Super Enhancer-Associated Circular RNA-CircKrt4 Regulates Hypoxic Pulmonary Artery Endothelial Cell Dysfunction in Mice
Arteriosclerosis, Thrombosis, and Vascular Biology
Abstract
Background:
Circular RNAs (circRNAs) have been implicated in pulmonary hypertension progression through largely unknown mechanisms. Pulmonary artery endothelial cell (PAEC) dysfunction is a hallmark in the pathogenesis of pulmonary hypertension. However, the specific role of circular RNAs in PAEC injury caused by hypoxia remains unclear.
Methods:
In this study, using the Western blotting, RNA pull down, Dual-luciferase reporter assay, immunohistochemistry, and immunofluorescence, we identified a novel circular RNA derived from alternative splicing of the keratin 4 gene (circKrt4).
Results:
CircKrt4 was upregulated in lung tissues and plasma and specifically in PAECs under hypoxic conditions. In the nucleus, circKrt4 induces endothelial-to-mesenchymal transition by interacting with the Pura (transcriptional activator protein Pur-alpha) to promote N-cadherin gene activation. In the cytoplasm, increased circKrt4 leads to mitochondrial dysfunction by inhibiting cytoplasmic-mitochondrial shuttling of mitochondrial-bound Glpk (glycerol kinase). Intriguingly, circKrt4 was identified as a super enhancer-associated circular RNA that is transcriptionally activated by a transcription factor, CEBPA (CCAAT enhancer binding protein alpha). Furthermore, RBM25 (RNA-binding-motif protein 25) was found to regulate circKrt4 cyclization by increase the back-splicing of Krt4 gene.
Conclusions:
These findings demonstrate that a super enhancer-associated circular RNA-circKrt4 modulates PAEC injury to promote pulmonary hypertension by targeting Pura and Glpk.
Graphical Abstract

Highlights
•
A new circular RNA derived from alternative splicing of the keratin 4 gene (circKrt4) is involved in the pathogenesis of hypoxia-induced pulmonary hypertension.
•
In the nucleus, upregulated circKrt4 in hypoxia could result in excessive endothelial-to-mesenchymal transition by contributing to the induction of Pura (transcriptional activator protein Pur-alpha) binding to the N-cadherin gene promoter.
•
In the cytoplasm, circKrt4 interacts with Glpk (glycerol kinase) and inhibits Glpk mitochondrial translocation, leading to oxidative stress and mitochondrial dysfunction.
•
CEBPA (CCAAT enhancer binding protein alpha) is found to control circKrt4 expression in hypoxia by regulating super enhancer at the Krt4 locus.
•
CircKrt4 inhibition is a new therapeutic strategy for hypoxic pulmonary hypertension.
Endothelial cells (ECs) line the luminal surface of cardiovascular systems and are continually exposed to injurious stimuli by interactions with platelets and inflammatory cells.1 Hypoxia, toxins, genetic factors, inflammatory cytokines, and pathological shear stress factors can trigger EC injury.2 As a consequence, EC dysfunction, which is characterized by the loss of homeostatic functions, enhanced thrombotic lesions and neointima formation, and secretion of vasoconstrictive and proliferative factors, represents the major feature in the pathogenesis of pulmonary hypertension (PH).3–5
Endothelial-to-mesenchymal transition (EndoMT) describes a cell transdifferentiation phenomenon by which ECs convert into a mesenchymal or myofibroblastic phenotype.6 EndoMT was observed in both human patients with PH nd experimental PH models induced by monocrotaline or by combined exposure to hypoxia and VEGFR (vascular endothelial growth factor receptor) blockade (SU5416 combined with hypoxia [SuHx]), whereby cells express high levels of α-SMA (α-smooth muscle actin) and phospho-vimentin and activate the EndoMT transcription factors TWIST1 (twist-related protein 1) and SNAIL1 (snail family transcriptional repressor 1).7–9 Studies have reported that HIF (hypoxia-inducible transcription factor) and TGFβ (transforming growth factor β) signaling pathways play essential roles in driving the pathogenesis of EndoMT in PH.2,10,11 Despite this, the regulative mechanisms of EndoMT in hypoxic PH remain elusive. Simultaneously, mitochondrial dysfunction also plays an essential role in increased pulmonary vascular resistance and PH development by contributing to the apoptosis-resistant and migrative phenotype of ECs.12 It has been proved that glycolysis-relevant genes, including LDHA (lactate dehydrogenase A), HK1/2/3 (hexokinase isoforms), and PKM1/2 (pyruvate kinase muscle isoforms) were associated with cell proliferation through a glycolytic shift in PH.13–15 Our previous studies have reported that AIF (apoptosis-inducing factor), which regulates the mitochondrial oxidative phosphorylation, frequently decreased in pulmonary artery smooth muscle cells (PASMCs) in hypoxia.16 However, many gene functions have still not been carefully studied in mitochondria, the mechanisms of pulmonary artery endothelial cell (PAEC) mitochondrial dysfunction, especially impaired mitochondrial oxidative phosphorylation, and oxidative stress remain unclear. These limitations collectively hinder our understanding of the importance of mitochondrial homeostasis in PAECs and therefore a better exploration of the mitochondrial landscape in PH is required.
Circular RNAs (circRNAs) are emerging as a new class of endogenous noncoding RNAs that have covalently closed loop structures and higher tolerance to exonuclease digestion.17,18 CircRNA-mediated gene regulation has been shown to contribute to the pathological process of PH and could be tested as diagnostic and therapeutic targets. Yuan et al19 showed that the levels of circGSAP (circular RNA gamma-secretase activating protein), a circRNA derives from the gene GSAP that encodes gamma-secretase activating proteins were decreased in lung tissues, B lymphocytes, and monocytes from idiopathic pulmonary arterial hypertension patients and were positively correlated with both the 6-minute walking distance and cardiac output of idiopathic pulmonary arterial hypertension patients. Jin et al20 demonstrated that overexpression of hsa_circNFXL1_009, a circRNA generated from the NFXL1 gene, can sponge hsa-miR-29b-2-5p and inhibit excessive proliferation and migration of PASMCs under hypoxic conditions. Despite the important role of circRNAs in certain diseases and PH development, their function in EndoMT and PAEC mitochondrial dysfunction needs further study.
In the present study, we explored the function and implication of a circRNA derived from the keratin 4 (Krt4) gene locus, circKrt4. In contrast to general circRNAs that function as miRNA (microRNA) sponges, circKrt4 regulated by CEBPA (CCAAT enhancer binding protein alpha) and RBM25 (RNA-binding-motif protein 25) is enriched in the nucleus and cytoplasm, regulates the Pura (transcriptional activator protein Pur-alpha) and Glpk (glycerol kinase), promotes EndoMT and mitochondrial dysfunction, and ultimately promotes PAEC proliferation, migration, and hypoxic PH progression.
Materials and Methods
The data that support the findings of this study are available from the corresponding author upon reasonable request. Research materials are listed in the Major Resources Table in the Supplemental Material.
Animal Treatments and Lung Tissue Preparation
Since female sex hormone, for example, estrogen, can attenuate chronic hypoxia and SuHx-induced PH,21,22 adult male C57BL/6 mice (6–9 weeks old) weighing 20 to 25 g from the Laboratory Animal Center of the Second Affiliated Hospital of Harbin Medical University were employed in this study. All experimental procedures were performed in accordance with the ethical standards in the 1964 Declaration of Helsinki and its later amendments and approved by the Ethics Committees of Harbin Medical University (HMUDQ20220317001). The corresponding target RNA cloning construction and serotype 5 adenovirus-associated virus (AAV5) were packaged by Genechem (Shanghai, China). Mice were randomly divided into different groups and infected with the AAV5 vector at 1011 genome equivalents in 20 to 30 µL Hank balanced salt solution after isoflurane anesthesia, followed by nasal drops. After 7 to 14 days, the mice were assigned to normoxia (Fi, O2 0.21) and hypoxia (Fi, O2 0.10) as described in a previous study.16,23 The normoxic group was kept in the same place adjacent to the hypoxic group, and the concentration of oxygen was monitored continuously using an oxygen analyzer (P110, BioSpherix New York) for 4 weeks. For SuHx-mediated PH model, SU5416 (S8442, Sigma-Aldrich, Darmstadt, Germany) was injected to mice subcutaneously at 20 mg/kg, and control mice were injected with the same volume of vehicle alone during the next 3 weeks in 10% oxygen and followed by reexposure to normoxia 2 weeks. Lung tissues were taken for corresponding experiments after anesthesia, and the right ventricular (RV) hypertrophy index (ratio of RV free wall weight over the sum of septum (S) plus left ventricular (LV) free wall weight), RV/(LV+S) was calculated.
Echocardiography and RV Systolic Pressure Measurements
At the end of hypoxic and SuHx treatment, mice were subjected to echocardiography using a Vevo2100 imaging system equipped with a 18- to 38-MHz (MS400, mouse cardiovascular) transducer probe (VisualSonics, Inc, Toronto, Ontario, Canada), as previously described.16 Mice were anesthetized with inhaled isoflurane, administered at 1% to 3% by an Advanced Table Top Anesthesia machine (HARVARD APPARATUS LIMITED). Mice were secured on a prewarmed (37 °C) imaging platform and heart and respiratory rates were observed continuously throughout the echocardiography procedure. Pulsed-wave Doppler was used to measure pulmonary artery acceleration time and the pulmonary artery velocity time integral. Pulmonary artery acceleration time was measured from the pulsed-wave Doppler flow velocity profile of the RV outflow tract in the parasternal short-axis view and was defined as the interval from the onset to the maximal velocity of forward flow. LV ejection fraction was measured at the parasternal short-axis view, the LV area was traced during end diastole and end systole, and the LV ejection fraction was calculated with: LV ejection fraction (%)=(LVEDV [left ventricular end-diastolic volume]−LVESV [left ventricular end-systolic volume])/LVEDV×100. All measurements were performed by the same experienced investigator. Off-line data analysis was performed using Vevo2100 software by investigator in a blind manner.
The RV systolic pressure was measured with PowerLab monitoring equipment (AD Instruments, Colorado Springs, CO). A 1.2 French Pressure Catheter (Scisense Inc) was inserted into the superior vena cava and finally into the RV vein, and the RV systolic pressure was continuously recorded for 20 to 40 minutes.
Morphometric Analysis
Hematoxylin and eosin staining and Masson trichrome staining were performed, as previously described.16 In brief, lung tissues of mice were immersed in 4% paraformaldehyde for 48 hours. After dehydration, clearing, and embedding in paraffin wax, the blocks were cut into 5-μm–thick sections and stained as appropriate. For immunohistochemistry, the sections were incubated with α-SMA (Boster, Wuhan, China). Then, the tissue sections were stained with 3,3-diaminobenzidine and restrained with hematoxylin. The total wall thickness and positive staining area in the vascular walls were quantified using a color-recognition algorithm in Image-Pro Plus 6.0 software.
Cell Culture and Cell Counting Kit 8 Assay
At least 4 different batches of PAECs (catalog no. CP-M011) from male mice were provided by Procell Life Science & Technology (Wuhan, China). Cells were maintained in endothelial cell medium (ScienCell, 1001, CA) containing 15% FBS and 1% penicillin streptomycin at 37 °C, 5% CO2, and 100% relative humidity. A tri-gas incubator (Thermo Fisher, MA) with an atmosphere comprising 3% O2, 5% CO2, and 92% N2 was used for hypoxic culture. All experiments were performed with cells less than passages 4. For the Cell Culture and Cell Counting Kit 8 assay, PAECs were seeded in 96-well plates and treated according to the different experimental groups for 24 to 48 hours. After adding 10 µL, Cell Culture and Cell Counting Kit 8 (Biosharp, Hefei, China) reagent to each well for 2 hours, the results were detected at 450 nm wavelength using a spectrophotometer.
Ethynyl-2’-Deoxyuridine Incorporation and Flow Cytometry Assay
PAECs were treated with 50 μmol/L 5-ethynyl-2’-deoxyuridine (C0071S, Beyotime, Shanghai, China) and incubated for 4 hours at 37 °C. The cells were fixed with 4% formaldehyde for 20 minutes and exposed to 0.5% Triton X-100 for 15 minutes. 4′,6-diamidino-2-phenylindole (5 μg/mL) was used to stain the DNA of cells in each well for 30 minutes. Then, the images were observed under a fluorescence microscope, and the results are shown as the percentage of proliferating cells (ethynyl-2’-deoxyuridine–positive cells) with 4′,6-diamidino-2-phenylindole. To assess the cell cycle distribution in the different experimental groups, a Cycle Test Plus DNA Reagent Kit (C1052, Beyotime, Shanghai, China) was used. Cells were fixed with 70% ethanol and exposed to 500 µL of propidium iodide. A BD FACSCalibur flow cytometer was used to measure DNA fluorescence.
Cell Transfection
Cell transfection was performed according to the manufacturer’s instructions for the Lipofectamine 2000 Reagent (11668019; Thermo Fisher, MA), and the procedure for transfection was described previously.16 Small interfering RNA (siRNA) against Pura, CEBPA, RBM25, and nontargeted control siRNA were designed and synthesized by GenePharma (Suzhou, China). The sequences are shown in Table S1. Glpk overexpression and mutant plasmids were constructed using the GV658 vector by GeneChem (Shanghai, China), and the vector alone was used as a negative control (NC).
RNA Isolation and Quantitative Real-Time Polymerase Chain Reaction
Total RNA samples were extracted from cultured PAECs and lung tissues using TRIzol reagent (Thermo Fisher, MA) according to the manufacturer’s protocol. RNA was extracted from blood/plasma using a Blood/Plasma RNA Extraction Kit (HaiGene, Harbin, China) according to the manufacturer’s protocol. Cytoplasmic and nuclear RNAs were isolated and purified using Norgen’s Cytoplasmic & Nuclear RNA Purification Kit (Thorold, ON, Canada). According to the manufacturer’s instructions, PAECs were lysed with ice-cold lysis buffer, cytoplasmic and nuclear RNA were bound to the column, and the mixture was separated by RNA elution. For each sample, 500 ng of total RNA was converted to cDNA using a Superscript First-Strand cDNA Synthesis Kit (HaiGene, Harbin, China). Real-time polymerase chain reaction (PCR) was performed in a LightCycler 480 II real-time PCR system (Roche, Basel, Switzerland) with SYBR Green (TOYOBO, Osaka, Japan). The threshold cycle (Ct) was determined, and the data were analyzed using the 2-ΔΔCT method. The primers (5′-3′) are shown in Table S2.
Western Blot Analysis
Protein samples (20–50 μg) extracted from lung tissues and PAECs were separated by SDS–PAGE (8%–12%) and transferred onto nitrocellulose membranes. The membranes were blocked in 5% nonfat dry milk for 1 hour and then incubated with the indicated antibodies (see Major Resources Table in the Supplemental Material for information about the antibodies) overnight at 4 °C. After washing with TBST buffer, the membranes were incubated with horseradish peroxidase-conjugated secondary antibodies and subjected to chemiluminescence reagent imaging.
Endothelial Cell Migration and Network Formation Assay
For the transwell assay, resuspended PAECs were added to the upper chamber of a modified Boyden chamber (Costar, Corning, NY) with a polycarbonic membrane (6.5 mm in diameter and 8 µm pore size). The migrated cells were fixed with 4% paraformaldehyde and stained with 0.4% crystal violet, and the number of stained migrated cells was counted under an inverted microscope (Nikon, Japan). For the network formation assay, 96-well culture plates were coated with 30 μL growth factor-reduced Matrigel (BD Biosciences, NY) for 30 minutes at 37 °C. PAECs were trypsinized and resuspended, and 200 μL of cell suspension was added to solidified Matrigel. Network formation of PAECs was observed under an inverted microscope and measured using Image-Pro Plus 6.0.
Immunofluorescence Staining and Fluorescent In Situ Hybridization
PAECs were fixed with 4% paraformaldehyde for 15 minutes and permeabilized with 0.01% Triton X-100. After blocking with 5% BSA, the cells were incubated with antibodies against N-cadherin (1:100), Pura (1:100), Glpk (1:100), and anti-DNA/RNA Damage (8-hydroxy-2 deoxyguanosine, 1:100) at 4 °C. The next day, the cells were incubated with CY3 (cyanine-3)/FITC (fluorescein isothiocyanate)–conjugated secondary antibody (1:100) at 37 °C for 2 hours and 4′,6-diamidino-2-phenylindole away from light for 10 minutes. The results of immunofluorescence staining were recorded by a live cell workstation (AF6000; Leica, Wetzlar, Germany). Fluorescent in situ hybridization (FISH) experiments for PAECs were performed using a Fluorescent In Situ Kit following the manufacturer’s instructions, and Cy3-conjugated circKrt4 probes were synthesized by RiboBio (Guangzhou, China). After treatment with agents according to the different experimental groups, PAECs were washed, fixed with 4% paraformaldehyde, and permeabilized with 0.3% Triton X-100. After the cells were blocked with prehybridization solution, they were incubated with hybridization solution containing circKrt4, 18S, and U6 probes followed by 4′,6-diamidino-2-phenylindole incubation. FISH experiments for tissues samples were performed using a circRNA FISH Probe Mix following the manufacturer’s instructions synthesized by EXONBIO (Guangzhou, China). Briefly, 5-μm-thick lung tissue sections were pretreated in PBS and incubated with digoxigenin-labeled denature circKrt4 probes in hybridization buffer at 37 °C overnight. Following incubation with CY3-labeled anti-digoxigenin antibodies in blocking buffer and 4′,6-diamidino-2-phenylindole, results of FISH staining were recorded by a live cell workstation (AF6000; Leica, Wetzlar, Germany).
Protein Coimmunoprecipitation and RNA Immunoprecipitation Assays
For the protein coimmunoprecipitation assay, lysed PAECs were collected and centrifuged at 15 000 revolutions per minute for 30 minutes at 4 °C to collect the supernatant, followed by the addition of 5 μg of target antibody or IgG and incubation at 4 °C for 6 hours. Protein A+G agarose beads were added overnight in a 4 °C shaker. The antibody-protein complexes were washed, and the pellet was resuspended in protein loading buffer (2×) followed by Western blotting. The RNA immunoprecipitation (RIP) assay was performed according to the manufacturer’s instructions (Bes5101; BersinBioTM, Guangzhou, China). Briefly, 1×107 PAECs were collected and lysed with RIP buffer. Then, the samples were incubated with protein A/G bead–conjugated anti-Pura (5 μg, GTX00901; GeneTex, CA), anti-Glpk (4 μg, 13360-1-AP; Proteintech, IL) and anti-RBM25 (1.5 μg, 25297-1-AP, Proteintech, IL) antibodies overnight at 4 °C. After extracting the RNA from immunoprecipitation and input samples, 1000 ng of the total RNA was converted to cDNA and subjected to quantitative real-time polymerase chain reaction (qRT-PCR) assays. The primers (5′–3′) are shown in Table S2.
Mitochondrial Oxidative Phosphorylation and Glycolysis Assays
The oxygen consumption rate (OCR, a hallmark of mitochondrial oxidative phosphorylation) and extracellular acidification rate (a hallmark of glycolysis) were measured using a 24-well extracellular flux analyzer (Agilent Technologies Co, Ltd) following the specific manufacturer’s protocol of the Seahorse XF Cell Mito Stress Test Kit and Seahorse XF Glycolysis Stress Test Kit (103015-100 and 103020-100; Agilent, CA). For OCR analysis, PAECs plated in an XFp cell culture miniplate (102340-100; Agilent, CA) and ATP synthase inhibitor oligomycin (trifluoromethoxy carbonylcyanide phenylhydrazone; 1 μM), which decreases OCR levels, followed by the electron transport chain accelerator FCCP (1 μM), which causes maximal respiration, and finally rotenone+antimycin A (both 1 μM), which are mitochondrial complex I and III inhibitors, were sequentially injected into cells. The results were calculated from the OCR values in picomoles per minute of oxygen consumed and were normalized with total protein concentration. For the glycolysis assay, cells were glucose starved in XF assay medium in a CO2-free XF prep station and then treated with glucose (2 mg/mL), oligomycin (1 μM), and 2-deoxy-D-glucose (100 mmol/L). All measurements were normalized with total protein concentration on each well and differences in glycolytic capacity were represented as mpH/min per protein.
Dual-Luciferase Reporter Assay
A dual-luciferase reporter assay was performed according to the protocol of the Dual-Lumi Luciferase Reporter Gene Assay Kit (RG088S, Beyotime, Shanghai, China). The Krt4 promoter fragment containing the CEBPA binding site was cloned into the GV238 plasmid expressing luciferase (GenePharma, Suzhou, China). PAECs were cotransfected with the CEBPA expression plasmid and Krt4 plasmid expressing luciferase with Lipofectamine 2000 for 48 hours. Then, the luciferase activities were measured by the dual-luciferase reporter assay system (GloMax Multi Detection System, Promega, Madison).
Reactive Oxygen Species Detection
For intracellular reactive oxygen species (ROS) assay, fresh media containing 10 μM 2′,7′-dichlorodihydroflfluorescein diacetate probes (S0033S, Beyotime, Shanghai, China) was treated for cells in a 37 °C humidified incubator. Green fluorescence was captured (488 nm excitation/525 nm emission) from ≥3 optical fields. Mitochondrial ROS activity was measured with a redox-sensitive fluorescent probe, mitochondrially targeted superoxide indicator (MitoSOX Red; M36008; Thermo Fisher, MA). PAECs were incubated with 5 µmol/L MitoSOX probe for 20 minutes. The cells were washed with PBS, and red fluorescence was captured (514 nm excitation/585 nm emission) from ≥3 optical fields. Total cellular ROS and mitochondrial ROS in the fluorescence intensity of treated cells was quantified by Image J.
Superoxide Dismutase and Glutathione Peroxidase Activity Assay
SOD (superoxide dismutase) activity was measured using a colorimetric CuZn/Mn-SOD activity assay kit (S0103, Beyotime, Shanghai, China). Briefly, 20 µL of the sample was mixed with WST working solution and enzyme working solution, and activities of SOD were calculated by measuring the absorbance at 450 nm following incubation at 33 °C for 20 minutes. GPx (glutathione peroxidase) activity was measured using a GPx activity assay kit (S0058, Beyotime, Shanghai, China). The collected cells were washed and lysed with 200 µL cold Assay Buffer. Following centrifugation 15 minutes at 4 °C at 10 000×g to remove any insoluble material, 10 µL sample was added to the mixture reaction system (40 mmol/L NADPH solution, glutathione reductase, and glutathione). The concentration of NADPH was measured at 340 nm at 0 and 5 minutes after cumene hydroperoxide addition, and GPx activities were calculated from the changes in NADPH concentrations.
Electrophoretic Mobility Shift Assay
The specific biotinylated probe containing the Krt4 promoter (−2000/−1580) fragment was synthesized by GenePharma (Suzhou, China). The Electrophoretic Mobility Shift Assay was performed following the instructions of a chemiluminescent Electrophoretic Mobility Shift Assay Kit (GS009, Beyotime, Shanghai, China). The probe and the extracted nuclear protein were reacted in binding reaction buffer for 20 minutes. Then, the sample was added to a 4% nondenatured polyacrylamide gel for electrophoresis purposes and transferred onto a nylon membrane, followed by UV cross-linking at 245 nm. Finally, the membrane was incubated with streptavidin-horseradish peroxidase conjugate, followed by enhanced chemiluminescent reagent imaging.
Chromatin Immunoprecipitation–Quantitative PCR
A chromatin immunoprecipitation (ChIP) assay was performed using a ChIP assay kit (P2078, Beyotime, Shanghai, China). Briefly, PAECs were cross-linked with a final concentration of 1% formaldehyde and quenched by a final concentration of 125 mmol/L glycine. After the cells were lysed with SDS supplemented with PMSF (phenylmethanesulfonyl fluoride), they were lysed by ultrasound to generate chromatin fragments of 100 to 1000 base pair. Antibodies against acetyl H3K27 (H3K27ac [acetyl-histone 3 lysine 27]; 1:100, A7253, ABclonal, Wuhan, China), monomethyl H3K4 (H3K4me1 [methyl-histone 3 lysine 4]; 1:100, A2355, ABclonal, Wuhan, China), CEBPA (3.75 μg, 18311-1-AP, Proteintech, IL), Pura (5 μg, GTX00901, GeneTex, SA), and normal rabbit IgG were used for immunoprecipitation. The cross-linking between histone and DNA was then removed. A DNA purification kit (D0033; Beyotime, Shanghai, China) was used for DNA purification. Finally, qRT-PCR was performed to measure the enrichment of H3k27ac, H3K4me1, CEBPA, and Pura to DNA molecules of interest. The primers (5′–3′) are shown in Table S2.
RNA Pull-Down Assays
Biotinylated RNA pull-down assays were performed using the BersinBioTM RNA pull-down Kit (Bes5102, Guangzhou, China) according to the manufacturer’s protocol. A total of 1×107 collected cells were lysed, and nucleic acids were removed by incubating with DNase at 25 °C. After adding streptavidin beads to prewash, RNA probes with streptavidin beads were added after washing to form probe and streptavidin beads complexes. Finally, the eluent was added to a shaker at 37 °C for 2 hours, and 30 μL collected protein samples bound to RNA were used for Western blotting.
Statistical Analysis
Statistical analysis was performed with GraphPad Prism 8 software. Data were checked for normal distribution and equal variance (F test) before statistical testing. Student t test (unpaired) was used for 2-group analysis with equal variance, and Welch correction test was used for 2-group analysis with unequal variance. One-way ANOVA with Tukey post hoc test was used to compare multiple groups with equal variance, and Brown-Forsythe and Welch ANOVA with Tamhane T2 post hoc test was used to compare multiple groups with unequal variance. Nonparametric analyses, including the Mann-Whitney U test for 2 groups or Kruskal-Wallis test followed by Dunn post test for >2 groups, were performed for nonnormally distributed data. Data are presented as the means±SEM, and P<0.05 was considered statistically significant.
Results
CircKrt4 Is a Bona Fide CircRNA Upregulated in Hypoxia
CircKrt4 was identified by circRNA sequencing from our previous study24 with log2 (fold change=7.28) and P<0.001. CircKrt4 was generated from the Krt4 gene and located on mouse chromosome 15 (101920551-101922975), and the circular structure of circKrt4 is illustrated in Figure 1A. Sanger sequencing further confirmed the head-to-tail junction, which was consistent with mmu_circKrt4 annotation (Figure 1B). We then revealed that circKrt4 amplified by divergent primers was detectable in cDNA rather than genomic DNA (Figure 1C). As shown in Figure 1D, compared with linear Krt4, circKrt4 was more resistant to RNase R digestion, confirming that circKrt4 was a bona fide circRNA. qRT-PCR assays verified that the expression of circKrt4 was higher in hypoxic lung tissues and plasma than in normoxia (Figure 1E). Next, we performed FISH analysis to determine the distribution of circKrt4 in lung tissues and found that circKrt4 was expressed in pulmonary arteries (Figure 1F). Furthermore, we found that circKrt4 was expressed in both PASMCs and PAECs, with significantly increased expression in PAECs exposed to hypoxia (Figure 1G). By subcellular fractionation and RNA FISH analysis, we clarified the expression of circKrt4 both in the cytoplasm and nucleus, which was upregulated by hypoxia exposure (Figure 1H and 1I).

Silencing CircKrt4 Inhibited Hypoxia-Induced PH In Vitro and In Vivo
To assess the function of circKrt4 in hypoxic PH, we designed loss-of-function assays by silencing circKrt4 in PAECs via circKrt4 ASO (antisense oligonucleotide; Figure S1A). The qRT-PCR results showed that ASO-circKrt4 effectively inhibited circKrt4 expression in PAECs (Figure S1B). Cytoplasmic and nuclear RNA isolation further confirmed the decreased level of circKrt4 by ASO-circKrt4 both in the cytoplasm and nucleus (Figure S1C). We found that linear Krt4 was upregulated under hypoxic conditions and that ASO-circKrt4 had no significant effect on the expression of Krt4 at the mRNA and protein levels (Figure S1D and S1E). Due to the essential role of HIFs (hypoxia-inducible factors) in PH, especially HIF-2 constitutes a universal mechanism of endothelium adaptation to prolonged hypoxia.25,26 We detect the role of circKrt4 in the regulation of HIF-2α in PAECs and found that the expression of HIF-2α was not affected by ASO-circKrt4 (Figure S1F). CCK-8 (Figure 2A) and ethynyl-2’-deoxyuridine assay (Figure 2B) were performed and showed that circKrt4 silencing retarded hypoxia-induced proliferation of PAECs. Western blotting analysis showed that hypoxia increased the expression of PCNA (proliferating cell nuclear antigen), cyclin D, and cyclin E, which was reversed by knockdown of circKrt4 under the same conditions (Figure 2C). Additionally, the effects of circKrt4 on cell cycle progression were verified by flow cytometry, in which circKrt4 silencing reduced the percentage of cells in S+G2/M phase under hypoxic conditions (Figure 2D).

To determine the role of circKrt4 in hypoxic PH in vivo, we used AAV5 to interfere with circKrt4 and constructed a hypoxia-PH model and a SuHx-PH model. We evaluated RV systolic pressure (Figure 2E; Figure S2A) and RV hypertrophy (Figure 2F; Figure S2B), and found that inhibition of circKrt4 inhibited the above PH index induced by hypoxia and SuHx. According to echocardiographic measurements, circKrt4 silencing attenuated the decrease of pulmonary artery velocity time integral and pulmonary artery acceleration time induced by hypoxia and SuHx which had no affect on LV ejection fraction (Figure 2G; Figure S2C). Furthermore, we found that knockdown of circKrt4 reversed hypoxia and SuHx-induced distal pulmonary vascular remodeling by measuring vascular wall thickening, vascular muscularization (Figure 2H; Figure S2D and S2E), and collagenation (Figure S2F). The interference efficiency of AAV5-circKrt4 is shown in Figure S2G, and the body weight of experimental animals was not affected (Figure S2H). The results suggested that circKrt4 plays a critical role in promoting PAEC proliferation and PH progression associated with hypoxia.
Role of CircKrt4 in PAEC Dysfunction
To explore the function of circKrt4 in PAEC dysfunction, we first sought to characterize the altered cellular phenotypes-EndoMT and the cell migration ability. Knockdown of circKrt4 significantly reversed hypoxia-induced decreased expression of E-cadherin (epithelial-cadherin) and increased expression of N-cadherin (neural-cadherin), Snail, and Vimentin (Figure 3A). Similarly, N-cadherin staining indicated that PAEC EndoMT was markedly decreased upon circKrt4 silencing (Figure 3B). Transwell and network formation assays revealed that circKrt4 deficiency inhibited the migration abilities of PAECs (Figure 3C). Increasing evidence demonstrates that mitochondrial injury and metabolic reprogramming are observed during epithelial-mesenchymal transition.27,28 To evaluate the role of circKrt4 in mitochondrial function in PAECs, oxidative stress, and metabolic changes were determined. Hypoxia-induced oxidative damage, as evidenced by fluorescence staining and ELISA assays demonstrating increases in 8-hydroxy-2 deoxyguanosine, mitochondrial matrix O2- and cellular ROS, and decreases in SOD and GPx activities when compared with those of the normoxic group, whereas, ASO-circKrt4 administration partly counteracted the afore-mentioned variations (Figure 3D through 3F). Besides, hypoxia upregulated the expression of HK II (hexokinase II) and PKM2 (pyruvate kinase 2) and decreased the expression of PDH (pyruvate dehydrogenase), which were reversed by circKrt4 knockdown (Figure 3G). In addition, there was a clear decline in OCR (maximal respiration, spare respiration capacity, and ATP production) in mitochondrial function after hypoxia exposure, while circKrt4 silencing treatment alleviated oxidative respiratory dysfunction (Figure 3H). Meanwhile, glycolysis was monitored in real time using a Seahorse XFe24 Extracellular Flux Analyzer. As expected, circKrt4 knockdown decreased the glycolytic stress activation stimulated by hypoxia (Figure 3I). These results indicated that upregulated circKrt4 in hypoxia could result in excessive EndoMT and mitochondrial dysfunction in PAECs.

CircKrt4 Interacts With Pura and Glpk
To investigate the molecular mechanism by which circKrt4 mediates hypoxic PH and PAEC dysfunction, we first predicted that circKrt4 may adsorb miRNAs based on bioinformatics (miRanda, pita, and RNAhybrid) and performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses (Figure S3A and S3B). qRT-PCR was used to detect the expression of miRNAs with the highest interaction scores, including miR-107-3p, miR-326-3p, miR-6972-5p, and miR-7665-3p. The results showed that there were no detectable changes in these target miRNAs after silencing circKrt4 in hypoxia (Figure S3C). Recent studies have shown that circRNAs can function as protein scaffolds and interact with RBPs (RNA-binding proteins).29 Therefore, we predicted target proteins interacting with circKrt4 by using the catRAPID program. Bioinformatics analysis predicted that the transcriptional activator protein Pura and mitochondrial-bound Glpk (glycerol kinase) could interact with circKrt4 (Figure 4A and 4B). We then used the HNADOCK Server to predict and visualize the 3-dimensional structural docking of circKrt4-Pura and circKrt4-Glpk (Figure 4C and 4D). To confirm such binding, RIP (RNA immunoprecipitation) experiments were performed and revealed that Pura and Glpk directly bound to circKrt4 in hypoxia, more obviously than the linear transcript Krt4 (Figure 4E and 4F). Then, according to the predicted binding site of circKrt4 and target genes (Figure S4A and S4B), we designed biotin-labeled specific probes to pull down the proteins directly bound to circKrt4, and the results showed that Pura and Glpk were indeed among the pulled protein complexes (Figure 4G). Consistent with these results, the FISH assay showed that circKrt4 colocalized with Pura and Glpk in the nucleus and cytoplasm, respectively (Figure 4H and 4I). Furthermore, as shown in Figure 4J and 4K, the expression of Pura and Glpk was increased in hypoxia; however, when circKrt4 was knocked down by ASO, the expression of Pura was markedly decreased, and the expression of Glpk was not affected.

CircKrt4 Regulated EndoMT via the Pura/N-Cadherin Axis
Knowing that circKrt4 is abundant both in the nucleus and cytoplasm, we first sought to examine the function of circKrt4-associated protein Pura in the nucleus. Therefore, siRNAs for Pura were synthesized, and siRNA-3 showed the highest efficiency for Pura silencing and was used in the following assay (Figure 5A). Accordingly, silencing Pura led to decreased migratory capability of hypoxic PAECs (Figure 5B). To investigate how Pura modulates PAEC function, we sought to identify the transcription target genes regulated by Pura, specifically EndoMT-associated proteins. Immunoblotting assays demonstrated that the expression of EndoMT key regulators, including E-cadherin, Snail, and Vimentin, remained unaltered after Pura siRNA transfection (Figure 5C). However, Pura knockdown significantly inhibited the expression of N-cadherin in PAECs (Figure 5C and 5D), suggesting a functional connection between Pura and N-cadherin. To clarify whether Pura affects N-cadherin transcription, we divided the N-cadherin promoter into 5 segments (P1–P5; Figure 5E). The ChIP-qPCR results verified that Pura directly bound to promoter (P) 1 (P1) and P3 within the N-cadherin promoter in hypoxic PAECs, which was reduced by silencing circKrt4 (Figure 5F). Together, the data demonstrated that Pura, as a transcription factor, is required for hypoxia-induced N-cadherin and EndoMT activation.

CircKrt4 Affected Glpk Translocation Through the TRFLV Motif to Regulate Mitochondrial Homeostasis
Next, we studied the role of circKrt4 in the cytoplasm, especially in mitochondria associated with Glpk. Having determined that knockdown of circKrt4 did not affect the expression of the Glpk protein in cells (Figure 4K), the interaction of circKrt4 and Glpk prompted us to determine the role of circKrt4 in the mitochondrial translocation of Glpk. We then examined the expression of Glpk through cytoplasmic and mitochondrial protein isolation and found that the mitochondrial localization of Glpk decreased under hypoxia, and when circKrt4 was silenced, the translocation of Glpk into mitochondria was significantly increased (Figure 6A). Accordingly, an immunofluorescence assay showed that knockdown of circKrt4 promoted Glpk mitochondrial translocation in PAECs (Figure 6B), suggesting that the interaction of circKrt4 and Glpk blocked the mitochondrial shuttling of Glpk in hypoxia. Then, we identified a mitochondria targeting sequence in the Glpk protein using the PSORT II prediction tool30 and found that there was a TRFLV (twin-arginine translocation–related flanking region with conserved Leu-Val residues) motif in the N-FGGY domain of Glpk (Figure 6C). To validate this prediction, we constructed a Glpk-overexpressing plasmid with an unaltered mitochondrial targeting motif (wild-type) and a mutated mitochondrial targeting motif (MUT) and confirmed the overexpression plasmid transfection efficiency (Figure S5A). When the TRFLV motif was mutated, Glpk translocation to mitochondria was diminished compared with wild-type–Glpk (Figure 6D). Then, we tested the influence of the translocation of Glpk from the cytoplasm to mitochondria on PAECs. Glpk mitochondrial translocation inhibition (MUT-Glpk) induced increased levels of mitochondrial ROS, total intracellular ROS, and oxidative DNA damage (8-hydroxy-2 deoxyguanosine; Figure 6E through 6G). In addition, there was a clear decline in maximal respiration capacity, spare respiration, and ATP production in mitochondrial function after transfection with the MUT-Glpk plasmid compared with wild-type–Glpk (Figure 6H). There was no significant difference in the extracellular acidification rate of the glycolysis index in the PAECs after mitochondrial targeting motif mutation (Figure 6I). As shown in Figure S5B and S5C, MUT-Glpk increased the proliferation of PAECs. These molecular data demonstrated that Glpk mitochondrial translocation inhibition resulted in PAEC dysfunction by inducing mitochondrial injury.

CEBPA Controls CircKrt4 Expression in Hypoxia by Regulating H3K27ac and H3K4me1 Enrichment at the Krt4 Locus
Super enhancer (SE) is a large cluster of enhancers that drives gene transcription, including protein-coding genes and noncoding RNAs, by binding transcription factors and coactivators.31,32 Analysis of SE active histone marker H3K27ac ChIP-seq data generated in mouse small-cell lung cancer identified a SE around Krt4 (Figure S6A), suggesting that Krt4 may be regulated by SE. To determine whether hypoxia-induced upregulation of circKrt4 expression is associated with SE in PAECs, we treated cells with JQ1 (BET bromodomain inhibitor), a SE inhibitor, and found that JQ1 specifically inhibited the expression of both Krt4 and circKrt4 in hypoxia (Figure 7A). ChIP-qPCR assays revealed that hypoxia promoted the binding of the H3K27ac to the individual constituent promoter (P1–P5) of Krt4 and H3K4me1 enriched in the promoter (P2, P3, and P5) of Krt4 in PAECs (Figure 7B). We next attempted to identify candidate transcription factors that contribute to the aberrant activation of circKrt4 in hypoxia. Knowing that Pura interacts with circKrt4 as a transcription factor (Figure 4), we first sought to examine whether the expression of circKrt4 is related to Pura. qRT-PCR showed that knockdown of Pura did not affect the expression of circKrt4 (Figure S6B). After, according to an epigenetic database (CHIP-Atlas) and promoter databases (JASPAR and AnimaiTFDB), we identified CEBPA, which could potentially combine with the promoter region of Krt4 (Figure 7C and 7D). Although the expression of CEBPA did not change in PAECs after hypoxia treatment (Figure 7E), ChIP-qPCR assays revealed that CEBPA could directly bind to the individual constituent promoter (P5) of Krt4 in PAECs under hypoxia (Figure 7F). The coimmunoprecipitation assay results verified the interaction of CEBPA with H3K4me1 and H3K27ac (Figure 7G). Electrophoretic Mobility Shift Assay results of nuclear proteins extracted from PAECs showed that the addition of a CEBPA-specific antibody in the binding reaction led to a super shifted band, indicating that CEBPA was indeed complexed with Krt4 probe (Figure 7H). Meanwhile, PAECs treatment with CEBPA siRNA (Figure S6C) counteracted hypoxia-mediated upregulation of Krt4 and circKrt4 (Figure 7I). We next cloned the promoter region of Krt4 into luciferase reporter vectors and measured their activities. The wild-type but not P5 MUT promoter region was activated by CEBPA in PAECs (Figure 7J). All of the above results indicated that circKrt4 expression is tightly regulated by the interaction between CEBPA and Krt4-acossiated SE (H3K27ac and H3K4me1).

RBM25 Regulates CircKrt4 Cyclization by Increase the Alternative Splicing of the Krt4 Gene
To further explore the formation of circKrt4, we investigated the potential alternative splicing of the Krt4 gene by bioinformatic prediction (RBPmap and catRAPID) and highlighted candidate RBPs (RBM25 [RNA-binding-motif protein 25], KSHRP [KH-type splicing regulatory protein], RBM15B [RNA-binding motif protein 15B], UNK [unkempt family zinc finger], and MATR3 [matrin3]) possibly binding to the flanking sequences of exon 2 and exon 5 (intron 1 and intron 5) of Krt4, in which RBM25 (RNA-binding-motif protein 25) was reported to regulate a large number of alternating splicing exons and was confirmed to promote circRNA formation33 (Figure 7K). To assess whether RBM25 was indeed able to regulate Krt4 splicing, we performed RIP assay and the results revealed significant binding between Krt4 pre-mRNA with RBM25 by anti-RBM25 compared with anti-IgG (Figure 7L). Subsequently, we knocked down RBM25 using siRNA (Figure S6D) and examined the expression of circKrt4. The results showed that circKrt4 but not Krt4 was significantly downregulated in which RBM25 was knocked down (Figure 7M). Therefore, we speculated that RBM25 was able to bind to flanking sequences of exon 2 and exon 5 to increase circKrt4 formation.
Discussion
In this study, we identified a new functional circRNA named circKrt4 in PH, especially in PAEC injury. Both in vitro and in vivo experiments demonstrated that circKrt4 silencing inhibited PAEC proliferation and pulmonary vascular remodeling under hypoxic conditions. We evaluated 2 mechanisms, as summarized in Figure 8. First, in the nucleus, hypoxia-induced Krt4 transcription by recruitment of the activating transcription factor CEBPA binding to the promoter marked by SE histone proteins (H3K27ac and H3K4me1). RBM25 promoted the cyclization of circKrt4 and finally increased the expression of circKrt4. Pura, a protein that interacts with circKrt4, transcriptionally activates N-cadherin expression and thereby stimulates EndoMT in PAECs. Second, in the cytoplasm, circKrt4 combined with Glpk and decreased mitochondrial enrichment of Glpk, leading to oxidative damage, mitochondrial dysfunction, and finally PAEC proliferation and migration.

Most studies, including ours, have demonstrated that circRNAs function as miRNA sponges in cardiovascular disease and PH.29,34 In previous studies, we found that circ-calm4 (circular RNA calmodulin 4) served as miR-337-3p and miR-124-3p sponge to regulate PASMC proliferation and pyroptosis.24,35 Circ-CDR1 (circular RNA cerebellar degeneration–related protein 1 antisense RNA) as mediates PASMC osteoblastic differentiation and calcification by sponging miR-7-5p.36 Accordingly, in the present study, we analyzed potential miRNAs binding with circKrt4 and performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. Unfortunately, qRT-PCR analysis results showed that there was no correlation of circKrt4 with the potential miRNA targets. However, we do not rule out that this may be because there are miRNAs binding with circKrt4 that have not been found, and in this paper, we first discuss that circKrt4 might work through diverse functions beyond being miRNA sponges.
Transcriptional activator protein Pura is a 322 aa nuclear protein located near many promoters and origins of replication that has been implicated in gene transcription and DNA replication initiation.37,38 A previous study indicated that Pura and YY (Yin Yang) 1 function as transcriptional activators of the neuron-specific FE65 gene promoter in Chinese hamster ovary cells.39 Importantly, Pura was found to be a hypoxia response factor binding to the HIF-1-independent β2 integrin promoters responsible for CD11b and CD11c activation in inflammation.40 Furthermore, Pura interacts with circular RNA; for example, circular RNA Cwc27 (spliceosome-associated protein CWC27 homolog) contributes to Alzheimer disease pathogenesis by repressing Pura activity, and circSamd4 (circular RNA sterile alpha motif domain containing 4; derived from exon 3 of the mouse gene Samd4) binds and sequesters Pura proteins and thereby prevents them from repressing the transcription of the myosin heavy chain gene.41,42 In this study, our results indicate that hypoxia coordinately induces PAEC EndoMT mechanisms that involve both circKrt4 and Pura. Specifically, we demonstrate that circKrt4 might contribute to the induction of Pura binding to the N-cadherin gene promoter and that Pura is required for hypoxia induction of N-cadherin gene activation. However, the effects of circKrt4/Pura complex interaction or competition with other EndoMT initial proteins or EndoMT inhibitory factors in the nucleus cannot be excluded.
Mitochondrial-bound Glpk is involved in step 1 of the subpathway that synthesizes sn (stereospecifically numbering)-glycerol 3-phosphate from glycerol and has been linked to obesity and type 2 diabetes.43,44 Here, we identified Glpk as a novel protein interactor of circKrt4 in PAECs and focused on the function of Glpk in mitochondrial homeostasis, which may indicate mechanisms relevant to PAEC proliferation as well as hypoxic PH. Interestingly, no detectable change in the total protein expression of Glpk was found after silencing circKrt4. However, we found that the mitochondrial localization of Glpk was obviously decreased under hypoxia, and the effect was reversed when circKrt4 was silenced. Moreover, when the mitochondrial localization sequence of Glpk was mutated, Glpk was trapped in the cytoplasm further exacerbated oxidative stress and mitochondrial oxidative phosphorylation damage, suggesting that the mitochondrial localization of Glpk is at least in part required for mediating mitochondrial function in PAECs. We further explored the catalytic activity of Glpk in regulating glycolysis and found the mutation of Glpk does not affect the basal extracellular acidification rate. Since glycolysis occurs within the cytoplasm, this result suggests that the role of Glpk in the cytoplasm may not be affected by circKrt4.
Exon-type circRNAs are generally localized to the cytoplasm45; however, the mechanisms of circKrt4 translocation between the nucleus and cytoplasm remain elusive. Huang et al46 reported that UAP56 (spliceosome RNA helicase DDX39B) and URH49 (ATP-dependent RNA helicase DDX39A) are key modulators of circRNA nuclear export by sensing the lengths of mature circRNAs. Chen et al47 revealed that m6A-modified circNSUN2 (derived from the NOP2 [nucleolar protein 2]/Sun RNA methyltransferase family member 2 locus) is exported by YTHDC1 (YTH domain-containing protein 1) from the nucleus to the cytoplasm. CircKrt4-protein interactions play vital roles in both the nucleus and cytoplasm, driving excessive PAEC proliferation and survival. However, the transport molecule involved in circKrt4 translocation in hypoxic PAECs remains to be fully elucidated and would be interesting to characterize in future studies.
SEs are a large cluster of transcriptional enhancers occupied by H3K4me1, H3K27ac, p300, and master transcription factors that drive gene expression to control cell identity.48,49 Importantly, SE-associated ncRNAs (non-coding RNAs) regulate myocardial infarction, red blood cell development, and tumor pathogenesis.31,50,51 However, how SEs regulate circRNAs in hypoxic PH has not been extensively investigated. Interestingly, we found that H3K27ac, H3K4me1, and CEBPA occupy the promoter of Krt4 and activate its transcription. Previous studies have demonstrated that CEBPA promotes chromatin accessibility and increases the components of chromatin-modifying complexes present at SEs, including Brd4 (bromodomain-containing protein 4), Med1 (mediator of RNA polymerase II transcription subunit 1), and Cdk9 (cyclin-dependent kinase 9).52 Similarly, in this study, we demonstrated that CEBPA is one cofactor that promotes Krt4 transcription initiation as the bridge between enhancers and promoters in PAECs and then activates circKrt4. Notwithstanding, the specific SE site of the regulation of Krt4 in response to hypoxia is still unknown and remains a limitation of this study.
Emerging evidence has demonstrated that RBM25 regulates a large fraction of alternatively spliced exons including the selective splicing of Bcl-x (Bcl-2-like protein 1) and BIN1 (Myc box-dependent–interacting protein 1) pre-mRNA.53–55 In addition, RBM25 is crucial for the formation of a subset of circRNAs, such as circAMOTL1L (circular RNA angiomotin-like 1L homeolog)56 and circCNTNAP3 (circular RNA contactin-associated protein-like 3).33 In this study, we identified that RBM25 interacts with Krt4 pre-mRNA, which form circKrt4, probably by binding to introns 1 and 5 of Krt4 gene. However, our study did not clarify the specific splicing site of RBM25 which is another limitation of this study.
This study is also limited in that we did not detect the expression and function of circKrt4 in PAECs from patients with PH. Because the characteristics of PAECs from patients with PH are completely different from those from healthy controls, it is valuable to distinguish the expression of circKrt4 in cells from patients with idiopathic PAH, hypoxic PH, and PH associated with left heart disease. In addition to PAECs, the roles of circKrt4 in other types of human lung vascular cells including PASMCs, fibroblasts, microvascular ECs, and pericytes deserve further efforts to clarify in the future. Although we confirmed the involvement of circKrt4 in the development of PH in 2 murine PH models, hypoxic model, and SuHx model, which mimic the pathological features of advanced human PH, we failed to investigate the role of circKrt4 in a vascular-specific circKrt4 transgenic mouse, which may yield more convincing evidence. Lastly, no clinical data were acquired concerning the inhibition of circKrt4 on human PH. However, with the continuous development in tissue engineering, it is possible to offer various humanized mouse models and organoid models mimicking the disease condition to predict the value of circKrt4 in the diagnosis and treatment of PH.
Conclusions
Together, we demonstrate for the first time that circKrt4 regulated by CEBPA and RBM25 is located both in the nucleus and cytoplasm; in the nucleus, it is critical in regulating EndoMT by interacting with the Pura/N-cadherin axis. In the cytoplasm, circKrt4 captured Glpk and decreased its translocation into the mitochondria, leading to oxidative stress and attenuation of oxidative phosphorylation. These findings imply that circKrt4 has the potential to become a novel target of nucleic acids to treat PAEC dysfunction in hypoxic PH.
Article Information
Supplemental Material
Figure S1–S6
Table S1–S2
Major Resources Table
Uncropped Blots
Footnote
Nonstandard Abbreviations and Acronyms
- α-SMA
- α-smooth muscle actin
- AAV5
- serotype 5 adenovirus-associated virus
- ASO
- antisense oligonucleotide
- CEBPA
- CCAAT enhancer binding protein alpha
- ChIP
- chromatin immunoprecipitation assay
- CircRNAs
- circular RNAs
- EndoMT
- endothelial-to-mesenchymal transition
- FISH
- fluorescent in situ hybridization
- Glpk
- glycerol kinase
- GPx
- glutathione peroxidase
- HK II
- hexokinase II
- NC
- negative control
- OCR
- oxygen consumption rate
- PAEC
- pulmonary artery endothelial cell
- PASMC
- pulmonary artery smooth muscle cell
- PDH
- pyruvate dehydrogenase
- PH
- pulmonary hypertension
- PKM2
- pyruvate kinase 2
- Pura
- transcriptional activator protein Pur-alpha
- RBM25
- RNA-binding-motif protein 25
- SE
- super enhancer
- SOD
- superoxide dismutase
- SuHx
- SU5416 combined with hypoxia
- TGFβ
- transforming growth factor β
Supplemental Material
File (atvb_atvb-2022-318842d_supp2.pdf)
- Download
- 4.25 MB
References
1.
Wong BW, Marsch E, Treps L, Baes M, Carmeliet P. Endothelial cell metabolism in health and disease: impact of hypoxia. EMBO J. 2017;36:2187–2203. doi: 10.15252/embj.201696150
2.
Kurakula K, Smolders V, Tura-Ceide O, Jukema JW, Quax PHA, Goumans MJ. Endothelial dysfunction in pulmonary hypertension: cause or consequence?. Biomedicines. 2021;9:57. doi: 10.3390/biomedicines9010057
3.
Evans CE, Cober ND, Dai Z, Stewart DJ, Zhao YY. Endothelial cells in the pathogenesis of pulmonary arterial hypertension. Eur Respir J. 2021;58:2003957. doi: 10.1183/13993003.03957-2020
4.
Lawrie A, Francis SE. Frataxin and endothelial cell senescence in pulmonary hypertension. J Clin Invest. 2021;131:e149721. doi: 10.1172/JCI149721
5.
Pu X, Du L, Hu Y, Fan Y, Xu Q. Stem/progenitor cells and pulmonary arterial hypertension. Arterioscler Thromb Vasc Biol. 2021;41:167–178. doi: 10.1161/ATVBAHA.120.315052
6.
Li Y, Lui KO, Zhou B. Reassessing endothelial-to-mesenchymal transition in cardiovascular diseases. Nat Rev Cardiol. 2018;15:445–456. doi: 10.1038/s41569-018-0023-y
7.
Sanchez-Duffhues G, Garcia de Vinuesa A, Ten Dijke P. Endothelial-to-mesenchymal transition in cardiovascular diseases: developmental signaling pathways gone awry. Dev Dyn. 2018;247:492–508. doi: 10.1002/dvdy.24589
8.
Good RB, Gilbane AJ, Trinder SL, Denton CP, Coghlan G, Abraham DJ, Holmes AM. Endothelial to mesenchymal transition contributes to endothelial dysfunction in pulmonary arterial hypertension. Am J Pathol. 2015;185:1850–1858. doi: 10.1016/j.ajpath.2015.03.019
9.
Tang H, Babicheva A, McDermott KM, Gu Y, Ayon RJ, Song S, Wang Z, Gupta A, Zhou T, Sun X, et al. Endothelial hif-2alpha contributes to severe pulmonary hypertension due to endothelial-to-mesenchymal transition. Am J Physiol Lung Cell Mol Physiol. 2018;314:L256–L275. doi: 10.1152/ajplung.00096.2017
10.
Rol N, Kurakula KB, Happe C, Bogaard HJ, Goumans MJ. Tgf-beta and bmpr2 signaling in pah: two black sheep in one family. Int J Mol Sci. 2018;19:2585. doi: 10.3390/ijms19092585
11.
Zhang C, Lu W, Luo X, Liu S, Li Y, Zheng Q, Liu W, Wu X, Chen Y, Jiang Q, et al. Mitomycin c induces pulmonary vascular endothelial-to-mesenchymal transition and pulmonary veno-occlusive disease via smad3-dependent pathway in rats. Br J Pharmacol. 2021;178:217–235. doi: 10.1111/bph.15314
12.
Yu Q, Chan SY. Mitochondrial and metabolic drivers of pulmonary vascular endothelial dysfunction in pulmonary hypertension. Adv Exp Med Biol. 2017;967:373–383. doi: 10.1007/978-3-319-63245-2_24
13.
Zhang WH, Qiu MH, Wang XJ, Sun K, Zheng Y, Jing ZC. Up-regulation of hexokinase1 in the right ventricle of monocrotaline induced pulmonary hypertension. Respir Res. 2014;15:119. doi: 10.1186/s12931-014-0119-9
14.
Zhang H, Wang D, Li M, Plecita-Hlavata L, D’Alessandro A, Tauber J, Riddle S, Kumar S, Flockton A, McKeon BA, et al. Metabolic and proliferative state of vascular adventitial fibroblasts in pulmonary hypertension is regulated through a microrna-124/ptbp1 (polypyrimidine tract binding protein 1)/pyruvate kinase muscle axis. Circulation. 2017;136:2468–2485. doi: 10.1161/CIRCULATIONAHA.117.028069
15.
Caruso P, Dunmore BJ, Schlosser K, Schoors S, Dos Santos C, Perez-Iratxeta C, Lavoie JR, Zhang H, Long L, Flockton AR, et al. Identification of microrna-124 as a major regulator of enhanced endothelial cell glycolysis in pulmonary arterial hypertension via ptbp1 (polypyrimidine tract binding protein) and pyruvate kinase m2. Circulation. 2017;136:2451–2467. doi: 10.1161/CIRCULATIONAHA.117.028034
16.
Ma C, Wang X, He S, Zhang L, Bai J, Qu L, Qi J, Zheng X, Zhu X, Mei J, et al. Ubiquitinated aif is a major mediator of hypoxia-induced mitochondrial dysfunction and pulmonary artery smooth muscle cell proliferation. Cell Biosci. 2022;12:9. doi: 10.1186/s13578-022-00744-3
17.
Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, et al. Circular rnas are a large class of animal rnas with regulatory potency. Nature. 2013;495:333–338. doi: 10.1038/nature11928
18.
Chen LL. The biogenesis and emerging roles of circular rnas. Nat Rev Mol Cell Biol. 2016;17:205–211. doi: 10.1038/nrm.2015.32
19.
Yuan P, Wu WH, Gong SG, Jiang R, Zhao QH, Pudasaini B, Sun Y-Y, Li J-L, Liu J-M, Wang L. Impact of circgsap in peripheral blood mononuclear cells on idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2021;203:1579–1583. doi: 10.1164/rccm.202005-2052LE
20.
Jin X, Xu Y, Guo M, Sun Y, Ding J, Li L, Zheng X, Li S, Yuan D, Li S-S. Hsa_circnfxl1_009 modulates apoptosis, proliferation, migration, and potassium channel activation in pulmonary hypertension. Mol Ther Nucleic Acids. 2021;23:1007–1019. doi: 10.1016/j.omtn.2020.09.029
21.
Lahm T, Frump AL, Albrecht ME, Fisher AJ, Cook TG, Jones TJ, Yakubov B, Whitson J, Fuchs RK, Liu A, et al. 17beta-estradiol mediates superior adaptation of right ventricular function to acute strenuous exercise in female rats with severe pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2016;311:L375–L388. doi: 10.1152/ajplung.00132.2016
22.
Earley S, Resta TC. Estradiol attenuates hypoxia-induced pulmonary endothelin-1 gene expression. Am J Physiol Lung Cell Mol Physiol. 2002;283:L86–L93. doi: 10.1152/ajplung.00476.2001
23.
Ma C, Li Y, Ma J, Liu Y, Li Q, Niu S, Shen Z, Zhang L, Pan Z, Zhu D. Key role of 15-lipoxygenase/15-hydroxyeicosatetraenoic acid in pulmonary vascular remodeling and vascular angiogenesis associated with hypoxic pulmonary hypertension. Hypertension. 2011;58:679–688. doi: 10.1161/HYPERTENSIONAHA.111.171561
24.
Zhang J, Li Y, Qi J, Yu X, Ren H, Zhao X, Xin W, He S, Zheng X, Ma C, et al. Circ-calm4 serves as an mir-337-3p sponge to regulate myo10 (myosin 10) and promote pulmonary artery smooth muscle proliferation. Hypertension. 2020;75:668–679. doi: 10.1161/HYPERTENSIONAHA.119.13715
25.
Bartoszewski R, Moszynska A, Serocki M, Cabaj A, Polten A, Ochocka R, Dell’Italia L, Bartoszewska S, Króliczewski J, Dąbrowski M, et al. Primary endothelial cell-specific regulation of hypoxia-inducible factor (hif)-1 and hif-2 and their target gene expression profiles during hypoxia. FASEB J. 2019;33:7929–7941. doi: 10.1096/fj.201802650RR
26.
Hu CJ, Poth JM, Zhang H, Flockton A, Laux A, Kumar S, McKeon B, Mouradian G, Li M, Riddle S, et al. Suppression of hif2 signalling attenuates the initiation of hypoxia-induced pulmonary hypertension. Eur Respir J. 2019;54:1900378. doi: 10.1183/13993003.00378-2019
27.
Hua W, Ten Dijke P, Kostidis S, Giera M, Hornsveld M. Tgfbeta-induced metabolic reprogramming during epithelial-to-mesenchymal transition in cancer. Cell Mol Life Sci. 2020;77:2103–2123. doi: 10.1007/s00018-019-03398-6
28.
Ge X, Tang P, Rong Y, Jiang D, Lu X, Ji C, Wang J, Huang C, Duan A, Liu Y, et al. Exosomal mir-155 from m1-polarized macrophages promotes endomt and impairs mitochondrial function via activating nf-kappab signaling pathway in vascular endothelial cells after traumatic spinal cord injury. Redox Biol. 2021;41:101932. doi: 10.1016/j.redox.2021.101932
29.
Li H, Xu JD, Fang XH, Zhu JN, Yang J, Pan R, Yuan S-J, Zeng N, Yang Z-Z, Yang H, et al. Circular rna circrna_000203 aggravates cardiac hypertrophy via suppressing mir-26b-5p and mir-140-3p binding to gata4. Cardiovasc Res. 2020;116:1323–1334. doi: 10.1093/cvr/cvz215
30.
Guo W, Liu W, Chen Z, Gu Y, Peng S, Shen L, Shen Y, Wang X, Feng G-S, Sun Y, et al. Tyrosine phosphatase shp2 negatively regulates nlrp3 inflammasome activation via ant1-dependent mitochondrial homeostasis. Nat Commun. 2017;8:2168. doi: 10.1038/s41467-017-02351-0
31.
Huang S, Li X, Zheng H, Si X, Li B, Wei G, Li C, Chen Y, Chen Y, Liao W, et al. Loss of super-enhancer-regulated circrna nfix induces cardiac regeneration after myocardial infarction in adult mice. Circulation. 2019;139:2857–2876. doi: 10.1161/CIRCULATIONAHA.118.038361
32.
Liang W, Shi C, Hong W, Li P, Zhou X, Fu W, Lin L, Zhang J. Super-enhancer-driven lncrna-daw promotes liver cancer cell proliferation through activation of wnt/beta-catenin pathway. Mol Ther Nucleic Acids. 2021;26:1351–1363. doi: 10.1016/j.omtn.2021.10.028
33.
Wang H, Song X, Wang Y, Yin X, Liang Y, Zhang T, Xu L, Jiang F, Dong G. Circcntnap3-tp53-positive feedback loop suppresses malignant progression of esophageal squamous cell carcinoma. Cell Death Dis. 2020;11:1010. doi: 10.1038/s41419-020-03217-y
34.
Chen LL. The expanding regulatory mechanisms and cellular functions of circular rnas. Nat Rev Mol Cell Biol. 2020;21:475–490. doi: 10.1038/s41580-020-0243-y
35.
Jiang Y, Liu H, Yu H, Zhou Y, Zhang J, Xin W, Li Y, He S, Ma C, Zheng X, et al. Circular rna calm4 regulates hypoxia-induced pulmonary arterial smooth muscle cells pyroptosis via the circ-calm4/mir-124-3p/pdcd6 axis. Arterioscler Thromb Vasc Biol. 2021;41:1675–1693. doi: 10.1161/ATVBAHA.120.315525
36.
Ma C, Gu R, Wang X, He S, Bai J, Zhang L, Zhang J, Li Q, Qu L, Xin W, et al. Circrna cdr1as promotes pulmonary artery smooth muscle cell calcification by upregulating camk2d and cnn3 via sponging mir-7-5p. Mol Ther Nucleic Acids. 2020;22:530–541. doi: 10.1016/j.omtn.2020.09.018
37.
Molitor L, Bacher S, Burczyk S, Niessing D. The molecular function of pura and its implications in neurological diseases. Front Genet. 2021;12:638217. doi: 10.3389/fgene.2021.638217
38.
Krachmarov CP, Chepenik LG, Barr-Vagell S, Khalili K, Johnson EM. Activation of the jc virus tat-responsive transcriptional control element by association of the tat protein of human immunodeficiency virus 1 with cellular protein pur alpha. Proc Natl Acad Sci U S A. 1996;93:14112–14117. doi: 10.1073/pnas.93.24.14112
39.
Zambrano N, De Renzis S, Minopoli G, Faraonio R, Donini V, Scaloni A, Cimino F, Russo T. DNA-binding protein pur alpha and transcription factor yy1 function as transcription activators of the neuron-specific fe65 gene promoter. Biochem J. 1997;328(Pt 1):293–300. doi: 10.1042/bj3280293
40.
Kong T, Scully M, Shelley CS, Colgan SP. Identification of pur alpha as a new hypoxia response factor responsible for coordinated induction of the beta 2 integrin family. J Immunol. 2007;179:1934–1941. doi: 10.4049/jimmunol.179.3.1934
41.
Song C, Zhang Y, Huang W, Shi J, Huang Q, Jiang M, Qiu Y, Wang T, Chen H, Wang H. Circular rna cwc27 contributes to alzheimer’s disease pathogenesis by repressing pur-alpha activity. Cell Death Differ. 2022;29:393–406. doi: 10.1038/s41418-021-00865-1
42.
Pandey PR, Yang JH, Tsitsipatis D, Panda AC, Noh JH, Kim KM, Munk R, Nicholson T, Hanniford D, Argibay D, et al. Circsamd4 represses myogenic transcriptional activity of pur proteins. Nucleic Acids Res. 2020;48:3789–3805. doi: 10.1093/nar/gkaa035
43.
Rahib L, Sriram G, Harada MK, Liao JC, Dipple KM. Transcriptomic and network component analysis of glycerol kinase in skeletal muscle using a mouse model of glycerol kinase deficiency. Mol Genet Metab. 2009;96:106–112. doi: 10.1016/j.ymgme.2008.11.163
44.
Kaneko M, Kurokawa M, Ishibashi S. Binding and function of mitochondrial glycerol kinase in comparison with those of mitochondrial hexokinase. Arch Biochem Biophys. 1985;237:135–141. doi: 10.1016/0003-9861(85)90262-0
45.
Salzman J, Gawad C, Wang PL, Lacayo N, Brown PO. Circular rnas are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS One. 2012;7:e30733. doi: 10.1371/journal.pone.0030733
46.
Huang C, Liang D, Tatomer DC, Wilusz JE. A length-dependent evolutionarily conserved pathway controls nuclear export of circular rnas. Genes Dev. 2018;32:639–644. doi: 10.1101/gad.314856.118
47.
Chen RX, Chen X, Xia LP, Zhang JX, Pan ZZ, Ma XD, Han K, Chen J-W, Judde J-G, Deas O, et al. N(6)-methyladenosine modification of circnsun2 facilitates cytoplasmic export and stabilizes hmga2 to promote colorectal liver metastasis. Nat Commun. 2019;10:4695. doi: 10.1038/s41467-019-12651-2
48.
Tang F, Yang Z, Tan Y, Li Y. Super-enhancer function and its application in cancer targeted therapy. npj Precis Oncol. 2020;4:2. doi: 10.1038/s41698-020-0108-z
49.
Thandapani P. Super-enhancers in cancer. Pharmacol Ther. 2019;199:129–138. doi: 10.1016/j.pharmthera.2019.02.014
50.
Alvarez-Dominguez JR, Knoll M, Gromatzky AA, Lodish HF. The super-enhancer-derived alncrna-ec7/bloodlinc potentiates red blood cell development in trans. Cell Rep. 2017;19:2503–2514. doi: 10.1016/j.celrep.2017.05.082
51.
Xie JJ, Jiang YY, Jiang Y, Li CQ, Lim MC, An O, Mayakonda A, Ding L-W, Long L, Sun C, et al. Super-enhancer-driven long non-coding rna linc01503, regulated by tp63, is over-expressed and oncogenic in squamous cell carcinoma. Gastroenterology. 2018;154:2137–2151.e1. doi: 10.1053/j.gastro.2018.02.018
52.
Di Stefano B, Collombet S, Jakobsen JS, Wierer M, Sardina JL, Lackner A, Stadhouders R, Segura-Morales C, Francesconi M, Limone F, et al. C/ebpalpha creates elite cells for ipsc reprogramming by upregulating klf4 and increasing the levels of lsd1 and brd4. Nat Cell Biol. 2016;18:371–381. doi: 10.1038/ncb3326
53.
Zhou A, Ou AC, Cho A, Benz EJ, Huang SC. Novel splicing factor rbm25 modulates bcl-x pre-mrna 5’ splice site selection. Mol Cell Biol. 2008;28:5924–5936. doi: 10.1128/MCB.00560-08
54.
Carlson SM, Soulette CM, Yang Z, Elias JE, Brooks AN, Gozani O. Rbm25 is a global splicing factor promoting inclusion of alternatively spliced exons and is itself regulated by lysine mono-methylation. J Biol Chem. 2017;292:13381–13390. doi: 10.1074/jbc.M117.784371
55.
Ge Y, Schuster MB, Pundhir S, Rapin N, Bagger FO, Sidiropoulos N, Hashem N, Porse BT. The splicing factor rbm25 controls myc activity in acute myeloid leukemia. Nat Commun. 2019;10:172. doi: 10.1038/s41467-018-08076-y
56.
Yang Z, Qu CB, Zhang Y, Zhang WF, Wang DD, Gao CC, Ma L, Chen J-S, Liu K-L, Zheng B, et al. Dysregulation of p53-rbm25-mediated circamotl1l biogenesis contributes to prostate cancer progression through the circamotl1l-mir-193a-5p-pcdha pathway. Oncogene. 2019;38:2516–2532. doi: 10.1038/s41388-018-0602-8
Information & Authors
Information
Published In
Copyright
© 2023 American Heart Association, Inc.
Versions
You are viewing the most recent version of this article.
History
Received: 11 August 2022
Accepted: 18 April 2023
Published online: 4 May 2023
Published in print: July 2023
Keywords
Subjects
Authors
Disclosures
Disclosures None.
Sources of Funding
This study was supported by National Natural Science Foundation of China (grant numbers 82170059 and 81873412 to C. Ma and 31820103007, 31771276, and 31471095 to D. Zhu); Heilongjiang Touyan Innovation Team to DZ Program; National Natural Science Foundation of Heilongjiang Province (grant number LH2020H027 to L. Qu); Construction Project of Scientific Research and Innovation Team of Harbin Medical University-Daqing (grant number HD-CXTD-202001 to C. Ma).
Metrics & Citations
Metrics
Citations
Download Citations
If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.
- Mechanism of pulmonary arterial vascular cell dysfunction in pulmonary hypertension in broiler chickens, Avian Pathology, (1-12), (2025).https://doi.org/10.1080/03079457.2025.2480802
- Exonic CircGUCY1A2 inhibits pulmonary artery smooth muscle cells phenotypic switching via regulating O-glycosylation of COL3A1 in pulmonary hypertension, European Journal of Pharmacology, 992, (177328), (2025).https://doi.org/10.1016/j.ejphar.2025.177328
- Super‐Enhancer Target Gene CBP/p300‐Interacting Transactivator With Glu/Asp‐Rich C‐Terminal Domain, 2 Cooperates With Transcription Factor Forkhead Box J3 to Inhibit Pulmonary Vascular Remodeling , Cell Proliferation, (2025).https://doi.org/10.1111/cpr.13817
- Super-enhancer-associated circPVT1 promotes malignancy of hepatocellular carcinoma via YBX1-mediated RRM2 activation, Cancer Letters, 611, (217395), (2025).https://doi.org/10.1016/j.canlet.2024.217395
- Application Strategies of Super-Enhancer RNA in Cardiovascular Diseases, Biomedicines, 13, 1, (117), (2025).https://doi.org/10.3390/biomedicines13010117
- Panorama of artery endothelial cell dysfunction in pulmonary arterial hypertension, Journal of Molecular and Cellular Cardiology, 197, (61-77), (2024).https://doi.org/10.1016/j.yjmcc.2024.10.004
- Effect of elastin on abnormal proliferation of pulmonary artery smooth muscle cells at an early stage of hypoxic exposure, Journal of Receptors and Signal Transduction, 44, 4, (129-139), (2024).https://doi.org/10.1080/10799893.2024.2430489
- Super‐Enhancer–Driven Syndecan‐4 Regulates Intercellular Communication in Hypoxic Pulmonary Hypertension, Journal of the American Heart Association, 13, 21, (2024)./doi/10.1161/JAHA.124.036757
- Non-Coding RNA Networks in Pulmonary Arterial Hypertension, Pharmacology, (1-12), (2024).https://doi.org/10.1159/000541060
- Circular RNA HMGCS1 sponges MIR4521 to aggravate type 2 diabetes-induced vascular endothelial dysfunction, eLife, 13, (2024).https://doi.org/10.7554/eLife.97267.3
- See more
Loading...
View Options
Login options
Check if you have access through your login credentials or your institution to get full access on this article.
Personal login Institutional LoginPurchase Options
Purchase this article to access the full text.
eLetters(0)
eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.
Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.