Skip to main content
Journal of Smooth Muscle Research logoLink to Journal of Smooth Muscle Research
. 2022 Aug 10;58:50–62. doi: 10.1540/jsmr.58.50

Potential of the TRPM7 channel as a novel therapeutic target for pulmonary arterial hypertension

Keizo Hiraishi 1,2,*, Lin Hai Kurahara 1, Kaori Ishikawa 3, Tetsuhiko Go 4, Naoya Yokota 4, Yaopeng Hu 2, Takayuki Fujita 2, Ryuji Inoue 2, Katsuya Hirano 1
PMCID: PMC9364263  PMID: 35944979

Abstract

Pulmonary arterial hypertension (PAH) is an intractable vascular disease characterized by a progressive increase in pulmonary vascular resistance caused by pulmonary vascular remodeling, which ultimately leads to right-sided heart failure. PAH remains incurable, despite the development of PAH-targeted therapeutics centered on pulmonary artery relaxants. It is necessary to identify the target molecules that contribute to pulmonary artery remodeling. Transient receptor potential (TRP) channels have been suggested to modulate pulmonary artery remodeling. Our study focused on the transient receptor potential ion channel subfamily M, member 7, or the TRPM7 channel, which modulates endothelial-to-mesenchymal transition and smooth muscle proliferation in the pulmonary artery. In this review, we summarize the role and expression profile of TRPM7 channels in PAH progression and discuss TRPM7 channels as possible therapeutic targets. In addition, we discuss the therapeutic effect of a Chinese herbal medicine, Ophiocordyceps sinensis (OCS), on PAH progression, which partly involves TRPM7 inhibition.

Keywords: TRPM7, pulmonary arterial hypertension, cardiovascular remodeling

Overview of Pulmonary Arterial Hypertension (PAH)

Pulmonary hypertension is an intractable vascular disease characterized by a progressive increase in pulmonary vascular resistance (PVR). Pulmonary hypertension was divided into five groups according to clinical, hemodynamic, and etiological characteristics and treatment strategy, including idiopathic and heritable pulmonary arterial hypertension (PAH) (group 1), PAH due to left heart disease (group 2), PAH due to lung diseases and/or hypoxia (group 3), PAH associated with chronic thromboembolism (group 4), and PAH forms with unclear or multifaceted origins (group 5). The estimated incidence of primary PAH ranges from 1 to 2 cases per million people in the general population.

Under physiological conditions, blood flows from the right ventricle to the lungs via the pulmonary artery and contributes to the gas exchange. Endothelial cells generate constriction factors, such as endothelin (1), and relaxing factors, such as nitric oxide (NO) and prostacyclin (2). Under physiological conditions, endothelium-derived relaxing factors play an important role in maintaining tissue homeostasis, preventing thrombosis, and protecting vessel walls from remodeling. In contrast, PAH is characterized by vasoconstriction and vascular wall remodeling, which are associated with intimal and medial thickening and thrombosis, resulting in an increase in PVR and pulmonary arterial pressure (3).

The resultant thickening of the pulmonary arterial wall and narrowing of the vessel diameter increases the PVR and pulmonary arterial pressure to over 25 mmHg at rest (4,5,6). At the 6th World Symposium on Pulmonary Hypertension (Nice, France, 2018), it was recognized that the original hemodynamic definition of PAH was arbitrary, and it was recommended that the mean pulmonary artery pressure (MPAP) threshold should be lowered to 20 mmHg. Pulmonary arterial wedge pressure (PAWP) and PVR thresholds were ≤15 mmHg and >3 Wood units, respectively, as measured using right heart catheterization (7).

Increased cardiac afterload due to increased PVR causes right heart hypertrophy, which eventually results in fatal right heart failure. Delaying the progression of right ventricular insufficiency by targeting vasoconstriction and vascular remodeling is anticipated to alleviate symptoms and improve the survival of patients with PAH (8). Before the development of current therapeutic options, idiopathic PAH progressed rapidly, leading to right-sided heart failure and death. In a recent study, the survival rates of patients with PAH at 1-, 2, and 3 years of follow-up were 85.7%, 69.6%, and 54.9%, respectively (9). In Japan, the survival of patients with PAH has been significantly improved by more specific treatments; the 3- and 10 year survival showed 88.2–92.1% and 69.5%, respectively (10, 11).

Several causative conditions have been suggested in the pathogenesis of certain types of PAH. Portal hypertension (12), human immunodeficiency virus infection (13, 14) and appetite-suppressant drugs such as fenfluramine (15, 16) have been proposed as the causative conditions for a type of associated PAH. More than 20 years ago, a genetic predisposition to a heterozygous mutation in the bone morphogenetic protein receptor type II (BMPR2), a member of the transforming growth factor β (TGF-β) superfamily receptor, was discovered in heritable PAH. At least 16 other genes have been identified in patients with hereditary PAH, including activin A receptor type II-like 1 (ACVRL1), SMAD1, SMAD4, SMAD9, and caveolin 1 (CAV1) (17). However, in most cases of PAH, the precise mechanism of pathogenesis is poorly understood and may vary among cases. Endothelial-to-mesenchymal transition (EndoMT) of pulmonary artery endothelial cells (PAECs) and increased proliferative activity of pulmonary arterial smooth muscle cells (PASMCs) are the main mechanisms underlying pulmonary vascular remodeling (18,19,20,21). The transcription factors hypoxia-inducible factor (HIF), CCAAT-enhancer-binding protein (CEBP), runt-related transcription factor (RUNX), activator protein-1 (AP-1), C-terminal binding protein-1 (CtBP1), forkhead box M1 (FoxM1), pyruvate kinase muscle-2 (PKM2), nuclear factor-kappa B (NF-κB), β-catenin, twist family basic helix–loop–helix transcription factor 1 (TWIST1), and SLUG have been suggested to contribute to pulmonary vascular remodeling in PAH development and progression (7).

Pulmonary Artery Remodeling in PAH

The major remedies for the current treatment of PAH are prostacyclin analogs, endothelin receptor antagonists, and phosphodiesterase type 5 inhibitors (8). These vasodilators target pulmonary vasoconstriction in PAH. Vascular remodeling is another critical condition that contributes to the pathogenesis and progression of PAH. Therefore, our study focused on vascular remodeling, which may serve as a therapeutic target for the development of novel PAH treatment strategies.

EndoMT is a biological process in which endothelial cells progressively change their endothelial phenotype to a mesenchymal or myofibroblastic phenotype (22). It contributes to vascular fibrosis and angiogenesis, which are often closely associated with cardiac neovascularization, atherosclerosis, arteriosclerosis, and PAH (19, 23). In the pulmonary artery in cases of PAH, most cells within various intimal lesions show a phenotype of myofibroblasts that are positive for smooth muscle-type α actin (α-SMA) but negative for endothelial markers, such as nitric oxide synthase (eNOS), vascular endothelial cadherin, and cluster of differentiation (CD) 31 (24,25,26). Among the various pathological insults to endothelial cells, including oxidative stress (15, 27), mechanical stress (28) and inflammatory cytokines (29), TGF-β is one of the most relevant inducers of EndoMT in PAH as it augments myofibroblast transformation in numerous cell types and contributes to tissue fibrosis. Several TGF isotypes, such as TGF-β1, TGF-β2, and TGF-β3 (30), contribute to angiogenesis (31, 32). The canonical TGF-β signaling pathway commences with the binding of TGF-β to the TGF-β type 2 receptor (TGF-βR2), which phosphorylates and activates the type 1 receptor (TGF-βRl) via its constitutive kinase activity. Activated TGF-βRl phosphorylates the transcription factors SMAD-2 and SMAD-3, which in turn promotes collagen synthesis (33, 34). Fibrotic diseases are characterized by EndoMT. In particular, upregulation of mesenchymal markers, such as α-SMA and type 1 collagen, and downregulation of endothelial markers, such as vascular endothelial (VE)-cadherin, caused by the TGF-β/Smad signaling pathway, have been shown (35).

The proliferation of PASMCs is another key element in vascular remodeling during PAH progression (36). PASMCs derived from patients with PAH show enhanced proliferation compared with those derived from patients without PAH (37). Increased proliferative activity is associated with the increased activity of cyclin-dependent kinases. Moreover, the calcineurin/nuclear factor of activated T-cell signaling pathway has been reported to contribute to increased proliferation and decreased apoptosis in PASMCs derived from PAH model rats (38).

The aforementioned signaling pathways that contribute to EndoMT and the proliferation of PASMCs are assumed to be good targets for the treatment of PAH, especially when focusing on vascular remodeling.

Role of Transient Receptor Potential (TRP) Channels in PAH Pathophysiology

Intracellular Ca2+ signaling contributes not only to vasoconstriction but also to many events of vascular remodeling, such as EndoMT, migration, proliferation, and production of extracellular matrix. An influx of extracellular Ca2+ and release of intracellularly stored Ca2+ are the main mechanisms underlying the increase in intracellular calcium concentration ([Ca2+]i). Calcium entry involves various plasmalemmal Ca2+ channels, including the superfamily of stretch-activated channels. Stretch-activated channels are nonselective Ca2+-permeable cation channels. Some transient receptor potential (TRP) channels and members of the Piezo channel superfamily serve as stretch-activated channels.

The TRP channel superfamily is a group of non-selective cation channels that are permeable to mono- and divalent cations such as Na+, K+, Cs+, Li+, Ca2+, and Mg2+ (39). TRP channels were initially discovered during a study on Drosophila phototransduction (40, 41). Twenty-eight distinct members of the TRP channel superfamily have been identified, 20 of which are expressed in mammals. All members of the TRP channel superfamily share a six-transmembrane structure, comprising 553–2022 amino acid residues. The development of X-ray diffraction analysis and electron microscopy is helpful in elucidating the structure of TRP channels (42, 43). Based on their structural differences, TRP channels can be categorized into six types: ankyrin (TRPA), canonical (TRPC), melastatin (TRPM), polycystin (TRPP), vanilloid (TRPV), and mucolipin (TRPML).

The expression of TRPC1, TRPC2, TRPC3, TRPC4, TRPC5, TRPC6, TRPC7, TRPV1, TRPV2, TRPV3, TRPV4, TRPM2, TRPM3, TRPM4, TRPM7, and TRPM8 has been detected in the pulmonary arteries of rats, mice, and humans. More precisely, TRPC1, TRPC2, TRPC3, TRPC4, TRPC5, TRPC6, TRPC7, TRPV1, TRPV2, TRPV4, TRPM1, TRPM3, TRPM4, TRPM6, TRPM7, and TRPM8 have been detected in PAEC (39).

The expression of TRPC channels was reported to be upregulated in PASMCs of PAH model mice, and these channels contribute to an increase in [Ca2+]i (44). Table 1 summarizes the influence of various PAH-inducing conditions on the expression of TRP channels, with the exception of TRPM7. The enhanced expression and function of TRP channels in PAH highlight their importance as potential targets for pharmacological interventions.

Table 1. Altered expression of TRPCs, TRPMs (except for M7), and TRPVs in PAH-related conditions.

Cell or tissue (animal species) PAH-related stimulation TRP channel Expression Ref.
PASMC (rat) Hypoxia C1, C6 Upregulation (44)
PASMC (rat) Hypoxia C1, C6 Upregulation (81)
Pulmonary artery (rat) MCT C1, C4 C3 Upregulation (C1, C4)Downregulation (C3) (82)
PASMC (mouse) Hypoxia C1 Upregulation (83)
Lung (rat) Chronic intermittent hypoxia C1, C4, C6 Upregulation (84)
PASMC (human) Hypoxia C6 Upregulation (85)
PASMC (human) Hypoxia C6 Upregulation (86)
PASMC (rat) Hypoxia C1, C6 Upregulation (87)
Pulmonary artery (rat) Hypoxia MCT M8 Downregulation (88)
PASMC (rat) Hypoxia M8 Downregulation (89)
PASMC (rat) Hypoxia M2 Upregulation (90)
Pulmonary artery (rat) Hypoxia V4 Upregulation (91)
Lung (rat) Hypoxia V3 Upregulation (92)
Lung microvascular endothelial cell (rat) SuHx V4 No changing, but regulate migration and proliferation (93)
Adventitia (rat) Hypoxia MCT V4 Upregulation (94)

PASMC: pulmonary arterial smooth muscle cell; MCT: monocrotaline; SuHx: Sugen5416 + hypoxia.

The involvement of TRPM7 channels in cardiovascular remodeling has been reported (45,46,47,48,49,50,51,52). TRPM7 channels are activated by oxidative, mechanical, and osmotic stress, thereby contributing to various physiological events, such as inflammation, fibrosis, cell migration, contraction, and cell growth (53). The TRPM7 channel is a non-selective cation channel with unique features of divalent (Ca2+, Mg2+) conductance and a structure involving the alpha-kinase domain in its C-terminal region (54, 55). Mg2+ permeability in TRPM7-transfected cell lines is reportedly slightly higher than that of Ca2+, while the relative permeability of Na:Ca:Mg is estimated to be 1:1.23:1.29 (56, 57). Moreover, the difference between intracellular and extracellular Ca2+ concentrations ([Ca2+]o / [Ca2+]i ≥ 104) was much larger than that of Mg2+([Mg2+]o / [Mg2+]i ≥ 2). Endogenous TRPM7 currents are typically small; this is particularly true for Mg2+ currents because the driving force (Nernst potential) for Mg2+ across the plasma membrane is much smaller than that for Ca2+ (58, 59).

Ca2+ permeation via TRPM7 channels is associated with several important events such as those related to tissue remodeling. Ca2+ influx via TRPM7 contributes to angiotensin II-induced cardiac fibrosis (60) and epithelial-to-mesenchymal transition and migration in breast cancer cells (61). TRPM7-mediated activation of extracellular signal-regulated kinase (ERK) 1/2 and signal transducer and activator of transcription 3 (STAT3) essentially plays important roles in these processes. Similarly, TRPM7 may contribute to endothelial remodeling. Indeed, TRPM7 channel activation has been shown to induce EndoMT by increasing [Ca2+]i in sepsis (62). TRPM7 also plays a critical role in EndoMT of PAECs. In addition, TRPM7-mediated Ca2+ influx enhances endothelial cell migration (63).

The TRPM7 channel, which is associated with shear stress, regulates physiological and pathophysiological events such as cell migration, differentiation, and fibrogenesis (64,65,66). Shear stress-induced by fluid flow upregulates TRPM7 channel expression and activates it in vascular smooth muscle cells derived from the aorta (67). This indicates that vessel damage caused by shear stress triggers TRPM7 channel upregulation and activation. In the pulmonary artery, TRPM7 and TRPV4 are required for shear stress-induced Ca2+ increases in [Ca2+]i in PASMCs (51). TRPM7 is also responsible for the shear stress-induced Mg2+ influx. Moreover, the shear stress-induced Mg2+ increase observed in PASMCs derived from patients with PAH was greater than that observed in normal PASMCs (51). Accordingly, TRPM7 expression in PASMC was remarkably higher in PAH lungs than in non-PAH lungs. The TRPM7 channel is involved in platelet-derived growth factor BB (PDGF-BB)-induced proliferation of hepatic stellate cells (68). PDGF-BB is known to promote proliferation and migration of human PASMCs (69). The miR-1181/STAT3 axis has been suggested to contribute to the PDGF-BB-induced proliferation and migration of PAMSCs. TRPM7 activation induces STAT3 phosphorylation in glioma cells (70). In the lungs of patients with PAH, increased STAT3 phosphorylation has been implicated in the proliferative and survival phenotypes of cells that comprise plexogenic lesions (71). STAT3 is a cytoplasmic transcription factor. Upon phosphorylation of its tyrosine 705 residue in response to cytokines, such as interleukin-6 (IL-6) and PDGF (69, 72), STAT3 translocates to the nucleus and binds to DNA after dimerization, thereby regulating gene expression (73). In contrast, the IL-6/STAT3 axis modulates TRPM7 function (74).

Extracts of Medicinal Mushroom Ophiocordyceps sinensis Ameliorate PAH Partially via TRPM7 Inhibition

In addition to the current therapeutic agents for PAH, it is necessary to develop highly safe agents that target vascular remodeling. Therefore, we explored these agents as ingredients in natural herbal medicines. FTY-720, which is synthesized from a component derived from the medicinal mushroom Isaria sinclairii, is now known to be a TRPM7 inhibitor (Supplementary Fig. 1) (75, 76). Another medicinal mushroom, Ophiocordyceps sinensis (OCS), known as Yarsagumba, has long been used as a folk remedy for altitude sickness, which is usually encountered above 4,000 m sea level (77). OCS enhances cardiopulmonary function and exerts therapeutic effects on fibrotic lesions and malignant tumors (78, 79). Therefore, we examined the therapeutic effects of OCS and FTY-720 on the pathophysiology of PAH (45). We observed that the OCS extract ameliorated PAH and suppressed TRPM7 channel activity. However, the specific ingredients of OCS targeting the TRPM7 channel are undefined. Figure 1 summarizes the proposed mechanism underlying the therapeutic effects of OCS in PAH, based on our own investigation.

Fig. 1.

Fig. 1.

Proposed mechanism for the therapeutic effects of Ophiocordyceps sinensis (OCS) on vascular remodeling in pulmonary hypertension.

In human pulmonary artery endothelial cells (HPAEC), inhibition of TRPM7 channels by OCS inhibits the phosphorylation of STAT3 and Smad2, and endothelial-to-mesenchymal transition (EndoMT). In human pulmonary artery smooth muscle cells (HPASMC), inhibition of the TRPM7 channel by OCS inhibits the phosphorylation of STAT3 and Akt and cell proliferation.

In vitro experiments showed that FTY-720 and OCS inhibited TGF-β-induced Smad2 phosphorylation and EndoMT in human PAECs and the proliferation of PASMCs derived from patients with PAH. Furthermore, inhibition of TRPM7 by the OCS extract and FTY-720, as well as the knockdown of TRPM7 channel expression, suppressed the phosphorylation of STAT3 and Akt induced by IL-6 in PAECs, which are intracellular signaling pathways relevant to the EndoMT. Upregulation of TRPM7 channel expression, colocalized with αSMA in the medial wall and occlusive lesions, was also observed in the pulmonary arteries of patients with PAH. Furthermore, OCS extracts and FTY-720 induced relaxation in the human pulmonary artery during contraction induced by the thromboxane A2 analog, U46619.

In vivo experiments using a rat model of PAH induced by monocrotaline showed that treatment with the OCS extract improved survival, right ventricular hypertrophy and dysfunction, and vascular remodeling. In PAH rats, TRPM7 channel expression is upregulated in α-SMA-positive cells. Administration of OCS extracts also ameliorated pathological progression in a mouse model generated by monocrotaline pyrrole. Furthermore, in TRPM7 channel knockout mice, monocrotaline pyrrole-induced increases in the right ventricular pressure and muscularization of the pulmonary artery were ameliorated compared with those in wild-type mice.

Our results suggest that targeting TRPM7 may be a novel strategy for PAH treatment, with the aim of ameliorating pulmonary artery remodeling. However, other studies have suggested that TRPM7 inhibition leads to PAH progression (80). One study showed that PASMCs derived from hypoxia-induced PAH model rats exhibited decreased expression of TRPM7 protein, and that TRPM7 knockdown and treatment with waixenicin A, a TRPM7 inhibitor, enhanced proliferation and apoptosis in PASMCs, suggesting that the TRPM7 channel is a negative regulator of PAH progression. Mechanistically, TRPM7 channel-mediated Mg2+ transport underlies attenuation of PAH progression. Furthermore, Mg2+ supplementation was shown to improve pulmonary arterial pressure, right heart hypertrophy, and medial wall thickening in a rat model of severe PAH (52). Mg2+ supplementation also inhibited PASMC proliferation and migration and enhanced the apoptosis of PASMCs. These reports contradict our findings and should thus be considered. The contribution of the TRPM7 channel to the pathogenesis of PAH may vary depending on the experimental PAH model. In other words, the therapeutic effects of targeting TRPM7 may depend on the type of PAH.

Summary

Despite significant advancements in therapeutic strategies over the last two decades, PAH remains an incurable disease. The present study proposes OCS, a traditional Chinese medicine, as a new treatment option for PAH. OCS ameliorates pathological changes in a rodent model of PAH. At least part of the observed anti-remodeling effects associated with EndoMT and STAT3 signaling involve the TRPM7 channel. TRPM7 has been suggested to be a therapeutic target for OCS. In addition, OCS improved PAH by inhibiting the proliferation and contraction of pulmonary artery smooth muscle. Based on these results, OCS may be a novel therapeutic agent for PAH.

Conflict of Interest

No conflict of interest needs to be disclosed.

Supplementary Material

Supplementary Material
jsmr-58-050-s001.pdf (310.8KB, pdf)

Acknowledgments

This research was supported by a grant from the MEXT-Supported Program supporting the research activities of female researchers and a grant from the Gender Equality Promotion Office of Kagawa University.

References

  • 1.Lüscher TF, Boulanger CM, Dohi Y, Yang ZH. Endothelium-derived contracting factors. Hypertension. 1992; 19(2): 117–30. doi: 10.1161/01.HYP.19.2.117 [DOI] [PubMed] [Google Scholar]
  • 2.Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ Res. 2000; 87(10): 840–4. doi: 10.1161/01.RES.87.10.840 [DOI] [PubMed] [Google Scholar]
  • 3.Kuwabara Y, Tanaka-Ishikawa M, Abe K, Hirano M, Hirooka Y, Tsutsui H, et al. Proteinase-activated receptor 1 antagonism ameliorates experimental pulmonary hypertension. Cardiovasc Res. 2019; 115(8): 1357–68. doi: 10.1093/cvr/cvy284 [DOI] [PubMed] [Google Scholar]
  • 4.Rubin LJ. Primary pulmonary hypertension. N Engl J Med. 1997; 336(2): 111–7. doi: 10.1056/NEJM199701093360207 [DOI] [PubMed] [Google Scholar]
  • 5.Rubin LJ. Primary pulmonary hypertension. Chest. 1993; 104(1): 236–50. doi: 10.1378/chest.104.1.236 [DOI] [PubMed] [Google Scholar]
  • 6.Montani D, Günther S, Dorfmüller P, Perros F, Girerd B, Garcia G, et al. Pulmonary arterial hypertension. Orphanet J Rare Dis. 2013; 8: 97. doi: 10.1186/1750-1172-8-97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Humbert M, Guignabert C, Bonnet S, Dorfmüller P, Klinger JR, Nicolls MR, et al. Pathology and pathobiology of pulmonary hypertension: state of the art and research perspectives. Eur Respir J. 2019; 53(1): 1801887. doi: 10.1183/13993003.01887-2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Humbert M, Sitbon O, Simonneau G. Treatment of pulmonary arterial hypertension. N Engl J Med. 2004; 351(14): 1425–36. doi: 10.1056/NEJMra040291 [DOI] [PubMed] [Google Scholar]
  • 9.Humbert M, Sitbon O, Chaouat A, Bertocchi M, Habib G, Gressin V, et al. Survival in patients with idiopathic, familial, and anorexigen-associated pulmonary arterial hypertension in the modern management era. Circulation. 2010; 122(2): 156–63. doi: 10.1161/CIRCULATIONAHA.109.911818 [DOI] [PubMed] [Google Scholar]
  • 10.Ogawa A, Satoh T, Tamura Y, Fukuda K, Matsubara H. Survival of Japanese patients with idiopathic/heritable pulmonary arterial hypertension. Am J Cardiol. 2017; 119(9): 1479–84. doi: 10.1016/j.amjcard.2017.01.015 [DOI] [PubMed] [Google Scholar]
  • 11.Tamura Y, Kumamaru H, Satoh T, Miyata H, Ogawa A, Tanabe N, et al. Japan PH Registry (JAPHR) Network. Effectiveness and outcome of pulmonary arterial hypertension-specific therapy in Japanese patients with pulmonary arterial hypertension. Circ J. 2017; 82(1): 275–82. doi: 10.1253/circj.CJ-17-0139 [DOI] [PubMed] [Google Scholar]
  • 12.Hervé P, Lebrec D, Brenot F, Simonneau G, Humbert M, Sitbon O, et al. Pulmonary vascular disorders in portal hypertension. Eur Respir J. 1998; 11(5): 1153–66. doi: 10.1183/09031936.98.11051153 [DOI] [PubMed] [Google Scholar]
  • 13.Speich R, Jenni R, Opravil M, Pfab M, Russi EW. Primary pulmonary hypertension in HIV infection. Chest. 1991; 100(5): 1268–71. doi: 10.1378/chest.100.5.1268 [DOI] [PubMed] [Google Scholar]
  • 14.Coplan NL, Shimony RY, Ioachim HL, Wilentz JR, Posner DH, Lipschitz A, et al. Primary pulmonary hypertension associated with human immunodeficiency viral infection. Am J Med. 1990; 89(1): 96–9. doi: 10.1016/0002-9343(90)90105-M [DOI] [PubMed] [Google Scholar]
  • 15.Brenot F, Herve P, Petitpretz P, Parent F, Duroux P, Simonneau G. Primary pulmonary hypertension and fenfluramine use. Br Heart J. 1993; 70(6): 537–41. doi: 10.1136/hrt.70.6.537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.McMurray J, Bloomfield P, Miller HC. Irreversible pulmonary hypertension after treatment with fenfluramine. Br Med J (Clin Res Ed). 1986; 292(6515): 239–40. doi: 10.1136/bmj.292.6515.239-a [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Machado RD, Southgate L, Eichstaedt CA, Aldred MA, Austin ED, Best DH, et al. Pulmonary arterial hypertension: a current perspective on established and emerging molecular genetic defects. Hum Mutat. 2015; 36(12): 1113–27. doi: 10.1002/humu.22904 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Good RB, Gilbane AJ, Trinder SL, Denton CP, Coghlan G, Abraham DJ, et al. Endothelial to mesenchymal transition contributes to endothelial dysfunction in pulmonary arterial hypertension. Am J Pathol. 2015; 185(7): 1850–8. doi: 10.1016/j.ajpath.2015.03.019 [DOI] [PubMed] [Google Scholar]
  • 19.Ranchoux B, Antigny F, Rucker-Martin C, Hautefort A, Péchoux C, Bogaard HJ, et al. Endothelial-to-mesenchymal transition in pulmonary hypertension. Circulation. 2015; 131(11): 1006–18. doi: 10.1161/CIRCULATIONAHA.114.008750 [DOI] [PubMed] [Google Scholar]
  • 20.Zhang S, Fantozzi I, Tigno DD, Yi ES, Platoshyn O, Thistlethwaite PA, et al. Bone morphogenetic proteins induce apoptosis in human pulmonary vascular smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2003; 285(3): L740–54. doi: 10.1152/ajplung.00284.2002 [DOI] [PubMed] [Google Scholar]
  • 21.McMurtry MS, Archer SL, Altieri DC, Bonnet S, Haromy A, Harry G, et al. Gene therapy targeting survivin selectively induces pulmonary vascular apoptosis and reverses pulmonary arterial hypertension. J Clin Invest. 2005; 115(6): 1479–91. doi: 10.1172/JCI23203 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Shu DY, Butcher E, Saint-Geniez M. EMT and EndMT: emerging roles in age-related macular degeneration. Int J Mol Sci. 2020; 21(12): 4271. doi: 10.3390/ijms21124271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ubil E, Duan J, Pillai ICL, Rosa-Garrido M, Wu Y, Bargiacchi F, et al. Mesenchymal-endothelial transition contributes to cardiac neovascularization. Nature. 2014; 514(7524): 585–90. doi: 10.1038/nature13839 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Arciniegas E, Frid MG, Douglas IS, Stenmark KR. Perspectives on endothelial-to-mesenchymal transition: potential contribution to vascular remodeling in chronic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2007; 293(1): L1–8. doi: 10.1152/ajplung.00378.2006 [DOI] [PubMed] [Google Scholar]
  • 25.Yi ES, Kim H, Ahn H, Strother J, Morris T, Masliah E, et al. Distribution of obstructive intimal lesions and their cellular phenotypes in chronic pulmonary hypertension. A morphometric and immunohistochemical study. Am J Respir Crit Care Med. 2000; 162(4 Pt 1): 1577–86. doi: 10.1164/ajrccm.162.4.9912131 [DOI] [PubMed] [Google Scholar]
  • 26.Frid MG, Kale VA, Stenmark KR. Mature vascular endothelium can give rise to smooth muscle cells via endothelial-mesenchymal transdifferentiation: in vitro analysis. Circ Res. 2002; 90(11): 1189–96. doi: 10.1161/01.RES.0000021432.70309.28 [DOI] [PubMed] [Google Scholar]
  • 27.Fraisl P, Mazzone M, Schmidt T, Carmeliet P. Regulation of angiogenesis by oxygen and metabolism. Dev Cell. 2009; 16(2): 167–79. doi: 10.1016/j.devcel.2009.01.003 [DOI] [PubMed] [Google Scholar]
  • 28.Davies PF. Flow-mediated endothelial mechanotransduction. Physiol Rev. 1995; 75(3): 519–60. doi: 10.1152/physrev.1995.75.3.519 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N Engl J Med. 2000; 342(18): 1350–8. doi: 10.1056/NEJM200005043421807 [DOI] [PubMed] [Google Scholar]
  • 30.Böttner M, Krieglstein K, Unsicker K. The transforming growth factor-betas: structure, signaling, and roles in nervous system development and functions. J Neurochem. 2000; 75(6): 2227–40. doi: 10.1046/j.1471-4159.2000.0752227.x [DOI] [PubMed] [Google Scholar]
  • 31.Risau W. Mechanisms of angiogenesis. Nature. 1997; 386(6626): 671–4. doi: 10.1038/386671a0 [DOI] [PubMed] [Google Scholar]
  • 32.Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat Med. 2000; 6(4): 389–95. doi: 10.1038/74651 [DOI] [PubMed] [Google Scholar]
  • 33.Medina C, Santos-Martinez MJ, Santana A, Paz-Cabrera MC, Johnston MJ, Mourelle M, et al. Transforming growth factor-beta type 1 receptor (ALK5) and Smad proteins mediate TIMP-1 and collagen synthesis in experimental intestinal fibrosis. J Pathol. 2011; 224(4): 461–72. doi: 10.1002/path.2870 [DOI] [PubMed] [Google Scholar]
  • 34.Biancheri P, Giuffrida P, Docena GH, MacDonald TT, Corazza GR, Di Sabatino A. The role of transforming growth factor (TGF)-β in modulating the immune response and fibrogenesis in the gut. Cytokine Growth Factor Rev. 2014; 25(1): 45–55. doi: 10.1016/j.cytogfr.2013.11.001 [DOI] [PubMed] [Google Scholar]
  • 35.Gwon MG, An HJ, Kim JY, Kim WH, Gu H, Kim HJ, et al. Anti-fibrotic effects of synthetic TGF-β1 and Smad oligodeoxynucleotide on kidney fibrosis in vivo and in vitro through inhibition of both epithelial dedifferentiation and endothelial-mesenchymal transitions. FASEB J. 2020; 34(1): 333–49. doi: 10.1096/fj.201901307RR [DOI] [PubMed] [Google Scholar]
  • 36.Guignabert C, Tu L, Le Hiress M, Ricard N, Sattler C, Seferian A, et al. Pathogenesis of pulmonary arterial hypertension: lessons from cancer. Eur Respir Rev. 2013; 22(130): 543–51. doi: 10.1183/09059180.00007513 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Weiss A, Neubauer MC, Yerabolu D, Kojonazarov B, Schlueter BC, Neubert L, et al. Targeting cyclin-dependent kinases for the treatment of pulmonary arterial hypertension. Nat Commun. 2019; 10(1): 2204. doi: 10.1038/s41467-019-10135-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.He RL, Wu ZJ, Liu XR, Gui LX, Wang RX, Lin MJ. Calcineurin/NFAT signaling modulates pulmonary artery smooth muscle cell proliferation, migration and apoptosis in monocrotaline-induced pulmonary arterial hypertension rats. Cell Physiol Biochem. 2018; 49(1): 172–89. doi: 10.1159/000492852 [DOI] [PubMed] [Google Scholar]
  • 39.Firth AL, Remillard CV, Yuan JX. TRP channels in hypertension. Biochim Biophys Acta. 2007; 1772(8): 895–906. doi: 10.1016/j.bbadis.2007.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Clapham DE, Runnels LW, Strübing C. The TRP ion channel family. Nat Rev Neurosci. 2001; 2(6): 387–96. doi: 10.1038/35077544 [DOI] [PubMed] [Google Scholar]
  • 41.Earley S, Brayden JE. Transient receptor potential channels in the vasculature. Physiol Rev. 2015; 95(2): 645–90. doi: 10.1152/physrev.00026.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Fujiwara Y, Minor DL, Jr . X-ray crystal structure of a TRPM assembly domain reveals an antiparallel four-stranded coiled-coil. J Mol Biol. 2008; 383(4): 854–70. doi: 10.1016/j.jmb.2008.08.059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Liao M, Cao E, Julius D, Cheng Y. Structure of the TRPV1 ion channel determined by electron cryo-microscopy. Nature. 2013; 504(7478): 107–12. doi: 10.1038/nature12822 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lin MJ, Leung GP, Zhang WM, Yang XR, Yip KP, Tse CM, et al. Chronic hypoxia-induced upregulation of store-operated and receptor-operated Ca2+ channels in pulmonary arterial smooth muscle cells: a novel mechanism of hypoxic pulmonary hypertension. Circ Res. 2004; 95(5): 496–505. doi: 10.1161/01.RES.0000138952.16382.ad [DOI] [PubMed] [Google Scholar]
  • 45.Hiraishi K, Kurahara LH, Feng J, Yamamura A, Cui Y, Yahiro E, et al. Substantial involvement of TRPM7 inhibition in the therapeutic effect of Ophiocordyceps sinensis on pulmonary hypertension. Transl Res. 2021; 233: 127–43. doi: 10.1016/j.trsl.2021.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Li Y, Jiang H, Ruan C, Zhong J, Gao P, Zhu D, et al. The interaction of transient receptor potential melastatin 7 with macrophages promotes vascular adventitial remodeling in transverse aortic constriction rats. Hypertens Res. 2014; 37(1): 35–42. doi: 10.1038/hr.2013.110 [DOI] [PubMed] [Google Scholar]
  • 47.Liu J, Chen L, Huang J, Guo S, Zhu D, Gao P. Transient receptor potential melastatin 7 promotes vascular adventitial fibroblasts phenotypic transformation and inflammatory reaction induced by mechanical stretching stress via p38 MAPK/JNK pathway. J Vasc Res. 2021; 58(2): 108–20. doi: 10.1159/000512595 [DOI] [PubMed] [Google Scholar]
  • 48.Negri S, Faris P, Berra-Romani R, Guerra G, Moccia F. Endothelial transient receptor potential channels and vascular remodeling: extracellular Ca2 + entry for angiogenesis, arteriogenesis and vasculogenesis. Front Physiol. 2020; 10: 1618. doi: 10.3389/fphys.2019.01618 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Paravicini TM, Yogi A, Mazur A, Touyz RM. Dysregulation of vascular TRPM7 and annexin-1 is associated with endothelial dysfunction in inherited hypomagnesemia. Hypertension. 2009; 53(2): 423–9. doi: 10.1161/HYPERTENSIONAHA.108.124651 [DOI] [PubMed] [Google Scholar]
  • 50.Rios FJ, Zou ZG, Harvey AP, Harvey KY, Nosalski R, Anyfanti P, et al. Chanzyme TRPM7 protects against cardiovascular inflammation and fibrosis. Cardiovasc Res. 2020; 116(3): 721–35. doi: 10.1093/cvr/cvz164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Song S, Yamamura A, Yamamura H, Ayon RJ, Smith KA, Tang H, et al. Flow shear stress enhances intracellular Ca2+ signaling in pulmonary artery smooth muscle cells from patients with pulmonary arterial hypertension. Am J Physiol Cell Physiol. 2014; 307(4): C373–83. doi: 10.1152/ajpcell.00115.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wang D, Zhu ZL, Lin DC, Zheng SY, Chuang KH, Gui LX, et al. Magnesium supplementation attenuates pulmonary hypertension via regulation of magnesium transporters. Hypertension. 2021; 77(2): 617–31. doi: 10.1161/HYPERTENSIONAHA.120.14909 [DOI] [PubMed] [Google Scholar]
  • 53.Yogi A, Callera GE, Antunes TT, Tostes RC, Touyz RM. Transient receptor potential melastatin 7 (TRPM7) cation channels, magnesium and the vascular system in hypertension. Circ J. 2011; 75(2): 237–45. doi: 10.1253/circj.CJ-10-1021 [DOI] [PubMed] [Google Scholar]
  • 54.Nadler MJ, Hermosura MC, Inabe K, Perraud AL, Zhu Q, Stokes AJ, et al. LTRPC7 is a Mg.ATP-regulated divalent cation channel required for cell viability. Nature. 2001; 411(6837): 590–5. doi: 10.1038/35079092 [DOI] [PubMed] [Google Scholar]
  • 55.Takezawa R, Schmitz C, Demeuse P, Scharenberg AM, Penner R, Fleig A. Receptor-mediated regulation of the TRPM7 channel through its endogenous protein kinase domain. Proc Natl Acad Sci USA. 2004; 101(16): 6009–14. doi: 10.1073/pnas.0307565101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Li M, Jiang J, Yue L. Functional characterization of homo- and heteromeric channel kinases TRPM6 and TRPM7. J Gen Physiol. 2006; 127(5): 525–37. doi: 10.1085/jgp.200609502 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hanano T, Hara Y, Shi J, Morita H, Umebayashi C, Mori E, et al. Involvement of TRPM7 in cell growth as a spontaneously activated Ca2+ entry pathway in human retinoblastoma cells. J Pharmacol Sci. 2004; 95(4): 403–19. doi: 10.1254/jphs.FP0040273 [DOI] [PubMed] [Google Scholar]
  • 58.Visser D, Middelbeek J, van Leeuwen FN, Jalink K. Function and regulation of the channel-kinase TRPM7 in health and disease. Eur J Cell Biol. 2014; 93(10-12): 455–65. doi: 10.1016/j.ejcb.2014.07.001 [DOI] [PubMed] [Google Scholar]
  • 59.Gunn RB, Curran PF. Membrane potentials and ion permeability in a cation exchange membrane. Biophys J. 1971; 11(7): 559–71. doi: 10.1016/S0006-3495(71)86235-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yu Y, Chen S, Xiao C, Jia Y, Guo J, Jiang J, et al. TRPM7 is involved in angiotensin II induced cardiac fibrosis development by mediating calcium and magnesium influx. Cell Calcium. 2014; 55(5): 252–60. doi: 10.1016/j.ceca.2014.02.019 [DOI] [PubMed] [Google Scholar]
  • 61.Davis FM, Azimi I, Faville RA, Peters AA, Jalink K, Putney JW, Jr , et al. Induction of epithelial-mesenchymal transition (EMT) in breast cancer cells is calcium signal dependent. Oncogene. 2014; 33(18): 2307–16. doi: 10.1038/onc.2013.187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Echeverría C, Montorfano I, Hermosilla T, Armisén R, Velásquez LA, Cabello-Verrugio C, et al. Endotoxin induces fibrosis in vascular endothelial cells through a mechanism dependent on transient receptor protein melastatin 7 activity. PLoS One. 2014; 9(4): e94146. doi: 10.1371/journal.pone.0094146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sarmiento D, Montorfano I, Cáceres M, Echeverría C, Fernández R, Cabello-Verrugio C, et al. Endotoxin-induced vascular endothelial cell migration is dependent on TLR4/NF-κB pathway, NAD(P)H oxidase activation, and transient receptor potential melastatin 7 calcium channel activity. Int J Biochem Cell Biol. 2014; 55: 11–23. doi: 10.1016/j.biocel.2014.08.001 [DOI] [PubMed] [Google Scholar]
  • 64.Thilo F, Vorderwülbecke BJ, Marki A, Krueger K, Liu Y, Baumunk D, et al. Pulsatile atheroprone shear stress affects the expression of transient receptor potential channels in human endothelial cells. Hypertension. 2012; 59(6): 1232–40. doi: 10.1161/HYPERTENSIONAHA.111.183608 [DOI] [PubMed] [Google Scholar]
  • 65.Yankaskas CL, Bera K, Stoletov K, Serra SA, Carrillo-Garcia J, Tuntithavornwat S, et al. The fluid shear stress sensor TRPM7 regulates tumor cell intravasation. Sci Adv. 2021; 7(28): eabh3457. doi: 10.1126/sciadv.abh3457 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Liu YS, Liu YA, Huang CJ, Yen MH, Tseng CT, Chien S, et al. Mechanosensitive TRPM7 mediates shear stress and modulates osteogenic differentiation of mesenchymal stromal cells through Osterix pathway. Sci Rep. 2015; 5: 16522. doi: 10.1038/srep16522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Oancea E, Wolfe JT, Clapham DE. Functional TRPM7 channels accumulate at the plasma membrane in response to fluid flow. Circ Res. 2006; 98(2): 245–53. doi: 10.1161/01.RES.0000200179.29375.cc [DOI] [PubMed] [Google Scholar]
  • 68.Fang L, Zhan S, Huang C, Cheng X, Lv X, Si H, et al. TRPM7 channel regulates PDGF-BB-induced proliferation of hepatic stellate cells via PI3K and ERK pathways. Toxicol Appl Pharmacol. 2013; 272(3): 713–25. doi: 10.1016/j.taap.2013.08.009 [DOI] [PubMed] [Google Scholar]
  • 69.Qian Z, Li Y, Yang H, Chen J, Li X, Gou D. PDGFBB promotes proliferation and migration via regulating miR-1181/STAT3 axis in human pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2018; 315(6): L965–76. doi: 10.1152/ajplung.00224.2018 [DOI] [PubMed] [Google Scholar]
  • 70.Liu M, Inoue K, Leng T, Guo S, Xiong ZG. TRPM7 channels regulate glioma stem cell through STAT3 and Notch signaling pathways. Cell Signal. 2014; 26(12): 2773–81. doi: 10.1016/j.cellsig.2014.08.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Paulin R, Meloche J, Bonnet S. STAT3 signaling in pulmonary arterial hypertension. JAK-STAT. 2012; 1(4): 223–33. doi: 10.4161/jkst.22366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wen Z, Darnell JE, Jr . Mapping of Stat3 serine phosphorylation to a single residue (727) and evidence that serine phosphorylation has no influence on DNA binding of Stat1 and Stat3. Nucleic Acids Res. 1997; 25(11): 2062–7. doi: 10.1093/nar/25.11.2062 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Bromberg JF, Horvath CM, Besser D, Lathem WW, Darnell JE, Jr . Stat3 activation is required for cellular transformation by v-src. Mol Cell Biol. 1998; 18(5): 2553–8. doi: 10.1128/MCB.18.5.2553 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Liu A, Zhao F, Wang J, Zhao Y, Luo Z, Gao Y, et al. Regulation of TRPM7 Function by IL-6 through the JAK2-STAT3 Signaling Pathway. PLoS One. 2016; 11(3): e0152120. doi: 10.1371/journal.pone.0152120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chiba K. FTY720, a new class of immunomodulator, inhibits lymphocyte egress from secondary lymphoid tissues and thymus by agonistic activity at sphingosine 1-phosphate receptors. Pharmacol Ther. 2005; 108(3): 308–19. doi: 10.1016/j.pharmthera.2005.05.002 [DOI] [PubMed] [Google Scholar]
  • 76.Qin X, Yue Z, Sun B, Yang W, Xie J, Ni E, et al. Sphingosine and FTY720 are potent inhibitors of the transient receptor potential melastatin 7 (TRPM7) channels. Br J Pharmacol. 2013; 168(6): 1294–312. doi: 10.1111/bph.12012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Baral B. Entomopathogenicity and biological attributes of Himalayan treasured fungus ophiocordyceps sinensis (Yarsagumba). J Fungi (Basel). 2017; 3(1): E4. doi: 10.3390/jof3010004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Gao BA, Yang J, Huang J, Cui XJ, Chen SX, Den HY, et al. Cordyceps sinensis extract suppresses hypoxia-induced proliferation of rat pulmonary artery smooth muscle cells. Saudi Med J. 2010; 31(9): 974–9. [PubMed] [Google Scholar]
  • 79.Luitel H, Novoyatleva T, Sydykov A, Petrovic A, Mamazhakypov A, Devkota B, et al. Yarsagumba is a promising therapeutic option for treatment of pulmonary hypertension due to the potent anti-proliferative and vasorelaxant properties. Medicina (Kaunas). 2020; 56(3): E131. doi: 10.3390/medicina56030131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Xing J, Wang M, Hong J, Gao Y, Liu Y, Gu H, et al. TRPM7 channel inhibition exacerbates pulmonary arterial hypertension through MEK/ERK pathway. Aging (Albany NY). 2019; 11(12): 4050–65. doi: 10.18632/aging.102036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wang J, Weigand L, Lu W, Sylvester JT, Semenza GL, Shimoda LA. Hypoxia inducible factor 1 mediates hypoxia-induced TRPC expression and elevated intracellular Ca2+ in pulmonary arterial smooth muscle cells. Circ Res. 2006; 98(12): 1528–37. doi: 10.1161/01.RES.0000227551.68124.98 [DOI] [PubMed] [Google Scholar]
  • 82.Liu XR, Zhang MF, Yang N, Liu Q, Wang RX, Cao YN, et al. Enhanced store-operated Ca2+ entry and TRPC channel expression in pulmonary arteries of monocrotaline-induced pulmonary hypertensive rats. Am J Physiol Cell Physiol. 2012; 302(1): C77–87. doi: 10.1152/ajpcell.00247.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Malczyk M, Veith C, Fuchs B, Hofmann K, Storch U, Schermuly RT, et al. Classical transient receptor potential channel 1 in hypoxia-induced pulmonary hypertension. Am J Respir Crit Care Med. 2013; 188(12): 1451–9. doi: 10.1164/rccm.201307-1252OC [DOI] [PubMed] [Google Scholar]
  • 84.Castillo-Galán S, Arenas GA, Reyes RV, Krause BJ, Iturriaga R. Stim-activated TRPC-ORAI channels in pulmonary hypertension induced by chronic intermittent hypoxia. Pulm Circ. 2020; 10(1 Suppl): 13–22. doi: 10.1177/2045894020941484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Qin Y, Zhu B, Li L, Wang D, Qiao Y, Liu B, et al. Overexpressed lncRNA AC068039.4 contributes to proliferation and cell cycle progression of pulmonary artery smooth muscle cells via sponging miR-26a-5p/TRPC6 in hypoxic pulmonary arterial hypertension. Shock. 2021; 55(2): 244–55. doi: 10.1097/SHK.0000000000001606 [DOI] [PubMed] [Google Scholar]
  • 86.Wang Y, Li N, Wang Y, Zheng G, An J, Liu C, et al. NF-κB/p65 competes with peroxisome proliferator-activated receptor gamma for transient receptor potential channel 6 in hypoxia-induced human pulmonary arterial smooth muscle cells. Front Cell Dev Biol. 2021; 9: 656625. doi: 10.3389/fcell.2021.656625 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Xu J, Wen X, Fu Z, Jiang Y, Hong W, Liu R, et al. Chronic hypoxia promoted pulmonary arterial smooth muscle cells proliferation through upregulated calcium-sensing receptorcanonical transient receptor potential 1/6 pathway. Microcirculation. 2021; 28(6): e12715. doi: 10.1111/micc.12715 [DOI] [PubMed] [Google Scholar]
  • 88.Liu XR, Liu Q, Chen GY, Hu Y, Sham JS, Lin MJ. Down-regulation of TRPM8 in pulmonary arteries of pulmonary hypertensive rats. Cell Physiol Biochem. 2013; 31(6): 892–904. doi: 10.1159/000350107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Mu YP, Lin DC, Zheng SY, Jiao HX, Sham JSK, Lin MJ. Transient receptor potential melastatin-8 activation induces relaxation of pulmonary artery by inhibition of store-operated calcium entry in normoxic and chronic hypoxic pulmonary hypertensive rats. J Pharmacol Exp Ther. 2018; 365(3): 544–55. doi: 10.1124/jpet.117.247320 [DOI] [PubMed] [Google Scholar]
  • 90.Song JL, Zheng SY, He RL, Gui LX, Lin MJ, Sham JSK. Serotonin and chronic hypoxic pulmonary hypertension activate a NADPH oxidase 4 and TRPM2 dependent pathway for pulmonary arterial smooth muscle cell proliferation and migration. Vascul Pharmacol. 2021; 138: 106860. doi: 10.1016/j.vph.2021.106860 [DOI] [PubMed] [Google Scholar]
  • 91.Yang XR, Lin AH, Hughes JM, Flavahan NA, Cao YN, Liedtke W, et al. Upregulation of osmo-mechanosensitive TRPV4 channel facilitates chronic hypoxia-induced myogenic tone and pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2012; 302(6): L555–68. doi: 10.1152/ajplung.00005.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zhang Q, Cao Y, Luo Q, Wang P, Shi P, Song C, et al. The transient receptor potential vanilloid-3 regulates hypoxia-mediated pulmonary artery smooth muscle cells proliferation via PI3K/AKT signaling pathway. Cell Prolif. 2018; 51(3): e12436. doi: 10.1111/cpr.12436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Suresh K, Servinsky L, Jiang H, Bigham Z, Yun X, Kliment C, et al. Reactive oxygen species induced Ca2+ influx via TRPV4 and microvascular endothelial dysfunction in the SU5416/hypoxia model of pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol. 2018; 314(5): L893–907. doi: 10.1152/ajplung.00430.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Cussac LA, Cardouat G, Tiruchellvam Pillai N, Campagnac M, Robillard P, Montillaud A, et al. TRPV4 channel mediates adventitial fibroblast activation and adventitial remodeling in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2020; 318(1): L135–46. doi: 10.1152/ajplung.00084.2019 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Material
jsmr-58-050-s001.pdf (310.8KB, pdf)

Articles from Journal of Smooth Muscle Research are provided here courtesy of Japan Society of Smooth Muscle Research

RESOURCES