algorithm to recapitulate the results (Qiu, et al. 2017). The “differentialGeneTest” function was employed to identify differentially expressed genes among clusters, followed by dimensionality reduction using the DDRTree method. Finally, cells were ordered and differentiation trajectories were built, while significant differentially expressed genes (qval < 0.1) were clustered to illustrate distinct expression patterns along the trajectory.
Enrichment analysis
The “FindAllMarkers” function was used to list cell populations’ markers to further explore their functions. Next, enrichment analysis was conducted through the “RunEnrichment” function in the SCP package (https://github.com/zhanghao-njmu/SCP). Identified differentially expressed genes were subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) terms enrichment analyses to depict functional differences along the pseudotime trajectory (Gene 2015; Kanehisa, et al. 2017). For the lung tissue RNA-sequencing cohort, gene set enrichment analysis (GSEA) was conducted to evaluate the biological pathway activities based on the ranked differential gene expression matrix (Subramanian, et al. 2005).
Single-cell metabolic activity analysis
The assessment of metabolic activity at the single-cell level was conducted using scMetabolism, which utilized metabolic pathways from KEGG and Reactome entries to quantify metabolic activity through a single-cell matrix file and the Vision algorithm (Gillespie, et al. 2022; Wu, et al. 2022).
Pivotal module genes identification
To identify pivotal genes associated with endothelial differentiation, high dimensional weighted gene co-expression network analysis (hdWGCNA) was executed as follows: (i) Construct an expression matrix from the single-cell object undergoing PCA and UMAP dimensional reduction; (ii) Based on the expression matrix, construct averaged “metacells” by grouping cells according to cell types; (iii) Normalize the metacell matrix and determine a suitable soft threshold β to create a topological overlap matrix (TOM); (iv) Establish the co-expression network and calculate the eigenvectors of the modules in order to identify module feature genes. The foregoing analyses were implemented based on hdWGCNA R package (Morabito, et al. 2021). Finally, the pivotal module and contained genes were recognized after evaluating the expression of module genes in distinct disease groups and endothelial subtypes.
Following the identification of pivotal module genes, EC3 significant altered genes were selected through FindAllMarkers function, applying a significance threshold of average log2 Fold Change > 1 and adjusted P value < 0.05. Their overlap genes were defined as the key genes involved in the EndMT process, called EndMT pattern genes (ETPGs).
PETS generated from integrative machine-learning program
To explore a PAH endothelial-mesenchymal transition signature (PETS), we leveraged nine machine learning algorithms, comprising elastic-net-regularized generalized linear model (glmNet), bootstrap aggregation classification and regression trees (Bagged CART), Naive Bayes (NB), Partial Least Squares (pls), Backpropagation neural network (NNet), K-Nearest Neighbors (KNN), random forest (RF), boosted generalized linear model (glmBoost), classification and regression trees (CART) (Zhang, Cui, et al. 2023; Zhang, et al. 2024). The final signature was generated following such a pipeline: (i) The initial investigation of the signature was carried out in the cohort GSE117261, which was randomly split into discovery and testing cohorts at a ratio of 7:3. (ii) Nine machine learning algorithms were employed on ETPGs to individually fit models. In order to mitigate the risk of overfitting resulting from the excessive complexity of the model, a tenfold 10-repeated cross-validation approach was implemented to enhance the generalization capability of the discovery cohort. (iii) In the testing cohort, a consensus evaluation strategy was applied to assess effectiveness and applicability of all models, encompassing the accuracy, Harrell’s concordance index (C-index), F1-score, precision, recall, and the root mean square of the residuals (RMSR). The glmNet model was recognized as the optimal scheme.
Cell lines and cultures
Primary human and mouse pulmonary artery endothelial cells (PAECs) were obtained from Wuhan Procell Life Technology company Wuhan Procell Life Science & Technology Corporation. All cells were cultured in Dulbecco's Modified Eagle Medium supplemented with 10% fetal bovine serum and maintained in a humidified incubator with 5% CO2 at 37 °C. The hypoxic PAECs were cultured in a 1% O2 incubator (Galaxy R; RS Bitotech, Alloa, UK) continually gassed with 5% CO2 and 90% N2.
RNA extraction and real-time quantitative PCR
Total RNA was isolated using Trizol (Takara, Dalian, China) according to the manufacture’s instruction. RNA concentration was determined by Nanodrop analysis. cDNA was synthesized from total RNA using PrimeScript RT reagent kit (Takara, Dalian, China). Quantitative RT-PCR was performed using SYBR Green Mix (Takara, Dalian, China). The primer sequences were as follows: Rarres2 forward, 5′-TTGCTGATCTCCCTAGCCCTA-3′, reverse, 5′-TGGGTGTTTGTGGAACTCCTC-3′. Tc2n forward, 5′-CACCTGATTTAAGTAGGCGCTT-3′, reverse, 5′-GGATGAATCGCTGAGATGAAGTT-3′. RARRES2 forward, 5′-AGAAACCCGAGTGCAAAGTCA-3′, reverse, 5′-AGAACTTGGGTCTCTATGGGG-3′. TC2N forward, 5′-TGGCTGTACTGAGGATTATTTGC-3′, reverse, 5′-TGTGAAGGAGTTTCTTGTGTCC-3′. CBY1 forward, 5′-TCTTTGGGAATACGTTCAGTCCG-3′, reverse, 5′-CCAGGTTCATAGTCGGGGA-3′. CKS1B forward, 5′-TATTCGGACAAATACGACGACG-3′, reverse, 5′-CGCCAAGATTCCTCCATTCAGA-3′. MSRB3 forward, 5′-CGGTTCAGGTTGGCCTTCATT-3′, reverse, 5′-GTGCATCCCATAGGAAAAGTCA-3′. SMAGP forward, 5′-ACCAGCCTCCTGACTACTCC-3′, reverse, 5′-CTTGCCACAGGACACCTTCA-3′.
The cycling conditions were as follows: initial denaturation for 10 min at 95 °C followed by 40 cycles of denaturation (15 s at 95 °C), annealing and elongation (30 s at 60 °C). The relative expression of gene was calculated using 2 − △△Ct method using β-Actin as the reference gene.
Mouse hypoxic PAH model and immunofluorescence Staining
Adult male Sprague–Dawley C57BL/6 J mice were purchased from the Laboratory Animal Center, Tongji Medical College. The mouse were placed in a normobaric chamber (Oxycycler model A84XOV; BioSpherix, Lacona, NY), and the oxygen concentration was adjusted to 10% for 4 weeks for hypoxia treatment. Hemodynamic studies and right ventricular hypertrophy were evaluated as previously reported (Tang, et al. 2022). The lungs of the mice were fixed by 4% paraformaldehyde and routinely processed into paraffin sections, 4 mm in thickness, for immunostaining examination. Paraffin sections of the lung were stained with DAPI (4′,6-diamidino-2-phenylindole), anti-CD31 (1:100; Proteintech Group), and anti-chemerin (1:100; Proteintech Group).
Statistical analysis
All data processing, plotting, and statistical analysis were conducted in R (version 4.1.2). Kruskal–Wallis test was applied for oxidative phosphorylation level comparison among three EC subsets. The T test was applied to compare continuous variables. For all statistical tests, a two-sided P < 0.05 and FDR < 0.05 was deemed statistically significant (Fig. 1).
Results
scRNA‑seq analysis revealed cell landscape and characteristics in PAH
The 31,444 cells from three PAH samples and six control samples were analyzed via 10X scRNA-seq. After quality control, 28,906 cells were ultimately retained (Figure S1). Subsequently, UMAP analysis revealed 21 cell clusters (Figure S2A), which were further annotated into the following cell types: B cells, endothelial cells (ECs), epithelial cells, fibroblasts, macrophages/monocytes, mast cells, natural killer (NK) cells, smooth muscle cells (SMCs), and T cells (Figure S2B, Fig. 2A). We then quantified PAH-driven alterations in the cell type proportions. As depicted in Fig. 2B and S3, there was an increase in ECs, SMCs, and fibroblasts in the PAH group.
scRNA-seq depicted the cell landscape in PAH. A Cell landscapes of 28906 cells from 9 types (B cells, endothelial cells, epithelial cells, fibroblasts, macrophages/monocytes, mast cells, natural killer cells, smooth muscle cells, and T cells). B Cell proportion in PAH and control groups. C Line chart showing tissue prevalence for each cell type estimated by Ro/e score. D Pie chart exhibiting contribution score of distinct cell types. E Dot plots showing differentially enriched pathways in the global cell types
To measure the tissue enrichment of cell populations, we carried out Ro/e analysis. Similarly, most immune cells preferred to distribute themselves in the normal lungs, whereas mast cells and stromal cells were preferentially located in the PAH lung tissues (Fig. 2C). Afterward, we assessed distinct cell populations’ contribution scores. For differences caused by PAH at the single-cell resolution, macrophages/monocytes and ECs accounted for significant contribution, while NK cells, T cells, and mast cells exhibited limited impact (Fig. 2D, Table S1, 2).
Immunology, metabolism, signaling, and proliferation hallmark gene sets were used to investigate major variations in pathways between the PAH and control group (Table S3). Notably, several specific pathways displayed significant enrichment in all stromal cells, such as coagulation, epithelial mesenchymal transition (EMT), apical junction, myogenesis, and TGF-β signaling pathway, implying the presence of potential shared biological processes in PAH (Fig. 2E). Of these, the most significant changes occurred in endothelial cells, which were more enriched in oxidative phosphorylation and MYC target V1 pathways.
Cell–cell communications of cell populations
Relying on CellChat established ligand-receptor pairs and their cofactors, we constructed a cell–cell communication network (Figure S4A). Attractively, elevated intercellular interactions numbers and strength were detected in the PAH group (Fig. 3A). Remarkable enhanced interactions of ECs and fibroblasts with B cells, T cells, NK cells, and macrophages/monocytes probably accounted for it (Fig. 3B). Then, we identified 18 enriched signaling pathways based on intercellular interaction information. MK, VSFATIN, GAS, and TWEAK signaling pathways occupied a dominant position in the PAH group, while ANGPTL, CALCR, GDF, and CSF3 signaling pathways were specifically enriched in the control group (Fig. 3C). In regard to these pathways, a detailed analysis of changes in signaling-receptor levels revealed that PAH-dominant signaling pathways were induced by ECs, fibroblasts, and macrophages/monocytes (Fig. 3D). Interestingly, in the PAH group, ECs, epithelial cells, and fibroblasts exhibited significantly reduced activity in control-dominant signaling pathways (Fig. 3D).
Cell–cell interaction differences in PAH. A Bar plot demonstrating elevated intercellular interaction number and strength in PAH. B Heatmap showing alterations in cell–cell interactions among distinct cell types. C-D Diverging activities between two groups and among specific cell types within each group in 18 enriched signaling pathways. E Variation of out-degree and in-degree of cell populations within the communication network in PAH
Finally, we deduced the transformation of cell population roles based on changes in out-degree and in-degree within the communication network (Fig. 3E). In PAH, epithelial cells and fibroblasts displayed increased outgoing interactions, whereas NK cells and T cells exhibited higher incoming interactions. Notably, ECs possessed lower incoming interactions and higher outgoing interactions in the PAH group, which signified the change from the signaling sender to a signaling receiver. Overall, further investigation of ECs is pressing due to extensive and significant changes in comprehensive exploration containing cell proportion, Ro/e analysis, cell contribution, and cell–cell communication.
Trajectory analyses revealed EndMT in endothelial populations
To gain a deeper understanding of the dynamics of cell differentiation, we conducted pseudotime developmental trajectory analysis. Following further dimensionality reduction and clustering, endothelial cells were categorized into three distinct populations: EC1, EC2, and EC3 (Fig. 4A). Initial Slingshot pseudotime trajectory analysis inferred the lineage structure with EC1 at the starting point position and EC3 at the end point position (Fig. 4B). We further examined subsets of ECs to elucidate the functional differentiation. Compared to EC1 and EC2, EC3 exhibited obvious heterogeneity and possessed 602 significant marker genes (Fig. 4C, Table S4). As depicted in Fig. 4D and S4B, immune-related pathways were activated during the initial stages of differentiation, including response to bacterial molecules and leukocyte migration. Subsequently, as cell differentiation progressed, there was an up-regulation in mitochondrial gene expression, followed by enrichment in mitochondrial transport and oxidative phosphorylation pathways.
Trajectory analysis of endothelial populations. A UMAP plots showed that a total of 3165 cells from the PAH and control group were recruited for further trajectory analysis. B The developmental trajectory of endothelial cells identified by Slingshot. C Volcano plot exhibited the DEGs among EC1, EC2, and EC3. D Alterations of biological function enrichment along the developmental trajectory. E The cell differentiation trajectory identified by Monocle 2, coloured-coded by the associated EC subsets (top), pseudotime (middle), and states (bottom). F Heatmap demonstrated different expressed patterns of clustered DEGs (termed C1, C2, and C3) along the pseudotime trajectory. G Bar plot showed selected enriched pathways related to C1, C2, and C3
We further reconstructed the cell differentiation trajectory using the monocle2 algorithm, and obtaining a similar pseudotime trajectory (Fig. 4E). By clustering pseudotime correlating genes, we identified three distinct gene expression patterns (Fig. 4F), namely Cluster1 (C1), Cluster2 (C2), and Cluster3 (C3). C1, which exhibited high expression at the onset of differentiation, was enriched in immune-related pathways such as myeloid leukocyte activation, T cell proliferation, and cell–cell adhesion-related pathways (Fig. 4G). C2, on the other hand, showed high expression during mid-differentiation and was associated with mesenchyme development, negative regulation of cell–cell adhesion, and negative chemotaxis pathways (Fig. 4G). Finally, C3 displayed high expression towards the end of differentiation and possessed correlation with pathways associated with mesenchymal transition, including extracellular matrix organization, cell transition, and positive regulation of extracellular matrix organization (Fig. 4G). For cell–cell communications, EC3 showed enhanced connection with fibroblasts and lacked communication with immune cells compared to EC1 (Figure S4C, D). Besides, we performed the quantitative metabolic activity analysis to validate our results, which indicated the most metabolically active in oxidative phosphorylation in EC3 (Fig. 5A, Figure S5A).
Functional enrichment analysis in the PAH patient cohort
We performed further analysis of the lung tissue RNA-seq data from PAH patients to confirm our conclusions from the single-cell level. After analyzing differentially expressed genes, 38 up-regulated genes and 30 down-regulated genes were identified (Fig. 5B, Table S5). GSEA was conducted to evaluate the biological processes associated with the ranked differential gene expression matrix. Likewise, we observed enrichment of mesenchymal differentiation-related pathways in PAH patients, including mesenchymal cell differentiation, collagen-containing extracellular matrix, EMT, extracellular matrix structure, and collagen fibril organization (Fig. 5C). In addition, the activation of TGF-β pathway and down-regulation of fatty acid metabolism pathway matched our findings obtained at the single-cell level (Fig. 5C).
Identification of EndMT-related modules and genes
Collectively, scRNA-seq and bulk RNA-seq analysis unveiled the presence of EndMT in PAH ECs. As described, EC3 showed the most significant connection with EndMT. Therefore, we further performed hdWGCNA analysis on EC3. The single-cell transcriptome co-expression matrix was constructed with the optimal soft threshold of 24 (fraction = 0.05) (Fig. 6A). According to the scale-free network structure, 13 modules were identified. Meanwhile, their dendrogram, module eigengenes, and module connectivity (kMEs) were demonstrated (Fig. 6B, C). Interestingly, the greenyellow module possessed a higher expression level in both EC3 and the PAH group and was recognized as the EndMT-related module (Fig. 6D, E). The intersection of 66 greenyellow module genes and 602 significant EC3 marker genes were defined as EndMT pattern genes (ETPGs), including RARRES2, CBY1, MSRB3, TC2N, CKS1B, and SMAGP (Fig. 6F, Table S6). We evaluated the ability of each gene to recognize PAH separately, with RARRES2 standing out (Figure S5B). Besides, compared to the control group, elevated mRNA expression levels of RARRES2, TC2N, CKS1B, and SMAGP were detected in human hypoxia-induced PAECs (Figure S6A). Though higher expression of CBY1 and MSRB1 were detected in human hypoxia-induced PAECs, the difference compared to the control group does not achieve statistical significance (Figure S6A).
hdWGCNA identified key genes associated with EC3. A Scale-free topological indices at various soft-thresholding powers (left); correlation analysis between the soft-thresholding powers and mean connectivity ofthe network. Setting the soft power of β to 24, resulted in a high scale-free R2 (> 0.8), indicating a good fit to the scale-free topology model, and the stabilization ofmean connectivity (B) Dendrogram showed 13 modules identified from the topological overlap matrix. C Module eigengenes and their connectivity (kMEs) of 13 modules. D Heatmap showed average expression of each module between two groups and among three EC subsets. E Network plots showed hub genes of the greenyellow module. F Venn plots showed six identified EndMT pattern genes
Machine learning-based integrative program generates PETS
ETPGs were incorporated into our machine learning-based integrative program to establish PETS. We employed nine classical learners using tenfold cross-validation with 10 repetitions to fit models and evaluated their performance using five metrics, including accuracy, C-index, F1-score, precision, recall, and root-mean-square of the residuals (RMSR), in the testing cohort. The generalized linear model demonstrated superior classification capabilities based on these performance metrics (Fig. 7A, Table S7). Following the determination of optimal regularization parameters by considering accuracy variation in cross-validation, the final model was identified as follows: PETS = 2.6926723 × RARRES2 + 0.7410611 × TC2N (Fig. 7B, C). Importantly, PAH patients exhibited higher PETS levels compared to healthy individuals, and PETS levels showed a better ability to recognize PAH (Fig. 7D), suggesting the potential of PETS levels as a biomarker for PAH. Meanwhile, in mouse hypoxia-induced PAECs, elevated mRNA expression levels of RARRES2 and TC2N were detected, consistent with our conclusions (Fig. 7E). As the most prominent factor in PETS, we conducted immunofluorescence staining with the protein encoded by RARRES2 (chemerin) and the EC marker gene in hypoxic PAH mice lung paraffin sections. As illustrated in Figure S6B, chemerin was upregulated in PAH ECs.
Machine learning-based integrative program generates PETS. A Comprehensive performances of nine types of learners. Boxplots depicted the distribution of the residuals, with red highlighted dots representing root-mean-square of residuals (RMSR). Circles showed the distribution of recall, precision, F1-score, C-index, and accuracy of each learner. B-C The determination of the optimal λ was obtained when the partial likelihood deviance reached the minimum value, and further generated glmNet coefficients of the most useful prognostic genes. D Boxplot showed PETS levels in PAH and control group. E Bar plot showed mRNA expression levels of RARRES2 and TC2N in hypoxia induced mouse PAECs and control
Discussion
PAH was a chronic, progressive and rapidly fatal disease with a poor prognosis, high mortality rate and lack of curative treatment options (Hassoun 2021). The early diagnosis and treatment of PAH have remained a significant challenge for clinicians (Vachiery and Gaine 2012; Poch and Mandel 2021). As a multifactorial disease, the development of PAH has been demonstrated to involve numerous biological processes that affect ECs, SMCs, and fibroblasts (Maron, et al. 2021). Fortunately, rapid development of high-throughput sequencing technology provided an opportunity to explore these biological processes. In recent years, researchers have completed a number of studies on PAH using this technology (Yao, et al. 2021; Tang, et al. 2022; He, et al. 2023). However, most of them focused on bulk RNA sequencing, and there is still limited research on the application of single-cell sequencing to explore the specific biological processes of PAH. In this study, we demonstrated that there were complex and heterogeneous changes in cell proportion, cell function, distributed preference, and cell–cell communication among PAH patients at the single-cell resolution. By integrating scRNA-seq and bulk RNA-seq data, we confirmed the occurrence of the EndMT process in the early stages of PAH, which was crucial for PAH development. Through further analysis of the EndMT process, we identified EndMT pattern genes (ETPGs). Based on ETPGs, our machine learning-based integrative program developed an optimal classifier (referred to as PETS), which effectively distinguished between PAH patients and healthy individuals.
The histologic features of PAH were complex and varied, but the uncontrolled growth of ECs, SMCs, and fibroblasts was a common feature (Southgate, et al. 2020). Consistently, we observed an elevated proportion of these cells and mast cells in PAH, which also exhibited preference and leading contribution to the disease. Furthermore, enhanced intercellular interactions were observed in PAH, particularly involving ECs and SMCs interacting with other cells. In recent years, ECs have gained interest, as they are affected in early PAH. For instance, Liang et al. Reported that TGF-β is a crucial mediator of EndMT (Liang, et al. 2022). Wang et al. Found a low level of mediator complex subunit 1 (MED1) in PAH patients’ lung tissue and endothelial cells, which promoted vascular remodeling via the TGF-β signaling pathway (Wang, et al. 2022). Notably, ECs from PAH patients revealed significant biological processes related to alterations in cell proliferation and EndMT, including oxidative phosphorylation, myogenesis, and the TGF-β signaling pathway. TGF-β signaling pathway activation, mesenchymal cells production and differentiation were detected in the bulk RNA-seq dataset as well. Moreover, ECs showed lower incoming interactions and higher outgoing interactions in the PAH group, which might indicate functional dysfunction.
These rousing findings prompted us to conduct further research on ECs. Surprisingly, we identified a unique unreported subgroup, which possessed heterogeneity and located at the terminal differentiation stage (termed EC3). At the gene expression level, it displayed significant differences compared to other ECs, mainly pointing to elevated oxidative phosphorylation activity. Subsequently, EC3 demonstrated mesenchymal differentiation and down-regulation of lipid metabolism gene expression patterns in the differentiation fate. Consistent findings occurred in the PAH lung tissue RNA-seq cohort, which refined the understanding of EC3. Of note, EC3 displayed enhanced cell–cell communications with fibroblasts. EndMT has the potential to induce the transformation of endothelial cells into smooth muscle-like cells with enhanced abilities in proliferation and migration, thereby directly contributing to the development of PAH or indirectly through paracrine effects on vascular intimal and medial proliferation (Suzuki, et al. 2018; Gorelova, et al. 2021). Therefore, it was of consequence to identify early diagnosis and treatment targets based on EC3.
At the single-cell level, we further constructed the topological overlap matrix and established the co-expression network to determine meaningful modules and feature genes. Based on the EC3-specific module and EC3 marker genes, we identified six EndMT pattern genes (ETPGs), including RARRES2, CBY1, MSRB3, TC2N, CKS1B, and SMAGP. Then, we aimed to develop an EndMT-related signature for the diagnosis and treatment of PAH patients. Nowadays, machine learning is emerging as a robust method for medical decision-making, demonstrating superior performance compared to traditional models in disease diagnosis, prognosis assessment, and disease progression prediction (Liu, et al. 2022; Silva, et al. 2022; Sun, et al. 2022). However, predictive power deficiency caused by researchers’ selection bias and knowledge limitations hindered the clinical application of many biomarkers (Shimron, et al. 2022; Volovici, et al. 2022). In contrast, we developed a systematic framework to fully exploit nine machine learning algorithms and performed comprehensive evaluations. Finally, the glmNet algorithm provided the optimal PETS, which exhibited powerful predictive performance. Therefore, we believe PETS might serve as a key biomarker of PAH diagnosis and a target for anti-EndMT therapy.
Retinoic acid receptor responder 2 (RARRES2), also known as chemerin, is an important adipokine in obesity, inflammation, and cardiovascular disease. Previous studies have reported that chemerin could promote angiogenesis and ROS production and decrease insulin signaling and nitric oxide production in vascular endothelial cells (Bozaoglu, et al. 2010; Neves, et al. 2018; Ferland, et al. 2020). Recently, serum chemerin levels have received increasing attention from clinical researchers, including cardiovascular and renal disease-related evaluations (Bonomini and Pandolfi 2016; Zhou, et al. 2019; Szpakowicz, et al. 2021). Relying on serum chemerin and TC2N levels, our PETS was a potential tool to indicate the risk of PAH at an early stage in clinical practice, which was conducive for early intervention and timely treatment. Additionally, new therapeutic drugs targeting chemerin and related signaling proteins, such as the receptor chemokine-like receptor 1 (CMKLR1), are also in progress. A CMKLR1 inhibitor, CCX832, has been extensively studied in vitro and in vivo, exhibiting significant ability to ameliorate chemerin-induced vascular dysfunction (Xie and Liu 2022). Thus, CCX832 may be a treatment option for PAH patients, but only after rigorous preclinical studies.
The following limitations in this study should be recognized: (1) Procedures bias from the system biology potentially cannot fully recapitulate the overall disease exacerbation. (2) All samples were retrospective data in our current research, a prospective study should be performed. (3) Cell dissociation-associated spatial information deficiency influenced the prediction of cell communication, further single-cell multi-omics studies would further expound the pathogenesis of PAH. (4) Since the current study is based on a high-throughput RNA data, multi-omics’ comprehensive studies would be crucial addition.
Conclusion
In conclusion, we further depicted the biological landscape in PAH by incorporating both scRNA-seq and bulk RNA-seq data. Using a computational biology approach, we explored unique alterations in ECs and developed a robust signature (PETS) for PAH diagnosis. These findings offered clues for future biological and clinical research and should prove valuable in prospective therapy strategies, specifically targeting EndMT.
Availability of data and materials
No datasets were generated or analysed during the current study.
Code availability
The code that supports the findings of this study is available from the corresponding author upon reasonable request.
Abbreviations
- PAH:
-
Pulmonary arterial hypertension
- EC:
-
Endothelial cell
- EndMT:
-
Endothelial-to-mesenchymal transition
- scRNA-seq:
-
Single-cell RNA sequencing
- ETPG:
-
EndMT pattern gene
- DEG:
-
Differentially expressed gene
- HVG:
-
Highly variable gene
- UMAP:
-
Uniform manifold approximation and projection
- KEGG:
-
Kyoto encyclopedia of genes and genomes
- GO:
-
Gene ontology
- GSEA:
-
Gene set enrichment analysis
- EMT:
-
Epithelial mesenchymal transition
- RMSR:
-
Root mean square of the residuals
- SMC:
-
Smooth muscle cell
References
Aibar S, Gonzalez-Blas CB, Moerman T, Huynh-Thu VA, Imrichova H, Hulselmans G, Rambow F, Marine JC, Geurts P, Aerts J, et al. SCENIC: single-cell regulatory network inference and clustering. Nat Methods. 2017;14:1083–6.
Aldridge S, Teichmann SA. Single cell transcriptomics comes of age. Nat Commun. 2020;11:4307.
Alvandi Z, Bischoff J. Endothelial-mesenchymal transition in cardiovascular disease. Arterioscler Thromb Vasc Biol. 2021;41:2357–69.
Becht E, McInnes L, Healy J, Dutertre CA, Kwok IWH, Ng LG, Ginhoux F, Newell EW. Dimensionality reduction for visualizing single-cell data using UMAP. Nat Biotechnol. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nbt.4314.
Bonomini M, Pandolfi A. Chemerin in renal dysfunction and cardiovascular disease. Vascul Pharmacol. 2016;77:28–34.
Bozaoglu K, Curran JE, Stocker CJ, Zaibi MS, Segal D, Konstantopoulos N, Morrison S, Carless M, Dyer TD, Cole SA, et al. Chemerin, a novel adipokine in the regulation of angiogenesis. J Clin Endocrinol Metab. 2010;95:2476–85.
Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36:411–20.
Dai Z, Zhu MM, Peng Y, Machireddy N, Evans CE, Machado R, Zhang X, Zhao YY. Therapeutic targeting of vascular remodeling and right heart failure in pulmonary arterial hypertension with a HIF-2alpha inhibitor. Am J Respir Crit Care Med. 2018;198:1423–34.
Daniloski Z, Jordan TX, Wessels HH, Hoagland DA, Kasela S, Legut M, Maniatis S, Mimitou EP, Lu L, Geller E, et al. Identification of required host factors for SARS-CoV-2 infection in human cells. Cell. 2021;184(92–105): e116.
Dave J, Jagana V, Janostiak R, Bisserier M. Unraveling the epigenetic landscape of pulmonary arterial hypertension: implications for personalized medicine development. J Transl Med. 2023;21:477.
Evans CE, Cober ND, Dai Z, Stewart DJ, Zhao YY. Endothelial cells in the pathogenesis of pulmonary arterial hypertension. Eur Respir J. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1183/13993003.03957-2020.
Ferland DJ, Mullick AE, Watts SW. Chemerin as a driver of hypertension: a consideration. Am J Hypertens. 2020;33:975–86.
Fernandez DM, Giannarelli C. Immune cell profiling in atherosclerosis: role in research and precision medicine. Nat Rev Cardiol. 2022;19:43–58.
Ferrian S, Cao A, McCaffrey EF, Saito T, Greenwald NF, Nicolls MR, Bruce T, Zamanian RT, Del Rosario P, Rabinovitch M, et al. Single-cell imaging maps inflammatory cell subsets to pulmonary arterial hypertension vasculopathy. Am J Respir Crit Care Med. 2024;209:206–18.
Gene OC. Gene ontology consortium: going forward. Nucleic Acids Res. 2015;43:D1049-1056.
Gillespie M, Jassal B, Stephan R, Milacic M, Rothfels K, Senff-Ribeiro A, Griss J, Sevilla C, Matthews L, Gong C, et al. The reactome pathway knowledgebase 2022. Nucleic Acids Res. 2022;50:D687–92.
Gorelova A, Berman M, Al GI. Endothelial-to-mesenchymal transition in pulmonary arterial hypertension. Antioxid Redox Signal. 2021;34:891–914.
Hao Y, Hao S, Andersen-Nissen E, Mauck WM 3rd, Zheng S, Butler A, Lee MJ, Wilk AJ, Darby C, Zager M, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184(3573–3587): e3529.
Hassoun PM. Pulmonary arterial hypertension. N Engl J Med. 2021;385:2361–76.
He X, Fang J, Gong M, Zhang J, Xie R, Zhao D, Gu Y, Ma L, Pang X, Cui Y. Identification of immune-associated signatures and potential therapeutic targets for pulmonary arterial hypertension. J Cell Mol Med. 2023;27:3864–77.
Humbert M, Kovacs G, Hoeper MM, Badagliacca R, Berger RMF, Brida M, Carlsen J, Coats AJS, Escribano-Subias P, Ferrari P, et al. 2022 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Heart J. 2022;43:3618–731.
Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan CH, Myung P, Plikus MV, Nie Q. Inference and analysis of cell-cell communication using Cell Chat. Nat Commun. 2021;12:1088.
Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45:D353–61.
Li Y, Lui KO, Zhou B. Reassessing endothelial-to-mesenchymal transition in cardiovascular diseases. Nat Rev Cardiol. 2018;15:445–56.
Li H, Jiang Y, Hu J, Xu J, Chen L, Zhang G, Zhao J, Zong S, Guo Z, Li X, et al. The U2AF65/circNCAPG/RREB1 feedback loop promotes malignant phenotypes of glioma stem cells through activating the TGF-beta pathway. Cell Death Dis. 2023;14:23.
Liang G, Wang S, Shao J, Jin YJ, Xu L, Yan Y, Gunther S, Wang L, Offermanns S. Tenascin-X mediates flow-induced suppression of EndMT and atherosclerosis. Circ Res. 2022;130:1647–59.
Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. 2011;27:1739–40.
Liu Q, Sun D, Wang Y, Li P, Jiang T, Dai L, Duo M, Wu R, Cheng Z. Use of machine learning models to predict prognosis of combined pulmonary fibrosis and emphysema in a Chinese population. BMC Pulm Med. 2022;22:327.
Maron BA, Abman SH, Elliott CG, Frantz RP, Hopper RK, Horn EM, Nicolls MR, Shlobin OA, Shah SJ, Kovacs G, et al. Pulmonary arterial hypertension: diagnosis, treatment, and novel advances. Am J Respir Crit Care Med. 2021;203:1472–87.
Mocumbi A, Humbert M, Saxena A, Jing ZC, Sliwa K, Thienemann F, Archer SL, Stewart S. Pulmonary Hypertension Nat Rev Dis Primers. 2024;10:1.
Morabito S, Miyoshi E, Michael N, Shahin S, Martini AC, Head E, Silva J, Leavy K, Perez-Rosendahl M, Swarup V. Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer’s disease. Nat Genet. 2021;53:1143–55.
Neves KB, Nguyen Dinh Cat A, Alves-Lopes R, Harvey KY, Costa RMD, Lobato NS, Montezano AC, Oliveira AM, Touyz RM, Tostes RC. Chemerin receptor blockade improves vascular function in diabetic obese mice via redox-sensitive and Akt-dependent pathways. Am J Physiol Heart Circ Physiol. 2018;315:H1851–60.
Poch D, Mandel J. Pulmonary Hypertension. Ann Intern Med. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.7326/AITC202104200.
Qiu X, Mao Q, Tang Y, Wang L, Chawla R, Pliner HA, Trapnell C. Reversed graph embedding resolves complex single-cell trajectories. Nat Methods. 2017;14:979–82.
Ranchoux B, Antigny F, Rucker-Martin C, Hautefort A, Pechoux C, Bogaard HJ, Dorfmuller P, Remy S, Lecerf F, Plante S, et al. Endothelial-to-mesenchymal transition in pulmonary hypertension. Circulation. 2015;131:1006–18.
Ren Y, Wu R, Li C, Liu L, Li L, Weng S, Xu H, Xing Z, Zhang Y, Wang L, et al. Single-cell RNA sequencing integrated with bulk RNA sequencing analysis identifies a tumor immune microenvironment-related lncRNA signature in lung adenocarcinoma. BMC Biol. 2024;22:69.
Rodor J, Chen SH, Scanlon JP, Monteiro JP, Caudrillier A, Sweta S, Stewart KR, Shmakova A, Dobie R, Henderson BEP, et al. Single-cell RNA sequencing profiling of mouse endothelial cells in response to pulmonary arterial hypertension. Cardiovasc Res. 2022;118:2519–34.
Ruopp NF, Cockrill BA. Diagnosis and treatment of pulmonary arterial hypertension: a review. JAMA. 2022;327:1379–91.
Saygin D, Tabib T, Bittar HET, Valenzi E, Sembrat J, Chan SY, Rojas M, Lafyatis R. Transcriptional profiling of lung cell populations in idiopathic pulmonary arterial hypertension. Pulm Circ. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/2045894020908782.
Shimron E, Tamir JI, Wang K, Lustig M. Implicit data crimes: machine learning bias arising from misuse of public data. Proc Natl Acad Sci U S A. 2022;119: e2117203119.
Silva GFS, Fagundes TP, Teixeira BC, ChiavegattoFilho ADP. Machine learning for hypertension prediction: a systematic review. Curr Hypertens Rep. 2022;24:523–33.
Simonneau G, Montani D, Celermajer DS, Denton CP, Gatzoulis MA, Krowka M, Williams PG, Souza R. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1183/13993003.01913-2018.
Southgate L, Machado RD, Graf S, Morrell NW. Molecular genetic framework underlying pulmonary arterial hypertension. Nat Rev Cardiol. 2020;17:85–95.
Street K, Risso D, Fletcher RB, Das D, Ngai J, Yosef N, Purdom E, Dudoit S. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics. 2018;19:477.
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–50.
Sun D, Wang Y, Liu Q, Wang T, Li P, Jiang T, Dai L, Jia L, Zhao W, Cheng Z. Prediction of long-term mortality by using machine learning models in Chinese patients with connective tissue disease-associated interstitial lung disease. Respir Res. 2022;23:4.
Suzuki T, Carrier EJ, Talati MH, Rathinasabapathy A, Chen X, Nishimura R, Tada Y, Tatsumi K, West J. Isolation and characterization of endothelial-to-mesenchymal transition cells in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol. 2018;314:L118–26.
Szpakowicz A, Szpakowicz M, Lapinska M, Paniczko M, Lawicki S, Raczkowski A, Kondraciuk M, Sawicka E, Chlabicz M, Kozuch M, et al. Serum Chemerin concentration is associated with proinflammatory status in chronic coronary syndrome. Biomolecules. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom11081149.
Tang S, Liu Y, Liu B. Integrated bioinformatics analysis reveals marker genes and immune infiltration for pulmonary arterial hypertension. Sci Rep. 2022;12:10154.
Vachiery JL, Gaine S. Challenges in the diagnosis and treatment of pulmonary arterial hypertension. Eur Respir Rev. 2012;21:313–20.
Volovici V, Syn NL, Ercole A, Zhao JJ, Liu N. Steps to avoid overuse and misuse of machine learning in clinical research. Nat Med. 2022;28:1996–9.
Wang C, Xing Y, Zhang J, He M, Dong J, Chen S, Wu H, Huang HY, Chou CH, Bai L, et al. MED1 regulates BMP/TGF-beta in endothelium: implication for pulmonary hypertension. Circ Res. 2022;131:828–41.
Wu Y, Yang S, Ma J, Chen Z, Song G, Rao D, Cheng Y, Huang S, Liu Y, Jiang S, et al. Spatiotemporal immune landscape of colorectal cancer liver metastasis at single-cell level. Cancer Discov. 2022;12:134–53.
Xie Y, Liu L. Role of Chemerin/ChemR23 axis as an emerging therapeutic perspective on obesity-related vascular dysfunction. J Transl Med. 2022;20:141.
Xue C, Senchanthisai S, Sowden M, Pang J, White RJ, Berk BC. Endothelial-to-mesenchymal transition and inflammation play key roles in cyclophilin a-induced pulmonary arterial hypertension. Hypertension. 2020;76:1113–23.
Yao M, Zhang C, Gao C, Wang Q, Dai M, Yue R, Sun W, Liang W, Zheng Z. Exploration of the shared gene signatures and molecular mechanisms between systemic lupus erythematosus and pulmonary arterial hypertension: evidence from transcriptome data. Front Immunol. 2021;12: 658341.
Zhang L, Yu X, Zheng L, Zhang Y, Li Y, Fang Q, Gao R, Kang B, Zhang Q, Huang JY, et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature. 2018;564:268–72.
Zhang G, Liu Z, Deng J, Liu L, Li Y, Weng S, Guo C, Zhou Z, Zhang L, Wang X, et al. Smooth muscle cell fate decisions decipher a high-resolution heterogeneity within atherosclerosis molecular subtypes. J Transl Med. 2022;20:568.
Zhang G, Cui X, Qin Z, Wang Z, Lu Y, Xu Y, Xu S, Tang L, Zhang L, Liu G, et al. Atherosclerotic plaque vulnerability quantification system for clinical and biological interpretability. iScience. 2023a;26:107587.
Zhang R, Zhang J, Zhang YL, Gong SG, Zhao QH, Wang XJ, Zhao JY, Jiang R, Qiu HL, Li HT, et al. Single-cell transcriptome analysis of peripheral neutrophils from patients with idiopathic pulmonary arterial hypertension. Hypertension. 2023b;80:1784–94.
Zhang S, Zhang G, Wang W, Guo SB, Zhang P, Wang F, Zhou Q, Zhou Z, Wang Y, Sun H, et al. An assessment system for clinical and biological interpretability in ulcerative colitis. Aging. 2024;16:3856–79.
Zhong Y, Yu PB. Decoding the link between inflammation and pulmonary arterial hypertension. Circulation. 2022;146:1023–5.
Zhou X, Tao Y, Chen Y, Xu W, Qian Z, Lu X. Serum Chemerin as a novel prognostic indicator in chronic heart failure. J Am Heart Assoc. 2019;8: e012091.
Acknowledgements
Not applicable.
Funding
This work was funded by National Natural Science Foundation of China (82170037) and the Medical Science and Technology Research Project of Henan Province (LHGJ20230169).
Author information
Authors and Affiliations
Contributions
Z.C, and R.W. contributed study design and paper revising. Y.L, T.J., P.L. and Y.W. contributed project oversight. G.Z. and R.W. contributed data analysis, visualization, and paper writing. M.G. and L.Q. conducted experiments. K.W., Y.L., and Z.H. contributed paper revisiting. All authors approved this manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not required.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
10020_2024_963_MOESM2_ESM.jpg
Additional file 2: Figure S2: (A) UMAP plot showed 21 identified cell clusters. (B) Dot plots of canonical marker genes.
10020_2024_963_MOESM3_ESM.jpg
Additional file 3: Figure S3: Boxplots depicted the percentage of the distinct cell types between PAH and control groups.
10020_2024_963_MOESM4_ESM.jpg
Additional file 4: Figure S4: (A) Circle plot displayed the number of interactions. Red lines indicated increased communication in PAH compared with control group, while blue lines represented decreased communication. The line thickness was proportional to unique ligand-receptor interactions. Autocrine circuits were symbolized by loops. (B) Enriched pathways for three EC subsets. (C-D) Cell-cell communications analysis for EC1 and EC3.
10020_2024_963_MOESM5_ESM.jpg
Additional file 5: Figure S5: (A) Metabolism activity for three EC subsets. (B) Respective ROC curve for PAH distinguishment of ETPGs.
10020_2024_963_MOESM6_ESM.jpg
Additional file 6: Figure S6: (A) Barplot showed mRNA expression levels of RARRES2, TC2N, CBY1, CKS1B, MSRB3, and SMAGP in hypoxia-induced human PAECs and control. (B) Hypoxic PAH mice lung paraffin sections were stained with chemerin (green) and CD31 (red), and were observed by immunofluorescence microscopy. DAPI staining was used to visualize the nuclear areas. The scale bar represents 20 μm.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Wu, R., Zhang, G., Guo, M. et al. Assessing personalized molecular portraits underlying endothelial-to-mesenchymal transition within pulmonary arterial hypertension. Mol Med 30, 189 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-024-00963-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-024-00963-z