Lymphatic vasculature is increasingly recognized as an important factor both in the regulation of normal tissue homeostasis and immune response and in many diseases, such as inflammation, cancer, obesity, and hypertension. In the last few years, in addition to the central role of vascular endothelial growth factor (VEGF)-C/VEGF receptor-3 signaling in lymphangiogenesis, significant new insights were obtained about Notch, transforming growth factor β/bone morphogenetic protein, Ras, mitogen-activated protein kinase, phosphatidylinositol 3 kinase, and Ca2+/calcineurin signaling pathways in the control of growth and remodeling of lymphatic vessels. An emerging picture of lymphangiogenic signaling is complex and in many ways distinct from the regulation of angiogenesis. This complexity provides new challenges, but also new opportunities for selective therapeutic targeting of lymphatic vasculature.

Lymphatic vessel structure and function

The vascular system consists of blood and lymphatic vessels, both of which are lined by endothelial cells. Lymphatic vasculature is composed of a branched network of capillaries and collecting lymphatic vessels, present in most organs (Figure 1).1  Unlike the blood vasculature, lymphatic capillaries are blind-ended. Small capillaries drain into precollecting and collecting vessels that join the thoracic duct. Lymph flows through collecting vessels and lymph nodes before ultimately being returned to venous circulation at junctions with the subclavian vein.

Figure 1

Organization and the role of lymphatic vasculature in physiology and human disease. Lymphatic capillaries uptake interstitial fluid, lipids, and proteins and serve as entry points to immune cells. Lymphatic collecting vessels transport the lymph toward lymph nodes and to blood circulation. Intraluminal lymphatic valves and SMCs coordinate lymph propulsion and the direction of flow.

Figure 1

Organization and the role of lymphatic vasculature in physiology and human disease. Lymphatic capillaries uptake interstitial fluid, lipids, and proteins and serve as entry points to immune cells. Lymphatic collecting vessels transport the lymph toward lymph nodes and to blood circulation. Intraluminal lymphatic valves and SMCs coordinate lymph propulsion and the direction of flow.

Close modal

The specialized, discontinuous, “button-like” junctions between endothelial cells of lymphatic capillaries represent sites of fluid and immune cell entry into lymphatic vasculature.2  Unlike blood capillaries, lymphatic capillaries have a sparse basement membrane and lack pericytes. They are also attached to extracellular matrix by anchoring filaments. Collecting lymphatic vessels have continuous cell-cell junctions, and they are covered with basement membrane and smooth muscle cells (SMCs). Collecting lymphatic vessels contain intraluminal valves, which consist of 2 semilunar leaflets, covered by a specialized endothelium attached to the core of extracellular matrix.3  High lymph pressure upstream of a valve opens the valve and enables lymph flow, whereas flow in the reverse direction pushes leaflets against each other and closes the valve, depending on changes in fluid pressure within the collecting vessels. SMCs that cover the lymphangions contract and regulate the lymph flow.4 

Main steps of lymphatic vascular development in mammals

The early studies of lymphatic vasculature suggested the 2 potential sources of lymphatic endothelial cells (LECs) during development: blood vessels5  and mesenchymal cells.6  Recent studies, using high-resolution imaging of developing mouse embryos and lineage tracing approaches,7,8  suggest that the majority of LECs in mammals are generated through trans-differentiation from embryonic veins. LEC commitment requires Sry-related Hmg-box 18 (Sox18) and the orphan nuclear receptor chicken ovalbumin upstream promoter transcription factor (COUP-TFII), which up-regulate prospero-related homeodomain transcription factor (Prox1), essential for lymphatic endothelial-specific programming in mammals, but not zebrafish.9-11  Prox1 down-regulates blood markers and vascular endothelial growth factor receptor (VEGFR)-3, which is important for LEC survival, migration, and proliferation. LECs then start to bud by following the gradient of VEGFR-3 ligand, VEGF-C.1  In mice, LECs begin to sprout from the cardinal veins and intersomitic vessels around embryonic day (E)10.7-9  LECs emigrate from veins as nonlumenized, loosely connected strings of cells, so that the integrity of the blood vessel is not disturbed.7,8  LECs further coalesce to form 2 large primordial vessels: the dorsal peripheral longitudinal lymphatic vessel and the ventral primordial thoracic duct, which are also commonly called “lymph sacs” in earlier publications.7  The primordial thoracic duct establishes a connection with the cardinal vein to provide a path for the return of lymph into the blood circulation. The connection is protected by the lymphovenous valve, formed by a small subpopulation of LEC progenitors remaining in the veins,12  which is important for preventing the backflow of blood into the thoracic duct.

Embryonic lymphatic vasculature is established by further sprouting from the primordial lymphatic structures. During late gestation and the postnatal period, the lymphatic capillary plexus is remodeled to establish functional compartments of lymphatic vasculature, capillaries, and the collecting lymphatic vessels.1  During this process, collecting lymphatic vessels develop intraluminal valves and acquire SMCs and basement membrane coverage, whereas lymphatic capillaries remodel cell junctions from the continuous “zipper-like” to the discontinuous button-like state.1,13  Lymphangiogenesis and vessel remodeling continues in many organs, including skin and intestine during early postnatal development, postnatal mouse mammary gland morphogenesis,14  and ovarian follicle growth in adult mice.15 

Lymphatic vessels in disease

In the adult tissues, the majority of lymphatic vessels are quiescent, with the exception of reproductive organs during the ovarian cycle and gestation15 ; however, reactivation of vessels occurs in a variety of pathological conditions. Such vessels may show loss of specialized junctions in capillaries, observed in inflammation,13  and ectopic recruitment of SMCs to lymphatic capillaries, such as in lymphedema.16 

Lymphangiogenesis has been observed in many human inflammatory diseases, including psoriasis and rheumatoid arthritis, and during transplant rejection.17,18  Inflammation-induced lymphangiogenesis regulates fluid drainage, immune cell migration, and removal of inflammatory mediators, ultimately accelerating the resolution of inflammation.19  In contrast, following organ transplantation, enhanced lymphangiogenesis induces reactivation of immune system in the draining lymph node, which results in organ rejection.18  Inflammatory lymphangiogenesis has frequently been shown to occur via secretion of lymphangiogenic growth factors, such as Vegf-c, by macrophages.20  In addition, neutrophils are also recruited to sites of inflammation where they modulate lymphangiogenesis via Vegf-a bioavailability and, to a lesser extent, secretion of Vegf-d.21  Hence, stimulation or inhibition of lymphangiogenesis represents an attractive novel therapeutic strategy for reducing chronic inflammation or transplant rejection.

In cancer, lymph node status is an important factor in determining the stage of disease progression. Many cancers exploit lymphatic vasculature to spread to lymph nodes.22,23  Adding to the disease burden is the development of secondary lymphedema in patients who have undergone radical axillary lymph node dissection.24  Many cancer types induce lymphangiogenesis via release of VEGF-C or VEGF-D, and blocking VEGFR-3 signaling inhibits tumor lymphangiogenesis as well as lymph node metastasis in animal models.25  Increased lymphatic vessel density is correlated with a poor prognosis in a number of solid tumors.22  Intratumoral vessels are thought to be collapsed and nonfunctional due to the high intratumoral fluid pressure.26  In contrast, peritumoral lymphatics are often dilated containing clusters of tumor cells.27,28  15-Lipoxygenase-1-expressing tumor cells, which secrete arachidonic acid metabolites which help to invade lymphatic vessels by inducing the formation of holes in LEC plasma membrane.27  Dilation of collecting vessels through the action of tumor-produced VEGF-C and prostaglandins further contributes to cancer dissemination to lymph nodes.29,30  Lymphangiogenesis in sentinel lymph nodes even before tumors cells metastasize has been shown in mouse models,31  and the extent of lymph node lymphangiogenesis correlates with the disease prognosis in several human cancer types.32,33  Lymph node lymphatic sinuses, but not peripheral lymphatic vessels, express CC chemokine ligand (CCL)-1 chemokine, which regulates the entry of CC chemokine receptor (CCR)-8+ tumor cells into the lymph node.34 

Lymphatic vessel dysfunction has been linked to obesity, and cardiovascular disease such as atherosclerosis. Impaired lymphatic function, such as in Prox1+/− mice, which have leaky lymphatic vessels, leads to adult-onset obesity and inflammation.35  Conversely, lymphatic function is impaired in obese patients.36  Lymphatic vasculature has a direct role in the reverse cholesterol transport, a process critical for the removal of cholesterol from peripheral tissues including the arterial wall.37,38  Interestingly, excessive accumulation of cholesterol in tissues, such as that found in hypercholesterolemic Apoe−/− mice, leads to structural and functional defects of lymphatic vessels.39  In the same mouse model, restoration of lymphatic drainage improved cholesterol clearance.38  Thus, normal lymphatic function is essential for lipid clearance and could be a target for prevention or treatment of atherosclerotic vascular disease.

Skin lymphatic vessels have also been implicated in systemic blood pressure control through regulation of the electrolyte clearance.40  In turn, in hypertension, osmotic stress leads to inflammatory cell recruitment and increased lymphangiogenic growth factor production in the skin.40,41 

In summary, lymphatic vasculature plays a key role in a number of physiological and pathological conditions. Studies of molecular mechanisms and signaling pathways that regulate development of lymphatic vasculature and the pathological lymphangiogenesis are actively underway. In this review, we describe the current view of several critical pathways regulating lymphangiogenesis with a major focus on the mechanisms in mammals (Figure 2). The latest view of lymphangiogenesis in zebrafish is presented in Impel and Schulte-Merker,42  whereas lymphatic vascular remodeling and the role of nonendothelial cells are reviewed in Schulte-Merker et al,1  Harvey and Gordon,20  and Alitalo.43 

Figure 2

Signaling pathways in lymphatic vessels. Lymphatic sprouting and proliferation is regulated by a variety of external stimuli and intracellular signaling pathways. Loss of Vegf-c/Vegfr-3 or Ccbe1 completely prevents formation of lymphatic vessels, demonstrating a central role in lymphangiogenesis. Nrp2 and EphrinB2 enhance VEGF-C-dependent lymphangiogenic sprouting and signaling. Vegfr-2 also contributes to lymphangiogenic response. TGFβ/BMP and angiopoietin-2 are important both for lymphatic capillary patterning and maturation of collecting lymphatic vessels. Dll4/Notch signaling restricts LEC response to Vegf-a in adult tissues, whereas it has a prolymphangiogenic function during postnatal development. The Ras-RAF-mitogen-activated protein kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway promotes lymphatic endothelial proliferation, likely downstream of VEGFR-3. Class I PI3 kinases are required for growth and remodeling of lymphatic vasculature in some vascular beds. Although strongly activated by (lymph)angiogenic growth factors in LECs in vitro, in vivo, the Ca2+/calcineurin pathway is mostly implicated in lymphatic collecting vessel development, in cooperation with Foxc2. Some pathways are also important for establishment of lymphatic endothelial cell identity, such as Notch and Raf/MEK/ERK1/2. Blue boxes indicate pathways also involved in sprouting. Foxc2 and calcineurin/Nfatc1 cooperatively regulate collecting vessel maturation (green box).

Figure 2

Signaling pathways in lymphatic vessels. Lymphatic sprouting and proliferation is regulated by a variety of external stimuli and intracellular signaling pathways. Loss of Vegf-c/Vegfr-3 or Ccbe1 completely prevents formation of lymphatic vessels, demonstrating a central role in lymphangiogenesis. Nrp2 and EphrinB2 enhance VEGF-C-dependent lymphangiogenic sprouting and signaling. Vegfr-2 also contributes to lymphangiogenic response. TGFβ/BMP and angiopoietin-2 are important both for lymphatic capillary patterning and maturation of collecting lymphatic vessels. Dll4/Notch signaling restricts LEC response to Vegf-a in adult tissues, whereas it has a prolymphangiogenic function during postnatal development. The Ras-RAF-mitogen-activated protein kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway promotes lymphatic endothelial proliferation, likely downstream of VEGFR-3. Class I PI3 kinases are required for growth and remodeling of lymphatic vasculature in some vascular beds. Although strongly activated by (lymph)angiogenic growth factors in LECs in vitro, in vivo, the Ca2+/calcineurin pathway is mostly implicated in lymphatic collecting vessel development, in cooperation with Foxc2. Some pathways are also important for establishment of lymphatic endothelial cell identity, such as Notch and Raf/MEK/ERK1/2. Blue boxes indicate pathways also involved in sprouting. Foxc2 and calcineurin/Nfatc1 cooperatively regulate collecting vessel maturation (green box).

Close modal

VEGF-C–VEGFR-3 signaling

VEGF-C, a member of the VEGF growth factor family, is a major regulator of formation and growth of lymphatic vessels, both in physiological and pathological contexts.1,43  In Vegfc−/− mice, LECs fail to migrate out of the veins to form primordial lymphatic vessels, and consequently, mice do not develop lymphatic vasculature.44  This function of Vegf-c is evolutionary conserved, as it is also required for the development of the zebrafish lymphatic vascular system.45  The lymphangiogenic activity of Vegf-c has been demonstrated in a range of animal models involving transgenic and knockout mice, viral gene delivery, and in in vitro assays, where VEGF-C promotes LEC proliferation, survival, and migration.25,46 

Full-length VEGF-C binds receptor tyrosine kinase VEGFR-3 with high affinity, and further proteolytic processing generates a shorter isoform capable of also interacting with VEGFR-2.47 Vegfr-3 is initially expressed in the blood vasculature before becoming restricted to lymphatic vessels around midgestation. Vegfr-3 knockout mice die because of early blood vascular defects.48  Heterozygous missense mutations in Vegfr-3 have been linked to lymphedema in humans and in Chy mice.49,50  A loss-of-function VEGF-C mutation was also recently described in a family with the autosomal dominant form of lymphedema,51  and a truncation mutation in zebrafish Vegf-c severely affected lymphatic development.52  Vegf-c-dependent activation of Vegfr-3 is enhanced by mechanical stretch in an integrin β1-dependent manner, providing a mechanism of how increased interstitial pressure guides expansion of lymphatic vasculature.53 

Neuronal guidance molecule neuropilin-2 (Nrp2) is highly expressed on lymphangiogenic vessels, where it acts as a Vegfr-3 coreceptor during Vegf-c-induced lymphatic sprouting. Nrp2−/− deficient mice have severe hypoplasia of lymphatic capillaries from E13 to birth.54  Furthermore, reduction of lymphatic vessel sprouting was observed in Nrp2+/−;Vegfr3+/− mice, but not in Nrp2+/−;Vegfr2+/− mice, providing genetic evidence for the role of Nrp2 in the Vegf-c/Vegfr-3 signaling cascade.55  Nrp2 levels are reduced in mice with endothelial-specific inactivation of COUP-TFII, and COUP-TFII acts as a direct regulator of Nrp2 expression in LECs.56  In addition, transcription factors GATA binding protein 1 (GATA1), GATA2, and LIM-domain-only protein (LMO)-2 regulate the expression Nrp2 on endothelial cells in vitro.57  Nrp2 is re-expressed during tumor lymphangiogenesis, and treatment with neutralizing anti-Nrp2 antibody decreases tumor lymphangiogenesis and metastasis to sentinel lymph node and distant organs.58 

In addition to Vegfr-3, lymphatic vessels also express high levels of Vegfr-2, whereas Vegfr-1 is predominantly expressed by blood vessels.59  Inactivation of Vegfr-2 in LECs results in decreased lymphatic sprouting and proliferation, without affecting vessel caliber.60  Interestingly, the VEGFR-2-specific ligand, VEGF-E, induces circumferential growth without affecting lymphatic sprouting.61  Thus, it is possible that ligand-dependent Vegfr-2 signaling contributes to LEC proliferation, whereas sprouting is regulated through a ligand-independent Vegfr-2 modulation of Vegfr-3/Nrp2 signaling (eg, via Vegfr-2/3 heterodimers) present in the LEC filipodia.62 

Collagen- and calcium-binding epidermal growth factor-like domains 1

Collagen- and calcium-binding epidermal growth factor-like domains 1 (Ccbe1) is a secreted extracellular matrix-binding protein, mutated in patients with Hennekam syndrome,63  a rare disease that presents with lymphedema, lymphangiectasia, and other pathological features.64  The function of Ccbe1 is evolutionary conserved, as it is required for embryonic lymphangiogenesis in zebrafish65  and mice.66  Ccbe1 is expressed in the somitic mesoderm during thoracic duct formation in zebrafish and in the areas adjacent to the anterior cardinal vein in mouse embryos, where it acts non–cell autonomously to regulate Vegf-c processing and bioavailability.65,67,68  Lymphangiogenesis is also potently induced on coadministration of Ccbe1 and Vegf-c in a corneal lymphangiogenesis mouse model.66 Ccbe1−/− mice develop severe anemia because of the defective definitive erythropoiesis, suggesting that Ccbe1 has additional,Vegf-c-independent function(s).69  Ccbe1 is a direct target of atypical E2F transcription factors E2F7/8. Importantly, inactivation of e2f7/8 in zebrafish reduced Ccbe1 expression and impaired venous sprouting and lymphangiogenesis, whereas overexpression of e2f7/8 rescued Ccbe1- and Vegfr-3-dependent lymphangiogenesis.70 

Ephrin-Eph

Eph receptor tyrosine kinase receptors and their ligands, ephrins, are key regulators of axon guidance during nervous system development, and they also contribute to many aspects of blood vascular morphogenesis.71  In addition to acting as Eph-activating ligands (so-called forward signaling), ephrins are also able to initiate reverse signaling, which in the case of ephrinB ligands is achieved through the recruitment of adaptor proteins in the cytoplasmic portion. Lymphatic vessels express both EphrinB2 and EphB4, an ephrinB2 receptor.72,73  EphrinB2 promotes Vegf-c/Vegfr-3 signaling in the developing LECs through the regulation of Vegfr-3 internalization, important for full activation of downstream signaling pathways, such as Akt (also known as protein kinase B) and ERK.74  EphrinB2 has a similar function in the regulation of Vegfr-2 on blood vessels and platelet-derived growth factor receptor ß signaling in pericytes.75,76  Blocking ephrinB2 activity using antibodies prevents tumor angiogenesis and lymphangiogenesis, suggesting a potential therapeutic approach for targeting tumor neovascularization.77 

Loss of the C-terminal PDZ (post synaptic density protein [PSD95], Drosophila disc large tumor suppressor [Dlg1], zonula occludens-1 protein [zo-1]) domain of ephrinB2, implicated in reverse signaling, also leads to defective remodeling of primary lymphatic plexus and failure to form collecting lymphatic vessels and lymphatic valves.72  Studies of adult corneal lymphatic vessels showed that EphB4 is highly expressed in the valves where administration of EphB4-Fc fusion proteins prevented regeneration of valves after corneal injury.73 

Angiopoietins and Tie receptors

Tie1 and Tie2 endothelial receptor tyrosine kinases and their angiopoietin (Ang) ligands play important roles in blood vessel maturation, patterning, and stability, both in physiological and pathological conditions.78  The current view of Ang-Tie signaling in blood vessels suggests that Ang1 acts to stabilize vessels and limit angiogenesis,78  although this was challenged by a genetic study that found that Ang1 is dispensable for maintenance of quiescent vasculature in adult animals.79  Ang2 is in many cases antagonistic to Ang1, but it can be also agonistic under certain conditions, such as at high concentration and/or in tumor endothelial cells. Ang1, Tie1, and Tie2, but not Ang2, are essential for embryonic vascular development, and their inactivation leads to embryonic lethality at E12.5 (reviewed in Eklund and Saharinen78 ). Decreased dosage of Tie1 leads to embryonic edema and mispatterning of early lymphatic vasculature, without affecting the lymphatic endothelial commitment.80,81  It is not known whether these defects are a result of modified Ang1 or Tie2 signaling in LECs or whether Tie1 has an angiopoietin-independent role in lymphatic endothelium. In contrast to its limited role during physiological angiogenesis, Ang2 plays an essential role in lymphatic vasculature. Angpt2−/− mice have hypoplastic lymphatic capillaries and fail to form collecting vessels, which leads to lymphatic dysfunction and impaired postnatal survival.82,83  Interestingly, Ang1 rescues the lymphatic vascular, but not blood vascular defects in Angpt2−/− mice, and overexpression of Ang1 induces both physiological and pathological lymphangiogenesis in a Vegfr-3-dependent manner,82,84,85  highlighting the differences in the use of angiopoietin ligands by the 2 vascular systems. Studies of blood vasculature revealed important roles of the Ang-Tie pathway in a variety of pathological conditions, such as inflammation, fibrosis, and cancer, underlining their importance as therapeutic targets.78,86,87  However, the relative contribution of Ang-Tie signaling in lymphatic vessels to these processes still needs to be fully examined.

Notch signaling

Notch is a fundamental signaling pathway, which plays a key role in blood vascular development and function. The Notch pathway is mediated by Notch receptors (Notch1-Notch4), as well as Delta-like (Dll1, Dll3, Dll4) and Jagged (Jag1 and Jag2) ligands. Notch receptors are single-pass transmembrane proteins that function both as cell surface receptors and transcriptional regulators. Canonical Notch signaling is triggered via the interaction of Notch receptor and ligands on neighboring cells, which induces a conformational change in Notch receptors, cleavage of the Notch intracellular domain (NICD), and translocation of the NICD in the nucleus. The NICD interacts with DNA-binding protein RbpJ, constitutively recruited to the promoters of Notch target genes, and replaces the associated corepressor complex. Primary Notch target genes include Hes and Hey transcription factors.88  In blood vessels, Notch signaling regulates angiogenic sprouting, by controlling the selection of stalk and tip cells, the recruitment of SMC/pericytes, and arterio-venous differentiation.89 

Cultured human LECs express high levels of DLL4, NOTCH1, NOTCH4, and JAG1.90-92  Blocking Notch signaling in cultured LECs and in adult mice using the Dll4-Fc fusion protein leads to lymphatic endothelial hypersprouting and increases responsiveness of LECs to Vegf-a, indicating that under these conditions Notch function is analogous to its role in blood vessels, ie, restriction of (lymph)angiogenic potential of endothelial cells.92  However, inactivation of Notch during early postnatal development or in a wound healing model in mice using Dll4 or Notch1 blocking antibodies or in zebrafish embryos using a morpholino oligo knockdown approach results in decreased lymphatic vessel sprouting.91,93 

In vitro studies also suggested the role of NOTCH signaling in lymphatic endothelial differentiation through the repression of PROX1 and COUP-TFII.94  Down-regulation of Notch activity in vivo resulted in increased generation of LEC progenitors, whereas overexpression of NICD in LECs between E9.75 and E13.5 repressed COUP-TFII and Prox1,95  demonstrating that Notch acts as a negative regulator of LEC specification. Taken together, these data reveal a complex picture of Notch signaling in lymphatic vasculature, where it can be either pro- or antilymphangiogenic or regulate venous vs LEC fate decisions, probably depending on the developmental stage or tissues investigated.

Transforming growth factor/bone morphogenetic protein signaling

Transforming growth factor (TGF) and bone morphogenetic protein (BMP) signaling pathways induce pleiotropic tissue-specific responses by regulating proliferation, differentiation, migration, and cellular survival. TGF/BMP ligands (BMP2, BMP7, GDF5, BMP9, BMP10, AMH, TGF-β1/2/3, Activins, and Nodals) bind TGF receptors type II (TGFRII) (BMPR2, ACVR2A/B, and TGFβRII), which phosphorylate TGFRI (Alk1-7) via Ser/Thr tyrosine kinase activity. Downstream signaling pathways are transmitted to the cell nucleus via phosphorylation of Mad (mothers against decapentaplegic gene in Drosophila) and Sma genes (SMAD) transcription factors, which trigger target gene transcription.96  In addition, the noncanonical TGFβ signaling pathway regulates diverse cellular responses involving mitogen-activated protein kinases (MAPK), ρ-like GTPases, and PI3k/Akt.97  Endothelial cells signal through 2 type I TGFRs: Alk5 (TGFβRI) and Alk1 (ACVRL1), which activate SMAD2/3 and SMAD1/5, respectively.97  Mutations of the components of these signaling pathways cause several human hereditary vascular diseases, such as hereditary hemorrhagic telangiectasia (ENDOGLIN or ALK1) or pulmonary arterial hypertension (BMPR2).97  Studies of genetic mouse models revealed critical roles of TGF-β/BMP signaling in angiogenic sprouting.98-100  TGF/BMP also play an important role in the regulation of vascular integrity in the brain,101  maintenance of the quiescent vasculature,102  and SMC differentiation and recruitment to blood endothelial cells.103-105 

Cultured LECs express Alk1, Alk2, Alk4, Alk5, ACVR2B, BMPR2, Endoglin, and TGFβRII receptors.106  In vitro treatment of LECs with TGFβ-1 reduces cell proliferation, cord formation, and expression of lymphatic markers Prox1 and lymphatic vessel endothelial hyaluronic acid receptor 1 (Lyve-1).107  TGFβ-1 inhibits lymphatic endothelial commitment and down-regulates related markers, such as COUP-TFII and Sox18, in murine embryonic stem cell-derived Flk1+ cardiovascular progenitors,107  although in another study, TGFβ-2 treatment increased expression of Vegfr-3 and Nrp2, both involved in lymphatic sprouting events rather that lymphatic endothelial commitment.108  Similarly, BMP9 suppresses the expression of LYVE1 and PROX1.109,110 

In contrast to the in vitro results, endothelial-specific loss of TgfbrI (Alk5) or TgfbrII during embryogenesis did not affect lymphatic endothelial cell commitment. Instead, it impaired formation of tip cells and reduced complexity of skin lymphatic network, leading to hyperplasia of cutaneous lymphatic vessels.108  These in vivo results suggest that TGFβ signaling is an important regulator of lymphatic network patterning.108  BMP2 is known to interact with BMPR2, ACVR2A, or ACVR2B as type II TGF receptors and Alk-2, Alk-3, or Alk-4 as type I receptors. In zebrafish embryos, Bmp2 signaling pathway acts as a negative regulator of LEC emergence from veins, and overexpression of Bmp2b suppresses formation of lymphatic vascular structures, presumably due to the loss of Prox1a.111  However, it should be noted that the genetic inactivation of Prox1a does not affect zebrafish lymphangiogenesis.11 

Inhibition of TGFβ/BMP signaling during the early postnatal development using Alk1, Acvr2b, and Bmbpr2 blocking antibodies prevented growth of lymphatic vessels in several organs, including the skin and intestine, without affecting collecting lymphatic vessels.106  This effect was further enhanced by blocking Vegfr-3 signaling. Thus, Alk1 signaling appears to play a major role in postnatal lymphangiogenesis, and in its absence, lymphatic vessels become more sensitive to Vegfr-3–Vegf-c/d depletion.106  At present, it is not clear what ligand is responsible for this effect. Indeed, targeted inactivation of high-affinity Alk1 ligand Bmp9 has an opposite effect, as it induces lymphatic vascular hyperplasia and lymphatic valve defects109,110 ; thus, additional Alk1 ligands that may be contributing to lymphangiogenesis remain to be investigated.

Several studies also addressed the question of the role of TGFβ signaling in pathological lymphangiogenesis. Treatment using a small-molecule TGFβRI inhibitor increased lymphangiogenesis in chronic peritonitis and tumor xenografts.107  In a model of tail lymphedema, treatment with blocking TGFβ antibody similarly increased lymphangiogenesis and alleviated inflammation, lymphedema, and fibrosis.112 

To conclude, TGF/BMP signaling is important for several steps of lymphatic vascular development, including sprouting/tip cell formation, regulation of cell proliferation, and formation of specialized lymphatic structures, such as intraluminal valves.107,108,113  The future challenge will be to elucidate the precise roles of different receptors and ligands in these processes, as well as to understand potential combinatorial interactions with other signaling pathways, such as Notch, already revealed in blood vasculature.99,100 

Lymphangiogenesis is regulated by a number of ligand-receptor interactions, which in turn activate intracellular signaling pathways responsible for transmitting information within the cell, and ultimately endothelial cell responses, such as proliferation, survival, or migration. Much effort is dedicated to studies of such ligands and receptors, because they represent important targets for drug development. However, in the last few years, novel knowledge has also been accumulated on the intracellular pathways, which in many cases revealed surprising differences between the blood and lymphatic vasculature (Table 1 114-128 ). In the following sections, we will describe the current knowledge on the major intracellular signaling pathways in lymphangiogenesis.

Table 1

Comparison of blood and lymphatic vascular phenotypes in loss-of-function genetic mouse models

GenesBlood vessel phenotypeLymphatic vessel phenotype
Ligands and receptors   
 Vegf-c Vegfc−/−: Normal developmental angiogenesis44  Vegfc−/−: No sprouting of LECs, absence of lymphatic vessels44 
Vegfc+/−: Chylous ascites, hypoplasia44  
 Vegfr-3 Vegfr3−/−: Early blood vessel remodeling defects, death at E9.5-1048 
Vegfr3f/f;Pdgfb-CreERT2: Vessel hypersprouting114  
Vegfr3+/Chy: Chylous ascites, hypoplasia50 
Vegfr3ΔLBD/ΔLBD: No sprouting of LECs due to the removal of ligand-binding (LBD) domain of Vegfr-3115  
 Nrp2 Nrp2−/−: Normal developmental angiogenesis54  Nrp2−/−: Hypoplasia of capillaries E13 to birth54 
Nrp2+/−;Vegfr3+/−: Decreased vessel sprouting55  
 EphrinB2 Efnb2f/f;Cdh5-CreERT2: Reduced vascular network complexity, sprouting and endothelial cell proliferation74 
Efnb2ΔV/ΔV: Normal developmental angiogenesis72  
Efnb2f/f;Cdh5-CreERT2: Decreased vessel sprouting74 
Efnb2ΔV/ΔV: Defective lymphatic vascular remodeling, hyperplasia, valve agenesis72,74  
 Vegfr-2 Vegfr2−/−: Lack of development of the blood islands and embryonic vasculature, death at E8.5-E9.5116 
Vegfrf/f;Lyve-1Cre: Decreased blood vessel density in the yolk sac, liver and lung60  
Vegfrf/f;Lyve-1Cre: Hypoplasia, decreased vessel sprouting60  
 Bmp9 Bmp9−/−: No phenotype in the retinal blood vessels117  Bmp9−/−: Postnatal vessel hyperplasia, decreased maturation of valves. Adults: enlarged lymphatics, decreased number of lymphatic valves, impaired lymph drainage109,110  
 TGFβ2 Tgfb2−/−: Blood vascular remodeling occurs normally108  Tgfb2−/−: Mild edema, increased cellular proliferation, decreased complexity and sprouting, enlarged vessels108  
 TGFβRI TgfbrIf/f;Tie1Cre: Defective vascular network, pericardial effusion in the heart, abnormalities in the vasculature of the yolk sac at E9.5, lethal at E10.5103 
TgfbrIf/f;Tie2Cre: Hypoplastic endocardial cushion in the arterio-ventricular canal; thinner, poorly trabeculated myocardium, lethal soon after E13118 
TgfbrIf/f;Alk1GFPCre/+: Enlarged pericardial cavity, underdeveloped heart, lethal at E14.5102  
TgfbrIf/fProx1+/GFPCre: Deletion at E9.5-10.5. Edema at E14.5 with blood filled lymphatics. Lymphatic network and sprouting reduction106 
TgfbrIf/f;Prox1-CreERT2: Deletion at E12.5 impaired cell sprouting and hyperplasia106 
TgfbrIf/f;VEC-CreERT2: Deletion at E12.5, mild edema, hyperplasia, lymphatic network and sprouting reduction106  
 TGFβRII TgfbrIIf/f;Tie1Cre: Pericardial effusion in the heart, abnormalities in the vasculature of the yolk sac at E9.5, lethal at around E10.5103 
TgfbrIIf/fl;Tie2Cre: Cardiac defects and lethality at around E12.5119 
TgfbrIIf/f;Pdgfb-CreERT2: Hemorrhagic blood vessels in retina, impaired vascular development96  
TgfbrIIf/f;Prox1+/GFPCre: Deletion at E9.5-10.5. Edema at E14.5 with blood – filled lymphatics. Lymphatic network and sprouting reduction106 
TgfbrIIf/f;Prox1-CreERT2: Deletion at E12.5 presence of dysmorphogenic lymphatic vessels and reduced lymphatic branching106 
TgfbrIIf/f;VEC-CreERT2: Deletion at E12.5, mild edema, hyperplasia, lymphatic network and sprouting reduction106  
 Notch1 Notch1f/f;Cdh5-CreERT2: Regional retinal vessel hypersprouting120  Notch1f/f;Prox1CreERT2: Overproduction of LECs, edema, blood-filled lymphatics and incorporation of BECs into the peripheral lymphatics95  
 Ccbe1 Ccbe1−/−: Normal physiological angiogenesis66  Ccbe1−/−: No sprouting of LECs, absence of lymphatic vessels66 
Ccbe1+/−: Early irregular formation of intersegmental veins; no lymphatic phenotype later7  
 Angipoietins/Tie Angpt1−/−, Tie1−/−, Tie2−/−: Defective embryonic blood vessel remodeling, lethality at E12.548121 
Angpt2−/−: Normal embryonic angiogenesis, defective hyaloid vessel regression82  
Angpt2−/−: Hypoplasia of lymphatic capillaries, failure to form collecting vessels82,83 
Tie1−/−: Abnormal patterning, dilated and disorganized
lymphatic vessels80,81  
Intracellular pathways   
 H/N/Kras Nras+/−;Kras+/−: Normal developmental angiogenesis122  Nras+/−;Kras+/−: Lymphatic hypoplasia, chylous ascites122  
 Rasa1 Rasa1−/−: Early blood vascular remodeling defects, lethality at E9.5123 
Rasa1f/f;UB-ERT2Cre: Systemic inactivation in adults-normal blood vessels124  
Rasa1f/f;UB-ERT2Cre: Systemic inactivation in adults leading to hyperplasia, increased leakage, chylothorax, chylous ascites124  
 PI3K/Akt1 Pi3kca−/−(p85/p55/p50): Minor defects of developmental angiogenesis125 
Akt1−/−: Normal physiological angiogenesis126  
Pi3kca−/−(p85/p55/p50): Impaired valve development, organ-specific hypoplasia125 
Pi3kp110Rbd/Rbd: Chylous ascites, decreased branching and network complexity125 
Akt1−/−: Reduced capillary density, defective valve development140  
 Cnb1 Cnb1f/f;Tie2-Cre: Defective coronary vasculature127 
Cnb1f/f;Pdgfb-CreERT2: Normal physiological angiogenesis128  
Cnb1f/f;Prox1-CreERT2: Defective lymphatic vessel maturation and valve development128  
GenesBlood vessel phenotypeLymphatic vessel phenotype
Ligands and receptors   
 Vegf-c Vegfc−/−: Normal developmental angiogenesis44  Vegfc−/−: No sprouting of LECs, absence of lymphatic vessels44 
Vegfc+/−: Chylous ascites, hypoplasia44  
 Vegfr-3 Vegfr3−/−: Early blood vessel remodeling defects, death at E9.5-1048 
Vegfr3f/f;Pdgfb-CreERT2: Vessel hypersprouting114  
Vegfr3+/Chy: Chylous ascites, hypoplasia50 
Vegfr3ΔLBD/ΔLBD: No sprouting of LECs due to the removal of ligand-binding (LBD) domain of Vegfr-3115  
 Nrp2 Nrp2−/−: Normal developmental angiogenesis54  Nrp2−/−: Hypoplasia of capillaries E13 to birth54 
Nrp2+/−;Vegfr3+/−: Decreased vessel sprouting55  
 EphrinB2 Efnb2f/f;Cdh5-CreERT2: Reduced vascular network complexity, sprouting and endothelial cell proliferation74 
Efnb2ΔV/ΔV: Normal developmental angiogenesis72  
Efnb2f/f;Cdh5-CreERT2: Decreased vessel sprouting74 
Efnb2ΔV/ΔV: Defective lymphatic vascular remodeling, hyperplasia, valve agenesis72,74  
 Vegfr-2 Vegfr2−/−: Lack of development of the blood islands and embryonic vasculature, death at E8.5-E9.5116 
Vegfrf/f;Lyve-1Cre: Decreased blood vessel density in the yolk sac, liver and lung60  
Vegfrf/f;Lyve-1Cre: Hypoplasia, decreased vessel sprouting60  
 Bmp9 Bmp9−/−: No phenotype in the retinal blood vessels117  Bmp9−/−: Postnatal vessel hyperplasia, decreased maturation of valves. Adults: enlarged lymphatics, decreased number of lymphatic valves, impaired lymph drainage109,110  
 TGFβ2 Tgfb2−/−: Blood vascular remodeling occurs normally108  Tgfb2−/−: Mild edema, increased cellular proliferation, decreased complexity and sprouting, enlarged vessels108  
 TGFβRI TgfbrIf/f;Tie1Cre: Defective vascular network, pericardial effusion in the heart, abnormalities in the vasculature of the yolk sac at E9.5, lethal at E10.5103 
TgfbrIf/f;Tie2Cre: Hypoplastic endocardial cushion in the arterio-ventricular canal; thinner, poorly trabeculated myocardium, lethal soon after E13118 
TgfbrIf/f;Alk1GFPCre/+: Enlarged pericardial cavity, underdeveloped heart, lethal at E14.5102  
TgfbrIf/fProx1+/GFPCre: Deletion at E9.5-10.5. Edema at E14.5 with blood filled lymphatics. Lymphatic network and sprouting reduction106 
TgfbrIf/f;Prox1-CreERT2: Deletion at E12.5 impaired cell sprouting and hyperplasia106 
TgfbrIf/f;VEC-CreERT2: Deletion at E12.5, mild edema, hyperplasia, lymphatic network and sprouting reduction106  
 TGFβRII TgfbrIIf/f;Tie1Cre: Pericardial effusion in the heart, abnormalities in the vasculature of the yolk sac at E9.5, lethal at around E10.5103 
TgfbrIIf/fl;Tie2Cre: Cardiac defects and lethality at around E12.5119 
TgfbrIIf/f;Pdgfb-CreERT2: Hemorrhagic blood vessels in retina, impaired vascular development96  
TgfbrIIf/f;Prox1+/GFPCre: Deletion at E9.5-10.5. Edema at E14.5 with blood – filled lymphatics. Lymphatic network and sprouting reduction106 
TgfbrIIf/f;Prox1-CreERT2: Deletion at E12.5 presence of dysmorphogenic lymphatic vessels and reduced lymphatic branching106 
TgfbrIIf/f;VEC-CreERT2: Deletion at E12.5, mild edema, hyperplasia, lymphatic network and sprouting reduction106  
 Notch1 Notch1f/f;Cdh5-CreERT2: Regional retinal vessel hypersprouting120  Notch1f/f;Prox1CreERT2: Overproduction of LECs, edema, blood-filled lymphatics and incorporation of BECs into the peripheral lymphatics95  
 Ccbe1 Ccbe1−/−: Normal physiological angiogenesis66  Ccbe1−/−: No sprouting of LECs, absence of lymphatic vessels66 
Ccbe1+/−: Early irregular formation of intersegmental veins; no lymphatic phenotype later7  
 Angipoietins/Tie Angpt1−/−, Tie1−/−, Tie2−/−: Defective embryonic blood vessel remodeling, lethality at E12.548121 
Angpt2−/−: Normal embryonic angiogenesis, defective hyaloid vessel regression82  
Angpt2−/−: Hypoplasia of lymphatic capillaries, failure to form collecting vessels82,83 
Tie1−/−: Abnormal patterning, dilated and disorganized
lymphatic vessels80,81  
Intracellular pathways   
 H/N/Kras Nras+/−;Kras+/−: Normal developmental angiogenesis122  Nras+/−;Kras+/−: Lymphatic hypoplasia, chylous ascites122  
 Rasa1 Rasa1−/−: Early blood vascular remodeling defects, lethality at E9.5123 
Rasa1f/f;UB-ERT2Cre: Systemic inactivation in adults-normal blood vessels124  
Rasa1f/f;UB-ERT2Cre: Systemic inactivation in adults leading to hyperplasia, increased leakage, chylothorax, chylous ascites124  
 PI3K/Akt1 Pi3kca−/−(p85/p55/p50): Minor defects of developmental angiogenesis125 
Akt1−/−: Normal physiological angiogenesis126  
Pi3kca−/−(p85/p55/p50): Impaired valve development, organ-specific hypoplasia125 
Pi3kp110Rbd/Rbd: Chylous ascites, decreased branching and network complexity125 
Akt1−/−: Reduced capillary density, defective valve development140  
 Cnb1 Cnb1f/f;Tie2-Cre: Defective coronary vasculature127 
Cnb1f/f;Pdgfb-CreERT2: Normal physiological angiogenesis128  
Cnb1f/f;Prox1-CreERT2: Defective lymphatic vessel maturation and valve development128  

RBD, Ras-binding domain.

Ras/Raf/MEK/ERK

The Ras family of GTPases (Hras, Nras, and Kras) plays essential roles in a variety of cellular functions, such as proliferation, migration, differentiation, and apoptosis, frequently downstream of receptor tyrosine kinases. Ras is a small molecule GTPase, which in its active, GTP-bound state, can activate multiple downstream effectors, including the Raf/MEK/ERK pathway and class I phosphatidylinositol 3 kinases (PI3Ks). A variety of associated proteins (guanine-nucleotide exchange factors) increase Ras GTPase activity by promoting GDP-GTP exchange, whereas GTPase-activating proteins, or Ras-GAPs, stimulate the hydrolysis of GTP on Ras and are important for Ras inactivation.129  In the Raf/MEK/ERK pathway, active Ras interacts with RAF1 Ser/Thr kinase, which in turn activates downstream MEK kinases. MEK kinases then phosphorylate and activate ERK1/2 MAPK.130 

Ras signaling plays an important role in lymphatic vessel development and function, likely downstream of the VEGFR-3 pathway. Nras+/−Kras+/− mice display lymphatic hypoplasia and chylous ascites, which are rescued by overexpression of Hras.122  Conversely, mice that overexpress Hras in endothelial cells have lymphatic vessel hyperplasia, driven by increased levels of Vegfr-3 and enhanced MAPK signaling. However, such hyperplastic vessels are not fully functional, as transgenic mice develop edema and chylothorax. In humans, chylous ascites, chylothorax and lymphedema are associated with activating mutations in KRAS in cardiofaciocutaneous syndrome131  and HRAS in Costello syndrome.132,133  The work in mouse models also revealed differential sensitivity of blood vs lymphatic vessels to altered Ras signaling, as blood vessel development or function was not affected on Hras overexpression or in Kras+/−, Nras+/− mice.122 

Negative regulator of Ras activity p120-RasGap (RASA1) is mutated in human hereditary disease capillary malformation–arteriovenous malformation (CM-AVM), characterized by multiple cutaneous vascular lesions and high risk for development of fast-flow blood vascular lesions.134  Lymphatic malformations were observed in some CM-AVM patients,135  and recent advanced imaging analysis revealed hyperplastic cutaneous lymphatic vessels in a CM-AVM patient, suggesting that lymphatic vascular dysfunction may be frequent in CM-AVM.136  Inactivation of Rasa1 in adult mice causes extensive Vegfr-3-dependent lymphatic vessel hyperplasia and lymphatic vessel leakage, leading to chylothorax, chylous ascites, and animal death.124  Importantly, no blood vasculature phenotype was observed on loss of Rasa1 in adult mice, unlike in Rasa1-deficient embryos,123  implying that blood vessels need Ras regulation only during embryonic angiogenesis, whereas lymphatic vessels require a lifelong suppression of Ras signaling. A number of other Ras signaling effectors and regulators have been implicated in vascular development and angiogenesis, such as Rasip1137 ; therefore, it will be interesting to investigate and compare their function in lymphatic vessels as well.

Studies of blood vasculature revealed important interactions of PI3K-Akt and Erk signaling in the regulation of arterio-venous differentiation, in which high PI3K-Akt signaling favors venous fate and vein formation, whereas arterial morphogenesis is induced by Erk1/2 MAPK under conditions of attenuated PI3K-Akt activation.138,139  In addition to the regulation of lymphatic hyperplasia downstream of VEGFR-3 signaling, as discussed above, MAPK/ERK cascade might be also important for the establishment of LEC fate. Endothelial-specific expression of activated MAPK via upstream regulator Raf1, increased commitment of venous endothelial cells to the lymphatic fate and even induced lymphatic markers in arterial endothelial cells through the induction of Sox18 and Prox1.140  Spatially localized, yet unknown, signal may therefore regulate Erk activation in embryonic veins and lead to the establishment of lymphatic endothelial fate in a subpopulation of venous ECs.

Class I PI3Ks

PI3Ks are a family of enzymes that phosphorylate the 3-OH group of inositol membrane lipids and produce a variety of 3-phosphorylated phosphoinositides. The latter propagate intracellular signaling by providing docking sites for pleckstrin-homology domains of diverse signaling proteins. Downstream effectors of PI3K include the serine/threonine kinase Akt that, among other pathways, activates mammalian target of rapamycin (mTOR) signaling, which is central for cellular proliferation, survival, and metabolism. The PI3K/Akt/mTOR pathway is frequently activated in cancers, and a number of PI3K and mTOR inhibitors are either in clinical trials or already approved for treatment.141,142 

Based on their primary structure, regulation, and substrate specificity, 3 classes of PI3Ks (Ia, Ib, II, and III) were identified. Class Ia PI3K is a group of dimeric proteins containing a catalytic subunit (p110α, p110β, or p110δ) and a regulatory subunit (p85α, p55α, p50α, p85β, or p55γ) that are activated by a variety of receptor tyrosine kinases. Regulatory p85 subunit directly binds phosphotyrosines on receptor tyrosine kinases or adaptor proteins, which leads to PI3K activation. PI3K can also be activated directly by Ras, which interacts with the p110 subunit.143  Studies of animal models have uncovered multiple functions of class I PI3Ks in blood vascular development, such as regulation of blood vessel integrity, angiogenic sprouting, and remodeling, as well as arterio-venous differentiation.144  VEGF-A, VEGF-C, and VEGF-D activate the PI3K-Akt signaling cascade in cultured LECs, where this pathway is important for migration145  via a direct interaction with VEGFR-3.146  LECs express p110α, -β, and -δ but not -γ isoforms, where p110α is a major isoform, responsible for VEGF-C-dependent Akt activation and LEC migration.147  Germ-line deletion of Pik3r1, encoding the regulatory subunits p85α, p55α, and p50α of class I PI3Ks, led to chylous ascites, suggesting lymphatic vascular dysfunction.148  Further analysis of these mice revealed organ-specific defects in postnatal lymphatic sprouting and lymphatic maturation, without major impact on blood vessel development.125  Mutations in the Pi3kca encoding the catalytic p110α isoform, which block its interaction with Ras, led to lymphatic vessel hypoplasia, reduced sprouting, and perinatal chylous ascites.149  In a tumor setting, p110α signaling downstream of VEGF-C is important for the activation of integrin α4β1 on lymph node lymphatic endothelium and subsequent adhesion of metastatic tumor cells.147  In human disease, somatic activating mutations in PI3KCA were found in patients with Congenital Lipomatous Overgrowth, Vascular Malformations and Epidermal Nevi Syndrome and Klippel-Trenaunay-Weber syndrome, which present with malformation or overgrowth of lymphatic vessels in addition to tissue overgrowth and other vascular anomalies.150 

Three highly related isoforms of Akt serine/threonine kinase (Akt1, Akt2, and Akt3) represent the major signaling arm of PI3K. All isoforms are expressed in LECs; however, Akt1 has a dominant role in lymphatic vasculature, as Akt1−/−, but not Akt2−/− or Akt3−/−, mice display reduced capillary density and defective valve development.126  Interestingly, VEGF-C-induced lymphangiogenesis is normal in Akt1−/− mice, indicating either compensation by other isoforms or a predominant role in cell survival rather than migration or proliferation.126  As observed in mice with mutant Ras or PI3K signaling components, the physiological angiogenesis was not affected in the absence of Akt1. The activating AKT1 mutation underlies Proteus syndrome, characterized by general tissue overgrowth, as well as cutaneous vascular lymphatic or lympho-venous malformation.151 

Ca2+/calcineurin/NFAT cells signaling

Calcineurin is a calcium-activated protein phosphatase, which plays a major role in calcium signaling in different cells types, and it is a target of immunosuppressive drugs cyclosporine A and tacrolimus. An increase in intracellular calcium activates calcineurin and leads to dephosphorylation and nuclear translocation of the transcription factors of the nuclear factor of activated T cells (NFAT) family, which act as main effectors of the Ca2+/calcineurin pathway. In vitro studies suggested that calcineurin/NFAT mediates most VEGF/VEGFR-2-induced responses of endothelial cells, including cell proliferation, migration, and tube formation.152-154  However, mice with constitutive endothelium-specific inactivation of calcineurin display coronary vessel-specific, but not generalized, angiogenesis defects.127  Angiogenesis proceeds normally on inducible inactivation of calcineurin in blood vasculature after E13.5, when the requirements for calcineurin signaling in heart development are bypassed.128  These data suggest a limited vascular bed-specific role for endothelial calcineurin signaling during physiological angiogenesis. Calcineurin may be more important for pathological angiogenesis, as mice deficient in Down syndrome critical region gene 1, an endogenous inhibitor of calcineurin, have decreased tumor angiogenesis.155 

To date, of the 5 members of the NFAT family, only NFATc1 (NFAT2) is known to be implicated in lymphatic vascular development.156,157  In LECs, expression of NFATc1 is controlled by PROX1, whereas its nuclear translocation can be induced by VEGF-C through the activation of VEGFR-2 or by flow shear stress. The Ca2+/calcineurin/NFAT pathway cooperates with FOXC2, a major regulator of collecting vessel phenotype.128,156  Pharmacological inhibition or lymphatic-endothelial-specific inactivation of calcineurin prevents maturation of lymphatic vessels and arrests the development of lymphatic valves.128,156  Thus, similar to blood vasculature, the role of calcineurin signaling in lymphatic vessels in physiological conditions is restricted to a specific vascular compartment.

MicroRNAs and lymphangiogenesis

MicroRNAs (miRs) are short noncoding single stranded RNAs, which act as post-transcriptional regulators by inhibiting mRNA translation and/or promoting mRNA degradation. The same miR can regulate many mRNAs; therefore, miRs act as broad regulators of gene networks, akin to the transcription factors, although miRs act not as binary on and off switches, but rather to fine-tune the genetic program. The number of known miRs that regulate angiogenesis either in a cell autonomous manner or through the regulation of angiogenic factors in other cells is constantly increasing.158-160  However, in contrast to the wealth of information on miRs in angiogenesis, little is known about specific miR functions in LECs. miR-181a is expressed at higher levels in blood endothelial cells (BECs) in comparison with LECs, and it targets Prox1 mRNA for degradation. Increasing miR-181a levels in LECs reduced Prox1 expression and shifted LECs toward a blood vascular phenotype, suggesting that miR-181a acts to maintain the blood vascular phenotype.161  In another study, quantitative reverse transcription-polymerase chain reaction profiling of 157 miRs in cultured LECs and BECs identified LEC-specific miR-95 and miR-326 and BEC-specific miR-137, miR-31, miR-125b, and miR-99a.162  Further analysis demonstrated that miR-31 suppresses a number of lymphatic endothelial-specific genes, and this effect is at least partially due to the direct suppression of PROX1.162  Thus, higher expression of miR-31 and miR-181a may be important for preventing the acquisition of LEC identity by BECs. Factors regulating the expression of miR-31 and miR-181a in endothelial cells include BMP2.111 

Lymphatic vessels perform a vast array of functions both in physiological and pathological conditions. Understanding molecular mechanisms regulating lymphatic vascular growth and remodeling should result in the design of new therapies for many human diseases. In the last few years, in addition to the well-established roles of VEGF-C/VEGFR-3 signaling in lymphangiogenesis, important contributions were uncovered for the Notch, TGFβ/BMP, Ras, MAPK/ERK, PI3K/Akt, and Ca2+/calcineurin pathways. It is noteworthy that in many cases lymphatic and blood vasculature demonstrates differential responses or use of these signaling pathways (Table 1). One of the future challenges will be to decipher the complexity of such signaling in different lymphatic vascular beds and disease conditions, and to identify critical determinants, which could be used for modulation of lymphatic function.

The authors apologize for not being able to cite all original research contributions due to space limitations. The authors thank Dr Jeremiah Bernier-Latmani and Dr Amélie Sabine for critical reading of the manuscript.

The research in T. V. Petrova’s laboratory is supported by Swiss National Science Foundation grants PPP0033-114898 and CRSII3-141811, Oncosuisse grant 2863-08-2011, the Leenaards Foundation, the Gebert Rüf Foundation, and the People Programme (Marie Curie Actions) of the European Union's Seventh Framework Programme FP7/2007-2013/ under REA grant agreement 317250.

Contribution: S.C., E.B., and T.V.P. conceived and wrote the manuscript and gave final approval for submission.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Tatiana V. Petrova, Department of Oncology, CHUV-UNIL, Ch. des Boveresses 155, CH-1066 Epalinges, Switzerland; e-mail: tatiana.petrova@unil.ch.

1
Schulte-Merker
 
S
Sabine
 
A
Petrova
 
TV
Lymphatic vascular morphogenesis in development, physiology, and disease.
J Cell Biol
2011
, vol. 
193
 
4
(pg. 
607
-
618
)
2
Baluk
 
P
Fuxe
 
J
Hashizume
 
H
, et al. 
Functionally specialized junctions between endothelial cells of lymphatic vessels.
J Exp Med
2007
, vol. 
204
 
10
(pg. 
2349
-
2362
)
3
Sabine
 
A
Petrova
 
T
Kiefer
 
F
Schulte-Merker
 
S
V. Interplay of mechanotransduction, FOXC2, connexins, and calcineurin signaling in lymphatic valve formation.
Developmental Aspects of the Lymphatic Vascular System
2014
, vol. 
vol. 214
 
Springer, Vienna, Austrial
(pg. 
67
-
80
)
4
Zawieja
 
DC
Contractile physiology of lymphatics.
Lymphat Res Biol
2009
, vol. 
7
 
2
(pg. 
87
-
96
)
5
Sabin
 
FR
On the origin of the lymphatic system from the veins, and the development of the lymph hearts and thoracic duct in the pig.
Am J Anat
1902
, vol. 
1
 
3
(pg. 
367
-
389
)
6
Huntington
 
GS
The genetic interpretation of the development of the lymphatic system.
Anat Rec
1908
, vol. 
2
 
292
(pg. 
19
-
46
)
7
Hägerling
 
R
Pollmann
 
C
Andreas
 
M
, et al. 
A novel multistep mechanism for initial lymphangiogenesis in mouse embryos based on ultramicroscopy.
EMBO J
2013
, vol. 
32
 
5
(pg. 
629
-
644
)
8
Yang
 
Y
García-Verdugo
 
JM
Soriano-Navarro
 
M
, et al. 
Lymphatic endothelial progenitors bud from the cardinal vein and intersomitic vessels in mammalian embryos.
Blood
2012
, vol. 
120
 
11
(pg. 
2340
-
2348
)
9
Wigle
 
JT
Oliver
 
G
Prox1 function is required for the development of the murine lymphatic system.
Cell
1999
, vol. 
98
 
6
(pg. 
769
-
778
)
10
François
 
M
Caprini
 
A
Hosking
 
B
, et al. 
Sox18 induces development of the lymphatic vasculature in mice.
Nature
2008
, vol. 
456
 
7222
(pg. 
643
-
647
)
11
van Impel
 
A
Zhao
 
Z
Hermkens
 
DMA
, et al. 
Divergence of zebrafish and mouse lymphatic cell fate specification pathways.
Development
2014
, vol. 
141
 
6
(pg. 
1228
-
1238
)
12
Srinivasan
 
RS
Oliver
 
G
Prox1 dosage controls the number of lymphatic endothelial cell progenitors and the formation of the lymphovenous valves.
Genes Dev
2011
, vol. 
25
 
20
(pg. 
2187
-
2197
)
13
Yao
 
L-C
Baluk
 
P
Srinivasan
 
RS
Oliver
 
G
McDonald
 
DM
Plasticity of button-like junctions in the endothelium of airway lymphatics in development and inflammation.
Am J Pathol
2012
, vol. 
180
 
6
(pg. 
2561
-
2575
)
14
Betterman
 
KL
Paquet-Fifield
 
S
Asselin-Labat
 
M-L
, et al. 
Remodeling of the lymphatic vasculature during mouse mammary gland morphogenesis is mediated via epithelial-derived lymphangiogenic stimuli.
Am J Pathol
2012
, vol. 
181
 
6
(pg. 
2225
-
2238
)
15
Rutkowski
 
JM
Ihm
 
JE
Lee
 
ST
, et al. 
VEGFR-3 neutralization inhibits ovarian lymphangiogenesis, follicle maturation, and murine pregnancy.
Am J Pathol
2013
, vol. 
183
 
5
(pg. 
1596
-
1607
)
16
Petrova
 
TV
Karpanen
 
T
Norrmén
 
C
, et al. 
Defective valves and abnormal mural cell recruitment underlie lymphatic vascular failure in lymphedema distichiasis.
Nat Med
2004
, vol. 
10
 
9
(pg. 
974
-
981
)
17
Aebischer
 
D
Iolyeva
 
M
Halin
 
C
The inflammatory response of lymphatic endothelium.
Angiogenesis
2013
 
18
Kim
 
H
Kataru
 
RP
Koh
 
GY
Regulation and implications of inflammatory lymphangiogenesis.
Trends Immunol
2012
, vol. 
33
 
7
(pg. 
350
-
356
)
19
Huggenberger
 
R
Ullmann
 
S
Proulx
 
ST
Pytowski
 
B
Alitalo
 
K
Detmar
 
M
Stimulation of lymphangiogenesis via VEGFR-3 inhibits chronic skin inflammation.
J Exp Med
2010
, vol. 
207
 
10
(pg. 
2255
-
2269
)
20
Harvey
 
NL
Gordon
 
EJ
Deciphering the roles of macrophages in developmental and inflammation stimulated lymphangiogenesis.
Vasc Cell
2012
, vol. 
4
 
1
pg. 
15
 
21
Tan
 
KW
Chong
 
SZ
Wong
 
FHS
, et al. 
Neutrophils contribute to inflammatory lymphangiogenesis by increasing VEGF-A bioavailability and secreting VEGF-D.
Blood
2013
, vol. 
122
 
22
(pg. 
3666
-
3677
)
22
Sleeman
 
JP
Thiele
 
W
Tumor metastasis and the lymphatic vasculature.
Int J Cancer
2009
, vol. 
125
 
12
(pg. 
2747
-
2756
)
23
Leong
 
SPL
Nakakura
 
EK
Pollock
 
R
, et al. 
Unique patterns of metastases in common and rare types of malignancy.
J Surg Oncol
2011
, vol. 
103
 
6
(pg. 
607
-
614
)
24
Mortimer
 
P
Arm lymphoedema after breast cancer.
Lancet Oncol
2013
, vol. 
14
 
6
(pg. 
442
-
443
)
25
Alitalo
 
A
Detmar
 
M
Interaction of tumor cells and lymphatic vessels in cancer progression.
Oncogene
2012
, vol. 
31
 
42
(pg. 
4499
-
4508
)
26
Jain
 
RK
Fenton
 
BT
Intratumoral lymphatic vessels: a case of mistaken identity or malfunction?
J Natl Cancer Inst
2002
, vol. 
94
 
6
(pg. 
417
-
421
)
27
Kerjaschki
 
D
Bago-Horvath
 
Z
Rudas
 
M
, et al. 
Lipoxygenase mediates invasion of intrametastatic lymphatic vessels and propagates lymph node metastasis of human mammary carcinoma xenografts in mouse.
J Clin Invest
2011
, vol. 
121
 
5
(pg. 
2000
-
2012
)
28
Kuroda
 
K
Horiguchi
 
A
Asano
 
T
Asano
 
T
Hayakawa
 
M
Prediction of lymphatic invasion by peritumoral lymphatic vessel density in prostate biopsy cores.
Prostate
2008
, vol. 
68
 
10
(pg. 
1057
-
1063
)
29
Karnezis
 
T
Shayan
 
R
Caesar
 
C
, et al. 
VEGF-D promotes tumor metastasis by regulating prostaglandins produced by the collecting lymphatic endothelium.
Cancer Cell
2012
, vol. 
21
 
2
(pg. 
181
-
195
)
30
He
 
Y
Rajantie
 
I
Ilmonen
 
M
, et al. 
Preexisting lymphatic endothelium but not endothelial progenitor cells are essential for tumor lymphangiogenesis and lymphatic metastasis.
Cancer Res
2004
, vol. 
64
 
11
(pg. 
3737
-
3740
)
31
Hirakawa
 
S
Brown
 
LF
Kodama
 
S
Paavonen
 
K
Alitalo
 
K
Detmar
 
M
VEGF-C-induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites.
Blood
2007
, vol. 
109
 
3
(pg. 
1010
-
1017
)
32
Feng
 
Y
Wang
 
W
Hu
 
J
Ma
 
J
Zhang
 
Y
Zhang
 
J
Expression of VEGF-C and VEGF-D as significant markers for assessment of lymphangiogenesis and lymph node metastasis in non-small cell lung cancer.
Anat Rec (Hoboken)
2010
, vol. 
293
 
5
(pg. 
802
-
812
)
33
Dadras
 
SS
Lange-Asschenfeldt
 
B
Velasco
 
P
, et al. 
Tumor lymphangiogenesis predicts melanoma metastasis to sentinel lymph nodes.
Mod Pathol
2005
, vol. 
18
 
9
(pg. 
1232
-
1242
)
34
Das
 
S
Sarrou
 
E
Podgrabinska
 
S
, et al. 
Tumor cell entry into the lymph node is controlled by CCL1 chemokine expressed by lymph node lymphatic sinuses.
J Exp Med
2013
, vol. 
210
 
8
(pg. 
1509
-
1528
)
35
Harvey
 
NL
Srinivasan
 
RS
Dillard
 
ME
, et al. 
Lymphatic vascular defects promoted by Prox1 haploinsufficiency cause adult-onset obesity.
Nat Genet
2005
, vol. 
37
 
10
(pg. 
1072
-
1081
)
36
Greene
 
AK
Grant
 
FD
Slavin
 
SA
Lower-extremity lymphedema and elevated body-mass index.
N Engl J Med
2012
, vol. 
366
 
22
(pg. 
2136
-
2137
)
37
Martel
 
C
Li
 
W
Fulp
 
B
, et al. 
Lymphatic vasculature mediates macrophage reverse cholesterol transport in mice.
J Clin Invest
2013
, vol. 
123
 
4
(pg. 
1571
-
1579
)
38
Lim
 
HY
Thiam
 
CH
Yeo
 
KP
, et al. 
Lymphatic vessels are essential for the removal of cholesterol from peripheral tissues by SR-BI-mediated transport of HDL.
Cell Metab
2013
, vol. 
17
 
5
(pg. 
671
-
684
)
39
Lim
 
HY
Rutkowski
 
JM
Helft
 
J
, et al. 
Hypercholesterolemic mice exhibit lymphatic vessel dysfunction and degeneration.
Am J Pathol
2009
, vol. 
175
 
3
(pg. 
1328
-
1337
)
40
Wiig
 
H
Schröder
 
A
Neuhofer
 
W
, et al. 
Immune cells control skin lymphatic electrolyte homeostasis and blood pressure.
J Clin Invest
2013
, vol. 
123
 
7
(pg. 
2803
-
2815
)
41
Machnik
 
A
Neuhofer
 
W
Jantsch
 
J
, et al. 
Macrophages regulate salt-dependent volume and blood pressure by a vascular endothelial growth factor-C-dependent buffering mechanism.
Nat Med
2009
, vol. 
15
 
5
(pg. 
545
-
552
)
42
Impel
 
A
Schulte-Merker
 
S
Kiefer
 
F
Schulte-Merker
 
S
A fisheye view on lymphangiogenesis.
Developmental Aspects of the Lymphatic Vascular System
2014
, vol. 
vol. 214
 
Springer, Vienna, Austria
(pg. 
153
-
165
)
43
Alitalo
 
K
The lymphatic vasculature in disease.
Nat Med
2011
, vol. 
17
 
11
(pg. 
1371
-
1380
)
44
Karkkainen
 
MJ
Haiko
 
P
Sainio
 
K
, et al. 
Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins.
Nat Immunol
2004
, vol. 
5
 
1
(pg. 
74
-
80
)
45
Küchler
 
AM
Gjini
 
E
Peterson-Maduro
 
J
Cancilla
 
B
Wolburg
 
H
Schulte-Merker
 
S
Development of the zebrafish lymphatic system requires VEGFC signaling.
Curr Biol
2006
, vol. 
16
 
12
(pg. 
1244
-
1248
)
46
Jeltsch
 
M
Leppänen
 
V-M
Saharinen
 
P
Alitalo
 
K
Receptor tyrosine kinase-mediated angiogenesis.
Cold Spring Harb Perspect Biol
2013
, vol. 
5
 
9
(pg. 
1
-
22
)
47
Joukov
 
V
Sorsa
 
T
Kumar
 
V
, et al. 
Proteolytic processing regulates receptor specificity and activity of VEGF-C.
EMBO J
1997
, vol. 
16
 
13
(pg. 
3898
-
3911
)
48
Dumont
 
DJ
Jussila
 
L
Taipale
 
J
, et al. 
Cardiovascular failure in mouse embryos deficient in VEGF receptor-3.
Science
1998
, vol. 
282
 
5390
(pg. 
946
-
949
)
49
Karkkainen
 
MJ
Ferrell
 
RE
Lawrence
 
EC
, et al. 
Missense mutations interfere with VEGFR-3 signalling in primary lymphoedema.
Nat Genet
2000
, vol. 
25
 
2
(pg. 
153
-
159
)
50
Karkkainen
 
MJ
Saaristo
 
A
Jussila
 
L
, et al. 
A model for gene therapy of human hereditary lymphedema.
Proc Natl Acad Sci USA
2001
, vol. 
98
 
22
(pg. 
12677
-
12682
)
51
Gordon
 
K
Schulte
 
D
Brice
 
G
, et al. 
Mutation in vascular endothelial growth factor-C, a ligand for vascular endothelial growth factor receptor-3, is associated with autosomal dominant milroy-like primary lymphedema.
Circ Res
2013
, vol. 
112
 
6
(pg. 
956
-
960
)
52
Villefranc
 
JA
Nicoli
 
S
Bentley
 
K
, et al. 
A truncation allele in vascular endothelial growth factor c reveals distinct modes of signaling during lymphatic and vascular development.
Development
2013
, vol. 
140
 
7
(pg. 
1497
-
1506
)
53
Planas-Paz
 
L
Strilić
 
B
Goedecke
 
A
Breier
 
G
Fässler
 
R
Lammert
 
E
Mechanoinduction of lymph vessel expansion.
EMBO J
2012
, vol. 
31
 
4
(pg. 
788
-
804
)
54
Yuan
 
L
Moyon
 
D
Pardanaud
 
L
, et al. 
Abnormal lymphatic vessel development in neuropilin 2 mutant mice.
Development
2002
, vol. 
129
 
20
(pg. 
4797
-
4806
)
55
Xu
 
Y
Yuan
 
L
Mak
 
J
, et al. 
Neuropilin-2 mediates VEGF-C-induced lymphatic sprouting together with VEGFR3.
J Cell Biol
2010
, vol. 
188
 
1
(pg. 
115
-
130
)
56
Lin
 
F-J
Chen
 
X
Qin
 
J
Hong
 
Y-K
Tsai
 
M-J
Tsai
 
SY
Direct transcriptional regulation of neuropilin-2 by COUP-TFII modulates multiple steps in murine lymphatic vessel development.
J Clin Invest
2010
, vol. 
120
 
5
(pg. 
1694
-
1707
)
57
Coma
 
S
Allard-Ratick
 
M
Akino
 
T
van Meeteren
 
LA
Mammoto
 
A
Klagsbrun
 
M
GATA2 and Lmo2 control angiogenesis and lymphangiogenesis via direct transcriptional regulation of neuropilin-2.
Angiogenesis
2013
, vol. 
16
 
4
(pg. 
939
-
952
)
58
Caunt
 
M
Mak
 
J
Liang
 
WC
, et al. 
Blocking neuropilin-2 function inhibits tumor cell metastasis.
Cancer Cell
2008
, vol. 
13
 
4
(pg. 
331
-
342
)
59
Murakami
 
M
Zheng
 
Y
Hirashima
 
M
, et al. 
VEGFR1 tyrosine kinase signaling promotes lymphangiogenesis as well as angiogenesis indirectly via macrophage recruitment.
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 
4
(pg. 
658
-
664
)
60
Dellinger
 
MT
Meadows
 
SM
Wynne
 
K
Cleaver
 
O
Brekken
 
RA
Vascular endothelial growth factor receptor-2 promotes the development of the lymphatic vasculature.
PLoS ONE
2013
, vol. 
8
 
9
pg. 
e74686
 
61
Wirzenius
 
M
Tammela
 
T
Uutela
 
M
, et al. 
Distinct vascular endothelial growth factor signals for lymphatic vessel enlargement and sprouting.
J Exp Med
2007
, vol. 
204
 
6
(pg. 
1431
-
1440
)
62
Nilsson
 
I
Bahram
 
F
Li
 
X
, et al. 
VEGF receptor 2/-3 heterodimers detected in situ by proximity ligation on angiogenic sprouts.
EMBO J
2010
, vol. 
29
 
8
(pg. 
1377
-
1388
)
63
Alders
 
M
Hogan
 
BM
Gjini
 
E
, et al. 
Mutations in CCBE1 cause generalized lymph vessel dysplasia in humans.
Nat Genet
2009
, vol. 
41
 
12
(pg. 
1272
-
1274
)
64
Van Balkom
 
IDC
Alders
 
M
Allanson
 
J
, et al. 
Lymphedema-lymphangiectasia-mental retardation (Hennekam) syndrome: a review.
Am J Med Genet
2002
, vol. 
112
 
4
(pg. 
412
-
421
)
65
Hogan
 
BM
Bos
 
FL
Bussmann
 
J
, et al. 
Ccbe1 is required for embryonic lymphangiogenesis and venous sprouting.
Nat Genet
2009
, vol. 
41
 
4
(pg. 
396
-
398
)
66
Bos
 
FL
Caunt
 
M
Peterson-Maduro
 
J
, et al. 
CCBE1 is essential for mammalian lymphatic vascular development and enhances the lymphangiogenic effect of vascular endothelial growth factor-C in vivo.
Circ Res
2011
, vol. 
109
 
5
(pg. 
486
-
491
)
67
Le Guen
 
L
Karpanen
 
T
Schulte
 
D
, et al. 
Ccbe1 regulates Vegfc-mediated induction of Vegfr3 signaling during embryonic lymphangiogenesis.
Development
2014
, vol. 
141
 
6
(pg. 
1239
-
1249
)
68
Jeltsch
 
M
Jha
 
SK
Tvorogov
 
D
, et al. 
 
CCBE1 enhances lymphangiogenesis via ADAMTS3-mediated VEGF-C activation. Circulation. Epub: February 19, 2014; doi:10.1161/CIRCULATIONAHA.113.002779
69
Zou
 
Z
Enis
 
DR
Bui
 
H
, et al. 
The secreted lymphangiogenic factor CCBE1 is essential for fetal liver erythropoiesis.
Blood
2013
, vol. 
121
 
16
(pg. 
3228
-
3236
)
70
Weijts
 
BGMW
van Impel
 
A
Schulte-Merker
 
S
de Bruin
 
A
Atypical E2fs control lymphangiogenesis through transcriptional regulation of Ccbe1 and Flt4.
PLoS ONE
2013
, vol. 
8
 
9
pg. 
e73693
 
71
Eilken
 
HM
Adams
 
RH
Dynamics of endothelial cell behavior in sprouting angiogenesis.
Curr Opin Cell Biol
2010
, vol. 
22
 
5
(pg. 
617
-
625
)
72
Mäkinen
 
T
Adams
 
RH
Bailey
 
J
, et al. 
PDZ interaction site in ephrinB2 is required for the remodeling of lymphatic vasculature.
Genes Dev
2005
, vol. 
19
 
3
(pg. 
397
-
410
)
73
Katsuta
 
H
Fukushima
 
Y
Maruyama
 
K
, et al. 
EphrinB2-EphB4 signals regulate formation and maintenance of funnel-shaped valves in corneal lymphatic capillaries.
Invest Ophthalmol Vis Sci
2013
, vol. 
54
 
6
(pg. 
4102
-
4108
)
74
Wang
 
Y
Nakayama
 
M
Pitulescu
 
ME
, et al. 
Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis.
Nature
2010
, vol. 
465
 
7297
(pg. 
483
-
486
)
75
Sawamiphak
 
S
Seidel
 
S
Essmann
 
CL
, et al. 
Ephrin-B2 regulates VEGFR2 function in developmental and tumour angiogenesis.
Nature
2010
, vol. 
465
 
7297
(pg. 
487
-
491
)
76
Nakayama
 
A
Nakayama
 
M
Turner
 
CJ
Höing
 
S
Lepore
 
JJ
Adams
 
RH
Ephrin-B2 controls PDGFRβ internalization and signaling.
Genes Dev
2013
, vol. 
27
 
23
(pg. 
2576
-
2589
)
77
Abéngozar
 
MA
de Frutos
 
S
Ferreiro
 
S
, et al. 
Blocking ephrinB2 with highly specific antibodies inhibits angiogenesis, lymphangiogenesis, and tumor growth.
Blood
2012
, vol. 
119
 
19
(pg. 
4565
-
4576
)
78
Eklund
 
L
Saharinen
 
P
Angiopoietin signaling in the vasculature.
Exp Cell Res
2013
, vol. 
319
 
9
(pg. 
1271
-
1280
)
79
Jeansson
 
M
Gawlik
 
A
Anderson
 
G
, et al. 
Angiopoietin-1 is essential in mouse vasculature during development and in response to injury.
J Clin Invest
2011
, vol. 
121
 
6
(pg. 
2278
-
2289
)
80
Qu
 
X
Tompkins
 
K
Batts
 
LE
Puri
 
M
Baldwin
 
S
Abnormal embryonic lymphatic vessel development in Tie1 hypomorphic mice.
Development
2010
, vol. 
137
 
8
(pg. 
1285
-
1295
)
81
D’Amico
 
G
Korhonen
 
EA
Waltari
 
M
Saharinen
 
P
Laakkonen
 
P
Alitalo
 
K
Loss of endothelial Tie1 receptor impairs lymphatic vessel development-brief report.
Arterioscler Thromb Vasc Biol
2010
, vol. 
30
 
2
(pg. 
207
-
209
)
82
Gale
 
NW
Thurston
 
G
Hackett
 
SF
, et al. 
Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by Angiopoietin-1.
Dev Cell
2002
, vol. 
3
 
3
(pg. 
411
-
423
)
83
Dellinger
 
M
Hunter
 
R
Bernas
 
M
, et al. 
Defective remodeling and maturation of the lymphatic vasculature in Angiopoietin-2 deficient mice.
Dev Biol
2008
, vol. 
319
 
2
(pg. 
309
-
320
)
84
Fagiani
 
E
Lorentz
 
P
Kopfstein
 
L
Christofori
 
G
Angiopoietin-1 and −2 exert antagonistic functions in tumor angiogenesis, yet both induce lymphangiogenesis.
Cancer Res
2011
, vol. 
71
 
17
(pg. 
5717
-
5727
)
85
Tammela
 
T
Saaristo
 
A
Lohela
 
M
, et al. 
Angiopoietin-1 promotes lymphatic sprouting and hyperplasia.
Blood
2005
, vol. 
105
 
12
(pg. 
4642
-
4648
)
86
Koh
 
GY
Orchestral actions of angiopoietin-1 in vascular regeneration.
Trends Mol Med
2013
, vol. 
19
 
1
(pg. 
31
-
39
)
87
Gerald
 
D
Chintharlapalli
 
S
Augustin
 
HG
Benjamin
 
LE
Angiopoietin-2: an attractive target for improved antiangiogenic tumor therapy.
Cancer Res
2013
, vol. 
73
 
6
(pg. 
1649
-
1657
)
88
Roca
 
C
Adams
 
RH
Regulation of vascular morphogenesis by Notch signaling.
Genes Dev
2007
, vol. 
21
 
20
(pg. 
2511
-
2524
)
89
Phng
 
LK
Gerhardt
 
H
Angiogenesis: a team effort coordinated by notch.
Dev Cell
2009
, vol. 
16
 
2
(pg. 
196
-
208
)
90
Shawber
 
CJ
Funahashi
 
Y
Francisco
 
E
, et al. 
Notch alters VEGF responsiveness in human and murine endothelial cells by direct regulation of VEGFR-3 expression.
J Clin Invest
2007
, vol. 
117
 
11
(pg. 
3369
-
3382
)
91
Niessen
 
K
Zhang
 
G
Ridgway
 
JB
, et al. 
The Notch1-Dll4 signaling pathway regulates mouse postnatal lymphatic development.
Blood
2011
, vol. 
118
 
7
(pg. 
1989
-
1997
)
92
Zheng
 
W
Tammela
 
T
Yamamoto
 
M
, et al. 
Notch restricts lymphatic vessel sprouting induced by vascular endothelial growth factor.
Blood
2011
, vol. 
118
 
4
(pg. 
1154
-
1162
)
93
Geudens
 
I
Herpers
 
R
Hermans
 
K
, et al. 
Role of delta-like-4/Notch in the formation and wiring of the lymphatic network in zebrafish.
Arterioscler Thromb Vasc Biol
2010
, vol. 
30
 
9
(pg. 
1695
-
1702
)
94
Kang
 
J
Yoo
 
J
Lee
 
S
, et al. 
An exquisite cross-control mechanism among endothelial cell fate regulators directs the plasticity and heterogeneity of lymphatic endothelial cells.
Blood
2010
, vol. 
116
 
1
(pg. 
140
-
150
)
95
Murtomaki
 
A
Uh
 
MK
Choi
 
YK
, et al. 
Notch1 functions as a negative regulator of lymphatic endothelial cell differentiation in the venous endothelium.
Development
2013
, vol. 
140
 
11
(pg. 
2365
-
2376
)
96
Jakobsson
 
L
van Meeteren
 
LA
Transforming growth factor β family members in regulation of vascular function: in the light of vascular conditional knockouts.
Exp Cell Res
2013
, vol. 
319
 
9
(pg. 
1264
-
1270
)
97
ten Dijke
 
P
Arthur
 
HM
Extracellular control of TGFbeta signalling in vascular development and disease.
Nat Rev Mol Cell Biol
2007
, vol. 
8
 
11
(pg. 
857
-
869
)
98
Park
 
SO
Lee
 
YJ
Seki
 
T
, et al. 
ALK5- and TGFBR2-independent role of ALK1 in the pathogenesis of hereditary hemorrhagic telangiectasia type 2.
Blood
2008
, vol. 
111
 
2
(pg. 
633
-
642
)
99
Moya
 
IM
Umans
 
L
Maas
 
E
, et al. 
Stalk cell phenotype depends on integration of Notch and Smad1/5 signaling cascades.
Dev Cell
2012
, vol. 
22
 
3
(pg. 
501
-
514
)
100
Larrivée
 
B
Prahst
 
C
Gordon
 
E
, et al. 
ALK1 signaling inhibits angiogenesis by cooperating with the Notch pathway.
Dev Cell
2012
, vol. 
22
 
3
(pg. 
489
-
500
)
101
Nguyen
 
HLLY
Lee
 
YJ
Shin
 
J
, et al. 
TGF-β signaling in endothelial cells, but not neuroepithelial cells, is essential for cerebral vascular development.
Lab Invest
2011
, vol. 
91
 
11
(pg. 
1554
-
1563
)
102
David
 
L
Mallet
 
C
Keramidas
 
M
, et al. 
Bone morphogenetic protein-9 is a circulating vascular quiescence factor.
Circ Res
2008
, vol. 
102
 
8
(pg. 
914
-
922
)
103
Carvalho
 
RL
Itoh
 
F
Goumans
 
MJ
, et al. 
Compensatory signalling induced in the yolk sac vasculature by deletion of TGFbeta receptors in mice.
J Cell Sci
2007
, vol. 
120
 
Pt 24
(pg. 
4269
-
4277
)
104
Armulik
 
A
Genové
 
G
Betsholtz
 
C
Pericytes: developmental, physiological, and pathological perspectives, problems, and promises.
Dev Cell
2011
, vol. 
21
 
2
(pg. 
193
-
215
)
105
Langlois
 
D
Hneino
 
M
Bouazza
 
L
, et al. 
Conditional inactivation of TGF-β type II receptor in smooth muscle cells and epicardium causes lethal aortic and cardiac defects.
Transgenic Res
2010
, vol. 
19
 
6
(pg. 
1069
-
1082
)
106
Niessen
 
K
Zhang
 
G
Ridgway
 
JB
Chen
 
H
Yan
 
M
ALK1 signaling regulates early postnatal lymphatic vessel development.
Blood
2010
, vol. 
115
 
8
(pg. 
1654
-
1661
)
107
Oka
 
M
Iwata
 
C
Suzuki
 
HI
, et al. 
Inhibition of endogenous TGF-β signaling enhances lymphangiogenesis.
Blood
2008
, vol. 
111
 
9
(pg. 
4571
-
4579
)
108
James
 
JM
Nalbandian
 
A
Mukouyama
 
YS
TGFβ signaling is required for sprouting lymphangiogenesis during lymphatic network development in the skin.
Development
2013
, vol. 
140
 
18
(pg. 
3903
-
3914
)
109
Levet
 
S
Ciais
 
D
Merdzhanova
 
G
, et al. 
Bone morphogenetic protein 9 (BMP9) controls lymphatic vessel maturation and valve formation.
Blood
2013
, vol. 
122
 
4
(pg. 
598
-
607
)
110
Yoshimatsu
 
Y
Lee
 
YG
Akatsu
 
Y
, et al. 
Bone morphogenetic protein-9 inhibits lymphatic vessel formation via activin receptor-like kinase 1 during development and cancer progression.
Proc Natl Acad Sci USA
2013
, vol. 
110
 
47
(pg. 
18940
-
18945
)
111
Dunworth
 
WP
Cardona-Costa
 
J
Bozkulak
 
EC
, et al. 
Bone morphogenetic protein 2 signaling negatively modulates lymphatic development in vertebrate embryos.
Circ Res
2014
, vol. 
114
 
1
(pg. 
56
-
66
)
112
Avraham
 
T
Daluvoy
 
S
Zampell
 
J
, et al. 
Blockade of transforming growth factor-β1 accelerates lymphatic regeneration during wound repair.
Am J Pathol
2010
, vol. 
177
 
6
(pg. 
3202
-
3214
)
113
Vittet
 
D
Merdzhanova
 
G
Prandini
 
M-H
Feige
 
J-J
Bailly
 
S
TGFβ1 inhibits lymphatic endothelial cell differentiation from mouse embryonic stem cells.
J Cell Physiol
2012
, vol. 
227
 
11
(pg. 
3593
-
3602
)
114
Tammela
 
T
Zarkada
 
G
Nurmi
 
H
, et al. 
VEGFR-3 controls tip to stalk conversion at vessel fusion sites by reinforcing Notch signalling.
Nat Cell Biol
2011
, vol. 
13
 
10
(pg. 
1202
-
1213
)
115
Zhang
 
L
Zhou
 
F
Han
 
W
, et al. 
VEGFR-3 ligand-binding and kinase activity are required for lymphangiogenesis but not for angiogenesis.
Cell Res
2010
, vol. 
20
 
12
(pg. 
1319
-
1331
)
116
Shalaby
 
F
Rossant
 
J
Yamaguchi
 
TP
, et al. 
Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice.
Nature
1995
, vol. 
376
 
6535
(pg. 
62
-
66
)
117
Ricard
 
N
Ciais
 
D
Levet
 
S
, et al. 
BMP9 and BMP10 are critical for postnatal retinal vascular remodeling.
Blood
2012
, vol. 
119
 
25
(pg. 
6162
-
6171
)
118
Sridurongrit
 
S
Larsson
 
J
Schwartz
 
R
Ruiz-Lozano
 
P
Kaartinen
 
V
Signaling via the Tgf-β type I receptor Alk5 in heart development.
Dev Biol
2008
, vol. 
322
 
1
(pg. 
208
-
218
)
119
Jiao
 
K
Langworthy
 
M
Batts
 
L
Brown
 
CB
Moses
 
HL
Baldwin
 
HS
Tgfbeta signaling is required for atrioventricular cushion mesenchyme remodeling during in vivo cardiac development.
Development
2006
, vol. 
133
 
22
(pg. 
4585
-
4593
)
120
Hellström
 
M
Phng
 
L-K
Hofmann
 
JJ
, et al. 
Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis.
Nature
2007
, vol. 
445
 
7129
(pg. 
776
-
780
)
121
Sato
 
TN
Tozawa
 
Y
Deutsch
 
U
, et al. 
Distinct roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation.
Nature
1995
, vol. 
376
 
6535
(pg. 
70
-
74
)
122
Ichise
 
T
Yoshida
 
N
Ichise
 
HH
H-, N- and Kras cooperatively regulate lymphatic vessel growth by modulating VEGFR3 expression in lymphatic endothelial cells in mice.
Development
2010
, vol. 
137
 
6
(pg. 
1003
-
1013
)
123
Henkemeyer
 
M
Rossi
 
DJ
Holmyard
 
DP
, et al. 
Vascular system defects and neuronal apoptosis in mice lacking ras GTPase-activating protein.
Nature
1995
, vol. 
377
 
6551
(pg. 
695
-
701
)
124
Lapinski
 
PE
Kwon
 
S
Lubeck
 
BA
, et al. 
RASA1 maintains the lymphatic vasculature in a quiescent functional state in mice.
J Clin Invest
2012
, vol. 
122
 
2
(pg. 
733
-
747
)
125
Mouta-Bellum
 
C
Kirov
 
A
Miceli-Libby
 
L
, et al. 
Organ-specific lymphangiectasia, arrested lymphatic sprouting, and maturation defects resulting from gene-targeting of the PI3K regulatory isoforms p85α, p55α, and p50α.
Dev Dyn
2009
, vol. 
238
 
10
(pg. 
2670
-
2679
)
126
Zhou
 
F
Chang
 
Z
Zhang
 
L
, et al. 
Akt/Protein kinase B is required for lymphatic network formation, remodeling, and valve development.
Am J Pathol
2010
, vol. 
177
 
4
(pg. 
2124
-
2133
)
127
Zeini
 
M
Hang
 
CT
Lehrer-Graiwer
 
J
Dao
 
T
Zhou
 
B
Chang
 
C-P
Spatial and temporal regulation of coronary vessel formation by calcineurin-NFAT signaling.
Development
2009
, vol. 
136
 
19
(pg. 
3335
-
3345
)
128
Sabine
 
A
Agalarov
 
Y
Maby-El Hajjami
 
H
, et al. 
Mechanotransduction, PROX1, and FOXC2 cooperate to control connexin37 and calcineurin during lymphatic-valve formation.
Dev Cell
2012
, vol. 
22
 
2
(pg. 
430
-
445
)
129
Castellano
 
E
Downward
 
J
Rommel
 
C
Vanhaesebroeck
 
B
Vogt
 
PK
Role of RAS in the regulation of PI 3-kinase.
Phosphoinositide 3-Kinase in Health and Disease
2011
, vol. 
vol. 346
 
Springer, Berlin, Germany
(pg. 
143
-
169
)
130
Pearson
 
G
Robinson
 
F
Beers Gibson
 
T
, et al. 
Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions.
Endocr Rev
2001
, vol. 
22
 
2
(pg. 
153
-
183
)
131
Schubbert
 
S
Zenker
 
M
Rowe
 
SL
, et al. 
Germline KRAS mutations cause Noonan syndrome.
Nat Genet
2006
, vol. 
38
 
3
(pg. 
331
-
336
)
132
Lo
 
IFM
Brewer
 
C
Shannon
 
N
, et al. 
Severe neonatal manifestations of Costello syndrome.
J Med Genet
2008
, vol. 
45
 
3
(pg. 
167
-
171
)
133
Kerr
 
B
Delrue
 
MA
Sigaudy
 
S
, et al. 
Genotype-phenotype correlation in Costello syndrome: HRAS mutation analysis in 43 cases.
J Med Genet
2006
, vol. 
43
 
5
(pg. 
401
-
405
)
134
Boon
 
LM
Mulliken
 
JB
Vikkula
 
M
RASA1: variable phenotype with capillary and arteriovenous malformations.
Curr Opin Genet Dev
2005
, vol. 
15
 
3
(pg. 
265
-
269
)
135
de Wijn
 
RS
Oduber
 
CE
Breugem
 
CC
Alders
 
M
Hennekam
 
RC
van der Horst
 
CM
Phenotypic variability in a family with capillary malformations caused by a mutation in the RASA1 gene.
Eur J Med Genet
2012
, vol. 
55
 
3
(pg. 
191
-
195
)
136
Burrows
 
PE
Gonzalez-Garay
 
ML
Rasmussen
 
JC
, et al. 
Lymphatic abnormalities are associated with RASA1 gene mutations in mouse and man.
Proc Natl Acad Sci USA
2013
, vol. 
110
 
21
(pg. 
8621
-
8626
)
137
Xu
 
K
Sacharidou
 
A
Fu
 
S
, et al. 
Blood vessel tubulogenesis requires Rasip1 regulation of GTPase signaling.
Dev Cell
2011
, vol. 
20
 
4
(pg. 
526
-
539
)
138
Ren
 
B
Deng
 
Y
Mukhopadhyay
 
A
, et al. 
ERK1/2-Akt1 crosstalk regulates arteriogenesis in mice and zebrafish.
J Clin Invest
2010
, vol. 
120
 
4
(pg. 
1217
-
1228
)
139
Hong
 
CC
Peterson
 
QP
Hong
 
J-Y
Peterson
 
RT
Artery/vein specification is governed by opposing phosphatidylinositol-3 kinase and MAP kinase/ERK signaling.
Curr Biol
2006
, vol. 
16
 
13
(pg. 
1366
-
1372
)
140
Deng
 
Y
Atri
 
D
Eichmann
 
A
Simons
 
M
Endothelial ERK signaling controls lymphatic fate specification.
J Clin Invest
2013
, vol. 
123
 
3
(pg. 
1202
-
1215
)
141
Vanhaesebroeck
 
B
Stephens
 
L
Hawkins
 
P
PI3K signalling: the path to discovery and understanding.
Nat Rev Mol Cell Biol
2012
, vol. 
13
 
3
(pg. 
195
-
203
)
142
Engelman
 
JA
Targeting PI3K signalling in cancer: opportunities, challenges and limitations.
Nat Rev Cancer
2009
, vol. 
9
 
8
(pg. 
550
-
562
)
143
Castellano
 
E
Downward
 
J
RAS interaction with PI3K.
Genes Cancer
2011
, vol. 
2
 
3
(pg. 
261
-
274
)
144
Graupera
 
M
Potente
 
M
Regulation of angiogenesis by PI3K signaling networks.
Exp Cell Res
2013
, vol. 
319
 
9
(pg. 
1348
-
1355
)
145
Mäkinen
 
T
Veikkola
 
T
Mustjoki
 
S
, et al. 
Isolated lymphatic endothelial cells transduce growth, survival and migratory signals via the VEGF-C/D receptor VEGFR-3.
EMBO J
2001
, vol. 
20
 
17
(pg. 
4762
-
4773
)
146
Coso
 
S
Zeng
 
Y
Opeskin
 
K
Williams
 
ED
Vascular endothelial growth factor receptor-3 directly interacts with phosphatidylinositol 3-kinase to regulate lymphangiogenesis.
PLoS ONE
2012
, vol. 
7
 
6
pg. 
e39558
 
147
Garmy-Susini
 
B
Avraamides
 
CJ
Desgrosellier
 
JS
, et al. 
PI3Kα activates integrin α4β1 to establish a metastatic niche in lymph nodes.
Proc Natl Acad Sci USA
2013
, vol. 
110
 
22
(pg. 
9042
-
9047
)
148
Fruman
 
DA
Snapper
 
SB
Yballe
 
CM
, et al. 
Impaired B cell development and proliferation in absence of phosphoinositide 3-kinase p85α.
Science
1999
, vol. 
283
 
5400
(pg. 
393
-
397
)
149
Gupta
 
S
Ramjaun
 
AR
Haiko
 
P
, et al. 
Binding of ras to phosphoinositide 3-kinase p110α is required for ras-driven tumorigenesis in mice.
Cell
2007
, vol. 
129
 
5
(pg. 
957
-
968
)
150
Kurek
 
KC
Luks
 
VL
Ayturk
 
UM
, et al. 
Somatic mosaic activating mutations in PIK3CA cause CLOVES syndrome.
Am J Hum Genet
2012
, vol. 
90
 
6
(pg. 
1108
-
1115
)
151
Hoey
 
SEH
Eastwood
 
D
Monsell
 
F
Kangesu
 
L
Harper
 
JI
Sebire
 
NJ
Histopathological features of Proteus syndrome.
Clin Exp Dermatol
2008
, vol. 
33
 
3
(pg. 
234
-
238
)
152
Johnson
 
EN
Lee
 
YM
Sander
 
TL
, et al. 
NFATc1 mediates vascular endothelial growth factor-induced proliferation of human pulmonary valve endothelial cells.
J Biol Chem
2003
, vol. 
278
 
3
(pg. 
1686
-
1692
)
153
Zaichuk
 
TA
Shroff
 
EH
Emmanuel
 
R
Filleur
 
S
Nelius
 
T
Volpert
 
OV
Nuclear factor of activated T cells balances angiogenesis activation and inhibition.
J Exp Med
2004
, vol. 
199
 
11
(pg. 
1513
-
1522
)
154
Schweighofer
 
B
Testori
 
J
Sturtzel
 
C
, et al. 
The VEGF-induced transcriptional response comprises gene clusters at the crossroad of angiogenesis and inflammation.
Thromb Haemost
2009
, vol. 
102
 
3
(pg. 
544
-
554
)
155
Minami
 
T
Jiang
 
S
Schadler
 
K
, et al. 
The calcineurin-NFAT-angiopoietin-2 signaling axis in lung endothelium is critical for the establishment of lung metastases.
Cell Rep
2013
, vol. 
4
 
4
(pg. 
709
-
723
)
156
Norrmén
 
C
Ivanov
 
KI
Cheng
 
J
, et al. 
FOXC2 controls formation and maturation of lymphatic collecting vessels through cooperation with NFATc1.
J Cell Biol
2009
, vol. 
185
 
3
(pg. 
439
-
457
)
157
Kulkarni
 
RM
Greenberg
 
JM
Akeson
 
AL
NFATc1 regulates lymphatic endothelial development.
Mech Dev
2009
, vol. 
126
 
5-6
(pg. 
350
-
365
)
158
Fish
 
JE
Cybulsky
 
MI
Taming endothelial activation with a microRNA.
J Clin Invest
2012
, vol. 
122
 
6
(pg. 
1967
-
1970
)
159
Hartmann
 
D
Thum
 
T
MicroRNAs and vascular (dys)function.
Vascul Pharmacol
2011
, vol. 
55
 
4
(pg. 
92
-
105
)
160
Boon
 
RA
MicroRNAs control vascular endothelial growth factor signaling.
Circ Res
2012
, vol. 
111
 
11
(pg. 
1388
-
1390
)
161
Kazenwadel
 
J
Michael
 
MZ
Harvey
 
NL
Prox1 expression is negatively regulated by miR-181 in endothelial cells.
Blood
2010
, vol. 
116
 
13
(pg. 
2395
-
2401
)
162
Pedrioli
 
DM
Karpanen
 
T
Dabouras
 
V
, et al. 
miR-31 functions as a negative regulator of lymphatic vascular lineage-specific differentiation in vitro and vascular development in vivo.
Mol Cell Biol
2010
, vol. 
30
 
14
(pg. 
3620
-
3634
)
Sign in via your Institution