Next Article in Journal
Molecular Response of Simmental Cows to Negative Energy Balance: Regulation of Interleukin-6 and Plasminogen During Early Lactation
Previous Article in Journal
The Role of Osteoblasts in Phenotypic Variability of Dominant Osteogenesis Imperfecta: Evidence from Patients and Murine Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endothelial-to-Mesenchymal Transition in Health and Disease: Molecular Insights and Therapeutic Implications

1
Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea
2
Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50611, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(23), 11724; https://doi.org/10.3390/ijms262311724
Submission received: 19 October 2025 / Revised: 30 November 2025 / Accepted: 1 December 2025 / Published: 3 December 2025

Abstract

Endothelial-to-mesenchymal transition (EndMT) is a cellular program implicated in fibrosis, vascular remodeling, and the tumor microenvironment across multiple organs. We synthesize mechanistic pathways including TGF-β/SMAD, non-canonical (MAPK, PI3K/AKT, Rho/ROCK), Notch, and Wnt/β-catenin cascades. Their crosstalk with hypoxia, inflammatory cues, and epigenetic mechanisms can drive loss of endothelial identity and acquisition of mesenchymal characteristics. We outline disease contexts in the heart, lungs, kidneys, liver, central nervous system, and cancer, highlighting context-dependent contributory roles of EndMT. Therapeutically, we review pathway-targeted agents, epigenetic inhibitors, microRNA-based strategies, antibodies/biologics, small molecules and natural compounds, and cell- and gene-based interventions. Finally, we outline a translational roadmap that pairs patient-derived iPSC/organoid and organ-on-a-chip platforms to stratify EndMT states and prioritize targets. We also explore combination regimens that integrate multi-pathway modulation with epigenetic and immune approaches, aiming to deliver clinically meaningful anti-fibrotic benefits while better preserving physiological signaling.

1. Introduction

Endothelial cells (ECs) play a central role in the maintenance of vascular integrity and tissue homeostasis by sensing and integrating mechanical and biochemical stimuli [1]. They respond to shear stress, inflammatory cytokines, and oxidative stress, which affect the structural and functional integrity of the endothelial barrier and leukocyte–EC interactions [2,3,4,5]. ECs constitute a monolayer lining the luminal surface of blood vessels, serving as a dynamic barrier that regulates vascular tone, permeability, and interactions between blood components and surrounding tissues [6,7]. Disruption of endothelial homeostasis under pathophysiological conditions leads to endothelial dysfunction and endothelial-to-mesenchymal transition (EndMT) [8]. This process is implicated in vascular pathologies and is characterized by the loss of endothelial identity and the acquisition of mesenchymal traits such as cytoskeletal remodeling, cellular motility, and contractility [9]. Over the past decades, in vitro and in vivo studies have implicated endothelial phenotypic transition in pathological remodeling across multiple organs, including the cardiovascular system, lungs, kidneys, liver, and central nervous system [8,10,11,12,13,14]. Accumulating evidence indicates that EndMT contributes to disease processes such as cardiac fibrosis, pulmonary vascular remodeling/pulmonary arterial hypertension (PAH), kidney and liver fibrosis, cerebral cavernous malformations (CCMs), and context-dependent blood–brain barrier (BBB) dysfunction. Mapping of key EndMT pathways, including transforming growth factor-β (TGF-β)/SMAD, Notch, Wnt/β-catenin, and YAP/TAZ, has identified potential therapeutic targets, supported by preclinical and early-phase clinical studies [8,15,16,17]. Collectively, pathway mapping and target identification support the development of EndMT-targeted therapeutics for human diseases involving cellular transdifferentiation.
To enhance transparency and enable readers to assess the scope and potential sources of bias, we summarize below the literature search strategy and selection criteria used in this narrative review.

Scope and Literature Selection

This review provides a narrative synthesis of EndMT in fibrosis, vascular remodeling, and the tumor microenvironment. We prioritized peer-reviewed primary studies and authoritative reviews searched in PubMed, Web of Science, and Scopus (databases last searched on 27 November 2025), using combinations of keywords including “endothelial-to-mesenchymal transition”, “EndMT”, “EndoMT”, “partial EndMT”, “fibrosis”, “vascular remodeling”, “barrier dysfunction”, “spatial transcriptomics”, “organoid”, and “organ-on-chip”. Additional relevant articles were identified through citation tracking of key papers. Inclusion criteria were as follows: (i) studies explicitly addressing EndMT (or endothelial mesenchymal activation/partial EndMT) in vascular or organ-specific endothelium; and/or (ii) studies providing mechanistic, methodological, or therapeutic modulation evidence relevant to EndMT. Evidence was categorized by experimental context (in vitro, in vivo, ex vivo, and human tissues/datasets). Exclusion criteria included articles not relevant to EndMT, studies lacking EndMT-related endpoints, and non-research items. Where cited, preprints were used sparingly to highlight emerging technologies and explicitly labeled as such.

2. Molecular Mechanisms and Pathophysiological Roles of EndMT

EndMT is orchestrated by various signaling pathways, including TGF-β/SMAD, Notch, Wnt/β-catenin, PI3K/AKT cascades, as well as inflammatory cytokines. It is governed by transcriptional networks comprising SNAI1, SNAI2, ZEB1/2, and TWIST1 [8,9,15,18,19,20,21]. Noncoding RNAs and epigenetic regulators also modulate the process [22,23,24]. Phenotypically, EndMT is commonly characterized by loss of endothelial markers (CD31, vWF, VE-cadherin) and gain of mesenchymal markers (α-SMA, fibronectin, vimentin) across developmental and pathological conditions [9,22,23]. EndMT is a subtype of epithelial-to-mesenchymal transition (EMT) that shares key regulatory pathways and transcriptional effectors [8,25]. It is essential for embryonic heart development and contributes to various pathological processes in postnatal tissues, including fibrosis, inflammation, and vascular remodeling [26,27]. EndMT is regulated by extrinsic cues (TGF-β, inflammatory cytokines), hypoxia, and oxidative stress [9,27]. The intrinsic transcriptional network also drives phenotypic transitions that are further shaped by epigenetic mechanisms, including DNA methylation, histone modifications, and microRNAs (miRs) [9,22,23,24] (Table 1).

2.1. Transforming Growth Factor-β

TGF-β signaling drives a wide range of organ-specific pathological processes across the lungs, kidneys, heart, and tumors [18]. It stimulates fibroblast-to-myofibroblast differentiation, extracellular matrix (ECM) deposition, pulmonary fibrosis and remodeling [28]. TGF-β induces tubulointerstitial and glomerular fibrosis through SMAD-dependent pathways in the kidneys [57]. TGF-β also facilitates post-infarcted and hypertrophic remodeling in myocardium and activates cancer-associated fibroblasts, which remodel the ECM and promote cancer progression and metastasis in the tumor microenvironment [58,59]. In ECs, TGF-β binds TGF-β receptor (TβR) II and activates TβRI to initiate the canonical SMAD2/3/4 signaling and non-canonical cascades [15,29]. In the canonical pathway, phosphorylated SMAD2/3 forms complexes with SMAD4 and translocates to the nucleus, where it induces transcriptional programs involving EndMT-associated transcription factors such as SNAI1, SNAI2, and TWIST [15,29]. In non-canonical signaling, p38 MAPK/JNK, PI3K/AKT, Rho GTPases, and NF-κB are engaged in cytoskeletal reorganization, cell motility and survival, and intersect with SMAD signaling to help coordinate the transcriptional reprogramming associated with EndMT initiation and progression [15,60].

2.2. Wnt/β-Catenin Signaling in EndMT

Canonical Wnt/β-catenin signaling has been implicated in EndMT, particularly in fibrotic and inflammatory contexts [25,61,62]. In several EndMT models, β-catenin-dependent transcription has been shown to attenuate endothelial gene programs and promote mesenchymal features, often accompanied by barrier dysfunction and profibrotic remodeling [51,63,64]. Mechanistically, Wnt ligands engage Frizzled and LRP5/6 co-receptors to stabilize β-catenin, allowing its nuclear translocation and transcriptional activation with TCF/LEF [65]. Genetic evidence also supports a requirement for endothelial β-catenin during developmental EndMT in cardiac cushion formation [66]. In the disease context, aberrant Wnt activation has been associated with EndMT-like programs in the infarcted heart and diabetic kidney disease [63,64,67]. Importantly, functional coupling between TGF-β and Wnt/β-catenin signaling has been reported in EndMT, indicating that their cross-talk may modulate the magnitude and persistence of EndMT responses [25,62].

2.3. Transcriptional Regulation of EndMT

Downstream transcription factors drive EndMT by repressing endothelial genes and simultaneously activating mesenchymal programs, whereas coupling to Rho GTPase family–dependent cytoskeletal remodeling enhances cell motility [9,60]. TGF-β signaling can upregulate MMP-2 and MMP-9, degrading the basement membrane/ECM and reinforcing EndMT progression in fibrosis [68,69]. Over the past decade, numerous studies have identified SNAI2, TWIST1 and ZEB1/2 as key transcription factors implicated in EndMT [9,25]. SNAI2 is essential for endocardial cushion morphogenesis during cardiac development, and implicated in repressing endothelial markers and promoting mesenchymal traits in disease contexts [19]. TWIST1 can be modulated in a context-dependent manner by developmental and mechanotransductive cues (Notch-dependent regulation, flow-responsive programs), and promotes EC motility and cytoskeletal remodeling in atherosclerosis [12,20,25,70]. ZEB transcription factors act as key drivers of EndMT by suppressing endothelial identity and activating mesenchymal programs [25]. The Notch pathway can modulate EndMT via Notch–SNAI2 in cardiac cushion morphogenesis or MMP-9-Notch signaling in kidney transdifferentiation [19,68]. Collectively, these mechanisms drive EndMT under pathological contexts.

2.4. Epigenetics

Epigenetics refers to the heritable changes in gene expression without alterations in the DNA sequence, primarily via DNA methylation, histone modifications, and noncoding RNA-mediated regulation [71]. These mechanisms have been implicated in modulating transcription factor networks that influence EndMT in developmental contexts (heart valve/endocardial cushion formation) and in pathological states such as cardiac fibrosis, diabetic cardiomyopathy, and vascular remodeling [8,15,47,72].

2.4.1. DNA Methylation

DNA methylation is a key epigenetic mechanism regulating EndMT [24]. Under specific pathological cues, notably disturbed flow and inorganic phosphate, DNA methyltransferase 1 (DNMT1)-dependent promoter hypermethylation and gene silencing have been reported in ECs undergoing mesenchymal activation [24,36,43]. These changes can diminish endothelial identity programs, such as KLF4 locus methylation, and are accompanied by epigenetic repression, favoring EndMT-associated transcriptional states [36,37,73]. Pharmacologic DNMT inhibition or genetic targeting of DNMT1 mitigates EndMT features in cellular and vascular models [24,43].

2.4.2. Histone Modification

The histone tails of the core octamers (H2A, H2B, H3, and H4) are the principal substrates for epigenetic modifications that control chromatin accessibility and gene expression in ECs [74]. Acetylation and methylation dynamically tune EndMT-related transcriptional programs [23]. Class I/II histone deacetylase (HDAC) activity, particularly HDAC9 and HDAC3, can promote EndMT and adverse vascular remodeling, whereas HDAC9 loss-of-function or pharmacologic HDAC3 inhibition attenuates EndMT in preclinical models [22,44,75]. The histone demethylase JMJD2B represses H3K9me3 at EndMT-associated promoters (SULF1 and CNN1) to enhance their expression [23]. In contrast, EZH2-mediated H3K27me3 has been linked to silencing of endothelial-maintaining or anti-fibrotic programs and facilitating EndMT and fibrogenic responses [45,46]. Together, pharmacological targeting of these enzymes can partially reverse mesenchymal reprogramming, suggesting that histone modification is a therapeutic candidate for EndMT-associated diseases.

2.4.3. MicroRNA

miRs are short noncoding RNAs that repress gene expression post-transcriptionally by base-pairing with target mRNAs that bidirectionally control EndMT promotion or inhibition [46]. EndMT-relevant miRs can modulate TGF-β/SMAD signaling at multiple nodes, and in certain contexts, they also influence related pathways such as Wnt/β-catenin, biasing ECs toward either endothelial maintenance or mesenchymal activation programs [49,76,77,78,79,80]. MiR-20a targets TβRII and SARA to dampen canonical TGF-β signaling and inhibit EndMT [76]. Members of the miR-200 family, especially miR-200b, help preserve endothelial identity by blocking ZEB1/2 and EndMT-promoting pathways [48]. In contrast, miR-21, miR-27 and miR-155 can facilitate TGF-β-driven EndMT and vascular remodeling [49,77,78]. Notably, emerging evidence suggests context-dependent pro-EndMT roles for specific miRs, including miR-200c-3p [81,82]. In addition, miR-632 has been linked to EndMT activation and fibrosis in Marfan syndrome aortopathy [83].
Collectively, epigenetic and post-transcriptional mechanisms intersect with transcriptional programs to coordinate EndMT progression, and targeting these integrated networks offers a promising strategy for preventing or treating EndMT-mediated pathologies [22,23,24,72].

2.5. Plasticity and Reversibility of EndMT

EndMT often manifests as a spectrum, including partial or intermediate states, and its apparent reversibility is highly context-dependent [15]. In vitro, induction of withdrawal paradigms and reinforcement of pro-endothelial pathways (KLF2/4, BMP7, and NO signaling) can partially restore endothelial markers and reduce mesenchymal features [84,85,86]. In vivo, endothelial lineage-tracing and time-resolved disease models support an endothelial contribution to mesenchymal-like populations [87]. However, evidence for sustained reversibility remains model- and readout-dependent and can be confounded by Cre-driver specificity/efficiency and marker overlap across stromal lineages [88,89,90]. Ex vivo/human tissues and single-cell/spatial datasets largely provide cross-sectional snapshots of mixed endothelial/mesenchymal programs and inferred trajectories rather than direct state transitions, underscoring the need for longitudinal sampling and lineage-aware, orthogonal validation when feasible [91,92,93].
Taken together, these signaling and epigenetic mechanisms provided a shared mechanistic framework that is deployed in an organ- and disease context-dependent manner [8,9,15]. In the following section, we discuss how canonical TGF-β/SMAD and non-canonical MAPK and PI3K/AKT cascades, Notch and Wnt/β-catenin signaling, hypoxia/HIF-driven responses, and epigenetic modifiers such as DNMTs, HDACs, and histone methyltransferases converge to drive EndMT in specific organ systems [15,24,25,29,60]. We also highlight how differences in hemodynamic load, local inflammatory milieu, and metabolic stress bias the contribution of these pathways to EndMT in the cardiovascular system, lungs, kidneys, liver, central nervous system, and tumor microenvironment [2,7,10,11,12,13,69] (Figure 1).

3. Organ System Pathologies Associated with EndMT

EndMT, which is recognized as a developmental program during endocardial cushion formation and valvulogenesis, has emerged as a disease-related mechanism in adult organs [8]. It contributes to atherosclerosis and vascular remodeling of the cardiovascular system, pulmonary hypertension, lung remodeling, and renal fibrosis via EndMT programs [8,9]. It can also contribute to tumor progression, partly through endothelial-derived carcinoma-associated fibroblasts, CCMs in the nervous system and hepatic pathology [9,94,95]. Collectively, these studies support EndMT as a mechanistic contributor to fibrogenesis, chronic inflammation, and tissue remodeling in multiple organ systems.

3.1. Cardiovascular System

EndMT is essential during embryogenesis and is increasingly recognized as a contributor to adult cardiovascular diseases, including cardiac fibrosis, valvular disorders, and atherosclerosis [8,96,97]. ECs undergo EndMT and generate fibroblast-like or myofibroblast populations that secrete the ECM, increasing collagen deposition and ventricular stiffening [8,30,98]. Lineage-tracing and animal studies indicate that a subset of cardiac fibroblasts derives from ECs via EndMT [30,87,98]. Moreover, the experimental suppression of EndMT mitigates interstitial and perivascular fibrosis after MI, linking EndMT to adverse ventricular remodeling and diastolic dysfunction [8,50,99].
Valvular ECs also undergo EndMT in calcific aortic valve disease, which contributes to fibrotic and calcific remodeling of stenotic valves [31,96]. Human calcified aortic valve and ApoE−/− mouse valves have reported cell populations co-expressing endothelial and mesenchymal markers, consistent with in situ EndMT [31]. Pro-calcific or pro-fibrotic cues, including TGF-β, BMP, and Notch, modulated by inflammatory and biomechanical stress, can induce EndMT in valvular ECs, supporting EndMT as a mechanistic contributor and potential therapeutic target in valvular remodeling [32,100,101].
In atherosclerosis, ECs located in regions exposed to disturbed flow exhibit EndMT-like programs, losing their endothelial identity and acquiring mesenchymal traits [43]. This transition is associated with increased ECM production and pro-inflammatory signaling within plaques, which contribute to the feature of plaque complexity and vulnerability [22,102]. Collectively, these findings underscore EndMT as a pathogenic contributor to vascular remodeling and highlight it as a potential therapeutic target in atherosclerotic disease.
Mechanistically, cardiovascular EndMT is driven predominantly by TGF-β/SMAD signaling and its non-canonical branches (p38/JNK, PI3K/AKT, and RhoA/ROCK), which are activated by pressure overload, neurohumoral activation, and inflammatory cytokines [15,103,104,105]. Disturbed flow at an atheroprone site further engages Notch signaling, TWIST1, and DNMT1-dependent epigenetic reprogramming, while hypoxia and oxidative stress stabilize mesenchymal gene programs [36,37,40,43]. These coordinated inputs link hemodynamic and inflammatory stress to endothelial plasticity, fibroblast accumulation, and adverse cardiac and vascular remodeling [7,8,30].

3.2. Lung

In idiopathic pulmonary fibrosis (IPF), lung ECs exposed to chronic injury and pro-fibrotic stimuli (TGF-β, hypoxia) have been reported to undergo EndMT, contributing to the emergence of fibroblast-like or myofibroblast populations, excessive ECM deposition and interstitial scarring that disrupts the alveolar-capillary interface and impairs gas exchange [106,107,108]. In PAH, pulmonary ECs subjected to disturbed shear and inflammatory cytokines exhibit EndMT features, promoting intimal thickening and vascular remodeling that elevate pulmonary vascular resistance and pressures, with downstream right ventricular strain [10,109]. TGF-β/SMAD, BMP and PDGF cascades intersect with biomechanical stress to influence EndMT, highlighting these pathways as potential therapeutic targets under investigation in chronic lung disease [10].
In these pulmonary settings, EndMT is orchestrated mainly by TGF-β/SMAD and BMP signaling in conjunction with hypoxia/HIF-1α-TWIST1 pathways and PDGF-driven proliferative cues, which together promote the transition of pulmonary ECs toward a mesenchymal and pro-fibrotic phenotype [10,40,107].

3.3. Kidney

EndMT contributes to renal fibrosis in chronic kidney disease (CKD), as lineage-tracing has shown that a subset of renal fibroblasts derives from ECs and is associated with microvascular rarefaction and disease progression [110,111]. The TGF-β/SMAD pathway, along with non-canonical cascades (ERK, p38 MAPK, PI3K/AKT), and Wnt/β-catenin and Notch signaling, can drive renal EndMT [40,61,86]. These pathways are activated by angiotensin II, oxidative stress/ROS and hyperglycemia, as well as inflammatory cytokines (IL-1β, TNF-α) [112]. Under chronic hypoxic conditions, endothelial HIF-dependent signaling may promote EndMT and microvascular rarefaction, which worsens oxygen delivery and causes peritubular injury, accelerating renal remodeling in CKD [111]. Thus, renal EndMT exemplifies how canonical TGF-β/SMAD, Wnt/β-catenin, and Notch signailing intersect with metabolic and oxidative stress to drive microvascular rarefaction and progressive tubulointerstitial fibrosis [11,57,68,110,112,113].

3.4. Liver

In chronic liver disease, liver sinusoidal ECs undergo capillarization and can adopt partial EndMT features under pro-fibrotic cues (TGF-β, hypoxia), contributing to excess ECM deposition and fibrotic remodeling in experimental in vivo models [114]. Ex vivo analyses of human fibrotic liver tissues/datasets further report endothelial populations with mixed endothelial and mesenchymal signatures consistent with partial EndMT [115]. Although stellate cells remain the principal source of myofibroblasts, context-dependent liver sinusoidal EC-EndMT may augment fibrogenesis and vascular remodeling as the disease advances [114,116]. Available evidence suggests that sinusoidal EndMT in chronic liver disease in driven largely by TGF-β/SMAD and hypoxia/HIF pathways, potentially modulated by inflammatory cytokines and epigenetic changes, amplifying fibrogenic and vascular remodeling responses as disease progresses [12,114,117,118].

3.5. Central Nervous System

In the central nervous system (CNS), neuroinflammatory cues (TGF-β, IL-1β) can induce EndMT in brain microvascular ECs, contributing to loss of BBB integrity and enhanced leukocyte infiltration in multiple sclerosis and experimental autoimmune encephalomyelitis [13,116]. Regulators, such as ETS1, modulate EndMT and BBB integrity. Inhibition of ETS1 has been associated with EndMT-linked BBB dysfunction in these models [13]. Genetic and cellular evidence from cerebral cavernous malformation analyses further supports a pathogenic role of EndMT in CNS vascular dysfunction [95]. At the molecular level, CNS EndMT appears to be governed by TGF-β/SMAD signaling together with inflammatory pathways such as NF-κB and transcriptional regulators, including ETS1, linking neuroinflammation and BBB breakdown to endothelial plasticity [13,119].

3.6. Tumor Microenvironment

In cancer, ECs undergoing EndMT lose endothelial markers and acquire mesenchymal traits, contributing to a subset of cancer-associated fibroblasts and secreting cytokines/ECM components that can promote tumor progression, angiogenesis, and immune evasion [94]. This phenomenon has been documented in breast, colon, and pancreatic cancers and is linked to inflammatory cues and TGF-β-driven EndMT programs [69,119,120]. Tumor-associated EndMT is primarily driven by TGF-β/SMAD signaling in concert with inflammatory cytokines and pro-angiogenic cues, accompanied by transcriptional and epigenetic reprogramming that generates EndMT-derived cancer-associated fibroblasts and reinforces an immune-evasive, pro-fibrotic tumor microenvironment [69,94,120,121].

4. Disease Models and Assessment Methods for EndMT

Refined in vitro (TGF-β, hypoxia, 2D-3D matrices, microfluidic shear) and in vivo (endothelial lineage-tracing, disease-specific) models with standardized marker panels, functional readouts and single-cell sequencing have facilitated EndMT research and informed therapeutic discovery across vascular and fibrotic diseases [37,91,122,123,124] (Table 2).

4.1. In Vitro Model Systems

4.1.1. Mechanochemical Stimulation-Based Models

In vivo, ECs are consistently exposed to dynamic mechanical forces, including shear stress, cyclic stretch, and ECM-derived cues that help maintain vascular homeostasis [38,136]. However, disturbed or pathological mechanics can reprogram endothelial phenotype and promote EndMT by engaging pathways such as Notch and TGF-β, with downstream effectors including SNAI1 and TWIST, while cell–matrix traction and substrate stiffness regulate barrier function and transcriptional states [38,136]. In vitro models using oscillatory-flow systems, hypoxic chambers, and stiff substrates have shown that disturbed shear and hypoxia can induce EndMT via Notch- and HIF-dependent pathways, whereas pathological matrix stiffness drives ECs toward the mesenchymal state [16,40,127]. These platforms provide mechanistic insight into how aberrant mechanical environments promote endothelial dysfunction and EndMT in vitro.

4.1.2. Two-Dimensional Monolayer Culture Models

Two-dimensional monolayer cultures of primary ECs (HUVECs, HAECs) are widely used platforms for studying EndMT, because they enable tightly controlled stimulation with defined inducers including TGF-β, TNF-α and hypoxia [16,125,137]. These models are convenient and reproducible; however, they lack the spatial and cellular heterogeneity of native tissues and microenvironments [126]. After EndMT induction, progression is quantified by shifts in endothelial/mesenchymal marker expression, barrier readouts such as transendothelial electrical resistance or FITC-dextran permeability, and morphological/cytoskeletal remodeling [13,38,40]. To overcome 2D limitations, many studies combine hypoxia or controlled flow with stiff substrates and transition to 3D and microfluidic systems that better recapitulate endothelial mechanochemical conditions [16,127,131,132].

4.1.3. Three-Dimensional Models and Organoid Systems

Three-dimensional collagen or Matrigel cultures are suitable for direct studies of cell–matrix interactions and structural remodeling during EndMT [128]. More advanced platforms, such as iPSC-derived vascular organoids and microfluidic organ-on-a-chip systems, incorporate physical cues (shear stress and hypoxia) to better approximate in vivo environments and provide more physiologically relevant EndMT dynamics [129,132]. In these systems, EndMT is tracked by dual-marker shifts, functional barrier readouts, and collagen deposition, including via second-harmonic generation imaging [138]. Organoids are valuable for capturing multicellular crosstalk and hypoxic gradients, while microfluidic chips enable precise control of shear profiles and real-time readouts under live imaging [130].
However, these platforms can exhibit batch-to-batch variability and differentiation-dependent heterogeneity in cellular composition and microenvironmental parameters [129,130,132]. Therefore, reproducibility is improved by standardized differentiation/operating protocols and transparent reporting of core QC metrics alongside EndMT readouts [129,130,132].

4.1.4. Hypoxia-Based In Vitro Models

Coronary microvascular rarefaction exacerbates tissue hypoxia and is associated with cardiac fibrosis and adverse ventricular remodeling [41,111,139]. Pathological observations have shown microvascular rarefaction and dysfunction with myocardial fibrosis and left ventricular remodeling, while hypoxia/HIF-1α signaling is implicated in these processes [41,111,139]. Hypoxia can induce EndMT via HIF-1α/TWIST1 and TGF-β/SMAD pathways, mediating the phenotypic transitions reported in PAH and cardiac fibrosis and supporting hypoxic ECs culture as an informative in vitro platform for EndMT research [16,42]. In PAH models, hypoxia/HIF-1α signaling engages TWIST1/PDGFB and TGF-β/SMAD, which promote intimal thickening and arterial remodeling, supporting these pathways as potential therapeutic targets [42,109].

4.2. In Vivo Model Systems

Genetically engineered mouse models for EndMT use endothelial lineage-specific Cre drivers (Tie2-Cre, VE-cadherin-CreERT2) with Rosa26 reporters to trace endothelial progeny [133,134,135]. Complementary functional genetics, such as Cre-induced TβRII deletion and SMAD3 deficiency or perturbation, allow tests of necessity across canonical fibrosis models, including MI, unilateral ureteral obstruction, and bleomycin-induced pulmonary fibrosis [57,140,141,142]. Zebrafish, with optical transparency and rapid development, enable real-time vascular-/fibrosis-related imaging using transgenic reporters and live biosensors [143,144]. However, because injured zebrafish often display transient or limited fibrotic scarring due to robust regeneration, caution is warranted when translating findings to persistent mammalian fibrosis [145]. In contrast, rat models can offer closer physiological concordance for organ-level fibrotic outcomes and are suitable for multimodal readouts, including MRI and PET/CT [146].

4.3. EndMT Detection and Quantification Methods

4.3.1. Marker-Panels and 3D Tissue Imaging

Marker-based EndMT detection typically relies on identifying loss of endothelial markers alongside gain of mesenchymal markers, quantified by immunofluorescence, flow cytometry, and immunoblotting [9,122]. While single markers are neither necessary nor sufficient to establish EndMT progression, and some fibroblast markers lack specificity, dual or multiplex panels are recommended to reduce misclassification and to support phenotypic calls within biologically coherent programs [8,88]. 3D tissue-level assessment of EndMT within fibrotic niches can be achieved with tissue clearing and light-sheet microscopy [14]. When these approaches are combined with appropriate lineage reporters, they enable whole-organ visualization of endothelial progeny and spatial quantification of their positions and fractions relative to collagen-dense scars [14,89].
In practice, the sensitivity of imaging-based detection depends on antibody validation and signal-to-noise, and on whole-mount immunolabeling performance and clearing/optional conditions [8,14,88,89]. Specificity is limited by marker overlap across endothelial, mural, and fibroblast-like lineages. Therefore, EndMT calls should be supported by coherent multi-marker programs rather than single markers [8,9,15,88]. To improve inter-laboratory reproducibility, studies should report the exact marker panel/antibody clones, clearing and imaging parameters, and quantification thresholds, and confirm key findings using an orthogonal modality when feasible [89].

4.3.2. EndMT Fate Mapping and Transcriptional Resolution

In vivo lineage tracing of EndMT commonly employs endothelial-specific Cre drivers crossed with Rosa26 reporters, providing permanent genetic labeling of endothelial progeny and supporting an endothelial contribution [133,134,135]. As recombination efficiency and specificity depend on the induction timing/dose and controls, best practice includes dual-recombinase intersectional strategies to increase lineage specificity [89,147]. Ex vivo single-cell transcriptional approaches use scRNA-seq to resolve endothelial subpopulations and transcriptional states during EndMT progression [124]. Integration of scRNA-seq with scATAC-seq has also identified motif activity inferred from chromatin accessibility [37]. Spatial transcriptomics has localized signatures consistent with EndMT programs within organ-specific niches (post-myocardial infarction (MI) heart, IPF lungs, atherosclerotic plaques), enabling spatial mapping of putative transitions from endothelial identity loss to mesenchymal acquisition [91,102,148].
For sequencing-based readouts, sensitivity for rare or transient EndMT states is influenced by sampling depth and transcript dropout, while specificity can be affected by dissociation-induced transcript dropout, ambient RNA contamination, and cell doublets that blur endothelial–mesenchymal boundaries [149,150]. Accordingly, trajectory outputs should be interpreted as inference and anchored to phenotypic/lineage-aware validation where possible [92]. Finally, cross-study comparability requires reproducibility safeguards, including clear reporting of tissue processing, QC metrics, batch-aware computational workflows, and clearly defined EndMT signature criteria/thresholds [149,151].

4.4. Organ-Specific Considerations for EndMT Models and Assessment

From a practical standpoint, EndMT research typically combines a disease-relevant in vivo model with a standardized detection workflow (Section 4.1, Section 4.2 and Section 4.3). In the cardiovascular studies, MI or pressure-overload models are commonly used to link EndMT signatures to fibrotic remodeling, whereas atherosclerosis models under disturbed-flow conditions are used to examine plaque-associated EndMT [22,30,43]. In pulmonary diseases, bleomycin-induced fibrosis and hypoxia-based pulmonary hypertension models are frequently employed to assess EndMT-associated vascular remodeling using multi-marker staining and morphometric readouts [40,54,142]. Renal EndMT is often evaluated in unilateral ureteral obstruction and diabetic or hypertensive nephropathy models, alongside endpoints such as collagen deposition and microvascular rarefaction [57,63,110]. In more context-dependent settings (liver, CNS, and cancer), chronic injury or inflammatory disease models combined with multi-marker staining and, where applicable, single-cell or spatial profiling, are increasingly used to localize EndMT-like states within disease niches [88,91,114,120,127].

5. Therapeutic Regulation of EndMT

5.1. Signaling Pathway Inhibitors

5.1.1. Inhibition of the TGF-β Pathway

The TGF-β pathway is a central and extensively studied driver of EndMT. Pharmacologic blockade of TβRI with small molecules such as SB-431542 suppresses TGF-β-induced EndMT by preventing SMAD2/3 phosphorylation in vitro and in disease contexts [33,152]. A-83-01, another TβRI inhibitor, similarly suppresses TGF-β-driven transdifferentiation [34]. Galunisertib (LY2157299), an oral TβRI inhibitor, has shown anti-fibrotic/anti-tumor activity in preclinical studies and early clinical signals in hepatocellular carcinoma and pancreatic cancer trials [153,154]. Because biomarker-guided patient selection has emerged as important in clinical experiences with TGF-β pathway inhibition, integrating pharmacodynamic markers into study design is advisable [153]. In addition, TGF-β signaling engages non-canonical branches (p38/JNK/ERK, RhoA/ROCK, PI3K/AKT) in EndMT, pairing TβRI blockade with barrier-function readouts, cytoskeletal remodeling, and transcriptional programs is commonly employed [29,60].

5.1.2. Modulation of Notch Signaling

Notch signaling, which is activated through juxtacrine interactions, is implicated in EndMT in developmental and pathological contexts, including atherosclerosis, cardiac fibrosis, and vascular malformations [27,155]. Pharmacologic γ-secretase inhibition with DAPT, a Notch pathway inhibitor, has been reported to attenuate TGF-β-driven EndMT in endothelial models [156]. Disturbed flow-activated JAG1-NOTCH4 promotes vascular dysfunction and EndMT-like programs in atherosclerosis, indicating that Notch1 and Notch4 are context-dependent regulators of endothelial fate [39]. Given that broad γ-secretase/Notch blockade has raised concerns about endothelial homeostasis and dose-limiting toxicities, context-selective targeting of disturbed-flow-activated JAG1-NOTCH4 has been proposed as a strategy that may suppress pathological EndMT while better preserving physiological Notch signaling [38,39].

5.1.3. Modulation of Wnt/β-Catenin Pathway

Wnt/β-catenin signaling is an important regulator implicated in EndMT, particularly in fibrotic and inflammatory conditions [25]. Pharmacologic disruption of the β-catenin/CBP interaction with ICG-001, a selective β-catenin/CBP inhibitor, dampens mesenchymal gene programs and can partially restore endothelial features [51,52]. Notably, β-catenin inhibition rescues AKT1-suppression-driven EndMT and vascular remodeling in vivo [51]. PRI-724, a second-generation β-catenin/CBP antagonist, shares the same mechanism as ICG-001 and supports a strategy to modulate β-catenin/CBP-dependent transcription without broadly shutting down Wnt signaling [53].

5.2. Epigenetic and ncRNA-Based Therapy Strategies

5.2.1. Epigenetic Inhibitors: HDAC and DNMT Inhibition

HDACs and DNMTs are key epigenetic regulators of EndMT [10,22]. Activity of HDAC9 and HDAC3 has been implicated in promoting mesenchymal reprogramming during EndMT, while HDAC inhibition has been reported to restore histone acetylation and attenuate TGF-β-driven EndMT features [22,44,157]. Conversely, disturbed flow can induce DNMT1-dependent hypermethylation at mechanosensitive promoters, such as KLF4 [36,43]. DNMT inhibition with 5-aza-2′-deoxycytidine has been reported to attenuate EndMT and endothelial dysfunction in preclinical models [10].

5.2.2. microRNA-Based Therapeutics

The miR-200 family (miR-200a/-200b/-429) suppresses EndMT by directly targeting ZEB1/2 and its loss under fibrotic or inflammatory stress promotes mesenchymal reprogramming [48,72]. Conversely, miR-21 is upregulated by TGF-β and accelerates EndMT via SMAD7 repression and AKT signaling [49,50]. Restoring anti-EndMT miRs, such as miR-200b, or inhibiting miR-21 attenuates mesenchymal marker expression and fibrosis in preclinical models [47,158]. Exosomes, lipid nanoparticles, and polymeric carriers can improve the stability and tissue targeting of miR therapeutics [159,160,161]. Endothelial-trophic delivery strategies underscore the importance of cell-specific targeting to minimize off-target pathway perturbations [162].

5.3. Antibodies and Biologic Agents

Monoclonal antibodies that modulate TGF-β signaling have shown preclinical and early clinical signals against fibrosis and EndMT-associated pathologies [35]. Fresolimumab, a pan-TGF-β-neutralizing antibody, suppresses TGF-β-driven fibrotic programs and showed clinical activity in early systemic sclerosis studies [163]. The anti-endoglin antibody TRC105 antagonizes endoglin co-receptor activity, ameliorates endothelial dysfunction, and targets pathological angiogenesis, consistent with the context-dependent role of endoglin in EndMT and vascular remodeling [164,165]. Because broad TGF-β blockade can cause dose-limiting toxicities, isoform- or co-receptor-targeted strategies, such as endoglin-directed modulation, have been proposed to better suppress pathological EndMT while sparing physiological signaling [17,166].

5.4. Small Molecule- and Compound-Based Strategies

Small molecule- and compound-based approaches have been reported to modulate pathological EndMT under fibrotic conditions [167]. Pirfenidone preserves endothelial network integrity and attenuates mesenchymal programs, in part via inhibition of Rho-kinase activity [55]. Nintedanib suppresses EndMT by reducing FAK activity and improves bleomycin-induced pulmonary fibrosis and experimental PAH [54,56]. Polyphenols, such as resveratrol and curcumin, have been shown to suppress EndMT/Endothelial-interstitial transitions in endothelial models [168,169]. Resveratrol counters NOX-mediated EndMT via PKC inhibition under high-glucose conditions, and curcumin reduces endothelial-interstitial transformation [168,169]. Combination strategies with anti-TGF-β or anti-inflammatory agents have been proposed to enhance anti-EndMT efficacy while limiting off-target effects [96,167].

5.5. Cell- and Gene-Based Therapeutic Strategies

iPSC-derived ECs (iPSC-ECs) have shown promise in preclinical models of vascular repair and attenuation of fibrotic remodeling [170,171]. CRISPR/Cas9 editing is feasible in primary ECs and iPSC-EC systems and provides a platform to modulate EndMT-relevant pathways in experimental settings [172,173]. Mechanistically, the miR-200 family suppresses EndMT, consistent with its direct repression of ZEB1/2 described in foundational EMT studies and validated in endothelial contexts [48]. In renal models, endothelial EndMT compromises vascular integrity and drives fibrosis via Myc-linked metabolic reprogramming, underscoring transcriptional regulators, such as SNAI1, as potential nodes for intervention and supporting cell-/gene-based strategies to stabilize engineered ECs [174]. Strategic vector selection, rigorous off-target profiling, and endothelial-tropic delivery are pivotal for translational success [162]. PECAM-1-targeting enables ApoE-independent lung-endothelial delivery of mRNA-loaded lipid nanoparticles, while PECAM-1/VCAM-1-directed DNA-lipid nanoparticles enhance the magnitude, duration, and organ specificity of endothelial transgene expression [162,175] (Table 3).

6. Conclusions

EndMT is increasingly recognized as an important contributor to fibrosis, vascular remodeling, and the tumor microenvironment [30,176]. The process appears organ- and time-dependent, creating opportunities for selective therapeutic interventions [7,124]. Recent advances in ex vivo single-cell omics have enabled the high-resolution profiling of EndMT-related populations across tissues and disease states, helping resolve cellular heterogeneity and infer transient versus more stable transition states [37,124]. When integrated with multiplexed and spatial imaging, these datasets can localize EndMT populations within tissue niches and map their interactions with stromal and immune cells, strengthening pathophysiological interpretation [91,102,148]. Additionally, patient-derived iPSC models and organoid/organ-on-a-chip systems may help establish disease-specific EndMT progression, supporting target discovery and exploration of patient-to-patient variability in drug responses [177,178,179]. However, translating findings from reductionist in vitro systems or emerging vascular organoid/on-chip platforms to human physiology remains challenging [126,130,132]. Key limitations include incomplete endothelial subtype maturation, reduced immune-stromal complexity, and cross-platform variability that complicates benchmarked endpoint alignment [129,132,180]. Finally, therapeutic strategies that combine pathway-directed agents with epigenetic modulation and immunoregulation may offer broader control of EndMT in preclinical models than single-pathway blockade, with potential to translate into meaningful anti-fibrotic effects [18,44,122].
Despite these advances, several conceptual and technical challenges remain. In vivo evidence often relies on endothelial lineage tracing and marker-based definitions [8,108,109,110]. However, lineage-tracing readouts can be confounded by Cre-driver recombination specificity/efficiency and induction timing and dose [109,110]. Marker-based definitions are limited by overlap and imperfect specificity of commonly used endothelial and fibroblast markers across related stromal lineages [8,108]. Single-cell and spatial datasets also provide largely static snapshots that infer, rather than directly observe, real-time state transitions [40,112]. Accordingly, integrating lineage information with longitudinal sampling and orthogonal functional readouts will be essential to distinguish EndMT trajectories from stable mesenchymal or perivascular phenotypes [31,109,110]. Key unresolved questions include the quantitative contribution of EndMT to fibroblast-like pools across organs and stages, whether partial EndMT reflects stable fate change versus adaptive endothelial activation, and the extent of reversibility in vivo under physiologically relevant conditions [15,30,181].
Most EndMT-targeted interventions remain at the preclinical stage, and safety, specificity, and delivery constraints continue to limit clinical translation [167,182]. While broad TGF-β pathway inhibition can attenuate EndMT and fibrosis, it is constrained by dose-limiting toxicities and the essential physiological roles of TGF-β signaling [35,183]. Likewise, miR- and gene-based approaches require rigorous off-target profiling and endothelial-tropic delivery to achieve durable and safe control in vivo [160,161,162,182]. Given the redundancy and extensive cross-talk among EndMT-regulatory networks, translational development will require mechanism-aware, context-selective strategies rather than single-pathway blockade [8,9,15]. Single-cell and spatial profiling can nominate tissue- and stage-resolved, program-level EndMT signature and trajectory-informed biomarker panels that support patient/lesion stratification and pharmacodynamic monitoring [91,93,124]. However, because snapshot omics infer rather than directly observe dynamic transitions, these signatures should be corroborated by orthogonal validation frameworks to establish in vivo dynamics under physiologically relevant conditions [89,92,144].

Author Contributions

Conceptualization, all authors; investigation, R.K.; writing—original draft preparation, R.K.; writing—review and editing, W.C.; supervision, W.C.; funding acquisition, W.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Bio & Medical Technology Development Program of the National Research Foundation (NRF) funded by the Ministry of Science & ICT (RS-2024-00399681).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ApoEApolipoprotein E
BBBBlood–Brain Barrier
CBPCREB-Binding Protein
CCMCerebral Cavernous Malformation
CKDChronic Kidney Disease
CNSCentral Nervous System
CREBcAMP Response Element-Binding Transcription Factor
DNMTDNA Methyltransferase
ECEndothelial Cell
ECMExtracellular Matrix
EndMTEndothelial-to-Mesenchymal Transition
ERKExtracellular Signal-Regulated Kinase
FITCFluorescein Isothiocyanate
HAECHuman Aortic Endothelial Cell
HDACHistone Deacetylase
HIFHypoxia-Inducible Factor
HUVECHuman Umbilical Vein Endothelial Cell
ILInterleukin
IPFIdiopathic Pulmonary Fibrosis
iPSCInduced Pluripotent Stem Cell
MAPKMitogen-Activated Protein Kinase
MIMyocardial Infarction
miRmicroRNA
MMPMatrix Metalloproteinase
PAHPulmonary Arterial Hypertension
PDGFPlatelet-Derived Growth Factor
PI3KPhosphoinositide 3-Kinase
ROSReactive Oxygen Species
scRNA-seqSingle Cell RNA Sequencing
TβRTransforming Growth Factor-Beta Receptor
TGF-βTransforming Growth Factor-Beta
TNF-αTumor Necrosis Factor-Alpha
vWFvon Willebrand Factor

References

  1. Peng, Z.; Shu, B.; Zhang, Y.; Wang, M. Endothelial Response to Pathophysiological Stress. Arterioscler. Thromb. Vasc. Biol. 2019, 39, e233–e243. [Google Scholar] [CrossRef]
  2. Chen, L.; Qu, H.; Liu, B.; Chen, B.C.; Yang, Z.; Shi, D.Z.; Zhang, Y. Low or oscillatory shear stress and endothelial permeability in atherosclerosis. Front. Physiol. 2024, 15, 1432719. [Google Scholar] [CrossRef]
  3. Aveleira, C.A.; Lin, C.M.; Abcouwer, S.F.; Ambrósio, A.F.; Antonetti, D.A. TNF-α Signals Through PKCζ/NF-κB to Alter the Tight Junction Complex and Increase Retinal Endothelial Cell Permeability. Diabetes 2010, 59, 2872–2882. [Google Scholar] [CrossRef]
  4. Sawa, Y.; Sugimoto, Y.; Ueki, T.; Ishikawa, H.; Sato, A.; Nagato, T.; Yoshida, S. Effects of TNF-alpha on leukocyte adhesion molecule expressions in cultured human lymphatic endothelium. J. Histochem. Cytochem. 2007, 55, 721–733. [Google Scholar] [CrossRef]
  5. He, P.; Talukder, M.A.H.; Gao, F. Oxidative Stress and Microvessel Barrier Dysfunction. Front. Physiol. 2020, 11, 472. [Google Scholar] [CrossRef] [PubMed]
  6. Dejana, E. Endothelial cell-cell junctions: Happy together. Nat. Rev. Mol. Cell. Biol. 2004, 5, 261–270. [Google Scholar] [CrossRef] [PubMed]
  7. Pober, J.S.; Sessa, W.C. Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 2007, 7, 803–815. [Google Scholar] [CrossRef]
  8. Kovacic, J.C.; Dimmeler, S.; Harvey, R.P.; Finkel, T.; Aikawa, E.; Krenning, G.; Baker, A.H. Endothelial to Mesenchymal Transition in Cardiovascular Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 190–209. [Google Scholar] [CrossRef]
  9. Piera-Velazquez, S.; Jimenez, S.A. Endothelial to Mesenchymal Transition: Role in Physiology and in the Pathogenesis of Human Diseases. Physiol. Rev. 2019, 99, 1281–1324. [Google Scholar] [CrossRef]
  10. Gorelova, A.; Berman, M.; Al Ghouleh, I. Endothelial-to-Mesenchymal Transition in Pulmonary Arterial Hypertension. Antioxid. Redox Signal. 2021, 34, 891–914. [Google Scholar] [CrossRef]
  11. Zeisberg, E.M.; Potenta, S.E.; Sugimoto, H.; Zeisberg, M.; Kalluri, R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J. Am. Soc. Nephrol. 2008, 19, 2282–2287. [Google Scholar] [CrossRef] [PubMed]
  12. Li, Z.; Chen, B.; Dong, W.; Kong, M.; Fan, Z.; Yu, L.; Wu, D.; Lu, J.; Xu, Y. MKL1 promotes endothelial-to-mesenchymal transition and liver fibrosis by activating TWIST1 transcription. Cell Death Dis. 2019, 10, 899. [Google Scholar] [CrossRef]
  13. Luo, Y.; Yang, H.; Wan, Y.; Yang, S.; Wu, J.; Chen, S.; Li, Y.; Jin, H.; He, Q.; Zhu, D.Y.; et al. Endothelial ETS1 inhibition exacerbate blood–brain barrier dysfunction in multiple sclerosis through inducing endothelial-to-mesenchymal transition. Cell Death Dis. 2022, 13, 462. [Google Scholar] [CrossRef]
  14. Chung, K.; Wallace, J.; Kim, S.Y.; Kalyanasundaram, S.; Andalman, A.S.; Davidson, T.J.; Mirzabekov, J.J.; Zalocusky, K.A.; Mattis, J.; Denisin, A.K.; et al. Structural and molecular interrogation of intact biological systems. Nature 2013, 497, 332–337. [Google Scholar] [CrossRef]
  15. Xu, Y.; Kovacic, J.C. Endothelial to Mesenchymal Transition in Health and Disease. Annu. Rev. Physiol. 2023, 85, 245–267. [Google Scholar] [CrossRef]
  16. Matsuo, E.; Okamoto, T.; Ito, A.; Kawamoto, E.; Asanuma, K.; Wada, K.; Shimaoka, M.; Takao, M.; Shimamoto, A. Substrate stiffness modulates endothelial cell function via the YAP-Dll4-Notch1 pathway. Exp. Cell Res. 2021, 408, 112835. [Google Scholar] [CrossRef]
  17. Han, L.W.; Jamalian, S.; Hsu, J.C.; Sheng, X.R.; Yang, X.; Yang, X.; Monemi, S.; Hassan, S.; Yadav, R.; Tuckwell, K.; et al. A Phase 1a Study to Evaluate Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of RO7303509, an Anti-TGFβ3 Antibody, in Healthy Volunteers. Rheumatol. Ther. 2024, 11, 755–771. [Google Scholar] [CrossRef]
  18. Deng, Z.; Fan, T.; Xiao, C.; Tian, H.; Zheng, Y.; Li, C.; He, J. TGF-β signaling in health, disease and therapeutics. Signal Transduct. Target. Ther. 2024, 9, 61. [Google Scholar] [CrossRef] [PubMed]
  19. Niessen, K.; Fu, Y.; Chang, L.; Hoodless, P.A.; McFadden, D.; Karsan, A. Slug is a direct Notch target required for initiation of cardiac cushion cellularization. J. Cell Biol. 2008, 182, 315–325. [Google Scholar] [CrossRef]
  20. Mahmoud, M.M.; Kim, H.R.; Xing, R.; Hsiao, S.; Mammoto, A.; Chen, J.; Serbanovic-Canic, J.; Feng, S.; Bowden, N.P.; Maguire, R.; et al. TWIST1 Integrates Endothelial Responses to Flow in Vascular Dysfunction and Atherosclerosis. Circ. Res. 2016, 119, 450–462. [Google Scholar] [CrossRef]
  21. Kim, R.; Kim, S.; Kim, H.; Jeong, S.; Moon, H.; Kim, J.; Song, B.W.; Chang, W. The antioxidant effects of decursin inhibit EndMT progression through PI3K/AKT/NF-κB and Smad signaling pathways. BMB Rep. 2025, 58, 406–414. [Google Scholar] [CrossRef]
  22. Lecce, L.; Xu, Y.; V’Gangula, B.; Chandel, N.; Pothula, V.; Caudrillier, A.; Santini, M.P.; d’Escamard, V.; Ceholski, D.K.; Gorski, P.A.; et al. Histone deacetylase 9 promotes endothelial-mesenchymal transition and an unfavorable atherosclerotic plaque phenotype. J. Clin. Investig. 2021, 131, e131178. [Google Scholar] [CrossRef]
  23. Glaser, S.F.; Heumüller, A.W.; Tombor, L.; Hofmann, P.; Muhly-Reinholz, M.; Fischer, A.; Günther, S.; Kokot, K.E.; Hassel, D.; Kumar, S.; et al. The histone demethylase JMJD2B regulates endothelial-to-mesenchymal transition. Proc. Natl. Acad. Sci. USA 2020, 117, 4180–4187, Erratum in Proc. Natl. Acad. Sci. USA 2020, 117, 8657. [Google Scholar] [CrossRef]
  24. Tan, X.; Xu, X.; Zeisberg, M.; Zeisberg, E.M. DNMT1 and HDAC2 Cooperate to Facilitate Aberrant Promoter Methylation in Inorganic Phosphate-Induced Endothelial-Mesenchymal Transition. PLoS ONE 2016, 11, e0147816. [Google Scholar] [CrossRef] [PubMed]
  25. Sánchez-Duffhues, G.; García de Vinuesa, A.; Ten Dijke, P. Endothelial-to-mesenchymal transition in cardiovascular diseases: Developmental signaling pathways gone awry. Dev. Dyn. 2018, 247, 492–508. [Google Scholar] [CrossRef] [PubMed]
  26. Ma, L.; Lu, M.F.; Schwartz, R.J.; Martin, J.F. Bmp2 is essential for cardiac cushion epithelial-mesenchymal transition and myocardial patterning. Development 2005, 132, 5601–5611. [Google Scholar] [CrossRef]
  27. Anbara, T.; Sharifi, M.; Aboutaleb, N. Endothelial to Mesenchymal Transition in the Cardiogenesis and Cardiovascular Diseases. Curr. Cardiol. Rev. 2020, 16, 306–314. [Google Scholar] [CrossRef]
  28. Saito, A.; Horie, M.; Nagase, T. TGF-β Signaling in Lung Health and Disease. Int. J. Mol. Sci. 2018, 19, 2460. [Google Scholar] [CrossRef]
  29. Van Meeteren, L.A.; Ten Dijke, P. Regulation of endothelial cell plasticity by TGF-β. Cell Tissue Res. 2011, 347, 177–186. [Google Scholar] [CrossRef]
  30. Zeisberg, E.M.; Tarnavski, O.; Zeisberg, M.; Dorfman, A.L.; McMullen, J.R.; Gustafsson, E.; Chandraker, A.; Yuan, X.; Pu, W.T.; Roberts, A.B.; et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat. Med. 2007, 13, 952–961. [Google Scholar] [CrossRef] [PubMed]
  31. Ma, X.; Zhao, D.; Yuan, P.; Li, J.; Yun, Y.; Cui, Y.; Zhang, T.; Ma, J.; Sun, L.; Ma, H.; et al. Endothelial-to-Mesenchymal Transition in Calcific Aortic Valve Disease. Acta Cardiol. Sin. 2020, 36, 183–194. [Google Scholar] [PubMed]
  32. Paranya, G.; Vineberg, S.; Dvorin, E.; Kaushal, S.; Roth, S.J.; Rabkin, E.; Schoen, F.J.; Bischoff, J. Aortic Valve Endothelial Cells Undergo Transforming Growth Factor-β-Mediated and Non-Transforming Growth Factor-β-Mediated Transdifferentiation in Vitro. Am. J. Pathol. 2001, 159, 1335–1343. [Google Scholar] [CrossRef]
  33. Medici, D.; Potenta, S.; Kalluri, R. Transforming growth factor-β2 promotes Snail-mediated endothelial–mesenchymal transition through convergence of Smad-dependent and Smad-independent signaling. Biochem. J. 2011, 437, 515–520. [Google Scholar] [CrossRef]
  34. Tojo, M.; Hamashima, Y.; Hanyu, A.; Kajimoto, T.; Saitoh, M.; Miyazono, K.; Node, M.; Imamura, T. The ALK-5 inhibitor A-83--01 inhibits Smad signaling and epithelial-to-mesenchymal transition by transforming growth factor-β. Cancer Sci. 2005, 96, 791–800. [Google Scholar] [CrossRef]
  35. Budi, E.H.; Schaub, J.R.; Decaris, M.; Turner, S.; Derynck, R. TGF-β as a driver of fibrosis: Physiological roles and therapeutic opportunities. J. Pathol. 2021, 254, 358–373. [Google Scholar] [CrossRef]
  36. Jiang, Y.Z.; Jiménez, J.M.; Ou, K.; McCormick, M.E.; Zhang, L.D.; Davies, P.F. Hemodynamic disturbed flow induces differential DNA methylation of endothelial Kruppel-Like Factor 4 promoter in vitro and in vivo. Circ. Res. 2014, 115, 32–43. [Google Scholar] [CrossRef]
  37. Andueza, A.; Kumar, S.; Kim, J.; Kang, D.W.; Mumme, H.L.; Perez, J.I.; Villa-Roel, N.; Jo, H. Endothelial Reprogramming by Disturbed Flow Revealed by Single-Cell RNA and Chromatin Accessibility Study. Cell Rep. 2020, 33, 108491. [Google Scholar] [CrossRef]
  38. Mack, J.J.; Mosqueiro, T.S.; Archer, B.J.; Jones, W.M.; Sunshine, H.; Faas, G.C.; Briot, A.; Aragón, R.L.; Su, T.; Romay, M.C.; et al. NOTCH1 is a mechanosensor in adult arteries. Nat. Commun. 2017, 8, 1620. [Google Scholar] [CrossRef]
  39. Souilhol, C.; Tardajos Ayllon, B.; Li, X.; Diagbouga, M.R.; Zhou, Z.; Canham, L.; Roddie, H.; Pirri, D.; Chambers, E.V.; Dunning, M.J.; et al. JAG1-NOTCH4 mechanosensing drives atherosclerosis. Sci. Adv. 2022, 8, eabo7958. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, B.; Niu, W.; Dong, H.Y.; Liu, M.L.; Luo, Y.; Li, Z.C. Hypoxia induces endothelial-mesenchymal transition in pulmonary vascular remodeling. Int. J. Mol. Med. 2018, 42, 270–278. [Google Scholar] [CrossRef] [PubMed]
  41. Sato, T.; Takeda, N. The roles of HIF-1α signaling in cardiovascular diseases. J. Cardiol. 2023, 81, 202–208. [Google Scholar] [CrossRef]
  42. Mammoto, A.; Hendee, K.; Muyleart, M.; Mammoto, T. Endothelial Twist1-PDGFB signaling mediates hypoxia-induced proliferation and migration of αSMA-positive cells. Sci. Rep. 2020, 10, 7563. [Google Scholar] [CrossRef]
  43. Zhao, J.; Zhao, C.; Yang, F.; Jiang, Z.; Zhu, J.; Yao, W.; Pang, W.; Zhou, J. DNMT1 mediates the disturbed flow-induced endothelial to mesenchymal transition through disrupting β-alanine and carnosine homeostasis. Theranostics 2023, 13, 4392–4411. [Google Scholar] [CrossRef]
  44. Chen, L.; Shang, C.; Wang, B.; Wang, G.; Jin, Z.; Yao, F.; Yue, Z.; Bai, L.; Wang, R.; Zhao, S.; et al. HDAC3 inhibitor suppresses endothelial-to-mesenchymal transition via modulating inflammatory response in atherosclerosis. Biochem. Pharmacol. 2021, 192, 114716. [Google Scholar] [CrossRef]
  45. Vanchin, B.; Sol, M.; Gjaltema, R.A.F.; Brinker, M.; Kiers, B.; Pereira, A.C.; Harmsen, M.C.; Moonen, J.A.J.; Krenning, G. Reciprocal regulation of endothelial–mesenchymal transition by MAPK7 and EZH2 in intimal hyperplasia and coronary artery disease. Sci. Rep. 2021, 11, 17764. [Google Scholar] [CrossRef]
  46. Fledderus, J.; Brouwer, L.; Kuiper, T.; Harmsen, M.C.; Krenning, G. H3K27Me3 abundance increases fibrogenesis during endothelial-to-mesenchymal transition via the silencing of microRNA-29c. Front. Cardiovasc. Med. 2024, 11, 1373279. [Google Scholar] [CrossRef]
  47. Feng, B.; Cao, Y.; Chen, S.; Chu, X.; Chu, Y.; Chakrabarti, S. miR-200b Mediates Endothelial-to-Mesenchymal Transition in Diabetic Cardiomyopathy. Diabetes 2016, 65, 768–779. [Google Scholar] [CrossRef]
  48. Korpal, M.; Lee, E.S.; Hu, G.; Kang, Y. The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. J. Biol. Chem. 2008, 283, 14910–14914. [Google Scholar] [CrossRef]
  49. Kumarswamy, R.; Volkmann, I.; Jazbutyte, V.; Dangwal, S.; Park, D.H.; Thum, T. Transforming growth factor-β-induced endothelial-to-mesenchymal transition is partly mediated by microRNA-21. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 361–369. [Google Scholar] [CrossRef] [PubMed]
  50. Li, Q.; Yao, Y.; Shi, S.; Zhou, M.; Zhou, Y.; Wang, M.; Chiu, J.J.; Huang, Z.; Zhang, W.; Liu, M.; et al. Inhibition of miR-21 alleviated cardiac perivascular fibrosis via repressing EndMT in T1DM. J. Cell. Mol. Med. 2020, 24, 910–920. [Google Scholar] [CrossRef]
  51. Sabbineni, H.; Verma, A.; Artham, S.; Anderson, D.; Amaka, O.; Liu, F.; Narayanan, S.P.; Somanath, P.R. Pharmacological inhibition of β-catenin prevents EndMT in vitro and vascular remodeling in vivo resulting from endothelial Akt1 suppression. Biochem. Pharmacol. 2019, 164, 205–215. [Google Scholar] [CrossRef] [PubMed]
  52. Emami, K.H.; Nguyen, C.; Ma, H.; Kim, D.H.; Jeong, K.W.; Eguchi, M.; Moon, R.T.; Teo, J.L.; Kim, H.Y.; Moon, S.H.; et al. A small molecule inhibitor of beta-catenin/CREB-binding protein transcription. Proc. Natl. Acad. Sci. USA 2004, 101, 12682–12687. [Google Scholar] [CrossRef] [PubMed]
  53. Okazaki, H.; Sato, S.; Koyama, K.; Morizumi, S.; Abe, S.; Azuma, M.; Chen, Y.; Goto, H.; Aono, Y.; Ogawa, H.; et al. The novel inhibitor PRI-724 for Wnt/β-catenin/CBP signaling ameliorates bleomycin-induced pulmonary fibrosis in mice. Exp. Lung Res. 2019, 45, 188–199. [Google Scholar] [CrossRef]
  54. Tsutsumi, T.; Nagaoka, T.; Yoshida, T.; Wang, L.; Kuriyama, S.; Suzuki, Y.; Nagata, Y.; Harada, N.; Kodama, Y.; Takahashi, F.; et al. Nintedanib ameliorates experimental pulmonary arterial hypertension via inhibition of endothelial mesenchymal transition and smooth muscle cell proliferation. PLoS ONE 2019, 14, e0214697. [Google Scholar] [CrossRef]
  55. Nakamura, Y.; Shimizu, Y.; Fujimaki-Shiraishi, M.; Uchida, N.; Takemasa, A.; Niho, S. A Protective Effect of Pirfenidone in Lung Fibroblast–Endothelial Cell Network via Inhibition of Rho-Kinase Activity. Biomedicines 2023, 11, 2259. [Google Scholar] [CrossRef]
  56. Yu, W.K.; Chen, W.C.; Su, V.Y.; Shen, H.C.; Wu, H.H.; Chen, H.; Yang, K.Y. Nintedanib Inhibits Endothelial Mesenchymal Transition in Bleomycin-Induced Pulmonary Fibrosis via Focal Adhesion Kinase Activity Reduction. Int. J. Mol. Sci. 2022, 23, 8193. [Google Scholar] [CrossRef]
  57. Sato, M.; Muragaki, Y.; Saika, S.; Roberts, A.B.; Ooshima, A. Targeted disruption of TGF-β1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J. Clin. Investig. 2003, 112, 1486–1494. [Google Scholar] [CrossRef]
  58. Dobaczewski, M.; Chen, W.; Frangogiannis, N.G. Transforming Growth Factor (TGF)-β signaling in cardiac remodeling. J. Mol. Cell. Cardiol. 2010, 51, 600–606. [Google Scholar] [CrossRef]
  59. Wu, F.; Yang, J.; Liu, J.; Wang, Y.; Mu, J.; Zeng, Q.; Deng, S.; Zhou, H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct. Target. Ther. 2021, 6, 218. [Google Scholar] [CrossRef]
  60. Ciszewski, W.M.; Wawro, M.E.; Sacewicz-Hofman, I.; Sobierajska, K. Cytoskeleton Reorganization in EndMT—The Role in Cancer and Fibrotic Diseases. Int. J. Mol. Sci. 2021, 22, 11607. [Google Scholar] [CrossRef]
  61. Tan, R.J.; Zhou, D.; Zhou, L.; Liu, Y. Wnt/β-catenin signaling and kidney fibrosis. Kidney Int. Suppl. 2014, 4, 84–90. [Google Scholar] [CrossRef] [PubMed]
  62. Ma, J.; Sanchez-Duffhues, G.; Goumans, M.J.; Ten Dijke, P. TGF-β-Induced Endothelial to Mesenchymal Transition in Disease and Tissue Engineering. Front. Cell Dev. Biol. 2020, 8, 260. [Google Scholar] [CrossRef]
  63. Zhang, J.; Chen, S.; Xiang, H.; Xiao, J.; Zhao, S.; Shu, Z.; Chai, Y.; Ouyang, J.; Liu, H.; Wang, X.; et al. S1PR2/Wnt3a/RhoA/ROCK1/β-catenin signaling pathway promotes diabetic nephropathy by inducting endothelial mesenchymal transition and impairing endothelial barrier function. Life Sci. 2023, 328, 121853. [Google Scholar] [CrossRef] [PubMed]
  64. Li, L.; Chen, L.; Zang, J.; Tang, X.; Liu, Y.; Zhang, J.; Bai, L.; Yin, Q.; Lu, Y.; Cheng, J.; et al. C3a and C5a receptor antagonists ameliorate endothelial-myofibroblast transition via the Wnt/β-catenin signaling pathway in diabetic kidney disease. Metabolism 2015, 64, 597–610. [Google Scholar] [CrossRef]
  65. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef] [PubMed]
  66. Liebner, S.; Cattelino, A.; Gallini, R.; Rudini, N.; Iurlaro, M.; Piccolo, S.; Dejana, E. Beta-catenin is required for endothelial-mesenchymal transformation during heart cushion development in the mouse. J. Cell Biol. 2004, 166, 359–367. [Google Scholar] [CrossRef]
  67. Aisagbonhi, O.; Rai, M.; Ryzhov, S.; Atria, N.; Feoktistov, I.; Hatzopoulos, A.K. Experimental myocardial infarction triggers canonical Wnt signaling and endothelial-to-mesenchymal transition. Dis. Model. Mech. 2011, 4, 469–483. [Google Scholar] [CrossRef]
  68. Zhao, Y.; Qiao, X.; Tan, T.K.; Zhao, H.; Zhang, Y.; Liu, L.; Zhang, J.; Wang, L.; Cao, Q.; Wang, Y.; et al. Matrix metalloproteinase 9-dependent Notch signaling contributes to kidney fibrosis through peritubular endothelial–mesenchymal transition. Nephrol. Dial. Transplant. 2017, 32, 781–791. [Google Scholar] [CrossRef] [PubMed]
  69. Ciszewski, W.M.; Sobierajska, K.; Wawro, M.E.; Klopocka, W.; Chefczyńska, N.; Muzyczuk, A.; Siekacz, K.; Wujkowska, A.; Niewiarowska, J. The ILK-MMP9-MRTF axis is crucial for EndMT differentiation of endothelial cells in a tumor microenvironment. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 2283–2296. [Google Scholar] [CrossRef]
  70. Tian, Y.; Xu, Y.; Fu, Q.; Chang, M.; Wang, Y.; Shang, X.; Wan, C.; Marymont, J.V.; Dong, Y. Notch inhibits chondrogenic differentiation of mesenchymal progenitor cells by targeting Twist1. Mol. Cell. Endocrinol. 2015, 403, 30–38. [Google Scholar] [CrossRef]
  71. Gibney, E.R.; Nolan, C.M. Epigenetics and gene expression. Heredity 2010, 105, 4–13. [Google Scholar] [CrossRef]
  72. Kim, J. MicroRNAs as critical regulators of the endothelial to mesenchymal transition in vascular biology. BMB Rep. 2018, 51, 65–72. [Google Scholar] [CrossRef]
  73. Li, Z.; Song, S.; Zha, S.; Wang, C.; Chen, S.; Wang, F. MeCP2 promotes endothelial-to-mesenchymal transition in human endothelial cells by downregulating BMP7 expression. Exp. Cell Res. 2019, 375, 82–89. [Google Scholar] [CrossRef] [PubMed]
  74. Fledderus, J.; Vanchin, B.; Rots, M.G.; Krenning, G. The Endothelium as a Target for Anti-Atherogenic Therapy: A Focus on the Epigenetic Enzymes EZH2 and SIRT1. J. Pers. Med. 2021, 11, 103. [Google Scholar] [CrossRef]
  75. Jang, S.; Choi, N.; Park, J.H.; Yoo, K.H. Contribution of histone deacetylases (HDACs) to the regulation of histone and non-histone proteins: Implications for fibrotic diseases. BMB Rep. 2025, 58, 313–324. [Google Scholar] [CrossRef]
  76. Correia, A.C.; Moonen, J.R.; Brinker, M.G.; Krenning, G. FGF2 inhibits endothelial–mesenchymal transition through microRNA-20a-mediated repression of canonical TGF-β signaling. J. Cell Sci. 2016, 129, 569–579. [Google Scholar] [CrossRef]
  77. Suzuki, H.I.; Katsura, A.; Mihira, H.; Horie, M.; Saito, A.; Miyazono, K. Regulation of TGF-β-mediated endothelial-mesenchymal transition by microRNA-27. J. Biochem. 2017, 161, 417–420. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, J.; He, W.; Xu, X.; Guo, L.; Zhang, Y.; Han, S.; Shen, D. The mechanism of TGF-β/miR-155/c-Ski regulates endothelial–mesenchymal transition in human coronary artery endothelial cells. Biosci. Rep. 2017, 37, BSR20160603. [Google Scholar] [CrossRef] [PubMed]
  79. Miscianinov, V.; Martello, A.; Rose, L.; Parish, E.; Cathcart, B.; Mitić, T.; Gray, G.A.; Meloni, M.; Al Haj Zen, A.; Caporali, A. MicroRNA-148b Targets the TGF-β Pathway to Regulate Angiogenesis and Endothelial-to-Mesenchymal Transition during Skin Wound Healing. Mol. Ther. 2018, 26, 1996–2007. [Google Scholar] [CrossRef]
  80. Wang, Z.; Wang, Z.; Gao, L.; Xiao, L.; Yao, R.; Du, B.; Li, Y.; Wu, L.; Liang, C.; Huang, Z.; et al. miR-222 inhibits cardiac fibrosis in diabetic mice heart via regulating Wnt/β-catenin-mediated endothelium to mesenchymal transition. J. Cell. Physiol. 2020, 235, 2149–2160. [Google Scholar] [CrossRef]
  81. Chen, D.; Zhang, C.; Chen, J.; Yang, M.; Afzal, T.A.; An, W.; Maguire, E.M.; He, S.; Luo, J.; Wang, X.; et al. miRNA-200c-3p promotes endothelial to mesenchymal transition and neointimal hyperplasia in artery bypass grafts. J. Pathol. 2021, 253, 209–224. [Google Scholar] [CrossRef]
  82. Mao, Y.; Jiang, L. MiR-200c-3p promotes ox-LDL-induced endothelial to mesenchymal transition in human umbilical vein endothelial cells through SMAD7/YAP pathway. J. Physiol. Sci. 2021, 71, 30. [Google Scholar] [CrossRef]
  83. Terriaca, S.; Scioli, M.G.; Pisano, C.; Ruvolo, G.; Ferlosio, A.; Orlandi, A. miR-632 Induces DNAJB6 Inhibition Stimulating Endothelial-to-Mesenchymal Transition and Fibrosis in Marfan Syndrome Aortopathy. Int. J. Mol. Sci. 2023, 24, 15133. [Google Scholar] [CrossRef]
  84. Sangwung, P.; Zhou, G.; Nayak, L.; Chan, E.R.; Kumar, S.; Kang, D.W.; Zhang, R.; Liao, X.; Lu, Y.; Sugi, K.; et al. KLF2 and KLF4 control endothelial identity and vascular integrity. JCI Insight 2017, 2, e91700. [Google Scholar] [CrossRef]
  85. Zeisberg, M.; Hanai, J.; Sugimoto, H.; Mammoto, T.; Charytan, D.; Strutz, F.; Kalluri, R. BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat. Med. 2003, 9, 964–968. [Google Scholar] [CrossRef]
  86. Cyr, A.R.; Huckaby, L.V.; Shiva, S.S.; Zuckerbraun, B.S. Nitric Oxide and Endothelial Dysfunction. Crit. Care Clin. 2020, 36, 307–321. [Google Scholar] [CrossRef]
  87. Zhang, S.; Li, Y.; Huang, X.; Liu, K.; Wang, Q.D.; Chen, A.F.; Sun, K.; Lui, K.O.; Zhou, B. Seamless Genetic Recording of Transiently Activated Mesenchymal Gene Expression in Endothelial Cells During Cardiac Fibrosis. Circulation 2021, 144, 2004–2020. [Google Scholar] [CrossRef] [PubMed]
  88. Kong, P.; Christia, P.; Saxena, A.; Su, Y.; Frangogiannis, N.G. Lack of specificity of fibroblast-specific protein 1 in cardiac remodeling and fibrosis. Am. J. Physiol. Heart Circ. Physiol. 2013, 305, H1363–H1372. [Google Scholar] [CrossRef]
  89. Liu, K.; Jin, H.; Zhou, B. Genetic lineage tracing with multiple DNA recombinases: A user’s guide for conducting more precise cell fate mapping studies. J. Biol. Chem. 2020, 295, 6413–6424. [Google Scholar] [CrossRef]
  90. Payne, S.; De Val, S.; Neal, A. Endothelial-Specific Cre Mouse Models. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 2550–2561. [Google Scholar] [CrossRef]
  91. Kuppe, C.; Ramirez Flores, R.O.; Li, Z.; Hayat, S.; Levinson, R.T.; Liao, X.; Hannani, M.T.; Tanevski, J.; Wünnemann, F.; Nagai, J.S.; et al. Spatial multi-omic map of human myocardial infarction. Nature 2022, 608, 766–777. [Google Scholar] [CrossRef]
  92. Weinreb, C.; Wolock, S.; Tusi, B.K.; Socolovsky, M.; Klein, A.M. Fundamental limits on dynamic inference from single-cell snapshots. Proc. Natl. Acad. Sci. USA 2018, 115, E2467–E2476. [Google Scholar] [CrossRef]
  93. Bressan, D.; Battistoni, G.; Hannon, G.J. The dawn of spatial omics. Science 2023, 381, eabq4964. [Google Scholar] [CrossRef]
  94. Zeisberg, E.M.; Potenta, S.; Xie, L.; Zeisberg, M.; Kalluri, R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 2007, 67, 10123–10128. [Google Scholar] [CrossRef]
  95. Maddaluno, L.; Rudini, N.; Cuttano, R.; Bravi, L.; Giampietro, C.; Corada, M.; Ferrarini, L.; Orsenigo, F.; Papa, E.; Boulday, G.; et al. EndMT contributes to the onset and progression of cerebral cavernous malformations. Nature 2013, 498, 492–496. [Google Scholar] [CrossRef]
  96. Alvandi, Z.; Bischoff, J. Endothelial-Mesenchymal Transition in Cardiovascular Disease. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 2357–2369. [Google Scholar] [CrossRef]
  97. Wesseling, M.; Sakkers, T.R.; De Jager, S.C.A.; Pasterkamp, G.; Goumans, M.J. The morphological and molecular mechanisms of epithelial/endothelial-to-mesenchymal transition and its involvement in atherosclerosis. Vascul. Pharmacol. 2018, 106, 1–8. [Google Scholar] [CrossRef]
  98. Cheng, W.; Li, X.; Liu, D.; Cui, C.; Wang, X. Endothelial-to-Mesenchymal Transition: Role in Cardiac Fibrosis. J. Cardiovasc. Pharmacol. Ther. 2021, 26, 3–11. [Google Scholar] [CrossRef] [PubMed]
  99. Zhang, H.; Hui, H.; Li, Z.; Pan, J.; Jiang, X.; Wei, T.; Cui, H.; Li, L.; Yuan, X.; Sun, T.; et al. Pigment epithelium-derived factor attenuates myocardial fibrosis via inhibiting Endothelial-to-Mesenchymal Transition in rats with acute myocardial infarction. Sci. Rep. 2017, 7, 41932. [Google Scholar] [CrossRef]
  100. Garside, V.C.; Chang, A.C.; Karsan, A.; Hoodless, P.A. Co-ordinating Notch, BMP, and TGF-β signaling during heart valve development. Cell. Mol. Life Sci. 2013, 70, 2899–2917. [Google Scholar] [CrossRef]
  101. Mahler, G.J.; Frendl, C.M.; Cao, Q.; Butcher, J.T. Effects of shear stress pattern and magnitude on mesenchymal transformation and invasion of aortic valve endothelial cells. Biotechnol. Bioeng. 2014, 111, 2326–2337. [Google Scholar] [CrossRef]
  102. Sun, J.; Singh, P.; Shami, A.; Kluza, E.; Pan, M.; Djordjevic, D.; Michaelsen, N.B.; Kennbäck, C.; van der Wel, N.N.; Orho-Melander, M.; et al. Spatial Transcriptional Mapping Reveals Site-Specific Pathways Underlying Human Atherosclerotic Plaque Rupture. J. Am. Coll. Cardiol. 2023, 81, 2213–2227. [Google Scholar] [CrossRef]
  103. Jin, Y.G.; Yuan, Y.; Wu, Q.Q.; Zhang, N.; Fan, D.; Che, Y.; Wang, Z.P.; Xiao, Y.; Wang, S.S.; Tang, Q.Z. Puerarin Protects against Cardiac Fibrosis Associated with the Inhibition of TGF- β 1/Smad2-Mediated Endothelial-to-Mesenchymal Transition. PPAR Res. 2017, 2017, 2647129. [Google Scholar] [CrossRef]
  104. Li, Z.; Kong, X.; Zhang, Y.; Zhang, Y.; Yu, L.; Guo, J.; Xu, Y. Dual roles of chromatin remodeling protein BRG1 in angiotensin II-induced endothelial-mesenchymal transition. Cell Death Dis. 2020, 11, 549. [Google Scholar] [CrossRef]
  105. Yoshimatsu, Y.; Wakabayashi, I.; Kimuro, S.; Takahashi, N.; Takahashi, K.; Kobayashi, M.; Maishi, N.; Podyma-Inoue, K.A.; Hida, K.; Miyazono, K.; et al. TNF-α enhances TGF-β-induced endothelial-to-mesenchymal transition via TGF-β signal augmentation. Cancer Sci. 2020, 111, 2385–2399. [Google Scholar] [CrossRef]
  106. May, J.; Mitchell, J.A.; Jenkins, R.G. Beyond epithelial damage: Vascular and endothelial contributions to idiopathic pulmonary fibrosis. J. Clin. Investig. 2023, 133, e1720. [Google Scholar] [CrossRef]
  107. Gaikwad, A.V.; Lu, W.; Dey, S.; Bhattarai, P.; Haug, G.; Larby, J.; Chia, C.; Jaffar, J.; Westall, G.; Singhera, G.K.; et al. Endothelial-to-mesenchymal transition: A precursor to pulmonary arterial remodelling in patients with idiopathic pulmonary fibrosis. ERJ Open Res. 2023, 9, 00487–02022. [Google Scholar] [CrossRef] [PubMed]
  108. Jia, W.; Wang, Z.; Gao, C.; Wu, J.; Wu, Q. Trajectory modeling of endothelial-to-mesenchymal transition reveals galectin-3 as a mediator in pulmonary fibrosis. Cell Death Dis. 2021, 12, 327. [Google Scholar] [CrossRef]
  109. Stenmark, K.R.; Frid, M.; Perros, F. Endothelial-to-Mesenchymal Transition: An Evolving Paradigm and a Promising Therapeutic Target in PAH. Circulation 2016, 133, 1734–1737. [Google Scholar] [CrossRef]
  110. Guerrot, D.; Dussaule, J.C.; Kavvadas, P.; Boffa, J.J.; Chadjichristos, C.E.; Chatziantoniou, C. Progression of renal fibrosis: The underestimated role of endothelial alterations. Fibrogenesis Tissue Repair. 2012, 5 (Suppl. 1), S15. [Google Scholar] [CrossRef]
  111. Xiong, A.; Liu, Y. Targeting Hypoxia Inducible Factors-1α As a Novel Therapy in Fibrosis. Front. Pharmacol. 2017, 8, 326. [Google Scholar] [CrossRef]
  112. Lu, L.; Li, X.; Zhong, Z.; Zhou, W.; Zhou, D.; Zhu, M.; Miao, C. KMT5A downregulation participated in High Glucose-mediated EndMT via Upregulation of ENO1 Expression in Diabetic Nephropathy. Int. J. Biol. Sci. 2021, 17, 4093–4107. [Google Scholar] [CrossRef]
  113. Liang, X.; Duan, N.; Wang, Y.; Shu, S.; Xiang, X.; Guo, T.; Yang, L.; Zhang, S.; Tang, X.; Zhang, J. Advanced oxidation protein products induce endothelial-to-mesenchymal transition in human renal glomerular endothelial cells through induction of endoplasmic reticulum stress. J. Diabetes Its Complicat. 2016, 30, 573–579. [Google Scholar] [CrossRef]
  114. Ruan, B.; Duan, J.L.; Xu, H.; Tao, K.S.; Han, H.; Dou, G.R.; Wang, L. Capillarized Liver Sinusoidal Endothelial Cells Undergo Partial Endothelial-Mesenchymal Transition to Actively Deposit Sinusoidal ECM in Liver Fibrosis. Front. Cell Dev. Biol. 2021, 9, 671081. [Google Scholar] [CrossRef]
  115. Ribera, J.; Pauta, M.; Melgar-Lesmes, P.; Córdoba, B.; Bosch, A.; Calvo, M.; Rodrigo-Torres, D.; Sancho-Bru, P.; Mira, A.; Jiménez, W.; et al. A small population of liver endothelial cells undergoes endothelial-to-mesenchymal transition in response to chronic liver injury. Am. J. Physiol.-Gastrointest. Liver Physiol. 2017, 313, G492–G504. [Google Scholar] [CrossRef] [PubMed]
  116. Yamashita, M.; Aoki, H.; Hashita, T.; Iwao, T.; Matsunaga, T. Inhibition of transforming growth factor beta signaling pathway promotes differentiation of human induced pluripotent stem cell-derived brain microvascular endothelial-like cells. Fluids Barriers CNS 2020, 17, 36. [Google Scholar] [CrossRef] [PubMed]
  117. Brenner, D.A.; Kisseleva, T.; Scholten, D.; Paik, Y.H.; Iwaisako, K.; Inokuchi, S.; Schnabl, B.; Seki, E.; De Minicis, S.; Oesterreicher, C.; et al. Origin of myofibroblasts in liver fibrosis. Fibrogenesis Tissue Repair. Fibrogenesis Tissue Repair. 2012, 5 (Suppl. 1), S17. [Google Scholar] [CrossRef]
  118. Dong, W.; Kong, M.; Zhu, Y.; Shao, Y.; Wu, D.; Lu, J.; Guo, J.; Xu, Y. Activation of TWIST Transcription by Chromatin Remodeling Protein BRG1 Contributes to Liver Fibrosis in Mice. Front. Cell Dev. Biol. 2020, 8, 340. [Google Scholar] [CrossRef]
  119. Derada Troletti, C.; Fontijn, R.D.; Gowing, E.; Charabati, M.; van Het Hof, B.; Didouh, I.; van der Pol, S.M.A.; Geerts, D.; Prat, A.; van Horssen, J.; et al. Inflammation-induced endothelial to mesenchymal transition promotes brain endothelial cell dysfunction and occurs during multiple sclerosis pathophysiology. Cell Death Dis. 2019, 10, 45. [Google Scholar] [CrossRef] [PubMed]
  120. Potenta, S.; Zeisberg, E.; Kalluri, R. The role of endothelial-to-mesenchymal transition in cancer progression. Br. J. Cancer 2008, 99, 1375–1379. [Google Scholar] [CrossRef]
  121. Li, Z.X.; Chenm, J.X.; Zheng, Z.J.; Cai, W.J.; Yang, X.B.; Huang, Y.Y.; Gong, Y.; Xu, F.; Chen, Y.S.; Lin, L. TGF-β1 promotes human breast cancer angiogenesis and malignant behavior by regulating endothelial-mesenchymal transition. Front. Oncol. 2022, 12, 1051148. [Google Scholar] [CrossRef]
  122. Kovacic, J.C.; Mercader, N.; Torres, M.; Boehm, M.; Fuster, V. Epithelial- and Endothelial- to Mesenchymal Transition: From Cardiovascular Development to Disease. Circulation 2012, 125, 1795–1808. [Google Scholar] [CrossRef]
  123. Islam, S.; Boström, K.I.; Di Carlo, D.; Simmons, C.A.; Tintut, Y.; Yao, Y.; Hsu, J.J. The Mechanobiology of Endothelial-to-Mesenchymal Transition in Cardiovascular Disease. Front. Physiol. 2021, 12, 734215. [Google Scholar] [CrossRef]
  124. Tombor, L.S.; John, D.; Glaser, S.F.; Luxán, G.; Forte, E.; Furtado, M.; Rosenthal, N.; Baumgarten, N.; Schulz, M.H.; Wittig, J.; et al. Single cell sequencing reveals endothelial plasticity with transient mesenchymal activation after myocardial infarction. Nat. Commun. 2021, 12, 681. [Google Scholar] [CrossRef]
  125. Mimouni, M.; Lajoix, A.D.; Desmetz, C. Experimental Models to Study Endothelial to Mesenchymal Transition in Myocardial Fibrosis and Cardiovascular Diseases. Int. J. Mol. Sci. 2023, 25, 382. [Google Scholar] [CrossRef] [PubMed]
  126. Duval, K.; Grover, H.; Han, L.H.; Mou, Y.; Pegoraro, A.F.; Fredberg, J.; Chen, Z. Modeling Physiological Events in 2D vs. 3D Cell Culture. Physiology 2017, 32, 266–277. [Google Scholar] [CrossRef] [PubMed]
  127. Karthika, C.L.; Venugopal, V.; Sreelakshmi, B.J.; Krithika, S.; Thomas, J.M.; Abraham, M.; Kartha, C.C.; Rajavelu, A.; Sumi, S. Oscillatory shear stress modulates Notch-mediated endothelial mesenchymal plasticity in cerebral arteriovenous malformations. Cell. Mol. Biol. Lett. 2023, 28, 22. [Google Scholar] [CrossRef]
  128. Montañez, E.; Casaroli-Marano, R.P.; Vilaró, S.; Pagan, R. Comparative study of tube assembly in three-dimensional collagen matrix and on Matrigel coats. Angiogenesis 2002, 5, 167–172. [Google Scholar] [CrossRef]
  129. Naderi-Meshkin, H.; Cornelius, V.A.; Eleftheriadou, M.; Potel, K.N.; Setyaningsih, W.A.W.; Margariti, A. Vascular organoids: Unveiling advantages, applications, challenges, and disease modelling strategies. Stem Cell Res. Ther. 2023, 14, 292. [Google Scholar] [CrossRef]
  130. Zhao, X.; Xu, Z.; Xiao, L.; Shi, T.; Xiao, H.; Wang, Y.; Li, Y.; Xue, F.; Zeng, W. Review on the Vascularization of Organoids and Organoids-on-a-Chip. Front. Bioeng. Biotechnol. 2021, 9, 637048. [Google Scholar] [CrossRef]
  131. de Haan, L.; Suijker, J.; van Roey, R.; Berges, N.; Petrova, E.; Queiroz, K.; Strijker, W.; Olivier, T.; Poeschke, O.; Garg, S.; et al. A Microfluidic 3D Endothelium-on-a-Chip Model to Study Transendothelial Migration of T Cells in Health and Disease. Int. J. Mol. Sci. 2021, 22, 8234. [Google Scholar] [CrossRef]
  132. Mandrycky, C.J.; Howard, C.C.; Rayner, S.G.; Shin, Y.J.; Zheng, Y. Organ-on-a-chip systems for vascular biology. J. Mol. Cell. Cardiol. 2021, 159, 1–13. [Google Scholar] [CrossRef]
  133. Kisanuki, Y.Y.; Hammer, R.E.; Miyazaki, J.; Williams, S.C.; Richardson, J.A.; Yanagisawa, M. Tie2-Cre transgenic mice: A new model for endothelial cell-lineage analysis in vivo. Dev. Biol. 2001, 230, 230–242. [Google Scholar] [CrossRef]
  134. Monvoisin, A.; Alva, J.A.; Hofmann, J.J.; Zovein, A.C.; Lane, T.F.; Iruela-Arispe, M.L. VE-cadherin-CreERT2 transgenic mouse: A model for inducible recombination in the endothelium. Dev. Dyn. 2006, 235, 3413–3422. [Google Scholar] [CrossRef]
  135. Madisen, L.; Zwingman, T.A.; Sunkin, S.M.; Oh, S.W.; Zariwala, H.A.; Gu, H.; Ng, L.L.; Palmiter, R.D.; Hawrylycz, M.J.; Jones, A.R.; et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 2010, 13, 133–140. [Google Scholar] [CrossRef]
  136. Dessalles, C.A.; Leclech, C.; Castagnino, A.; Barakat, A.I. Integration of substrate- and flow-derived stresses in endothelial cell mechanobiology. Commun. Biol. 2021, 4, 764. [Google Scholar] [CrossRef] [PubMed]
  137. Ou, S.; Kim, T.Y.; Jung, E.; Shin, S.Y. p38 mitogen-activated protein kinase contributes to TNFα-induced endothelial tube formation of bone-marrow-derived mesenchymal stem cells by activating the JAK/STAT/TIE2 signaling axis. BMB Rep. 2024, 57, 238–243. [Google Scholar] [CrossRef]
  138. Mostaço-Guidolin, L.; Rosin, N.L.; Hackett, T.L. Imaging Collagen in Scar Tissue: Developments in Second Harmonic Generation Microscopy for Biomedical Applications. Int. J. Mol. Sci. 2017, 18, 1772. [Google Scholar] [CrossRef]
  139. Zeng, H.; Chen, J.X. Microvascular Rarefaction and Heart Failure With Preserved Ejection Fraction. Front. Cardiovasc. Med. 2019, 6, 15. [Google Scholar] [CrossRef]
  140. Chytil, A.; Magnuson, M.A.; Wright, C.V.; Moses, H.L. Conditional inactivation of the TGF-beta type II receptor using Cre:Lox. Genesis 2002, 32, 73–75. [Google Scholar] [CrossRef]
  141. Kong, P.; Shinde, A.V.; Su, Y.; Russo, I.; Chen, B.; Saxena, A.; Conway, S.J.; Graff, J.M.; Frangogiannis, N.G. Opposing Actions of Fibroblast and Cardiomyocyte Smad3 Signaling in the Infarcted Myocardium. Circulation 2018, 137, 707–724. [Google Scholar] [CrossRef] [PubMed]
  142. Zhao, J.; Shi, W.; Wang, Y.L.; Chen, H.; Bringas, P., Jr.; Datto, M.B.; Frederick, J.P.; Wang, X.F.; Warburton, D. Smad3 deficiency attenuates bleomycin-induced pulmonary fibrosis in mice. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2002, 282, L585–L593. [Google Scholar] [CrossRef]
  143. Choe, C.P.; Choi, S.Y.; Kee, Y.; Kim, M.J.; Kim, S.H.; Lee, Y.; Park, H.C.; Ro, H. Transgenic fluorescent zebrafish lines that have revolutionized biomedical research. Lab. Anim. Res. 2021, 37, 26. [Google Scholar] [CrossRef]
  144. Okuda, K.S.; Keyser, M.S.; Gurevich, D.B.; Sturtzel, C.; Mason, E.A.; Paterson, S.; Chen, H.; Scott, M.; Condon, N.D.; Martin, P.; et al. Live-imaging of endothelial Erk activity reveals dynamic and sequential signalling events during regenerative angiogenesis. eLife 2021, 10, e62196. [Google Scholar] [CrossRef]
  145. Ryan, R.; Moyse, B.R.; Richardson, R.J. Zebrafish cardiac regeneration-looking beyond cardiomyocytes to a complex microenvironment. Histochem. Cell Biol. 2020, 154, 533–548. [Google Scholar] [CrossRef]
  146. Mahmutovic Persson, I.; Falk Håkansson, H.; Örbom, A.; Liu, J.; von Wachenfeldt, K.; Olsson, L.E. Imaging Biomarkers and Pathobiological Profiling in a Rat Model of Drug-Induced Interstitial Lung Disease Induced by Bleomycin. Front. Physiol. 2020, 11, 584. [Google Scholar] [CrossRef]
  147. Madisen, L.; Garner, A.R.; Shimaoka, D.; Chuong, A.S.; Klapoetke, N.C.; Li, L.; van der Bourg, A.; Niino, Y.; Egolf, L.; Monetti, C.; et al. Transgenic mice for intersectional targeting of neural sensors and effectors with high specificity and performance. Neuron 2015, 85, 942–958. [Google Scholar] [CrossRef] [PubMed]
  148. Mayr, C.H.; Santacruz, D.; Jarosch, S.; Bleck, M.; Dalton, J.; McNabola, A.; Lempp, C.; Neubert, L.; Rath, B.; Kamp, J.C.; et al. Spatial transcriptomic characterization of pathologic niches in IPF. Sci. Adv. 2024, 10, eadl5473. [Google Scholar] [CrossRef]
  149. Luecken, M.D.; Theis, F.J. Current best practices in single-cell RNA-seq analysis: A tutorial. Mol. Syst. Biol. 2019, 15, e8746. [Google Scholar] [CrossRef] [PubMed]
  150. van den Brink, S.C.; Sage, F.; Vértesy, Á.; Spanjaard, B.; Peterson-Maduro, J.; Baron, C.S.; Robin, C.; van Oudenaarden, A. Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations. Nat. Methods 2017, 14, 935–936. [Google Scholar] [CrossRef]
  151. Haque, A.; Engel, J.; Teichmann, S.A.; Lönnberg, T. A practical guide to single-cell RNA-sequencing for biomedical research and clinical applications. Genome Med. 2017, 9, 75. [Google Scholar] [CrossRef]
  152. Halder, S.K.; Beauchamp, R.D.; Datta, P.K. A Specific Inhibitor of TGF-β Receptor Kinase, SB-431542, as a Potent Antitumor Agent for Human Cancers. Neoplasia 2005, 7, 509–521. [Google Scholar] [CrossRef]
  153. Herbertz, S.; Sawyer, J.S.; Stauber, A.J.; Gueorguieva, I.; Driscoll, K.E.; Estrem, S.T.; Cleverly, A.L.; Desaiah, D.; Guba, S.C.; Benhadji, K.A.; et al. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des. Dev. Ther. 2015, 9, 4479–4499. [Google Scholar] [CrossRef]
  154. Melisi, D.; Garcia-Carbonero, R.; Macarulla, T.; Pezet, D.; Deplanque, G.; Fuchs, M.; Trojan, J.; Oettle, H.; Kozloff, M.; Cleverly, A.; et al. Galunisertib plus gemcitabine vs. gemcitabine for first-line treatment of patients with unresectable pancreatic cancer. Br. J. Cancer 2018, 119, 1208–1214. [Google Scholar] [CrossRef]
  155. Murphy, P.A.; Lam, M.T.; Wu, X.; Kim, T.N.; Vartanian, S.M.; Bollen, A.W.; Carlson, T.R.; Wang, R.A. Endothelial Notch4 signaling induces hallmarks of brain arteriovenous malformations in mice. Proc. Natl. Acad. Sci. USA 2008, 105, 10901–10906. [Google Scholar] [CrossRef]
  156. Li, C.; Dong, F.; Jia, Y.; Du, H.; Dong, N.; Xu, Y.; Wang, S.; Wu, H.; Liu, Z.; Li, W. Notch signal regulates corneal endothelial-to-mesenchymal transition. Am. J. Pathol. 2013, 183, 786–795. [Google Scholar] [CrossRef]
  157. Milan, M.; Pace, V.; Maiullari, F.; Chirivì, M.; Baci, D.; Maiullari, S.; Madaro, L.; Maccari, S.; Stati, T.; Marano, G.; et al. Givinostat reduces adverse cardiac remodeling through regulating fibroblasts activation. Cell Death Dis. 2018, 9, 108. [Google Scholar] [CrossRef] [PubMed]
  158. Glover, E.K.; Jordan, N.; Sheerin, N.S.; Ali, S. Regulation of Endothelial-to-Mesenchymal Transition by microRNAs in Chronic Allograft Dysfunction. Transplantation 2019, 103, e64–e73. [Google Scholar] [CrossRef] [PubMed]
  159. Kang, I.S.; Kwon, K. Potential application of biomimetic exosomes in cardiovascular disease: Focused on ischemic heart disease. BMB Rep. 2022, 55, 30–38. [Google Scholar] [CrossRef] [PubMed]
  160. Liu, G.W.; Guzman, E.B.; Menon, N.; Langer, R.S. Lipid Nanoparticles for Nucleic Acid Delivery to Endothelial Cells. Pharm. Res. 2023, 40, 3–25. [Google Scholar] [CrossRef]
  161. Dosta, P.; Tamargo, I.; Ramos, V.; Kumar, S.; Kang, D.W.; Borrós, S.; Jo, H. Delivery of Anti-miR-712 to Inflamed Endothelial Cells Using Poly(β-amino ester) Nanoparticles Conjugated with VCAM-1 Targeting Peptide. Adv. Healthc. Mater. 2021, 10, e2001894. [Google Scholar] [CrossRef]
  162. Parhiz, H.; Shuvaev, V.V.; Pardi, N.; Khoshnejad, M.; Kiseleva, R.Y.; Brenner, J.S.; Uhler, T.; Tuyishime, S.; Mui, B.L.; Tam, Y.K.; et al. PECAM-1 directed re-targeting of exogenous mRNA providing two orders of magnitude enhancement of vascular delivery and expression in lungs independent of apolipoprotein E-mediated uptake. J. Control. Release 2018, 291, 106–115. [Google Scholar] [CrossRef]
  163. Rice, L.M.; Padilla, C.M.; McLaughlin, S.R.; Mathes, A.; Ziemek, J.; Goummih, S.; Nakerakanti, S.; York, M.; Farina, G.; Whitfield, M.L.; et al. Fresolimumab treatment decreases biomarkers and improves clinical symptoms in systemic sclerosis patients. J. Clin. Investig. 2015, 125, 2795–2807. [Google Scholar] [CrossRef]
  164. Tripska, K.; Igreja Sá, I.C.; Vasinova, M.; Vicen, M.; Havelek, R.; Eissazadeh, S.; Svobodova, Z.; Vitverova, B.; Theuer, C.; Bernabeu, C.; et al. Monoclonal anti-endoglin antibody TRC105 (carotuximab) prevents hypercholesterolemia and hyperglycemia-induced endothelial dysfunction in human aortic endothelial cells. Front. Med. 2022, 9, 845918. [Google Scholar] [CrossRef] [PubMed]
  165. Schoonderwoerd, M.J.A.; Goumans, M.T.H.; Hawinkels, L.J.A.C. Endoglin: Beyond the Endothelium. Biomolecules 2020, 10, 289. [Google Scholar] [CrossRef]
  166. Danielpour, D. Advances and Challenges in Targeting TGF-β Isoforms for Therapeutic Intervention of Cancer: A Mechanism-Based Perspective. Pharmaceuticals 2024, 17, 533. [Google Scholar] [CrossRef]
  167. Man, S.; Sanchez Duffhues, G.; Ten Dijke, P.; Baker, D. The therapeutic potential of targeting the endothelial-to-mesenchymal transition. Angiogenesis 2019, 22, 3–13. [Google Scholar] [CrossRef]
  168. Giordo, R.; Nasrallah, G.K.; Posadino, A.M.; Galimi, F.; Capobianco, G.; Eid, A.H.; Pintus, G. Resveratrol-Elicited PKC Inhibition Counteracts NOX-Mediated Endothelial to Mesenchymal Transition in Human Retinal Endothelial Cells Exposed to High Glucose. Antioxidants 2021, 10, 224. [Google Scholar] [CrossRef] [PubMed]
  169. Chen, X.; Chen, X.; Shi, X.; Gao, Z.; Guo, Z. Curcumin attenuates endothelial cell fibrosis through inhibiting endothelial–interstitial transformation. Clin. Exp. Pharmacol. Physiol. 2020, 47, 1182–1192. [Google Scholar] [CrossRef]
  170. Pera, R.; Gambhir, S.S.; Cooke, J.P. Endothelial cells derived from human iPSCs increase capillary density and improve perfusion in a mouse model of peripheral arterial disease. Arterioscler. Thromb. Vasc. Biol. 2011, 31, e72–e79. [Google Scholar]
  171. Cheng, Y.C.; Hsieh, M.L.; Lin, C.J.; Chang, C.M.C.; Huang, C.Y.; Puntney, R.; Wu Moy, A.; Ting, C.Y.; Herr Chan, D.Z.; Nicholson, M.W.; et al. Combined Treatment of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes and Endothelial Cells Regenerate the Infarcted Heart in Mice and Non-Human Primates. Circulation 2023, 148, 1395–1409. [Google Scholar] [CrossRef]
  172. Brandt, C.B.; Fonager, S.V.; Haskó, J.; Helmig, R.B.; Degn, S.; Bolund, L.; Jessen, N.; Lin, L.; Luo, Y. HIF1A Knockout by Biallelic and Selection-Free CRISPR Gene Editing in Human Primary Endothelial Cells with Ribonucleoprotein Complexes. Biomolecules 2022, 13, 23. [Google Scholar] [CrossRef]
  173. Lazovic, B.; Nguyen, H.T.; Ansarizadeh, M.; Wigge, L.; Kohl, F.; Li, S.; Carracedo, M.; Kettunen, J.; Krimpenfort, L.; Elgendy, R.; et al. Human iPSC and CRISPR targeted gene knock-in strategy for studying the somatic TIE2L914F mutation in endothelial cells. Angiogenesis 2024, 27, 523–542, Erratum in Angiogenesis 2024, 27, 543–544. [Google Scholar] [CrossRef]
  174. Lovisa, S.; Fletcher-Sananikone, E.; Sugimoto, H.; Hensel, J.; Lahiri, S.; Hertig, A.; Taduri, G.; Lawson, E.; Dewar, R.; Revuelta, I.; et al. Endothelial-to-mesenchymal transition compromises vascular integrity to induce Myc-mediated metabolic reprogramming in kidney fibrosis. Sci. Signal. 2020, 13, eaaz2597. [Google Scholar] [CrossRef]
  175. Marzolini, N.; Brysgel, T.V.; Essien, E.O.; Rahman, R.J.; Wu, J.; Majumder, A.; Patel, M.N.; Tiwari, S.; Espy, C.L.; Shuvaev, V.V.; et al. Targeting DNA-LNPs to Endothelial Cells Improves Expression Magnitude, Duration, and Specificity. bioRxiv 2025. [Google Scholar] [CrossRef]
  176. Hall, I.F.; Kishta, F.; Xu, Y.; Baker, A.H.; Kovacic, J.C. Endothelial to mesenchymal transition: At the axis of cardiovascular health and disease. Cardiovasc. Res. 2024, 120, 223–236. [Google Scholar] [CrossRef]
  177. Wimmer, R.A.; Leopoldi, A.; Aichinger, M.; Wick, N.; Hantusch, B.; Novatchkova, M.; Taubenschmid, J.; Hämmerle, M.; Esk, C.; Bagley, J.A.; et al. Human blood vessel organoids as a model of diabetic vasculopathy. Nature 2019, 565, 505–510. [Google Scholar] [CrossRef] [PubMed]
  178. Shakeri, A.; Wang, Y.; Zhao, Y.; Landau, S.; Perera, K.; Lee, J.; Radisic, M. Engineering organ-on-a-chip systems for vascular diseases. Arterioscler. Thromb. Vasc. Biol. 2023, 43, 2241–2255. [Google Scholar] [CrossRef] [PubMed]
  179. Dao, L.; You, Z.; Lu, L.; Xu, T.; Sarkar, A.K.; Zhu, H.; Liu, M.; Calandrelli, R.; Yoshida, G.; Lin, P.; et al. Modeling blood-brain barrier formation and cerebral cavernous malformations in human PSC-derived organoids. Cell Stem Cell 2024, 31, 818–833.e11. [Google Scholar] [CrossRef]
  180. Cheroni, C.; Trattaro, S.; Caporale, N.; López-Tobón, A.; Tenderini, E.; Sebastiani, S.; Troglio, F.; Gabriele, M.; Bressan, R.B.; Pollard, S.M.; et al. Benchmarking brain organoid recapitulation of fetal corticogenesis. Transl. Psychiatry 2022, 12, 520. [Google Scholar] [CrossRef]
  181. Fang, J.S.; Hultgren, N.W.; Hughes, C.C.W. Regulation of Partial and Reversible Endothelial-to-Mesenchymal Transition in Angiogenesis. Front. Cell Dev. Biol. 2021, 9, 702021. [Google Scholar] [CrossRef]
  182. Cong, X.; Zhang, Z.; Li, H.; Yang, Y.G.; Zhang, Y.; Sun, T. Nanocarriers for targeted drug delivery in the vascular system: Focus on endothelium. J. Nanobiotechnol. 2024, 22, 620. [Google Scholar] [CrossRef] [PubMed]
  183. Tie, Y.; Tang, F.; Peng, D.; Zhang, Y.; Shi, H. TGF-beta signal transduction: Biology, function and therapy for diseases. Mol. Biomed. 2022, 3, 45. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of EndMT from endothelial identity to mesenchymal-like state across organs. Abbreviations: EndMT, endothelial-to-mesenchymal transition; EC, endothelial cell; MI, myocardial infarction; ECM, extracellular matrix; IPF, idiopathic pulmonary fibrosis; PAH, pulmonary arterial hypertension; HIF, hypoxia-inducible factor; AngII, angiotensin II; ROS, reactive oxygen species. Symbols/arrows: ↑ indicate increased expression/activity; → indicate “leads to/induces”. The central downward/upward arrow indicates progression from an endotelilal to a mesenchymal-like phenotype during EndMT and, where applicable, phenotypic plasticity/partial reversion toward an endothelial-like state, respectively.
Figure 1. Overview of EndMT from endothelial identity to mesenchymal-like state across organs. Abbreviations: EndMT, endothelial-to-mesenchymal transition; EC, endothelial cell; MI, myocardial infarction; ECM, extracellular matrix; IPF, idiopathic pulmonary fibrosis; PAH, pulmonary arterial hypertension; HIF, hypoxia-inducible factor; AngII, angiotensin II; ROS, reactive oxygen species. Symbols/arrows: ↑ indicate increased expression/activity; → indicate “leads to/induces”. The central downward/upward arrow indicates progression from an endotelilal to a mesenchymal-like phenotype during EndMT and, where applicable, phenotypic plasticity/partial reversion toward an endothelial-like state, respectively.
Ijms 26 11724 g001
Table 1. EndMT inducers, pathways, and disease contexts.
Table 1. EndMT inducers, pathways, and disease contexts.
Inducer/
Stimulus
Major
Pathway
Key TFs/
Regulators
Core
Readouts
Disease
Context
References
TGF-βTβR1→SMAD2/3;
PI3K/AKT, p38/JNK
SNAI1/2
TWIST1
ZEB1/2
↓CD31/VE-cadherin;
↑α-SMA, FN1, COL1A1
Cardiac fibrosis and valvular disease, aortic aneurysms, renal and
pulmonary fibrosis
[8,15,28,29,30,31,32,33,34,35]
TNF-α/IL-1βNF-κB, MAPKsNF-κB
(p65)
Barrier loss (TEER↓),
Leukocyte adhesion↑;
Synergizes with TGF-β to promote EndMT
Inflammatory
vasculopathies,
atherosclerosis
[3,5,7]
Disturbed/
oscillatory shear
Notch1,
JAG-NOTCH4;
DNMT1-mediated KLF4 promoter
methylation
TWIST1
NOTCH1/4
DNMT1
EndMT program↑
EC identity↓
Atherosclerosis[2,20,36,37,38,39]
HypoxiaHIF-1α→TIWST1-PDGFB axisHIF-1α
TWIST1
EndMT markers↑
(α-SMA, FN1, SNAI1/2)
PAH,
lung remodeling
[10,40,41,42]
Epigenetic
regulation
HDAC9/HDAC3;DNMT1; JMJD2B;EZH2/H3K27me3HDAC9/3
DNMT1
JMJD2B
EZH2
H3K27me3 changes,
KLF4 promoter
methylation↑,
miR-29c silencing
Atherosclerosis,
Neointima,
Fibrosis
[22,23,24,43,44,45,46]
miR-200 familyDirect targeting of ZEB1/2→EMT/EndMT suppressionmiR-200a/b/cZEB1/2↓,
Maintenance of EC
Identity
Diabetic complications,
Fibrosis
[47,48]
miR-21Represses SMAD inhibitor→strengthens TGF-β miR-21EndMT markers↑,
anti-miR-21 attenuates EndMT
Perivascular/
Cardiac fibrosis
[49,50]
Wnt/β-catenin
(Therapy)
Inhibition of β-catenin/CBP transcriptional complex-EndMT suppression,
Improved vascular
Remodeling
Pulmonary fibrosis,
Vasculopathy
[51,52,53]
Small molecules/
natural products(Therapy)
Rho-kinase/FAK↓-EndMT↓,
Amelioration of fibrosis/PAH
Pulmonary fibrosis,
PAH
[54,55,56]
Note: Arrows indicate direction of change relative to baseline ECs (↑up-regulated/increased; ↓down-regulated/decreased). The “→” symbol denoted a directional signaling/cause-effect relationship (activates/induces/leads to) between components. “-” is not applicable. Listed references are representative rather than exhaustive and were selected based on the inclusion/exclusion criteria.
Table 2. Key methodological platform for EndMT studies: strengths, limitations, and standardization considerations.
Table 2. Key methodological platform for EndMT studies: strengths, limitations, and standardization considerations.
PlatformMajor StrengthsKey Limitations/PitfallsMinimal Reporting/
QC Items
References
2D EC monolayersHigh control, scalable,
Convenient/reproducible
Baseline
Limited tissue context
Over-simplify transient/partial states
Cell source/passage; inducer dose/time; marker panel; replicate design[125,126]
MechnochemicalmodelsMimics shear/stiffness-
driven programs
Device-to-device variability; sensitivity to setup; lab-to-lab reproducibility issuesFlow/shear parameters; substrate stiffness; calibration method[38,127]
2D ECMCell–matrix interaction,
Structural remodeling
Matrix batch variability; limited cellular heterogeneityMatrix composition/concentration; gel protocol; imaging quantification[126,128]
iPSC-vascular organoidsMulticellular crosstalk,
More physiological
Gradients
Batch-to-batch variability; maturation state affects specificityDifferentiation QC; cell composition metrics; batch controls[129,130]
Organ-on-a-chip/microfluidicsControlled shear+
Real-time readouts
Fabrication/operation variability; throughput constraintsDevice specs; shear profiles; barrier readouts; standardized operating protocol[131,132]
In vivo disease modelsSystem-level context,
Causality testing
Species differences; model-specific confoundersModel details; time-course; endpoints; randomization/blinding if used[125]
Endothelial lineagetracingLineage-informed “contribution” evidenceRecombination specificity/efficiency; time/dose confoundsDriver line; induction regimen; recombination efficiency controls[133,134,135]
Marker panelsWidely accessible; quantitative protein-level callsSpecificity limited by marker overlap; single-marker false positivesDefine “program-level” criteria; recombination efficiency controls[8,9,88]
Tissue cleaningWhole-organ 3D localization/quantificationAntibody penetration; signal-to-noise impacts sensitivity; protocol-dependentClearing protocol; imaging settings; segmentation method; validation modality[14]
scRNA-seq/scATAC-seqUnbiased state discovery; regulatory inferenceDropout/dissociation bias affects sensitivity; snapshot→trajectory inferenceCell numbers; QC metrics; batch correction; signature definitions[37,92,124]
Spatial transcriptomicsNiche localization of putative transitionsResolution limits; transcript capture affects sensitivity; snapshot inferencePlatform/resolution; tissue handling; normalization; validation markers[91,93]
Note: Sensitivity/specificity refer to the confidence of EndMT calls; the symbol “→” denotes a directional transition of inference; transient/partial states are often rare and time-dependent; reproducibility requires standardized reporting of protocols, QC metrics, and EndMT signature/threshold definitions.
Table 3. EndMT-targeted interventions.
Table 3. EndMT-targeted interventions.
Strategy/MechanismRepresentative AgentsModel &
Readouts
EffectReferences
Pathway inhibitors:TβR1/canonical SMADSB-431542, A-83-01;
Galunisertib
ECs; MI/IPF/PAH models; marker panelEndMT markers↓,
migration↓;
fibrosis burden↓
[8,28,32,145,146,147,148,160]
Non-canonical:Rho/ROCK, FAK, PI3K/AKT, MAPKROCK/FAK inhibitorsAtheroprone-flow ECs; lung vascular modelsStress fiber↓,
mesenchymal
program↓
[54,55,56]
Epigenetic:HDAC/DNMT/EZH2/JMJD2BHDAC inhibitors;
5-aza-2′-deoxycytidine
In vitro ECs;
fibrosis models
Chromatin
re-opening→EndMT↓
[22,23,24,49,53,55,56]
miRNA therapeutics:restore anti-EndMT/block pro-miRsmiR-200b mimic; anti-miR-21 (LNP/exosome)ECs, iPSC-ECs;
injury models
ZEB1/2↓; EndMT↓;
maintain EC identity
[47,58,63,84,157,158,159]
Biologics:anti-TGF-β/endoglinaxismAbs/ligand traps
(context-dependent)
Fibrosis/PAH modelsSMAD-driven
EndMT↓
[8,17,18,160,162]
Cell/Gene:iPSC-ECs; CRISPR editsSNAI/ZEB KO;
miR-cassettes KI
Graft stability assays;
in vivo repair
EndMT resistance↑,
preserve function
[72,128,129,130,131,132,133,138,139,170,171,172,173]
Natural products/small molecules:multi-pathwayResveratrol; CurcuminEC EndMT assays;
fibrosis models
ROS/NF-κB↓;EndMT↓[168,169]
Note: Arrows indicate direction of change relative to baseline ECs (↑ up-regulated/increased; ↓ down-regulated/decreased).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, R.; Chang, W. Endothelial-to-Mesenchymal Transition in Health and Disease: Molecular Insights and Therapeutic Implications. Int. J. Mol. Sci. 2025, 26, 11724. https://doi.org/10.3390/ijms262311724

AMA Style

Kim R, Chang W. Endothelial-to-Mesenchymal Transition in Health and Disease: Molecular Insights and Therapeutic Implications. International Journal of Molecular Sciences. 2025; 26(23):11724. https://doi.org/10.3390/ijms262311724

Chicago/Turabian Style

Kim, Ran, and Woochul Chang. 2025. "Endothelial-to-Mesenchymal Transition in Health and Disease: Molecular Insights and Therapeutic Implications" International Journal of Molecular Sciences 26, no. 23: 11724. https://doi.org/10.3390/ijms262311724

APA Style

Kim, R., & Chang, W. (2025). Endothelial-to-Mesenchymal Transition in Health and Disease: Molecular Insights and Therapeutic Implications. International Journal of Molecular Sciences, 26(23), 11724. https://doi.org/10.3390/ijms262311724

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop