Endothelial cells (ECs) migrate, sprout, and proliferate in response to (lymph)angiogenic mitogens, such as vascular endothelial growth factors. When ECs reach high confluency and encounter spatial confinement, they establish mature cell–cell junctions, reduce proliferation, and enter a quiescent state through a process known as contact inhibition. However, EC quiescence is modulated not only by spatial confinement but also by other mechano-environmental factors, including blood or lymph flow and extracellular matrix properties. Changes in physical forces and intracellular signaling can disrupt contact inhibition, resulting in aberrant proliferation and vascular dysfunction. Therefore, it is critical to understand the mechanisms by which endothelial cells regulate contact inhibition. While contact inhibition has been well studied in blood endothelial cells (BECs), its regulation in lymphatic endothelial cells (LECs) remains largely unexplored. Here, we review the current knowledge on extrinsic stimuli and intrinsic molecular pathways that govern endothelial contact inhibition and highlight nuanced differences between BECs and LECs. Furthermore, we provide perspectives for future research on lymphatic contact inhibition. A deeper understanding of the BEC and LEC-specific pathways underlying contact inhibition may enable targeted modulation of this process in blood or lymphatic vessels with relevance to lymphatic or blood vascular-specific disorders.

The term ‘contact inhibition’ was first introduced in the 1950s when Abercrombie and Heaysman observed that fibroblast-like cells derived from chick heart explants altered their direction of movement to avoid colliding with other cells in a two-dimensional (2D) culture [1]. This phenomenon, referred to as contact inhibition of locomotion, facilitated the maintenance of a cell monolayer architecture by preventing cells from overgrowing one another [1]. A decade later, the term ‘contact inhibition’ was also adopted to describe the cell cycle arrest observed for many cell types in culture, a process termed contact inhibition of proliferation [2]. Although initially studied in in vitro cell cultures of fibroblasts [3], epithelial cells [4-6], endothelial cells (ECs) [7-9], or cardiomyocytes [10], contact inhibition has been demonstrated to be a fundamental mechanism in tissue growth and development [11-14], as well as wound healing and homeostasis [15,16]. As early as 1967, it was demonstrated that melanoma cells maintained exponential colony growth rates, whereas fibroblasts shifted to a linear growth phase as cell density increased [17]. Concordantly, the loss of contact inhibition was recognized as a hallmark of cancer cells in tumors [18].

Cell cycle regulation

Contact inhibition is intrinsically linked to cell cycle regulation, as increased cell density induces arrest in the gap 0 (G0)/gap 1 (G1) phase, effectively preventing further progression through the cell cycle. The cell cycle is historically divided into G1, DNA synthesis (S), gap 2 (G2) – together forming the interphase – and mitosis (M) phases. Cell cycle transitions from G1 to S phase, as well as from G2 to M phase, are tightly controlled by cyclins and cyclin-dependent kinase (CDK) complexes [19]. Specifically, cyclin D-CDK4/CDK6 regulates G1 phase progression, the cyclin E-CDK2 complex modulates the transition from G1 to S phase, cyclin A-CDK2 are involved in S phase, cyclin A-CDK1 regulate G2 phase, and, lastly, cyclin B-CDK1 complex is involved in G2 to M transition [19,20]. Moreover, the transition to the S phase requires further inactivation of the Retinoblastoma (Rb) protein through its phosphorylation by the cyclin D-CDK4/CDK6 complex. This phosphorylation event releases the E2F-DP (dimerization partner) complex, enabling the subsequent expression of cyclins E and A and the progression through the cell cycle [21,22].

Each phase of the cell cycle can be inhibited by cyclin-dependent kinase inhibitors (CDKIs), which are categorized into two families: the Cip/Kip family (or CDKN1 family), comprising p21 (human gene name: CDKN1A) [23], p27 (CDKN1B), and p57 (CDKN1C) [24] and the INK4 family [25], consisting of p16 and p14ARF (or p19ARF in mice, both encoded by CDKN2A and Cdkn2, respectively), as well as p15 (CDKN2B), p18 (CDKN2C), and p19 (CDKN2D). The two families of CDKIs encode distinct proteins with different biochemical and functional characteristics, reflecting their specific roles in cell cycle regulation [26,27]. The Cip/Kip family primarily inhibits CDK1 and CDK2, effectively regulating the cell cycle at various checkpoints [28]. In contrast, the members of the INK4 family predominantly target CDK4 and CDK6, playing a crucial role in controlling the transition from G1 to S phase. Interestingly, global knockout mice for the majority of the Cdkn1 and Cdkn2 genes do not show major developmental or proliferation defects, likely due to genetic redundancy or compensation [29]. Differential organ-specific expression profiles of CDKIs and various post-translational modifications [30,31] further influence their function independently of their transcriptional dynamics.

In addition to the four canonical phases of the cell cycle, cells can also enter the G0 phase. During this phase, inhibitors from the CDKN2 family are typically up-regulated, and transcriptional repression of cell cycle activators occurs, resulting in a state of quiescence and cell cycle arrest[32]. Upon removal of the stimuli that induce cell cycle arrest, cells can exit their quiescent state and reactivate the cell cycle. However, the time required to re-enter the S phase depends on the ‘depth of quiescence’ experienced; the longer the cells remain quiescent, the more time they require to initiate the S phase [33,34]. This relationship highlights the dynamic nature of cellular responses to prolonged inactivity and their capacity for re-entry into active proliferation. It is important to note that quiescence is not synonymous with senescence. In contrast with quiescent cells that retain their potential for reactivation, senescent cells are incapable of responding to mitogenic stimuli and, as a result, cannot re-enter the cell cycle [35].

The concept of contact inhibition

Contact inhibition is established in stages, beginning with an initial increase in cell size that triggers entry into the S phase, leading to subsequent cell proliferation [36]. This increase in cell size must reach a substrate-dependent threshold, highlighting a crucial role of the mechanical microenvironment in the regulation of contact inhibition [36,37]. Substrate stiffening was also shown to sensitize epithelial cells to epidermal growth factor (EGF) as moderate stiffening of the matrix reduced the threshold amount of EGF needed to over-ride contact inhibition by over 100-fold 38.

Subsequently, in a confluent monolayer, contact inhibition occurs following the suppression of cell motility and is triggered when mechanical constraints on local expansion cause cell divisions to reduce individual cell area [39]. Indeed, the mechanical pressure of confinement also plays a crucial role in establishing long-term contact inhibition. This process involves the translocation of β-catenin from cell junctions to the nucleus, indicating that β-catenin mediates the establishment of a pressure threshold that triggers contact inhibition [40].

However, even in confluent monolayer, regions with higher proliferation rates can be identified in both epithelial and EC cultures [41]. This has been correlated with mechanical stimulation caused by the geometry of the environment, such as the corners and edges of cell culture dishes [41]. Interestingly, not only do cells that directly experience mechanical stimuli, such as available space, re-enter the cell cycle, but also cells located farther away from these stimuli also begin to proliferate, indicating that these mechanical cues can be sensed or transmitted over a certain distance from the source [41].

These concepts have been reinforced recently through computational models where tissue growth varies depending on the cell sensitivity to contact inhibition (i.e. intrinsic response to growth factors) and the elastic modulus of the substrate (stiffness) [42]. Mechanical feedback between tissue confinement and individual cell growth enhances cell proliferation at tissue boundaries, while growth in the center mass is suppressed, reflecting experimental observations in epithelial cells previously described [36,41]. Notably, adjusting the model to higher elasticity (stiffness) led to increased bulk growth, in line with the previous notion that many cell types exhibit greater proliferative capacity when exposed to stiffer substrates [43]. Intermediate levels of both contact inhibition and stiffness replicate the growth patterns observed in tissues during development and homeostasis, suggesting a complex interplay between microenvironmental and intrinsic cues [42]. Other modeling approaches demonstrate that, before cells encounter constraints or confinement and when contact inhibition is minimal, cell colonies grow exponentially over time, driven solely by the proliferation rate [44]. Later, the colony boundary moves at a constant speed, determined exclusively by the migration rate of individual cells and independent of the proliferation rate [44].

Moreover, significant differences in cell velocity, age, and stress distributions were observed between non-migrating and migrating cells, as analyzed using a multiphase field model [45]. Importantly, numerical simulations show that, consistent with experimental findings, the loss of contact inhibition is a sufficient mechanism to explain the increase in the proportion of tumor cells [46], demonstrating that the molecular mechanisms that define contact inhibition may constitute druggable targets. Computational models primarily focused on angiogenic processes at the tissue level [47] could potentially be expanded in the future to include modeling of contact inhibition.

Adhesion molecules and the Hippo pathway: key regulators of contact inhibition

Many studies highlight the importance of cell adhesion molecules in the establishment of contact inhibition. For example, in epithelial carcinoma cells, E-cadherin controls an increase in expression of the CDKN1 protein p27 to trigger cell cycle arrest [4]. Notably, nonadherent mouse mammary carcinoma cells transfected with E-cadherin showed increased adhesion within multicellular spheroids and reduced proliferation [4]. In contrast, in HCT116 human colon carcinoma cell line, the CDKI p21 was shown to reduce E-cadherin expression, which was necessary to form multicellular spheroids that can proliferate [48]. Lack of functional p21 and E-cadherin caused proliferation stop and apoptosis [48].

In another model using the SW480 colorectal tumor cell line, it was, however, shown that cells lacking E-cadherin expression rather increased their proliferative potential through the activation of the β-catenin/TCF pathway, demonstrating that not only the actual adhesive function of junctional proteins but also cytoplasmic effectors of cell–cell contacts are involved in the regulation of contact inhibition [49].

Although growth factors are among the major players inducing cell proliferation (reviewed in [50-53]), the cellular response to them may vary depending also on the state of cell–cell adhesion. For example, modulating cell–cell contacts through different substrates or E-cadherin overexpression can induce contact inhibition even at higher EGF concentrations typically permissive for proliferation. This suggests that the threshold required to activate growth factor-mediated cell cycle activity is adjustable and depends on the local balance between growth factor levels and cell–cell contact states [54].

E-cadherin and its associated β-catenin are also responsive to mechanical stress in epithelial cells, with static biaxial stretch leading to nuclear translocation of YAP1 (effector of the Hippo pathway) and β-catenin [55]. While YAP1 remains in the nucleus for only a few hours, β-catenin localizes in the nucleus for up to 24-hour post-stimulus. This suggests that the Hippo pathway is more important in sensing local and transient changes in the mechanical microenvironment and cell–cell contacts and leads to a more rapid response [40,55]. Similarly, high cellular density in epithelial cells or a soft extracellular matrix (ECM) activates the Hippo pathway, increases LATS1/2 kinase activity, and subsequently inactivates YAP1/TAZ [6]. The inactivation or cytosolic translocation of YAP1/TAZ reduces the expression of myosin-II genes and leads to the loss of actin stress fibers, which, in turn, impairs autophagosome activity and reduces proliferation [6]. In line with this, the F-actin-capping/severing proteins Cofilin, CapZ, and Gelsolin have been identified as essential gatekeepers that limit YAP/TAZ activity in epithelial cells experiencing low mechanical stresses during contact inhibition [56]. Furthermore, more recent studies have shown that the activation of YAP1 through the annexin A2 inhibitor PY-60 leads to escape from contact inhibition, allowing keratinocytes to continue proliferating even at high cell density [57].

Blood endothelial cells (BECs) are sensitive to contact inhibition both in vitro and in vivo, where BECs undergo low turnover in large, as well as small, caliber vessels [58,59]. The homeostatic, quiescent state of BECs can be considered that of a confluent monolayer, as mature vessels are lined with an EC monolayer naturally attached in a 2D manner to the underlying ECM [43]. While the exact cell cycle arrest states of different BEC subtypes have not been comprehensively mapped, human umbilical vein ECs (HUVECs), bovine pulmonary arterial ECs, and human corneal ECs have been described to arrest in the G1 phase [60,61]. However, in case of vessel injuries, tumor angiogenesis [62], and in vascular diseases [63], BECs are reactivated to induce proliferation and migration. The shift between the quiescent, contact-inhibited state and the proliferative state in BECs is also driven by growth factors and junctional rearrangements, which activate or repress intracellular pathways [64].

Mitogenic control of blood endothelial cell contact inhibition

For example, the binding of the major angiogenic mitogen vascular endothelial growth factor (VEGF) to VEGF receptor 2 (VEGFR2) activates and internalizes the receptor, triggering EC proliferation to support the expansion of new and tumor-associated blood vessels (reviewed in [65]). Other mitogenic stimuli, such as fibroblast growth factor (FGF) or fetal calf serum, or low-density culture conditions, activate p42/p44 mitogen-activated protein kinase (MAPK), also known as ERK1/2, in BECs and allow them to enter the cell cycle [66], in a manner similarly described for epithelial cells [67] and fibroblasts [68]. Notably, this process can be reversed by the depletion of mitogenic signals and/or an increase in monolayer confluency, demonstrating a finely tuned regulatory mechanism for cell cycle arrest of the blood endothelium [66]. Interestingly, recent work by Pontes-Quero et al. challenges the traditional assumption that increased growth factor concentration, as well as the resulting mitogenic activity, drives both endothelial proliferation and sprouting [69]. Instead, very high mitogenic stimulation induced by VEGF or Notch inhibition actually arrested the proliferation of angiogenic tip cells in the retina [69]. The study identified a bell-shaped dose–response to VEGF and MAPK activity, regulated by Notch and p21, which determines whether BECs sprout/migrate, proliferate, or enter a quiescent state [69]. If and how the adhesive properties of BECs co-determine these cellular responses remains to be explored.

In contact-inhibited BECs, the activity of phosphatases is increased leading to down-regulation of ERK and phosphatidylinositol 3-kinase (PI3 K)/Akt signaling [70]. The process is reversible, as demonstrated by the use of phosphatase inhibitors, which induce cell cycle progression [70].

Junctional control of blood endothelial cell contact inhibition

Vascular endothelial (VE)-cadherin is the major adhesion molecule in blood endothelial cell–cell contacts [71-73]. Similar to epithelial E-cadherin, the absence of VE-cadherin in BECs results in continuous proliferation [7]. Vice versa, high expression of VE-cadherin was associated with reduced activity of VEGFR2 and ERK signaling, as well as increased activity of the high cell density–enhanced protein tyrosine phosphatase 1 (DEP-1)/CD148 [7]. Furthermore, β-catenin-mediated association of VE-cadherin and VEGFR2 was necessary to induce cell cycle arrest [7] and VE-cadherin retained VEGFR2 at the membrane, thereby preventing its internalization into signaling compartments [74]. In contrast with low ERK activity triggered by contact inhibition, shear stress-induced up-regulation of connexin 37 in BECs requires ERK phosphorylation to increase p27 expression and promote G1 arrest [75], indicating that absolute levels of ERK and its activity are not the sole determinators of BEC proliferation.

Another mechanism by which VE-cadherin regulates the cell cycle and contact inhibition in BECs is through phosphorylation of its tyrosine 685 residue and its subsequent interaction with the protein tyrosine kinase C-terminal Src kinase (Csk) [76]. In correlation with cell monolayer density, Csk then transduces the signal of tight cell–cell contact formation intracellularly to induce contact inhibition [76]. While not directly involved in BEC contact inhibition, VE-cadherin-mediated up-regulation of Claudin 5 (CLDN5) is necessary for forming tight junctions in BECs, as demonstrated in in vitro and ex vivo allantois explants. However, during blood vessel homeostasis, CLDN5 is differentially expressed in arteries versus veins [77] and does not control HUVEC barrier properties [78], suggesting its potential vascular bed-specific role in contact inhibition.

Other junctional regulators have been shown to be differentially expressed in sub-confluent versus confluent BEC cultures and, therefore, may play a role in the establishment of contact inhibition. N-cadherin is present at the junctions of sub-confluent BECs, where it is involved in forming the first cell–cell connections, but N-cadherin is excluded from stable cell–cell junctions with increasing monolayer density [79]. ICAM-2 expression, although already present in sub-confluent BECs, was found to control N-cadherin and VE-cadherin recruitment into endothelial junctions through recruitment and activity of ezrin, radixin, and moesin (ERM) and Rac1 proteins to induce contact inhibition [79]. Notably, in vivo deletion of VE-cadherin did not result in N-cadherin localization to junctions [71], suggesting a more complex regulation of the cadherins in vivo with potential consequences for cell cycle states.

Mechanical control of blood endothelial cell contact inhibition

BECs constantly interact with mechanical forces at their luminal side, such as flow-induced shear stress, and on their abluminal side via changes in stiffness and stretch [43]. Recent work has highlighted molecular differences in laminar flow-induced quiescence among BEC types. Arterial ECs exposed to flow initially entered deep quiescence and then transitioned to a shallow homeostatic quiescence, while venous ECs maintained a stable deep quiescence [80]. p27 was essential for flow-mediated quiescence, with expression levels correlating with quiescence depth. Mechanistically, the Notch and bone morphogenetic protein (BMP) target HES1 and ID3, respectively, and act as p27 repressors, adjusting quiescence depth by lowering p27 levels to promote shallow quiescence [80].

In ECs, proliferation decreases in soft 2D substrates and increases in stiff 2D substrates [43]. For example, HUVECs cultured on more compliant matrices (1 kilopascal, kPa) showed increased expression and clustering of VEGFR2, while stiffer matrices (10 kPa) induced increased VEGFR2 internalization and signaling [81]. This switch was mediated by Rho activity and actin contractility [81]. In a confluent endothelial monolayer, however, stiffness-enhanced VEGF signaling is no longer observed, suggesting that this mechanism is specific to actively proliferating cells and angiogenic processes and suppressed once contact inhibition is established [81]. Notably, not only ECM stiffness but also the ECM’s capacity to retain mitogens might play a role in BEC contact inhibition [82]. Furthermore, mechanosensitive YAP1 signaling has been shown to regulate endothelial contact inhibition, as the loss of the YAP1 regulator DLC1 (deleted in liver cancer 1) leads to a loss of contact inhibition, while YAP silencing prevents this effect [83].

Notch signaling as a central regulator of contact inhibition in blood endothelial cells

Notch signaling has been implicated in cell fate control during development, where it requires cell–cell contact to activate lateral inhibition [84]. Since the early 2000s, several studies have demonstrated how Notch employs a similar mechanism to induce contact inhibition in BECs. When BECs are plated at low, medium, or high density, downstream Notch genes are up-regulated in correlation with increasing cell confluency [8,85]. An increase in Notch activation correlates with a reduction in p21 and an increase in p27 expression, indicating that Notch plays a key role in establishing BEC quiescence and contact inhibition [8,85]. Additionally, Notch signaling controlled BEC contact inhibition through the regulation of the minichromosome maintenance (MCM) proteins 2 and 6 [86]. In this case, Notch activation down-regulates MCM2 and MCM6 expressions, which, in turn, reduces Rb phosphorylation, thereby blocking cell cycle progression [86].

While in vitro studies have shown that active Notch signaling consistently acts as a contact inhibition signal, in vivo models of retinal angiogenesis reveal that its role is highly context-dependent [69] discussed above. At very low VEGF signaling levels, retinal ECs remain quiescent with active Notch signaling that suppresses ERK activity and cell proliferation. Stalk cells operate under balanced Notch and VEGF signaling, producing an ERK activity level optimal for controlled proliferation. In contrast, tip cells experience high VEGF and low Notch signaling, leading to elevated ERK activity, which induces p21, cell cycle arrest, and promotes cell sprouting and migration [69].

Notch signaling has also been implicated in arterialization, where it is coupled with the suppression of the BEC cell cycle. Using inducible genetic mosaics, it was shown that although BECs with high VEGF and Notch signaling are preferentially located in arterial vascular beds, Notch does not directly activate an arterial genetic program but instead suppresses MYC-driven metabolic and cell cycle activities [87]. Consistent with this, microRNA-218 (miR-218) has been identified as a downstream effector of active Notch signaling in quiescent BECs [88]. Induction of miR-218 expression attenuates MYC activity, thereby repressing BEC proliferation and promoting contact inhibition [88].

Although contact inhibition has been extensively studied in BECs, its regulation in lymphatic endothelial cells (LECs) remains largely unknown (Figure 1). The authors and others have shown that the regulation of endothelial barrier function employs different molecules in BECs and LECs, even when expression patterns and levels appear similar across lymphatic and blood vascular beds and organs. This applies to molecules such as VE-cadherin [71,89] and CLDN5 [77,90], as well as EphB4 [90,91], and suggests that certain aspects of contact inhibition in LECs may also differ from those in BECs.

Mechanisms of contact inhibition in lymphatic and blood endothelial cells.

Figure 1:
Mechanisms of contact inhibition in lymphatic and blood endothelial cells.

Blank spaces indicate that the extrinsic stimulus or molecular pathway has not been studied in the context of lymphatic or blood endothelial contact inhibition. (✓) denotes context-specific regulation of contact inhibition. Notably, several key regulators of contact inhibition have not yet been specifically studied in lymphatic endothelial cells.

Figure 1:
Mechanisms of contact inhibition in lymphatic and blood endothelial cells.

Blank spaces indicate that the extrinsic stimulus or molecular pathway has not been studied in the context of lymphatic or blood endothelial contact inhibition. (✓) denotes context-specific regulation of contact inhibition. Notably, several key regulators of contact inhibition have not yet been specifically studied in lymphatic endothelial cells.

Close modal

The PDE2A/cGMP/p38/MAPK/Notch axis controls lymphatic endothelial cell contact inhibition

Phosphodiesterase 2A (PDE2A) is a phosphodiesterase with dual specificity, which hydrolyzes cyclic adenosine monophosphate (cAMP) to AMP and cyclic guanosine monophosphate (cGMP) to GMP [92]. The loss of PDE2A in BECs was correlated with increased cAMP levels and dysregulated blood vessel barriers during homeostasis and inflammation [93,94]. However, despite these roles, PDE2A is not essential for proper blood vessel formation in vitro and in vivo [14].

Recently, we have shown that LECs induce contact inhibition through a previously unappreciated PDE2A-controlled cGMP/p38 MAPK/Notch axis [14]. We demonstrated that the loss of lymphatic PDE2A causes embryonic lymphatic dysplasia in vivo, as well as increased cGMP levels, defective junctions, and down-regulation of the major lymphatic junctional regulator CLDN5 in vitro [14]. Interestingly, VE-cadherin expression was not significantly altered in the absence of PDE2A [14].

An RNA sequencing approach comparing low-confluency (CLDN5Low) and high-confluency (CLDN5High) LECs revealed both similar and distinct gene expression changes in junctional and contact inhibition/proliferation genes, compared with BECs. Notably, FLT4 (the coding gene for VEGFR3) expression was significantly elevated in contact-inhibited LECs but not BECs [14], potentially suggesting the distinct responsiveness to VEGFC compared with VEGF/VEGFR2 in LECs versus BECs, respectively.

Using the same rationale but comparing CLDN5high LECs in the presence and absence of PDE2A reversed gene expression, with proliferation genes being up-regulated and junctional genes being down-regulated in the absence of PDE2A. In the absence of Pde2a in lymphatic vessels, LECs showed moderate but continuous proliferation in mouse embryonic back skins which coincided with compromised LEC contact inhibition [14]. The moderate, rather than hyperproliferative, growth is likely due to a decline in mitogenic signaling at later stages of lymphatic development.

The lymphatic defects observed upon PDE2A loss were attributed to the enzyme’s unique function in LECs, where it selectively hydrolyzes cGMP over cAMP. The resulting elevation in cGMP levels in LECs disrupts junctional integrity, leading to the loss of contact inhibition. Elevated cGMP levels also led to increased p38 phosphorylation, a pathway previously implicated in contact inhibition of other cell types [95,96] but not in BECs. Downstream of p38, LECs activated Notch signaling to promote contact inhibition [14], aligning with the previously established role of Notch in BEC contact inhibition.

Mechanical control of lymphatic endothelial cell contact inhibition

Unlike BECs [80], LECs do not initiate contact inhibition and quiescence when exposed to steady laminar flow [97,98]. For example, in response to laminar flow, LECs, but not BECs, showed increased proliferation through regulation of VEGFA, VEGFC, FGFR3, and p53/CDKN1C. ORAI1, a subunit of the calcium release-activated calcium channel, was identified as an early mediator of these shear stress responses and proliferation in LECs [97]. Moreover, flow-mediated proliferation was accompanied by the loss of lymphatic contact inhibition during homeostasis in mice, indicating that LECs possess unique regulatory mechanisms to finetune contact inhibition and proliferation.

Furthermore, these mechanisms display heterogeneity throughout the lymphatic vascular tree. Flow also controlled the loss of lymphatic contact inhibition during homeostasis in mice. Adult LEC turnover and proliferation was shown to be confined to valve regions of collecting vessels, with valve cells displaying the shortest lifespan [99]. Exposure to low recirculating flow, modeled by oscillatory shear stress (OSS) in vitro, induced valve cell proliferation via mTORC1 signaling to support the renewal of valve LECs, which are naturally subjected to higher mechanical stress [99]. In contrast, high recirculating, disturbed flow in lymphatic vessels (and high OSS in vitro) cooperates with the transcription factor FOXC2 to induce quiescence and ensure lifelong stability of the lymphatic vasculature. The loss of FOXC2 conferred abnormal shear stress sensing, activated YAP1/TAZ signaling, and promoted junction disassembly and entry into the cell cycle in vitro [98].

Additionally, both fluid accumulation (by increasing the amount of interstitial fluid in mouse embryos in ‘gain-of-fluid’ experiments) and resulting LEC stretching were shown to induce VEGFR3/β1-integrin-mediated proliferation [100]. Conversely, ‘loss-of-fluid’ experiments revealed reduced LEC proliferation [100], suggesting that this mechanism could also play a role for LEC contact inhibition at a later stage.

Finally, and importantly, studies in zebrafish have shown that precise regulation of the CDKIs p27 and p21 is essential to control lymphatic sprouting [101,102]. Whether these underlying mechanisms also contribute to lymphatic contact inhibition later in development remains to be determined.

Stable and quiescent endothelial cell junctions are of vital importance to guarantee blood and lymph flow without leakage. Endothelial contact inhibition controls quiescence and prevents uncontrolled proliferation, a condition associated with various vascular diseases, such as hemangiomas, vascular malformations, atherosclerosis, and psoriasis, as well as tumor (lymph)angiogenesis. Blood endothelial contact inhibition has been extensively studied, showing that many of its key regulators are shared across cell types, including fibroblasts and epithelial cells. In contrast, mechanisms of lymphatic contact inhibition are still understudied. Interestingly, while Notch activation plays a similar role in promoting contact inhibition in both BECs and LECs, our findings and those of others indicate that certain extrinsic factors, such as differential types of fluid flow, and molecular pathways, like the PDE2A/cGMP/p38 MAPK axis, are specific to BECs or LECs (Figure 1). EC-specific control of contact inhibition could provide the opportunity to modulate contact inhibition in selected vessel types.

Perspectives
  • In high-confluency monolayers and under spatial confinement, endothelial cells form mature cell–cell junctions and reduce proliferation. This process, referred to as contact inhibition, controls endothelial quiescence and prevents uncontrolled proliferation, a condition associated with various vascular diseases.

  • Blood endothelial contact inhibition has been extensively studied showing that many of its key regulators, such as PI3K/AKT, Notch, Yap, and cadherins, are shared across cell types, including fibroblasts and epithelial cells. Furthermore, many additional signaling pathways have been implicated in regulating endothelial proliferation and the formation of endothelial contacts. However, whether these pathways are also linked to adhesive processes during contact inhibition remains to be experimentally determined.

  • The regulation of contact inhibition in lymphatic endothelial cells is, however, still understudied. A select few studies show that while LECs and BECs share common mechanisms of contact inhibition, nuanced differences are now being identified, which could allow for targeted modulation of blood versus lymphatic vessels.

The authors declare that they have no conflict of interest.

This work was supported by an Exploration Grant of the Boehringer Ingelheim Foundation (BIS) and the German Research Foundation (DFG) grant FR4239/1-1 (to M.F.).

Writing – Original Draft, C.C., L.M.H.L. and M.F.; Writing – Review & Editing, C.C., L.M.H.L., M.B. and M.F.; Visualization, M.F.; Funding Acquisition, M.F.

Figure 1 was partially created in BioRender (2025, https://biorender.com/r55q684).

BECs

Blood Endothelial Cells

BMP

bone morphogenetic protein

CDK

Cyclin-Dependent Kinases

CDKIs

Cyclin-Dependent Kinase Inhibitors

ECM

Extracellular Matrix

EGF

Epidermal Growth Factor

ERM

Ezrin, Radixin, and Moesin

FGF

Fibroblast Growth Factor

HUVECs

Human Umbilical Vein Endothelial Cells

LECs

Lymphatic Endothelial Cells

MAPK

Mitogen-Activated Protein Kinase

MCM

Minichromosome Maintenance

Rb

Retinoblastoma Protein

VE

Vascular Endothelial

VEGFs

Vascular Endothelial Growth Factors

cAMP

Cyclic Adenosine Monophosphate

cGMP

Cyclic Guanosine Monophosphate

1
ABERCROMBIE
,
M.
and
HEAYSMAN
,
J.E
. (
1954
)
Observations on the social behaviour of cells in tissue culture. II. Monolayering of fibroblasts
.
Exp. Cell Res.
6
,
293
306
https://doi.org/10.1016/0014-4827(54)90176-7
2
Stoker
,
M.G.P.
and
Rubin
,
H
. (
1967
)
Density dependent inhibition of cell growth in culture
.
Nature New Biol.
215
,
171
172
https://doi.org/10.1038/215171a0
3
Lemons
,
J.M.S.
,
Feng
,
X.-J.
,
Bennett
,
B.D.
,
Legesse-Miller
,
A.
,
Johnson
,
E.L.
,
Raitman
,
I
, et al.
(
2010
)
Quiescent fibroblasts exhibit high metabolic activity
.
PLOS Biol
8
, e1000514 https://doi.org/10.1371/journal.pbio.1000514
4
St. Croix
,
B.
,
Sheehan
,
C.
,
Rak
,
J.W.
,
Flørenes
,
V.A.
,
Slingerland
,
J.M.
and
Kerbel
,
R.S
. (
1998
)
E-cadherin–dependent growth suppression is mediated by the cyclin-dependent kinase inhibitor p27KIP1
.
J. Cell Biol.
142
,
557
571
https://doi.org/10.1083/jcb.142.2.557
5
Kim
,
N.-G.
,
Koh
,
E.
,
Chen
,
X.
and
Gumbiner
,
B.M
. (
2011
)
E-cadherin mediates contact inhibition of proliferation through Hippo signaling-pathway components
.
Proc. Natl. Acad. Sci. U.S.A
108
,
11930
11935
https://doi.org/10.1073/pnas.1103345108
6
Pavel
,
M.
,
Renna
,
M.
,
Park
,
S.J.
,
Menzies
,
F.M.
,
Ricketts
,
T.
,
Füllgrabe
,
J
, et al.
(
2018
)
Contact inhibition controls cell survival and proliferation via YAP/TAZ-autophagy axis
.
Nat. Commun
2961
,
2961
https://doi.org/10.1038/s41467-018-05388-x
7
Grazia Lampugnani
,
M.
,
Zanetti
,
A.
,
Corada
,
M.
,
Takahashi
,
T.
,
Balconi
,
G.
,
Breviario
,
F.
et al.
(
2003
)
Contact inhibition of VEGF-induced proliferation requires vascular endothelial cadherin, beta-catenin, and the phosphatase DEP-1/CD148
.
J. Cell Biol.
161
,
793
804
https://doi.org/10.1083/jcb.200209019
8
Noseda
,
M.
,
Chang
,
L.
,
McLean
,
G.
,
Grim
,
J.E.
,
Clurman
,
B.E.
,
Smith
,
L.L.
et al.
(
2004
)
Notch activation induces endothelial cell cycle arrest and participates in contact inhibition: role of p21Cip1 repression
.
Mol. Cell. Biol.
24
,
8813
8822
https://doi.org/10.1128/MCB.24.20.8813-8822.2004
9
Vlodavsky
,
I.
,
Fielding
,
P.E.
,
Fielding
,
C.J.
and
Gospodarowicz
,
D
. (
1978
)
Role of contact inhibition in the regulation of receptor-mediated uptake of low density lipoprotein in cultured vascular endothelial cells
.
Proc. Natl. Acad. Sci. U.S.A
75
,
356
360
https://doi.org/10.1073/pnas.75.1.356
10
Buikema
,
J.W.
,
Lee
,
S.
,
Goodyer
,
W.R.
,
Maas
,
R.G.
,
Chirikian
,
O.
,
Li
,
G
, et al.
(
2020
)
Wnt activation and reduced cell-cell contact synergistically induce massive expansion of functional human iPSC-derived cardiomyocytes
.
Cell Stem Cell
27
,
50
63
https://doi.org/10.1016/j.stem.2020.06.001
11
Bard
,
J.B.
and
Hay
,
E.D.
, eds. (
1975
)
The behavior of fibroblasts from the developing avian cornea
.
Morphology and movement in situ and in vitro. J Cell Biol
67
,
400
418
https://doi.org/10.1083/jcb.67.2.400
12
Davis
,
J.R.
,
Huang
,
C.-Y.
,
Zanet
,
J.
,
Harrison
,
S.
,
Rosten
,
E.
,
Cox
,
S.
et al.
(
2012
)
Emergence of embryonic pattern through contact inhibition of locomotion
.
Development
139
,
4555
4560
https://doi.org/10.1242/dev.082248
13
Carmona-Fontaine
,
C.
,
Matthews
,
H.K.
,
Kuriyama
,
S.
,
Moreno
,
M.
,
Dunn
,
G.A.
,
Parsons
,
M.
et al.
(
2008
)
Contact inhibition of locomotion in vivo controls neural crest directional migration
.
Nature New Biol.
456
,
957
961
https://doi.org/10.1038/nature07441
14
Carlantoni
,
C.
,
Liekfeld
,
L.M.H.
,
Hemkemeyer
,
S.A.
,
Schreier
,
D.
,
Saygi
,
C.
,
Kurelic
,
R
, et al.
(
2024
)
The phosphodiesterase 2A controls lymphatic junctional maturation via cGMP-dependent notch signaling
.
Dev. Cell
59
,
308
325
https://doi.org/10.1016/j.devcel.2023.12.002
15
Lanosa
,
X.A.
and
Colombo
,
J.A
. (
2008
)
Cell contact-inhibition signaling as part of wound-healing processes in brain
.
Neuron Glia Biol.
4
,
27
34
https://doi.org/10.1017/S1740925X09000039
16
Zanca
,
A.
,
Flegg
,
J.A.
and
Osborne
,
J.M
. (
2022
)
Push or pull? Cell proliferation and migration during wound healing
.
Front. Syst. Biol.
2
https://doi.org/10.3389/fsysb.2022.876075
17
Fisher
,
H.W.
and
Yeh
,
J
. (
1967
)
Contact inhibition in colony formation
.
Science
155
,
581
582
https://doi.org/10.1126/science.155.3762.581
18
Hanahan
,
D.
and
Weinberg
,
R.A
. (
2000
)
The hallmarks of cancer
.
Cell
100
,
57
70
https://doi.org/10.1016/s0092-8674(00)81683-9
19
Matthews
,
H.K.
and
Bertoli
,
C
. (
2022
)
Cell cycle control in cancer
.
Nat. Rev. Mol. Cell Biol.
23
,
74
88
https://doi.org/10.1038/s41580-021-00404-3
20
Martínez-Alonso
,
D.
and
Malumbres
,
M
. (
2020
)
Mammalian cell cycle cyclins
.
Semin. Cell Dev. Biol.
107
,
28
35
https://doi.org/10.1016/j.semcdb.2020.03.009
21
Goodrich
,
D.W.
,
Wang
,
N.P.
,
Qian
,
Y.-W.
,
Lee
,
E.Y.H.P.
and
Lee
,
W.-H
. (
1991
)
The retinoblastoma gene product regulates progression through the G1 phase of the cell cycle
.
Cell
67
,
293
302
https://doi.org/10.1016/0092-8674(91)90181-w
22
Shao
,
Z.
and
Robbins
,
P.D
. (
1995
)
Differential regulation of E2F and Sp1-mediated transcription by G1 cyclins
.
Oncogene
10
,
221
228
23
Harper
,
J.W.
,
Elledge
,
S.J.
,
Keyomarsi
,
K.
,
Dynlacht
,
B.
,
Tsai
,
L.H.
,
Zhang
,
P.
et al.
(
1995
)
Inhibition of cyclin-dependent kinases by p21
.
Mol. Biol. Cell
6
,
387
400
https://doi.org/10.1091/mbc.6.4.387
24
Nakayama
,
K.
and
Nakayama
,
K
. (
1998
)
Cip/Kip cyclin-dependent kinase inhibitors: brakes of the cell cycle engine during development
.
Bioessays
20
,
1020
1029
https://doi.org/10.1002/(sici)1521-1878(199812)20:12<1020::Aid-bies8>3.0.Co;2-d
25
Kim
,
W.Y.
and
Sharpless
,
N.E
. (
2006
)
The regulation of INK4/ARF in cancer and aging
.
Cell
127
,
265
275
https://doi.org/10.1016/j.cell.2006.10.003
26
Satyanarayana
,
A.
and
Kaldis
,
P
. (
2009
)
Mammalian cell-cycle regulation: several Cdks, numerous cyclins and diverse compensatory mechanisms
.
Oncogene
28
,
2925
2939
https://doi.org/10.1038/onc.2009.170
27
Lim
,
S.
and
Kaldis
,
P
. (
2013
)
Cdks, cyclins and CKIs: roles beyond cell cycle regulation
.
Development
140
,
3079
3093
https://doi.org/10.1242/dev.091744
28
Schirripa
,
A.
,
Sexl
,
V.
and
Kollmann
,
K
. (
2022
)
Cyclin-dependent kinase inhibitors in malignant hematopoiesis
.
Front. Oncol.
12
, 916682 https://doi.org/10.3389/fonc.2022.916682
29
Mühleder
,
S.
,
Fernández-Chacón
,
M.
,
Garcia-Gonzalez
,
I.
and
Benedito
,
R
. (
2021
)
Endothelial sprouting, proliferation, or senescence: tipping the balance from physiology to pathology
.
Cell. Mol. Life Sci.
78
,
1329
1354
https://doi.org/10.1007/s00018-020-03664-y
30
Lu
,
Z.
and
Hunter
,
T
. (
2010
)
Ubiquitylation and proteasomal degradation of the p21(Cip1), p27(Kip1) and p57(Kip2) CDK inhibitors
.
Cell Cycle
9
,
2342
2352
https://doi.org/10.4161/cc.9.12.11988
31
Alaiz Noya
,
M.
,
Berti
,
F.
and
Dietrich
,
S
. (
2022
)
Comprehensive expression analysis for the core cell cycle regulators in the chicken embryo reveals novel tissue-specific synexpression groups and similarities and differences with expression in mouse, frog and zebrafish
.
J. Anat.
241
,
42
66
https://doi.org/10.1111/joa.13629
32
Coller
,
H.A.
,
Sang
,
L.
and
Roberts
,
J.M
. (
2006
)
A new description of cellular quiescence
.
PLoS Biol
4
, e83 https://doi.org/10.1371/journal.pbio.0040083
33
Fujimaki
,
K.
,
Li
,
R.
,
Chen
,
H.
,
Della Croce
,
K.
,
Zhang
,
H.H.
,
Xing
,
J
, et al.
(
2019
)
Graded regulation of cellular quiescence depth between proliferation and senescence by a lysosomal dimmer switch
.
Proc. Natl. Acad. Sci. U.S.A
116
,
22624
22634
https://doi.org/ 10.1073/pnas.1915905116
34
Kwon
,
J.S.
,
Everetts
,
N.J.
,
Wang
,
X.
,
Wang
,
W.
,
Della Croce
,
K.
,
Xing
,
J
, et al.
(
2017
)
Controlling depth of cellular quiescence by an Rb-E2F network switch
.
Cell Rep
20
,
3223
3235
https://doi.org/ 10.1016/j.celrep.2017.09.007
35
Herranz
,
N.
and
Gil
,
J
. (
2018
)
Mechanisms and functions of cellular senescence
.
J. Clin. Invest
128
,
1238
1246
https://doi.org/ 10.1172/jci95148
36
Streichan
,
S.J.
,
Hoerner
,
C.R.
,
Schneidt
,
T.
,
Holzer
,
D.
and
Hufnagel
,
L
. (
2014
)
Spatial constraints control cell proliferation in tissues
.
Proc. Natl. Acad. Sci. U.S.A
111
,
5586
5591
https://doi.org/10.1073/pnas.1323016111
37
Devany
,
J.
,
Falk
,
M.J.
,
Holt
,
L.J.
,
Murugan
,
A.
and
Gardel
,
M.L
. (
2023
)
Epithelial tissue confinement inhibits cell growth and leads to volume-reducing divisions
.
Dev. Cell
58
,
1462
1476
https://doi.org/10.1016/j.devcel.2023.05.018
38
Kim
,
J.-H.
and
Asthagiri
,
A.R
. (
2011
)
Matrix stiffening sensitizes epithelial cells to EGF and enables the loss of contact inhibition of proliferation
.
J. Cell. Sci.
124
,
1280
1287
https://doi.org/10.1242/jcs.078394
39
Puliafito
,
A.
,
Hufnagel
,
L.
,
Neveu
,
P.
,
Streichan
,
S.
,
Sigal
,
A.
,
Fygenson
,
D.K.
et al.
(
2012
)
Collective and single cell behavior in epithelial contact inhibition
.
Proc. Natl. Acad. Sci. U.S.A
109
,
739
744
https://doi.org/10.1073/pnas.1007809109
40
Di Meglio
,
I.
,
Trushko
,
A.
,
Guillamat
,
P.
,
Blanch-Mercader
,
C.
,
Abuhattum
,
S.
and
Roux
,
A
. (
2022
)
Pressure and curvature control of the cell cycle in epithelia growing under spherical confinement
.
Cell Rep.
40
, 111227 https://doi.org/10.1016/j.celrep.2022.111227
41
Nelson
,
C.M.
,
Jean
,
R.P.
,
Tan
,
J.L.
,
Liu
,
W.F.
,
Sniadecki
,
N.J.
,
Spector
,
A.A.
et al.
(
2005
)
Emergent patterns of growth controlled by multicellular form and mechanics
.
Proc. Natl. Acad. Sci. U.S.A
102
,
11594
11599
https://doi.org/10.1073/pnas.0502575102
42
Carpenter
,
L.C.
,
Pérez-Verdugo
,
F.
and
Banerjee
,
S
. (
2024
)
Mechanical control of cell proliferation patterns in growing epithelial monolayers
.
Biophys. J
123
,
909
919
https://doi.org/10.1016/j.bpj.2024.03.002
43
Gordon
,
E.
,
Schimmel
,
L.
and
Frye
,
M
. (
2020
)
The importance of mechanical forces for in vitro endothelial cell biology
.
Front. Physiol
11
,
684
https://doi.org/10.3389/fphys.2020.00684
44
Schnyder
,
S.K.
,
Molina
,
J.J.
and
Yamamoto
,
R
. (
2020
)
Control of cell colony growth by contact inhibition
.
Sci. Rep
10
,
6713
https://doi.org/10.1038/s41598-020-62913-z
45
Jain
,
H.P.
,
Wenzel
,
D.
and
Voigt
,
A
. (
2022
)
Impact of contact inhibition on collective cell migration and proliferation
.
Phys. Rev. E
105
,
034402
https://doi.org/10.1103/PhysRevE.105.034402
46
Morais
,
M.C.C.
,
Stuhl
,
I.
,
Sabino
,
A.U.
,
Lautenschlager
,
W.W.
,
Queiroga
,
A.S.
,
Tortelli
,
T.C
, et al.
(
2017
)
Stochastic model of contact inhibition and the proliferation of melanoma in situ
.
Sci. Rep.
https://doi.org/10.1038/s41598-017-07553-6
47
Zhang
,
Y.
,
Wang
,
H.
,
Oliveira
,
R.H.M.
,
Zhao
,
C.
and
Popel
,
A.S
. (
2022
)
Systems biology of angiogenesis signaling: Computational models and omics
.
WIREs Mech Dis
14
, e1550
48
Mueller
,
S.
and
Cadenas
,
E
. (
2000
)
p21WAF1 Regulates Anchorage-independent Growth of HCT116 Colon Carcinoma Cells via E-Cadherin Expression1
.
Cancer Res.
60
,
156
163
49
Gottardi
,
C.J.
,
Wong
,
E.
and
Gumbiner
,
B.M
. (
2001
)
E-Cadherin Suppresses Cellular Transformation by Inhibiting β-Catenin Signaling in an Adhesion-Independent Manner
.
J. Cell Biol.
153
,
1049
1060
50
Rodrigues
,
M.
,
Griffith
,
L.G.
and
Wells
,
A
. (
2010
)
Growth factor regulation of proliferation and survival of multipotential stromal cells
.
Stem Cell Res. Ther.
1
,
32
https://doi.org/10.1186/scrt32
51
Aaronson
,
S.A
. (
1991
)
Growth factors and cancer
.
Science
254
,
1146
1153
https://doi.org/10.1126/science.1659742
52
Maddaluno
,
L.
,
Urwyler
,
C.
and
Werner
,
S
. (
2017
)
Fibroblast growth factors: key players in regeneration and tissue repair
.
Development
144
,
4047
4060
https://doi.org/10.1242/dev.152587
53
Ferrara
,
N
. (
2004
)
Vascular endothelial growth factor: basic science and clinical progress
.
Endocr. Rev.
25
,
581
611
https://doi.org/10.1210/er.2003-0027
54
Kim
,
J.-H.
,
Kushiro
,
K.
,
Graham
,
N.A.
and
Asthagiri
,
A.R
. (
2009
)
Tunable interplay between epidermal growth factor and cell–cell contact governs the spatial dynamics of epithelial growth
.
Proc. Natl. Acad. Sci. U.S.A
106
,
11149
11153
https://doi.org/10.1073/pnas.0812651106
55
Benham-Pyle
,
B.W.
,
Pruitt
,
B.L.
and
Nelson
,
W.J
. (
2015
)
Cell adhesion. Mechanical strain induces E-cadherin-dependent Yap1 and β-catenin activation to drive cell cycle entry
.
Science
348
,
1024
1027
https://doi.org/10.1126/science.aaa4559
56
Aragona
,
M.
,
Panciera
,
T.
,
Manfrin
,
A.
,
Giulitti
,
S.
,
Michielin
,
F.
,
Elvassore
,
N
, et al.
(
2013
)
A mechanical checkpoint controls multicellular growth through YAP/TAZ regulation by actin-processing factors
.
Cell
154
,
1047
1059
https://doi.org/10.1016/j.cell.2013.07.042
57
Shalhout
,
S.Z.
,
Yang
,
P.-Y.
,
Grzelak
,
E.M.
,
Nutsch
,
K.
,
Shao
,
S.
,
Zambaldo
,
C.
et al.
(
2021
)
YAP-dependent proliferation by a small molecule targeting annexin A2
.
Nat. Chem. Biol.
17
,
767
775
https://doi.org/10.1038/s41589-021-00755-0
58
Li
,
Y.
,
Liu
,
Z.
,
Han
,
X.
,
Liang
,
F.
,
Zhang
,
Q.
,
Huang
,
X.
et al.
(
2024
)
Dynamics of endothelial cell generation and turnover in arteries during homeostasis and diseases
.
Circulation
149
,
135
154
https://doi.org/10.1161/CIRCULATIONAHA.123.064301
59
D’Amore
,
P.A
. (
1992
)
Mechanisms of endothelial growth control
.
Am. J. Respir. Cell Mol. Biol.
6
,
1
8
https://doi.org/10.1165/ajrcmb/6.1.1
60
Joyce
,
N.C
. (
2010
) Cell cycle control and replication in corneal endothelium.
In
In Cornea and External Eye Disease: Corneal Allotransplantation, Allergic Disease and Trachoma
(
Reinhard
,
T.
and
Larkin
,
F.
, eds),
pp
.
69
86
, , https://doi.org/10.1007/978-3-540-85544-6_6
61
Hanai
,
J. -i.
,
Dhanabal
,
M.
,
Karumanchi
,
S.A.
,
Albanese
,
C.
,
Waterman
,
M.
,
Chan
,
B
, et al.
(
2002
)
Endostatin causes G
.
J. Biol. Chem.
277
,
16464
16469
https://doi.org/10.1074/jbc.M112274200
62
Lugano
,
R.
,
Ramachandran
,
M.
and
Dimberg
,
A
. (
2020
)
Tumor angiogenesis: causes, consequences, challenges and opportunities
.
Cell. Mol. Life Sci.
77
,
1745
1770
https://doi.org/10.1007/s00018-019-03351-7
63
Abdelilah-Seyfried
,
S.
and
Ola
,
R
. (
2024
)
Shear stress and pathophysiological PI3K involvement in vascular malformations
.
J. Clin. Invest.
134
https://doi.org/10.1172/JCI172843
64
Lampugnani
,
M.G.
and
Dejana
,
E
. (
1997
)
Interendothelial junctions: structure, signalling and functional roles
.
Curr. Opin. Cell Biol.
9
,
674
682
https://doi.org/10.1016/s0955-0674(97)80121-4
65
Simons
,
M.
,
Gordon
,
E.
and
Claesson-Welsh
,
L
. (
2016
)
Mechanisms and regulation of endothelial VEGF receptor signalling
.
Nat. Rev. Mol. Cell Biol.
17
,
611
625
https://doi.org/10.1038/nrm.2016.87
66
Viñals
,
F.
and
Pouysségur
,
J
. (
1999
)
Confluence of vascular endothelial cells induces cell cycle exit by inhibiting p42/p44 mitogen-activated protein kinase activity
.
Mol. Cell. Biol.
19
,
2763
2772
https://doi.org/10.1128/MCB.19.4.2763
67
Li
,
S.
,
Gerrard
,
E.R.
and
Balkovetz
,
D.F
. (
2004
)
Evidence for ERK1/2 phosphorylation controlling contact inhibition of proliferation in Madin-Darby canine kidney epithelial cells
.
American Journal of Physiology-Cell Physiology
287
,
C432
C439
https://doi.org/10.1152/ajpcell.00020.2004
68
Wayne
,
J.
,
Sielski
,
J.
,
Rizvi
,
A.
,
Georges
,
K.
and
Hutter
,
D
. (
2006
)
ERK regulation upon contact inhibition in fibroblasts
.
Mol. Cell. Biochem.
286
,
181
189
https://doi.org/10.1007/s11010-005-9089-z
69
Pontes-Quero
,
S.
,
Fernández-Chacón
,
M.
,
Luo
,
W.
,
Lunella
,
F.F.
,
Casquero-Garcia
,
V.
,
Garcia-Gonzalez
,
I.
et al.
(
2019
)
High mitogenic stimulation arrests angiogenesis
.
Nat. Commun.
10
,
2016
https://doi.org/10.1038/s41467-019-09875-7
70
Suzuki
,
E.
,
Nagata
,
D.
,
Yoshizumi
,
M.
,
Kakoki
,
M.
,
Goto
,
A.
,
Omata
,
M
, et al.
(
2000
)
Reentry into the cell cycle of contact-inhibited vascular endothelial cells by a phosphatase inhibitor: possible involvement of extracellular signal-regulated kinase and phosphatidylinositol 3-kinase
.
J. Biol. Chem.
275
,
3637
3644
https://doi.org/10.1074/jbc.275.5.3637
71
Frye
,
M.
,
Dierkes
,
M.
,
Küppers
,
V.
,
Vockel
,
M.
,
Tomm
,
J.
,
Zeuschner
,
D.
et al.
(
2015
)
Interfering with VE-PTP stabilizes endothelial junctions in vivo via Tie-2 in the absence of VE-cadherin
.
J. Exp. Med.
212
,
2267
2287
https://doi.org/10.1084/jem.20150718
72
Wessel
,
F.
,
Winderlich
,
M.
,
Holm
,
M.
,
Frye
,
M.
,
Rivera-Galdos
,
R.
,
Vockel
,
M.
et al.
(
2014
)
Leukocyte extravasation and vascular permeability are each controlled in vivo by different tyrosine residues of VE-cadherin
.
Nat. Immunol.
15
,
223
230
https://doi.org/10.1038/ni.2824
73
Drexler
,
H.C.A.
,
Vockel
,
M.
,
Polaschegg
,
C.
,
Frye
,
M.
,
Peters
,
K.
and
Vestweber
,
D
. (
2019
)
Vascular endothelial receptor tyrosine phosphatase: identification of novel substrates related to junctions and a ternary complex with EPHB4 and TIE2[S].
.
Mol. Cell Proteomics
18
,
2058
2077
https://doi.org/10.1074/mcp.RA119.001716
74
Lampugnani
,
M.G.
,
Orsenigo
,
F.
,
Gagliani
,
M.C.
,
Tacchetti
,
C.
and
Dejana
,
E
. (
2006
)
Vascular endothelial cadherin controls VEGFR-2 internalization and signaling from intracellular compartments
.
J. Cell Biol.
174
,
593
604
https://doi.org/10.1083/jcb.200602080
75
Acharya
,
B.R.
,
Fang
,
J.S.
,
Jeffery
,
E.D.
,
Chavkin
,
N.W.
,
Genet
,
G.
,
Vasavada
,
H.
et al.
(
2023
)
Connexin 37 sequestering of activated-ERK in the cytoplasm promotes p27-mediated endothelial cell cycle arrest
.
Life Sci. Alliance
6
, e202201685 https://doi.org/10.26508/lsa.202201685
76
Baumeister
,
U.
,
Funke
,
R.
,
Ebnet
,
K.
,
Vorschmitt
,
H.
,
Koch
,
S.
and
Vestweber
,
D
. (
2005
)
Association of Csk to VE-cadherin and inhibition of cell proliferation
.
EMBO J.
24
,
1686
1695
https://doi.org/10.1038/sj.emboj.7600647
77
Richards
,
M.
,
Nwadozi
,
E.
,
Pal
,
S.
,
Martinsson
,
P.
,
Kaakinen
,
M.
,
Gloger
,
M.
et al.
(
2022
)
Claudin5 protects the peripheral endothelial barrier in an organ and vessel-type-specific manner
.
Elife
11
, e78517 https://doi.org/10.7554/eLife.78517
78
Kluger
,
M.S.
,
Clark
,
P.R.
,
Tellides
,
G.
,
Gerke
,
V.
and
Pober
,
J.S
. (
2013
)
Claudin-5 controls intercellular barriers of human dermal microvascular but not human umbilical vein endothelial cells
.
Arterioscler. Thromb. Vasc. Biol.
33
,
489
500
https://doi.org/10.1161/ATVBAHA.112.300893
79
Amsellem
,
V.
,
Dryden
,
N.H.
,
Martinelli
,
R.
,
Gavins
,
F.
,
Almagro
,
L.O.
,
Birdsey
,
G.M.
et al.
(
2014
)
ICAM-2 regulates vascular permeability and N-cadherin localization through ezrin-radixin-moesin (ERM) proteins and Rac-1 signalling
.
Cell Commun. Signal
12
,
12
https://doi.org/10.1186/1478-811X-12-12
80
Tanke
,
N.T.
,
Liu
,
Z.
,
Gore
,
M.T.
,
Bougaran
,
P.
,
Linares
,
M.B.
,
Marvin
,
A.
et al.
(
2024
)
Endothelial cell flow-mediated quiescence is temporally regulated and utilizes the cell cycle inhibitor p27
.
Arterioscler. Thromb. Vasc. Biol.
44
,
1265
1282
https://doi.org/10.1161/ATVBAHA.124.320671
81
LaValley
,
D.J.
,
Zanotelli
,
M.R.
,
Bordeleau
,
F.
,
Wang
,
W.
,
Schwager
,
S.C.
and
Reinhart-King
,
C.A
. (
2017
)
Matrix stiffness enhances VEGFR-2 internalization, signaling, and proliferation in endothelial cells
.
Converg. Sci. Phys. Oncol
3
,
044001
https://doi.org/10.1088/2057-1739/aa9263
82
Chen
,
T.T.
,
Luque
,
A.
,
Lee
,
S.
,
Anderson
,
S.M.
,
Segura
,
T.
and
Iruela-Arispe
,
M.L
. (
2010
)
Anchorage of VEGF to the extracellular matrix conveys differential signaling responses to endothelial cells
.
J. Cell Biol.
188
,
595
609
https://doi.org/10.1083/jcb.200906044
83
Ritchey
,
L.
,
Ha
,
T.
,
Otsuka
,
A.
,
Kabashima
,
K.
,
Wang
,
D.
,
Wang
,
Y.
et al.
(
2019
)
DLC1 deficiency and YAP signaling drive endothelial cell contact inhibition of growth and tumorigenesis
.
Oncogene
38
,
7046
7059
https://doi.org/10.1038/s41388-019-0944-x
84
Zhou
,
B.
,
Lin
,
W.
,
Long
,
Y.
,
Yang
,
Y.
,
Zhang
,
H.
,
Wu
,
K.
et al.
(
2022
)
Notch signaling pathway: architecture, disease, and therapeutics
.
Signal Transduct. Target. Ther.
7
,
95
,
95
https://doi.org/10.1038/s41392-022-00934-y
85
Rostama
,
B.
,
Turner
,
J.E.
,
Seavey
,
G.T.
,
Norton
,
C.R.
,
Gridley
,
T.
,
Vary
,
C.P.H.
et al.
(
2015
)
Dll4/Notch1 and BMP9 interdependent signaling induces human endothelial cell quiescence via p27KIP1 and thrombospondin-1
.
Arterioscler. Thromb. Vasc. Biol.
35
,
2626
2637
https://doi.org/10.1161/ATVBAHA.115.306541
86
Noseda
,
M.
,
Niessen
,
K.
,
McLean
,
G.
,
Chang
,
L.
and
Karsan
,
A
. (
2005
)
Notch-dependent cell cycle arrest is associated with downregulation of minichromosome maintenance proteins
.
Circ. Res.
97
,
102
104
https://doi.org/10.1161/01.RES.0000174380.06673.81
87
Luo
,
W.
,
Garcia-Gonzalez
,
I.
,
Fernández-Chacón
,
M.
,
Casquero-Garcia
,
V.
,
Sanchez-Muñoz
,
M.S.
,
Mühleder
,
S.
et al.
(
2021
)
Arterialization requires the timely suppression of cell growth
.
Nature New Biol.
589
,
437
441
https://doi.org/10.1038/s41586-020-3018-x
88
Sun
,
J.-X.
,
Dou
,
G.-R.
,
Yang
,
Z.-Y.
,
Liang
,
L.
,
Duan
,
J.-L.
,
Ruan
,
B.
et al.
(
2021
)
Notch activation promotes endothelial quiescence by repressing MYC expression via miR-218
.
Mol. Ther. Nucleic Acids
25
,
554
566
https://doi.org/10.1016/j.omtn.2021.07.023
89
Hägerling
,
R.
,
Hoppe
,
E.
,
Dierkes
,
C.
,
Stehling
,
M.
,
Makinen
,
T.
,
Butz
,
S.
et al.
(
2018
)
Distinct roles of VE-cadherin for development and maintenance of specific lymph vessel beds
.
EMBO J.
37
, e98271 https://doi.org/10.15252/embj.201798271
90
Frye
,
M.
,
Stritt
,
S.
,
Ortsäter
,
H.
,
Hernandez Vasquez
,
M.
,
Kaakinen
,
M.
,
Vicente
,
A.
et al.
(
2020
)
EphrinB2-EphB4 signalling provides Rho-mediated homeostatic control of lymphatic endothelial cell junction integrity
.
Elife
9
, e57732 https://doi.org/10.7554/eLife.57732
91
Luxán
,
G.
,
Stewen
,
J.
,
Díaz
,
N.
,
Kato
,
K.
,
Maney
,
S.K.
,
Aravamudhan
,
A.
et al.
(
2019
)
Endothelial EphB4 maintains vascular integrity and transport function in adult heart
.
Elife
8
, e45863 https://doi.org/10.7554/eLife.45863
92
Weber
,
S.
,
Zeller
,
M.
,
Guan
,
K.
,
Wunder
,
F.
,
Wagner
,
M.
and
El-Armouche
,
A
. (
2017
)
PDE2 at the crossway between cAMP and cGMP signalling in the heart
.
Cell. Signal
38
,
76
84
https://doi.org/10.1016/j.cellsig.2017.06.020
93
Surapisitchat
,
J.
,
Jeon
,
K.-I.
,
Yan
,
C.
and
Beavo
,
J.A
. (
2007
)
Differential regulation of endothelial cell permeability by cGMP via phosphodiesterases 2 and 3
.
Circ. Res.
101
,
811
818
https://doi.org/10.1161/CIRCRESAHA.107.154229
94
Chen
,
W.
,
Spitzl
,
A.
,
Mathes
,
D.
,
Nikolaev
,
V.O.
,
Werner
,
F.
,
Weirather
,
J.
et al.
(
2016
)
Endothelial actions of ANP enhance myocardial inflammatory infiltration in the early phase after acute infarction
.
Circ. Res.
119
,
237
248
https://doi.org/10.1161/CIRCRESAHA.115.307196
95
Slisz
,
M.
,
Rothenberger
,
E.
and
Hutter
,
D
. (
2008
)
Attenuation of p38 MAPK activity upon contact inhibition in fibroblasts
.
Mol. Cell. Biochem.
308
,
65
73
https://doi.org/10.1007/s11010-007-9613-4
96
Faust
,
D.
,
Dolado
,
I.
,
Cuadrado
,
A.
,
Oesch
,
F.
,
Weiss
,
C.
,
Nebreda
,
A.R.
et al.
(
2005
)
p38α MAPK is required for contact inhibition
.
Oncogene
24
,
7941
7945
https://doi.org/10.1038/sj.onc.1208948
97
Choi
,
D.
,
Park
,
E.
,
Jung
,
E.
,
Seong
,
Y.J.
,
Hong
,
M.
,
Lee
,
S.
et al.
(
2017
)
Orai1 activates proliferation of lymphatic endothelial cells in response to laminar flow through Krüppel-like factors 2 and 4.
.
Circ. Res.
120
,
1426
1439
https://doi.org/10.1161/CIRCRESAHA.116.309548
98
Sabine
,
A.
,
Bovay
,
E.
,
Demir
,
C.S.
,
Kimura
,
W.
,
Jaquet
,
M.
,
Agalarov
,
Y.
et al.
(
2015
)
FOXC2 and fluid shear stress stabilize postnatal lymphatic vasculature
.
J. Clin. Invest.
125
,
3861
3877
, 80454 https://doi.org/10.1172/JCI80454
99
Saygili Demir
,
C.
,
Sabine
,
A.
,
Gong
,
M.
,
Dormond
,
O.
and
Petrova
,
T.V
. (
2023
)
Mechanosensitive mTORC1 signaling maintains lymphatic valves
.
J. Cell Biol.
222
, e202207049. https://doi.org/10.1083/jcb.202207049
100
Planas-Paz
,
L.
,
Strilić
,
B.
,
Goedecke
,
A.
,
Breier
,
G.
,
Fässler
,
R.
and
Lammert
,
E
. (
2012
)
Mechanoinduction of lymph vessel expansion
.
EMBO J.
31
,
788
804
https://doi.org/10.1038/emboj.2011.456
101
Koltowska
,
K.
,
Okuda
,
K.S.
,
Gloger
,
M.
,
Rondon-Galeano
,
M.
,
Mason
,
E.
,
Xuan
,
J.
et al.
(
2021
)
The RNA helicase Ddx21 controls Vegfc-driven developmental lymphangiogenesis by balancing endothelial cell ribosome biogenesis and p53 function
.
Nat. Cell Biol.
23
,
1136
1147
https://doi.org/10.1038/s41556-021-00784-w
102
Jerafi-Vider
,
A.
,
Bassi
,
I.
,
Moshe
,
N.
,
Tevet
,
Y.
,
Hen
,
G.
,
Splittstoesser
,
D.
et al.
(
2021
)
VEGFC/FLT4-induced cell-cycle arrest mediates sprouting and differentiation of venous and lymphatic endothelial cells
.
Cell Rep.
35
,
109255
https://doi.org/10.1016/j.celrep.2021.109255
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).