Home Medicine Tanshinone ⅡA suppresses hypoxia-induced human pulmonary artery smooth muscle cell over-proliferation via the LINC01013/miR-548c-3p/IL6 pathway
Article Open Access

Tanshinone ⅡA suppresses hypoxia-induced human pulmonary artery smooth muscle cell over-proliferation via the LINC01013/miR-548c-3p/IL6 pathway

  • Nuan Zhang ORCID logo and Kai Xing ORCID logo EMAIL logo
Published/Copyright: November 3, 2025

Abstract

Objectives

The high proliferation of pulmonary artery endothelial cells and smooth muscle cells (PASMCs) is one of the mechanisms of pulmonary hypertension (PH), which can eventually lead to death. The purpose of this study is to explore the effect of tanshinone ⅡA on PASMC proliferation and its action mechanism.

Methods

PASMCs were treated with hypoxia to simulate the PH cell model, and they were cultured by the medium containing different concentrations of tanshinone ⅡA to evaluate its effect on cell viability. CCK-8 reagent was for the viability detection of PASMCs. The expressions of LINC01013, miR-548c-3p, and interleukin 6 (IL6) were detected by RT-qPCR or western blotting, and their targeting relationships were demonstrated by a dual-luciferase reporter assay. The proliferation capacity of PASMCs was evaluated by CCK-8 and the expressions of markers. Glycolysis was measured by detecting phosphofructokinase-1 (PFK-1) activity and pyruvic acid level.

Results

Tanshinone ⅡA restrained hypoxic PASMC viability in a concentration-dependent manner. Overexpression of LINC01013 decreased miR-548c-3p level by binding to it. IL6 was a target of miR-548c-3p, and its expression was hindered by the miR-548c-3p mimics. LINC01013 and IL6 promoted the proliferation and glycolysis of hypoxic PASMCs, and miR-548c-3 acted with inhibiting effects on which. Tanshinone ⅡA suppressed LINC01013/IL6 expression and up-regulated miR-548c-3p.

Conclusions

Tanshinone ⅡA inhibited the glycolysis-mediated growth in hypoxic PASMCs by regulating the LINC01013/miR-548c-3p/IL6 pathway.

Introduction

The changes in the structure or function of pulmonary blood vessels caused by diseases or other factors can cause increased vascular resistance, leading to the occurrence of PH ultimately. The right ventricle is responsible for pumping blood to the lungs, and continuous PH increases its burden. This prolonged high load may eventually progress to right heart failure [1], [2], [3]. Therefore, if PH is not treated promptly and effectively, its further deterioration may lead to the death of patients.

The pathogenesis of PH mainly includes pulmonary vasoconstriction, remodeling, and thrombosis. The disruption of the balance between vasoconstrictor/dilator substances and the changes in ion channel activity are important factors leading to pulmonary vasoconstriction [4]. The proliferation of endothelial cells, smooth muscle cells, and fibroblasts in the pulmonary artery is an important process of vascular remodeling, often accompanied by abnormal accumulation of collagen fibers and elastin, which is irreversible and will lead to the media thickening of the lung intima/blood vessel and lumen stenosis [5]. Moreover, inflammation, endothelium-mesenchymal transformation, dysregulation of proliferative/apoptotic signaling, or genetic changes can lead to vascular remodeling. Persistent inflammation is also a potential risk for pulmonary thrombosis, and other factors include platelet activation and endothelial dysfunction, etc. [6].

In the view of traditional Chinese medicine, Radix Salviae, the dried root and rhizome of the Labiatae plant “Salvia miltiorrhiza Bge.”, has the effect of promoting blood circulation and removing blood stasis, cooling blood to eliminate the carbuncle, and relieving pain. It can be used to treat heart disease, hypertension, and other diseases [7], 8]. Tanshinone IIA is the main active ingredient of Radix Salviae, which is a natural product for treating PH. Tanshinone IIA has been reported to be effective in alleviating monocrotaline-induced PH in rats, which can inhibit the intimamedia thickening of the pulmonary artery [9]. Sodium tanshinone IIA sulfonate is a water-soluble salt of tanshinone IIA which can improve hypoxia-induced PH in rats by inhibiting PI3K/AKT/mTOR pathway, promoting autophagy and reducing infiltration of inflammatory factors in the lung tissue, and it can suppress the proliferation of rats PASMCs by restoring PKG-PPAR-γ signaling [10], 11]. Here, we revealed a novel mechanism by which tanshinone IIA hindered PASMC proliferation by regulating the LINC01013/miR-548c-3p/IL6 pathway.

Materials and methods

Cell line and treatment

PASMCs (SUNNCELL, China) were grown in a specific complete medium (SNPM-H050) in a 37 °C incubator with 5 % CO2. They were cultured at 2 % O2, 5 % CO2, and 92 % N2 for 72 h to induce a hypoxic cell model. PASMCs were treated with different concentrations of tanshinone ⅡA (Sigma-Aldrich, USA) for 24 h to detect the effect of the drug on cell viability, and 20 μg/mL of the drug was for the subsequent experiments. Cell transfection was performed using Lipofectamine 2000 reagent (Invitrogen, USA).

CCK-8 assay

The cells were inoculated in a 96-well plate at a density of 8 × 103 cells/well. After treatment, a new medium (100 μL/well) was replaced and 10 μL CCK-8 reagent was added to each well. After the cells were cultured at 37 °C for 2 h, the optical density (OD) of each well at 450 nm was measured using a microplate reader (BiOTek Instruments, Inc, USA).

RT-qPCR assay

Total RNA was isolated by Trizol reagent (TaKaRa, JPN), and its concentration was measured using the QNano ultramicro spectrophotometer (YEASEN, China). RNA was reversely transcribed into cDNA using a reverse transcription kit (Vazyme, China). U6 small nuclear 1 (U6) was used as the control of miR-548c-3p, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as the control of LINC01013, proliferating cell nuclear antigen (PCNA), and MKI67 (Ki-67). SYBR Premix Ex Taq II (TaKaRa) was used for RT-qPCR analysis. Expression data was calculated using the 2−ΔΔCt method. The relevant primer sequences were collected in Supplementary Table 1.

The biochemical kits

The PFK activity test kit (Colorimetric) and pyruvic acid content detection kit were from Sigma-Aldrich (USA) and Solarbio (China).

Western blotting assay

The cells were lysed using a protein extraction kit (Beyotime, China). The collected supernatant after centrifugation was the protein sample, which was separated using 10 % SDS-PAGE. The protein was then transferred to the PVDF membrane (Millipore, USA), which was sealed with 5 % skim milk at room temperature for 2 h, and then incubated with primary antibody (4 °C overnight) and secondary antibody (1 h at room temperature). Finally, the protein bands were visualized using a chemiluminescent reagent (Beyotime). The anti-PFK-1 (1:1,000) antibody was purchased from Santa Cruz Biotechnology (USA), and the primary antibodies of IL6 (1:800) and GAPDH (1:2,000) were from Proteintech (China). Horseradish peroxidase-labeled mouse/rabbit secondary antibody was purchased from ThermoFisher (USA) with a using concentration of 1:10,000.

Bioinformatics analysis

The Gene Expression Omnibus database (https://www.ncbi.nlm.nih.gov/) was used to retrieve relevant datasets which were analyzed using the GEO2R function in this database. The GSE236251 dataset revealed the differentially expressed genes (DEGs) in the pulmonary arteries of left heart disease patients with or without PH. The non-coding RNA analysis in GSE188938 included the blood samples from patients with chronic thromboembolic pulmonary hypertension (CTEPH) and normal control individuals. GSE149413 is a high-throughput analysis dataset based on fibroblasts derived from the pulmonary artery thrombus and out-tunica of CTEPH patients. The target genes of tanshinone ⅡA were mined through the CTD database (https://ctdbase.org/). The lncRNASNP2 (https://guolab.wchscu.cn/) and miRDB (https://mirdb.org/) databases were used to predict the downstream miRNAs of LINC01013 and the upstream miRNAs of IL6. The GO and KEGG analyses were conducted on the Weisheng online platform (https://www.bioinformatics.com.cn/).

Dual-luciferase reporter assay

The miR-548c-3p mimics (MedChemExpress, USA) or negative controls were co-transfected into the cells transfected with wild-type or mutant-LINC01013/IL6 reporter vectors (Genecreate Biotech, China). After 48 h, a firefly luciferase reporter gene assay kit (Beyotime) was used to evaluate luciferase activity.

Statistical analysis

The data was analyzed using GraphPad Prism software. Differences between two or more sets of data were analyzed using an unpaired Student’s t-test or one-way ANOVA. A value of p<0.05 was considered statistically significant.

Results

Tanshinone ⅡA inhibited PASMC proliferation

After treatment with different concentrations of tanshinone ⅡA, the viability of hypoxia-induced PASMCs was significantly inhibited in a concentration-dependent manner, but normoxic PASMCs were relatively insensitive to it (Figure 1A and B). Since 20 μg/mL tanshinone ⅡA significantly suppressed PASMC activity in the hypoxia group but hardly affected normoxic PASMCs, this concentration was used for subsequent mechanism studies. Tanshinone ⅡA treatment reduced the hypoxia-induced expressions of cell proliferation markers PCNA and Ki-67 (Figure 1C and D). PFK-1 is a rate-limiting enzyme in glycolysis. Tanshinone ⅡA attenuated the induction of PFK-1 activation and pyruvic acid production by hypoxia (Figure 1E and F). The expression of PFK-1 was significantly down-regulated after transfection of its small interfering RNA (si-PFK-1) (Figure 1G). The down-regulation of PFK-1 expression further enhanced the tanshinone ⅡA-mediated inhibition on cell viability and PCNA expression of hypoxic PASMCs (Figure 1H and I). These results demonstrated that tanshinone ⅡA hindered the propagation of hypoxic PASMCs by inhibiting glycolysis.

Figure 1: 
Tanshinone ⅡA in the PASMC proliferation. (A–B) Sensitivity of hypoxic and normoxic PASMCs to different concentrations of tanshinone ⅡA. (C–D) The expressions of PCNA and Ki-67 in cells with hypoxia or tanshinone ⅡA treatment. (E–F) The influence of tanshinone ⅡA on PFK-1 activity and pyruvic acid level in hypoxic PASMCs. (G) Transfection with si-PFK-1 silenced PFK-1 expression. (H–I) PFK-1 silencing enhanced the inhibition of cell viability and PCNA expression by tanshinone ⅡA. *p<0.05, **p<0.01, ***p<0.001.
Figure 1:

Tanshinone ⅡA in the PASMC proliferation. (A–B) Sensitivity of hypoxic and normoxic PASMCs to different concentrations of tanshinone ⅡA. (C–D) The expressions of PCNA and Ki-67 in cells with hypoxia or tanshinone ⅡA treatment. (E–F) The influence of tanshinone ⅡA on PFK-1 activity and pyruvic acid level in hypoxic PASMCs. (G) Transfection with si-PFK-1 silenced PFK-1 expression. (H–I) PFK-1 silencing enhanced the inhibition of cell viability and PCNA expression by tanshinone ⅡA. *p<0.05, **p<0.01, ***p<0.001.

IL6 overexpression reversed the tanshinone ⅡA mediated-inhibition on PASMC growth

By making a VENN map using the target genes of tanshinone ⅡA and the DEGs in non-PH/PH arterial, 29 shared genes were obtained in the intersection (Figure S1A and Table S1). The GO analysis showed that the biological processes enriched by these genes included the proliferation of smooth muscle cells, etc. (Figure 2A). The KEGG analysis displayed that the enriched signaling pathways included TNF, HIF-1, and Toll-like receptor pathways, etc. (Figure 2B), which have been reported to be related to PH [12], [13], [14]. The key gene IL6 was screened through a VENN diagram of the genes involved in these biological processes and signaling pathways (Figure 2C). Hypoxia induced the expression of IL6 (Figure 2D). The role of IL-6 in PASMC proliferation was demonstrated by transfecting its overexpression vector (oe-IL6, Figure 2E). Overexpression of IL6 increased PASMC viability and PCNA expression in the tanshinone ⅡA-treated group (Figure 2F and G), which reversed the tanshinone ⅡA-induced decreases of PFK-1 activity and pyruvic acid level (Figure 2H and I), proving the acceleration of IL6 on PASMC growth.

Figure 2: 
IL6 in the PASMC proliferation. (A–B) The GO and KEGG analysis of genes. (C) VENN diagram of the genes involved in smooth muscle cell proliferation biological processes and TNF, HIF-1, and toll-like receptor signaling pathways. (D) The expression of IL6 in PASMCs. (E) IL6 was overexpressed after oe-IL6 transfection. Influence of IL6 overexpression on the growth inhibition (F–G) and glycolysis suppression (H–I) of hypoxic PASMCs by tanshinone ⅡA. *p<0.05, **p<0.01, ***p<0.001.
Figure 2:

IL6 in the PASMC proliferation. (A–B) The GO and KEGG analysis of genes. (C) VENN diagram of the genes involved in smooth muscle cell proliferation biological processes and TNF, HIF-1, and toll-like receptor signaling pathways. (D) The expression of IL6 in PASMCs. (E) IL6 was overexpressed after oe-IL6 transfection. Influence of IL6 overexpression on the growth inhibition (F–G) and glycolysis suppression (H–I) of hypoxic PASMCs by tanshinone ⅡA. *p<0.05, **p<0.01, ***p<0.001.

LINC01013 promoted IL6 expression by inhibiting miR-548c-3p

LINC01013 is a high-expressed lncRNA in both the blood and the thrombus fibroblasts of CTEPH patients (Figure S1B and Table S1). We then predicted the downstream miRNAs of LINC01013 and the upstream miRNAs of IL6 using the relevant database, and finally, miR-548c-3p was identified by a VENN diagram (Figure S1C and Table S1). Figure S1D shows the binding sequences of miR-548c-3p to LINC01013 and IL6 mRNA predicted by the database. After transfection of the mimics, miR-548c-3p was significantly upregulated (Figure 3A), which weakened the luciferase intensity in cells transfected with wild-type (wt)-LINC01013/-IL6 reporter vector but didn’t affect the mutant (mut) transfection group (Figure 3B and C), verifying the target relationship among them. The overexpression vector of LINC01013 (oe-LINC01013) was introduced to explore its regulation on miR-548c-3p/IL6 (Figure 3D), whose transfection down-regulated miR-548c-3p and increased IL6 expression (Figure 3E and F). Moreover, the miR-548c-3p mimics hindered IL6 expression, and its co-transfection reversed the facilitation of LINC01013 on IL6 expression (Figure 3F). All these experiments certify that LINC01013 enhanced IL6 expression by down-regulating miR-548c-3p.

Figure 3: 
Regulation of LINC01013 on the expression of miR-548c-3p/IL6 axis. (A) The efficiency of the mimics in up-regulating miR-548c-3p. (B–C) Dual-luciferase assay demonstrated the targeting effect of LINC01013 on miR-548c-3p and miR-548c-3p on IL6. (D) Effect of oe-LINC01013 on LINC01013 expression. (E) Regulation of miR-548c-3p expression by LINC01013. (F) The regulatory effect of LINC01013 on miR-548c-3p/IL6 expression. *p<0.05, **p<0.01, ***p<0.001.
Figure 3:

Regulation of LINC01013 on the expression of miR-548c-3p/IL6 axis. (A) The efficiency of the mimics in up-regulating miR-548c-3p. (B–C) Dual-luciferase assay demonstrated the targeting effect of LINC01013 on miR-548c-3p and miR-548c-3p on IL6. (D) Effect of oe-LINC01013 on LINC01013 expression. (E) Regulation of miR-548c-3p expression by LINC01013. (F) The regulatory effect of LINC01013 on miR-548c-3p/IL6 expression. *p<0.05, **p<0.01, ***p<0.001.

Tanshinone ⅡA inhibited hypoxic PASMC proliferation by regulating the LINC01013/miR-548c-3p/IL6 axis

As shown in Figure 4A–D, overexpression of LINC01013 further enhanced the glycolysis-mediated growth of hypoxic PASMCs, manifesting increased cell viability, PCNA expression, PFK-1 activity, and pyruvic acid level, which were reversed by co-transfection of the miR-548c-3p mimics. Overexpression of IL6 partially eliminated the suppression of miR-548c-3p mimics on cell proliferation and glycolysis process in hypoxic PASMCs (Figure 4A–D). Tanshinone ⅡA inhibited LINC01013/IL6 expression and up-regulated miR-548c-3p in hypoxia-induced PASMCs (Figure 4E–G). These results hint that tanshinone ⅡA can restrain hypoxic PASMC propagation by regulating the LINC01013/miR-548c-3p/IL6 axis.

Figure 4: 
The LINC01013/miR-548c-3p/IL6 axis mediated the suppression of tanshinone ⅡA on hypoxic PASMC growth. The roles of LINC01013/miR-548c-3p/IL6 axis in hypoxic PASMC proliferation (A–B) and glycolysis (C–D). (E–G) Effects of hypoxia and tanshinone ⅡA on the expressions of LINC01013, miR-548c-3p, and IL6. *p<0.05, **p<0.01, ***p<0.001.
Figure 4:

The LINC01013/miR-548c-3p/IL6 axis mediated the suppression of tanshinone ⅡA on hypoxic PASMC growth. The roles of LINC01013/miR-548c-3p/IL6 axis in hypoxic PASMC proliferation (A–B) and glycolysis (C–D). (E–G) Effects of hypoxia and tanshinone ⅡA on the expressions of LINC01013, miR-548c-3p, and IL6. *p<0.05, **p<0.01, ***p<0.001.

Discussion

The potential of tanshinone IIA for the treatment of PH is already self-evident. In the PH rat model, treatment of tanshinone IIA intraperitoneal injection can reduce pulmonary artery muscularization and intimal-media thickening, and improve pulmonary vascular morphology [9]. Given the cardiac burden caused by PH, Zhang et al. revealed the role of tanshinone IIA in reducing right ventricular systolic pressure and improving right ventricular fat index in PH rats. It can also inhibit hypoxia-induced proliferation of pulmonary artery smooth muscle cells by reducing transforming growth factor β-induced smad3 phosphorylation [15]. Another study revealed that targeting SOCE regulated by the PGG-PPAR-γ pathway is a potential strategy for the treatment of PH with sodium tanshinone IIA sulfonate [10]. However, its specific regulatory mechanisms remain to be explored. We report that tanshinone IIA inhibited the viability of hypoxic PASMCs in a concentration-dependent manner, but its low concentrations were relatively safe for normoxic cells.

It has been reported that the competitive endogenous RNA (ceRNA) network mediates the progression of PH. Simply put, many types of RNA (such as mRNA, lncRNA, circRNA, etc.) contain the miRNA response elements. LncRNA or circRNA can compete with mRNA to bind miRNA, thereby mediating the gene expression regulation by miRNA. Under hypoxia conditions, lncRNA HOXA-AS3 is up-regulated but miR-675-3p was down-regulated in PASMCs. HOXA-AS3 can promote PDE5A expression by sponging miR-675-3p, facilitating the progression of PH [16]. LncRNA Tug1 is overexpressed in hypoxic PH mice. Its silencing down-regulates FOXC1 expression by releasing miR-374c, thereby impeding pulmonary vascular remodeling [17]. LncRNA PAHRF is down-regulated in PH patients and hypoxic PASMCs. It can reduce the proliferation of PASMCs under hypoxia conditions by regulating the miR-23a-3p/MST1 axis [18]. CircGSAP levels are significantly reduced in the plasma of patients with idiopathic PH, and its overexpression is associated with high survival. CircGSAP can reduce the inhibition of miR-27a-3p on BMPR2 expression by binding it, thus hindering pulmonary microvascular endothelial cells from proliferating and migrating induced by hypoxia and inducing their death [19]. Here, we report a novel ceRNA network affecting PH. We found that hypoxia treatment induced the expressions of LINC01013 and IL6 and inhibited miR-548c-3p expression. Tanshinone IIA restrained IL6 by alleviating the targeting effect of LINC01013 on miR-548c-3p, thereby reducing the glycolysis-mediated PASMC proliferation.

Myocardial fibrosis greatly increases the risk of death from heart disease. Silencing of LINC01013 decreased the expression of pro-fibrotic markers and reduced the activation of fibroblasts [20]. Silencing LINC01013 in endothelial cells damages its ability to migrate and angiogenesis [21]. LINC01013 is upregulated in calcified aortic valve disease and can control the transcription of CCN2 by inducing NELF-E expression to mediate the fibrocalcific process of the disease [22]. MiR-548c-3p is an inhibitor of tumor cell growth. Circ_0002395 can enhance the expression of PDK1 by down-regulating miR-548c-3p and promoting the aerobic glycolysis-mediated proliferation of pancreatic cancer cells [23]. Another study showed that miR-548c-3p may be an important player in heart-related diseases. In the myocardium of rats with myocardial infarction, miR-548c-3p is absent. Its upregulation reduces myocardial fibrosis and improves heart function [24]. Notably, miR-548c-3p was shown to be a maladjusted miRNA in a PH pig model [25], but its specific role in PH remains unknown. As we mentioned in the “Introduction”, inflammation is an important factor leading to pulmonary vascular remodeling and thrombosis. Therefore, IL6, a pleiotropic proinflammatory cytokine, has a potential position in PH development. Cai et al. reported that overexpression of miR-125a-5p can inhibit the proliferation and induce apoptosis in PASMCs by reducing the production of IL6 and TGF-β1 [26].

In conclusion, we highlighted a new ceRNA network of which the LINC01013/miR-548c-3p/IL6 axis may be involved in the progression of PH and revealed the reliable suppression of tanshinone ⅡA on PASMC proliferation. Our study provides new insight into the understanding of PH pathogenesis and the use of tanshinone ⅡA in PH treatment.


Corresponding author: Kai Xing, Special Education College of Changchun University, No. 6543, Weixing road, 130000, Changchun, China, E-mail:

  1. Research ethics: Not applicable.

  2. Informed consent: Not applicable.

  3. Author contributions: Conceptualization, N.Z. and K.X.; Data curation, N.Z. and K.X.; Formal analysis, N.Z. and K.X.; Funding acquisition, K.X.; Investigation, N.Z. and K.X.; Methodology, N.Z. and K.X.; Project administration, K.X.; Resources, N.Z. and K.X.; Software, N.Z. and K.X.; Supervision, K.X.; Validation, N.Z. and K.X.; Visualization, N.Z.; Roles/Writing - original draft, N.Z.; Writing - review & editing, K.X.

  4. Use of Large Language Models, AI and Machine Learning Tools: None declared.

  5. Conflict of interest: Not applicable.

  6. Research funding: Not applicable.

  7. Data availability: The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

References

1. Chen, S, Zhong, J, Hu, B, Shao, N, Deng, C. Whole-genome microRNA sequencing analysis in patients with pulmonary hypertension. Front Genet 2023;14:1250629. https://doi.org/10.3389/fgene.2023.1250629.Search in Google Scholar PubMed PubMed Central

2. Kerkutluoglu, M, Gunes, H, Onus, AE, Dagli, M, Yucel, O. Predictive value of nesfatin-1 in heart failure mortality. Turk J Biochem 2023;48:426–31. https://doi.org/10.1515/tjb-2022-0227.Search in Google Scholar

3. Ozturk, D, Erdogan, DG, Tanyeli, A, Çomaklı, S, Baylan, H, Polat, E. The protective effects of urapidil on lung tissue after intestinal ischemia-reperfusion injury. Turk J Biochem 2019;44:539–48.10.1515/tjb-2018-0286Search in Google Scholar

4. Zainab, A, Gooch, M, Tuazon, DM. Acute respiratory distress syndrome in patients with cardiovascular disease. Methodist Debakey Cardiovasc J 2023;19:58–65. https://doi.org/10.14797/mdcvj.1244.Search in Google Scholar PubMed PubMed Central

5. Dierick, F, Solinc, J, Bignard, J, Soubrier, F, Nadaud, S. Progenitor/stem cells in vascular remodeling during pulmonary arterial hypertension. Cells 2021;10:1338. https://doi.org/10.3390/cells10061338.Search in Google Scholar PubMed PubMed Central

6. Renaud-Picard, B, Gallais, F, Ohana, M, Zeyons, F, Kretz, B, Andre, J, et al.. Bilateral acute cardioembolic limb ischemia after coronavirus disease 2019 pneumonia in a lung transplant recipient: a case report. Transplant Proc 2020;52:2715–8. https://doi.org/10.1016/j.transproceed.2020.06.024.Search in Google Scholar PubMed PubMed Central

7. Chai, R, Ye, Z, Xue, W, Shi, S, Wei, Y, Hu, Y, et al.. Tanshinone IIA inhibits cardiomyocyte pyroptosis through TLR4/NF-kappaB p65 pathway after acute myocardial infarction. Front Cell Dev Biol 2023;11:1252942.10.3389/fcell.2023.1252942Search in Google Scholar PubMed PubMed Central

8. Wang, S, Yuan, Y, Lin, Q, Zhou, H, Tang, B, Liu, Y, et al.. Antiosteoporosis effect of tanshinol in osteoporosis animal models: a systematic review and meta-analysis. Front Pharmacol 2022;13:937538. https://doi.org/10.3389/fphar.2022.937538.Search in Google Scholar PubMed PubMed Central

9. Zhang, X, Liu, S, Sun, YLG. [Tanshinone IIA alleviates monocrotaline-induced pulmonary hypertension in rats through the PI3K/Akt-eNOS signaling pathway]. Nanfang Yike Daxue Xuebao 2022;42:718–23. https://doi.org/10.12122/j.issn.1673-4254.2022.05.13.Search in Google Scholar PubMed PubMed Central

10. Jiang, Q, Lu, W, Yang, K, Hadadi, C, Fu, X, Chen, Y, et al.. Sodium tanshinone IIA sulfonate inhibits hypoxia-induced enhancement of SOCE in pulmonary arterial smooth muscle cells via the PKG-PPAR-gamma signaling axis. Am J Physiol Cell Physiol 2016;311:C136–49. https://doi.org/10.1152/ajpcell.00252.2015.Search in Google Scholar PubMed PubMed Central

11. Bao, YR, Chen, JW, Jiang, Y, Wang, LH, Xue, R, Qian, JX, et al.. Sodium tanshinone II sulfonate A ameliorates hypoxia-induced pulmonary hypertension. Front Pharmacol 2020;11:687. https://doi.org/10.3389/fphar.2020.00687.Search in Google Scholar PubMed PubMed Central

12. Deng, H, Tian, X, Sun, H, Liu, H, Lu, M, Wang, H. Calpain-1 mediates vascular remodelling and fibrosis via HIF-1alpha in hypoxia-induced pulmonary hypertension. J Cell Mol Med 2022;26:2819–30. https://doi.org/10.1111/jcmm.17295.Search in Google Scholar PubMed PubMed Central

13. Farkas, D, Thompson, AAR, Bhagwani, AR, Hultman, S, Ji, H, Kotha, N, et al.. Toll-like receptor 3 is a therapeutic target for pulmonary hypertension. Am J Respir Crit Care Med 2019;199:199–210. https://doi.org/10.1164/rccm.201707-1370oc.Search in Google Scholar PubMed PubMed Central

14. Liu, WY, Wang, L, Lai, YF. Hepcidin protects pulmonary artery hypertension in rats by activating NF-kappaB/TNF-alpha pathway. Eur Rev Med Pharmacol Sci 2019;23:7573–81.Search in Google Scholar

15. Zhang, N, Dong, M, Luo, Y, Zhao, F, Li, Y. Danshensu prevents hypoxic pulmonary hypertension in rats by inhibiting the proliferation of pulmonary artery smooth muscle cells via TGF-beta-smad3-associated pathway. Eur J Pharmacol 2018:8201–7. https://doi.org/10.1016/j.ejphar.2017.12.010.Search in Google Scholar PubMed

16. Li, ZK, Gao, LF, Zhu, XAXDK. LncRNA HOXA-AS3 promotes the progression of pulmonary arterial hypertension through mediation of miR-675-3p/PDE5A axis. Biochem Genet 2021;59:1158–72. https://doi.org/10.1007/s10528-021-10053-y.Search in Google Scholar PubMed

17. Yang, L, Liang, H, Shen, L, Guan, Z, Meng, X. LncRNA Tug1 involves in the pulmonary vascular remodeling in mice with hypoxic pulmonary hypertension via the microRNA-374c-mediated Foxc1. Life Sci 2019;237:116769. https://doi.org/10.1016/j.lfs.2019.116769.Search in Google Scholar PubMed

18. Liu, Y, Hu, R, Zhu, J, Nie, X, Jiang, Y, Hu, P, et al.. The lncRNA PAHRF functions as a competing endogenous RNA to regulate MST1 expression by sponging miR-23a-3p in pulmonary arterial hypertension. Vascul Pharmacol 2021;139:106886. https://doi.org/10.1016/j.vph.2021.106886.Search in Google Scholar PubMed

19. Sun, Y, Jiang, R, Hu, X, Gong, S, Wang, L, Wu, W, et al.. CircGSAP alleviates pulmonary microvascular endothelial cells dysfunction in pulmonary hypertension via regulating miR-27a-3p/BMPR2 axis. Respir Res 2022;23:322. https://doi.org/10.1186/s12931-022-02248-7.Search in Google Scholar PubMed PubMed Central

20. Quaife, NM, Chothani, S, Schulz, JF, Lindberg, EL, Vanezis, K, Adami, E, et al.. LINC01013 is a determinant of fibroblast activation and encodes a novel fibroblast-activating micropeptide. J Cardiovasc Transl Res 2023;16:77–85. https://doi.org/10.1007/s12265-022-10288-z.Search in Google Scholar PubMed PubMed Central

21. Pham, TP, Bink, DI, Stanicek, L, van Bergen, A, van Leeuwen, E, Tran, Y, et al.. Long non-coding RNA aerrie controls DNA damage repair via YBX1 to maintain endothelial cell function. Front Cell Dev Biol 2020;8:619079. https://doi.org/10.3389/fcell.2020.619079.Search in Google Scholar PubMed PubMed Central

22. Chignon, A, Argaud, D, Boulanger, MC, Mkannez, G, Bon-Baret, V, Li, Z, et al.. Genome-wide chromatin contacts of super-enhancer-associated lncRNA identify LINC01013 as a regulator of fibrosis in the aortic valve. PLoS Genet 2022;18:e1010010. https://doi.org/10.1371/journal.pgen.1010010.Search in Google Scholar PubMed PubMed Central

23. Shu, ML, Xia, JK, Yan, J, Feng, YJ, Qian, CJ, Teng, XS, et al.. Circ_0002395 promotes aerobic glycolysis and proliferation in pancreatic adenocarcinoma cells via miR-548c-3p/PDK1 axis. J Bioenerg Biomembr 2024;56:55–71. https://doi.org/10.1007/s10863-023-09995-2.Search in Google Scholar PubMed

24. Zhang, LX, Zhang, SH, Wang, CQ, Bing, Q, Zhao, Z, Wang, J, et al.. Role and mechanism of microRNA-548c-3p/c-Myb in myocardial infarction fibrosis in rats. Eur Rev Med Pharmacol Sci 2019;23:4908–16.Search in Google Scholar

25. Rothman, A, Restrepo, H, Sarukhanov, V, Evans, WN, Wiencek, RGJr., Williams, R, et al.. Assessment of microRNA and gene dysregulation in pulmonary hypertension by endoarterial biopsy. Pulm Circ 2017;7:455–64. https://doi.org/10.1177/2045893217704206.Search in Google Scholar PubMed PubMed Central

26. Cai, Z, Li, J, Zhuang, Q, Zhang, X, Yuan, A, Shen, L, et al.. MiR-125a-5p ameliorates monocrotaline-induced pulmonary arterial hypertension by targeting the TGF-beta1 and IL-6/STAT3 signaling pathways. Exp Mol Med 2018;50:1–11. https://doi.org/10.1038/s12276-018-0068-3.Search in Google Scholar PubMed PubMed Central


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/tjb-2025-0133).


Received: 2025-03-31
Accepted: 2025-09-26
Published Online: 2025-11-03

© 2025 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 11.1.2026 from https://www.degruyterbrill.com/document/doi/10.1515/tjb-2025-0133/html
Scroll to top button