Abstract
Background
Right ventricular (RV) dysfunction is a significant prognostic determinant of morbidity and mortality in pulmonary arterial hypertension (PAH). Despite the importance of RV function in PAH, the underlying molecular mechanisms of RV dysfunction secondary to PAH remain unclear. We aim to identify and compare molecular determinants of RV failure using RNA sequencing of RV tissue from 2 clinically relevant animal models of PAH.Methods
We performed RNA sequencing on RV from rats treated with monocrotaline or Sugen with hypoxia/normoxia. PAH and RV failure were confirmed by catheterization and echocardiography. We validated the RV transcriptome results using quantitative real-time polymerase chain reaction, immunofluorescence, and Western blot. Immunohistochemistry and immunofluorescence were performed on human RV tissue from control (n=3) and PAH-induced RV failure patients (n=5).Results
We identified similar transcriptomic profiles of RV from monocrotaline- and Sugen with hypoxia-induced RV failure. Pathway analysis showed genes enriched in epithelial-to-mesenchymal transition, inflammation, and metabolism. Histological staining of human RV tissue from patients with RV failure secondary to PAH revealed significant RV fibrosis and endothelial-to-mesenchymal transition, as well as elevated cellular communication network factor 2 (top gene implicated in epithelial-to-mesenchymal transition/endothelial-to-mesenchymal transition) expression in perivascular areas compared with normal RV.Conclusions
Transcriptomic signature of RV failure in monocrotaline and Sugen with hypoxia models showed similar gene expressions and biological pathways. We provide translational relevance of this transcriptomic signature using RV from patients with PAH to demonstrate evidence of epithelial-to-mesenchymal transition/endothelial-to-mesenchymal transition and protein expression of cellular communication network factor 2 (CTGF [connective tissue growth factor]). Targeting specific molecular mechanisms responsible for RV failure in monocrotaline and Sugen with hypoxia models may identify novel therapeutic strategies for PAH-associated RV failure.Free full text
Transcriptomic Analysis of Right Ventricular Remodeling in Two Rat models of Pulmonary Hypertension: Identification and Validation of EMT in Human Right Ventricular Failure
Abstract
Background:
Right ventricular (RV) dysfunction is a significant prognostic determinant of morbidity and mortality in pulmonary arterial hypertension (PAH). Despite the importance of RV function in PAH, the underlying molecular mechanisms of RV dysfunction secondary to PAH remain unclear. We aim to identify and compare molecular determinants of RV failure using RNA-sequencing of RV tissue from two clinically relevant animal models of PAH.
Methods:
We performed RNA-sequencing on RV from rats treated with monocrotaline (MCT) or Sugen with hypoxia/normoxia (SuHx). PAH and RV failure were confirmed by catheterization and echocardiography. We validated the RV transcriptome results using quantitative real-time PCR, immunofluorescence and Western blot. Immunohistochemistry and immunofluorescence were performed on human RV tissue from control (n=3) and PAH-induced RV failure patients (n=5).
Results:
We identified similar transcriptomic profiles of RV from MCT- and SuHx-induced RV-failure. Pathway analysis showed genes enriched in epithelial-to-mesenchymal transition (EMT), inflammation, and metabolism. Histological staining of human RV tissue from patients with RV-failure secondary to PAH revealed significant RV fibrosis and endothelial-to-mesenchymal transition (EndMT), as well as elevated CCN2 (top gene implicated in EMT/EndMT) expression in perivascular areas compared to normal RV.
Conclusions:
Transcriptomic signature of RV failure in MCT and SuHx models showed similar gene expressions and biological pathways. We provide translational relevance of this transcriptomic signature using RV from PAH patients to demonstrate evidence of EMT/EndMT and protein expression of CCN2 (CTGF). Targeting specific molecular mechanisms responsible for RV failure in MCT and SuHx models may identify novel therapeutic strategies for PAH-associated RV failure.
Introduction
Pulmonary arterial hypertension (PAH) is a progressive, fatal disease characterized by pathological arterial remodeling leading to increased pulmonary vascular resistance (PVR), right ventricular (RV) hypertrophy, RV failure, and death.1,2 Although current therapies targeting pulmonary vasculature have resulted in some functional improvement for patients with PAH-induced RV failure, survival remains poor.1,2 Previous studies have demonstrated that symptom severity and survival in PAH are closely linked to RV remodeling and function.2 RV remodeling due to chronic pressure overload causes a number of adaptations in size, shape, and structure, and can lead to significant RV dysfunction.3 It is a complex process that leads to cardiomyocyte hypertrophy, fibrosis, inflammation, angiogenesis, and metabolic changes. In PAH patient, the adaptation of the RV to high pulmonary arterial pressures is the most important determinant of prognosis. However, the underlying molecular mechanism of adverse RV remodeling and dysfunction is poorly understood, and there are no currently approved therapies that improve RV function.1
Recent evidence suggests RV fibrosis to play a crucial role in the development of RV failure. Increased fibrotic deposition, characterized by myofibroblast accumulation and excessive collagen secretion, can result in RV stiffness and severe dysfunction.4 Recent studies have implicated epithelial-to-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (EndMT) to play a role in adult cardiovascular disease and fibrosis. In response to cardiac injury, a significant portion of myofibroblasts arise from the conversion of epithelial or endothelial cells through the EMT or EndMT process, respectively, and ultimately lead to excessive collagen deposition, fibrosis and cardiac dysfunction.5,6 EndMT has been shown to play a critical role in myocardial fibrosis and diastolic dysfunction in pressure-overload rodent models.6,7
Monocrotaline (MCT) toxin and vascular endothelial growth factor (VEGF) receptor antagonist (SU5416) combined with chronic hypoxia followed by normoxia (SuHx) represent two important animal models that induce PAH and RV failure in response to pressure overload.8 Most studies in RV remodeling due to PAH have used MCT model, but off-target effects of MCT have greatly limited its clinical relevance.1 SuHx model is considered an important pre-clinical model of PAH due to its occlusive plexiform-like lesions in the lung, which closely resemble human PAH.9 This model also leads to severe RV dysfunction with marked hypertrophy, which are important features of RV failure due to PAH. Here, we performed comparative transcriptomic analysis of RV failure in MCT and SuHx PAH models using RNA sequencing. This study provides a comprehensive understanding of dysregulated genes and pathways involved in PAH rats with RV failure, and further validation of transcriptomic signature in human RV failure patients with PAH.
Materials and Methods
The data that support the findings of this study are available from the corresponding author upon reasonable request.
Development of RV failure in PAH rats
All animal studies were performed in accordance with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals. Adult male Sprague Dawley rats (250–350g) received either a single subcutaneous injection of endothelial toxin Monocrotaline (MCT, 60mg/kg, MCT group, n=7) and were followed for ~30 days or VEGF-receptor antagonist Sugen (SU5416, 20mg/kg, SuHx group, n=7) and kept in hypoxia (10% oxygen) for 3-weeks followed by 2-weeks of normoxia. Phosphate buffered saline (PBS) treated rats served as controls (CTRL group, n=7). Transthoracic echocardiography was performed to monitor cardiopulmonary hemodynamics using a Vevo 2100 high-resolution image system. The right ventricular systolic pressure (RVSP) was measured directly by a catheter connected to a pressure transducer (ADInstruments) into the RV.
RNA Sequencing and functional analysis
Libraries for RNA-seq were prepared using SMARTer Stranded Total RNA-Seq Kit v2 - Pico Input Mammalian (Takara Bio) and sequenced using Hiseq 3000 (Illumina). Differential expression analysis was performed using the DESeq2 R package version 1.25.16. DEGs with false discovery rate (FDR) <0.05 based on Benjamini Hochberg algorithm were considered statistically significant. Gene set enrichment analysis was performed using the Bioconductor (release 3.1) fgsea10 and R (version 3.6.1) software package. Hallmark11 and Reactome12 gene sets were obtained from molecular signature database (MSigDB).13 Enriched pathways considered statistically significant were defined by adjusted p-value <0.05.
RNA Extraction and quantitative real-time PCR
Total RNA was isolated from rat RV and LV tissue using Trizol (Invitrogen). Quantitative real-time PCR was performed with iTaq Universal SYBR (Bio-Rad). GADPH was used as an internal reference control, and relative gene expression was normalized to PBS-treated group.
Western Blot Analysis
Protein lysates were prepared from rat RV and LV tissue using modified RIPA lysis buffer containing protease and phosphatase inhibitor cocktails (Sigma). Proteins were diluted in 4x Laemmli sample buffer, boiled, separated on 10% gels (Bio-Rad) by SDS page and subsequently transferred onto nitrocellulose membranes (Bio-Rad) using semi-dry blotting (TransBlot Turbo System, Bio-Rad). After transfer, membranes were blocked with 5% bovine serum albumin (Sigma) and incubated with antibodies directed against CTGF (Abcam #ab6992; 1:200) and Vinculin (Sigma #V9131; 1:5000). IR Dye-conjugated secondary antibodies (LI-COR) were used for detection and blots were scanned and quantified using the LI-COR Odyssey Infrared Imaging System and Image Studio Lite.
Histologic assessment and immunofluorescence staining
The RV wall, the left ventricular (LV) wall, and the interventricular septum (IVS) were dissected and weighed. The ratio of the RV to LV plus septal weight [RV/(LV + IVS)] was calculated as the Fulton index of RV hypertrophy. Rat heart OCT sections were stained with antibodies against anti-αSMA (Sigma #A2547; 1:400), anti-CD31 (Novus Biologicals #NB100–2284; 1:100), or anti-CTGF (CCN2, Abcam #ab6992; 1:500). Paraffin embedded human RV free wall sections were stained with Masson’s trichrome staining according to manufacturer’s protocol (Sigma) or with antibodies against anti-αSMA, anti-VWF, or anti-CTGF (CCN2). Percent RV tissue fibrosis was determined as previously described.14 VWF and αSMA positive vessels were counted manually at 10x objective from at least 20 images across the RV section. Mean fluorescence intensity was calculated by at least 15 images across the RV sections and then analyzed with ImageJ software (http://rsb.info.nih.gov/ij/). Sections were imaged with Nikon confocal microscope.
Human RV Tissue acquisition and characteristics
We acquired human RV sections (RV free wall) from non-heart failure donor hearts (control group; n=3) and right heart failure patients with PAH (RV Failure; n=5) from the Department of Pathology at University of California, Los Angeles (UCLA). De-identified clinical dataset were obtained for each patient. Right heart dysfunction due to PAH was evaluated by right heart catheterization (Table E6).
Statistical Analysis
To assess differences between groups, Welch’s Unpaired t-test and one-way ANOVA tests were used due to potential assumption violations (equal variance) using the more standard tests. When significant differences were detected, individual mean values were compared by post-hoc tests that allowed for multiple comparisons with adequate type I error control (Bonferroni). Analyses were run using GraphPad Prism and p-values<0.05 was considered statistically significant. Values are expressed as mean ± SD.
Results
Development of severe PAH and RV failure in MCT and SuHx rats
We first confirmed severe PAH and RV failure using serial non-invasive echocardiography and terminal right heart catheterization in rats treated with MCT or SuHx compared to PBS-treated rats for control (Figure 1A). Both MCT and SuHx rats showed significant PAH as evidenced by increased RVSP (MCT: 90.6±16.0, p<0.0001; SuHx: 90.8±18.7, p<0.0001), and decreased pulmonary artery acceleration time (PAT) (MCT: 21.1±6.2, p=0.1; SuHx: 18.5±.2.3, p=0.01) and PAT/pulmonary ejection time (PET) ratio (MCT: 0.3±0.1, p=0.04; SuHx: 0.2±0.0, p=0.001) compared to control (Figure 1C; Table 1). RV dysfunction was demonstrated by increased RV internal diameter at end-diastole (RVIDd) (MCT: 3.0 ± 0.9, p=0.0006; SuHx: 2.7 ± 0.8, p=0.0043) and decreased RV fractional area change (RVFAC) in MCT and SuHx rats (MCT: 18.6±10.8, p=0.0044; SuHx: 15.8±5.7, p=0.0014) compared to control (Figure 1B, ,C;C; Table 1). MCT and SuHx rats also demonstrated an increase in RV hypertrophy index (RV/LV+IVS) (MCT: 0.7±0.2, p<0.0001; SuHx: 0.6±0.2, p=0.0003). No significant differences were observed between SuHx- and MCT-treated groups.
Table 1.
Control (n=7) | SuHx (n=7) | MCT (n=7) | |
---|---|---|---|
RVFAC (%) | 38.4 ± 12.0 | 15.8 ± 5.7** | 18.6 ± 10.8** |
PAT (ms) | 29.4 ± 4.2 | 18.5 ± 2.3*** | 21.1 ± 6.2** |
PAT/PET | 0.4 ± 0.0 | 0.2 ± 0.0*** | 0.3 ± 0.1*** |
RVIDd (mm) | 1.3 ± 0.3 | 2.7 ± 0.8** | 3.0 ± 0.9*** |
LVEF (%) | 81.7 ± 9.8 | 91.2 ± 9.0 | 81.6 ± 11.8 |
LVFS (%) | 53.1 ± 9.5 | 68.6 ± 16.3 | 57.7 ± 17.7 |
HR (bpm) | 346 ± 40 | 376 ± 117 | 302 ± 36 |
RVSP (mmHg) | 37.6 ± 2.0 | 90.8 ± 18.7*** | 90.6 ± 16.0*** |
RV/(LV+S) | 0.3 ± 0.0 | 0.6 ± 0.2*** | 0.7 ± 0.2*** |
Mean ± SD. N= 6–7 rats per group.
RVFAC: right ventricle fractional area change; PAT: pulmonary artery acceleration time; PET: pulmonary ejection time; RVIDd: right ventricle internal diameter at end-diastole; LVEF: left ventricular ejection fraction; LVFS: left ventricular fractional shortening; HR: heart rate; RVSP: right ventricular systolic pressure; RV: right ventricle, LV: left ventricle; S: septum.
Similar transcriptome signature of RV failure in MCT and SuHx rats
After confirmation of severe RV dysfunction, RNA sequencing of RV tissue from MCT (n=4) and SuHx (n=4) rats revealed 7,842 and 3,146 differentially expressed genes (DEG), respectively, compared to control rats (n=4) based on FDR <0.05 (Figure 2, ,3A,3A, Table E1 and E2). Hierarchical clustering of top DEGs (FDR <0.05) in the RV showed similar gene signatures between MCT and SuHx rats compared to control (Figure 2), with mild variability within the SuHx group. Both MCT- and SuHx-treated groups shared 2,693 DEGs in the RV that were either up- or down-regulated in a similar fashion (Figure 3A, ,3C).3C). The top 20 up- and down-regulated transcripts according to log2fold change and p-value threshold of <0.05 in RV of MCT and SuHx-treated groups are shown in Figure 3B. SPP1 and AHRR were the most up-regulated DEG in MCT and SuHx-treated group, respectively compared to control. Myo16 and Proz were the most down-regulated DEGs in MCT and SuHx-treated group, respectively compared to control.
Multiple pathways are commonly altered in RV of MCT- and SuHx-induced RV failure
We performed a ranked-based directional enrichment analysis of RV transcriptome from MCT and SuHx rats compared to control. Since previous studies reported RV failure being associated with RV fibrosis, inflammation, and angiogenesis in MCT- and SuHx- models15,16, we used a refined collection of gene sets (Hallmark) that includes these biological processes.11 We identified 32 and 30 Hallmark pathways significantly enriched in MCT and SuHx rats, respectively with 25 pathways overlapping (Figure 4A, Table E3). The top up-regulated enriched pathways in RV of MCT and SuHx rats according to FDR <0.05 were EMT and tumor-necrosis factor-alpha (TNF-α) signaling via nuclear factor-kB (NFkB). The top down-regulated enriched pathways in both MCT and SuHx rats were oxidative phosphorylation and fatty acid metabolism (Figure 4B). Inflammatory response and angiogenesis were also significantly enriched in both MCT- and SuHx-treated groups.
To further explore specific biological processes, we used Reactome pathway enrichment analysis12 of RV from MCT and SuHx rats. Extracellular matrix organization and interleukin-10 signaling were the top up-regulated enriched pathways in MCT and SuHx rats, respectively. The citric acid TCA cycle and respiratory electron transport were the top down-regulated enriched pathways in MCT and SuHx rats (Table E4).
qPCR validation of selected genes from enrichment pathway analysis
To validate the RNA sequencing results and gene expression changes, we selected DEGs from significantly enriched pathways that were identified by Hallmark analysis for qRT-PCR. PCR results of DEGs in the RV from EMT (SPP1, CCN2), TNF-α signaling via NFκB (IL7R), oxidative phosphorylation (ACAT1, COX7B), fatty acid metabolism (DECR1, HSP90A), inflammation (C5AR1) and angiogenesis (S100A4) were consistent with the RNA sequencing results (Figure 5A). In contrast, PCR results in the LV for SPP1, C5AR1, HSP90A, ACAT1, COX7B, and DECR1 showed no significant change in gene expression from SuHx and MCT rats compared to control. Though, we did observe significant up-regulation of S100A4 in SuHx (p=0.007) and MCT (p=0.003), CCN2 in SuHx (p<0.001), and IL7R in SuHx (p=0.006) compared to control (Figure 5B). No significant difference in gene expression levels between SuHx and MCT were noted.
Demonstration of EndMT/EMT in RV of MCT- and SuHx-induced RV failure
To evaluate the relevance of our RV transcriptome results in the context of RVF, we focused on EndMT/EMT, the top up-regulated enriched pathway in RV of MCT and SuHx rats. RV sections were stained with antibodies against α-SMA (myofibroblast marker) and CD31 (endothelial cell marker). Co-localization of α-SMA and CD31 is considered as evidence for EndMT (Figure 6).
To identify significant genes involved in EMT/EndMT, we created a list of genes based on Hallmark EMT gene set that were statistically significant (FDR <0.05) in both MCT and SuHx RV tissue (Table E5). Based on log2fold change, we identified SPP1 and CCN2 as the top up-regulated genes associated with EMT pathway. To validate their association with EMT and RV failure, we assessed CTGF (CCN2) immunolocalization in the RV which appears to be localized to vascular/perivascular areas and within the cardiomyocytes (Figure 7A). We also measured the expression of CTGF protein in MCT and SuHx rats using Western blot analysis. We found an increased RV expression of CTGF in SuHx (p=0.0362) and MCT rats (p=0.0016), whereas no significant differences were found in the LV (Figure 7B, ,CC).
Validation of rat RV transcriptome in human right heart failure
To evaluate the clinical relevance of our RV transcriptome results in the context of RVF, we focused on EMT—the top up-regulated enriched pathway in RV of MCT and SuHx rats—in human RV sections from patients with RV failure secondary to PAH (n=5) and normal RV controls (n=3) (Table E6). Given the role of EMT and EndMT in cardiac fibrosis during chronic pressure overload6,17, we first stained human RV tissues with Masson’s trichome. Fibrosis in both interstitial and perivascular areas increased in RV of human PAH compared to control (RVF: 49.5±9.0, Ctrl: 19.9±2.6, p=0.028) (Figure 8A, ,B).B). Human RV sections were further stained with antibodies against α-SMA (myofibroblast marker) and VWF (endothelial cell marker). We observed increased co-localization (RVF: 5.0±0.1; Ctrl: 11.6±2.3, p=0.047) and number of VWF and α-SMA positive major vessels in PAH RV sections (RVF: 71.6±4.4%; Ctrl: 53.7±2.2%, p=0.013) compared to control (Figure 8C, ,DD).
To validate the association of CTGF (CCN2) with EMT and RV failure, we measured the RV expression of CTGF protein in human PAH RV tissue using immunofluorescence. Similarly, we observed an increased expression of CTGF (CCN2) that localized primarily to perivascular areas in human PAH RV tissue compared to control (RVF: 101.9±5.9; Ctrl: 82.8± 0.6 MFI per vessel, p=0.047) (Figure 8E, ,FF).
Discussion
The present study is the first to conduct comparative RNA-seq analysis of RV between MCT- and SuHx-induced PAH animal models. We showed that MCT and SuHx rats shared similar gene expression profiles in RV. Furthermore, Hallmark and Reactome enrichment pathway analysis showed a significant shift in EMT, inflammation, and extracellular matrix organization, which is consistent with previous findings of RV failure from MCT- and SuHx-induced PAH models.15,16 Also, this study revealed significant impairment of pathways related to metabolism such as oxidative phosphorylation, fatty acid metabolism, and respiratory electron transport. We provide translational relevance of our rat RV transcriptome using RV tissue from PAH patients to show evidence of EMT/EndMT and protein expression of a top EMT gene CCN2 (CTGF).
PAH animal models of RV failure
Traditionally, MCT rat model has been used to study RV failure, but off-target effects of MCT have greatly limited its translational relevance.8,18 For example, myocarditis induced by MCT can affect both the left and right ventricle, which may complicate transcriptome results of RV hypertrophy and failure.19 A recent transcriptomic study showed the gene expression profile of RV failure using MCT rat model, and compared it with published microarray data from pulmonary arterial banding (PAB) mice model and patients with bone morphogenetic protein receptor type 2 mutations (BRMP2).20 Potus et al. showed significant metabolic changes involving mitochondria and electron transport chain, as well as fibrosis, inflammation, and angiogenesis.20 Our MCT-RV expression profile is also in agreement with their results and previous reported studies. However, we include for the first time, the expression profile of RV from SuHx animal model using RNA sequencing, which is considered a more relevant pre-clinical model due to its pulmonary neointimal lesions that closely resemble human plexiform lesions.9,21
PAB model has been used alternatively to single injection of endothelial toxins to study RV remodeling since it is better tolerated and does not induce systemic or toxic effects. PAB is considered a pure RV pressure overload model without any pulmonary vascular injury. It typically results in far less RV decompensation and RV failure compared to MCT and SuHx animal models, and thus considered a better representation of RV adaptive remodeling.2 A recent study comparing RV function of PAB with SuHx model showed that chronic progressive RV pressure overload alone does not cause RV failure, while RV failure from angioproliferative PAH results in exaggerated RVH, apoptosis, fibrosis, and capillary rarefaction.22 Clinically, RV function from adult patients with PAH can still deteriorate despite significant reduction in PVR23, and survival has been shown to be significantly associated with changes in RV ejection fraction as opposed to PVR or CO.24 Based on these observations, we find MCT and SuHx to be appropriate models for understanding the molecular fingerprints of maladaptive RVH as opposed to adaptive RVH. In this study, we defined maladaptive RVH and RV failure from MCT and SuHx rats by significant elevation of RVSP, decrease in PAT and PAT/PET ratio, RV dilation and fibrosis, which were confirmed prior to RNA sequencing. Thus, our results provide a comprehensive overview of genes involved in maladaptive RVH, but identifying key molecular determinants in the transition from adaptive to maladaptive RV remodeling are still needed.
Identification of top up-regulated genes in RV failure
Based on our differential expression analysis, the top up-regulated genes in the RV were SPP1 (Osteopontin, OPN) and AHRR (Aryl-Hydrocarbon Receptor Repressor) from MCT and SuHx rats, respectively. Previous reports have shown OPN, a matricellular cardiac fetal gene that is reactivated during cardiac stress, to be elevated in RV of MCT-induced RV failure rats, which can be reversed by estrogen.14,25 OPN deletion has been shown to attenuate collagen deposition and deteriorate systolic function in the heart of angiotensin II-induced cardiac hypertrophy mouse model.26 Also, RV remodeling and failure has been associated with extensive fibrosis, which is tightly linked to transformation growth factor beta (TGF-β) signaling, and changes in expression of extracellular matrix proteins and metalloproteinases.27 We found that immunostaining of OPN in human RV tissue of patients with RV failure secondary to PAH localizes to interstitial and perivascular fibrotic areas (data not shown). Thus, these observations suggest a potential role of OPN in PAH-induced RV fibrosis.
Previous studies indicated that AHRR regulates AHR (aryl-hydrocarbon receptor), a basic helix-loop-helix transcription factor that acts as a receptor for endogenous metabolites and xenobiotics such as cigarette smoking.28 AHRR mRNA is highly expressed in heart and brain29, and plays a role in monocyte-to-macrophage differentiation30, suppression of anti-inflammatory mediators31, and tumor biology.32 The role of AHRR in PAH and RV failure is unclear. Interestingly, studies have shown AHR and TGF-β signaling pathways to cross-regulate each other in a cell type-specific manner.33,34 Given the role of TGF-β signaling in RV fibrosis, AHRR elevation may be involved in RV maladaptive remodeling in the setting of PAH.
EMT/EndMT activation in RV failure secondary to PAH
Based on our Hallmark enrichment analysis of RV from MCT and SuHx models, we identified up-regulated genes that were highly enriched in functions associated with EMT. EMT and EndMT play an integral role in early cardiovascular development, and are reactivated in numerous chronic cardiovascular disease states.6,35 EMT/EndMT have been well characterized in myocardial fibrosis and diastolic dysfunction in pressure-overload rodent models7,36, and play a critical role in tissue fibrosis and pulmonary vascular remodeling in PAH.37 Given the evidence of EndMT in lungs of PAH patients37 and cardiac fibrosis, and evidence of EndMT in RV of SuHx and MCT rats (Figure 6), we hypothesized that EndMT is most likely involved in RV failure of PAH patients. In this study, we identified five patients who met hemodynamic criteria for right-sided heart failure and either WHO group 1 (n=4) or group 3 (n=1) PH classification. MCT and SuHx models are considered more reflective of maladaptive RV hypertrophy in patients with WHO group 1 PAH.2 We observed activation of EndMT by immunofluorescence using myofibroblast (α-SMA) and endothelial (VWF) markers. Further analysis of genes associated with EMT based on Hallmark gene set showed SPP1 and CCN2 as the common top up-regulated genes in RV failure from MCT and SuHx rats. CCN2, also known as connective tissue growth factor (CTGF), is a matricellular protein that is induced in the heart following cardiac injury38,39, and has been associated with the development of tissue fibrosis.40 We observed increased CTGF immunolabeling and protein expression in RV of SuHx and MCT rats (Figure 7). In human RV sections, we observed an increased expression of CTGF in perivascular areas. Our results are in agreement with a recent study showing up-regulation of CTGF in PAH-mediated RV failure samples compared to non-failing RV by immunoblot.41 Our findings using human RV tissue further validate the RV transcriptome from MCT and SuHx models, and highlight the importance of EMT/EndMT in RV failure secondary to PAH.
The role of inflammation in RV failure secondary to PAH
In our data, pro-inflammatory pathways were amongst some of the most up-regulated pathways in both models. Pro-inflammatory cytokines such as TNF-α, IL-1, and IL-6 have been associated with the development and progression of RV failure in PAH animal models.15, 42–45 In MCT rats and PAB mice, expression of TNF-α in RV was increased46 and contributed to adverse ventricular remodeling and dysfunction.47 Decompensated RV failure from MCT model was also associated with increased TNF-α serum levels compared to compensated RV hypertrophy, suggesting a role of TNF-α as a biomarker for RV disease progression.42,48 In addition, activation of NFkB has been associated with the development of cardiac hypertrophy and transition to heart failure.49,50 A recent study showed an up-regulation of NFkB in RV of MCT model, which was modulated by exercise.44 Attenuation of cardiovascular hemodynamics and RV hypertrophy in MCT-induced PAH by TNF-α antagonist suggests NFkB to be an important molecular mediator of TNF-α signaling.51
The role of metabolism in RV failure from PAH
We also identified multiple down-regulated genes in RV of MCT and SuHx rats that were involved with metabolism and fatty acid oxidation (FAO). During the process of RV adaptation and remodeling, there is a notable glycolytic shift in the RV of humans with PAH52 and animal models of PAH.53 For instance, RV hypertrophy from MCT- and PAB-induced PAH causes an increase in glycolysis and Glut1 expression, and pyruvate dehydrogenase kinase (PDK)-mediated inhibition of pyruvate dehydrogenase (PDH).53 This PDK-mediated metabolic switch is associated with impaired RV contractility and decreased cardiac output, and electrical remodeling.53,54
FAO involvement in metabolic remodeling in RV failure remains unclear. A reciprocal relationship between FAO and glucose oxidation has been described (Randle’s Cycle) in RVH, where inhibiting FAO and thereby increasing glucose oxidation can limit cellular damage.55,56 In fact, recent studies using FAO inhibitors, trimetazidine and ranolazine, showed improvement of RV function by increasing glucose oxidation in PAB55 and MCT models.57 Another study in MCT-induced PAH rats showed impaired mitochondrial function and ADP channeling within the RV, and decreased ratio of mitochondria per myofibrillar proteins58, which supports the role of energy metabolism in contributing to contractile dysfunction in the RV.59
Limitations
In this study, our RV transcriptomic signature represents maladaptive RV remodeling from hemodynamic pressure overload induced by MCT and SuHx. However, we cannot exclude the possibility that the direct toxic effects of MCT and SU5416 may have on the RV expression profile. Although both animal models induce PAH by different mechanisms, we identified significant overlap of DEGs and enriched pathways in the RV of MCT and SuHx rats. Given the similar expression profiles between MCT and SuHx rats, the development of RV failure was most likely initiated by the hemodynamic overload as opposed to direct systemic effects from endothelial toxins.
We also observed mild variability in expression profiles from animals exposed to SuHx as opposed to MCT. Previous reports showed hemodynamic variations in response to SuHx in different strains of rats or between colonies of the same strain.60 A study using a similar protocol as our present study showed maximum RVSP and RV hypertrophy at 5-weeks and 3-weeks post-SU5416 injection in rats, respectively, followed by regression during normoxia exposure.61 In this study, the exposure to normoxia prior to isolation of RV at 5-weeks post-injection may have attributed to the observed variability in RV hemodynamics and expression profiles between rats.
We acknowledge that our human RV tissue section from patients with PAH is a heterogenous group. It is plausible that patient’s comorbidities such as systemic scleroderma may have contributed to the variability in histological staining. In addition, availability of normal RV sections was limited, and we observed prominent age-related cardiac fibrosis in one control patient. Direct comparison of expression profiles between MCT/SuHx and human would be ideal. Unfortunately, human RV tissue for RNA sequencing and public genomics data was not available to us.
Conclusions
Our RV transcriptomic signature of RV failure in MCT and SuHx animal models showed similar patterns of gene expression changes and biological pathways. Despite the inherent differences in PAH induction by MCT and SuHx models, the underlying pathways involved in RV maladaptation in response to pressure-load are similar. Histological confirmation of EndMT pathway and up-regulation of CTGF (CCN2) protein in human PAH-induced RV failure patients provide further validation of our RV transcriptomic findings. Targeting specific molecular mechanisms responsible for RV failure in both MCT and SuHx animal models may identify novel therapeutic strategies for PAH-associated RV failure.
Supplementary Material
Supplemental Material
Supplemental Tables
Added Author Consent
Non-standard Abbreviations and Acronyms
PAH | Pulmonary arterial hypertension |
RV | Right ventricle |
PVR | Pulmonary vascular resistance |
EMT | Epithelial-to-mesenchymal transition |
EndMT | Endothelial-to-mesenchymal transition |
MCT | Monocrotaline |
SuHx | Sugen hypoxia |
RVSP | Right ventricular systolic pressure |
PBS | Phosphate buffered saline |
FDR | False discovery rate |
PAT | Pulmonary artery acceleration time |
RVIDd | RV internal diameter at end-diastole |
FAO | Fatty acid oxidation |
CTGF | Connective tissue growth factor |
References
Full text links
Read article at publisher's site: https://doi.org/10.1161/circheartfailure.120.007058
Read article for free, from open access legal sources, via Unpaywall: https://www.ahajournals.org/doi/pdf/10.1161/CIRCHEARTFAILURE.120.007058
Citations & impact
Impact metrics
Article citations
Pathophysiology of the right ventricle and its pulmonary vascular interaction.
Eur Respir J, 64(4):2401321, 31 Oct 2024
Cited by: 0 articles | PMID: 39209482 | PMCID: PMC11525331
Review Free full text in Europe PMC
Pulmonary Hypertension and Right Ventricle: A Pathophysiological Insight.
Clin Med Insights Cardiol, 18:11795468241274744, 09 Sep 2024
Cited by: 0 articles | PMID: 39257563 | PMCID: PMC11384539
Review Free full text in Europe PMC
Mast Cells in Cardiac Remodeling: Focus on the Right Ventricle.
J Cardiovasc Dev Dis, 11(2):54, 04 Feb 2024
Cited by: 0 articles | PMID: 38392268 | PMCID: PMC10889421
Review Free full text in Europe PMC
BBLN triggers CAMK2D pathology in mice under cardiac pressure overload and potentially in unrepaired hearts with tetralogy of Fallot.
Nat Cardiovasc Res, 2(11):1044-1059, 26 Oct 2023
Cited by: 0 articles | PMID: 38666071 | PMCID: PMC11041739
Multi-omic and multispecies analysis of right ventricular dysfunction.
J Heart Lung Transplant, 43(2):303-313, 01 Oct 2023
Cited by: 2 articles | PMID: 37783299
Go to all (17) article citations
Data
Data behind the article
This data has been text mined from the article, or deposited into data resources.
BioStudies: supplemental material and supporting data
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
Identification of Long Noncoding RNA H19 as a New Biomarker and Therapeutic Target in Right Ventricular Failure in Pulmonary Arterial Hypertension.
Circulation, 142(15):1464-1484, 23 Jul 2020
Cited by: 77 articles | PMID: 32698630
Analyses of long non-coding RNA and mRNA profiles in right ventricle myocardium of acute right heart failure in pulmonary arterial hypertension rats.
Biomed Pharmacother, 106:1108-1115, 17 Jul 2018
Cited by: 16 articles | PMID: 30119177
Ultrastructural Changes of the Right Ventricular Myocytes in Pulmonary Arterial Hypertension.
J Am Heart Assoc, 8(5):e011227, 01 Mar 2019
Cited by: 17 articles | PMID: 30807241 | PMCID: PMC6474942
Epigenetic Regulation of Pulmonary Arterial Hypertension-Induced Vascular and Right Ventricular Remodeling: New Opportunities?
Int J Mol Sci, 21(23):E8901, 24 Nov 2020
Cited by: 11 articles | PMID: 33255338 | PMCID: PMC7727715
Review Free full text in Europe PMC