1,403
Views
1
CrossRef citations to date
0
Altmetric
Review

A promising field: regulating imbalance of EndMT in cardiovascular diseases

, , , , , , , & show all
Pages 1477-1486 | Received 18 Nov 2020, Accepted 30 Jun 2021, Published online: 16 Jul 2021

ABSTRACT

Endothelial–mesenchymal transition (EndMT) is widely involved in the occurrence and development of cardiovascular diseases. Although there is no direct evidence, it is very promising as an effective target for the treatment of these diseases. Endothelial cells need to respond to the complex cardiovascular environment through EndMT, but sustained stimuli will cause the imbalance of EndMT. Blocking the signal transduction promoting EndMT is an effective method to control the imbalance of EndMT. In particular, we also discussed the potential role of endothelial cell apoptosis and autophagy in regulating the imbalance of EndMT. In addition, promoting mesenchymal-endothelial transformation (MEndT) is also a method to control the imbalance of EndMT. However, targeting EndMT to treat cardiovascular disease still faces many challenges. By reviewing the research progress of EndMT, we have put forward some insights and translated them into challenges and opportunities for new treatment strategies for cardiovascular diseases.

1. Endothelial–mesenchymal transition in the cardiovascular diseases

Endothelial–mesenchymal transition (EndMT), a subtype of epithelial–mesenchymal transition (EMT), is that endothelial cells (ECs) lose endothelial characteristics and acquire characteristics of mesenchymal cells. Cardiovascular System always accompanies EndMT: EndMT plays a key role in heart development; EndMT is a reversible and homeostasis process, which can be disrupted by post-developmental EndMT resulting in deterioration of vascular function and plaque formation observed in atherosclerosis [Citation1,Citation2]. EndMT is required for normal heart valve development, and EndMT occurs in endocardial cells activated by the transforming growth factor-β (TGF-β) and other ligands, triggering their migration to the endocardial cushion and forming the heart valve stroma [Citation3]. Endothelial fatty acid oxidation (FAO), a key regulator of EndMT, works through changes in intracellular acetyl-CoA levels, Xiong J et al. Found, and deletion of endothelial carnitine palmitoyl transferase II (Cpt2E-KO) disruping FAO increases the size of the embryo’s EndMT, leading to thickening of the heart valve [Citation4]. Studies have shown that endothelial cells transform to mesenchymal cells under hypoxic conditions, invade cardiac tissue to promote myocardial fibrosis, and release TGFβ-1 to induce myocardial cell apoptosis, leading to the development of heart failure in vivo [Citation5]. Though many studies show percentage of mesenchymal cells (derived from endothelial cells) rate is relatively low it is the mesenchymal cells (derived from endothelial cells) disturb the barrier function of the endothelium thus enable enhanced crosstalk of the endothelium and cardiomyocytes. These indicate that EndMT contributes to the accumulation of cardiac fibroblasts and loss of cardiomyocytes, which promotes myocardial fibrosis. In addition, endothelial-derived mesenchymal cells also promote diabetic myocardial fibrosis [Citation6], and chronic pulmonary hypertension [Citation7].

EndMT is widely involved in the occurrence and development of atherosclerosis. In addition to the aforementioned we mentioned that EndMT is involved in the formation of atherosclerotic plaques. The earliest stage of atherosclerotic lesions occurs in endothelial cells [Citation8]. Previous studies have shown that oscillatory-shear (OSS) [Citation9] and low-shear [Citation10] stress-induced EndMT is a potential pathogenesis of early atherosclerosis, and the anti-inflammatory and anticoagulant effects of uniform laminar flow shear stress (LSS) on endothelial cells are anti-atherosclerotic [Citation11]. Moonen JR’s group demonstrated that EndMT is involved in fibroproliferative vascular disease, and it promotes neointimal hyperplasia and atherosclerotic differentiation of endothelial cells. Importantly, they found that EndMT was regulated by shear stress in an ERK5-dependent manner, and showed that this is a new atherosclerotic protection mechanism for uniform layer shear stress, which explains to some extent focal vascular disease [Citation12]. What is important is the development of atherosclerosis, such as the formation of plaques, which will cause changes in local blood flow to form low shear stress and oscillating shear stress to promote EndMT and accelerate the process of atherosclerosis to form a vicious circle [Citation13]. A rise in Reactive Oxygen Species (ROS) beyond a “normal” or “physiological” threshold level results in a process called “oxidative stress”, which can also promote EndMT [Citation14]. In addition to oxidative stress EndMT mediated endothelial fibrosis are closely connected to the inflammatory conditions, and the key regulator of inflammation NF-κB has been reported to promote the development of EndMT, causing endothelial cells to differentiate into fibroblasts and impair function [Citation15]. There is also evidence supporting osteogenic cells derived from the vascular endothelium contributing vascular calcification [Citation16]. The enhanced BMP signaling results in EndMT was an essential step [Citation16]. Moreover sex determining region Y-box 2 mediates activation of specific serine proteases which is essential for initiating EndMT [Citation17]. Furthermore fibrodysplasia ossificans progressiva (FOP) is also a vascular disease based on EndMT induces a stem cell-like phenotype [Citation18]. Proteomics analysis of non-dilated ascending aorta by Maleki S et al. found that EndMT activity was increased in patients with bicuspid aortic valve (BAV), and speculated that the induction of EndMT/EMT in BAV ascending aorta may be related to the aorta’s constant exposure to non-physiological hemodynamics and/Or embryo developmental defects [Citation19]. EndMT leads to instability of endothelial cell connections and enhanced vascular permeability of the ascending aorta, which may lay the foundation for increased susceptibility to aneurysms [Citation20]. Inhibiting EndMT only reduces the possibility of aneurysm, and cannot reverse the deterioration of the aortic valve wall.

There is also evidence supporting EndMT is involved in angiogenesis [Citation21] which is a complex, multi-step process including a series of continuous events that begin with the proliferation and survival of ECs, then they migrate and finally differentiate into capillary-like networks. One of the characteristics of advanced proliferative diabetic retinopathy is the formation of abnormal new blood vessels on the surface of the retina [Citation22]. Based on previous research reports that high glucose can induce endothelial cells to undergo EndMT, Feng L et al. further investigated the role of RKIP, a Raf kinase inhibitor protein (RKIP), in the glucose-induced EndMT in capillary endothelial cells in Diabetic retinopathy (DR), and the result showed that it negatively regulates EndMT [Citation23]. The beneficial angiogenesis benefits from the regulation of EndMT dynamic balance. However, in most cases, EndMT cannot be well regulated and breaks the vascular homeostasis, leading to malignant results such as vascular remodeling. Therefore, whether angiogenesis can be beneficially developed has yet to be further studied. Maintaining the balance of EndMT may be a good choice. In aggregate, the imbalance of EndMT is closely related to myocardial fibrosis of the cardiovascular system, atherosclerosis, vascular calcification, aneurysms and angiogenesis.

2. Recent advances in regulating imbalance of EndMT

2.1 Block signaling pathways of promoting EndMT

The most direct strategy to inhibit endothelial mesenchymal transformation is to interfere with the signaling pathway that activates EndMT. The TGF-β superfamily are the most prominent participants in EndMT, and the pleiotropic effect of TGF-β on endothelial cells has been reported in detail [Citation24]. TGFβ signaling through Smad-dependent and independent pathways direct regulates EndMT [Citation24]. In TGF-β/Smad1 signal transduction, TGF-β1 binds to transforming growth factor-βRII, recruits and phosphorylates TGF-βRI, and activates Smad2/3/4 consisting of phosphorylated Smad2/3 and Smad4/3 and Smad4 complex [Citation25]. And then the complexs are shifted into the nucleus to induce RELM-β transcription [Citation25]. SB432542, a SMAD inhibitor, could reduce the protein levels of Resistin-like molecule-β (RELM-β), p-SMAD2, p-SMAD3, and SMAD4 which could ameliorate TGF-β1-induced EndMT in HUVECs and HPAECs [Citation25]. It has been reported that TGF‐β silenced sirtuin 1 (SIRT1) expression via promoter hypermethylation and histone modification [Citation26]. However, as a class III histone deacetylase, its activation inhibits TGF-β-induced EndMT in HUVEC cell lines with Smad4 deacetylation [Citation27]. Another study showed that upregulation of SIRT1 deacetylase activity attenuated TGF-β-induced the expression of extracelluar matrix proteins through inhibiting Smad3 in murine mesangial cell line cells [Citation7]. In addition, the activation of SIRT1 also suppressed that of TGF-βR1 expression and inhibited the nuclear translocation of Smad2/3 in TGF-β1-treated H5V cells [Citation28]. In TGF-β signaling through Smad-independent pathway, SIRT1 reportedly inhibited Akt signaling pathways in cardiac fibrosis [Citation29]. However, Sirt1 may promote TGF-β/Smad-dependent transcription in fibroblasts, due to Sirt1 negatively regulates the expression of Smad7, an endogenous inhibitor of canonical Smad signaling in fibroblasts [Citation27,Citation30]. The progress of EndMT through TGF-β/Smad and Akt/mTOR/p70S6K signaling pathways plays an important role in renal allograft recipients with renal interstitial fibrosis and chronic allograft dysfunction [Citation31], according to Wang Z et al., and in their further research they found that hepatocyte growth factor (HGF) inhibits TGF-β1-induced EndMT of HUVEC and HRGECs by reducing the levels of phosphorylated Akt, mTOR, p70S6K, Smad2, and Smad3 [Citation32].

Aside from TGF-β, the canonical Wnt signaling has also been reported to regulate EndMT. The natural regulation of the Wnt pathway occurs mainly at the level of inhibitors secreted extracellularly. In the study by Blyszczuk P et al. TGF-β-dependent myofibroblast differentiation is mediated by secreted WNTs, myocardial fibrosis depends on the bioavailability of extracellular WNTs, and TAK1-dependent Wnt protein secretion represents TGF-β-mediated human and mouse Myocarditis is a new downstream key mechanism of myocardial fibrosis and remodeling, and their data also suggest that inhibition of Wnt signaling can improve cardiac function [Citation33]. Dickkopf-1 (DKK-1) a powerful antagonist of wnt/β-catenin signaling, which can mediate endothelial cell activation via inhibiting the Wnt /β-catenin pathway. DKK-1 stimulation increased the expression of EndMT markers such as N-cadherin, Twist, and vimentin, while reducing the expression of endothelial cell marker VE-cadherin and promoted EndMT. Interestingly, it enhances angiogenesis in HUVEC cells by up-regulating VEGFR2 mRNA expression independent of Wnt signaling pathway [Citation34]. In addition, the standardized Wnt signal is an important component of aortic valve and vascular calcification. Cheng SL et al. found that MSX2 and WNT7 family members stabilized the bovine AoEC phenotype, while Dkk1 promoted EndMT in cultured bovine aortic endothelial cells (AoECs). The animal experiments demonstrate that Cdh5-Cre; Wnt7b(fl/fl); LDLR−/− mice exhibited more significant aortic fibrosis, collagen accumulation, and calcium deposition after challenge with atherogenic high-fat diets [Citation35]. Dickkopf-3 in abnormal endothelial cell secretions is a possible ligand of the Wnt/bcatenin pathway, which not only antagonizes the role of the known Wnt pathway inhibitor Dkk1 in the process of fibroblasts to myofibroblasts, but also induces EndMT [Citation36].

There are also reports that the Notch signaling pathway is involved in the EndMT process. In one study, one of the possible mechanisms by which Relaxin inhibits cardiac fibrosis is the inhibition of EndMT in fibrotic hearts, and the Notch pathway may mediate this process [Citation37]. According to Lin QQ et al., activation of Notch signaling pathway can induce the progression of EndMT and promote the development of atherosclerotic lesions [Citation38]. In the study by Zhang J et al., they found that miR-29a/b negatively regulate Notch2 expression and inhibit high glucose-induced EndMT in human retinal microvascular endothelial cells, which can be reversed by overexpression of Notch2 [Citation39]. Numb has been shown to be a negative regulator of EndMT, which negatively regulates Notch signaling pathway in EndMT of Diabetic nephropathy (DN) [Citation40]. In addition, Scutellarin prevents isoprenaline-induced myocardial fibrosis via inhibition of cardiac EndMT potentially, which is also associated with the Notch pathway [Citation41].

In addition to the above signaling factors, EndMT induced by TGF-β can be inhibited by fibroblast growth factor (FGF) by modulating let-7 miRNA [Citation42]. Vascular endothelial growth factor (VEGF) inhibits EndMT through VEGF-R2, but through VEGF-R1 promotes EndMT by reducing the bioavailability of VEGF [Citation43,Citation44]. From the current research, FGF and VEGF play an anti-EndMT role also by blocking TGF-β signaling pathway. These signal pathways crisscross and regulate the process of EndMT, which are effective targets for controlling EndMT imbalance. What is important is that the signal regulation network of EndMT and the apoptosis signal regulation network have an intersection. With reference to the relationship between EMT and apoptosis, we will discuss the feasibility of apoptosis in controlling EndMT imbalance. What’s more, we will also discuss the flexible application of autophagy strategies to inhibit EndMT.

2.2 Reasonable application of apoptosis

Apoptotic cells produce more substances which regulates EndMT, thus inhibit endothelial apoptosis helps controling EndMT. EMT is a mechanism that deceives cell death [Citation45], which is well interpreted in survival and metastasis of cancer cells. Similarly, EndMT becomes a buffer zone for endothelial cells to escape apoptosis. These indicate that there is a deep correlation between the regulation of EndMT and the regulation of apoptosis. Nicotinamide, the precursor of nicotinamide adenine dinucleotide (NAD), effectively inhibited TGF-β-1-induced corneal EndMT and decreased the levels of EndMT regulators snail and slug [Citation46]. Furthermore NAD+ precursors can effectively prevent the apoptosis of the corneal endothelium through reactivating AKT signaling [Citation47]. Vaccarin protects HUVECs from ox-LDL-induced EndMT and apoptosis by inhibiting ROS/p38MAPK signaling pathway [Citation48]. Naringin and vaccarin have similar endothelial protective effects, which is naringin modulated ox-LDL-induced apoptosis, EndMT, and inflammation in vitro, via the YAP pathway [Citation49]. Although researchers have regarded EndMT and apoptosis as important factors of endothelial damage, few people have studied the potential link between apoptosis and EndMT. It has been reported that vascular endothelial cell apoptosis is involved in various stages of atherosclerosis [Citation50]. It may be the continuous influence of these apoptotic factors that the dynamic balance of EndMT is broken and endothelial cells undergo EndMT to obtain an invasion ability, migrate into the middle layer of blood vessels and transdifferentiate into smooth muscle cells in the process of atherosclerosis [Citation51]. Compared with EndMT, the damage of endothelial cell apoptosis to blood vessels is greater, more rapid, and the consequences are more serious. In addition, the factors that induce EndMT are also partially the same as those that induce endothelial cell apoptosis, such as oxidative stress and low shear stress [Citation52]. TGF-β is thought to activate apoptosis [Citation53]. ECs apoptosis induced the production of TGF-β1 in both apoptotic and neighboring viable cells, Exacerbating EndMT imbalance, but it can be relieved by Blocking Smad3, PI3k/Akt, and MAPK/ERK signaling pathway [Citation54]. It shows that although endothelial cell apoptosis contributes to more endothelial cells undergoing EndMT, after breaking the downstream signal of TGF-β EndMT imbalance is relieved. Based on their research, it can be concluded that the ultimate goal of the stimulus is to cause the death of endothelial cells, EndMT delays this death process, and then the stimulus is removed, and there is no threat of death, EndMT is no longer activated.

When myocardial infarction occurs milk fat globule-epidermal growth factor 8-mediated myofibroblasts engulfment of apoptotic cells, myofibroblasts acquires antiinflammatory properties [Citation55]. Therefore, myofibroblasts controls the infarct area effectively, but myocardial fibrosis is relieved by promoting the apoptosis of myofibroblasts. CCN5 reverses cardiac fibrosis established in HF mouse models via inhibiting the trans differentiation of EndMT and fibroblasts into myofibroblasts, and in particular by increasing the apoptosis of myofibroblasts [Citation56]. Although the molecular mechanism is not fully understood, CCN5 exerts these effects at least in part by inhibiting the TGF-β signaling pathway [Citation56]. So reasonable application of apoptosis is also a Strategy to cope with EndMT.

2.3 Autophagy targeted therapy strategy

Targeted autophagy therapy, whether it is targeted at autophagy defects or enhanced autophagy, is very promising in regulating EndMT. With the continuous development of research on autophagy in cardiovascular disease, researchers combine autophagy-targeted therapeutic strategy with regulating EndMT [Citation57], although the precise role of Autophagy in EndMT has not been fully elucidated yet. The mechanism weakening EndMT by enhanced autophagy has been explored preliminarily. Myofibroblast-like cells (MFLCs) promote human pulmonary microvascular endothelial cells to undergo EndMT by inhibiting autophagy, moreover rapamycin reversed the phenotypic alterations and the gene expression changes in ECs co-cultured with MFLCs [Citation58]. Hypoxia triggered EndMT and autophagic flux increased in human cardiac microvascular endothelial cells (HCMECs) [Citation59]. In addition, improving the level of autophagy partially inhibited EndMT while blocking autophagy promoted EndMT [Citation59]. Autophagy attenuated EndMT induced by hypoxia via p62-dependent degradation of intracellular Snail (the key transcription factor in the downstream of TGF-β signaling pathway to trigger EndMT [Citation60]) and Twist (a key transcription factor that maintains the invasive mesenchymal phenotype [Citation59,Citation61,Citation62]. MiR-204 suppressed autophagy by targeting 3ʹUTR of ATG7 mRNA, down-regulation of miR-204 enhanced autophagy, thereby impacting p62-dependent degradation of Snail and Twist [Citation62]. When rat pulmonary artery intima and human pulmonary artery endothelial cells were hypoxic, miR-204 was down-regulated and its further down-regulation by using miR-204 inhibitor suppressed hypoxia-induced EndMT [Citation62]. The lack of autophagy exacerbates EndMT. Marchi S’s data point to the key role of autophagy defects which is highly correlated with EndMT in the pathogenesis of cerebral cavernous deformity [Citation63,Citation64]. According to reports, deletion of the essential autophagy gene ATG7 in endothelial cells leads to impaired autophagic energy flux of endothelial cells, and endothelial cells acquire a phenotype consistent with EndMT [Citation65]. Hammoutene A et al. found Hepatic endothelial cell autophagy defects lead to endothelial cell inflammation, EndMT, and endothelial cell apoptosis [Citation66]. It is interesting that autophagy defect-induced EndMT is dependent on IL6 in human microvascular endothelial cells and Il6-neutralizing antibody rescued metabolic defects and organ fibrosis in HFD-fed Atg5 Endo [Citation67].

However, some studies have shown that autophagy promotes EndMT. Liu C et al. suggested that hypoxia challenge activated the level of autophagic flux and resulting in obviously upregulated the expressions of collagen I and connective tissue growth factor (CTGF) at the protein level in systemic sclerosis fibroblasts, as well as the expressions of vimentin and α-SMA in HUVECs, while the expressions of VE-cadherin and CD31 were decreased compared with the normoxic treatment [Citation68]. What’s more, blockade of autophagic flux inhibited collagen synthesis and EndMT [Citation69]. Autophagy mediated by monocyte chemotactic protein-induced protein 1(MCPIP1) promoted the SiO2-induced loss of an endothelial phenotype by human umbilical vein endothelial cells [Citation70]. Autophagy induction of early endothelial progenitor cells can increase or decrease mesenchymal properties of renal endothelial cells [Citation71]. These illustrate the nature of the double-edged sword of autophagy to some extent. Actually controversies exist regarding the role of autophagy on EndMT [Citation72]. Autophagy is indeed involved in the regulation of EndMT homeostasis. When responding to environmental pressure, Autophagy and EndMT should work together to combat external stimuli. If autophagy can fully cope with it, EndMT becomes a burden and is inhibited, such as the degradation of related transcription factors mentioned above. But when autophagy cannot control the deterioration of the situation, EndMT cannot avoid the occurrence of which includes the secretion of several pro-inflammatory cytokines related to autophagy [Citation72] (especially the cytokines involved in promoting EndMT). When autophagy for EndMT regulation turns counterproductive, targeted autophagy inhibition is also an indispensable study.

Macroscopically speaking, autophagy is involved in regulating the balance of EndMT. When endothelial cells are induced to undergo phenotypic transformation, autophagy is activated to combat EndMT. However, the continuous activation of autophagy will become an independent stimulating factor for endothelial cells, which will have to initiate EndMT to combat the adverse consequences of autophagy such as apoptosis.

Figure. Endothelial cell autophagy and apoptosis have a deep relationship with EndMT. Endothelial cells can consume apoptosis “signals” through EndMT to avoid the worse effects caused by apoptosis. However, the intersection of EndMT and apoptosis is worth digging. From the intersection of autophagy and apoptosis, the dual effects of autophagy on EndMT can also be mapped. Autophagy can degrade the key transcription factors Snail and Twist that induce EndMT to inhibit EndMT. However, the complex relationship between autophagy and the secretion of pro-inflammatory cytokines, as well as some of the unknown complex regulatory networks of autophagy promote EndMT. Endothelial cell autophagy maintains EndMT homeostasis.

2.4 Promote mesenchymal–endothelial transition

Promoting mesenchymal-endothelial transformation (MEndT) is a strategy that inhibits EndMT that has not attracted attention or is easily overlooked. The plasticity of endothelial cells changes their phenotype from endothelial cells to mesenchymal cells, and vice versa, which are considered to be EndMT and MEndT [Citation73]. EndMT is a dynamic process, including endogenous expression of endothelial cells related proteins that maintain endothelial cell phenotype and acquired expression of mesenchymal cell-related proteins. In general, researchers ignore the self-sustaining ability of endothelial cells, that is, the glow of promoting MEndT is masked by inhibiting EndMT. The process of EndMT has not been divided into stages, and some researchers have proposed the concept of “Partial EndMT”[Citation74]. Corresponding to this is “Full EndMT”, which is the main reason for the dynamic imbalance of EndMT, because fully transformed endothelial cells are difficult to return to endothelial cells. EndMT is an instinctive activity of endothelial cells in response to stimulation. When the dynamic balance is broken, endothelial cells will become fibroblasts after undergoing EndMT and participate in the related progress of fibrotic diseases. MEndT refers to the transformation of stromal cells into endothelial cells, which is very similar to the reversal process of EndMT. This should arouse people’s attention. The most significant part of promoting MEndT should be to protect endothelial cells from the occurrence of “Full EndMT”. Certainly, if the fibroblasts transformed from endothelial cells can regain the endothelial phenotype and then restore the endothelial cell function, it is of great significance for fibrotic diseases.

At present, the research on MEndT has not introduced the inhibition of EndMT, but the connectivity between the two is worth exploring in depth. Cardiac fibroblasts are considered sentinel cells in the myocardium and actively respond to environmental stressors, including differentiation into a myofibroblast phenotype in response to ischemic and mechanical injury. Also, cardiac fibroblasts can undergo differentiation into endothelial-like cells. Studies have shown that cardiac fibroblasts undergo MEndT into neo-endothelial cells in damaged hearts, and have shown can be enhanced to promote heart repair, and the transcription factor p53 regulates such a switch in cardiac fibroblast fate [Citation75]. It is interesting that the MEndT plays an important role in increasing the number of coronary endothelial cells after myocardial infarction, but genetic lineage tracking shows that myocardial fibroblasts have expanded significantly after injury, but have no effect on new coronary vessels, indicating no contribution of MEndT to the formation of neovascularization, indicating that the mesenchymal-endothelial cells did not contribute any to the neovascularization [Citation76]. This seems to be contradictory, but the focus of our attention is not whether fibroblasts can be transformed into endothelial cells, but the protective effect of MEndT. The complete conversion of fibroblasts into endothelial cells does not play a decisive role in the repair of damage after myocardial infarction. It is still too early to increase the number of endothelial cells derived from fibroblasts. To some extent, MEndT can replace EndMT in response to environmental pressure and form a barrier to protect endothelial cells.

Perhaps the researchers’ expectations of MEndT are too high and the research is not deep enough. We all hope to make the “Full EndMT” completely reversed or the fibroblast “Full MEndT”, but this is difficult, and the goal can be to save the EndMT imbalance. Studies have suggested the hope of Full MEndT, which can reprogram adult fibroblasts into functional cardiomyocytes by combining rich lineage transcription factors [Citation77]. In addition, studies have reported that adult fibroblasts can be reprogrammed into functional cardiomyocytes by combining rich lineage transcription factors. Studies have suggested the hope of Full MEndT, which can reprogram adult fibroblasts into functional cardiomyocytes by combining rich lineage transcription factors. In fact, some studies have paid attention to the existence of the MEndT process in the dynamic balance of EndMT. In one study, hydrocortisone (HC) supplementation to the basal medium significantly enhances the barrier properties of human brain microvascular endothelial cells (HBMEC/ciβ), and these effects of HC on HBMEC/ciβ could be summarized as facilitating endothelial differentiation characteristics while concurrently retarding mesenchymal characteristics [Citation78]. Generally speaking, MEndT is considered to be an important process in tumorigenesis and embryonic development and in one study it is proposed for juvenile angiofibromas [Citation79]. In recent studies, Lin F et al. found that miR-133 promotes fibroblast angiogenesis and MEndT, thereby reducing myocardial fibrosis [Citation80], and the research by the Batlle R et al. shows that the kinase p38α via down-regulating TGF-β and JNK signals inhibits mesenchymal cell angiogenesis procedures, including the transition to an endothelial-like phenotype [Citation81]. There are few reports on the biological phenomenon of MEndT. It may be too early to use it as a strategy to control the imbalance of EndMT, but its role cannot be ignored.

3. Conclusions and future perspectives

EndMT is an effective target for the treatment of cardiovascular diseases. Endothelial cells adapt to the complex environment of the cardiovascular system through EndMT. Therefore, the signal transduction to promote EndMT is very active, and directly or indirectly blocking these signal pathways is an effective method to control EndMT imbalance. In addition, reasonable application of apoptosis and autophagy can also maintain the balance of EndMT. In fact, EndMT is an inevitable physiological activity. But EndMT imbalance will become the pathological basis of cardiovascular disease, and under the pathological conditions to exacerbate this imbalance to form a vicious circle. Therefore, it is necessary to fully describe the role of EndMT in the pathogenesis of various cardiovascular diseases. At the same time, it is necessary to deepen the research on the mechanism of regulating EndMT, so as to better control EndMT imbalance.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (9183910381, 670429, 81470435, to ZSJ), and “Double First-Class” project for innovative Group of Basic Medicine, University of South China (to ZSJ).

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This work was supported by the National Natural Science Foundation of China [670429]; National Natural Science Foundation of China [81470435]; National Natural Science Foundation of China [9183910381].

References

  • Evrard SM, Lecce L, Michelis KC, et al. Endothelial to mesenchymal transition is common in atherosclerotic lesions and is associated with plaque instability. Nat Commun. 2016;7(1):11853.
  • Xiao L, Dudley AC. Fine-tuning vascular fate during endothelial-mesenchymal transition. J Pathol. 2017;241(1):25–35.
  • Von Gise A, Pu WT. Endocardial and epicardial epithelial to mesenchymal transitions in heart development and disease. Circ Res. 2012;110(12):1628–1645.
  • Xiong J, Kawagishi H, Yan Y, et al. A metabolic basis for endothelial-to-mesenchymal transition. Mol Cell. 2018;69(4):e7.
  • Isabella S, Mona P, Jacqueline H, et al. Endothelial mesenchymal transition in hypoxic microvascular endothelial cells and paracrine induction of cardiomyocyte apoptosis are mediated via TGFβ1/SMAD Signaling. International Journal of Molecular Ences. 2017;18:307–316.
  • Chen XY, Lv RJ, Zhang W, et al. Inhibition of myocyte-specific enhancer factor 2A improved diabetic cardiac fibrosis partially by regulating endothelial-to-mesenchymal transition. Oncotarget. 2016;7(21):31053–31066.
  • Bochenek ML, Leidinger C, Rosinus NS, et al. Activated Endothelial TGFβ1 signaling promotes venous thrombus nonresolution in mice via Endothelin-1: potential Role for Chronic Thromboembolic Pulmonary Hypertension. Circ Res. 2020;126(2):162–181.
  • Ya-Meng H, Hou-Qin Y, Zhong R, et al. Endothelial to mesenchymal transition in atherosclerotic vascular remodeling. Clinica Chimica Acta International Journal of Clinical Chemistry. 2018;490:34-38.
  • Kouzbari K, Hossan MR, Arrizabalaga JH, et al. Oscillatory shear potentiates latent TGF-beta1 activation more than steady shear as demonstrated by a novel force generator. Sci Rep. 2019;9(1):6065.
  • Mahmoud MM, Serbanovic-Canic J, Feng S, et al. Shear stress induces endothelial-to-mesenchymal transition via the transcription factor Snail. Sci Rep. 2017;7(1):3375.
  • Ajami NE, Gupta S, Maurya MR, et al. Systems biology analysis of longitudinal functional response of endothelial cells to shear stress. Proc Natl Acad Sci U S A. 2017;114(41):10990.
  • Moonen JR, Lee ES, Schmidt M, et al. Endothelial-to-mesenchymal transition contributes to fibro-proliferative vascular disease and is modulated by fluid shear stress. Cardiovasc Res. 2015;108(3):377–386.
  • Lai B, Li Z, He M, et al. Atheroprone flow enhances the endothelial-to-mesenchymal transition. Am J Physiol Heart Circ Physiol. 2018;315(5):H1293–h303.
  • Burtenshaw D, Kitching M, Redmond EM, et al. Reactive Oxygen Species (ROS), Intimal Thickening, and Subclinical Atherosclerotic Disease. 2019;6:89.
  • Jiyuan C, Andrew P, L C P, et al. Loss of Smooth Muscle α-Actin Leads to NF-κB-Dependent Increased Sensitivity to Angiotensin II in Smooth Muscle Cells and Aortic Enlargement. Circ Res. 2017;120(12):1903-1915.
  • Yao Y, Jumabay M, Ly A, et al. A role for the endothelium in vascular calcification. Circ Res. 2013;113(5):495–504.
  • Yao J, Guihard PJ, Blazquez-Medela AM, et al. Serine protease activation essential for endothelial-mesenchymal transition in vascular calcification. Circ Res. 2015;117(9):758–769.
  • Medici D, Shore EM, Lounev VY, et al. Conversion of vascular endothelial cells into multipotent stem-like cells. Nat Med. 2010;16(12):1400–1406.
  • Shohreh M, Sanela K, Valentina P, et al. Mesenchymal state of intimal cells may explain higher propensity to ascending aortic aneurysm in bicuspid aortic valves. Sci Rep. 2016;6:35712.
  • Maleki S, Poujade FA, Bergman O, et al. Endothelial/ epithelial mesenchymal transition in ascending aortas of patients with bicuspid aortic valve. Front Cardiovasc Med. 2019;6:182.
  • Nicolini G, Forini F, Kusmic C, et al. Angiopoietin 2 signal complexity in cardiovascular disease and cancer. Life ences. 2019;239:117080.
  • Vingolo EM, Fragiotta S, Mafrici M, et al. Vitreous and plasma changes of endothelin-1, adrenomedullin and vascular endothelium growth factor in patients with proliferative diabetic retinopathy. European Review for Medical & Pharmacological Sciences. 2017;21:662.
  • Feng L, Zhang C, Liu G, et al. RKIP negatively regulates the glucose induced angiogenesis and endothelial-mesenchymal transition in retinal endothelial cells. Exp Eye Res. 2019;189:107851.
  • Goumans M-J, ten Dijke P. TGF-β Signaling in Control of Cardiovascular Function. Cold Spring Harb Perspect Biol. 2017;10(2):a022210.
  • Yongliang J, Xuanfen Z, Ruicheng H, et al. β1-induced SMAD2/3/4 activation promotes RELM-β transcription to modulate the endothelium-mesenchymal transition in human endothelial cells. Int J Biochem Biotechnol. 2018;105:52-60.
  • Volkmann I, Kumarswamy R, Pfaff N, et al. MicroRNA-mediated epigenetic silencing of sirtuin1 contributes to impaired angiogenic responses. Circ Res. 2013;113(8):997–1003.
  • Li Z, Wang F, Zha S, et al. SIRT1 inhibits TGF‐β‐induced endothelial‐mesenchymal transition in human endothelial cells with Smad4 deacetylation. J Cell Physiol. 2018;233(11):9007-9014.
  • Liu ZH, Zhang Y, Wang X, et al. SIRT1 activation attenuates cardiac fibrosis by endothelial-to-mesenchymal transition. Biomed Pharmacother. 2019;118:109227.
  • Lin C-H, Lin -C-C, Ting W-J, et al. Resveratrol enhanced FOXO3 phosphorylation via synergetic activation of SIRT1 and PI3K/Akt signaling to improve the effects of exercise in elderly rat hearts. Age (Omaha). 2014;36(5):9705.
  • Pawel Z, Katrin P-Z, Jingang H, et al. Sirt1 regulates canonical TGF-β signalling to control fibroblast activation and tissue fibrosis. Ann Rheum Dis. 2016.75(1):226-33.
  • Wang Z, Han Z, Tao J, et al. Role of endothelial-to-mesenchymal transition induced by TGF-β1 in transplant kidney interstitial fibrosis. J Cell Mol Med. 2017;21(10):2359–2369.
  • Wang Z, Fei S, Suo C, et al. Antifibrotic Effects of Hepatocyte Growth Factor on Endothelial-to-Mesenchymal Transition via Transforming Growth Factor-Beta1 (TGF-β1)/Smad and Akt/mTOR/P70S6K Signaling Pathways. Ann Transplant. 2018;23:1–10.
  • Przemyslaw B, Bjrn M-E, Tomas V, et al. Transforming growth factor-β-dependent Wnt secretion controls myofibroblast formation and myocardial fibrosis progression in experimental autoimmune myocarditis. Eur Heart J. 2017;38(18):1413–1425.
  • Choi SH, Kim H, Lee HG, et al. Dickkopf-1 induces angiogenesis via VEGF receptor 2 regulation independent of the Wnt signaling pathway. Oncotarget. 2017;8(35):58974-58984.
  • Cheng S-L, Shao J-S, Behrmann A, et al. Dkk1 and MSX2-Wnt7b signaling reciprocally regulate the endothelial-mesenchymal transition in aortic endothelial cells. Arteriosclerosis Thrombosis & Vascular Biology. 2013;33(7):1679–1689.
  • Mark L, Hassan D, Li JN, et al. Dickkopf-3 in aberrant endothelial secretome triggers renal fibroblast activation and endothelial-mesenchymal transition. Nephrol Dialysis Transplantation. 2018;34(1):49–62.
  • Zhou X, Chen X, Cai JJ, et al. Relaxin inhibits cardiac fibrosis and endothelial-mesenchymal transition via the Notch pathway. Drug Design Development & Therapy. 2015;9:4599-4611.
  • Lin QQ, Zhao J, Zheng CG, et al. Roles of notch signaling pathway and endothelial-mesenchymal transition in vascular endothelial dysfunction and atherosclerosis. Eur Rev Med Pharmacol Sci. 2018;22:6485–6491.
  • Zhang J, Zeng Y, Chen J, et al. miR-29a/b cluster suppresses high glucose-induced endothelial-mesenchymal transition in human retinal microvascular endothelial cells by targeting Notch2. Exp Ther Med. 2019;17:3108–3116.
  • Liu W, Wu Y, Yu F, et al. The implication of Numb-induced Notch signaling in endothelial-mesenchymal transition of diabetic nephropathy. J Diabetes Complications. 2018;32(10):889–899.
  • Zhou H, Chen X, Chen L, et al. Anti-fibrosis effect of scutellarin via inhibition of endothelial–mesenchymal transition on isoprenaline-induced myocardial fibrosis in rats. Molecules. 2014;19(10):15611–15623.
  • Chen PY, Qin L, Barnes C, et al. FGF Regulates TGF-β Signaling and Endothelial-to-Mesenchymal Transition via Control of let-7 miRNA Expression. Cell Rep. 2012;2(6):1684-1696.
  • Ben M-W, Illigens A, Casar B, et al. Vascular Endothelial Growth Factor Prevents Endothelial-to-Mesenchymal Transition in?Hypertrophy. Ann Thorac Surg. 2017;104(3):932-939.
  • Shi S, Srivastava SP, Kanasaki M, et al. Interactions of DPP-4 and integrin β1 influences endothelial-to-mesenchymal transition. Kidney Int. 2015;88(3):479–489.
  • Chakraborty S, Mir KB, Seligson ND, et al. Integration of EMT and cellular survival instincts in reprogramming of programmed cell death to anastasis. Cancer Metastasis Rev. 2020;39(2):553–566.
  • Zongyi L, Haoyun D, Wenjing Y, et al. Nicotinamide inhibits corneal endothelial mesenchymal transition and accelerates wound healing. Experimental eye research. 2019;184:227-233
  • Zhao C, Li W, Duan H, et al. NAD+ precursors protect corneal endothelial cells from UVB-induced apoptosis. Am J Physiol Cell Physiol. 2020;318(4):C796–c805.
  • Gong L, Lei Y, Liu Y, et al. Vaccarin prevents ox-LDL-induced HUVEC EndMT, inflammation and apoptosis by suppressing ROS/p38 MAPK signaling. Am J Transl Res. 2019;11:2140–2154.
  • Zhao H, Liu M, Liu H, et al. Naringin protects endothelial cells from apoptosis and inflammation by regulating the Hippo-YAP Pathway. Biosci Rep. 2020;40(3):BSR20193431.
  • Zhang CC, Yan Z, Zong Q, et al. Synergistic Effect of the γ-Secretase Inhibitor PF-03084014 and Docetaxel in Breast Cancer Models. Stem Cells Transl Med. 2013;2(3):233–242.
  • Souilhol C, Harmsen MC, Evans PC, et al. Endothelial-mesenchymal transition in atherosclerosis. Cardiovasc Res. 2018;114(4):565–577.
  • Mallat Z, Tedgui A. Apoptosis in the vasculature: mechanisms and functional importance. Br J Pharmacol. 2000;130(5):947-962.
  • Song J. EMT or apoptosis: a decision for TGF-beta. Cell Res. 2007;17(4):289–290.
  • Li J, Xiong J, Yang B, et al. Endothelial Cell Apoptosis Induces TGF-β Signaling-Dependent Host Endothelial-Mesenchymal Transition to Promote Transplant Arteriosclerosis. Am J Transplant. 2015;15(12):3095–3111.
  • Nakaya M, Watari K, Tajima M, et al. Cardiac myofibroblast engulfment of dead cells facilitates recovery after myocardial infarction. J Clin Investig. 2017;127(1):383.
  • Jeong D, Lee M, Li A, et al. Matricellular Protein CCN5 Reverses Established Cardiac Fibrosis. JOURNAL- AMERICAN COLLEGE OF CARDIOLOGY. 2016;67(13):1556-1568
  • Jin B, Shi H, Zhu J, et al. Up-regulating autophagy by targeting the mTOR-4EBP1 pathway: a possible mechanism for improving cardiac function in mice with experimental dilated cardiomyopathy. BMC Cardiovasc Disord. 2020;20(1):56.
  • Sakao S, Hao H, Tanabe N, et al. Endothelial-like cells in chronic thromboembolic pulmonary hypertension: crosstalk with myofibroblast-like cells. Respir Res. 2011;12(1):109.
  • Zou J, Liu Y, Li B, et al. Autophagy attenuates endothelial-to-mesenchymal transition by promoting Snail degradation in human cardiac microvascular endothelial cells. Biosci Rep. 2017;37(5):BSR20171049.
  • Pinto MT, Ferreira Melo FU, Malta TM, et al. Endothelial cells from different anatomical origin have distinct responses during SNAIL/TGF-β2-mediated endothelial-mesenchymal transition. Am J Transl Res. 2018;10:4065–4081.
  • Song S, Liu L, Yu Y, et al. Inhibition of BRD4 attenuates transverse aortic constriction- and TGF-β-induced endothelial-mesenchymal transition and cardiac fibrosis. J Mol Cell Cardiol. 2019;127:83–96.
  • Liu T, Zou XZ, Huang N, et al. Down-regulation of miR-204 attenuates endothelial-mesenchymal transition by enhancing autophagy in hypoxia-induced pulmonary hypertension. Eur J Pharmacol. 2019;863:172673.
  • Marchi S, Trapani E, Corricelli M, et al. Beyond multiple mechanisms and a unique drug: defective autophagy as pivotal player in cerebral cavernous malformation pathogenesis and implications for targeted therapies. Rare Dis (Austin, Tex). 2016;4(1):e1142640.
  • Marchi S, Corricelli M, Trapani E, et al. Defective autophagy is a key feature of cerebral cavernous malformations. EMBO Mol Med. 2015;7(11):1403–1417.
  • Singh KK, Lovren F, Pan Y, et al. The essential autophagy gene ATG7 modulates organ fibrosis via regulation of endothelial-to-mesenchymal transitionBSR20171049. J Biol Chem. 2015;290(5):2547-2559.
  • Hammoutene A, Biquard L, Lasselin J, et al. A defect in endothelial autophagy occurs in patients with non-alcoholic steatohepatitis and promotes inflammation and fibrosis. J Hepatol. 2020;72:528–538.
  • Takagaki Y, Lee SM, Dongqing Z, et al. Endothelial autophagy deficiency induces IL6 - dependent endothelial mesenchymal transition and organ fibrosis. Autophagy. 2020;16(10):1905-1914.
  • Chaofan L, Xing Z, Jinghao L, et al. Autophagy mediates 2-methoxyestradiol-inhibited scleroderma collagen synthesis and endothelial-to-mesenchymal transition induced by hypoxia. Rheumatology (Oxford). 2019;58(11):1966-1975.
  • Chao J, Wang X, Zhang Y, et al. Role of MCPIP1 in the Endothelial-mesenchymal transition induced by silica. Cellular Physiology &Biochemistry. 2016;40(1–2):309–325.
  • Patschan D, Schwarze K, Henze E, et al. Endothelial autophagy and Endothelial-to-Mesenchymal Transition (EndoMT) in eEPC treatment of ischemic AKI. J Nephrol. 2016;29(5):637–644.
  • Garg M. Epithelial plasticity, autophagy and metastasis: potential modifiers of the crosstalk to overcome therapeutic resistance. Stem Cell Rev Rep. 2020;16(3):503–510.
  • Rojas-Sanchez G, Cotzomi-Ortega I, Pazos-Salazar NG, et al. Autophagy and Its Relationship to Epithelial to Mesenchymal Transition: when Autophagy Inhibition for Cancer Therapy Turns Counterproductive. Biology (Basel). 2019;8(4):1–20.
  • Dejana E, Hirschi KK, Simons M. The molecular basis of endothelial cell plasticity. Nat Commun. 2017;8(1):14361.
  • Welch-Reardon KM, Wu N, Hughes CCW. A Role for Partial Endothelial-Mesenchymal Transitions in Angiogenesis? Arteriosclerosis Thrombosis & Vascular Biology. 2014;35(2):303-308.
  • Ubil E, Duan J, Pillai IC, et al. Mesenchymal-endothelial transition contributes to cardiac neovascularization. Nature. 2014;514(7524):585–590.
  • He L, Huang X, Kanisicak O, et al. Preexisting endothelial cells mediate cardiac neovascularization after injury. J Clin Investig. 2017;127(8):2968.
  • Song K, Nam YJ, Luo X, et al. Heart repair by reprogramming non-myocytes with cardiac transcription factors. Nature. 2012;485(7400):599–604.
  • Furihata T, Kawamatsu S, Ito R, et al. Hydrocortisone enhances the barrier properties of HBMEC/cibeta, a brain microvascular endothelial cell line, through mesenchymal-to-endothelial transition-like effects. Fluids Barriers CNS. 2015;12(1):7.
  • Kulas P, Willnecker V, Dlugaiczyk J, et al. Mesenchymal-endothelial transition in juvenile angiofibroma? Acta Otolaryngol. 2015;135(9):955–961.
  • Lin F, Zeng Z, Song Y, et al. YBX-1 mediated sorting of miR-133 into hypoxia/reoxygenation-induced EPC-derived exosomes to increase fibroblast angiogenesis and MEndoT. Stem Cell Res Ther. 2019;10(1):263.
  • Batlle R, Andrés E, Gonzalez L, et al. Regulation of tumor angiogenesis and mesenchymal-endothelial transition by p38α through TGF-β and JNK signaling. Nat Commun. 2019;10(1):3071.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.