Next Article in Journal
Uncovering the Localization and Function of a Novel Read-Through Transcript ‘TOMM40-APOE
Previous Article in Journal
CREB1 Facilitates GABAergic Neural Differentiation of Human Mesenchymal Stem Cells through BRN2 for Pain Alleviation and Locomotion Recovery after Spinal Cord Injury
Previous Article in Special Issue
A Cysteine Residue within the Kinase Domain of Zap70 Regulates Lck Activity and Proximal TCR Signaling
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dysregulation of Lymphatic Endothelial VEGFR3 Signaling in Disease

by
Kevin Kuonqui
,
Adana-Christine Campbell
,
Ananta Sarker
,
Arielle Roberts
,
Bracha L. Pollack
,
Hyeung Ju Park
,
Jinyeon Shin
,
Stav Brown
,
Babak J. Mehrara
* and
Raghu P. Kataru
*
Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
*
Authors to whom correspondence should be addressed.
Submission received: 15 November 2023 / Revised: 20 December 2023 / Accepted: 26 December 2023 / Published: 28 December 2023
(This article belongs to the Special Issue New Insights into Tyrosine Kinase Alterations in Human Diseases)

Abstract

:
Vascular endothelial growth factor (VEGF) receptor 3 (VEGFR3), a receptor tyrosine kinase encoded by the FLT4 gene, plays a significant role in the morphogenesis and maintenance of lymphatic vessels. Under both normal and pathologic conditions, VEGF-C and VEGF-D bind VEGFR3 on the surface of lymphatic endothelial cells (LECs) and induce lymphatic proliferation, migration, and survival by activating intracellular PI3K-Akt and MAPK-ERK signaling pathways. Impaired lymphatic function and VEGFR3 signaling has been linked with a myriad of commonly encountered clinical conditions. This review provides a brief overview of intracellular VEGFR3 signaling in LECs and explores examples of dysregulated VEGFR3 signaling in various disease states, including (1) lymphedema, (2) tumor growth and metastasis, (3) obesity and metabolic syndrome, (4) organ transplant rejection, and (5) autoimmune disorders. A more complete understanding of the molecular mechanisms underlying the lymphatic pathology of each disease will allow for the development of novel strategies to treat these chronic and often debilitating illnesses.

1. Introduction

VEGFR3 Signaling in Lymphatic Endothelial Cells

The lymphatic vasculature regulates immune cell trafficking and interstitial fluid homeostasis [1]. During fetal development, the proper formation of lymphatic networks is critical for the morphogenesis of nearly all organs [2]. Postnatally, stable lymphatic function is required to carry out physiologic activities of most organs, including the central nervous, cardiovascular, gastrointestinal, and integumentary systems [3].
Vascular endothelial growth factor (VEGF) receptor 3 (VEGFR3) is a receptor tyrosine kinase that plays a critical role in regulating growth of new lymphatic vessels (lymphangiogenesis) and new blood vessels (angiogenesis)—two processes that are essential for the development and maintenance of the vascular system. Binding of VEGF-C ligand to its cognate receptor VEGFR3 in lymphatic endothelial cells (LECs) triggers receptor homodimerization (VEGFR3-VEGFR3) or heterodimerization (VEGFR2-VEGFR3) and subsequently autophosphorylation of its cytoplasmic tyrosine kinase domains, which activate cytoplasmic secondary messengers [4,5,6]. Key VEGFR3-mediated downstream intracellular signaling interactions relevant to this review are illustrated in Figure 1. Intracellular adapter proteins including Src homology and collagen domain (Shc), growth factor receptor-bound protein 2 (Grb2), and Sh2 domain-containing protein tyrosine phosphatases play important roles in regulating downstream signaling cascade activation [7,8].
The Ras/mitogen-activated kinase (MAPK) and phosphoinositide-3-kinase (PI3K) signaling cascades serve as principal downstream VEGFR3 signaling effectors [9,10,11,12,13]. PI3K promotes downstream phosphorylation of protein kinase B (Akt), which in turn activates endothelial nitric oxide synthase (eNOS) and mammalian target of rapamycin (mTOR) [13,14,15]. mTOR subsequently mediates the phosphorylation of ribosomal protein S6 kinase beta 1 (p70S6K) and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) [16,17]. Phosphoinositide-dependent kinase 1 (PDK1) facilitates Akt phosphorylation, while phosphatase and tensin homology (PTEN) negatively regulate Akt activity [18,19]. On the other hand, protein kinase C (PKC)-dependent activation of the Raf1-MEK1/2 cascade results in downstream p42/p44 MAPK (ERK1/2) phosphorylation, which mediates the activation of the cAMP-response-element-binding protein (CREB) and Ets-domain 1 and 2 transcription factor [10,20,21,22]. Ras-GTPases serve as negative regulators of VEGF-C-mediated activation of downstream ERK signaling [23]. Classically, PI3K-Akt signaling is thought to promote the expression of pro-survival and anti-apoptotic genes, while MAPK/ERK signaling is mainly believed to mediate cellular proliferation [24,25]. Together, these pathways regulate LEC functions including proliferation, migration, and tubule formation [9,26].
Important molecular regulators of VEGFR3 activation include membrane coreceptors neuropilin 2 (Nrp2), which can bind either VEGF-C or class III semaphorin ligands to enhance or diminish VEGFR3 activation, respectively [27,28,29]. The EphB4 receptor facilitates VEGF-C/VEGFR3 complex internalization when stimulated by its cognate ligand EphrinB2 [30,31]. In the absence of VEGF-C, membrane integrins can transactivate VEGFR3 via recruitment of non-receptor-associated Src tyrosine kinases [32,33]. Other important modulators of VEGFR3 signal transduction include environmental mechanical forces and extracellular matrix (ECM) molecules, including fibronectin (FN), heparan sulfate (HS), and various other proteoglycans or glycoproteins [34,35,36].
In mature, healthy adults, the majority of lymphatic vessel networks throughout the body exist in a growth-quiescent state, with the exception of meningeal and intestinal lymphatics, which require continuous VEGF-C stimulation [37,38,39]. VEGFR3-mediated lymphatic remodeling can be activated as part of the inflammatory response, wound healing, obesity, tumor growth, and other physiologic or pathologic conditions [37,38,39]. Dysregulated LEC expression of VEGFR3 or its downstream mediators serves as a potential mechanism by which excessive or, more commonly, insufficient lymphangiogenesis contribute to pathological changes.
Over the past two decades, several studies have suggested that lymphatic vessel growth dysfunction contributes to numerous clinically relevant pathologies such as lymphedema; cancer; and autoimmune, metabolic, and inflammatory disorders. Therefore, understanding VEGFR3-related signaling pathways in LECs is important for developing therapeutic strategies to treat cancer, lymphedema, and other inflammatory and metabolic diseases. Targeted interventions aimed at modulating insufficient or excessive VEGFR3 signaling are actively being studied to address many of these lymphatic-associated diseases. While research in this field continues to expand, there currently is a paucity of literature outlining key pathologic findings related to dysregulated VEGFR3 signaling in lymphatic-related disorders.

2. Lymphedema and Other Lymphatic Anomalies

2.1. Primary Lymphedema and Other Primary Lymphatic Disorders

The development of the lymphatic vasculature is a well-defined process during which LEC progenitors initially bud from embryonic veins, eventually forming an early lymph sac from which all lymphatic vessels originate [40,41]. The VEGF-C/VEGFR3 signaling axis has been shown to play a critical role in the budding of early LECs from the cardinal veins [42,43,44,45]. Germline and somatic mutations have been linked to dysregulated VEGFR3 signaling in LECs, potentially leading to developmental lymphatic defects likely underlying a wide spectrum of primary lymphatic disorders (Figure 2a) [41,46]. For example, patients with Milroy’s disease, one of the most extensively studied hereditary congenital lymphedema disorders, have inactivating autosomal dominant mutations within the Fms-related tyrosine kinase 4 (FLT4) gene encoding VEGFR3 expression [47,48,49,50]. Some patients with Milroy-like hereditary primary lymphedema have inactivating VEGFC gene mutations resulting in variable degrees of lymphatic hypoplasia [51,52]. Additionally, collagen and calcium-binding EGF domain-containing protein 1 (CCBE1) mutations contribute to defective post-translational proteolytic activation of the VEGF-C ligand in Hennekam’s syndrome, a heritable disease characterized by congenital lymphedema and lymphangiectasia [53,54,55]. It is important to note, however, that mutations in genes encoding ADAM metallopeptidase with thrombospondin type 1 motif 3 (ADAMTS3) and the atypical cadherin FAT4 have also been associated with Hennekam’s syndrome, thus highlighting the complex pathophysiology underlying the development of primary lymphatic disorders [56,57]. Missense autosomal recessive FLT4 gene mutations have also been isolated in familial lymphedema patients with dermal lymphatic hypoplasia [58]. More recently, mutations affecting other molecular signaling pathways have been increasingly implicated in the pathogenesis of several primary lymphedema disorders, which are reviewed in more detail elsewhere [59,60,61,62,63]. VEGFR3 and neuropilin-2 transcription are elevated in various lymphovenous overgrowth malformations, suggesting the underlying hyperactivation of VEGFR3 and/or its downstream signaling pathways, in contrast to primary lymphedema disorders usually characterized by attenuated VEGFR3 signaling [64]. In experimental models of cerebral cavernous malformations (CCM), deletion of programmed cell death 10 (CCM3), a molecular regulator of cellular apoptosis, enhances VEGFR3 expression and ERK1/2 activation in LECs, leading to hyperplastic lymphatic development [65]. Importantly, the molecular defects associated with various lymphatic overgrowth malformations usually occur intracellularly downstream from the membrane VEGFR3 activation complex, which are discussed in subsequent paragraphs. Lastly, recent studies have linked aberrant VEGFR3 function with congenital heart defect development in both humans and mice, which are described extensively in a review by Monaghan et al. [66]. For example, truncating FLT4 gene variants have been identified in large-scale studies of patients with sporadic, non-syndromic Tetralogy of Fallot (TOF) [66,67,68,69]. Currently, there is a limited understanding of the molecular pathophysiology underlying early lymphatic disorders commonly associated with congenital heart diseases, including plastic bronchitis, chyloptysis, chylopericardium, and chylothorax [70,71]. Recently, variants of unknown significance (VUS) at the FLT4 locus were isolated in two primary isolated congenital chylothorax patients, thus highlighting the need for further characterization of VEGFR3 signaling in these disorders [72].
Further downstream in the VEGFR3 transduction cascade, a missense germline INPPL1 mutation affecting Sh2-domain-containing 5’-inositol phosphatase 2 (SHIP-2), an intracellular adapter protein that negatively regulates PI3K/Akt and MAPK/ERK activation, was isolated in some patients with primary lymphedema [73]. In vitro transfection of this SHIP-2 mutant enhances Akt and ERK activation following VEGF-C treatment. However, despite increased VEGFR3 signaling, mutant LECs have impaired migration, tube formation, and matrix adhesion, possibly due to increased LEC apoptosis resulting from excessive ERK activation [73].
Mutations, mostly of somatic origin, in the phosphoinositide-3-kinase (PI3K) pathway have also been identified in various lymphatic malformations. For example, somatic PIK3CA mutants have been reported in generalized lymphatic anomaly (GLA), a condition characterized by the development of multifocal or diffuse lymphatic lesions [74]. In a mouse model of GLA, hyperactive PI3K signaling resulted in lymphatic hyperplasia and impaired lymphatic transport [74]. In congenital lipomatous overgrowth, vascular malformations, epidermal nevi, and spinal/skeletal anomalies/scoliosis (CLOVES) syndrome, a disease affecting multiple organ systems, somatic activating PI3K mutations elicit pathologic lymphatic overgrowth anomalies [75,76]. Recently, Martinez-Corral et al. demonstrated that PIK3CAH1047R mutant-mediated microcystic lymphatic malformation overgrowth depends on intact VEGF-C/VEGFR3 and mTOR signaling in vivo, suggesting upstream signaling molecules play distinct roles in upregulating lymphangiogenic activities [77]. Somatic hyperactivating Akt and somatic inactivating PTEN mutations in Proteus syndrome also elicit similar phenotypic effects [74,78,79].
Alterations in the Ras signaling pathway may cause familial or sporadic lymphatic dysfunction, a common feature of ‘Rasopathy’ syndromes. For example, both germline and somatic inactivating mutations affecting H-Ras, N-Ras, K-Ras, and Son of sevenless homolog 1 (SOS1) have been linked to development of chylothorax and chylous ascites [75,80]. Activating somatic KRAS mutations have been reported in Gorham–Stout disease (GSD), where ectopic lymphatics destructively infiltrate bony structures [81]. The Dellinger group recapitulated this ectopic lymphatic growth phenotype in murine bone by generating a hyperactive KRASG12D mutant [82]. Functionally, these mutant lymphatics exhibit decreased in vivo vessel branching, increased vessel diameter, and reduced valve formation that are hypothesized to contribute to chylothorax-related death in these mice [82]. Blockade of the downstream Ras effector MEK1/2 using trametinib ameliorated the GSD phenotype, further confirming the role of hyperactive Ras signaling in LEC dysfunction. Gain-of-function Raf mutants commonly seen in central conducting lymphatic anomalies, and Noonan syndrome, a developmental disorder caused by germline mutations in one of several Ras pathway members, lead to cutaneous lymphatic malformations and lymphangiectasia in translational animal models [75,83,84,85,86]. Additionally, loss of negative Ras regulators, such as the GTPase activator p120-RasGAP (RASA1), may result in heritable lymphatic overgrowth phenotypes [23]. These experimental findings are clinically relevant, given that altered MEK/ERK RNA expression has been detected in blood samples from lymphatic malformation patients [87].
Using whole-exome sequencing, a novel variant of the EphB4 receptor, which regulates VEGFR3 internalization, was isolated in affected family members with central conducting lymphatic anomalies across four generations [88]. In zebrafish, transfection of this EphB4 mutant results in aberrant lymphatic vessel branching and increased mTORC1 and p70-S6K phosphorylation [88]. Rather unexpectedly, mTORC1 and MEK blockade both restored lymphatic branching morphology, thus also implicating EphB4 crosstalk with MAPK signaling [88]. Previous reports of EphrinB2-EphB4-p120-RasGAP-axis-driven hyperactivation of the mTORC1 pathway further illustrate the role of non-canonical Ras/MAPK-PI3K cross-talk in the regulation of downstream VEGFR3 signaling in LECs [89]. Further characterization and identification of dysregulated VEGFR3 molecular effectors may therefore enable the development of therapies to treat primary lymphedema disorders.

2.2. Secondary Lymphedema

VEGF-C, neuropilin-2, and FOXC2 polymorphisms have been identified in patients with breast cancer-related lymphedema (BCRL), suggesting that baseline differences in VEGFR3 activation or lymphatic function may contribute to the risk of developing post-surgical lymphedema [90,91]. This concept is supported by the finding that Prox-1 haploinsufficient mice with underlying subclinical lymphatic dysfunction have an increased propensity for developing lymphedema after lymphatic injury [92]. However, BCRL does not appear to be caused by a deficiency of VEGF-C because VEGF-C expression is increased in tissue biopsies and in the plasma of patients with lymphedema and in animal models of the disease [93,94,95]. Concordantly, VEGF-C overexpression in a mouse model of lymphedema results in a more rapid onset and more severe swelling due to increased vascular permeability and inflammation [96]. VEGF-C overexpression to treat BCRL led to inconclusive effects and has been largely abandoned as a therapeutic approach [97]. Genetic polymorphisms in inflammatory mediators such as IL-4, IL-10, and NF- κ B confer varying degrees of risk or protection against the development of secondary lymphedema, highlighting the importance of immune responses in regulating lymphatic function [91]. Together, these findings suggest that dysregulated VEGFR3 signaling or other mechanisms impair lymphatic regeneration and function in patients who develop secondary lymphedema.
Recent studies have suggested that alterations in regulatory microRNA (miRNA) expression may mediate the pathophysiologic features of various disease states, such as dermal inflammation, fibrosis, and immune dysregulation [98]. Expression of miR-1236 in LECs is associated with the inhibition of lymphangiogenesis via VEGFR3, Akt, and ERK downregulation [99] (Figure 2a). Similarly, the expression of miR128-3p is associated with reduced LEC proliferation via the attenuation of intracellular ERK and Ca2+ signaling [100]. A recent study showed that the expression of miR199a-3p and miR151a-3p is increased in the serum of patients with BCRL compared to serum of breast cancer patients without BCRL or healthy controls [101]. Interestingly, the expression of these microRNAs temporally correlates with lymphedema onset and is hypothesized to increase lymphedema risk by interacting with transforming growth factor-β (TGF-β), PI3K-Akt, and MAPK signaling cascades [101].
In conclusion, accumulating evidence in mice and humans shows that VEGFR3 dysregulation along with increased VEGF-C ligand is observed in secondary lymphedema, possibly due to accompanying chronic inflammatory conditions. Thus, therapies controlling inflammation might hold more potential in reversing VEGFR3 dysregulation and making use of the excess lymphangiogenic ligands for lymphatic regeneration. More detailed studies are warranted to understand the paradoxical condition of excess VEGF-C in lymphedema tissues and yet poor lymphatic function. This indirectly implicates that the problem might be in the dysregulation of VEGFR3 rather than the shortage of VEGF-C.

3. Tumor Growth and Metastatic Environment

Lymphangiogenesis and peritumoral lymphatics are associated with an increased risk of metastasis in several cancers including squamous cell cancer, breast cancer, and gastric cancer [102,103,104,105,106,107,108]. Tumoral lymphangiogenesis is fostered by tumor-derived VEGF-C and other lymphangiogenic growth factors such as angiopoietin 2 and fibroblast growth factor 2, which functionally interact with VEGFR3’s downstream effectors. Peritumoral lymphatics enhance access to local lymph nodes for tumor metastasis [109,110,111]. VEGF-C is also a potent chemoattractant for macrophages and other inflammatory cells that produce VEGF-C and modulate the tumor microenvironment [112,113]. Breast tumors also indirectly stimulate peritumoral lymphangiogenesis by inducing podoplanin expression in nearby macrophages by activating CLEC2A carbohydrate-binding receptors on LECs [114].
Aberrant expression of key molecular VEGFR3 regulators in malignant tumors further implicates lymphatic participation in metastasis progression (Figure 2b). In oral squamous cell carcinoma, elevated neuropilin-2 (Nrp2) expression is positively associated with tumor stage, lymphovascular invasion, and lymph node metastasis [115]. The expression of semaphorin 3F (SEMA3F), a Nrp2 ligand that negatively regulates VEGFR3 activation, is downregulated in esophageal squamous cell carcinoma and is associated with increased VEGF-C and Nrp2 expression [116]. Decreased semaphorin 3F expression correlates with nodal metastasis and poorer survival outcomes [116]. In hypopharyngeal cancer specimens, the elevated expression of eukaryotic translation initiation factor 4E (eIF4E), a downstream mTOR effector, is associated with increased lymphatic density within metastatic lymph nodes [117]. Enhanced integrin B1 signaling—a regulator of VEGFR3 transactivation—in peritumoral lymphatics increases the metastasis of B16 melanoma cells in mice [118].
Tumor cells also mediate lymphatic vessel recruitment by secreting various paracrine signaling molecules that act in parallel to VEGFR3. For example, tumor VEGF-D synthesis upregulates C-C motif chemokine receptor type 10 (CCR10) expression and increases LEC migration towards tumor-derived chemokine ligands 27 and 28 (CCL27/28) [119]. Similarly, tumor VEGF-C-mediated stimulation of LEC C-X-C motif chemokine receptor 4 (CXCR4) expression increases chemotactic responsiveness to peritumoral CXCL12 gradients [120]. In turn, VEGF-C-VEGFR3 signaling cascades induce LEC chemokine production, which reciprocally influences tumor activity [121,122,123]. The interaction of LEC-derived chemokine CCL21 with tumor CCR7 receptors promotes the formation of tumor–lymphatic interfaces that contribute to metastasis [102]. In in vitro studies of human skin cancers, IL-6 expression in LECs promotes tumor cell proliferation [109,124]. Breast tumor cells secrete PGE2 and induce peritumoral lymphangiogenesis in part by promoting autocrine VEGF-D expression and activating intracellular Akt and ERK signaling in murine and human LECs [114,125,126].
Although most studies report that lymphatic vessels primarily serve as enhancers of tumor metastasis, several studies have suggested that lymphatics may promote local antitumor effects by facilitating the presentation of tumor-associated antigens to the immune system [19]. Using a B16 melanoma model, Kimura et al. showed that mice with k-Cyclin deletion-mediated lymphatic dysfunction exhibited a decreased number of tumor-associated antigens in draining lymph nodes and increased primary tumor growth [127]. Transfer of CD8+ T cells from tumor-draining lymph nodes of kCYC+/− mice led to decreased immune cytotoxic activity against tumor cells in vitro and in vivo [127]. Despite increased primary tumor growth, there was a significant decrease in tumor metastasis, suggesting lymphatics regulate immune cell and tumor cell trafficking via distinct mechanisms [127]. B16 melanoma implantation studies in K14-VEGFR3-Ig and Chy mice further support these findings and show that abnormal skin lymphatics are associated with decreased intratumoral leukocyte infiltration and increased primary tumor growth [128]. Our research group has shown that the inducible ablation of peritumoral lymphatic vessels leads to increased peritumoral immunosuppressive cytokine expression, increased tumor PD-L1 expression, and decreased intratumor CD8+ T cell infiltration, which functionally correlate with increased tumor growth, suggesting tumor-associated lymphatics play active roles in regulating tumor immune responses [129]. Conversely, Song et al. found that increased VEGF-C-driven meningeal lymphatic drainage leads to improved glioblastoma tumor clearance in mice [130]. Zhou and Ma later showed that VEGF-C/VEGFR3-mediated CCL21 expression promotes dendritic cell trafficking and subsequent CD8+ T cell activation, leading to attenuated tumor growth in mouse glioma models [131,132]. A recently developed vaccine that overexpresses VEGF-C induces lymphangiogenesis and enhances T-cell-mediated antitumor immunity and sustained attenuated tumor growth in B16 melanoma mouse models [133]. Of note, some studies have reported that elevated lymphatic vessel density mediates increased immune tolerance to tumors [134,135].
Despite the evidence supporting the two faces of tumor lymphatic vessels, i.e., promoting tumor growth and progression and accelerating anti-tumor immune responses, it is not clear what warrants this dual nature. It will be interesting to understand whether tumor type dictates the pro or anti-tumor lymphatic function. Further research is needed to understand the dual nature of lymphatic vessels in regulating tumor growth and progression. Independent of the pro- or anti-tumor nature of tumor lymphatic vessels, it is getting clear that the dysregulation of VEGFR3 is critical in tumor lymphatics, and targeting VEGFR3 to regulate tumor lymphatic vessels could be an ideal way going forward.

4. Obesity and Metabolic Syndrome

The negative effects of obesity on lymphatic function have been extensively reported, but fewer studies have focused on characterizing the reciprocal contribution of dysregulated lymphangiogenic activity to the development of obesity and metabolic syndrome [136,137,138,139,140]. A link between lymphatic vessel dysfunction and metabolic syndrome is suggested by case series reporting increased serum levels of VEGF-C in patients with metabolic disorders [141,142,143]. Recently, the Rockson group published a single-center retrospective cohort analysis identifying a positive association between the presence of clinically diagnosed lymphatic disorders (lipedema, secondary lymphedema, or lymphovascular disease) and diabetes, further strengthening the hypotheses of lymphatic involvement in metabolic disease pathogenesis [144].
One of the earliest studies to implicate lymphatic dysfunction in the pathogenesis of adult-onset obesity was the discovery that haploinsufficiency of the multisystem developmental regulator Prox-1 causes this phenotype, along with the increased incidence of hepatic steatosis and elevations in circulating insulin and leptin [145] (Figure 2c). Other studies showed that VEGFR3 haploinsufficent mice (i.e., Chy mice) also have increased subcutaneous fat deposition [146,147]. Harvey et al. proposed that excessive lymph accumulation in affected tissues enhances lipid storage in existing adipocytes and increases adipogenesis, ultimately leading to increased ectopic fat deposition [145]. The same research group later validated this interpretation by demonstrating that the restoration of Prox-1 activity rescues this adult obesity phenotype in mice [148]. Consistent with these studies, our group showed that lymphatic fluid stasis resulting from surgical lymphatic disruption in mouse tail skin promotes increased adipose differentiation marker expression in affected tissues, further implicating lymphatic fluid stasis in adipogenesis [149]. These experimental studies collectively suggest that insufficient adipose tissue lymphangiogenesis or impaired lymphatic function may contribute to increased fat accumulation. In agreement with the above, Chakraborty et al. showed that adipose-specific transgenic VEGF-D overexpression in mice fed a high-fat diet (HFD) increases lymphangiogenesis, decreases macrophage infiltration, and improves systemic glucose and lipid metabolism markers [150].
Lymphatic VEGFR3 signaling is also a critical regulator of lipid and cholesterol transport in multiple cell types. The genetic deletion of VEGF-C results in lymphatic vessel regression within the lacteals and intestinal wall and decreases systemic triglyceride and cholesterol levels [151]. Using Chy mice and anti-VEGFR3 monoclonal antibodies, Martel et al. demonstrated the role of lymphatic VEGFR3 signaling in regulating macrophage reverse cholesterol transport from peripheral tissues [152]. In subsequent studies, the same researchers found that reversing hypercholesterolemia-induced lymphatic transport dysfunction with exogenous VEGF-C decreases the progression of atherosclerotic plaques in atherogenic mice on an HFD [153]. Deletion of the fatty acid transporter CD36 in LECs results in attenuated VEGF-C-induced Akt activation, impaired cellular oxidative metabolism, junctional VE-cadherin destabilization development of leaky gut lymphatics, and obesity in mice, suggesting that intracellular VEGFR3 signaling may be an attractive target for translational studies [154].
Obesity affects lymphatic function by promoting a chronic low-level inflammatory state in perivascular lymphatic tissues [136,155,156]. While studying the relationship between mesenteric lymphatic dysfunction and metabolic syndrome, Cao et al. reported that both HFD-fed obese mice and humans with obesity had leaky mesenteric lymphatics, along with elevated VEGF-C expression within extravasated lymph and neighboring visceral adipose tissue [157]. An intestinal lymphatic-targeted COX-2 inhibitor (orally administered) reduced mesenteric VEGF-C expression and concomitantly restored mesenteric lymphatic function via attenuation of macrophage-derived growth factor secretion, highlighting the importance of immune cells in regulating gastrointestinal VEGF-C homeostasis [157]. Transgenic overexpression of VEGF-C driven by the epithelial keratin-14 promoter in mouse dermal tissues results in increased weight gain and the development of insulin resistance in mutant mice, due in part to increased infiltration of pro-inflammatory M1 macrophages within subcutaneous white adipose tissue [158]. It will be interesting to observe how VEGFR3 expression and signaling on LECs change in the state of chronic low-grade inflammation with abundant VEGF-C in the tissues.
Obesity-associated inflammation also reduces lymphangiogenesis. Analyzing sorted LECs from HFD-fed C57BL/6J obesity-prone mice showed that the expression of VEGFR3 is decreased in obesity, while HFD-fed BALB/cJ and myostatin null (MSTNln) obesity-resistant mice had no significant changes to their lymphatics [159,160,161]. Based on findings of increased peri-lymphatic lipid droplet accumulation in HFD obese mice, we performed in vitro studies to determine the effects of long-chain free fatty acids on lymphatic function. LECs treated with stearic acid exhibited decreased VEGFR3 expression and downstream Akt and eNOS activation, which was reversible using pharmacologic PTEN blockade, illustrating a mechanism by which lymphangiogenesis can be modulated in disease states without the additional recruitment of immune cells [19,159]. These studies suggest obesity-associated inflammation confers increased LEC resistance to VEGFR3 activation, further contributing to the progression of metabolic dysfunction.
In conclusion, research shows that obesity decreases lymphatic function by the downregulation of VEGFR3 signaling via chronic low-grade inflammation, and, conversely, decreased lymphatic function also contributes to lipid accumulation causing obesity. Thus, specific targeting of VEGFR3 signaling to improve lymphatic function either by ligand (VEGF-C/D) overexpression or by controlling chronic inflammation with lymphatic dysfunction related obesity and additional metabolic disorders.

5. Transplant Allograft Rejection

The lymphatic vasculature is thought to contribute to chronic organ transplant rejection by promoting antigen-presenting cell (APC) trafficking and activation of adaptive immune responses (Figure 2d). This pathophysiologic process has been extensively studied in the mammalian cornea because it is avascular and immune-privileged under healthy conditions but becomes vascularized and inflamed under pathologic conditions [162,163,164,165,166,167]. In a murine allogeneic corneal transplant model, Dietrich and colleagues found that lymphangiogenesis had a greater effect on immune allograft rejection rates than angiogenesis [167]. Inhibition of angiogenesis using VEGF-Trap resulted in a lesser degree of graft survival improvement than the inhibition of lymphangiogenesis using either of the two VEGFR3 inhibitors [167]. Interestingly, galectin-8, a carbohydrate-binding protein found in LECs, has also been identified as a promoter of corneal allograft rejection via the upregulation of inflammatory lymphangiogenesis by mediating podoplanin-VEGFR3-integrin crosstalk [168].
Similarly, within a renal allograft mouse model, spontaneous regeneration of the renal lymphatic vasculature after surgery has been linked to an increase in the systemic trafficking of CCR7+ APCs involved in adaptive immune responses and progression of chronic transplant rejection [169]. Upon analyzing biopsies of renal transplant patients, these same researchers found patients experiencing chronic transplant rejection displayed greater lymphatic vessel area but not vessel number in their kidneys compared to transplant patients who were not experiencing organ rejection [169]. Additionally, VEGF-C, VEGF-D, and fibroblast growth factor 2 (FGF-2) expression was elevated within these transplant rejection patients, reflecting increased renal lymphangiogenic activity [169]. In another mouse renal transplant study, downstream inhibition of the VEGFR3 effector mTORC1 using sirolimus reduced lymphangiogenic activity, which elicited greater attenuation of chronic allograft transplant injury responses compared to calcineurin inhibition, thus implicating intracellular VEGFR3 signaling in the pathophysiologic development of chronic kidney rejection [170].
In cardiac allograft transplantation, lymphangiogenesis similarly supports immune cell infiltration to facilitate rejection. In a murine model, post-allograft ischemia–reperfusion injury stimulated VEGFR3 expression in LECs and promoted increased CD4+ T cell, CD8+ T cell, and ED1+ macrophage, and myeloperoxidase (MPO)+ neutrophil infiltration [171]. Accordingly, and in line with the protective effects of blocking VEGFR3 signaling in other transplant settings, inducible LEC-specific deletion of VEGFR3 prior to transplant surgery improves graft survival rates [171]. In another study, increased lymph flow and enhanced migration of donor passenger leukocytes from cardiac allografts to recipient draining lymph nodes was associated with increased allograft CD8+ T cell infiltration [172]. Furthermore, cardiac allograft vasculopathy, determined by the severity of arterial luminal occlusion, correlated with CD8+ T cell infiltration density and lymphatic vessel area, suggesting lymphangiogenic activity contributes to graft loss [172]. Inhibition of LEC VEGFR3 signaling in a rat cardiac allograft model reduced the tissue expression of CCL21, a chemokine promoter of dendritic cell migration, which reduced inflammatory cell infiltration and prolonged graft viability [123]. VEGFR3 has also been shown to promote the migration of peripherally injected naïve CD4+ T cells to draining lymph nodes by regulating extracellular heparan sulfate expression and resultant CCL21 gradients in a PI3K-dependent manner [173].
Many transplant rejection models conceptualize lymphangiogenesis as a pathogenic driver of allograft failure. However, based on experimental findings of decreased pulmonary lymphatic density and increased hyaluronan (HA) fragment accumulation after mouse lung transplantation, Cui et al. hypothesized that impaired hyaluronan clearance from inadequate lymphatic drainage may mediate acute lung transplant rejection [174]. Compared to controls and isografts, lung allografts exhibited a significantly higher proportion of apoptotic LECs and lower nuclear proliferation marker expression, thus confirming impaired lymphangiogenic capacity [174]. In contrast to other transplant settings, daily intravenous VEGF-C156S (the lymphangiogenesis-specific form of VEGF-C) injections enhanced lymphatic drainage and HA clearance and attenuated acute rejection responses in treated mice [174]. These findings appear to be clinically relevant, since reductions in tissue HA levels following treatment for acute lung transplant rejection were associated with improved respiratory functional outcomes [174].
In conclusion, corneal, renal, and cardiac transplant rejections are mainly due to activated VEGFR3 signaling, and blocking VEGFR3 signaling is a potential therapeutic target for longer transplant survival. On the contrary, lung transplant survival is proper lymphatic drainage dependent, and thus activation of VEGFR3 signaling through pro-lymphangiogenic agents seems to be critical. Further research has yet to reveal how lung transplants differ from other organ transplants in terms of opposite lymphatic and VEGFR3 signaling requirements.

6. Autoimmune and Autoinflammatory Disorders

Altered VEGFR3 signaling and the resulting lymphatic vessel dysfunction have also been implicated in the pathogenesis of multiple autoinflammatory disorders, including several glandular autoinflammatory conditions. Single-nucleotide polymorphisms (SNPs) in the gene encoding VEGF-C have been identified as potential risk factors for developing autoimmune thyroid diseases, including Grave’s disease and Hashimoto’s thyroiditis [175]. Within the minor salivary glands of primary Sjogren’s syndrome patients, newly expanded lymphatic capillaries display increased VEGFR3 expression and increased periductal inflammatory cell infiltration [176] (Figure 2e). Behçet’s disease patients experiencing uveitis exhibit higher circulating levels of soluble VEGFR3 (sVEGFR3) and a lower ratio of VEGF-C/sVEGFR3, suggesting dysregulated lymphangiogenesis [177].
Research shows that systemic lupus erythematosus (SLE) and systemic sclerosis (SS) display cutaneous lymphatic dysfunction with limited available information on VEGFR3 expression. SLE patients exhibit dilated cutaneous lymphatic vessels without significant changes in lymphatic density [178]. Ambler et al. recapitulated these findings in an ultra-violet radiation (UVR)-sensitive mouse model in which SLE is induced via chronic epicutaneous imiquimod treatment [178]. LEC-specific PTEN gene knockout, which enhances intracellular VEGFR3 activation and resulting lymphatic drainage, in imiquimod-treated mice following UVR exposure led to decreased inflammation and B-cell responses in draining lymph nodes [178]. Cutaneous biopsies from systemic sclerosis (SS) patients with multi-organ fibrosis display diminished lymphatic vessel density but increased VEGFR3 and VEGF-D mRNA expression compared to healthy controls [179]. Another study in SS patients noted an inverse correlation between the number of fingertip ulcers and cutaneous biopsy lymphatic vessel counts [180]. In vitro treatment of wildtype LECs with SS-patient-derived serum resulted in reduced VEGFR3 expression and impaired migratory, proliferative, and tube-forming capacity [181].
While lymphangiogenesis is usually regulated by VEGF-C/VEGFR3, a VEGF-A overexpression mouse model of psoriasis displayed persistently increased lymphatic vessel size, tortuosity, and proliferation as part of delayed-type hypersensitivity reactions. These changes were reversible with combined VEGFR1-VEGFR2 blockade, thereby implicating excessive LEC VEGFR2 signaling in the pathogenesis of at least one type of inflammatory skin disease [182].
Within a mouse model of alopecia areata, an autoimmune condition, lymphatic vessels within affected dermal tissues were distended, suggesting lymphatic contribution to hair growth [183]. Transgenic overexpression of VEGF-C and resulting increased dermal lymphatic density promoted prolonged anagen hair follicle growth via paracrine activation of dermal papilla cells [184,185]. Conversely, the expression of soluble VEGFR3 (sVEGFR3) extracellular domains produced the opposite results, indicating that VEGFR3 underactivity limits hair growth [184]. Additionally, recent seminal finding related to hair follicle stem cells (even though not directly related to alopecia) indicate that hair follicle stem cell (HFSC)-associated lymphatics have also been shown to promote stem cell quiescence [186]. Diphtheria-toxin-mediated dermal lymphatic ablation promoted precocious telogen-to-anagen transitions in hair follicles, which was also recapitulated following soluble VEGFR3 antibody treatment, implicating VEGFR3 signaling in this process [186]. In turn, HFSCs reciprocally regulate lymphatic activity via angiopoietin-like protein 7 and angiopoietin-like protein 4 expression [186]. Further studies in this direction are needed to fully characterize the role of lymphatic vessels in hair growth and regeneration and the link to autoimmunity in this context.
Certain VEGF-C SNPs confer the increased risk for developing rheumatoid arthritis (RA), suggesting a potential contribution of dysregulated VEGFR3 signaling [187]. Additionally, TNF-α, the primary driver of RA progression, has been shown to increase VEGF-C expression in the affected joint synovial fluid of RA patients, potentially disrupting endogenous growth factor gradients [188]. Several experimental RA models propose that lymphatic insufficiency may worsen disease progression, which is supported by clinical findings of impaired lymphatic clearance of indocyanine green (ICG) dye within the affected hands of RA patients and experimental findings of decreased lymphatic vessel maturity within the inflamed joints of transgenic TNF-expressing mice [189,190]. Within translational models of inflammatory arthritis, VEGFR3 blockade results in decreased lymphangiogenesis, impaired afferent lymphatic drainage, and increased synovial volumes in transgenic tumor necrosis factor (TNF)-overexpressing mice [191]. Conversely, treatment with adenovirus expressing VEGF-C has been shown to increase intraarticular lymphatic vessel density and concomitantly reduce joint swelling and inflammation [192]. Together, these findings indicate that impaired lymphangiogenesis and lymphatic function promotes the pathogenesis of RA [191,192]. In contrast, in patients with adult-onset Still’s disease, an incompletely understood autoinflammatory disorder characterized by fever, arthralgias, and lymphadenopathy, circulating VEGF-C has recently been found to be elevated [193]. Interestingly, serum VEGF-C concentrations correlated with active symptoms, inflammatory cytokine levels, and disease severity markers [193].
Elevated VEGF-C, VEGFR3, podoplanin, and LYVE-1 mRNA expression has been observed within the inflamed colonic mucosa of ulcerative colitis (UC) and Crohn’s disease (CD) patients [194,195]. This has been reproduced in dextran sodium sulfate (DSS)-induced mouse models of inflammatory bowel disease (IBD), in which the intestinal lymphatics exhibit increased vessel density, elevated VEGFR3 expression, and vessel dilation, suggesting that overactive lymphangiogenesis may be involved in disease pathogenesis [196,197]. However, this hypothesis is complicated by the finding that VEGFR3 blockade induces the development of dilated and tortuous lymphatics in the IL-10 knockout model of spontaneous inflammatory bowel disease (IBD), which was shown to worsen intestinal inflammation [198]. To better understand these relationships, several research groups have induced VEGF-C overexpression, leading to improvement in some studies and worse intestinal inflammation in others [196,199]. Indeed, it remains unknown if these histopathologic findings are causative or compensatory in nature, making it difficult to determine if insufficient versus excessive lymphangiogenesis is to blame [200]. However, dysfunctional lymphatics clearly exert some influence on intestinal inflammatory disease progression.
In conclusion, autoimmune pathologies seem to be highly heterogeneous, showing a disease specific up- and downregulation of VEGFR3 signaling. While SLE, SS, and alopecia exhibit reduced VEGFR3 signaling, RA, UC, CD, and IBD display increased VEGFR3 signaling; thus, both pro- and anti-lymphatic therapies are in play, depending on the disease type. For future studies, it will be interesting to elucidate the basis of the heterogeneity in VEGFR3 signaling between different autoimmune conditions to specifically target lymphatic vessels and VEGFR3 signaling.

7. Conclusions

Aberrant VEGFR3 signal transduction and altered lymphangiogenesis or lymphatic function may contribute to the development of many diseases, as summarized in Table 1. This effect is likely due to the central role of the lymphatic system in tissue fluid drainage, fat absorption, and immune cell trafficking, as well as the propensity of lymphatic vessels to become injured by chronic inflammatory reactions. The heterogeneity in VEGFR3 signaling across different pathologies shows that not all diseases arise uniformly with respect to alterations in lymphatic growth and function. For example, diseases like secondary lymphedema and obesity largely show decreased VEGFR3 signaling despite increased VEGF-C ligand expression in affected tissues, indicating a probable development of VEGF-C resistance in these pathologies, akin to the phenomenon of insulin resistance [201]. Thus, therapeutic interventions in these diseases should be focused on enhancing VEGFR3 signaling by regulating chronic inflammation rather than exogenous lymphangiogenic ligand supply. Demonstrating the concept of VEGF-C resistance in lymphatic pathologies and understanding the molecular basis of such phenomena could be a potential new direction for future studies. In contrast to lymphedema and obesity, many cancers and organ transplant rejection cases, when viewed from a lymphatic perspective, arise mainly due to increased VEGFR3 signaling and the pathologic activation of lymphatic growth. In these conditions, therapeutic strategies should rely on dampening VEGFR3 signaling. Finally, autoimmune pathologies involving lymphatic dysfunction appear to be a mixed bag, with some pathologies showing increased while others show decreased VEGFR3 signaling, therefore necessitating disease-specific therapeutic interventions. Overall, understanding the cellular mechanisms that regulate the crosstalk between lymphatic vessels and other tissues is an important research goal. This understanding may lead to novel targeted therapies that can impact not only primarily lymphatic diseases such as lymphedema but also a host of common diseases including obesity, metabolic syndrome, tumors, and autoimmune disorders.

Author Contributions

K.K., R.P.K. and B.J.M. conceptualized the review; K.K., R.P.K. and A.-C.C. drafted the manuscript; A.-C.C. made the figures; A.R. made the table; B.L.P., H.J.P., A.S., S.B. and J.S. helped in the literature search and initial draft of the manuscript. All the other authors reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by the National Institutes of Health through R01HL111130, R01CA278599, R21AR081076, and R21AG076132 (awarded to B.J.M.), and T32CA009501 (stipend for A.-C.C.), as well as the Cancer Center Support Grant P30 CA008748, which supports the research infrastructure at MSK.

Data Availability Statement

Data sharing is not applicable to this article.

Acknowledgments

The authors would like to acknowledge Dagmar Schnau and Jessica Moore, our colleagues from the Memorial Sloan Kettering Department of Surgery, for editorial and formatting assistance.

Conflicts of Interest

Outside of the submitted work, B.J.M. is the recipient of investigator-initiated research awards from Regeneron Corp and Pfizer and royalty payments from PureTech, as well as being a consultant for Mediflix Corp. The other authors have no relationships with outside entities to disclose. The funders had no role in the writing of the manuscript or in the decision to reference the selected publications.

References

  1. Petrova, T.V.; Koh, G.Y. Biological functions of lymphatic vessels. Science 2020, 369, eaax4063. [Google Scholar] [CrossRef] [PubMed]
  2. Wong, B.W.; Zecchin, A.; García-Caballero, M.; Carmeliet, P. Emerging Concepts in Organ-Specific Lymphatic Vessels and Metabolic Regulation of Lymphatic Development. Dev. Cell 2018, 45, 289–301. [Google Scholar] [CrossRef] [PubMed]
  3. Petrova, T.V.; Koh, G.Y. Organ-specific lymphatic vasculature: From development to pathophysiology. J. Exp. Med. 2018, 215, 35–49. [Google Scholar] [CrossRef] [PubMed]
  4. Galland, F.; Karamysheva, A.; Pebusque, M.J.; Borg, J.P.; Rottapel, R.; Dubreuil, P.; Rosnet, O.; Birnbaum, D. The FLT4 gene encodes a transmembrane tyrosine kinase related to the vascular endothelial growth factor receptor. Oncogene 1993, 8, 1233–1240. [Google Scholar]
  5. Kukk, E.; Lymboussaki, A.; Taira, S.; Kaipainen, A.; Jeltsch, M.; Joukov, V.; Alitalo, K. VEGF-C receptor binding and pattern of expression with VEGFR-3 suggests a role in lymphatic vascular development. Development 1996, 122, 3829–3837. [Google Scholar] [CrossRef]
  6. Zhang, L.; Zhou, F.; Han, W.; Shen, B.; Luo, J.; Shibuya, M.; He, Y. VEGFR-3 ligand-binding and kinase activity are required for lymphangiogenesis but not for angiogenesis. Cell Res. 2010, 20, 1319–1331. [Google Scholar] [CrossRef]
  7. Salameh, A.; Galvagni, F.; Bardelli, M.; Bussolino, F.; Oliviero, S. Direct recruitment of CRK and GRB2 to VEGFR-3 induces proliferation, migration, and survival of endothelial cells through the activation of ERK, AKT, and JNK pathways. Blood 2005, 106, 3423–3431. [Google Scholar] [CrossRef]
  8. Wang, J.F.; Zhang, X.; Groopman, J.E. Activation of vascular endothelial growth factor receptor-3 and its downstream signaling promote cell survival under oxidative stress. J. Biol. Chem. 2004, 279, 27088–27097. [Google Scholar] [CrossRef]
  9. Deng, Y.; Zhang, X.; Simons, M. Molecular controls of lymphatic VEGFR3 signaling. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 421–429. [Google Scholar] [CrossRef]
  10. Mäkinen, T.; Veikkola, T.; Mustjoki, S.; Karpanen, T.; Catimel, B.; Nice, E.C.; Wise, L.; Mercer, A.; Kowalski, H.; Kerjaschki, D.; et al. Isolated lymphatic endothelial cells transduce growth, survival and migratory signals via the VEGF-C/D receptor VEGFR-3. EMBO J. 2001, 20, 4762–4773. [Google Scholar] [CrossRef]
  11. Zhou, F.; Chang, Z.; Zhang, L.; Hong, Y.K.; Shen, B.; Wang, B.; Zhang, F.; Lu, G.; Tvorogov, D.; Alitalo, K.; et al. Akt/Protein kinase B is required for lymphatic network formation, remodeling, and valve development. Am. J. Pathol. 2010, 177, 2124–2133. [Google Scholar] [CrossRef] [PubMed]
  12. Deng, Y.; Atri, D.; Eichmann, A.; Simons, M. Endothelial ERK signaling controls lymphatic fate specification. J. Clin. Investig. 2013, 123, 1202–1215. [Google Scholar] [CrossRef] [PubMed]
  13. Coso, S.; Zeng, Y.; Opeskin, K.; Williams, E.D. Vascular endothelial growth factor receptor-3 directly interacts with phosphatidylinositol 3-kinase to regulate lymphangiogenesis. PLoS ONE 2012, 7, e39558. [Google Scholar] [CrossRef] [PubMed]
  14. Harris, T.K. PDK1 and PKB/Akt: Ideal targets for development of new strategies to structure-based drug design. IUBMB Life 2003, 55, 117–126. [Google Scholar] [CrossRef] [PubMed]
  15. Lahdenranta, J.; Hagendoorn, J.; Padera, T.P.; Hoshida, T.; Nelson, G.; Kashiwagi, S.; Jain, R.K.; Fukumura, D. Endothelial nitric oxide synthase mediates lymphangiogenesis and lymphatic metastasis. Cancer Res. 2009, 69, 2801–2808. [Google Scholar] [CrossRef]
  16. Huber, S.; Bruns, C.J.; Schmid, G.; Hermann, P.C.; Conrad, C.; Niess, H.; Huss, R.; Graeb, C.; Jauch, K.W.; Heeschen, C.; et al. Inhibition of the mammalian target of rapamycin impedes lymphangiogenesis. Kidney Int. 2007, 71, 771–777. [Google Scholar] [CrossRef]
  17. Luo, Y.; Liu, L.; Rogers, D.; Su, W.; Odaka, Y.; Zhou, H.; Chen, W.; Shen, T.; Alexander, J.S.; Huang, S. Rapamycin inhibits lymphatic endothelial cell tube formation by downregulating vascular endothelial growth factor receptor 3 protein expression. Neoplasia 2012, 14, 228–237. [Google Scholar] [CrossRef]
  18. Primo, L.; di Blasio, L.; Roca, C.; Droetto, S.; Piva, R.; Schaffhausen, B.; Bussolino, F. Essential role of PDK1 in regulating endothelial cell migration. J. Cell Biol. 2007, 176, 1035–1047. [Google Scholar] [CrossRef]
  19. Kataru, R.P.; Baik, J.E.; Park, H.J.; Ly, C.L.; Shin, J.; Schwartz, N.; Lu, T.T.; Ortega, S.; Mehrara, B.J. Lymphatic-specific intracellular modulation of receptor tyrosine kinase signaling improves lymphatic growth and function. Sci. Signal. 2021, 14, eabc0836. [Google Scholar] [CrossRef]
  20. Bui, K.; Hong, Y.K. Ras Pathways on Prox1 and Lymphangiogenesis: Insights for Therapeutics. Front. Cardiovasc. Med. 2020, 7, 597374. [Google Scholar] [CrossRef]
  21. Ichise, T.; Yoshida, N.; Ichise, H. Ras/MAPK signaling modulates VEGFR-3 expression through Ets-mediated p300 recruitment and histone acetylation on the Vegfr3 gene in lymphatic endothelial cells. PLoS ONE 2012, 7, e51639. [Google Scholar] [CrossRef]
  22. Takahashi, T.; Ueno, H.; Shibuya, M. VEGF activates protein kinase C-dependent, but Ras-independent Raf-MEK-MAP kinase pathway for DNA synthesis in primary endothelial cells. Oncogene 1999, 18, 2221–2230. [Google Scholar] [CrossRef] [PubMed]
  23. Lapinski, P.E.; Kwon, S.; Lubeck, B.A.; Wilkinson, J.E.; Srinivasan, R.S.; Sevick-Muraca, E.; King, P.D. RASA1 maintains the lymphatic vasculature in a quiescent functional state in mice. J. Clin. Investig. 2012, 122, 733–747. [Google Scholar] [CrossRef] [PubMed]
  24. Datta, S.R.; Brunet, A.; Greenberg, M.E. Cellular survival: A play in three Akts. Genes Dev. 1999, 13, 2905–2927. [Google Scholar] [CrossRef] [PubMed]
  25. Kandel, E.S.; Hay, N. The regulation and activities of the multifunctional serine/threonine kinase Akt/PKB. Exp. Cell Res. 1999, 253, 210–229. [Google Scholar] [CrossRef] [PubMed]
  26. Ren, B.; Deng, Y.; Mukhopadhyay, A.; Lanahan, A.A.; Zhuang, Z.W.; Moodie, K.L.; Mulligan-Kehoe, M.J.; Byzova, T.V.; Peterson, R.T.; Simons, M. ERK1/2-Akt1 crosstalk regulates arteriogenesis in mice and zebrafish. J. Clin. Investig. 2010, 120, 1217–1228. [Google Scholar] [CrossRef] [PubMed]
  27. Uchida, Y.; James, J.M.; Suto, F.; Mukouyama, Y.S. Class 3 semaphorins negatively regulate dermal lymphatic network formation. Biol. Open 2015, 4, 1194–1205. [Google Scholar] [CrossRef] [PubMed]
  28. Xu, Y.; Yuan, L.; Mak, J.; Pardanaud, L.; Caunt, M.; Kasman, I.; Larrivée, B.; Del Toro, R.; Suchting, S.; Medvinsky, A.; et al. Neuropilin-2 mediates VEGF-C-induced lymphatic sprouting together with VEGFR3. J. Cell Biol. 2010, 188, 115–130. [Google Scholar] [CrossRef]
  29. Parker, M.W.; Linkugel, A.D.; Goel, H.L.; Wu, T.; Mercurio, A.M.; Vander Kooi, C.W. Structural basis for VEGF-C binding to neuropilin-2 and sequestration by a soluble splice form. Structure 2015, 23, 677–687. [Google Scholar] [CrossRef]
  30. Wang, Y.; Nakayama, M.; Pitulescu, M.E.; Schmidt, T.S.; Bochenek, M.L.; Sakakibara, A.; Adams, S.; Davy, A.; Deutsch, U.; Lüthi, U.; et al. Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Nature 2010, 465, 483–486. [Google Scholar] [CrossRef]
  31. Wu, B.; Rockel, J.S.; Lagares, D.; Kapoor, M. Ephrins and Eph Receptor Signaling in Tissue Repair and Fibrosis. Curr. Rheumatol. Rep. 2019, 21, 23. [Google Scholar] [CrossRef] [PubMed]
  32. Galvagni, F.; Pennacchini, S.; Salameh, A.; Rocchigiani, M.; Neri, F.; Orlandini, M.; Petraglia, F.; Gotta, S.; Sardone, G.L.; Matteucci, G.; et al. Endothelial cell adhesion to the extracellular matrix induces c-Src-dependent VEGFR-3 phosphorylation without the activation of the receptor intrinsic kinase activity. Circ. Res. 2010, 106, 1839–1848. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, X.; Groopman, J.E.; Wang, J.F. Extracellular matrix regulates endothelial functions through interaction of VEGFR-3 and integrin alpha5beta1. J. Cell. Physiol. 2005, 202, 205–214. [Google Scholar] [CrossRef] [PubMed]
  34. Kumaravel, S.; Abbey, C.A.; Bayless, K.J.; Chakraborty, S. The β(1)-integrin plays a key role in LEC invasion in an optimized 3-D collagen matrix model. Am. J. Physiol. Cell Physiol. 2020, 319, C1045–C1058. [Google Scholar] [CrossRef] [PubMed]
  35. Yin, X.; Johns, S.C.; Lawrence, R.; Xu, D.; Reddi, K.; Bishop, J.R.; Varner, J.A.; Fuster, M.M. Lymphatic endothelial heparan sulfate deficiency results in altered growth responses to vascular endothelial growth factor-C (VEGF-C). J. Biol. Chem. 2011, 286, 14952–14962. [Google Scholar] [CrossRef]
  36. Sun, M.; Puri, S.; Mutoji, K.N.; Coulson-Thomas, Y.M.; Hascall, V.C.; Jackson, D.G.; Gesteira, T.F.; Coulson-Thomas, V.J. Hyaluronan Derived From the Limbus is a Key Regulator of Corneal Lymphangiogenesis. Investig. Ophthalmol. Vis. Sci. 2019, 60, 1050–1062. [Google Scholar] [CrossRef]
  37. Tan, K.W.; Chong, S.Z.; Angeli, V. Inflammatory lymphangiogenesis: Cellular mediators and functional implications. Angiogenesis 2014, 17, 373–381. [Google Scholar] [CrossRef]
  38. Kim, H.; Kataru, R.P.; Koh, G.Y. Regulation and implications of inflammatory lymphangiogenesis. Trends Immunol. 2012, 33, 350–356. [Google Scholar] [CrossRef]
  39. Wu, M.; Matar, D.Y.; Yu, Z.; Chen, Z.; Knoedler, S.; Ng, B.; Darwish, O.; Haug, V.; Friedman, L.; Orgill, D.P.; et al. Modulation of Lymphangiogenesis in Incisional Murine Diabetic Wound Healing using Negative Pressure Wound Therapy. Adv. Wound Care 2022, 12, 483–497. [Google Scholar] [CrossRef]
  40. Yang, Y.; García-Verdugo, J.M.; Soriano-Navarro, M.; Srinivasan, R.S.; Scallan, J.P.; Singh, M.K.; Epstein, J.A.; Oliver, G. Lymphatic endothelial progenitors bud from the cardinal vein and intersomitic vessels in mammalian embryos. Blood 2012, 120, 2340–2348. [Google Scholar] [CrossRef]
  41. Oliver, G. Lymphatic endothelial cell fate specification in the mammalian embryo: An historical perspective. Dev. Biol. 2022, 482, 44–54. [Google Scholar] [CrossRef] [PubMed]
  42. Karkkainen, M.J.; Haiko, P.; Sainio, K.; Partanen, J.; Taipale, J.; Petrova, T.V.; Jeltsch, M.; Jackson, D.G.; Talikka, M.; Rauvala, H.; et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat. Immunol. 2004, 5, 74–80. [Google Scholar] [CrossRef] [PubMed]
  43. Oliver, G.; Srinivasan, R.S. Endothelial cell plasticity: How to become and remain a lymphatic endothelial cell. Development 2010, 137, 363–372. [Google Scholar] [CrossRef] [PubMed]
  44. Jerafi-Vider, A.; Bassi, I.; Moshe, N.; Tevet, Y.; Hen, G.; Splittstoesser, D.; Shin, M.; Lawson, N.D.; Yaniv, K. VEGFC/FLT4-induced cell-cycle arrest mediates sprouting and differentiation of venous and lymphatic endothelial cells. Cell Rep. 2021, 35, 109255. [Google Scholar] [CrossRef] [PubMed]
  45. Srinivasan, R.S.; Escobedo, N.; Yang, Y.; Interiano, A.; Dillard, M.E.; Finkelstein, D.; Mukatira, S.; Gil, H.J.; Nurmi, H.; Alitalo, K.; et al. The Prox1-Vegfr3 feedback loop maintains the identity and the number of lymphatic endothelial cell progenitors. Genes Dev. 2014, 28, 2175–2187. [Google Scholar] [CrossRef] [PubMed]
  46. Francois, M.; Oszmiana, A.; Harvey, N.L. When form meets function: The cells and signals that shape the lymphatic vasculature during development. Development 2021, 148, dev167098. [Google Scholar] [CrossRef] [PubMed]
  47. Gordon, K.; Spiden, S.L.; Connell, F.C.; Brice, G.; Cottrell, S.; Short, J.; Taylor, R.; Jeffery, S.; Mortimer, P.S.; Mansour, S.; et al. FLT4/VEGFR3 and Milroy disease: Novel mutations, a review of published variants and database update. Hum. Mutat. 2013, 34, 23–31. [Google Scholar] [CrossRef]
  48. Irrthum, A.; Karkkainen, M.J.; Devriendt, K.; Alitalo, K.; Vikkula, M. Congenital hereditary lymphedema caused by a mutation that inactivates VEGFR3 tyrosine kinase. Am. J. Hum. Genet. 2000, 67, 295–301. [Google Scholar] [CrossRef]
  49. Karkkainen, M.J.; Ferrell, R.E.; Lawrence, E.C.; Kimak, M.A.; Levinson, K.L.; McTigue, M.A.; Alitalo, K.; Finegold, D.N. Missense mutations interfere with VEGFR-3 signalling in primary lymphoedema. Nat. Genet. 2000, 25, 153–159. [Google Scholar] [CrossRef]
  50. Ferrell, R.E.; Levinson, K.L.; Esman, J.H.; Kimak, M.A.; Lawrence, E.C.; Barmada, M.M.; Finegold, D.N. Hereditary lymphedema: Evidence for linkage and genetic heterogeneity. Hum. Mol. Genet. 1998, 7, 2073–2078. [Google Scholar] [CrossRef]
  51. Gordon, K.; Schulte, D.; Brice, G.; Simpson, M.A.; Roukens, M.G.; van Impel, A.; Connell, F.; Kalidas, K.; Jeffery, S.; Mortimer, P.S.; 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, 112, 956–960. [Google Scholar] [CrossRef] [PubMed]
  52. Balboa-Beltran, E.; Fernández-Seara, M.J.; Pérez-Muñuzuri, A.; Lago, R.; García-Magán, C.; Couce, M.L.; Sobrino, B.; Amigo, J.; Carracedo, A.; Barros, F. A novel stop mutation in the vascular endothelial growth factor-C gene (VEGFC) results in Milroy-like disease. J. Med. Genet. 2014, 51, 475–478. [Google Scholar] [CrossRef] [PubMed]
  53. Le Guen, L.; Karpanen, T.; Schulte, D.; Harris, N.C.; Koltowska, K.; Roukens, G.; Bower, N.I.; van Impel, A.; Stacker, S.A.; Achen, M.G.; et al. Ccbe1 regulates Vegfc-mediated induction of Vegfr3 signaling during embryonic lymphangiogenesis. Development 2014, 141, 1239–1249. [Google Scholar] [CrossRef] [PubMed]
  54. Frosk, P.; Chodirker, B.; Simard, L.; El-Matary, W.; Hanlon-Dearman, A.; Schwartzentruber, J.; Majewski, J.; Rockman-Greenberg, C. A novel CCBE1 mutation leading to a mild form of hennekam syndrome: Case report and review of the literature. BMC Med. Genet. 2015, 16, 28. [Google Scholar] [CrossRef]
  55. Jeltsch, M.; Jha, S.K.; Tvorogov, D.; Anisimov, A.; Leppänen, V.M.; Holopainen, T.; Kivelä, R.; Ortega, S.; Kärpanen, T.; Alitalo, K. CCBE1 enhances lymphangiogenesis via A disintegrin and metalloprotease with thrombospondin motifs-3-mediated vascular endothelial growth factor-C activation. Circulation 2014, 129, 1962–1971. [Google Scholar] [CrossRef]
  56. Brouillard, P.; Dupont, L.; Helaers, R.; Coulie, R.; Tiller, G.E.; Peeden, J.; Colige, A.; Vikkula, M. Loss of ADAMTS3 activity causes Hennekam lymphangiectasia-lymphedema syndrome 3. Hum. Mol. Genet. 2017, 26, 4095–4104. [Google Scholar] [CrossRef]
  57. Alders, M.; Al-Gazali, L.; Cordeiro, I.; Dallapiccola, B.; Garavelli, L.; Tuysuz, B.; Salehi, F.; Haagmans, M.A.; Mook, O.R.; Majoie, C.B.; et al. Hennekam syndrome can be caused by FAT4 mutations and be allelic to Van Maldergem syndrome. Hum. Genet. 2014, 133, 1161–1167. [Google Scholar] [CrossRef]
  58. Ghalamkarpour, A.; Holnthoner, W.; Saharinen, P.; Boon, L.M.; Mulliken, J.B.; Alitalo, K.; Vikkula, M. Recessive primary congenital lymphoedema caused by a VEGFR3 mutation. J. Med. Genet. 2009, 46, 399–404. [Google Scholar] [CrossRef]
  59. Leppänen, V.M.; Brouillard, P.; Korhonen, E.A.; Sipilä, T.; Jha, S.K.; Revencu, N.; Labarque, V.; Fastré, E.; Schlögel, M.; Ravoet, M.; et al. Characterization of ANGPT2 mutations associated with primary lymphedema. Sci. Transl. Med. 2020, 12, eaax8013. [Google Scholar] [CrossRef]
  60. Maltese, P.E.; Michelini, S.; Ricci, M.; Maitz, S.; Fiorentino, A.; Serrani, R.; Lazzerotti, A.; Bruson, A.; Paolacci, S.; Benedetti, S.; et al. Increasing evidence of hereditary lymphedema caused by CELSR1 loss-of-function variants. Am. J. Med. Genet. A 2019, 179, 1718–1724. [Google Scholar] [CrossRef]
  61. Finegold, D.N.; Schacht, V.; Kimak, M.A.; Lawrence, E.C.; Foeldi, E.; Karlsson, J.M.; Baty, C.J.; Ferrell, R.E. HGF and MET mutations in primary and secondary lymphedema. Lymphat. Res. Biol. 2008, 6, 65–68. [Google Scholar] [CrossRef] [PubMed]
  62. Gordon, K.; Varney, R.; Keeley, V.; Riches, K.; Jeffery, S.; Van Zanten, M.; Mortimer, P.; Ostergaard, P.; Mansour, S. Update and audit of the St George’s classification algorithm of primary lymphatic anomalies: A clinical and molecular approach to diagnosis. J. Med. Genet. 2020, 57, 653–659. [Google Scholar] [CrossRef] [PubMed]
  63. Ostergaard, P.; Simpson, M.A.; Mendola, A.; Vasudevan, P.; Connell, F.C.; van Impel, A.; Moore, A.T.; Loeys, B.L.; Ghalamkarpour, A.; Onoufriadis, A.; et al. Mutations in KIF11 cause autosomal-dominant microcephaly variably associated with congenital lymphedema and chorioretinopathy. Am. J. Hum. Genet. 2012, 90, 356–362. [Google Scholar] [CrossRef] [PubMed]
  64. Partanen, T.A.; Vuola, P.; Jauhiainen, S.; Lohi, J.; Salminen, P.; Pitkäranta, A.; Häkkinen, S.K.; Honkonen, K.; Alitalo, K.; Ylä-Herttuala, S. Neuropilin-2 and vascular endothelial growth factor receptor-3 are up-regulated in human vascular malformations. Angiogenesis 2013, 16, 137–146. [Google Scholar] [CrossRef] [PubMed]
  65. Qin, L.; Zhang, H.; Li, B.; Jiang, Q.; Lopez, F.; Min, W.; Zhou, J.H. CCM3 Loss-Induced Lymphatic Defect Is Mediated by the Augmented VEGFR3-ERK1/2 Signaling. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 2943–2960. [Google Scholar] [CrossRef] [PubMed]
  66. Monaghan, R.M.; Page, D.J.; Ostergaard, P.; Keavney, B.D. The physiological and pathological functions of VEGFR3 in cardiac and lymphatic development and related diseases. Cardiovasc. Res. 2021, 117, 1877–1890. [Google Scholar] [CrossRef] [PubMed]
  67. Jin, S.C.; Homsy, J.; Zaidi, S.; Lu, Q.; Morton, S.; DePalma, S.R.; Zeng, X.; Qi, H.; Chang, W.; Sierant, M.C.; et al. Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands. Nat. Genet. 2017, 49, 1593–1601. [Google Scholar] [CrossRef] [PubMed]
  68. Reuter, M.S.; Jobling, R.; Chaturvedi, R.R.; Manshaei, R.; Costain, G.; Heung, T.; Curtis, M.; Hosseini, S.M.; Liston, E.; Lowther, C.; et al. Haploinsufficiency of vascular endothelial growth factor related signaling genes is associated with tetralogy of Fallot. Genet. Med. 2019, 21, 1001–1007. [Google Scholar] [CrossRef]
  69. Page, D.J.; Miossec, M.J.; Williams, S.G.; Monaghan, R.M.; Fotiou, E.; Cordell, H.J.; Sutcliffe, L.; Topf, A.; Bourgey, M.; Bourque, G.; et al. Whole Exome Sequencing Reveals the Major Genetic Contributors to Nonsyndromic Tetralogy of Fallot. Circ. Res. 2019, 124, 553–563. [Google Scholar] [CrossRef]
  70. Ramirez-Suarez, K.I.; Tierradentro-García, L.O.; Biko, D.M.; Otero, H.J.; White, A.M.; Dori, Y.; Smith, C.L.; Vatsky, S.; Rapp, J.B. Lymphatic anomalies in congenital heart disease. Pediatr. Radiol. 2022, 52, 1862–1876. [Google Scholar] [CrossRef]
  71. Itkin, M.; Chidekel, A.; Ryan, K.A.; Rabinowitz, D. Abnormal pulmonary lymphatic flow in patients with paediatric pulmonary lymphatic disorders: Diagnosis and treatment. Paediatr. Respir. Rev. 2020, 36, 15–24. [Google Scholar] [CrossRef] [PubMed]
  72. Schneider, S.; Köllges, R.; Stegmann, J.D.; Thieme, F.; Hilger, A.C.; Waffenschmidt, L.; Fazaal, J.; Kalanithy, J.C.; Geipel, A.; Strizek, B.; et al. Resequencing of VEGFR3 pathway genes implicate GJC2 and FLT4 in the formation of primary congenital chylothorax. Am. J. Med. Genet. A 2022, 188, 1607–1611. [Google Scholar] [CrossRef] [PubMed]
  73. Agollah, G.D.; Gonzalez-Garay, M.L.; Rasmussen, J.C.; Tan, I.C.; Aldrich, M.B.; Darne, C.; Fife, C.E.; Guilliod, R.; Maus, E.A.; King, P.D.; et al. Evidence for SH2 domain-containing 5’-inositol phosphatase-2 (SHIP2) contributing to a lymphatic dysfunction. PLoS ONE 2014, 9, e112548. [Google Scholar] [CrossRef] [PubMed]
  74. Rodriguez-Laguna, L.; Agra, N.; Ibañez, K.; Oliva-Molina, G.; Gordo, G.; Khurana, N.; Hominick, D.; Beato, M.; Colmenero, I.; Herranz, G.; et al. Somatic activating mutations in PIK3CA cause generalized lymphatic anomaly. J. Exp. Med. 2018, 216, 407–418. [Google Scholar] [CrossRef] [PubMed]
  75. Brouillard, P.; Boon, L.; Vikkula, M. Genetics of lymphatic anomalies. J. Clin. Investig. 2014, 124, 898–904. [Google Scholar] [CrossRef] [PubMed]
  76. Osborn, A.J.; Dickie, P.; Neilson, D.E.; Glaser, K.; Lynch, K.A.; Gupta, A.; Dickie, B.H. Activating PIK3CA alleles and lymphangiogenic phenotype of lymphatic endothelial cells isolated from lymphatic malformations. Hum. Mol. Genet. 2015, 24, 926–938. [Google Scholar] [CrossRef] [PubMed]
  77. Martinez-Corral, I.; Zhang, Y.; Petkova, M.; Ortsäter, H.; Sjöberg, S.; Castillo, S.D.; Brouillard, P.; Libbrecht, L.; Saur, D.; Graupera, M.; et al. Blockade of VEGF-C signaling inhibits lymphatic malformations driven by oncogenic PIK3CA mutation. Nat. Commun. 2020, 11, 2869. [Google Scholar] [CrossRef]
  78. Zhou, X.P.; Marsh, D.J.; Hampel, H.; Mulliken, J.B.; Gimm, O.; Eng, C. Germline and germline mosaic PTEN mutations associated with a Proteus-like syndrome of hemihypertrophy, lower limb asymmetry, arteriovenous malformations and lipomatosis. Hum. Mol. Genet. 2000, 9, 765–768. [Google Scholar] [CrossRef]
  79. Lindhurst, M.J.; Sapp, J.C.; Teer, J.K.; Johnston, J.J.; Finn, E.M.; Peters, K.; Turner, J.; Cannons, J.L.; Bick, D.; Blakemore, L.; et al. A mosaic activating mutation in AKT1 associated with the Proteus syndrome. N. Engl. J. Med. 2011, 365, 611–619. [Google Scholar] [CrossRef]
  80. Sevick-Muraca, E.M.; King, P.D. Lymphatic vessel abnormalities arising from disorders of Ras signal transduction. Trends Cardiovasc. Med. 2014, 24, 121–127. [Google Scholar] [CrossRef]
  81. Nozawa, A.; Ozeki, M.; Niihori, T.; Suzui, N.; Miyazaki, T.; Aoki, Y. A somatic activating KRAS variant identified in an affected lesion of a patient with Gorham-Stout disease. J. Hum. Genet. 2020, 65, 995–1001. [Google Scholar] [CrossRef] [PubMed]
  82. Homayun-Sepehr, N.; McCarter, A.L.; Helaers, R.; Galant, C.; Boon, L.M.; Brouillard, P.; Vikkula, M.; Dellinger, M.T. KRAS-driven model of Gorham-Stout disease effectively treated with trametinib. JCI Insight 2021, 6, e149831. [Google Scholar] [CrossRef] [PubMed]
  83. Li, D.; March, M.E.; Gutierrez-Uzquiza, A.; Kao, C.; Seiler, C.; Pinto, E.; Matsuoka, L.S.; Battig, M.R.; Bhoj, E.J.; Wenger, T.L.; et al. ARAF recurrent mutation causes central conducting lymphatic anomaly treatable with a MEK inhibitor. Nat. Med. 2019, 25, 1116–1122. [Google Scholar] [CrossRef] [PubMed]
  84. Ekvall, S.; Wilbe, M.; Dahlgren, J.; Legius, E.; van Haeringen, A.; Westphal, O.; Annerén, G.; Bondeson, M.L. Mutation in NRAS in familial Noonan syndrome--case report and review of the literature. BMC Med. Genet. 2015, 16, 95. [Google Scholar] [CrossRef] [PubMed]
  85. Sarkozy, A.; Carta, C.; Moretti, S.; Zampino, G.; Digilio, M.C.; Pantaleoni, F.; Scioletti, A.P.; Esposito, G.; Cordeddu, V.; Lepri, F.; et al. Germline BRAF mutations in Noonan, LEOPARD, and cardiofaciocutaneous syndromes: Molecular diversity and associated phenotypic spectrum. Hum. Mutat. 2009, 30, 695–702. [Google Scholar] [CrossRef]
  86. Tartaglia, M.; Zampino, G.; Gelb, B.D. Noonan syndrome: Clinical aspects and molecular pathogenesis. Mol. Syndromol. 2010, 1, 2–26. [Google Scholar] [CrossRef]
  87. Kim, T.; Tafoya, E.; Chelliah, M.P.; Lekwuttikarn, R.; Li, J.; Sarin, K.Y.; Teng, J. Alterations of the MEK/ERK, BMP, and Wnt/β-catenin pathways detected in the blood of individuals with lymphatic malformations. PLoS ONE 2019, 14, e0213872. [Google Scholar] [CrossRef]
  88. Li, D.; Wenger, T.L.; Seiler, C.; March, M.E.; Gutierrez-Uzquiza, A.; Kao, C.; Bhoj, E.; Tian, L.; Rosenbach, M.; Liu, Y.; et al. Pathogenic variant in EPHB4 results in central conducting lymphatic anomaly. Hum. Mol. Genet. 2018, 27, 3233–3245. [Google Scholar] [CrossRef]
  89. Kawasaki, J.; Aegerter, S.; Fevurly, R.D.; Mammoto, A.; Mammoto, T.; Sahin, M.; Mably, J.D.; Fishman, S.J.; Chan, J. RASA1 functions in EPHB4 signaling pathway to suppress endothelial mTORC1 activity. J. Clin. Investig. 2014, 124, 2774–2784. [Google Scholar] [CrossRef]
  90. Miaskowski, C.; Dodd, M.; Paul, S.M.; West, C.; Hamolsky, D.; Abrams, G.; Cooper, B.A.; Elboim, C.; Neuhaus, J.; Schmidt, B.L.; et al. Lymphatic and angiogenic candidate genes predict the development of secondary lymphedema following breast cancer surgery. PLoS ONE 2013, 8, e60164. [Google Scholar] [CrossRef]
  91. Leung, G.; Baggott, C.; West, C.; Elboim, C.; Paul, S.M.; Cooper, B.A.; Abrams, G.; Dhruva, A.; Schmidt, B.L.; Kober, K.; et al. Cytokine candidate genes predict the development of secondary lymphedema following breast cancer surgery. Lymphat. Res. Biol. 2014, 12, 10–22. [Google Scholar] [CrossRef] [PubMed]
  92. Hespe, G.E.; Ly, C.L.; Kataru, R.P.; Mehrara, B.J. Baseline Lymphatic Dysfunction Amplifies the Negative Effects of Lymphatic Injury. Plast. Reconstr. Surg. 2019, 143, 77e–87e. [Google Scholar] [CrossRef] [PubMed]
  93. Jensen, M.R.; Simonsen, L.; Karlsmark, T.; Lanng, C.; Bülow, J. Higher vascular endothelial growth factor-C concentration in plasma is associated with increased forearm capillary filtration capacity in breast cancer-related lymphedema. Physiol. Rep. 2015, 3, e12403. [Google Scholar] [CrossRef] [PubMed]
  94. Rutkowski, J.M.; Moya, M.; Johannes, J.; Goldman, J.; Swartz, M.A. Secondary lymphedema in the mouse tail: Lymphatic hyperplasia, VEGF-C upregulation, and the protective role of MMP-9. Microvasc. Res. 2006, 72, 161–171. [Google Scholar] [CrossRef] [PubMed]
  95. Zampell, J.C.; Avraham, T.; Yoder, N.; Fort, N.; Yan, A.; Weitman, E.S.; Mehrara, B.J. Lymphatic function is regulated by a coordinated expression of lymphangiogenic and anti-lymphangiogenic cytokines. Am. J. Physiol. Cell Physiol. 2012, 302, C392–C404. [Google Scholar] [CrossRef] [PubMed]
  96. Gousopoulos, E.; Proulx, S.T.; Bachmann, S.B.; Dieterich, L.C.; Scholl, J.; Karaman, S.; Bianchi, R.; Detmar, M. An Important Role of VEGF-C in Promoting Lymphedema Development. J. Investig. Dermatol. 2017, 137, 1995–2004. [Google Scholar] [CrossRef] [PubMed]
  97. Plc, H.P. Herantis Announces Inconclusive Results from Phase II Study with Lymfactin in Breast Cancer Related Lymphedema®; Herantis Pharma Plc.: Espoo, Finland, 2021. [Google Scholar]
  98. Yusof, K.M.; Groen, K.; Rosli, R.; Avery-Kiejda, K.A. Crosstalk Between microRNAs and the Pathological Features of Secondary Lymphedema. Front. Cell Dev. Biol. 2021, 9, 732415. [Google Scholar] [CrossRef] [PubMed]
  99. Jones, D.; Li, Y.; He, Y.; Xu, Z.; Chen, H.; Min, W. Mirtron microRNA-1236 inhibits VEGFR-3 signaling during inflammatory lymphangiogenesis. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 633–642. [Google Scholar] [CrossRef]
  100. Zhou, J.; He, Z.; Guo, L.; Zeng, J.; Liang, P.; Ren, L.; Zhang, M.; Zhang, P.; Huang, X. MiR-128-3p directly targets VEGFC/VEGFR3 to modulate the proliferation of lymphatic endothelial cells through Ca2+ signaling. Int. J. Biochem. Cell Biol. 2018, 102, 51–58. [Google Scholar] [CrossRef]
  101. Yusof, K.M.; Groen, K.; Rosli, R.; Abdullah, M.; Mahmud, R.; Avery-Kiejda, K.A. Evaluation of Circulating MicroRNAs and Adipokines in Breast Cancer Survivors with Arm Lymphedema. Int. J. Mol. Sci. 2022, 23, 11359. [Google Scholar] [CrossRef]
  102. Ji, R.C. Lymphatic endothelial cells, tumor lymphangiogenesis and metastasis: New insights into intratumoral and peritumoral lymphatics. Cancer Metastasis Rev. 2006, 25, 677–694. [Google Scholar] [CrossRef] [PubMed]
  103. Franchi, A.; Gallo, O.; Massi, D.; Baroni, G.; Santucci, M. Tumor lymphangiogenesis in head and neck squamous cell carcinoma: A morphometric study with clinical correlations. Cancer 2004, 101, 973–978. [Google Scholar] [CrossRef] [PubMed]
  104. Kitadai, Y.; Kodama, M.; Cho, S.; Kuroda, T.; Ochiumi, T.; Kimura, S.; Tanaka, S.; Matsumura, S.; Yasui, W.; Chayama, K. Quantitative analysis of lymphangiogenic markers for predicting metastasis of human gastric carcinoma to lymph nodes. Int. J. Cancer 2005, 115, 388–392. [Google Scholar] [CrossRef] [PubMed]
  105. Valtola, R.; Salven, P.; Heikkilä, P.; Taipale, J.; Joensuu, H.; Rehn, M.; Pihlajaniemi, T.; Weich, H.; deWaal, R.; Alitalo, K. VEGFR-3 and its ligand VEGF-C are associated with angiogenesis in breast cancer. Am. J. Pathol. 1999, 154, 1381–1390. [Google Scholar] [CrossRef] [PubMed]
  106. Zhao, Y.C.; Ni, X.J.; Li, Y.; Dai, M.; Yuan, Z.X.; Zhu, Y.Y.; Luo, C.Y. Peritumoral lymphangiogenesis induced by vascular endothelial growth factor C and D promotes lymph node metastasis in breast cancer patients. World J. Surg. Oncol. 2012, 10, 165. [Google Scholar] [CrossRef] [PubMed]
  107. Kigure, W.; Fujii, T.; Sutoh, T.; Morita, H.; Katoh, T.; Yajima, R.N.; Yamaguchi, S.; Tsutsumi, S.; Asao, T.; Kuwano, H. The association of VEGF-C expression with tumor lymphatic vessel density and lymph node metastasis in patients with gastric cancer and gastrointestinal stromal tumor. Hepatogastroenterology 2013, 60, 277–280. [Google Scholar] [PubMed]
  108. Miyahara, M.; Tanuma, J.; Sugihara, K.; Semba, I. Tumor lymphangiogenesis correlates with lymph node metastasis and clinicopathologic parameters in oral squamous cell carcinoma. Cancer 2007, 110, 1287–1294. [Google Scholar] [CrossRef] [PubMed]
  109. He, M.; He, Q.; Cai, X.; Chen, Z.; Lao, S.; Deng, H.; Liu, X.; Zheng, Y.; Liu, X.; Liu, J.; et al. Role of lymphatic endothelial cells in the tumor microenvironment-a narrative review of recent advances. Transl. Lung Cancer Res. 2021, 10, 2252–2277. [Google Scholar] [CrossRef]
  110. Cao, R.; Ji, H.; Feng, N.; Zhang, Y.; Yang, X.; Andersson, P.; Sun, Y.; Tritsaris, K.; Hansen, A.J.; Dissing, S.; et al. Collaborative interplay between FGF-2 and VEGF-C promotes lymphangiogenesis and metastasis. Proc. Natl. Acad. Sci. USA 2012, 109, 15894–15899. [Google Scholar] [CrossRef]
  111. Holopainen, T.; Saharinen, P.; D’Amico, G.; Lampinen, A.; Eklund, L.; Sormunen, R.; Anisimov, A.; Zarkada, G.; Lohela, M.; Heloterä, H.; et al. Effects of angiopoietin-2-blocking antibody on endothelial cell-cell junctions and lung metastasis. J. Natl. Cancer Inst. 2012, 104, 461–475. [Google Scholar] [CrossRef]
  112. Jeon, B.H.; Jang, C.; Han, J.; Kataru, R.P.; Piao, L.; Jung, K.; Cha, H.J.; Schwendener, R.A.; Jang, K.Y.; Kim, K.S.; et al. Profound but dysfunctional lymphangiogenesis via vascular endothelial growth factor ligands from CD11b+ macrophages in advanced ovarian cancer. Cancer Res. 2008, 68, 1100–1109. [Google Scholar] [CrossRef] [PubMed]
  113. Volk-Draper, L.; Patel, R.; Bhattarai, N.; Yang, J.; Wilber, A.; DeNardo, D.; Ran, S. Myeloid-Derived Lymphatic Endothelial Cell Progenitors Significantly Contribute to Lymphatic Metastasis in Clinical Breast Cancer. Am. J. Pathol. 2019, 189, 2269–2292. [Google Scholar] [CrossRef] [PubMed]
  114. Elder, A.M.; Tamburini, B.A.J.; Crump, L.S.; Black, S.A.; Wessells, V.M.; Schedin, P.J.; Borges, V.F.; Lyons, T.R. Semaphorin 7A Promotes Macrophage-Mediated Lymphatic Remodeling during Postpartum Mammary Gland Involution and in Breast Cancer. Cancer Res. 2018, 78, 6473–6485. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, B.; Gao, Z.; Sun, M.; Li, H.; Fan, H.; Chen, D.; Zheng, J. Prognostic significance of VEGF-C, semaphorin 3F, and neuropilin-2 expression in oral squamous cell carcinomas and their relationship with lymphangiogenesis. J. Surg. Oncol. 2015, 111, 382–388. [Google Scholar] [CrossRef] [PubMed]
  116. Xie, Z.; Li, T.; Huang, B.; Liu, S.; Zhang, L.; Zhang, Q. Semaphorin 3F Serves as a Tumor Suppressor in Esophageal Squamous Cell Carcinoma and is Associated With Lymph Node Metastasis in Disease Progression. Technol. Cancer Res. Treat. 2020, 19, 1533033820928117. [Google Scholar] [CrossRef] [PubMed]
  117. Ernst, B.P.; Mikstas, C.; Stöver, T.; Stauber, R.; Strieth, S. Association of eIF4E and SPARC Expression with Lymphangiogenesis and Lymph Node Metastasis in Hypopharyngeal Cancer. Anticancer Res. 2018, 38, 699–706. [Google Scholar] [CrossRef]
  118. Lin, Y.C.; Ohbayashi, N.; Hongu, T.; Katagiri, N.; Funakoshi, Y.; Lee, H.; Kanaho, Y. Arf6 in lymphatic endothelial cells regulates lymphangiogenesis by controlling directional cell migration. Sci. Rep. 2017, 7, 11431. [Google Scholar] [CrossRef]
  119. Karnezis, T.; Farnsworth, R.H.; Harris, N.C.; Williams, S.P.; Caesar, C.; Byrne, D.J.; Herle, P.; Macheda, M.L.; Shayan, R.; Zhang, Y.F.; et al. CCL27/CCL28-CCR10 Chemokine Signaling Mediates Migration of Lymphatic Endothelial Cells. Cancer Res. 2019, 79, 1558–1572. [Google Scholar] [CrossRef]
  120. Zhuo, W.; Jia, L.; Song, N.; Lu, X.A.; Ding, Y.; Wang, X.; Song, X.; Fu, Y.; Luo, Y. The CXCL12-CXCR4 chemokine pathway: A novel axis regulates lymphangiogenesis. Clin. Cancer Res. 2012, 18, 5387–5398. [Google Scholar] [CrossRef]
  121. Chen, J.Y.; Lai, Y.S.; Chu, P.Y.; Chan, S.H.; Wang, L.H.; Hung, W.C. Cancer-Derived VEGF-C Increases Chemokine Production in Lymphatic Endothelial Cells to Promote CXCR2-Dependent Cancer Invasion and MDSC Recruitment. Cancers 2019, 11, 1120. [Google Scholar] [CrossRef]
  122. Tutunea-Fatan, E.; Majumder, M.; Xin, X.; Lala, P.K. The role of CCL21/CCR7 chemokine axis in breast cancer-induced lymphangiogenesis. Mol. Cancer 2015, 14, 35. [Google Scholar] [CrossRef] [PubMed]
  123. Nykänen, A.I.; Sandelin, H.; Krebs, R.; Keränen, M.A.I.; Tuuminen, R.; Kärpänen, T.; Wu, Y.; Pytowski, B.; Koskinen, P.K.; Ylä-Herttuala, S.; et al. Targeting Lymphatic Vessel Activation and CCL21 Production by Vascular Endothelial Growth Factor Receptor-3 Inhibition Has Novel Immunomodulatory and Antiarteriosclerotic Effects in Cardiac Allografts. Circulation 2010, 121, 1413–1422. [Google Scholar] [CrossRef] [PubMed]
  124. Van de Velde, M.; Ebroin, M.; Durré, T.; Joiret, M.; Gillot, L.; Blacher, S.; Geris, L.; Kridelka, F.; Noel, A. Tumor exposed-lymphatic endothelial cells promote primary tumor growth via IL6. Cancer Lett. 2021, 497, 154–164. [Google Scholar] [CrossRef] [PubMed]
  125. Nandi, P.; Girish, G.V.; Majumder, M.; Xin, X.; Tutunea-Fatan, E.; Lala, P.K. PGE2 promotes breast cancer-associated lymphangiogenesis by activation of EP4 receptor on lymphatic endothelial cells. BMC Cancer 2017, 17, 11. [Google Scholar] [CrossRef] [PubMed]
  126. Lala, P.K.; Nandi, P.; Majumder, M. Roles of prostaglandins in tumor-associated lymphangiogenesis with special reference to breast cancer. Cancer Metastasis Rev. 2018, 37, 369–384. [Google Scholar] [CrossRef] [PubMed]
  127. Kimura, T.; Sugaya, M.; Oka, T.; Blauvelt, A.; Okochi, H.; Sato, S. Lymphatic dysfunction attenuates tumor immunity through impaired antigen presentation. Oncotarget 2015, 6, 18081–18093. [Google Scholar] [CrossRef] [PubMed]
  128. Lund, A.W.; Wagner, M.; Fankhauser, M.; Steinskog, E.S.; Broggi, M.A.; Spranger, S.; Gajewski, T.F.; Alitalo, K.; Eikesdal, H.P.; Wiig, H.; et al. Lymphatic vessels regulate immune microenvironments in human and murine melanoma. J. Clin. Investig. 2016, 126, 3389–3402. [Google Scholar] [CrossRef] [PubMed]
  129. Kataru, R.P.; Ly, C.L.; Shin, J.; Park, H.J.; Baik, J.E.; Rehal, S.; Ortega, S.; Lyden, D.; Mehrara, B.J. Tumor Lymphatic Function Regulates Tumor Inflammatory and Immunosuppressive Microenvironments. Cancer Immunol. Res. 2019, 7, 1345–1358. [Google Scholar] [CrossRef]
  130. Song, E.; Mao, T.; Dong, H.; Boisserand, L.S.B.; Antila, S.; Bosenberg, M.; Alitalo, K.; Thomas, J.L.; Iwasaki, A. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature 2020, 577, 689–694. [Google Scholar] [CrossRef]
  131. Hu, X.; Deng, Q.; Ma, L.; Li, Q.; Chen, Y.; Liao, Y.; Zhou, F.; Zhang, C.; Shao, L.; Feng, J.; et al. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res. 2020, 30, 229–243. [Google Scholar] [CrossRef]
  132. Zhou, C.; Ma, L.; Xu, H.; Huo, Y.; Luo, J. Meningeal lymphatics regulate radiotherapy efficacy through modulating anti-tumor immunity. Cell Res. 2022, 32, 543–554. [Google Scholar] [CrossRef] [PubMed]
  133. Sasso, M.S.; Mitrousis, N.; Wang, Y.; Briquez, P.S.; Hauert, S.; Ishihara, J.; Hubbell, J.A.; Swartz, M.A. Lymphangiogenesis-inducing vaccines elicit potent and long-lasting T cell immunity against melanomas. Sci. Adv. 2021, 7, eabe4362. [Google Scholar] [CrossRef] [PubMed]
  134. Bordry, N.; Broggi, M.A.S.; de Jonge, K.; Schaeuble, K.; Gannon, P.O.; Foukas, P.G.; Danenberg, E.; Romano, E.; Baumgaertner, P.; Fankhauser, M.; et al. Lymphatic vessel density is associated with CD8(+) T cell infiltration and immunosuppressive factors in human melanoma. Oncoimmunology 2018, 7, e1462878. [Google Scholar] [CrossRef] [PubMed]
  135. Lund, A.W.; Duraes, F.V.; Hirosue, S.; Raghavan, V.R.; Nembrini, C.; Thomas, S.N.; Issa, A.; Hugues, S.; Swartz, M.A. VEGF-C promotes immune tolerance in B16 melanomas and cross-presentation of tumor antigen by lymph node lymphatics. Cell Rep. 2012, 1, 191–199. [Google Scholar] [CrossRef]
  136. Kataru, R.P.; Park, H.J.; Baik, J.E.; Li, C.; Shin, J.; Mehrara, B.J. Regulation of Lymphatic Function in Obesity. Front. Physiol. 2020, 11, 459. [Google Scholar] [CrossRef]
  137. Escobedo, N.; Oliver, G. The Lymphatic Vasculature: Its Role in Adipose Metabolism and Obesity. Cell Metab. 2017, 26, 598–609. [Google Scholar] [CrossRef]
  138. Norden, P.R.; Kume, T. The Role of Lymphatic Vascular Function in Metabolic Disorders. Front. Physiol. 2020, 11, 404. [Google Scholar] [CrossRef]
  139. Nitti, M.D.; Hespe, G.E.; Kataru, R.P.; García Nores, G.D.; Savetsky, I.L.; Torrisi, J.S.; Gardenier, J.C.; Dannenberg, A.J.; Mehrara, B.J. Obesity-induced lymphatic dysfunction is reversible with weight loss. J. Physiol. 2016, 594, 7073–7087. [Google Scholar] [CrossRef]
  140. Mehrara, B.J.; Greene, A.K. Lymphedema and obesity: Is there a link? Plast. Reconstr. Surg. 2014, 134, 154e–160e. [Google Scholar] [CrossRef]
  141. Zafar, M.I.; Mills, K.; Ye, X.; Blakely, B.; Min, J.; Kong, W.; Zhang, N.; Gou, L.; Regmi, A.; Hu, S.Q.; et al. Association between the expression of vascular endothelial growth factors and metabolic syndrome or its components: A systematic review and meta-analysis. Diabetol. Metab. Syndr. 2018, 10, 62. [Google Scholar] [CrossRef]
  142. Gómez-Ambrosi, J.; Catalán, V.; Rodríguez, A.; Ramírez, B.; Silva, C.; Gil, M.J.; Salvador, J.; Frühbeck, G. Involvement of serum vascular endothelial growth factor family members in the development of obesity in mice and humans. J. Nutr. Biochem. 2010, 21, 774–780. [Google Scholar] [CrossRef]
  143. Wada, H.; Ura, S.; Kitaoka, S.; Satoh-Asahara, N.; Horie, T.; Ono, K.; Takaya, T.; Takanabe-Mori, R.; Akao, M.; Abe, M.; et al. Distinct characteristics of circulating vascular endothelial growth factor-a and C levels in human subjects. PLoS ONE 2011, 6, e29351. [Google Scholar] [CrossRef]
  144. Rockson, S.G.; Zhou, X.; Zhao, L.; Hosseini, D.K.; Jiang, X.; Sweatt, A.J.; Kim, D.; Tian, W.; Snyder, M.P.; Nicolls, M.R. Exploring disease interrelationships in patients with lymphatic disorders: A single center retrospective experience. Clin. Transl. Med. 2022, 12, e760. [Google Scholar] [CrossRef]
  145. Harvey, N.L.; Srinivasan, R.S.; Dillard, M.E.; Johnson, N.C.; Witte, M.H.; Boyd, K.; Sleeman, M.W.; Oliver, G. Lymphatic vascular defects promoted by Prox1 haploinsufficiency cause adult-onset obesity. Nat. Genet. 2005, 37, 1072–1081. [Google Scholar] [CrossRef]
  146. Rutkowski, J.M.; Markhus, C.E.; Gyenge, C.C.; Alitalo, K.; Wiig, H.; Swartz, M.A. Dermal Collagen and Lipid Deposition Correlate with Tissue Swelling and Hydraulic Conductivity in Murine Primary Lymphedema. Am. J. Pathol. 2010, 176, 1122–1129. [Google Scholar] [CrossRef]
  147. Karkkainen, M.J.; Saaristo, A.; Jussila, L.; Karila, K.A.; Lawrence, E.C.; Pajusola, K.; Bueler, H.; Eichmann, A.; Kauppinen, R.; Kettunen, M.I.; et al. A model for gene therapy of human hereditary lymphedema. Proc. Natl. Acad Sci. USA 2001, 98, 12677–12682. [Google Scholar] [CrossRef]
  148. Escobedo, N.; Proulx, S.T.; Karaman, S.; Dillard, M.E.; Johnson, N.; Detmar, M.; Oliver, G. Restoration of lymphatic function rescues obesity in Prox1-haploinsufficient mice. JCI Insight 2016, 1, e85096. [Google Scholar] [CrossRef]
  149. Aschen, S.; Zampell, J.C.; Elhadad, S.; Weitman, E.; De Brot Andrade, M.; Mehrara, B.J. Regulation of adipogenesis by lymphatic fluid stasis: Part II. Expression of adipose differentiation genes. Plast. Reconstr. Surg. 2012, 129, 838–847. [Google Scholar] [CrossRef] [PubMed]
  150. Chakraborty, A.; Barajas, S.; Lammoglia, G.M.; Reyna, A.J.; Morley, T.S.; Johnson, J.A.; Scherer, P.E.; Rutkowski, J.M. Vascular Endothelial Growth Factor-D (VEGF-D) Overexpression and Lymphatic Expansion in Murine Adipose Tissue Improves Metabolism in Obesity. Am. J. Pathol. 2019, 189, 924–939. [Google Scholar] [CrossRef] [PubMed]
  151. Nurmi, H.; Saharinen, P.; Zarkada, G.; Zheng, W.; Robciuc, M.R.; Alitalo, K. VEGF-C is required for intestinal lymphatic vessel maintenance and lipid absorption. EMBO Mol. Med. 2015, 7, 1418–1425. [Google Scholar] [CrossRef] [PubMed]
  152. Martel, C.; Li, W.; Fulp, B.; Platt, A.M.; Gautier, E.L.; Westerterp, M.; Bittman, R.; Tall, A.R.; Chen, S.H.; Thomas, M.J.; et al. Lymphatic vasculature mediates macrophage reverse cholesterol transport in mice. J. Clin. Investig. 2013, 123, 1571–1579. [Google Scholar] [CrossRef] [PubMed]
  153. Milasan, A.; Smaani, A.; Martel, C. Early rescue of lymphatic function limits atherosclerosis progression in Ldlr(-/-) mice. Atherosclerosis 2019, 283, 106–119. [Google Scholar] [CrossRef] [PubMed]
  154. Cifarelli, V.; Appak-Baskoy, S.; Peche, V.S.; Kluzak, A.; Shew, T.; Narendran, R.; Pietka, K.M.; Cella, M.; Walls, C.W.; Czepielewski, R.; et al. Visceral obesity and insulin resistance associate with CD36 deletion in lymphatic endothelial cells. Nat. Commun. 2021, 12, 3350. [Google Scholar] [CrossRef] [PubMed]
  155. Savetsky, I.L.; Torrisi, J.S.; Cuzzone, D.A.; Ghanta, S.; Albano, N.J.; Gardenier, J.C.; Joseph, W.J.; Mehrara, B.J. Obesity increases inflammation and impairs lymphatic function in a mouse model of lymphedema. Am. J. Physiol. Heart Circ. Physiol. 2014, 307, H165–H172. [Google Scholar] [CrossRef]
  156. Rehal, S.; Kataru, R.P.; Hespe, G.E.; Baik, J.E.; Park, H.J.; Ly, C.; Shin, J.; Mehrara, B.J. Regulation of lymphatic function and injury by nitrosative stress in obese mice. Mol. Metab. 2020, 42, 101081. [Google Scholar] [CrossRef]
  157. Cao, E.; Watt, M.J.; Nowell, C.J.; Quach, T.; Simpson, J.S.; De Melo Ferreira, V.; Agarwal, S.; Chu, H.; Srivastava, A.; Anderson, D.; et al. Mesenteric lymphatic dysfunction promotes insulin resistance and represents a potential treatment target in obesity. Nat. Metab. 2021, 3, 1175–1188. [Google Scholar] [CrossRef]
  158. Karaman, S.; Hollmén, M.; Yoon, S.Y.; Alkan, H.F.; Alitalo, K.; Wolfrum, C.; Detmar, M. Transgenic overexpression of VEGF-C induces weight gain and insulin resistance in mice. Sci. Rep. 2016, 6, 31566. [Google Scholar] [CrossRef]
  159. García Nores, G.D.; Cuzzone, D.A.; Albano, N.J.; Hespe, G.E.; Kataru, R.P.; Torrisi, J.S.; Gardenier, J.C.; Savetsky, I.L.; Aschen, S.Z.; Nitti, M.D.; et al. Obesity but not high-fat diet impairs lymphatic function. Int. J. Obes. 2016, 40, 1582–1590. [Google Scholar] [CrossRef]
  160. Montgomery, M.K.; Hallahan, N.L.; Brown, S.H.; Liu, M.; Mitchell, T.W.; Cooney, G.J.; Turner, N. Mouse strain-dependent variation in obesity and glucose homeostasis in response to high-fat feeding. Diabetologia 2013, 56, 1129–1139. [Google Scholar] [CrossRef]
  161. McPherron, A.C.; Lee, S.J. Suppression of body fat accumulation in myostatin-deficient mice. J. Clin. Investig. 2002, 109, 595–601. [Google Scholar] [CrossRef]
  162. Hou, Y.; Bock, F.; Hos, D.; Cursiefen, C. Lymphatic Trafficking in the Eye: Modulation of Lymphatic Trafficking to Promote Corneal Transplant Survival. Cells 2021, 10, 1661. [Google Scholar] [CrossRef]
  163. Inomata, T.; Mashaghi, A.; Di Zazzo, A.; Lee, S.M.; Chiang, H.; Dana, R. Kinetics of Angiogenic Responses in Corneal Transplantation. Cornea 2017, 36, 491–496. [Google Scholar] [CrossRef]
  164. Zhang, H.; Grimaldo, S.; Yuen, D.; Chen, L. Combined blockade of VEGFR-3 and VLA-1 markedly promotes high-risk corneal transplant survival. Investig. Ophthalmol. Vis. Sci. 2011, 52, 6529–6535. [Google Scholar] [CrossRef]
  165. Hou, Y.; Le, V.N.H.; Tóth, G.; Siebelmann, S.; Horstmann, J.; Gabriel, T.; Bock, F.; Cursiefen, C. UV light crosslinking regresses mature corneal blood and lymphatic vessels and promotes subsequent high-risk corneal transplant survival. Am. J. Transpl. 2018, 18, 2873–2884. [Google Scholar] [CrossRef]
  166. Hos, D.; Bock, F.; Dietrich, T.; Onderka, J.; Kruse, F.E.; Thierauch, K.H.; Cursiefen, C. Inflammatory corneal (lymph)angiogenesis is blocked by VEGFR-tyrosine kinase inhibitor ZK 261991, resulting in improved graft survival after corneal transplantation. Investig. Ophthalmol. Vis. Sci. 2008, 49, 1836–1842. [Google Scholar] [CrossRef]
  167. Dietrich, T.; Bock, F.; Yuen, D.; Hos, D.; Bachmann, B.O.; Zahn, G.; Wiegand, S.; Chen, L.; Cursiefen, C. Cutting Edge: Lymphatic Vessels, Not Blood Vessels, Primarily Mediate Immune Rejections After Transplantation. J. Immunol. 2010, 184, 535. [Google Scholar] [CrossRef]
  168. Chen, W.S.; Cao, Z.; Sugaya, S.; Lopez, M.J.; Sendra, V.G.; Laver, N.; Leffler, H.; Nilsson, U.J.; Fu, J.; Song, J.; et al. Pathological lymphangiogenesis is modulated by galectin-8-dependent crosstalk between podoplanin and integrin-associated VEGFR-3. Nat. Commun. 2016, 7, 11302. [Google Scholar] [CrossRef]
  169. Lin, J.; Chen, Y.; Zhu, H.; Cheng, K.; Wang, H.; Yu, X.; Tang, M.; Chen, J. Lymphatic Reconstruction in Kidney Allograft Aggravates Chronic Rejection by Promoting Alloantigen Presentation. Front. Immunol. 2021, 12, 796260. [Google Scholar] [CrossRef] [PubMed]
  170. Palin, N.K.; Savikko, J.; Koskinen, P.K. Sirolimus inhibits lymphangiogenesis in rat renal allografts, a novel mechanism to prevent chronic kidney allograft injury. Transpl. Int. 2013, 26, 195–205. [Google Scholar] [CrossRef] [PubMed]
  171. Dashkevich, A.; Raissadati, A.; Syrjälä, S.O.; Zarkada, G.; Keränen, M.A.; Tuuminen, R.; Krebs, R.; Anisimov, A.; Jeltsch, M.; Leppänen, V.M.; et al. Ischemia-Reperfusion Injury Enhances Lymphatic Endothelial VEGFR3 and Rejection in Cardiac Allografts. Am. J. Transpl. 2016, 16, 1160–1172. [Google Scholar] [CrossRef] [PubMed]
  172. Edwards, L.A.; Nowocin, A.K.; Jafari, N.V.; Meader, L.L.; Brown, K.; Sarde, A.; Lam, C.; Murray, A.; Wong, W. Chronic Rejection of Cardiac Allografts Is Associated With Increased Lymphatic Flow and Cellular Trafficking. Circulation 2018, 137, 488–503. [Google Scholar] [CrossRef] [PubMed]
  173. Iwami, D.; Brinkman, C.C.; Bromberg, J.S. Vascular endothelial growth factor c/vascular endothelial growth factor receptor 3 signaling regulates chemokine gradients and lymphocyte migration from tissues to lymphatics. Transplantation 2015, 99, 668–677. [Google Scholar] [CrossRef] [PubMed]
  174. Cui, Y.; Liu, K.; Monzon-Medina, M.E.; Padera, R.F.; Wang, H.; George, G.; Toprak, D.; Abdelnour, E.; D’Agostino, E.; Goldberg, H.J.; et al. Therapeutic lymphangiogenesis ameliorates established acute lung allograft rejection. J. Clin. Investig. 2015, 125, 4255–4268. [Google Scholar] [CrossRef] [PubMed]
  175. Gao, C.; Zhu, J.; Qin, Q.; Yang, X.; Jiang, Y.; Zhang, J. The Relationship between VEGFC Gene Polymorphisms and Autoimmune Thyroiditis. Biomed. Res. Int. 2022, 2022, 2603519. [Google Scholar] [CrossRef] [PubMed]
  176. Alunno, A.; Ibba-Manneschi, L.; Bistoni, O.; Rosa, I.; Caterbi, S.; Gerli, R.; Manetti, M. Mobilization of lymphatic endothelial precursor cells and lymphatic neovascularization in primary Sjögren’s syndrome. J. Cell. Mol. Med. 2016, 20, 613–622. [Google Scholar] [CrossRef] [PubMed]
  177. Sertoglu, E.; Yücel, Ç.; Omma, A.; Hayran, Y.; Colak, S.; Sandıkçı, S.C.; Durukan, A.H.; Ozgurtas, T. Determination of serum vascular endothelial growth factor (VEGF) and VEGF receptor levels with VEGF gene polymorphisms in patients with Behçet’s uveitis. Adv. Clin. Exp. Med. 2022, 31, 231–240. [Google Scholar] [CrossRef] [PubMed]
  178. William, G.A.; Mir, H.; Madhavi Latha, S.C.; Ethan, S.S.; JiHyun, S.; Jinyeon, S.; Noa, S.; William, D.S., III; Dragos, D.; Camila, B.C.; et al. Lymphatic dysfunction in lupus contributes to cutaneous photosensitivity and lymph node B cell responses. bioRxiv, 2022; bioRxiv:2022.06.13.495930. [Google Scholar] [CrossRef]
  179. Honda, N.; Jinnin, M.; Kajihara, I.; Makino, T.; Fukushima, S.; Ihn, H. Impaired lymphangiogenesis due to excess vascular endothelial growth factor-D/Flt-4 signalling in the skin of patients with systemic sclerosis. Br. J. Dermatol. 2010, 163, 776–780. [Google Scholar] [CrossRef]
  180. Akhmetshina, A.; Beer, J.; Zwerina, K.; Englbrecht, M.; Palumbo, K.; Dees, C.; Reich, N.; Zwerina, J.; Szucs, G.; Gusinde, J.; et al. Decreased lymphatic vessel counts in patients with systemic sclerosis: Association with fingertip ulcers. Arthritis Rheum. 2010, 62, 1513–1522. [Google Scholar] [CrossRef]
  181. Manetti, M.; Romano, E.; Rosa, I.; Fioretto, B.S.; Guiducci, S.; Bellando-Randone, S.; Pigatto, E.; Cozzi, F.; Ibba-Manneschi, L.; Matucci-Cerinic, M. Systemic Sclerosis Serum Significantly Impairs the Multi-Step Lymphangiogenic Process: In Vitro Evidence. Int. J. Mol. Sci. 2019, 20, 6189. [Google Scholar] [CrossRef]
  182. Kunstfeld, R.; Hirakawa, S.; Hong, Y.K.; Schacht, V.; Lange-Asschenfeldt, B.; Velasco, P.; Lin, C.; Fiebiger, E.; Wei, X.; Wu, Y.; et al. Induction of cutaneous delayed-type hypersensitivity reactions in VEGF-A transgenic mice results in chronic skin inflammation associated with persistent lymphatic hyperplasia. Blood 2004, 104, 1048–1057. [Google Scholar] [CrossRef]
  183. Sundberg, J.P.; Pratt, C.H.; Silva, K.A.; Kennedy, V.E.; Stearns, T.M.; Sundberg, B.A.; King, L.E.; HogenEsch, H. Dermal lymphatic dilation in a mouse model of alopecia areata. Exp. Mol. Pathol. 2016, 100, 332–336. [Google Scholar] [CrossRef] [PubMed]
  184. Yoon, S.Y.; Dieterich, L.C.; Karaman, S.; Proulx, S.T.; Bachmann, S.B.; Sciaroni, C.; Detmar, M. An important role of cutaneous lymphatic vessels in coordinating and promoting anagen hair follicle growth. PLoS ONE 2019, 14, e0220341. [Google Scholar] [CrossRef] [PubMed]
  185. Yoon, S.Y.; Detmar, M. Sostdc1 Secreted from Cutaneous Lymphatic Vessels Acts as a Paracrine Factor for Hair Follicle Growth. Curr. Issues Mol. Biol. 2022, 44, 2167–2174. [Google Scholar] [CrossRef] [PubMed]
  186. Gur-Cohen, S.; Yang, H.; Baksh, S.C.; Miao, Y.; Levorse, J.; Kataru, R.P.; Liu, X.; de la Cruz-Racelis, J.; Mehrara, B.J.; Fuchs, E. Stem cell-driven lymphatic remodeling coordinates tissue regeneration. Science 2019, 366, 1218–1225. [Google Scholar] [CrossRef] [PubMed]
  187. Dai, C.; Kuo, S.J.; Hu, S.L.; Tsai, C.H.; Huang, Y.L.; Huang, C.C.; Wang, L.; Xu, G.; Su, C.M.; Tang, C.H. VEGF-C Gene Polymorphisms Increase Susceptibility to Rheumatoid Arthritis. Int. J. Med. Sci. 2019, 16, 1397–1403. [Google Scholar] [CrossRef] [PubMed]
  188. Cha, H.-S.; Bae, E.-K.; Koh, J.-H.; Chai, J.-Y.; Jeon, C.H.; Ahn, K.-S.; Kim, J.; Koh, E.-M. Tumor necrosis factor-alpha induces vascular endothelial growth factor-C expression in rheumatoid synoviocytes. J. Rheumatol. 2007, 34, 16–19. [Google Scholar] [PubMed]
  189. Bell, R.D.; Rahimi, H.; Kenney, H.M.; Lieberman, A.A.; Wood, R.W.; Schwarz, E.M.; Ritchlin, C.T. Altered Lymphatic Vessel Anatomy and Markedly Diminished Lymph Clearance in Affected Hands of Patients With Active Rheumatoid Arthritis. Arthritis Rheumatol. 2020, 72, 1447–1455. [Google Scholar] [CrossRef]
  190. Shi, J.X.; Liang, Q.Q.; Wang, Y.J.; Mooney, R.A.; Boyce, B.F.; Xing, L. Use of a whole-slide imaging system to assess the presence and alteration of lymphatic vessels in joint sections of arthritic mice. Biotech. Histochem. 2013, 88, 428–439. [Google Scholar] [CrossRef]
  191. Guo, R.; Zhou, Q.; Proulx, S.T.; Wood, R.; Ji, R.C.; Ritchlin, C.T.; Pytowski, B.; Zhu, Z.; Wang, Y.J.; Schwarz, E.M.; et al. Inhibition of lymphangiogenesis and lymphatic drainage via vascular endothelial growth factor receptor 3 blockade increases the severity of inflammation in a mouse model of chronic inflammatory arthritis. Arthritis Rheum. 2009, 60, 2666–2676. [Google Scholar] [CrossRef]
  192. Zhou, Q.; Guo, R.; Wood, R.; Boyce, B.F.; Liang, Q.; Wang, Y.J.; Schwarz, E.M.; Xing, L. Vascular endothelial growth factor C attenuates joint damage in chronic inflammatory arthritis by accelerating local lymphatic drainage in mice. Arthritis Rheum. 2011, 63, 2318–2328. [Google Scholar] [CrossRef]
  193. Chen, X.; Hu, Q.Y.; Wang, M.; Jia, J.; Teng, J.; Sun, Y.; Cheng, X.; Ye, J.; Su, Y.; Shi, H.; et al. Serum VEGF-C as an evaluation marker of disease activity in adult-onset Still’s disease. Rheumatol. Int. 2022, 42, 149–157. [Google Scholar] [CrossRef] [PubMed]
  194. Sato, H.; Higashiyama, M.; Hozumi, H.; Sato, S.; Furuhashi, H.; Takajo, T.; Maruta, K.; Yasutake, Y.; Narimatsu, K.; Yoshikawa, K.; et al. Platelet interaction with lymphatics aggravates intestinal inflammation by suppressing lymphangiogenesis. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 311, G276–G285. [Google Scholar] [CrossRef] [PubMed]
  195. Rahier, J.F.; De Beauce, S.; Dubuquoy, L.; Erdual, E.; Colombel, J.F.; Jouret-Mourin, A.; Geboes, K.; Desreumaux, P. Increased lymphatic vessel density and lymphangiogenesis in inflammatory bowel disease. Aliment Pharmacol. Ther. 2011, 34, 533–543. [Google Scholar] [CrossRef] [PubMed]
  196. D’Alessio, S.; Correale, C.; Tacconi, C.; Gandelli, A.; Pietrogrande, G.; Vetrano, S.; Genua, M.; Arena, V.; Spinelli, A.; Peyrin-Biroulet, L.; et al. VEGF-C-dependent stimulation of lymphatic function ameliorates experimental inflammatory bowel disease. J. Clin. Investig. 2014, 124, 3863–3878. [Google Scholar] [CrossRef] [PubMed]
  197. Ocansey, D.K.W.; Pei, B.; Xu, X.; Zhang, L.; Olovo, C.V.; Mao, F. Cellular and molecular mediators of lymphangiogenesis in inflammatory bowel disease. J. Transl. Med. 2021, 19, 254. [Google Scholar] [CrossRef] [PubMed]
  198. Jurisic, G.; Sundberg, J.P.; Detmar, M. Blockade of VEGF receptor-3 aggravates inflammatory bowel disease and lymphatic vessel enlargement. Inflamm. Bowel Dis. 2013, 19, 1983–1989. [Google Scholar] [CrossRef] [PubMed]
  199. Wang, X.L.; Zhao, J.; Qin, L.; Qiao, M. Promoting inflammatory lymphangiogenesis by vascular endothelial growth factor-C (VEGF-C) aggravated intestinal inflammation in mice with experimental acute colitis. Braz. J. Med. Biol. Res. 2016, 49, e4738. [Google Scholar] [CrossRef]
  200. Becker, F.; Yi, P.; Al-Kofahi, M.; Ganta, V.C.; Morris, J.; Alexander, J.S. Lymphatic dysregulation in intestinal inflammation: New insights into inflammatory bowel disease pathomechanisms. Lymphology 2014, 47, 3–27. [Google Scholar]
  201. Boucher, J.; Kleinridders, A.; Kahn, C.R. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb. Perspect. Biol. 2014, 6, a009191. [Google Scholar] [CrossRef]
  202. Ghalamkarpour, A.; Morlot, S.; Raas-Rothschild, A.; Utkus, A.; Mulliken, J.B.; Boon, L.M.; Vikkula, M. Hereditary lymphedema type I associated with VEGFR3 mutation: The first de novo case and atypical presentations. Clin. Genet. 2006, 70, 330–335. [Google Scholar] [CrossRef]
  203. Keppler-Noreuil, K.M.; Rios, J.J.; Parker, V.E.; Semple, R.K.; Lindhurst, M.J.; Sapp, J.C.; Alomari, A.; Ezaki, M.; Dobyns, W.; Biesecker, L.G. PIK3CA-related overgrowth spectrum (PROS): Diagnostic and testing eligibility criteria, differential diagnosis, and evaluation. Am. J. Med. Genet. A 2015, 167, 287–295. [Google Scholar] [CrossRef] [PubMed]
  204. Liu, M.; Smith, C.L.; Biko, D.M.; Li, D.; Pinto, E.; O’Connor, N.; Skraban, C.; Zackai, E.H.; Hakonarson, H.; Dori, Y.; et al. Genetics etiologies and genotype phenotype correlations in a cohort of individuals with central conducting lymphatic anomaly. Eur. J. Hum. Genet. 2022, 30, 1022–1028. [Google Scholar] [CrossRef] [PubMed]
  205. Roy, S.; Kumaravel, S.; Banerjee, P.; White, T.K.; O’Brien, A.; Seelig, C.; Chauhan, R.; Ekser, B.; Bayless, K.J.; Alpini, G.; et al. Tumor Lymphatic Interactions Induce CXCR2-CXCL5 Axis and Alter Cellular Metabolism and Lymphangiogenic Pathways to Promote Cholangiocarcinoma. Cells 2021, 10, 3093. [Google Scholar] [CrossRef] [PubMed]
  206. Di Tommaso, L.; Battista, S.; Destro, A.; Sciarra, A.; Morenghi, E.; Roncalli, M. Cracking spaces in Hashimoto Thyroiditis are lymphatic and prelymphatic vessels: A gift of immunohistochemistry for the centenary of Hashimoto’s description. Am. J. Surg. Pathol. 2010, 34, 1857–1861. [Google Scholar] [CrossRef]
  207. Choi, I.; Lee, S.; Kyoung Chung, H.; Suk Lee, Y.; Eui Kim, K.; Choi, D.; Park, E.K.; Yang, D.; Ecoiffier, T.; Monahan, J.; et al. 9-cis retinoic acid promotes lymphangiogenesis and enhances lymphatic vessel regeneration: Therapeutic implications of 9-cis retinoic acid for secondary lymphedema. Circulation 2012, 125, 872–882. [Google Scholar] [CrossRef]
Figure 1. Intracellular VEGFR3 signaling in LECS in response to the VEGF-C ligand. VEGF-C-mediated activation of lymphangiogenic VEGFR3 signaling in lymphatic endothelial cells (LECs) is regulated by numerous co-receptors, adapter proteins, and secondary messengers. The PI3K-Akt and Raf-ERK pathways serve as the principal downstream effectors of activated membrane VEGFR3. Downstream Erk phosphorylation leads to the activation of nuclear transcription factors CREB and ETS to promote cellular differentiation and proliferation activities. Downstream activation of the PI3K-Akt pathways promotes the activation of ribosomal regulatory proteins elF4E and p70S6K to promote protein synthesis and cell survival functions.
Figure 1. Intracellular VEGFR3 signaling in LECS in response to the VEGF-C ligand. VEGF-C-mediated activation of lymphangiogenic VEGFR3 signaling in lymphatic endothelial cells (LECs) is regulated by numerous co-receptors, adapter proteins, and secondary messengers. The PI3K-Akt and Raf-ERK pathways serve as the principal downstream effectors of activated membrane VEGFR3. Downstream Erk phosphorylation leads to the activation of nuclear transcription factors CREB and ETS to promote cellular differentiation and proliferation activities. Downstream activation of the PI3K-Akt pathways promotes the activation of ribosomal regulatory proteins elF4E and p70S6K to promote protein synthesis and cell survival functions.
Cells 13 00068 g001
Figure 2. Dysregulated VEGFR3 signaling in disease states. (a) VEGFR3 dysregulation in lymphedema. Primary LE: defective activation of VEGFR3 is associated with Milroy’s disease. VEGFR3 and neuropilin-2 transcription is elevated in lymphovenous overgrowth malformations. Missense mutations affecting SHIP-2 protein enhance VEGF-C-induced activation of Akt and ERK in primary lymphedema. Somatic activating PIK3CA mutations are seen in generalized lymphatic anomaly (GLA) and congenital lipomatous overgrowth, vascular malformations, epidermal nevi, and spinal/skeletal anomalies (CLOVES) syndrome. Secondary LE: VEGF-C expression is increased in the plasma of patients with LE. Expression of micro-RNA-1236 in LECs inhibits lymphangiogenesis by downregulating VEGFR3-mediated Akt and ERK activation. (b) VEGFR3 dysregulation and tumor growth and metastases. Tumor-derived VEGF-C is a potent chemoattractant and enhances peritumoral lymphatic access for tumor metastasis. Downregulation of SEMA3F, a NRP2 ligand that negatively regulates VEGFR3 activation, is downregulated in esophageal SCC. Enhanced integrin β1 signaling increases peritumoral metastasis in melanoma. Tumor-derived VEGF C-D expression activates intracellular Akt and ERK signaling to promote peritumoral lymphangiogenesis in breast tumors. (c) VEGFR3 dysregulation in obesity and metabolic syndrome. Increased serum levels of VEGF-C are seen with metabolic disorders. Haploinsufficiency of PROX-1 is associated with excessive lymph accumulation and increased insulin and leptin levels in affected tissues. (d)VEGFR-3 dysregulation in transplant rejection. Inflammatory lymphangiogenesis and corneal allograft rejection is promoted by galectin-8 mediated podoplanin-VEGFR3 cross-talk. VEGF-C-D elevation promotes increased renal lymphangiogenic activity and subsequent renal transplant rejection. (e) VEGFR3 dysregulation in autoimmune/autoinflammatory disorders. Single-nucleotide polymorphisms (SNPs) in the gene encoding VEGF-C are identified as potential risk factors for developing Graves’ disease, Hashimoto’s thyroiditis, and rheumatoid arthritis (RA). Elevated VEGF-C and VEGFR3 mRNA expression is observed in inflammatory bowel disorders like ulcerative colitis (UC) and Crohn’s disease.
Figure 2. Dysregulated VEGFR3 signaling in disease states. (a) VEGFR3 dysregulation in lymphedema. Primary LE: defective activation of VEGFR3 is associated with Milroy’s disease. VEGFR3 and neuropilin-2 transcription is elevated in lymphovenous overgrowth malformations. Missense mutations affecting SHIP-2 protein enhance VEGF-C-induced activation of Akt and ERK in primary lymphedema. Somatic activating PIK3CA mutations are seen in generalized lymphatic anomaly (GLA) and congenital lipomatous overgrowth, vascular malformations, epidermal nevi, and spinal/skeletal anomalies (CLOVES) syndrome. Secondary LE: VEGF-C expression is increased in the plasma of patients with LE. Expression of micro-RNA-1236 in LECs inhibits lymphangiogenesis by downregulating VEGFR3-mediated Akt and ERK activation. (b) VEGFR3 dysregulation and tumor growth and metastases. Tumor-derived VEGF-C is a potent chemoattractant and enhances peritumoral lymphatic access for tumor metastasis. Downregulation of SEMA3F, a NRP2 ligand that negatively regulates VEGFR3 activation, is downregulated in esophageal SCC. Enhanced integrin β1 signaling increases peritumoral metastasis in melanoma. Tumor-derived VEGF C-D expression activates intracellular Akt and ERK signaling to promote peritumoral lymphangiogenesis in breast tumors. (c) VEGFR3 dysregulation in obesity and metabolic syndrome. Increased serum levels of VEGF-C are seen with metabolic disorders. Haploinsufficiency of PROX-1 is associated with excessive lymph accumulation and increased insulin and leptin levels in affected tissues. (d)VEGFR-3 dysregulation in transplant rejection. Inflammatory lymphangiogenesis and corneal allograft rejection is promoted by galectin-8 mediated podoplanin-VEGFR3 cross-talk. VEGF-C-D elevation promotes increased renal lymphangiogenic activity and subsequent renal transplant rejection. (e) VEGFR3 dysregulation in autoimmune/autoinflammatory disorders. Single-nucleotide polymorphisms (SNPs) in the gene encoding VEGF-C are identified as potential risk factors for developing Graves’ disease, Hashimoto’s thyroiditis, and rheumatoid arthritis (RA). Elevated VEGF-C and VEGFR3 mRNA expression is observed in inflammatory bowel disorders like ulcerative colitis (UC) and Crohn’s disease.
Cells 13 00068 g002
Table 1. Proposed VEGFR3-signaling-related molecular alterations in lymphatic-related diseases.
Table 1. Proposed VEGFR3-signaling-related molecular alterations in lymphatic-related diseases.
DiseaseClinical ManifestationsHistopathologic
Findings
Proposed VEGFR3-Related Molecular PathologySelected
References
Milroy/Milroy-like
primary lymphedema
Lower extremity swellingHypoplastic lymphaticsVEGF-C, VEGFR3,
INPPL1 gene mutations
[48,49,51,52,58,73,202]
Hennekam’s
syndrome
Generalized lymphedema and
lymphangiectasia,
variable intellectual
disability, characteristic facial
dysmorphic features
Lymphatic vessel
dysplasia
CCBE-1, ADAMTS3 gene mutations[54,55,56]
Isolated lymphovenous malformationsCongenital lymphatic
malformations, cerebral cavernous
malformations (CCM)
Hyperplastic lymphaticsIncreased cellular VEGFR3, neuropilin-2, ERK 1/2 activity [64,65]
Generalized
lymphatic
anomaly (GLA)
Diffuse or multi-focal lymphatic malformations; cutaneous/soft tissue edema; chylous thoracic effusions, ascites, lymphorrheaHyperproliferative,
dilated lymphatics
PIK3CA mutation
(hyperactivating)
[74]
CLOVES syndromeCapillary, venous,
lymphatic vascular
malformations; thoracic lipomatous hyperplasia; asymmetric limb growth
Macrocystic and
microcystic malformations, or
combined lymphatic
lesions w/disorganized channels
PIK3CA mutation
(hyperactivating)
[76,203]
Proteus
syndrome
Cutaneous,
Musculoskeletal,
and vascular tissue
overgrowth lesions
Hyperplastic
lymphatics, abnormal
lymphovascular channels
AKT1 mutation
(hyperactivating)
PTEN (inactivating)
[78,79]
Gorham–Stout
disease
Spontaneous, progressive osteolysis; soft tissue lymphangioma;
chylothorax
Proliferative ectopic lymphatics within bony structuresKRAS mutation
(activating)
[81,82]
Noonan syndromeLymphedema of bilateral
lower limb and genitals; posterior cervical hygroma
Dilated hyperplastic
lymphatics
Ras-Raf, PTPN11, SHP2
mutations
[84,85,86]
Central conducting
lymphatic anomaly (CCLA)
Abnormal lymphatic drainage by large vessels within the trunkDilated channels, central
lymphatic obstruction
ARAF, EPHB4 mutations[83,88,204]
Tumor MetastasisClinical ManifestationsHistopathologic FindingsProposed VEGFR3-Related Molecular PathologySelected References
Breast cancerLocal tissue infiltration and metastasis to distant organsPeri-tumoral
lymphatic growth and tumor invasion of neighboring lymph nodes
PGE2-EP4 axis
stimulation, CLEC2A activation,
VEGF-C
overexpression
[114,125,126]
MelanomaLocal tissue
infiltration and metastasis to distant organs
Aberrant distribution of melanocytes, Pagetoid spread, dyscohesive nests of melanocytes ARF6 upregulation, integrin B1 upregulation,
k-Cyclin deletion
[118,127,128,133,134,135]
Oral SCCLocal tissue infiltration and metastasis to distant organsTumor budding, perineural and vascular invasion, sarcolemma spread, tumor-infiltrating T-lymphocytes, CD68+-tumor-associated macrophages, tumor-associated tissue eosinophilia, cellular cannibalismNeuropilin-2
upregulation, SEMA3F downregulation, VEGF-C overexpression
[108,115]
Hypopharyngeal cancerLocal tissue infiltration and metastasis to distant organsSquamous cell (most common), submucosal tumor extension EIF4E activation[117]
Esophageal SCCLocal tissue infiltration and metastasis to distant organsKeratin pearls, individual cell keratinization, intercellular bridgesSEMA3F downregulation[116]
CholangiocarcinomaLocal tissue infiltration and metastasis to distant organsAbundant fibrous
desmoplastic stroma
CXCR2-CXCL5 axis
activation
[205]
Autoimmune DisordersClinical ManifestationsHistopathologic FindingsProposed VEGFR3-Related Molecular Pathology Selected References
Autoimmune thyroid disease Grave’s disease; Hashimoto’s
thyroiditis; dysregulated thyroid
hormone secretion
Increased lymphatic vessel densityVEGF-C
polymorphisms
[175,206]
Sjogren’s syndromeDiminished lacrimal and salivary gland function, xerostomia, keratoconjunctivitis sicca, parotid gland enlargementExpansion of lymphatic
capillaries, periductal
inflammatory cell infiltration
VEGFR3,
VEGF-C
polymorphisms
[176]
Behcet’s diseaseMucocutaneous lesions, recurrent
genital ulcerations, uveitis, skin lesions
Expansion of lymphatic capillaries,
periductal inflammatory cell
infiltration
Increased
circulating sVEGFR3,
dysregulated VEGF-C/
VEGFR3 ratio
[177]
Alopecia areataDiscrete, round patches of hair lossDilated dermal lymphaticsElevated vascular endothelial growth factor expression[183]
Systemic lupus
erythematosis (SLE)
Progressive multi-organ tissue fibrosis, vascular damage and inflammationSLE: Dilated cutaneous lymphatic vessels without changes in lymphatic densityIncreased VEGF-C,
VEGF-D, VEGFR2, VEGFR3 activity
[178]
PsoriasisWell circumscribed, circular,
erythematous papules and plaques with dry scaling
Perivascular and dermal
inflammatory cell infiltration, vascular dilatation, edema of dermal papillae, parakeratosis
Increased VEGF-A
expression, VEGFR2
activation
[182]
Rheumatoid arthritisSystemic polyarthritis, articular destructionChronic perilymphatic inflammation, lymphatic leakinessVEGF-C
polymorphisms, impaired
VEGF-C/VEGFR3
signaling
[187,188,189,190]
Still’s diseaseAutoinflammatory fever, arthralgias, lymphadenopathy, joint pain,
persistent papules and plaques
Upper keratinocyte
dyskeratosis, scattered
superficial dermal neutrophils, vacuolar interface
changes, apoptotic
keratinocytes
Increased VEGF-C
expression
[193]
Inflammatory bowel
disease
Crohn’s disease, ulcerative colitis Increased lymphatic vessel densityImpaired
VEGFC/VEGFR3 axis
signaling
[194,195,196,197,198,199,200]
Allograft Transplant RejectionClinical ManifestationsHistopathologic FindingsProposed VEGFR3 Related Molecular PathologySelected References
Corneal transplant
rejection
Progressive end-organ dysfunction, corneal edema, anterior chamber
inflammation, increased intraocular pressure
Increased pathologic
lymphangiogenesis
Increased VEGFR3 activation; galectin-8-mediated integrin- podoplanin-VEGFR3 crosstalk[162,163,166,167,168]
Renal allograft rejectionMalaise, fever, oliguria, graft pain or tenderness, progressive end-organ dysfunctionEndothelial cell swelling and inflammation, dilated peri-tubular capillaries, thrombotic microangiopathy, subendothelial widening (acute), tubular hypertrophy, interstitial fibrosis, mononuclear inflammatory cell infiltrate (chronic)Increased VEGFR3,
VEGF-D, mTORC1
activation
[169,170]
Cardiac allograft
rejection
Arterial luminal occlusion,
diminished cardiac output,
hypotension, mechanical
abnormalities
CD4+ T cell, CD8+ T cell, ED1+ macrophage, myeloperoxidase +, neutrophil
infiltration, perivascular infiltration, interstitial inflammatory cells

Decreased pulmonary lymphatic vessel density, increased hyluronan, fragment accumulation, lymphocytic infiltrate
Increased VEGF-C/VEGFR3
activation
[123,171,172]
Pulmonary allograft
rejection
Dyspnea, cough, sputum production, respiratory distress (acute)Insufficient VEGF-C/VEGFR3
activation
[174]
Obesity/metabolic
syndrome
Hypertension, hyperglycemia, visceral adiposity, dyslipidemiaLeaky lymphatics, impaired lymph drainage, lymphatic fluid stasisProx-1 mutations, increased adipose tissue and serum VEGF-C levels, reduced VEGFR3-AKT-eNOS activation, impaired LEC CD36 activity
[141,142,143,144,150,154,157,158]
Secondary/Acquired LymphedemaClinical ManifestationsHistopathologic FindingsProposed VEGFR3-Related Molecular PathologySelected References
Breast-cancer-related lymphedema (BCRL)Subcutaneous lymphatic fluid stasis and accumulation, fibroadipose skin deposition, dermal thickening CD4+ T cell infiltration, fibrosis,
immature hyperplastic, leaky lymphatics
Prox-1, VEGF-C, Neuropilin-2
mutations, elevated tissue and systemic VEGF-C
expression, SHIP2 mutation,
epigenetic microRNA
alterations
[90,91,92,93,94,96,97,98,207]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kuonqui, K.; Campbell, A.-C.; Sarker, A.; Roberts, A.; Pollack, B.L.; Park, H.J.; Shin, J.; Brown, S.; Mehrara, B.J.; Kataru, R.P. Dysregulation of Lymphatic Endothelial VEGFR3 Signaling in Disease. Cells 2024, 13, 68. https://0-doi-org.brum.beds.ac.uk/10.3390/cells13010068

AMA Style

Kuonqui K, Campbell A-C, Sarker A, Roberts A, Pollack BL, Park HJ, Shin J, Brown S, Mehrara BJ, Kataru RP. Dysregulation of Lymphatic Endothelial VEGFR3 Signaling in Disease. Cells. 2024; 13(1):68. https://0-doi-org.brum.beds.ac.uk/10.3390/cells13010068

Chicago/Turabian Style

Kuonqui, Kevin, Adana-Christine Campbell, Ananta Sarker, Arielle Roberts, Bracha L. Pollack, Hyeung Ju Park, Jinyeon Shin, Stav Brown, Babak J. Mehrara, and Raghu P. Kataru. 2024. "Dysregulation of Lymphatic Endothelial VEGFR3 Signaling in Disease" Cells 13, no. 1: 68. https://0-doi-org.brum.beds.ac.uk/10.3390/cells13010068

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

Article Metrics

Back to TopTop