Introduction
Pulmonary arterial hypertension (PAH) is a condition characterized by progressive remodeling of the pulmonary arteries leading to increased pressure that if left untreated, results in right heart failure and death [1]. Underlying causes of PAH include heritable factors, connective tissue diseases (CTD), drugs and toxins, portal hypertension, congenital heart diseases, and human immunodeficiency virus. There is also a form without clear etiology named idiopathic PAH (iPAH) [2]. The most common etiologies of PAH are iPAH and PAH-associated with CTD [3,4]. The pathobiology of PAH includes endothelial dysfunction, smooth muscle cell hypertrophy and proliferation, inflammation, metabolic abnormalities, and thrombosis [5–7].
The prognosis of the disease depends on risk assessment and prompt initiation of PAH-specific therapies. Despite effective treatments for PAH the median survival is around 7 years [8]. There is a growing assortment of drugs to treat PAH with limited data to select specific agents. Treatment selection for PAH is currently based on severity of the disease and not on patient-specific molecular defects [2]. Except in the presence of comorbidities, current guidelines indicate dual or triple combination therapy for the treatment of PAH [2]. The recent approval of sotatercept, an agent with a novel mechanism of action, adds a different treatment pathway [9] which further extends the treatment approach [10–12]. A clear understanding of the mechanisms of disease and their therapeutic modification has been the goal in precision medicine [13–15]. Although there has been great progress in understanding of the pathobiology of PAH, mechanistic research has been restricted by critical factors. For instance, animal models cannot replicate the complex and heterogeneous pathogenesis of the human disease [16,17]. This is especially true in iPAH where the cause of the disease is unknown.
Based on the animal model limitations, it remains necessary to carry out studies using biological samples from PAH patients. Cellular material from patients with PAH is primarily obtained from lungs recovered at the time of autopsies or transplantation, which by their nature represent late stages of the disease [18,19]. These cells are of limited supply and may not reflect events that initiate or drive the disease given the potential alteration in their biological responses due to genetic instability, environmental stimuli, and treatment effects [20,21]. There have been attempts to obtain endothelial cells from PAH patients at earlier stages of the disease, including the a) recovery from balloons of pulmonary artery catheters or b) transformation from induced pluripotent stem cells (iPSCs) generated from fibroblast isolated from skin biopsies. These processes can be expensive and technically challenging [22–29], and in the case of iPSC‘s they can introduce new mutations and alter gene expression profiles.
Here we demonstrate a simple method to isolate and culture circulating endothelial cells (CECs) from pulmonary arterial blood obtained at the time of right heart catheterization (RHC) in PAH patients. We tested these endothelial cells using transcriptomics and found that they have the characteristic changes observed in PAH patients, including specific abnormalities in PAH treatment pathways, i.e. nitric oxide, endothelin, prostacyclin and BMP/activin pathways. Furthermore, CECs carried other abnormalities that could advance our understanding of the disease process. Our approach yields CECs with good recovery rates supporting the use of this cellular model for the ex-vivo mechanistic evaluation of critical PAH pathways that participate in the pathogenesis of the disease [30].
Methods
Patient selection
We included patients with iPAH or hPAH who underwent RHC for evaluation of PAH and signed the informed consent to participate in our Cleveland Clinic IRB approved study (# 06–245). All blood samples were collected between May 2018 and September 2019. All patients had precapillary PAH, defined as mPAP > 20 mmHg, PAWP ≥ 15 mmHg and PVR ≥ 3 Wood units, based on proceedings of the 6th World Symposium on PH [31]. Patients with iPAH had no identifiable cause that could explain the precapillary PH and all patients with hPAH had either a known genetical abnormality or a first-degree family member with PAH without a clear etiology [2,31].
Circulating endothelial cell isolation
At the time of RHC, we obtained 10–20 mL of blood in BD Vacutainer® EDTA tubes (BD bioscience, Franklin lakes, NJ) from the distal port of the pulmonary artery catheter, located in the right or left main pulmonary artery. An equal volume of 2% fetal bovine serum (FBS) in phosphate buffered saline (PBS) was added to the blood and the mixture was layered onto 15ml of Ficoll-Paque Plus (Cytiva, Wilmington DE). The sample was centrifuged at 19°C for 30 mins at 740g with maximum acceleration but minimum brake. After centrifugation, the top plasma layer was aspirated and discarded. The buffy coat was then transferred to a new tube, where 14 ml of 2% FBS in PBS was added. This sample was then centrifuged at 19°C for 5 mins at 500g. The supernatant was aspirated and discarded. The cell pellet was resuspended with 5ml 2% FBS in PBS and centrifuged at room temperature for 5 mins at 500g. The supernatant was aspirated, and the pellet resuspended in 2ml of endothelial cell media (EGM-2; Lonza). This sample was then placed onto rat tail collagen (BD bioscience, Franklin lakes, NJ) coated dishes. These plates were prepared by adding 2ml 50ug/ml of rat tail collagen in 0.02N acetic acid into 35mm dishes for 2hrs. After which they were washed twice in PBS and then used to plate isolated cells. The media on the cells was replaced after 24 hours and changed every day for 7 days and then every 48hrs thereafter, until clones appeared (14–21 days). At this stage the only cells that grow in the plates are endothelial cells, as validated by endothelial cell marker, CD31. No further sorting was required to isolate the endothelial cells. After clones appeared and grew sufficiently, they were expanded, used, or frozen for storage.
Circulating endothelial cell validation
CECs phenotype was confirmed by the endothelial cell-specific marker CD31 (1:30 dilution; Dako, Glostrup, Denmark). Fluorescence-activated cell sorting (FACS) for CD31 expression (Becton Dickinson, San Jose, Calif.) was used as previously described [32]. Cells were also tested for 2D tube formation using an angiogenesis assay kit (#ECM625, Millipore) using approximately 80k cells/well.
RNA isolation from endothelial cells
All endothelial cells were grown on fibronectin coated plates (50ng/ml) using endothelial cell media, (EGM-2, Lonza Walkerville, MD). Selected cells were passage 5 or 6. Control endothelial cells (cPAEC) from non-PAH subjects were purchased from Lonza. Cells from Lonza were derived from pulmonary arteries from donated lungs, after given permission for their use in research applications by informed consent or legal authorization. When cells were confluent, they were harvested using RNA isolation lysis buffer (RTL buffer + 10ul 2-Mercaptoethanol/ml; Qiagen). RNA was isolated using the RNeasy kit (#74104, Qiagen) and quantitated via NanoDrop and Qubit Assay. The quality of the RNA was assessed using RNA Integrity Number using a bioanalyzer assay.
RNA sequencing and transcriptomic analyses of endothelial cells
Upon passing quality control, Novogene created sequencing libraries using NEBNext Ultra RNA library prep kit for Illumina (NEB, USA). Between 200ng-1ug RNA was used as input material and libraries were created following the manufacturer’s protocols. Briefly, mRNA was purified from total RNA using poly-T oligos attached to magnetic beads. Fragmentation was carried out using divalent cations under elevated temps in NEBNext first strand synthesis reaction buffer. First strand synthesis was carried out using random hexamer oligos primers and M-MuLV reverse transcriptase. Second strand synthesis was performed using DNA polymerase I and RNAse H and overhangs were converted to blunt ends using exonuclease/polymerase activity. After adenylation of the DNA fragments, NEBnext adaptors with hairpin loop structures were ligated to prepare for hybridization. cDNA fragments of 150bp-200bps were purified using AMPure XF system (Beckman Coulter, Beverly, USA). Then USER enzyme was used with the size selected cDNA at 37°C followed by 5 mins at 95°C before PCR. The PCR was performed with Phusion High- Fidelity DNA polymerase, universal PCR primers and index primers. PCR products were then purified using the AMPure XP system and library quantity assessed on the Agilent Bioanalyzer 2100 system. Original image data files from high-throughput sequencing platforms are available in the NCBI Gene Expression Omnibus (GEO) data repository (accession number GSE277919).
Transcriptomic analyses of endothelial cells
Clustering and sequencing: Clustering of the index-coded samples was performed using a cBot Cluster Generation System on the PE Cluster Kit cBot-HS (Illumina) according to the manufacturers.
Data analysis-quality control: Raw data of FASTQ format were processed through fastp. Clean data were obtained by removing reads with adapter and poly-N sequences and reads with low quality from raw data (S1–S5 Tables). At the same time, Q20, Q30 and GC content of the clean data were calculated (S1 Table). All the downstream analyses were done on the high-quality clean data. Detailed workflow of RNA sequencing is presented in the (S1 File).
Mapping to reference genome: Reference genome and gene model annotation files were downloaded from the genome browser website (NCBI/UCSC/Ensembl). Clean reads of paired end were aligned to the reference genome using the Spliced Transcripts Alignment to a Reference (STAR) software, that uses sequential maximum mappable seed search in an uncompressed suffix array followed by a seed clustering and stitching procedure. Summary of mapping data for fragment is shown in S3 and S4 Tables.
Quantification of transcripts: Feature Counts were used to calculate the read numbers mapped of each gene. Then Reads Per Kilobase (RPKM) of each gene was calculated based on the length of the gene and reads count mapped to that gene. The summary of the gene expression level analysis (FPKM) is shown in S5 and S6 Tables.
Differential expression analysis: Comparisons were made between CECs and a) control pulmonary artery lung endothelial cells (cPAECs), as well as b) pulmonary lung endothelial cells from PAH patients isolated from patient lungs (LECs). Differential expression analysis between groups were performed using the DESeq2 R package, which provides a model based on the negative binomial distribution. The resulting p values were adjusted using the Benjamini and Hochberg’s approach for controlling the False Discovery Rate (FDR). Genes with an adjusted p value < 0.05 by DESeq2 were assigned as differentially expressed.
Ingenuity Pathway Analysis (IPA)
Core analysis was performed using IPA for DEGs with adjusted p value (padj) of <0.05 and log2 fold change of ≥ 2. The results of the core analysis for the CECs vs cPAECs were used for comparative analysis. Upstream regulators, downstream analysis, causal networks, and GO analysis was done by the QIAGEN IPA software.
Results
A) Isolation of CECs
During RHC, blood was collected from the pulmonary artery and processed as shown in Fig 1A. Representatives images of clones are illustrated in Fig 1B. These cells were CD31+ (Table 1) and produced 2D tubes on Matrigel confirming their endothelial cell behavior. We successfully generated CECs from ~ 60% of patients sampled.
[Figure omitted. See PDF.]
Panel A diagrammatic illustration of the CEC isolation procedure. Image was created using BioRender.com. Buffy coats from blood are isolated using a Ficoll-paque gradient and after twice washing cells in FBS/PBS, plated onto collagen coated plates. Clones generally appear after day 14–20. Panel B shows representative images of CEC clones.
[Figure omitted. See PDF.]
B) Transcriptomic analysis of CECs and LECs in PAH patients
We compared eight CECs and five LECs to five cPAEC (Table 1). When compared to cPAECs, we observed 566 differentially expressed genes (DEGs) that were significantly altered two-fold in CECs (padj<0.05), while 919 genes were altered two-fold in LECs and 206 were common in both groups, (Fig 2A). In CECs, 426 genes were upregulated, and 140 genes were downregulated, while, in LECs, 670 genes were upregulated, and 249 genes were downregulated. A volcano plot shows the top 30 genes that are altered in CECs (Fig 2B), with many involved in signaling, proliferation and clotting. A hierarchical clustered heat map of DEGs from CECs and LECs shows difference between cPAEC (Fig 3).
[Figure omitted. See PDF.]
Panel A shows a Venn diagram displaying differentially expressed genes (DEGs) in CECs and LECs compared to cPAECs. We found an overlap in 206 genes between CECs and LECs supporting the similarity between the cell lines. Panel B represents a Volcano plot showing the distribution of DEGs between CECs vs cPAECs. The top 30 altered DEGs are labeled.
[Figure omitted. See PDF.]
CECs and LECs from PAH patients were compared to cPAECs. Using hierarchical clustering, we show that DEG shows similarities between CECs and LECs, while revealing differences with cPAECs. Genes that were significantly different (pAdj<0.05) and with a Log2FC>2 were used to generate a heat map. Hues of red color reflect DEG upregulation, while shades of blue color reflect downregulation.
C) Differential expression of endothelial cells markers on isolated cells
To verify that the isolated cells were of endothelial origin, we checked the endothelial cell markers expression in CECs/LECs vs cPAECs. No statistical difference was observed between CECs or LECs in the pan-endothelial cell markers or arterial endothelial cells markers (Table 2). Due to low expression and variability of some genes, the fold differences may have been high but not significant. Data on the expression of genes shown here are provided in S7 Table. Interestingly, vasoactive intestinal peptide receptor 1 (VIPR1) (Table 2), a marker for lung specific endothelial cells know to be downregulated in PAH patients [33], was greatly reduced in CECs (log2FC = -4.21, pAdj = 3.09E-05) and in LECs (log2FC = -0.7582 pAdj = 0.21).
[Figure omitted. See PDF.]
D) Differential gene expression between circulating endothelial cells and PAH Lung endothelial cells
The top 20 genes that are up- and down-regulated in CECs and LECs are shown in Table 3. The most down-regulated genes in both CECs and LECs were the transcription factors GATA4 (CECs- Log2FC = -10.94, pAdj = 5.77E-13; LECs- Log2FC = -11.02, pAdj = 3.09E-11) and heart and neural crest derivatives expressed 2 (HAND2: CECs- Log2FC = -9.90, pAdj = 1.01E-25; LECs- Log2FC = -9.98, pAdj = 6.06E-19). These two proteins are involved in embryonic development and adult cell homeostasis. Defects in HAND2 can cause pathological cardiac remodeling [34–36]. HAND2 also interacts with NK2 transcription factor related locus 5 (NKX2-5) [37], which was also decreased in CECs (Log2FC = -5.51, pAdj = 1.18E-05 and LECs (Log2FC = -6.90, pAdj = 1.44E-08). Both GATA4 and NKX2-5 were also found to be reduced in endothelial cells isolated from pulmonary artery catheters balloons of PAH patients [23].
[Figure omitted. See PDF.]
E) Gene Ontology analysis on differentially expressed genes in circulating endothelial cells
Functional pathway enrichment for DEGs with greater than 2-fold change was carried out in CECs and LECs using Gene Ontology (GO) analysis. Results from the top biological and cellular processes are presented in Table 4. For both CECs and LECs, biological processes enrichment showed that the processes predominantly affected were angiogenesis, cell growth, blood circulation, and heart development, which are important in the pathobiology of PAH. In the molecular function analysis, the receptor regulatory activity involved in PAH was enriched in both CECs and PAH PAECs.
[Figure omitted. See PDF.]
F) Upstream regulator analysis
We analyzed upstream regulators which can identify transcriptional factors affecting the DEG. In CECs, the top upstream regulators of DEG were tumor necrosis factor (TNF; Z score = 7.84), interferon ƴ (IFNG; Z score = 6.26), thrombin (F2; Z score = 5.79) and transforming growth factor β1 (TGFB1; Z score = 5.44). Proteins known to play a significant role in PAH such as epidermal growth factor, SMAD3 (Z score = 4.48), endothelin 1 (EDN1- Z score = 4.34), hepatocyte growth factor (Z score = 4.19), and vascular endothelial growth factor A (VEGFA; Z score = 4.11) (Table 5) were activated [38–42].
[Figure omitted. See PDF.]
G) Causal network analysis
Causal network analysis demonstrated that the top master regulator was activin A with a depth of 3 (Z score = 7.63; pval of overlap = 1.64E-36). Activin A is elevated in PAH and downregulates BMPR2 by causing its degradation (Table 6) [43,44]. We also observed an increase in activin A (INHBA) transcription in CECs (log2FC = 2.53, pAdj = 0.04) and LECs (log2FC = 2.09, pAdj = 0.09).
[Figure omitted. See PDF.]
H) Alterations of genes important in PAH treatment pathways
Several vasodilator treatments are available in PAH that target nitric oxide, prostacyclin, endothelin and activin pathways. We investigated if defects in these pathways are present in CECs or LECs.
1. 1) Nitric oxide pathway: In both CECs and LECs, we found no significant difference in gene expression of the endothelial form of nitric oxide synthase (eNOS) (NOS3); however, cellular levels of eNOS are regulated by other factors such as CAV1 which stabilizes eNOS and correctly localize it into caveolae [45,46]. We found lower levels of cav-1 in CECs (Log2FC = -0.57, pAdj = 0.07) and LECs (log2FC = -0.86, pAdj = 1.49E-8), that would lead to decreased stability of eNOS, and/or decreased phosphorylation needed for eNOS activity [46–49].
Critical downstream components of NO signaling pathway include phosphodiesterase’s (PDEs). We found higher levels of PDE5 in both CECs (log2FC = 1.68, pAdj = 0.003) and LECs (log2 FC = 1.09, pAdj = 0.02) which are responsible for down regulating NO signaling [50,51]. In addition, we saw differences in other PDE enzymes that play a role in this and other signaling pathways [50,52,53], including, increased levels of PDE10A in CECs (log2FC = 1. 1.83, pAdj = 3.601E-07) and LECs (log2 FC = 0.85, pAdj = 0.009), and decreased levels of PDE3A in CECs (log2FC = -1.34, pAdj = 0.06) and LECs (log2FC = -1.25, pAdj = 0.0003) and PDE7B in CECs (log2FC = -1.28, pAdj = 0.04) and LECs (log2FC = -0.36, pAdj = 0.31) (Table 7).
[Figure omitted. See PDF.]
1. 2) Prostacyclin pathway: We found that CECs have reduced gene expression of the prostacyclin receptor (PTGIR) (log2FC = -1.46, pAdj = 0.02), consistent with the reduced levels observed in PAH patients [54]. Other prostaglandin receptors are also altered in PAH [55] and we noted that in both CECs and LECs there was increased expression of the PGE2 receptor, EP4 (PTGER4) in CECs (log2FC = 1.43, pAdj = 0.02) and LECs (log2 FC = 1.30, pAdj = 0.005). EP1 (PTGER1), another PGE2 receptor, was elevated over 11-fold in LECs (Log2FC = 3.52, pAdj = 0.001) while it was unchanged in CECs, possibly suggesting patient or disease stage differences. We saw an increased levels of the thromboxane A2 receptor (TBXA2R) in CECs (log2FC = 2.36, pAdj = 0.014) and LECs (log2 FC = 1.601, pAdj = 0.003). Thromboxane A2 is prostanoid that causes vasoconstriction and is elevated in PAH [56]. In CECs we saw an increase in Cox-2 expression (PTGS2; log2FC = 3.63, pAdj = 0.008). Over expression of Cox-2, which synthesized many different prostanoids under inflammatory conditions, leads to decreased apoptosis by elevating anti-apoptotic factors such as Bcl-2 [57–59] and increased neoangiogenesis [60]. We identified an increase in anti-apoptotic protein Bcl-2 (Log2FC = 1.97. pAdj = 5.99E-07) in CECs suggesting a mechanism for the decreased apoptosis seen in PAH.
2. 3) TGF/Activin pathway: Defects in the BMPR2 signaling pathway have also been recognized in iPAH [61,62]. We did not see a decreased expression of BMPR2 in CECs but did in LECs (log2FC = -0.70, pAdj = 0.03). We have previously shown that signaling through BMPR2 is downregulated by SMAD specific E3 ubiquitin protein ligase (SMURF) that increases the degradation of the mediator SMAD 1/5/8 [61,63–65]. We found that levels of SMURF1 and 2 are increased in CECs (SMURF1; log2FC = 0.52, pAdj = 0.07: SMURF2; log2FC = 1.57, pAdj = 0.0004) and LECs (SMURF1; log2FC = 0.59, pAdj = 0.0009: SMURF2; log2FC = 1.01, pAdj = 0.02).
Higher plasma levels of activin A are present in PAH patients [43]. We found increased expression of the activin A (INHBA) in both CECs (log2FC = 2.53, pAdj = 0.04) and LECs (log2FC = 2.10, pAdj = 0.09). Noggin (NOG), a downstream product of the activin pathway, sequesters BMPs and suppresses signaling through this pathway. We found higher levels of NOG in CECs (log2FC = 4.37, pAdj = 0.02) and LECs (log2FC = 2.43, pAdj = 0.04). Twisted gastrulation BMP signaling modulator 1 (TWSG1) is another inhibitor of BMPs that is increased in CECs (log2FC = 0.905, pAdj = 0.031) [66]. Additionally, ALK5 (TGFBR1) which is a co-receptor for activin signaling was elevated in CECs (log2FC = 1.50, pAdj = 0.007) which could enhance signaling through that pathway.
1. 4) Gene expression alteration in other key PAH pathways.
Many genes have been found to play a role in the pathogenesis of PAH. We saw ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1), a regulator of the pulmonary vasculature, was reduced in both CECs (log2FC = -1.41, pAdj = 0.02) and LECs (log2FC = -1.10, pAdj = 0.02). ENTPD1 is reduced in the pulmonary artery endothelium of patients with iPAH [67].
Interleukin-6,-7, -8, and CXCL1 are potent cytokines that play an important role in PAH [33,42,68,69]. We saw increased expression of the interleukin-8 (IL-8; CXCL8) in CECs (log2FC = 3.75, pAdj = 0.006) and LECs (log2 FC = 2.40, pAdj = 0.02) and elevated IL-6 in LECs (log2 FC = 1.54, pAdj = 0.02). CXCL1 was increased in CECs (log2FC = 1.77, pAdj = 0.007) and LECs (log2FC = 1.98, pAdj = 3.59E-05). IL7R, the receptor for IL-7, which is elevated in PAH [33,69], was increased in both CECs (log2FC = 4.08, pAdj = 0.0001) and LECs (log2FC = 2.38, pAdj = 0.0005) compared to cPAECs.
Dysfunction Wnt signaling is implicated in PAH [70] as reduced WNT7 or WNT5B expression leads to abnormal angiogenesis [71]. DKK1 and DKK3 are negative regulators of the Wnt pathway and were elevated in CECs (DKK3: log2FC = 1.77, pAdj = 3.42E-05; DKK1: log2FC = 2.67, pAdj = 0.0004) and LECs (DKK3: log2FC = 0.98, pAdj = 0.0001; DKK1: log2FC = 1.04, pAdj = 0.06). NEDD9 is elevated in plasma of PAH patients, promoting endothelial fibrosis [72,73]. We found that NEDD9 levels were elevated in CECs (log2FC = 2.31, pAdj = 0.003) and LECs (log2FC = 1.54, pAdj = 5.75E-06).
Discussion
In the present study we demonstrate that CECs have the characteristic pathway abnormalities seen in PAH and constitute a good model to test relevant disease processes in individual patients. When compared to LECs from PAH patients isolated at end-stage disease, CECs have a smaller number of DEGs with important similarities but some differences. Using our methodology, CECs were relatively easy to obtain from small amounts of blood, with the ability to culture them ~ 60% patients. This initial step for establishing an ex-vivo cell model for PAH is critical to test critical PAH pathways that can help understand the pathophysiology of the disease and move us closer to the optimal goal of personalized medicine in PAH.
Other approaches to generate endothelial cells from PAH patients involve generation of iPSCs or isolation from the balloon of pulmonary artery catheters [22–29]. Generation of iPSCs requires virus transformation of skin fibroblasts taken by biopsy. These iPSC are then transformed into endothelial cells using a mixture of different growth factors [27–29,74–77]. This process is lengthy, costly, and technically challenging and may not completely mimic the endothelial cells present in PAH patients as the process may introduce mutations and/or alter cellular transcriptomics/phenotype. There is also the possibility of lingering presence of viral fragments and the need for specialized viral handling space. However, it would be useful to make the comparison with endothelial cells generated by iPSCs, CECs and LECs to determine the usefulness in advancing PAH research.
The other alternative to isolate endothelial cells from the balloon of pulmonary artery catheter. Unfortunately, we were unsuccessful in recovering endothelial cells using this methodology, despite many modifications of our protocol (changing manufacturer of pulmonary artery catheter, longer or recurrent balloon inflation, using different media and temperature for transport, use of enzymatic product to dislodge cells from the balloon, collection with residual blood, balloon inflation while in the media, etc.). This led us to develop an alternative approach to isolate endothelial cells from PAH patients. CECs isolated by our methodology may be similar as endothelial cells isolated from the pulmonary artery balloon by other investigators, but this would require further investigation.
Although many of the important genes involved in PAH are similar in CECs and LECs, differences between them may be due to several factors, 1) number of samples analyzed with heterogeneity in disease pathways among PAH patients, 2) cells were obtained at different stages of the disease, as LECs reflect end-stage disease process that could be affected by additive mutations, epigenetics and PAH treatments, 3) cells may be different subtypes or diverse circulations [78]. Although we isolated our CEC from blood obtained from the pulmonary artery, we cannot be certain that our CECs originate in the pulmonary circulation. It is likely that PAH specific pathway abnormalities are present in a variety of endothelial cells and not just those originating from the pulmonary circulation. There is growing supports that PAH is a systemic disease [79], therefore even if our CECs are from another circulation, they clearly still manifest specific PAH pathway abnormalities, and therefore are of great value.
We have established that the CECs contain many of the defects characteristics of PAH and can be used to study the different aspects of the disease. These include pathways abnormalities involving the vascular tone, inflammation, angiogenesis, fibrosis, and thrombosis. Isolation of CECs will allow for deeper understanding of the pathogenesis of PAH and can be used as models to determine the interpatient variability. In CECs we see many of the genes that have been found to altered in PAH and are not current targets for drugs, including the Wnt pathway (DKK1, DKK3), plurinucleate pathways (ENTPD1), and the inflammatory pathways (IL6, 7, 8, CXCL1). CECs may provide a critical substrate to understand the interplay between known and novel PAH pathways.
The CECs would also greatly aid in achieving the goal of precision medicine in PAH, since current and future treatment pathways can be thoroughly assessed ex-vivo. This unique investigation can provide valuable guidance to determine the best treatment/s in every patient. Decades of research in PAH have produced many PAH-specific medications, that predominantly act at different points of the four disease pathways (nitric oxide, prostacyclin, endothelin and BMP/activin pathways). Current treatment guidelines direct PAH treatment selection based on risk stratification [2]. More than ever, we need to develop tools to base PAH treatment on mechanistic information and not only risk of complications. This patient specific treatment selection has the potential to reduce costs, minimize drug interactions and side effects, while improving the quality of life of our PAH patients [10–12,80].
All current treatment pathways, besides endothelin, were identified in our CECs and showed the alterations expected in the disease. In the NO pathway, we observed reduced levels of CAV1 and increased expression of PDE5A which reduces the NO signaling. In the prostacyclin pathway we noted a decrease in PTGIR and increase in EP4, EP1, thromboxane receptor and Cox-2; affecting the prostacyclin signaling. In the BMP/activin pathway we found factors that decrease BMP signaling, i.e. SMURF1 and 2, and noggin, as well as factors that increase activin signaling, i.e. activin A, and ALK5. As we have previously shown, there is heterogeneity in the pathways involved, which is essential to make informed treatment decisions. For example, prostacyclin agonists target the PGI2 but also activate several other prostaglandin receptors [67] (PTGIR, EP1, EP2, and EP4), which may impact treatment response and side effects of the prostacyclin analogue used.
Our study has limitations including a) transcriptomic analysis was performed in a small number of samples, potentially not detecting certain molecular differences between cells, and under recognizing interpatient variability, b) the origin of our CECs cannot be fully established since they were isolated from blood taken from the pulmonary catheter during RHC and there are no biological markers that would definitely attribute them to the pulmonary circulation, c) since the cellular isolation techniques are dissimilar, some of the differences observed may be due to the methodology used; however, we are focusing not only on the differences among cell lines, but predominantly on similarities regarding PH pathway abnormalities, d) culture of these CECs is only possible in ~60% of the cases, e) the endothelin pathway could not be evaluated in CECs, and f) no functional studies were performed, but are currently planned for the next stages of this research. Regardless of these limitations, this is the first study that comprehensively tested CECs isolated from a small amount of blood obtained from the pulmonary artery at the time of RHC in patients with PAH. We have shown significant differences between CECs and cPAECs in the characteristic PAH pathways. Even if the origin of our CECs is not completely clear, they carry characteristic PAH abnormalities, making them a useful cellular model to study PAH. We are currently making modifications to our protocol to increase the CECs recovery rate.
Conclusions
In conclusion, we have described a simple method to isolate CECs from pulmonary arterial blood, providing an excellent opportunity to investigate abnormalities in PAH specific pathways, identify new pathways responsible for the pathogenesis of the disease, and guide therapeutic decisions, moving the field towards the goal of precision medicine in PAH. For the first time, this approach will allow us to determine possible temporal changes associated with the disease process, discerning events that happen at early versus late stages of PAH.
Supporting information
S1 Table. Data quality summary.
Data quality summary for the raw and clean Reads for the RNA-seq.
https://doi.org/10.1371/journal.pone.0312535.s001
(XLSX)
S2 Table. Classification of raw reads.
Classification of the raw reads for the CEC, LEC and cPAEC for RNA-seq data.
https://doi.org/10.1371/journal.pone.0312535.s002
(XLSX)
S3 Table. MapStat summary.
Summary of the total reads one ones mapped to unique or multiple sites.
https://doi.org/10.1371/journal.pone.0312535.s003
(XLSX)
S4 Table. Distribution of sequencing reads of all samples to exons, introns and intergenic regions.
Distribution of sequencing reads to exons, introns and intergenic regions for all the CEC, LEC and cPAEC samples undergoing RNA-seq analysis.
https://doi.org/10.1371/journal.pone.0312535.s004
(XLSX)
S5 Table. CEC FPKM data.
FPKM data for all genes found in CEC.
https://doi.org/10.1371/journal.pone.0312535.s005
(XLSX)
S6 Table. LEC FPKM data.
FPKM data for all genes found in LEC.
https://doi.org/10.1371/journal.pone.0312535.s006
(XLSX)
S7 Table. Endothelial cells FPKM data for CEC and LEC.
FPKM data Specific for the endothelial cell markers.
https://doi.org/10.1371/journal.pone.0312535.s007
(XLSX)
S1 File. Supplemental methods.
Addition information about the processing of the RNA seq data.
https://doi.org/10.1371/journal.pone.0312535.s008
(DOCX)
References
1. 1. Tonelli AR, Arelli V, Minai OA, Newman J, Bair N, Heresi GA, et al. Causes and circumstances of death in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2013;188(3):365–9. pmid:23600433
* View Article
* PubMed/NCBI
* Google Scholar
2. 2. Humbert M, Kovacs G, Hoeper MM, Badagliacca R, Berger RMF, Brida M, et al. 2022 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 2023;61(1).
* View Article
* Google Scholar
3. 3. Farber HW, Miller DP, Poms AD, Badesch DB, Frost AE, Muros-Le Rouzic E, et al. Five-Year outcomes of patients enrolled in the REVEAL Registry. Chest. 2015;148(4):1043–54. pmid:26066077
* View Article
* PubMed/NCBI
* Google Scholar
4. 4. McGoon MD, Miller DP. REVEAL: a contemporary US pulmonary arterial hypertension registry. Eur Respir Rev. 2012;21(123):8–18. pmid:22379169
* View Article
* PubMed/NCBI
* Google Scholar
5. 5. Hassoun PM, Mouthon L, Barbera JA, Eddahibi S, Flores SC, Grimminger F, et al. Inflammation, growth factors, and pulmonary vascular remodeling. J Am Coll Cardiol. 2009;54(1 Suppl):S10–S9. pmid:19555853
* View Article
* PubMed/NCBI
* Google Scholar
6. 6. Humbert M, Guignabert C, Bonnet S, Dorfmuller P, Klinger JR, Nicolls MR, et al. Pathology and pathobiology of pulmonary hypertension: state of the art and research perspectives. Eur Respir J. 2019;53(1). pmid:30545970
* View Article
* PubMed/NCBI
* Google Scholar
7. 7. Rajagopal S, Yu YA. The Pathobiology of Pulmonary Arterial Hypertension. Cardiol Clin. 2022;40(1):1–12. pmid:34809910
* View Article
* PubMed/NCBI
* Google Scholar
8. 8. Benza RL, Miller DP, Barst RJ, Badesch DB, Frost AE, McGoon MD. An evaluation of long-term survival from time of diagnosis in pulmonary arterial hypertension from the REVEAL Registry. Chest. 2012;142(2):448–56. pmid:22281797
* View Article
* PubMed/NCBI
* Google Scholar
9. 9. Kingwell K. FDA approves Merck & Co’s pulmonary arterial hypertension drug sotatercept. Nat Rev Drug Discov. 2024.
* View Article
* Google Scholar
10. 10. Narechania S, Malesker MA. Drug Interactions Associated With Therapies for Pulmonary Arterial Hypertension. J Pharm Technol. 2022;38(6):349–59. pmid:36311309
* View Article
* PubMed/NCBI
* Google Scholar
11. 11. Ruopp NF, Cockrill BA. Diagnosis and Treatment of Pulmonary Arterial Hypertension: A Review. JAMA. 2022;327(14):1379–91. pmid:35412560
* View Article
* PubMed/NCBI
* Google Scholar
12. 12. Sun X, Chen R, Yao X, Zheng Z, Wang M, Wang C, et al. Treatment of Pulmonary Hypertension: Is Triple Therapy Necessarily Better than Monotherapy? Am J Respir Crit Care Med. 2021;204(12):1492–3. pmid:34672866
* View Article
* PubMed/NCBI
* Google Scholar
13. 13. Gurtu V, Michelakis ED. A Paradigm Shift Is Needed in the Field of Pulmonary Arterial Hypertension for Its Entrance Into the Precision Medicine Era. Circ Res. 2016;119(12):1276–9. pmid:27932471
* View Article
* PubMed/NCBI
* Google Scholar
14. 14. Kanwar MK. Biomarkers in pulmonary arterial hypertension: Moving closer toward precision medicine? J Heart Lung Transplant. 2020;39(4):287–8. pmid:32199588
* View Article
* PubMed/NCBI
* Google Scholar
15. 15. Kelly NJ, Chan SY. Pulmonary Arterial Hypertension: Emerging Principles of Precision Medicine across Basic Science to Clinical Practice. Rev Cardiovasc Med. 2022;23(11). pmid:36875282
* View Article
* PubMed/NCBI
* Google Scholar
16. 16. Boucherat O, Agrawal V, Lawrie A, Bonnet S. The Latest in Animal Models of Pulmonary Hypertension and Right Ventricular Failure. Circ Res. 2022;130(9):1466–86. pmid:35482834
* View Article
* PubMed/NCBI
* Google Scholar
17. 17. Wu XH, Ma JL, Ding D, Ma YJ, Wei YP, Jing ZC. Experimental animal models of pulmonary hypertension: Development and challenges. Animal Model Exp Med. 2022;5(3):207–16. pmid:35333455
* View Article
* PubMed/NCBI
* Google Scholar
18. 18. Comhair SA, Xu W, Mavrakis L, Aldred MA, Asosingh K, Erzurum SC. Human primary lung endothelial cells in culture. Am J Respir Cell Mol Biol. 2012;46(6):723–30. pmid:22427538
* View Article
* PubMed/NCBI
* Google Scholar
19. 19. Kessler S, Rho H, West G, Fiocchi C, Drazba J, de la Motte C. Hyaluronan (HA) deposition precedes and promotes leukocyte recruitment in intestinal inflammation. Clin Transl Sci. 2008;1(1):57–61. pmid:20443819
* View Article
* PubMed/NCBI
* Google Scholar
20. 20. Carethers JM, Pham TT. Mutations of transforming growth factor beta 1 type II receptor, BAX, and insulin-like growth factor II receptor genes in microsatellite unstable cell lines. In Vivo. 2000;14(1):13–20. pmid:10757056
* View Article
* PubMed/NCBI
* Google Scholar
21. 21. Yeager ME, Golpon HA, Voelkel NF, Tuder RM. Microsatellite mutational analysis of endothelial cells within plexiform lesions from patients with familial, pediatric, and sporadic pulmonary hypertension. Chest. 2002;121(3 Suppl):61S. pmid:11893687
* View Article
* PubMed/NCBI
* Google Scholar
22. 22. Passineau MJ, Gallo PH, Williams G, Perez R, Benza RL. Harvest of Endothelial Cells from the Balloon Tips of Swan-Ganz Catheters after Right Heart Catheterization. J Vis Exp. 2019(143). pmid:30735186
* View Article
* PubMed/NCBI
* Google Scholar
23. 23. Singh N, Eickhoff C, Garcia-Agundez A, Bertone P, Paudel SS, Tambe DT, et al. Transcriptional profiles of pulmonary artery endothelial cells in pulmonary hypertension. Sci Rep. 2023;13(1):22534. pmid:38110438
* View Article
* PubMed/NCBI
* Google Scholar
24. 24. Ventetuolo CE, Aliotta JM, Braza J, Chichger H, Dooner M, McGuirl D, et al. Culture of pulmonary artery endothelial cells from pulmonary artery catheter balloon tips: considerations for use in pulmonary vascular disease. Eur Respir J. 2020;55(3). pmid:31949110
* View Article
* PubMed/NCBI
* Google Scholar
25. 25. Pollett JB, Benza RL, Murali S, Shields KJ, Passineau MJ. Harvest of pulmonary artery endothelial cells from patients undergoing right heart catheterization. J Heart Lung Transplant. 2013;32(7):746–9. pmid:23684132
* View Article
* PubMed/NCBI
* Google Scholar
26. 26. Pu X, Du L, Hu Y, Fan Y, Xu Q. Stem/Progenitor Cells and Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol. 2021;41(1):167–78. pmid:33028095
* View Article
* PubMed/NCBI
* Google Scholar
27. 27. Sanchez-Duffhues G, Hiepen C. Human iPSCs as Model Systems for BMP-Related Rare Diseases. Cells. 2023;12(17). pmid:37681932
* View Article
* PubMed/NCBI
* Google Scholar
28. 28. Geti I, Ormiston ML, Rouhani F, Toshner M, Movassagh M, Nichols J, et al. A practical and efficient cellular substrate for the generation of induced pluripotent stem cells from adults: blood-derived endothelial progenitor cells. Stem Cells Transl Med. 2012;1(12):855–65. pmid:23283547
* View Article
* PubMed/NCBI
* Google Scholar
29. 29. Olmer R, Engels L, Usman A, Menke S, Malik MNH, Pessler F, et al. Differentiation of Human Pluripotent Stem Cells into Functional Endothelial Cells in Scalable Suspension Culture. Stem Cell Reports. 2018;10(5):1657–72. pmid:29681541
* View Article
* PubMed/NCBI
* Google Scholar
30. 30. Auth R, Klinger JR. Emerging pharmacotherapies for the treatment of pulmonary arterial hypertension. Expert Opin Investig Drugs. 2023;32(11):1025–42. pmid:37881882
* View Article
* PubMed/NCBI
* Google Scholar
31. 31. Simonneau G, Montani D, Celermajer DS, Denton CP, Gatzoulis MA, Krowka M, et al. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J. 2019;53(1). pmid:30545968
* View Article
* PubMed/NCBI
* Google Scholar
32. 32. Duong HT, Comhair SA, Aldred MA, Mavrakis L, Savasky BM, Erzurum SC, et al. Pulmonary artery endothelium resident endothelial colony-forming cells in pulmonary arterial hypertension. Pulm Circ. 2011;1(4):475–86. pmid:22530103
* View Article
* PubMed/NCBI
* Google Scholar
33. 33. Chen Y, Ouyang T, Yin Y, Fang C, Tang CE, Jiang L, et al. Identification of immune-related hub genes and analysis of infiltrated immune cells of idiopathic pulmonary artery hypertension. Front Cardiovasc Med. 2023;10:1125063. pmid:36926043
* View Article
* PubMed/NCBI
* Google Scholar
34. 34. Barnes RM, Firulli BA, VanDusen NJ, Morikawa Y, Conway SJ, Cserjesi P, et al. Hand2 loss-of-function in Hand1-expressing cells reveals distinct roles in epicardial and coronary vessel development. Circ Res. 2011;108(8):940–9. pmid:21350214
* View Article
* PubMed/NCBI
* Google Scholar
35. 35. Cohen ASA, Simotas C, Webb BD, Shi H, Khan WA, Edelmann L, et al. Haploinsufficiency of the basic helix-loop-helix transcription factor HAND2 causes congenital heart defects. Am J Med Genet A. 2020;182(5):1263–7. pmid:32134193
* View Article
* PubMed/NCBI
* Google Scholar
36. 36. Videira RF, Koop AMC, Ottaviani L, Poels EM, Kocken JMM, Dos Remedios C, et al. The adult heart requires baseline expression of the transcription factor Hand2 to withstand right ventricular pressure overload. Cardiovasc Res. 2022;118(12):2688–702. pmid:34550326
* View Article
* PubMed/NCBI
* Google Scholar
37. 37. Zeisberg EM, Ma Q, Juraszek AL, Moses K, Schwartz RJ, Izumo S, et al. Morphogenesis of the right ventricle requires myocardial expression of Gata4. J Clin Invest. 2005;115(6):1522–31. pmid:15902305
* View Article
* PubMed/NCBI
* Google Scholar
38. 38. Farha S, Asosingh K, Xu W, Sharp J, George D, Comhair S, et al. Hypoxia-inducible factors in human pulmonary arterial hypertension: a link to the intrinsic myeloid abnormalities. Blood. 2011;117(13):3485–93. pmid:21258008
* View Article
* PubMed/NCBI
* Google Scholar
39. 39. Park CS, Kim SH, Yang HY, Kim JH, Schermuly RT, Cho YS, et al. Sox17 Deficiency Promotes Pulmonary Arterial Hypertension via HGF/c-Met Signaling. Circ Res. 2022;131(10):792–806. pmid:36205124
* View Article
* PubMed/NCBI
* Google Scholar
40. 40. Radik M, Kmecova Z, Veteskova J, Malikova E, Doka G, Krenek P, et al. Hepatocyte growth factor plays a particular role in progression of overall cardiac damage in experimental pulmonary hypertension. Int J Med Sci. 2019;16(6):854–63. pmid:31337959
* View Article
* PubMed/NCBI
* Google Scholar
41. 41. Giannakoulas G, Mouratoglou SA, Gatzoulis MA, Karvounis H. Blood biomarkers and their potential role in pulmonary arterial hypertension associated with congenital heart disease. a systematic review. Int J Cardiol. 2014;174(3):618–23. pmid:24814894
* View Article
* PubMed/NCBI
* Google Scholar
42. 42. Saleby J, Bouzina H, Lundgren J, Radegran G. Angiogenic and inflammatory biomarkers in the differentiation of pulmonary hypertension. Scand Cardiovasc J. 2017;51(5):261–70. pmid:28776404
* View Article
* PubMed/NCBI
* Google Scholar
43. 43. Guignabert C, Savale L, Boucly A, Thuillet R, Tu L, Ottaviani M, et al. Serum and Pulmonary Expression Profiles of the Activin Signaling System in Pulmonary Arterial Hypertension. Circulation. 2023;147(24):1809–22. pmid:37096577
* View Article
* PubMed/NCBI
* Google Scholar
44. 44. Ryanto GRT, Ikeda K, Miyagawa K, Tu L, Guignabert C, Humbert M, et al. An endothelial activin A-bone morphogenetic protein receptor type 2 link is overdriven in pulmonary hypertension. Nat Commun. 2021;12(1):1720. pmid:33741934
* View Article
* PubMed/NCBI
* Google Scholar
45. 45. Chen Z, DSO S, Zimnicka AM, Jiang Y, Sharma , Chen S, et al. Reciprocal regulation of eNOS and caveolin-1 functions in endothelial cells. Mol Biol Cell. 2018;29(10):1190–202. pmid:29563255
* View Article
* PubMed/NCBI
* Google Scholar
46. 46. Harding IC, Mitra R, Mensah SA, Herman IM, Ebong EE. Pro-atherosclerotic disturbed flow disrupts caveolin-1 expression, localization, and function via glycocalyx degradation. J Transl Med. 2018;16(1):364. pmid:30563532
* View Article
* PubMed/NCBI
* Google Scholar
47. 47. Sessa WC. eNOS at a glance. J Cell Sci. 2004;117(Pt 12):2427–9. pmid:15159447
* View Article
* PubMed/NCBI
* Google Scholar
48. 48. Aulak KS, Barnes JW, Tian L, Mellor NE, Haque MM, Willard B, et al. Specific O-GlcNAc modification at Ser-615 modulates eNOS function. Redox Biol. 2020;36:101625. pmid:32863226
* View Article
* PubMed/NCBI
* Google Scholar
49. 49. Heiss EH, Dirsch VM. Regulation of eNOS enzyme activity by posttranslational modification. Curr Pharm Des. 2014;20(22):3503–13. pmid:24180389
* View Article
* PubMed/NCBI
* Google Scholar
50. 50. Murray F, Maclean MR, Insel PA. Role of phosphodiesterases in adult-onset pulmonary arterial hypertension. Handb Exp Pharmacol. 2011(204):279–305. pmid:21695645
* View Article
* PubMed/NCBI
* Google Scholar
51. 51. Murray F, Patel HH, Suda RY, Zhang S, Thistlethwaite PA, Yuan JX, et al. Expression and activity of cAMP phosphodiesterase isoforms in pulmonary artery smooth muscle cells from patients with pulmonary hypertension: role for PDE1. Am J Physiol Lung Cell Mol Physiol. 2007;292(1):L294–303. pmid:16980375
* View Article
* PubMed/NCBI
* Google Scholar
52. 52. Aulak KS, Al Abdi S, Li L, Crabb JS, Ghosh A, Willard B, et al. Disease-specific platelet signaling defects in idiopathic pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol. 2021;320(5):L739–L49. pmid:33596129
* View Article
* PubMed/NCBI
* Google Scholar
53. 53. He W, Su X, Chen L, Liu C, Lu W, Wang T, et al. Potential biomarkers and therapeutic targets of idiopathic pulmonary arterial hypertension. Physiol Rep. 2022;10(1):e15101. pmid:34981661
* View Article
* PubMed/NCBI
* Google Scholar
54. 54. Falcetti E, Hall SM, Phillips PG, Patel J, Morrell NW, Haworth SG, et al. Smooth muscle proliferation and role of the prostacyclin (IP) receptor in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2010;182(9):1161–70. pmid:20622039
* View Article
* PubMed/NCBI
* Google Scholar
55. 55. Patel JA, Shen L, Hall SM, Benyahia C, Norel X, McAnulty RJ, et al. Prostanoid EP(2) Receptors Are Up-Regulated in Human Pulmonary Arterial Hypertension: A Key Anti-Proliferative Target for Treprostinil in Smooth Muscle Cells. Int J Mol Sci. 2018;19(8).
* View Article
* Google Scholar
56. 56. Christman BW, McPherson CD, Newman JH, King GA, Bernard GR, Groves BM, et al. An imbalance between the excretion of thromboxane and prostacyclin metabolites in pulmonary hypertension. N Engl J Med. 1992;327(2):70–5. pmid:1603138
* View Article
* PubMed/NCBI
* Google Scholar
57. 57. Adams JM, Cory S. Bcl-2-regulated apoptosis: mechanism and therapeutic potential. Curr Opin Immunol. 2007;19(5):488–96. pmid:17629468
* View Article
* PubMed/NCBI
* Google Scholar
58. 58. Sheng H, Shao J, Morrow JD, Beauchamp RD, DuBois RN. Modulation of apoptosis and Bcl-2 expression by prostaglandin E2 in human colon cancer cells. Cancer Res. 1998;58(2):362–6. pmid:9443418
* View Article
* PubMed/NCBI
* Google Scholar
59. 59. Singh B, Cook KR, Vincent L, Hall CS, Berry JA, Multani AS, et al. Cyclooxygenase-2 induces genomic instability, BCL2 expression, doxorubicin resistance, and altered cancer-initiating cell phenotype in MCF7 breast cancer cells. J Surg Res. 2008;147(2):240–6. pmid:18498876
* View Article
* PubMed/NCBI
* Google Scholar
60. 60. Szweda M, Rychlik A, Babinska I, Pomianowski A. Significance of Cyclooxygenase-2 in Oncogenesis. J Vet Res. 2019;63(2):215–24. pmid:31276061
* View Article
* PubMed/NCBI
* Google Scholar
61. 61. Barnes JW, Kucera ET, Tian L, Mellor NE, Dvorina N, Baldwin WW 3rd, et al. Bone Morphogenic Protein Type 2 Receptor Mutation-Independent Mechanisms of Disrupted Bone Morphogenetic Protein Signaling in Idiopathic Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol. 2016;55(4):564–75. pmid:27187737
* View Article
* PubMed/NCBI
* Google Scholar
62. 62. Barnes JW, Tonelli AR, Heresi GA, Newman JE, Mellor NE, Grove DE, et al. Novel methods in pulmonary hypertension phenotyping in the age of precision medicine (2015 Grover Conference series). Pulm Circ. 2016;6(4):439–47. pmid:28090286
* View Article
* PubMed/NCBI
* Google Scholar
63. 63. Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K, Imamura T, et al. Smurf1 interacts with transforming growth factor-beta type I receptor through Smad7 and induces receptor degradation. J Biol Chem. 2001;276(16):12477–80. pmid:11278251
* View Article
* PubMed/NCBI
* Google Scholar
64. 64. Murakami K, Etlinger JD. Role of SMURF1 ubiquitin ligase in BMP receptor trafficking and signaling. Cell Signal. 2019;54:139–49. pmid:30395943
* View Article
* PubMed/NCBI
* Google Scholar
65. 65. Murakami K, Mathew R, Huang J, Farahani R, Peng H, Olson SC, et al. Smurf1 ubiquitin ligase causes downregulation of BMP receptors and is induced in monocrotaline and hypoxia models of pulmonary arterial hypertension. Exp Biol Med (Maywood). 2010;235(7):805–13. pmid:20558834
* View Article
* PubMed/NCBI
* Google Scholar
66. 66. Lockhart-Cairns MP, Lim KTW, Zuk A, Godwin ARF, Cain SA, Sengle G, et al. Internal cleavage and synergy with twisted gastrulation enhance BMP inhibition by BMPER. Matrix Biol. 2019;77:73–86. pmid:30125619
* View Article
* PubMed/NCBI
* Google Scholar
67. 67. Visovatti SH, Hyman MC, Goonewardena SN, Anyanwu AC, Kanthi Y, Robichaud P, et al. Purinergic dysregulation in pulmonary hypertension. Am J Physiol Heart Circ Physiol. 2016;311(1):H286–98. pmid:27208163
* View Article
* PubMed/NCBI
* Google Scholar
68. 68. Upton PD, Davies RJ, Trembath RC, Morrell NW. Bone morphogenetic protein (BMP) and activin type II receptors balance BMP9 signals mediated by activin receptor-like kinase-1 in human pulmonary artery endothelial cells. J Biol Chem. 2009;284(23):15794–804. pmid:19366699
* View Article
* PubMed/NCBI
* Google Scholar
69. 69. Rafikov R, Rischard F, Vasilyev M, Varghese MV, Yuan JX, Desai AA, et al. Cytokine profiling in pulmonary arterial hypertension: the role of redox homeostasis and sex. Transl Res. 2022;247:1–18. pmid:35405322
* View Article
* PubMed/NCBI
* Google Scholar
70. 70. de Jesus Perez V, Yuan K, Alastalo TP, Spiekerkoetter E, Rabinovitch M. Targeting the Wnt signaling pathways in pulmonary arterial hypertension. Drug Discov Today. 2014;19(8):1270–6. pmid:24955837
* View Article
* PubMed/NCBI
* Google Scholar
71. 71. Chakraborty A, Nathan A, Orcholski M, Agarwal S, Shamskhou EA, Auer N, et al. Wnt7a deficit is associated with dysfunctional angiogenesis in pulmonary arterial hypertension. Eur Respir J. 2023;61(6). pmid:37024132
* View Article
* PubMed/NCBI
* Google Scholar
72. 72. Samokhin AO, Hsu S, Yu PB, Waxman AB, Alba GA, Wertheim BM, et al. Circulating NEDD9 is increased in pulmonary arterial hypertension: A multicenter, retrospective analysis. J Heart Lung Transplant. 2020;39(4):289–99. pmid:31952977
* View Article
* PubMed/NCBI
* Google Scholar
73. 73. Samokhin AO, Stephens T, Wertheim BM, Wang RS, Vargas SO, Yung LM, et al. NEDD9 targets COL3A1 to promote endothelial fibrosis and pulmonary arterial hypertension. Sci Transl Med. 2018;10(445). pmid:29899023
* View Article
* PubMed/NCBI
* Google Scholar
74. 74. Devendran A, Kar S, Bailey R, Trivieri MG. The Role of Bone Morphogenetic Protein Receptor Type 2 (BMPR2) and the Prospects of Utilizing Induced Pluripotent Stem Cells (iPSCs) in Pulmonary Arterial Hypertension Disease Modeling. Cells. 2022;11(23). pmid:36497082
* View Article
* PubMed/NCBI
* Google Scholar
75. 75. Gu M, Shao NY, Sa S, Li D, Termglinchan V, Ameen M, et al. Patient-Specific iPSC-Derived Endothelial Cells Uncover Pathways that Protect against Pulmonary Hypertension in BMPR2 Mutation Carriers. Cell Stem Cell. 2017;20(4):490–504 e5. pmid:28017794
* View Article
* PubMed/NCBI
* Google Scholar
76. 76. Ormiston ML, Toshner MR, Kiskin FN, Huang CJ, Groves E, Morrell NW, et al. Generation and Culture of Blood Outgrowth Endothelial Cells from Human Peripheral Blood. J Vis Exp. 2015(106):e53384. pmid:26780290
* View Article
* PubMed/NCBI
* Google Scholar
77. 77. Usman A, Haase A, Merkert S, Gohring G, Hansmann G, Gall H, et al. Generation of pulmonary arterial hypertension patient-specific induced pluripotent stem cell lines from three unrelated patients with a heterozygous missense mutation in exon 12, a heterozygous in-frame deletion in exon 3 and a missense mutation in exon 11 of the BMPR2 gene. Stem Cell Res. 2021;55:102488. pmid:34388490
* View Article
* PubMed/NCBI
* Google Scholar
78. 78. Schupp JC, Adams TS, Cosme C Jr., Raredon MSB, Yuan Y, Omote N, et al. Integrated Single-Cell Atlas of Endothelial Cells of the Human Lung. Circulation. 2021;144(4):286–302. pmid:34030460
* View Article
* PubMed/NCBI
* Google Scholar
79. 79. Nickel NP, Yuan K, Dorfmuller P, Provencher S, Lai YC, Bonnet S, et al. Beyond the Lungs: Systemic Manifestations of Pulmonary Arterial Hypertension. Am J Respir Crit Care Med. 2020;201(2):148–57. pmid:31513751
* View Article
* PubMed/NCBI
* Google Scholar
80. 80. Boucly A, Savale L, Jais X, Bauer F, Bergot E, Bertoletti L, et al. Association between Initial Treatment Strategy and Long-Term Survival in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med. 2021;204(7):842–54. pmid:34185620
* View Article
* PubMed/NCBI
* Google Scholar
Citation: Aulak KS, Mavarakis L, Tian L, Paul D, Comhair SA, Dweik RA, et al. (2024) Characteristic disease defects in circulating endothelial cells isolated from patients with pulmonary arterial hypertension. PLoS ONE 19(10): e0312535. https://doi.org/10.1371/journal.pone.0312535
About the Authors:
Kulwant S. Aulak
Roles: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Validation, Writing – original draft, Writing – review & editing
Affiliation: Department of Immunology and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio, United States of America
Lori Mavarakis
Roles: Data curation, Investigation, Methodology, Writing – review & editing
Affiliation: Department of Immunology and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio, United States of America
Liping Tian
Roles: Formal analysis, Investigation, Methodology, Writing – original draft, Writing – review & editing
Affiliation: Department of Immunology and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio, United States of America
Deborah Paul
Roles: Data curation, Investigation, Methodology, Writing – original draft, Writing – review & editing
Affiliation: Department of Pulmonary, Cleveland Clinic, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland, Ohio, United States of America
ORICD: https://orcid.org/0000-0003-3666-7424
Suzy A. Comhair
Roles: Formal analysis, Investigation, Methodology, Resources, Supervision, Writing – original draft, Writing – review & editing
Affiliation: Department of Immunology and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio, United States of America
Raed A. Dweik
Roles: Conceptualization, Data curation, Formal analysis, Investigation, Resources, Writing – original draft, Writing – review & editing
Affiliation: Department of Pulmonary, Cleveland Clinic, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland, Ohio, United States of America
Adriano R. Tonelli
Roles: Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Supervision, Writing – original draft, Writing – review & editing
E-mail: [email protected]
Affiliation: Department of Pulmonary, Cleveland Clinic, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland, Ohio, United States of America
ORICD: https://orcid.org/0000-0002-2321-9545
1. Tonelli AR, Arelli V, Minai OA, Newman J, Bair N, Heresi GA, et al. Causes and circumstances of death in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2013;188(3):365–9. pmid:23600433
2. Humbert M, Kovacs G, Hoeper MM, Badagliacca R, Berger RMF, Brida M, et al. 2022 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 2023;61(1).
3. Farber HW, Miller DP, Poms AD, Badesch DB, Frost AE, Muros-Le Rouzic E, et al. Five-Year outcomes of patients enrolled in the REVEAL Registry. Chest. 2015;148(4):1043–54. pmid:26066077
4. McGoon MD, Miller DP. REVEAL: a contemporary US pulmonary arterial hypertension registry. Eur Respir Rev. 2012;21(123):8–18. pmid:22379169
5. Hassoun PM, Mouthon L, Barbera JA, Eddahibi S, Flores SC, Grimminger F, et al. Inflammation, growth factors, and pulmonary vascular remodeling. J Am Coll Cardiol. 2009;54(1 Suppl):S10–S9. pmid:19555853
6. Humbert M, Guignabert C, Bonnet S, Dorfmuller P, Klinger JR, Nicolls MR, et al. Pathology and pathobiology of pulmonary hypertension: state of the art and research perspectives. Eur Respir J. 2019;53(1). pmid:30545970
7. Rajagopal S, Yu YA. The Pathobiology of Pulmonary Arterial Hypertension. Cardiol Clin. 2022;40(1):1–12. pmid:34809910
8. Benza RL, Miller DP, Barst RJ, Badesch DB, Frost AE, McGoon MD. An evaluation of long-term survival from time of diagnosis in pulmonary arterial hypertension from the REVEAL Registry. Chest. 2012;142(2):448–56. pmid:22281797
9. Kingwell K. FDA approves Merck & Co’s pulmonary arterial hypertension drug sotatercept. Nat Rev Drug Discov. 2024.
10. Narechania S, Malesker MA. Drug Interactions Associated With Therapies for Pulmonary Arterial Hypertension. J Pharm Technol. 2022;38(6):349–59. pmid:36311309
11. Ruopp NF, Cockrill BA. Diagnosis and Treatment of Pulmonary Arterial Hypertension: A Review. JAMA. 2022;327(14):1379–91. pmid:35412560
12. Sun X, Chen R, Yao X, Zheng Z, Wang M, Wang C, et al. Treatment of Pulmonary Hypertension: Is Triple Therapy Necessarily Better than Monotherapy? Am J Respir Crit Care Med. 2021;204(12):1492–3. pmid:34672866
13. Gurtu V, Michelakis ED. A Paradigm Shift Is Needed in the Field of Pulmonary Arterial Hypertension for Its Entrance Into the Precision Medicine Era. Circ Res. 2016;119(12):1276–9. pmid:27932471
14. Kanwar MK. Biomarkers in pulmonary arterial hypertension: Moving closer toward precision medicine? J Heart Lung Transplant. 2020;39(4):287–8. pmid:32199588
15. Kelly NJ, Chan SY. Pulmonary Arterial Hypertension: Emerging Principles of Precision Medicine across Basic Science to Clinical Practice. Rev Cardiovasc Med. 2022;23(11). pmid:36875282
16. Boucherat O, Agrawal V, Lawrie A, Bonnet S. The Latest in Animal Models of Pulmonary Hypertension and Right Ventricular Failure. Circ Res. 2022;130(9):1466–86. pmid:35482834
17. Wu XH, Ma JL, Ding D, Ma YJ, Wei YP, Jing ZC. Experimental animal models of pulmonary hypertension: Development and challenges. Animal Model Exp Med. 2022;5(3):207–16. pmid:35333455
18. Comhair SA, Xu W, Mavrakis L, Aldred MA, Asosingh K, Erzurum SC. Human primary lung endothelial cells in culture. Am J Respir Cell Mol Biol. 2012;46(6):723–30. pmid:22427538
19. Kessler S, Rho H, West G, Fiocchi C, Drazba J, de la Motte C. Hyaluronan (HA) deposition precedes and promotes leukocyte recruitment in intestinal inflammation. Clin Transl Sci. 2008;1(1):57–61. pmid:20443819
20. Carethers JM, Pham TT. Mutations of transforming growth factor beta 1 type II receptor, BAX, and insulin-like growth factor II receptor genes in microsatellite unstable cell lines. In Vivo. 2000;14(1):13–20. pmid:10757056
21. Yeager ME, Golpon HA, Voelkel NF, Tuder RM. Microsatellite mutational analysis of endothelial cells within plexiform lesions from patients with familial, pediatric, and sporadic pulmonary hypertension. Chest. 2002;121(3 Suppl):61S. pmid:11893687
22. Passineau MJ, Gallo PH, Williams G, Perez R, Benza RL. Harvest of Endothelial Cells from the Balloon Tips of Swan-Ganz Catheters after Right Heart Catheterization. J Vis Exp. 2019(143). pmid:30735186
23. Singh N, Eickhoff C, Garcia-Agundez A, Bertone P, Paudel SS, Tambe DT, et al. Transcriptional profiles of pulmonary artery endothelial cells in pulmonary hypertension. Sci Rep. 2023;13(1):22534. pmid:38110438
24. Ventetuolo CE, Aliotta JM, Braza J, Chichger H, Dooner M, McGuirl D, et al. Culture of pulmonary artery endothelial cells from pulmonary artery catheter balloon tips: considerations for use in pulmonary vascular disease. Eur Respir J. 2020;55(3). pmid:31949110
25. Pollett JB, Benza RL, Murali S, Shields KJ, Passineau MJ. Harvest of pulmonary artery endothelial cells from patients undergoing right heart catheterization. J Heart Lung Transplant. 2013;32(7):746–9. pmid:23684132
26. Pu X, Du L, Hu Y, Fan Y, Xu Q. Stem/Progenitor Cells and Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol. 2021;41(1):167–78. pmid:33028095
27. Sanchez-Duffhues G, Hiepen C. Human iPSCs as Model Systems for BMP-Related Rare Diseases. Cells. 2023;12(17). pmid:37681932
28. Geti I, Ormiston ML, Rouhani F, Toshner M, Movassagh M, Nichols J, et al. A practical and efficient cellular substrate for the generation of induced pluripotent stem cells from adults: blood-derived endothelial progenitor cells. Stem Cells Transl Med. 2012;1(12):855–65. pmid:23283547
29. Olmer R, Engels L, Usman A, Menke S, Malik MNH, Pessler F, et al. Differentiation of Human Pluripotent Stem Cells into Functional Endothelial Cells in Scalable Suspension Culture. Stem Cell Reports. 2018;10(5):1657–72. pmid:29681541
30. Auth R, Klinger JR. Emerging pharmacotherapies for the treatment of pulmonary arterial hypertension. Expert Opin Investig Drugs. 2023;32(11):1025–42. pmid:37881882
31. Simonneau G, Montani D, Celermajer DS, Denton CP, Gatzoulis MA, Krowka M, et al. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J. 2019;53(1). pmid:30545968
32. Duong HT, Comhair SA, Aldred MA, Mavrakis L, Savasky BM, Erzurum SC, et al. Pulmonary artery endothelium resident endothelial colony-forming cells in pulmonary arterial hypertension. Pulm Circ. 2011;1(4):475–86. pmid:22530103
33. Chen Y, Ouyang T, Yin Y, Fang C, Tang CE, Jiang L, et al. Identification of immune-related hub genes and analysis of infiltrated immune cells of idiopathic pulmonary artery hypertension. Front Cardiovasc Med. 2023;10:1125063. pmid:36926043
34. Barnes RM, Firulli BA, VanDusen NJ, Morikawa Y, Conway SJ, Cserjesi P, et al. Hand2 loss-of-function in Hand1-expressing cells reveals distinct roles in epicardial and coronary vessel development. Circ Res. 2011;108(8):940–9. pmid:21350214
35. Cohen ASA, Simotas C, Webb BD, Shi H, Khan WA, Edelmann L, et al. Haploinsufficiency of the basic helix-loop-helix transcription factor HAND2 causes congenital heart defects. Am J Med Genet A. 2020;182(5):1263–7. pmid:32134193
36. Videira RF, Koop AMC, Ottaviani L, Poels EM, Kocken JMM, Dos Remedios C, et al. The adult heart requires baseline expression of the transcription factor Hand2 to withstand right ventricular pressure overload. Cardiovasc Res. 2022;118(12):2688–702. pmid:34550326
37. Zeisberg EM, Ma Q, Juraszek AL, Moses K, Schwartz RJ, Izumo S, et al. Morphogenesis of the right ventricle requires myocardial expression of Gata4. J Clin Invest. 2005;115(6):1522–31. pmid:15902305
38. Farha S, Asosingh K, Xu W, Sharp J, George D, Comhair S, et al. Hypoxia-inducible factors in human pulmonary arterial hypertension: a link to the intrinsic myeloid abnormalities. Blood. 2011;117(13):3485–93. pmid:21258008
39. Park CS, Kim SH, Yang HY, Kim JH, Schermuly RT, Cho YS, et al. Sox17 Deficiency Promotes Pulmonary Arterial Hypertension via HGF/c-Met Signaling. Circ Res. 2022;131(10):792–806. pmid:36205124
40. Radik M, Kmecova Z, Veteskova J, Malikova E, Doka G, Krenek P, et al. Hepatocyte growth factor plays a particular role in progression of overall cardiac damage in experimental pulmonary hypertension. Int J Med Sci. 2019;16(6):854–63. pmid:31337959
41. Giannakoulas G, Mouratoglou SA, Gatzoulis MA, Karvounis H. Blood biomarkers and their potential role in pulmonary arterial hypertension associated with congenital heart disease. a systematic review. Int J Cardiol. 2014;174(3):618–23. pmid:24814894
42. Saleby J, Bouzina H, Lundgren J, Radegran G. Angiogenic and inflammatory biomarkers in the differentiation of pulmonary hypertension. Scand Cardiovasc J. 2017;51(5):261–70. pmid:28776404
43. Guignabert C, Savale L, Boucly A, Thuillet R, Tu L, Ottaviani M, et al. Serum and Pulmonary Expression Profiles of the Activin Signaling System in Pulmonary Arterial Hypertension. Circulation. 2023;147(24):1809–22. pmid:37096577
44. Ryanto GRT, Ikeda K, Miyagawa K, Tu L, Guignabert C, Humbert M, et al. An endothelial activin A-bone morphogenetic protein receptor type 2 link is overdriven in pulmonary hypertension. Nat Commun. 2021;12(1):1720. pmid:33741934
45. Chen Z, DSO S, Zimnicka AM, Jiang Y, Sharma , Chen S, et al. Reciprocal regulation of eNOS and caveolin-1 functions in endothelial cells. Mol Biol Cell. 2018;29(10):1190–202. pmid:29563255
46. Harding IC, Mitra R, Mensah SA, Herman IM, Ebong EE. Pro-atherosclerotic disturbed flow disrupts caveolin-1 expression, localization, and function via glycocalyx degradation. J Transl Med. 2018;16(1):364. pmid:30563532
47. Sessa WC. eNOS at a glance. J Cell Sci. 2004;117(Pt 12):2427–9. pmid:15159447
48. Aulak KS, Barnes JW, Tian L, Mellor NE, Haque MM, Willard B, et al. Specific O-GlcNAc modification at Ser-615 modulates eNOS function. Redox Biol. 2020;36:101625. pmid:32863226
49. Heiss EH, Dirsch VM. Regulation of eNOS enzyme activity by posttranslational modification. Curr Pharm Des. 2014;20(22):3503–13. pmid:24180389
50. Murray F, Maclean MR, Insel PA. Role of phosphodiesterases in adult-onset pulmonary arterial hypertension. Handb Exp Pharmacol. 2011(204):279–305. pmid:21695645
51. Murray F, Patel HH, Suda RY, Zhang S, Thistlethwaite PA, Yuan JX, et al. Expression and activity of cAMP phosphodiesterase isoforms in pulmonary artery smooth muscle cells from patients with pulmonary hypertension: role for PDE1. Am J Physiol Lung Cell Mol Physiol. 2007;292(1):L294–303. pmid:16980375
52. Aulak KS, Al Abdi S, Li L, Crabb JS, Ghosh A, Willard B, et al. Disease-specific platelet signaling defects in idiopathic pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol. 2021;320(5):L739–L49. pmid:33596129
53. He W, Su X, Chen L, Liu C, Lu W, Wang T, et al. Potential biomarkers and therapeutic targets of idiopathic pulmonary arterial hypertension. Physiol Rep. 2022;10(1):e15101. pmid:34981661
54. Falcetti E, Hall SM, Phillips PG, Patel J, Morrell NW, Haworth SG, et al. Smooth muscle proliferation and role of the prostacyclin (IP) receptor in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2010;182(9):1161–70. pmid:20622039
55. Patel JA, Shen L, Hall SM, Benyahia C, Norel X, McAnulty RJ, et al. Prostanoid EP(2) Receptors Are Up-Regulated in Human Pulmonary Arterial Hypertension: A Key Anti-Proliferative Target for Treprostinil in Smooth Muscle Cells. Int J Mol Sci. 2018;19(8).
56. Christman BW, McPherson CD, Newman JH, King GA, Bernard GR, Groves BM, et al. An imbalance between the excretion of thromboxane and prostacyclin metabolites in pulmonary hypertension. N Engl J Med. 1992;327(2):70–5. pmid:1603138
57. Adams JM, Cory S. Bcl-2-regulated apoptosis: mechanism and therapeutic potential. Curr Opin Immunol. 2007;19(5):488–96. pmid:17629468
58. Sheng H, Shao J, Morrow JD, Beauchamp RD, DuBois RN. Modulation of apoptosis and Bcl-2 expression by prostaglandin E2 in human colon cancer cells. Cancer Res. 1998;58(2):362–6. pmid:9443418
59. Singh B, Cook KR, Vincent L, Hall CS, Berry JA, Multani AS, et al. Cyclooxygenase-2 induces genomic instability, BCL2 expression, doxorubicin resistance, and altered cancer-initiating cell phenotype in MCF7 breast cancer cells. J Surg Res. 2008;147(2):240–6. pmid:18498876
60. Szweda M, Rychlik A, Babinska I, Pomianowski A. Significance of Cyclooxygenase-2 in Oncogenesis. J Vet Res. 2019;63(2):215–24. pmid:31276061
61. Barnes JW, Kucera ET, Tian L, Mellor NE, Dvorina N, Baldwin WW 3rd, et al. Bone Morphogenic Protein Type 2 Receptor Mutation-Independent Mechanisms of Disrupted Bone Morphogenetic Protein Signaling in Idiopathic Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol. 2016;55(4):564–75. pmid:27187737
62. Barnes JW, Tonelli AR, Heresi GA, Newman JE, Mellor NE, Grove DE, et al. Novel methods in pulmonary hypertension phenotyping in the age of precision medicine (2015 Grover Conference series). Pulm Circ. 2016;6(4):439–47. pmid:28090286
63. Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K, Imamura T, et al. Smurf1 interacts with transforming growth factor-beta type I receptor through Smad7 and induces receptor degradation. J Biol Chem. 2001;276(16):12477–80. pmid:11278251
64. Murakami K, Etlinger JD. Role of SMURF1 ubiquitin ligase in BMP receptor trafficking and signaling. Cell Signal. 2019;54:139–49. pmid:30395943
65. Murakami K, Mathew R, Huang J, Farahani R, Peng H, Olson SC, et al. Smurf1 ubiquitin ligase causes downregulation of BMP receptors and is induced in monocrotaline and hypoxia models of pulmonary arterial hypertension. Exp Biol Med (Maywood). 2010;235(7):805–13. pmid:20558834
66. Lockhart-Cairns MP, Lim KTW, Zuk A, Godwin ARF, Cain SA, Sengle G, et al. Internal cleavage and synergy with twisted gastrulation enhance BMP inhibition by BMPER. Matrix Biol. 2019;77:73–86. pmid:30125619
67. Visovatti SH, Hyman MC, Goonewardena SN, Anyanwu AC, Kanthi Y, Robichaud P, et al. Purinergic dysregulation in pulmonary hypertension. Am J Physiol Heart Circ Physiol. 2016;311(1):H286–98. pmid:27208163
68. Upton PD, Davies RJ, Trembath RC, Morrell NW. Bone morphogenetic protein (BMP) and activin type II receptors balance BMP9 signals mediated by activin receptor-like kinase-1 in human pulmonary artery endothelial cells. J Biol Chem. 2009;284(23):15794–804. pmid:19366699
69. Rafikov R, Rischard F, Vasilyev M, Varghese MV, Yuan JX, Desai AA, et al. Cytokine profiling in pulmonary arterial hypertension: the role of redox homeostasis and sex. Transl Res. 2022;247:1–18. pmid:35405322
70. de Jesus Perez V, Yuan K, Alastalo TP, Spiekerkoetter E, Rabinovitch M. Targeting the Wnt signaling pathways in pulmonary arterial hypertension. Drug Discov Today. 2014;19(8):1270–6. pmid:24955837
71. Chakraborty A, Nathan A, Orcholski M, Agarwal S, Shamskhou EA, Auer N, et al. Wnt7a deficit is associated with dysfunctional angiogenesis in pulmonary arterial hypertension. Eur Respir J. 2023;61(6). pmid:37024132
72. Samokhin AO, Hsu S, Yu PB, Waxman AB, Alba GA, Wertheim BM, et al. Circulating NEDD9 is increased in pulmonary arterial hypertension: A multicenter, retrospective analysis. J Heart Lung Transplant. 2020;39(4):289–99. pmid:31952977
73. Samokhin AO, Stephens T, Wertheim BM, Wang RS, Vargas SO, Yung LM, et al. NEDD9 targets COL3A1 to promote endothelial fibrosis and pulmonary arterial hypertension. Sci Transl Med. 2018;10(445). pmid:29899023
74. Devendran A, Kar S, Bailey R, Trivieri MG. The Role of Bone Morphogenetic Protein Receptor Type 2 (BMPR2) and the Prospects of Utilizing Induced Pluripotent Stem Cells (iPSCs) in Pulmonary Arterial Hypertension Disease Modeling. Cells. 2022;11(23). pmid:36497082
75. Gu M, Shao NY, Sa S, Li D, Termglinchan V, Ameen M, et al. Patient-Specific iPSC-Derived Endothelial Cells Uncover Pathways that Protect against Pulmonary Hypertension in BMPR2 Mutation Carriers. Cell Stem Cell. 2017;20(4):490–504 e5. pmid:28017794
76. Ormiston ML, Toshner MR, Kiskin FN, Huang CJ, Groves E, Morrell NW, et al. Generation and Culture of Blood Outgrowth Endothelial Cells from Human Peripheral Blood. J Vis Exp. 2015(106):e53384. pmid:26780290
77. Usman A, Haase A, Merkert S, Gohring G, Hansmann G, Gall H, et al. Generation of pulmonary arterial hypertension patient-specific induced pluripotent stem cell lines from three unrelated patients with a heterozygous missense mutation in exon 12, a heterozygous in-frame deletion in exon 3 and a missense mutation in exon 11 of the BMPR2 gene. Stem Cell Res. 2021;55:102488. pmid:34388490
78. Schupp JC, Adams TS, Cosme C Jr., Raredon MSB, Yuan Y, Omote N, et al. Integrated Single-Cell Atlas of Endothelial Cells of the Human Lung. Circulation. 2021;144(4):286–302. pmid:34030460
79. Nickel NP, Yuan K, Dorfmuller P, Provencher S, Lai YC, Bonnet S, et al. Beyond the Lungs: Systemic Manifestations of Pulmonary Arterial Hypertension. Am J Respir Crit Care Med. 2020;201(2):148–57. pmid:31513751
80. Boucly A, Savale L, Jais X, Bauer F, Bergot E, Bertoletti L, et al. Association between Initial Treatment Strategy and Long-Term Survival in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med. 2021;204(7):842–54. pmid:34185620
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2024 Aulak et al. This is an open access article distributed under the terms of the Creative Commons Attribution License: http://creativecommons.org/licenses/by/4.0/ (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by elevated pulmonary arterial pressures that can lead to right heart failure and death. No cure exists for this disease, but therapeutic advancements have extended its median survival from 2 to 7 years. Mechanistic research in PAH has been limited by factors including that a) animal models do not fully recapitulate the disease or provide insights into its pathogenesis, and b) cellular material from PAH patients is primarily obtained from donor lungs during autopsy or transplantation, which reflect end-stage disease. Therefore, there is a need to identify tools that can elucidate the specific mechanisms of human disease in individual patients, a critical step to guide treatment decisions based on specific pathway abnormalities. Here we demonstrate a simple method to isolate and culture circulating endothelial cells (CECs) obtained at the time of right heart catheterization in PAH patients. We tested these CECs using transcriptomics and found that they have typical traits of PAH, including those involving key treatment pathways, i.e. nitric oxide, endothelin, prostacyclin and BMP/activin pathways. CECs show important gene expression changes in other central PAH disease pathways. In summary, we present a new cellular model for the ex-vivo mechanistic evaluation of critical PAH pathways that participate in the pathogenesis of the disease and may help personalized therapeutic decisions.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer