Entry - *136352 - FMS-LIKE TYROSINE KINASE 4; FLT4 - OMIM
* 136352

FMS-LIKE TYROSINE KINASE 4; FLT4


Alternative titles; symbols

VASCULAR ENDOTHELIAL GROWTH FACTOR RECEPTOR 3; VEGFR3


HGNC Approved Gene Symbol: FLT4

Cytogenetic location: 5q35.3     Genomic coordinates (GRCh38): 5:180,601,506-180,650,298 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
5q35.3 Congenital heart defects, multiple types, 7 618780 AD 3
Hemangioma, capillary infantile, somatic 602089 3
Lymphatic malformation 1 153100 AD 3

TEXT

Cloning and Expression

By screening a placenta cDNA library with a mouse Flt3 probe, Galland et al. (1992) isolated a human gene encoding a putative receptor-type tyrosine kinase. The deduced amino acid sequence of the intracellular portion of the molecule showed that it was strongly related to FLT1 (165070) and KDR (191306) and to a lesser degree to members of the class III receptor-type tyrosine kinases: FMS (164770), PDGFR (173490, 173410), KIT (164920), and FLT3 (136351).


Mapping

Galland et al. (1992) mapped FLT4 to chromosome 5q34-q35, telomeric to the FMS and PDGFRB genes, by in situ hybridization. They assigned the mouse homolog to chromosome 11 by the same method. In the process of creating a radiation hybrid map of 18 genes, Warrington et al. (1992) demonstrated that the FLT4 gene is located on distal 5q between GABRA1 (137160) at 5q34-q35 and DRD1 (126449) at 5q35.1. Aprelikova et al. (1992) also mapped the FLT4 gene to chromosome 5q33-qter.


Gene Function

Among the factors stimulating angiogenesis, the acidic and basic fibroblast growth factors FGF1 (131220) and FGF2 (134920) and the vascular endothelial growth factor VEGF (192240) exert their effects via specific cell surface receptor tyrosine kinases: for FGF1 and FGF2, FGF receptor-1 (FGFR1; 136350), also known as FLT2, and the endothelial-specific FMS-like tyrosine kinase-1; and for VEGF, the KDR/FLK1 receptor. The protein product of the FLT4 receptor tyrosine kinase cDNA is structurally similar to the FLT1 and KDR/FLK1 receptors (Pajusola et al., 1992), but FLT4 does not bind VEGF (Pajusola et al., 1994). Lee et al. (1996) identified and characterized a vascular endothelial growth factor-related protein (VEGFC; 601528) that specifically binds to the extracellular domain of Flt4 and stimulates tyrosine phosphorylation and mitogenesis of endothelial cells.

Kaipainen et al. (1995) analyzed the expression of FLT4 by in situ hybridization during mouse embryogenesis and in adult human tissues. The FLT4 mRNA signals first became detectable in the angioblasts of head mesenchyme, the cardinal vein, and extraembryonally in the allantois of 8.5-day postcoitus (p.c.) embryos. In 12.5-day p.c. embryos, the FLT4 signal decorated developing venous and presumptive lymphatic endothelia, but arterial endothelia were negative. During later stages of development, FLT4 mRNA became restricted to vascular plexuses devoid of red cells, representing developing lymphatic vessels. In adult human tissues, only the lymphatic endothelia and some high endothelial venules expressed FLT4 mRNA. Increased expression occurred in lymphatic sinuses in metastatic lymph nodes and in lymphangioma. The results suggested that FLT4 is a marker for lymphatic vessels and some high endothelial venules in human adult tissues. They also supported the theory of the venous origin of lymphatic vessels.

Vascular endothelial growth factor is a key regulator of blood vessel development in embryos and angiogenesis in adult tissues. Unlike VEGF, the related VEGFC stimulates the growth of lymphatic vessels through its specific lymphatic endothelial receptor VEGFR3. Dumont et al. (1998) showed that targeted inactivation of the VEGFR3 gene in mice resulted in defective blood vessel development in early embryos. Vasculogenesis and angiogenesis occurred, but large vessels became abnormally organized with defective lumens, leading to fluid accumulation in the pericardial cavity and cardiovascular failure at embryonic day 9.5. Thus, VEGFR3 has an essential role in the development of the embryonic cardiovascular system before the emergence of the lymphatic vessels.

In studies of the mouse cornea in vivo, Cursiefen et al. (2006) demonstrated that the presence of Vegfr3 in the corneal epithelium is critical for suppressing inflammatory corneal angiogenesis, by acting as a decoy receptor and binding the angiogenic growth factors VEGFC and VEGFD (FIGF; 300091). Cursiefen et al. (2006) concluded that VEGFR3, which provides proangiogenic signaling when expressed on endothelium, may also have antiangiogenic properties when expressed at an avascular site by nonendothelial cells.

In studies in zebrafish to investigate the role of Notch (see 190198) signaling in angiogenesis, Siekmann and Lawson (2007) found that Flt4 is expressed in segmental artery tip cells and becomes ectopically expressed throughout the sprout in the absence of Notch. Loss of Flt4 could partially restore normal endothelial cell number in Rbpsuh (147183)-deficient segmental arteries. Finally, loss of the Notch ligand Dll4 (605185) also led to an increased number of endothelial cells within segmental arteries. Siekmann and Lawson (2007) concluded that their studies taken together indicated that proper specification of cell identity, position, and behavior in a developing blood vessel sprout is required for normal angiogenesis, and implicated the Notch signaling pathway in this process.

Tammela et al. (2008) demonstrated that VEGFR3 is highly expressed in angiogenic sprouts, and that genetic targeting of VEGFR3 or blocking of VEGFR3 signaling with monoclonal antibodies results in decreased sprouting, vascular density, vessel branching, and endothelial cell proliferation in mouse angiogenesis models. Stimulation of VEGFR3 augmented VEGF-induced angiogenesis and sustained angiogenesis even in the presence of VEGFR2 (191306) inhibitors, whereas antibodies against VEGFR3 and VEGFR2 in combination resulted in additive inhibition of angiogenesis and tumor growth. Furthermore, genetic or pharmacologic disruption of the Notch signaling pathway led to widespread endothelial VEGFR3 expression and excessive sprouting, which was inhibited by blocking VEGFR3 signals. Tammela et al. (2008) concluded that their results implicated VEGFR3 as a regulator of vascular network formation. The authors suggested that targeting VEGFR3 may provide additional efficacy for antiangiogenic therapies, especially towards vessels that are resistant to VEGF or VEGFR2 inhibitors.

Wang et al. (2010) demonstrated with genetic experiments in mouse and zebrafish that ephrin-B2 (EFNB2; 600527), a transmembrane ligand for Ephrin receptor tyrosine kinases, promotes sprouting behavior and motility in the angiogenic endothelium. Wang et al. (2010) linked this proangiogenic function to a crucial role of ephrin-B2 in the VEGF signaling pathway, which they studied in detail for VEGFR3, the receptor for VEGF-C. In the absence of ephrin-B2, the internalization of VEGFR3 in cultured cells and mutant mice is defective, which compromises downstream signal transduction by the small GTPase Rac1 (602048), Akt (164730), and the mitogen-activated protein kinase Erk (601795). Wang et al. (2010) concluded that VEGFR3 signaling is coupled to receptor internalization. Ephrin-B2 is a key regulator of this process and thereby controls angiogenic and lymphangiogenic growth.

Benedito et al. (2012) used inducible loss-of-function genetics in combination with inhibitors in vivo to demonstrate that DLL4 (605185) protein expression in retinal tip cells is only weakly modulated by VEGFR2 (191306) signaling. Surprisingly, Notch (190198) inhibition also had no significant impact on VEGFR2 expression and induced deregulated endothelial sprouting and proliferation even in the absence of VEGFR2, which is the most important VEGFA (192240) receptor and is considered to be indispensable for these processes. By contrast, VEGFR3, the main receptor for VEGFC (601528), was strongly modulated by Notch. VEGFR3 kinase activity inhibitors but not ligand-blocking antibodies suppressed the sprouting of endothelial cells that had low Notch signaling activity. Benedito et al. (2012) concluded that their results established that VEGFR2 and VEGFR3 are regulated in a highly differential manner by Notch. They proposed that successful antiangiogenic targeting of these receptors and their ligands will strongly depend on the status of endothelial Notch signaling.

By screening immunity-related proteins and their receptors for bacteria- or LPS-induced expression, Zhang et al. (2014) detected upregulation of VEGFR3 and VEGFC in macrophages. Serum VEGFC was also increased in patient and mouse models of septic shock. Ligation of VEGFR3 by VEGFC attenuated proinflammatory cytokine production. In the absence of either the ligand-binding domain or tyrosine kinase activity of Vegfr3, mice became more sensitive to septic shock. Vegfr3 restrained Tlr4 (603030)-NFKB (see 164011) activation by regulating the PI3K (see 601232)-Akt signaling pathway and Socs1 (603597) expression. Zhang et al. (2014) proposed that in addition to targeting lymphatic vessels, VEGFR3 signaling via VEGFC prevents microphagic overreaction to infections complicated by lymphedema.

Lorenz et al. (2018) used the developing liver as a model organ to study angiocrine signals and showed that the growth rate of the liver correlates both spatially and temporally with blood perfusion to this organ. By manipulating blood flow through the liver vasculature, Lorenz et al. (2018) demonstrated that vessel perfusion activates beta-1 integrin (135630) and VEGFR3. Notably, both beta-1 integrin and VEGFR3 are strictly required for normal production of hepatocyte growth factor, survival of hepatocytes, and liver growth. Ex vivo perfusion of adult mouse liver and in vitro mechanical stretching of human hepatic endothelial cells illustrated that mechanotransduction alone is sufficient to turn on angiocrine signals. When the endothelial cells are mechanically stretched, angiocrine signals trigger in vitro proliferation and survival of primary human hepatocytes. Lorenz et al. (2018) concluded that their findings uncovered a signaling pathway in vascular endothelial cells that translates blood perfusion and mechanotransduction into organ growth and maintenance.


Molecular Genetics

Lymphatic Malformation 1

In affected members of a family with lymphatic malformation-1 (LMPHM1; 153100), Ferrell et al. (1998) identified a mutation in the FLT4 gene (136352.0005).

Karkkainen et al. (2000) identified mutations at the FLT4 locus in several families with hereditary lymphedema. They found that all disease-associated alleles analyzed had missense mutations and encoded proteins with an inactive tyrosine kinase, preventing downstream gene activation. These studies established that vascular endothelial growth factor receptor-3 is important for normal lymphatic vascular function.

In a family with hereditary lymphedema, Irrthum et al. (2000) identified a mutation in the FLT4 gene (136352.0006) that cosegregated with the disease. In vitro expression showed that this mutation inhibited the autophosphorylation of the receptor.

Evans et al. (2003) identified 8 different heterozygous mutations in the FLT4 gene (see, e.g., 136352.0011) in affected members of 12 different Caucasian families with hereditary lymphedema. All the mutations occurred in the tyrosine kinase domains. Several families showed incomplete penetrance of the phenotype.

Kim and Dumont (2003) reviewed molecular mechanisms in lymphangiogenesis and their implications for human disease. In addition to VEGFR3 and FOXC2 (602402), 6 'lymphangiogenic markers' were reviewed. The role of some of these lymphangiogenetic mechanisms in cancer and metastasis was also reviewed.

Ghalamkarpour et al. (2006) identified mutations in the FLT4 gene (see, e.g., 136352.0008-136352.0009) in affected members from 3 unrelated families with autosomal dominant lymphedema and in a sporadic case.

In 14 affected and 2 unaffected members of a 3-generation consanguineous Israeli family of Muslim Arab origin with hereditary lymphedema, Spiegel et al. (2006) identified heterozygosity for a missense mutation in the VEGFR3 gene (136352.0010).

Connell et al. (2009) identified mutations in the FLT4 gene, including 14 novel mutations, in 22 (42%) of 52 patients with primary lymphedema. Mutation prevalence was 75% in patients with a typical Milroy phenotype and a positive family history, and 68% if positive family history was not a diagnostic criterion. No mutations were found outside the kinase domains, showing that analysis of nonkinase domains of FLT4 is not useful for Milroy disease patients. No mutations were identified in the VEGFC gene (601528), which encodes the FLT4 ligand.

In a Hispanic female with congenital lymphedema who was born of first-cousin parents, Ghalamkarpour et al. (2009) identified homozygosity for a missense mutation in the ATP-binding domain of the FLT4 gene (136352.0012). Her unaffected parents were heterozygous for the hypomorphic mutation, which was not found in 110 controls.

Congenital Heart Defects, Multiple Types, 7

In a cohort of 2,871 probands with congenital heart disease, comprising 2,645 parent-offspring trios and 226 singletons, Jin et al. (2017) performed whole-exome sequencing and identified 10 probands who were heterozygous for frameshift or nonsense mutations in the FLT4 gene. In 2 probands the mutations appeared to have arisen de novo and in 2 probands the mutations were inherited from an affected parent; however, in 6 probands, the mutation was inherited from an unaffected parent. Cardiac diagnoses in affected individuals included tetralogy of Fallot, pulmonary stenosis or atresia, absent pulmonary valve, right aortic arch, double aortic arch, and major aortopulmonary collateral arteries. The authors stated that FLT4 mutation carriers had no extracardiac malformations (unspecified), but elsewhere in the text stated that 1 proband had an extracardiac congenital anomaly.

Reuter et al. (2019) sequenced 175 adult patients with tetralogy of Fallot and 56 with other congenital cardiac anomalies for loss-of-function and deleterious mutations in FLT4 and other VEGF pathway genes. They identified 9 (5.1%) probands with novel FLT4 variants, all of whom came from the group of individuals with tetralogy of Fallot. Seven of the variants were predicted to have a loss-of-function effect, implicating haploinsufficiency, and comprised 2 stopgain, 3 frameshift, 1 splice site, and 1 multiexon deletion mutation; the other 2 mutations, a missense and an in-frame deletion, were predicted to be deleterious. In addition to variants in FLT4, multiple variants were also identified in the KDR (191306) gene.


Animal Model

The Chy mouse mutant, characterized by accumulation of chylous ascites and swelling of the limbs, was obtained by ethylnitrosourea-induced mutagenesis (Lyon and Glenister (1984, 1986)). The phenotype is linked to mouse chromosome 11. Karkkainen et al. (2001) sequenced the Vegfr3 candidate gene on chromosome 11 in Chy mice and found a heterozygous 3157A-T mutation resulting in an ile1053-to-phe (I1053F) substitution in the tyrosine kinase domain. This mutation was located in a highly conserved catalytic domain of the receptor, in close proximity to the VEGFR3 mutations in human primary lymphedema. The I1053F mutant receptor was tyrosine kinase inactive. Although lymphedema patients with heterozygous missense mutations of VEGFR3 retain some receptor activity because of the presence of the wildtype allele (Karkkainen et al., 2000), the mutant VEGFR3 can be classified as a dominant-negative receptor similar to certain mutant KIT receptors in piebaldism (172800) and RET receptors (164761) in Hirschsprung disease (142623). Karkkainen et al. (2001) found that neuropilin-2 (NRP2; 602070) bound VEGFC and was expressed in the visceral, but not in the cutaneous, lymphatic endothelia. This may explain why hypoplastic cutaneous, but not visceral, lymphatic vessels were found in the Chy mice. Using virus-mediated VEGFC gene therapy, Karkkainen et al. (2001) generated functional lymphatic vessels in the lymphedema mice. The results suggested that growth factor gene therapy is applicable to human lymphedema as well and provided a paradigm for other diseases associated with mutant receptors, i.e., ligand therapy.


ALLELIC VARIANTS ( 17 Selected Examples):

.0001 MOVED TO 136352.0005


.0002 LYMPHATIC MALFORMATION 1

FLT4, GLY857ARG
  
RCV000017647...

In a family with lymphatic malformation-1 (LMPHM1; 153100) in members of 3 generations, Karkkainen et al. (2000) identified a heterozygous G-A transition in the FLT4 gene, resulting in a gly857-to-arg (G857R) substitution.


.0003 LYMPHATIC MALFORMATION 1

FLT4, ARG1041PRO
  
RCV000017648

In a family with hereditary lymphedema (LMPHM1; 153100) in at least 4 generations, Karkkainen et al. (2000) identified a heterozygous mutation in the FLT4 gene, resulting in an arg1041-to-pro (R1041P) substitution.


.0004 LYMPHATIC MALFORMATION 1

FLT4, LEU1044PRO
  
RCV000017649

In a large family with autosomal dominant lymphedema (LMPHM1; 153100) in 5 generations and many different sibships, Karkkainen et al. (2000) identified heterozygosity for a transition in the FLT4 gene, resulting in a leu1044-to-pro (L1044P) substitution.


.0005 LYMPHATIC MALFORMATION 1

FLT4, PRO1114LEU
  
RCV000017650

In a mother and 2 daughters with primary lymphedema (LMPHM1; 153100), Karkkainen et al. (2000) identified a heterozygous pro1114-to-leu (P1114L) missense mutation of the FLT4 gene.

Ferrell et al. (1998) had originally described the mutation in this family as a 3360G-A transition in the FLT4 gene, predicted to cause a nonconservative PRO1126LEU (P1126L) substitution in the mature receptor (Karkkainen et al., 2000).


.0006 LYMPHATIC MALFORMATION 1

FLT4, HIS1035ARG
  
RCV000017651

In a family in which the father and 4 of 7 children had congenital lymphedema (LMPHM1; 153100), Irrthum et al. (2000) demonstrated a heterozygous his1035-to-arg (H1035R) missense mutation in the FLT4 gene.


.0007 HEMANGIOMA, CAPILLARY INFANTILE, SOMATIC

FLT4, PRO954SER
  
RCV000017652...

In 1 of 15 infantile hemangioma (602089) specimens, Walter et al. (2002) found a pro954-to-ser (P954S) missense mutation in the kinase insert of the FLT4 gene. This result, and the finding of a somatic missense mutation in the VEGFR2 gene (191306.0001) in another of the 15 specimens, suggested that alteration of the FLT4 signaling pathway in endothelial and/or pericytic cells may be a mechanism involved in hemangioma formation.


.0008 LYMPHATIC MALFORMATION 1

FLT4, VAL878MET
  
RCV000017653

In 4 affected individuals of a family with autosomal dominant lymphedema (LMPHM1; 153100), Ghalamkarpour et al. (2006) identified a heterozygous 2632G-A transition in the FLT4 gene, resulting in a val878-to-met (V878M) substitution in the tyrosine kinase domain I of the protein. One affected family member was a 22-week-old fetus who was found to have fetal hydrops with bilateral leg edema, pleural effusions, hydrothorax, and pulmonary hypoplasia on ultrasound. The pregnancy was terminated. Other affected family members had congenital lymphedema of the legs with variable severity.


.0009 LYMPHATIC MALFORMATION 1

FLT4, ILE1086THR
  
RCV000017654...

In affected members of a family with autosomal dominant lymphedema (LMPHM1; 153100) spanning 5 generations, Ghalamkarpour et al. (2006) identified a heterozygous 3257T-C transition in the FLT4 gene, resulting in an ile1086-to-thr (I1086T) substitution in the tyrosine kinase II domain of the protein. The proband had a severe phenotype with elephantiasis up to the inguinal ligament bilaterally, chronic venous ulcerations, cellulitis, and papillomatosis.


.0010 LYMPHATIC MALFORMATION 1

FLT4, GLU1106LYS
  
RCV000017655

In 14 affected and 2 unaffected members of a 3-generation consanguineous Israeli family of Muslim Arab origin with hereditary lymphedema (LMPHM1; 153100), Spiegel et al. (2006) identified heterozygosity for a 3316G-A transition in the FLT4 gene, resulting in a glu1106-to-lys (E1106K) substitution. The mutation was not found in 110 control individuals.


.0011 LYMPHATIC MALFORMATION 1

FLT4, 3-BP DEL, 3323TCT
  
RCV000017656...

In affected members of a family with hereditary lymphedema (LMPHM1; 153100), Evans et al. (2003) identified a heterozygous 3-bp in-frame deletion (3323delTCT), resulting in the deletion of residue phe1108 in the tyrosine kinase II domain.


.0012 LYMPHATIC MALFORMATION 1

FLT4, ALA855THR
  
RCV000017657...

In a Hispanic female with congenital lymphedema (LMPHM1; 153100) who was born of first-cousin parents, Ghalamkarpour et al. (2009) identified homozygosity for a 2563G-A transition in exon 18 of the FLT4 gene, resulting in an ala855-to-thr (A855T) substitution in the ATP-binding domain of the receptor. The unaffected parents were heterozygous for the mutation, which was not found in 110 controls. Assessment of receptor function showed reduced phosphorylation of the receptor, with impaired ligand-induced internalization and defective downstream signaling.


.0013 CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, TYR361TER
   RCV001003426

In a mother and son (family 1-04970), both with tetralogy of Fallot (CHTD7; 618780), Jin et al. (2017) identified a tyrosine-to-stop substitution at codon 361 of the FLT4 protein (Y361X). The son additionally had right-sided aortic arch. Hamosh (2020) noted that this variant was absent from the gnomAD database on February 17, 2020.


.0014 CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, 2-BP DEL
  
RCV001003427...

In 2 unrelated probands (1-03410 and 1-05967) with tetralogy of Fallot, pulmonary atresia, and major aortopulmonary collateral arteries (CHTD7; 618780), Jin et al. (2017) identified deletion of a C nucleotide in the FLT4 gene resulting in a proline-to-arginine substitution at codon 30, followed by a premature termination codon 3 amino acids downstream (Pro30ArgfsTer3). In family 1-03410, the mutation occurred as a de novo event. The proband also had right-sided aortic arch. In family 1-05967, the proband had inherited the mutation from her unaffected mother. She additionally had double aortic arch.

Reuter et al. (2019) identified this mutation in a patient (CGC-034) with tetralogy of Fallot, pulmonary atresia, and major aortopulmonary collateral arteries, as well as congenital lymphedema. She had inherited the mutation from her mother, who had normal echocardiography results. The mother's father had bradycardia. Reuter et al. (2019) recorded the mutation as c.89delC (c.89delC, NM_182925.4).


.0015 CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, EX25-29DEL
   RCV001003428

In a 23-year-old female (TOF293) with tetralogy of Fallot, right-sided aortic arch, and absent pulmonary valve (CHTD7; 618780), Reuter et al. (2019) identified heterozygous deletion of exons 25 through 29 of the FLT4 gene (chr5:180,031,767-180,040,470del).


.0016 CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, GLN1192TER
  
RCV001003429

In a 79-year-old man (TOF158) with tetralogy of Fallot, right-sided aortic arch, and atrial flutter requiring ablation (CHTD7; 618780), and in his daughter with tetralogy of Fallot, Reuter et al. (2019) identified a C-to-T transition at nucleotide 3574 (c.3574C-T, NM_182925.4) of the FLT4 gene, resulting in a glutamine-to-termination substitution at codon 1192 (Q1192X). The variant was absent from gnomAD.


.0017 CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, 1-BP DUP, 1622G
  
RCV001003430

In a 29-year-old man (TOF284) with tetralogy of Fallot, major aortopulmonary collateral arteries, and aortic valve replacement (CHTD7; 618780), Reuter et al. (2019) identified duplication of a G at nucleotide 1622 (c.1622dupG, NM_182925.4) of the FLT4 gene, resulting in a frameshift and premature termination (Gln542ProfsTer3). The variant was absent from gnomAD.


REFERENCES

  1. Aprelikova, O., Pajusola, K., Partanen, J., Armstrong, E., Alitalo, R., Bailey, S. K., McMahon, J., Wasmuth, J., Huebner, K., Alitalo, K. FLT4, a novel class III receptor tyrosine kinase in chromosome 5q33-qter. Cancer Res. 52: 746-748, 1992. [PubMed: 1310071, related citations]

  2. Benedito, R., Rocha, S. F., Woeste, M., Zamykal, M., Radtke, F., Casanovas, O., Duarte, A., Pytowski, B., Adams, R. H. Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF-VEGFR2 signalling. Nature 484: 110-114, 2012. [PubMed: 22426001, related citations] [Full Text]

  3. Connell, F. C., Ostergaard, P., Carver, C., Brice, G., Williams, N., Mansour, S., Mortimer, P. S., Jeffery, S., Lymphoedema Consortium. Analysis of the coding regions of VEGFR3 and VEGFC in Milroy disease and other primary lymphoedemas. Hum. Genet. 124: 625-631, 2009. Note: Erratum: Hum. Genet. 125: 237 only, 2009. [PubMed: 19002718, related citations] [Full Text]

  4. Cursiefen, C., Chen, L., Saint-Geniez, M., Hamrah, P., Jin, Y., Rashid, S., Pytowksi, B., Persaud, K., Wu, Y., Streilein, J. W., Dana, R. Nonvascular VEGF receptor 3 expression by corneal epithelium maintains avascularity and vision. Proc. Nat. Acad. Sci. 103: 11405-11410, 2006. [PubMed: 16849433, images, related citations] [Full Text]

  5. Dumont, D. J., Jussila, L., Taipale, J., Lymboussaki, A., Mustonen, T., Pajusola, K., Breitman, M., Alitalo, K. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science 282: 946-949, 1998. [PubMed: 9794766, related citations] [Full Text]

  6. Evans, A. L., Bell, R., Brice, G., Comeglio, P., Lipede, C., Jeffery, S., Mortimer, P., Sarfarazi, M., Child, A. H. Identification of eight novel VEGFR-3 mutations in families with primary lymphoedema. J. Med. Genet. 40: 697-703, 2003. [PubMed: 12960217, related citations] [Full Text]

  7. 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. Molec. Genet. 7: 2073-2078, 1998. [PubMed: 9817924, related citations] [Full Text]

  8. Galland, F., Karamysheva, A., Mattei, M.-G., Rosnet, O., Marchetto, S., Birnbaum, D. Chromosomal localization of FLT4, a novel receptor-type tyrosine kinase gene. Genomics 13: 475-478, 1992. [PubMed: 1319394, related citations] [Full Text]

  9. 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. 46: 399-404, 2009. [PubMed: 19289394, related citations] [Full Text]

  10. 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. 70: 330-335, 2006. [PubMed: 16965327, related citations] [Full Text]

  11. Hamosh, A. Personal Communication. Baltimore, Md. 02/17/2020.

  12. 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. 67: 295-301, 2000. [PubMed: 10856194, images, related citations] [Full Text]

  13. Jin, S. C., Homsy, J., Zaidi, S., Lu, Q., Morton, S., DePalma, S. R., Zeng, X., Qi, H., Chang, W., Sierant, M. C., Hung, W.-C., Haider, S., and 33 others. Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands. Nature Genet. 49: 1593-1601, 2017. [PubMed: 28991257, related citations] [Full Text]

  14. Kaipainen, A., Korhonen, J., Mustonen, T., van Hinsbergh, V. W. M., Fang, G.-H., Dumont, D., Breitman, M., Alitalo, K. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc. Nat. Acad. Sci. 92: 3566-3570, 1995. [PubMed: 7724599, related citations] [Full Text]

  15. 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. Nature Genet. 25: 153-159, 2000. [PubMed: 10835628, related citations] [Full Text]

  16. Karkkainen, M. J., Saaristo, A., Jussila, L., Karila, K. A., Lawrence, E. C., Pajusola, K., Bueler, H., Eichmann, A., Kauppinen, R., Kettunen, M. I., Yla-Herttuala, S., Finegold, D. N., Ferrell, R. E., Alitalo, K. A model for gene therapy of human hereditary lymphedema. Proc. Nat. Acad. Sci. 98: 12677-12682, 2001. [PubMed: 11592985, images, related citations] [Full Text]

  17. Kim, H., Dumont, D. J. Molecular mechanisms in lymphangiogenesis: model systems and implications in human disease. Clin. Genet. 64: 282-292, 2003. [PubMed: 12974730, related citations] [Full Text]

  18. Lee, J., Gray, A., Yuan, J., Luoh, S.-M., Avraham, H., Wood, W. I. Vascular endothelial growth factor-related protein: a ligand and specific activator of the tyrosine kinase receptor Flt4. Proc. Nat. Acad. Sci. 93: 1988-1992, 1996. [PubMed: 8700872, related citations] [Full Text]

  19. Lorenz, L., Axnick, J., Buschmann, T., Henning, C., Urner, S., Fang, S., Nurmi, H., Eichhorst, N., Holtmeier, R., Bodis, K., Hwang, J.-H., Mussig, K., Eberhard, D., Stypmann, J., Kuss, O., Roden, M., Alitalo, K., Haussinger, D., Lammert, E. Mechanosensing by beta-1 integrin induces angiocrine signals for liver growth and survival. Nature 562: 128-132, 2018. [PubMed: 30258227, related citations] [Full Text]

  20. Lyon, M. F., Glenister, P. H. New Mutants. Mouse Newsletter 71: 26 only, 1984.

  21. Lyon, M. F., Glenister, P. H. Gene order of Chy-vt-Re on chromosome 11. Mouse Newsletter 74: 96 only, 1986.

  22. Milroy, W. F. An undescribed variety of hereditary oedema. New York Med. J. 56: 505-508, 1892.

  23. Offori, T. W., Platt, C. C., Stephens, M., Hopkinson, G. B. Angiosarcoma in congenital hereditary lymphoedema (Milroy's disease): diagnostic beacons and a review of the literature. Clin. Exp. Derm. 18: 174-177, 1993. [PubMed: 8482001, related citations] [Full Text]

  24. Pajusola, K., Aprelikova, O., Korhonen, J., Kaipainen, A., Pertovaara, L., Alitalo, R., Alitalo, K. FLT4 receptor tyrosine kinase contains seven immunoglobulin-like loops and is expressed in multiple human tissues and cell lines. Cancer Res. 52: 5738-5743, 1992. Note: Erratum: Cancer Res. 53: 3845 only, 1993. [PubMed: 1327515, related citations]

  25. Pajusola, K., Aprelikova, O., Pelicci, G., Weich, H., Claesson-Welsh, L., Alitalo, K. Signalling properties of FLT4, a proteolytically processed receptor tyrosine kinase related to two VEGF receptors. Oncogene 9: 3545-3555, 1994. [PubMed: 7970715, related citations]

  26. Reuter, M. S., Jobling, R., Chaturvedi, R. R., Manshaei, R., Costain, G., Heung, T., Curtis, M., Hosseini, S. M., Liston, E., Lowther, C., Oechslin, E., Sticht, H., and 9 others. Haploinsufficiency of vascular endothelial growth factor related signaling genes is associated with tetralogy of Fallot. Genet. Med. 21: 1001-1007, 2019. [PubMed: 30232381, related citations] [Full Text]

  27. Siekmann, A. F., Lawson, N. D. Notch signalling limits angiogenic cell behaviour in developing zebrafish arteries. Nature 445: 781-784, 2007. [PubMed: 17259972, related citations] [Full Text]

  28. Spiegel, R., Ghalamkarpour, A., Daniel-Spiegel, E., Vikkula, M., Shalev, S. A. Wide clinical spectrum in a family with hereditary lymphedema type I due to a novel missense mutation in VEGFR3. J. Hum. Genet. 51: 846-850, 2006. [PubMed: 16924388, related citations] [Full Text]

  29. Tammela, T., Zarkada, G., Wallgard, E., Murtomaki, A., Suchting, S., Wirzenius, M., Waltari, M., Hellstrom, M., Schomber, T., Peltonen, R., Freitas, C., Duarte, A., and 9 others. Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation. Nature 454: 656-660, 2008. [PubMed: 18594512, related citations] [Full Text]

  30. Walter, J. W., North, P. E., Waner, M., Mizeracki, A., Blei, F., Walker, J. W. T., Reinisch, J. F., Marchuk, D. A. Somatic mutation of vascular endothelial growth factor receptors in juvenile hemangioma. Genes Chromosomes Cancer 33: 295-303, 2002. [PubMed: 11807987, related citations] [Full Text]

  31. Wang, Y., Nakayama, M., Pitulescu, M. E., Schmidt, T. S., Bochenek, M. L., Sakakibara, A., Adams, S., Davy, A., Deutsch, U., Luthi, U., Barberis, A., Benjamin, L. E., Makinen, T., Nobes, C. D., Adams, R. H. Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Nature 465: 483-486, 2010. [PubMed: 20445537, related citations] [Full Text]

  32. Warrington, J. A., Bailey, S. K., Armstrong, E., Aprelikova, O., Alitalo, K., Dolganov, G. M., Wilcox, A. S., Sikela, J. M., Wolfe, S. F., Lovett, M., Wasmuth, J. J. A radiation hybrid map of 18 growth factor, growth factor receptor, hormone receptor, or neurotransmitter receptor genes on the distal region of the long arm of chromosome 5. Genomics 13: 803-808, 1992. [PubMed: 1322355, related citations] [Full Text]

  33. Zhang, Y., Lu, Y., Ma, L., Cao, X., Xiao, J., Chen, J., Jiao, S., Gao, Y., Liu, C., Duan, Z., Li, D., He, Y., Wei, B., Wang, H. Activation of vascular endothelial growth factor receptor-3 in macrophages restrains TLR4-NF-kappa-beta signaling and protects against endotoxin shock. Immunity 40: 501-514, 2014. [PubMed: 24656836, related citations] [Full Text]


Ada Hamosh - updated : 02/17/2020
Ada Hamosh - updated : 09/13/2019
Marla J. F. O'Neill - updated : 04/17/2018
Paul J. Converse - updated : 9/19/2014
Ada Hamosh - updated : 4/24/2012
Ada Hamosh - updated : 6/30/2010
Cassandra L. Kniffin - updated : 6/10/2009
Cassandra L. Kniffin - updated : 2/25/2009
Ada Hamosh - updated : 10/20/2008
Cassandra L. Kniffin - updated : 4/18/2008
Ada Hamosh - updated : 6/26/2007
Marla J. F. O'Neill - updated : 12/29/2006
Cassandra L. Kniffin - updated : 11/1/2006
Marla J. F. O'Neill - updated : 9/29/2006
Cassandra L. Kniffin - reorganized : 11/19/2003
Victor A. McKusick - updated : 11/4/2003
Victor A. McKusick - updated : 3/14/2002
Victor A. McKusick - updated : 1/14/2002
Victor A. McKusick - updated : 10/3/2000
Victor A. McKusick - updated : 5/25/2000
Victor A. McKusick - updated : 2/10/1999
Victor A. McKusick - updated : 1/6/1999
Victor A. McKusick - updated : 11/10/1998
Victor A. McKusick - updated : 10/27/1998
Creation Date:
Victor A. McKusick : 6/1/1992
alopez : 06/25/2020
alopez : 02/18/2020
carol : 02/18/2020
alopez : 02/17/2020
alopez : 02/17/2020
alopez : 09/13/2019
carol : 12/18/2018
alopez : 11/07/2018
carol : 04/18/2018
carol : 04/17/2018
mgross : 09/29/2014
carol : 9/22/2014
mcolton : 9/19/2014
carol : 1/30/2013
carol : 1/29/2013
carol : 1/2/2013
alopez : 4/25/2012
terry : 4/24/2012
alopez : 6/30/2010
terry : 6/30/2010
wwang : 9/28/2009
wwang : 7/1/2009
ckniffin : 6/10/2009
wwang : 6/1/2009
ckniffin : 5/20/2009
wwang : 3/5/2009
ckniffin : 2/25/2009
alopez : 10/22/2008
terry : 10/20/2008
wwang : 4/18/2008
alopez : 7/2/2007
terry : 6/26/2007
wwang : 12/29/2006
ckniffin : 11/6/2006
carol : 11/6/2006
ckniffin : 11/1/2006
wwang : 9/29/2006
wwang : 9/29/2006
carol : 11/19/2003
ckniffin : 11/12/2003
tkritzer : 11/6/2003
terry : 11/4/2003
cwells : 3/20/2002
cwells : 3/18/2002
terry : 3/14/2002
terry : 3/8/2002
carol : 1/20/2002
mcapotos : 1/14/2002
mcapotos : 1/14/2002
carol : 10/5/2000
terry : 10/3/2000
alopez : 5/25/2000
alopez : 11/18/1999
terry : 3/9/1999
carol : 3/3/1999
carol : 2/10/1999
terry : 2/10/1999
carol : 1/18/1999
terry : 1/7/1999
terry : 1/6/1999
carol : 11/18/1998
carol : 11/17/1998
terry : 11/16/1998
terry : 11/10/1998
dkim : 11/6/1998
alopez : 10/29/1998
terry : 10/27/1998
dkim : 7/2/1998
carol : 7/20/1992
carol : 6/1/1992

* 136352

FMS-LIKE TYROSINE KINASE 4; FLT4


Alternative titles; symbols

VASCULAR ENDOTHELIAL GROWTH FACTOR RECEPTOR 3; VEGFR3


HGNC Approved Gene Symbol: FLT4

SNOMEDCT: 399889006;  


Cytogenetic location: 5q35.3     Genomic coordinates (GRCh38): 5:180,601,506-180,650,298 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
5q35.3 Congenital heart defects, multiple types, 7 618780 Autosomal dominant 3
Hemangioma, capillary infantile, somatic 602089 3
Lymphatic malformation 1 153100 Autosomal dominant 3

TEXT

Cloning and Expression

By screening a placenta cDNA library with a mouse Flt3 probe, Galland et al. (1992) isolated a human gene encoding a putative receptor-type tyrosine kinase. The deduced amino acid sequence of the intracellular portion of the molecule showed that it was strongly related to FLT1 (165070) and KDR (191306) and to a lesser degree to members of the class III receptor-type tyrosine kinases: FMS (164770), PDGFR (173490, 173410), KIT (164920), and FLT3 (136351).


Mapping

Galland et al. (1992) mapped FLT4 to chromosome 5q34-q35, telomeric to the FMS and PDGFRB genes, by in situ hybridization. They assigned the mouse homolog to chromosome 11 by the same method. In the process of creating a radiation hybrid map of 18 genes, Warrington et al. (1992) demonstrated that the FLT4 gene is located on distal 5q between GABRA1 (137160) at 5q34-q35 and DRD1 (126449) at 5q35.1. Aprelikova et al. (1992) also mapped the FLT4 gene to chromosome 5q33-qter.


Gene Function

Among the factors stimulating angiogenesis, the acidic and basic fibroblast growth factors FGF1 (131220) and FGF2 (134920) and the vascular endothelial growth factor VEGF (192240) exert their effects via specific cell surface receptor tyrosine kinases: for FGF1 and FGF2, FGF receptor-1 (FGFR1; 136350), also known as FLT2, and the endothelial-specific FMS-like tyrosine kinase-1; and for VEGF, the KDR/FLK1 receptor. The protein product of the FLT4 receptor tyrosine kinase cDNA is structurally similar to the FLT1 and KDR/FLK1 receptors (Pajusola et al., 1992), but FLT4 does not bind VEGF (Pajusola et al., 1994). Lee et al. (1996) identified and characterized a vascular endothelial growth factor-related protein (VEGFC; 601528) that specifically binds to the extracellular domain of Flt4 and stimulates tyrosine phosphorylation and mitogenesis of endothelial cells.

Kaipainen et al. (1995) analyzed the expression of FLT4 by in situ hybridization during mouse embryogenesis and in adult human tissues. The FLT4 mRNA signals first became detectable in the angioblasts of head mesenchyme, the cardinal vein, and extraembryonally in the allantois of 8.5-day postcoitus (p.c.) embryos. In 12.5-day p.c. embryos, the FLT4 signal decorated developing venous and presumptive lymphatic endothelia, but arterial endothelia were negative. During later stages of development, FLT4 mRNA became restricted to vascular plexuses devoid of red cells, representing developing lymphatic vessels. In adult human tissues, only the lymphatic endothelia and some high endothelial venules expressed FLT4 mRNA. Increased expression occurred in lymphatic sinuses in metastatic lymph nodes and in lymphangioma. The results suggested that FLT4 is a marker for lymphatic vessels and some high endothelial venules in human adult tissues. They also supported the theory of the venous origin of lymphatic vessels.

Vascular endothelial growth factor is a key regulator of blood vessel development in embryos and angiogenesis in adult tissues. Unlike VEGF, the related VEGFC stimulates the growth of lymphatic vessels through its specific lymphatic endothelial receptor VEGFR3. Dumont et al. (1998) showed that targeted inactivation of the VEGFR3 gene in mice resulted in defective blood vessel development in early embryos. Vasculogenesis and angiogenesis occurred, but large vessels became abnormally organized with defective lumens, leading to fluid accumulation in the pericardial cavity and cardiovascular failure at embryonic day 9.5. Thus, VEGFR3 has an essential role in the development of the embryonic cardiovascular system before the emergence of the lymphatic vessels.

In studies of the mouse cornea in vivo, Cursiefen et al. (2006) demonstrated that the presence of Vegfr3 in the corneal epithelium is critical for suppressing inflammatory corneal angiogenesis, by acting as a decoy receptor and binding the angiogenic growth factors VEGFC and VEGFD (FIGF; 300091). Cursiefen et al. (2006) concluded that VEGFR3, which provides proangiogenic signaling when expressed on endothelium, may also have antiangiogenic properties when expressed at an avascular site by nonendothelial cells.

In studies in zebrafish to investigate the role of Notch (see 190198) signaling in angiogenesis, Siekmann and Lawson (2007) found that Flt4 is expressed in segmental artery tip cells and becomes ectopically expressed throughout the sprout in the absence of Notch. Loss of Flt4 could partially restore normal endothelial cell number in Rbpsuh (147183)-deficient segmental arteries. Finally, loss of the Notch ligand Dll4 (605185) also led to an increased number of endothelial cells within segmental arteries. Siekmann and Lawson (2007) concluded that their studies taken together indicated that proper specification of cell identity, position, and behavior in a developing blood vessel sprout is required for normal angiogenesis, and implicated the Notch signaling pathway in this process.

Tammela et al. (2008) demonstrated that VEGFR3 is highly expressed in angiogenic sprouts, and that genetic targeting of VEGFR3 or blocking of VEGFR3 signaling with monoclonal antibodies results in decreased sprouting, vascular density, vessel branching, and endothelial cell proliferation in mouse angiogenesis models. Stimulation of VEGFR3 augmented VEGF-induced angiogenesis and sustained angiogenesis even in the presence of VEGFR2 (191306) inhibitors, whereas antibodies against VEGFR3 and VEGFR2 in combination resulted in additive inhibition of angiogenesis and tumor growth. Furthermore, genetic or pharmacologic disruption of the Notch signaling pathway led to widespread endothelial VEGFR3 expression and excessive sprouting, which was inhibited by blocking VEGFR3 signals. Tammela et al. (2008) concluded that their results implicated VEGFR3 as a regulator of vascular network formation. The authors suggested that targeting VEGFR3 may provide additional efficacy for antiangiogenic therapies, especially towards vessels that are resistant to VEGF or VEGFR2 inhibitors.

Wang et al. (2010) demonstrated with genetic experiments in mouse and zebrafish that ephrin-B2 (EFNB2; 600527), a transmembrane ligand for Ephrin receptor tyrosine kinases, promotes sprouting behavior and motility in the angiogenic endothelium. Wang et al. (2010) linked this proangiogenic function to a crucial role of ephrin-B2 in the VEGF signaling pathway, which they studied in detail for VEGFR3, the receptor for VEGF-C. In the absence of ephrin-B2, the internalization of VEGFR3 in cultured cells and mutant mice is defective, which compromises downstream signal transduction by the small GTPase Rac1 (602048), Akt (164730), and the mitogen-activated protein kinase Erk (601795). Wang et al. (2010) concluded that VEGFR3 signaling is coupled to receptor internalization. Ephrin-B2 is a key regulator of this process and thereby controls angiogenic and lymphangiogenic growth.

Benedito et al. (2012) used inducible loss-of-function genetics in combination with inhibitors in vivo to demonstrate that DLL4 (605185) protein expression in retinal tip cells is only weakly modulated by VEGFR2 (191306) signaling. Surprisingly, Notch (190198) inhibition also had no significant impact on VEGFR2 expression and induced deregulated endothelial sprouting and proliferation even in the absence of VEGFR2, which is the most important VEGFA (192240) receptor and is considered to be indispensable for these processes. By contrast, VEGFR3, the main receptor for VEGFC (601528), was strongly modulated by Notch. VEGFR3 kinase activity inhibitors but not ligand-blocking antibodies suppressed the sprouting of endothelial cells that had low Notch signaling activity. Benedito et al. (2012) concluded that their results established that VEGFR2 and VEGFR3 are regulated in a highly differential manner by Notch. They proposed that successful antiangiogenic targeting of these receptors and their ligands will strongly depend on the status of endothelial Notch signaling.

By screening immunity-related proteins and their receptors for bacteria- or LPS-induced expression, Zhang et al. (2014) detected upregulation of VEGFR3 and VEGFC in macrophages. Serum VEGFC was also increased in patient and mouse models of septic shock. Ligation of VEGFR3 by VEGFC attenuated proinflammatory cytokine production. In the absence of either the ligand-binding domain or tyrosine kinase activity of Vegfr3, mice became more sensitive to septic shock. Vegfr3 restrained Tlr4 (603030)-NFKB (see 164011) activation by regulating the PI3K (see 601232)-Akt signaling pathway and Socs1 (603597) expression. Zhang et al. (2014) proposed that in addition to targeting lymphatic vessels, VEGFR3 signaling via VEGFC prevents microphagic overreaction to infections complicated by lymphedema.

Lorenz et al. (2018) used the developing liver as a model organ to study angiocrine signals and showed that the growth rate of the liver correlates both spatially and temporally with blood perfusion to this organ. By manipulating blood flow through the liver vasculature, Lorenz et al. (2018) demonstrated that vessel perfusion activates beta-1 integrin (135630) and VEGFR3. Notably, both beta-1 integrin and VEGFR3 are strictly required for normal production of hepatocyte growth factor, survival of hepatocytes, and liver growth. Ex vivo perfusion of adult mouse liver and in vitro mechanical stretching of human hepatic endothelial cells illustrated that mechanotransduction alone is sufficient to turn on angiocrine signals. When the endothelial cells are mechanically stretched, angiocrine signals trigger in vitro proliferation and survival of primary human hepatocytes. Lorenz et al. (2018) concluded that their findings uncovered a signaling pathway in vascular endothelial cells that translates blood perfusion and mechanotransduction into organ growth and maintenance.


Molecular Genetics

Lymphatic Malformation 1

In affected members of a family with lymphatic malformation-1 (LMPHM1; 153100), Ferrell et al. (1998) identified a mutation in the FLT4 gene (136352.0005).

Karkkainen et al. (2000) identified mutations at the FLT4 locus in several families with hereditary lymphedema. They found that all disease-associated alleles analyzed had missense mutations and encoded proteins with an inactive tyrosine kinase, preventing downstream gene activation. These studies established that vascular endothelial growth factor receptor-3 is important for normal lymphatic vascular function.

In a family with hereditary lymphedema, Irrthum et al. (2000) identified a mutation in the FLT4 gene (136352.0006) that cosegregated with the disease. In vitro expression showed that this mutation inhibited the autophosphorylation of the receptor.

Evans et al. (2003) identified 8 different heterozygous mutations in the FLT4 gene (see, e.g., 136352.0011) in affected members of 12 different Caucasian families with hereditary lymphedema. All the mutations occurred in the tyrosine kinase domains. Several families showed incomplete penetrance of the phenotype.

Kim and Dumont (2003) reviewed molecular mechanisms in lymphangiogenesis and their implications for human disease. In addition to VEGFR3 and FOXC2 (602402), 6 'lymphangiogenic markers' were reviewed. The role of some of these lymphangiogenetic mechanisms in cancer and metastasis was also reviewed.

Ghalamkarpour et al. (2006) identified mutations in the FLT4 gene (see, e.g., 136352.0008-136352.0009) in affected members from 3 unrelated families with autosomal dominant lymphedema and in a sporadic case.

In 14 affected and 2 unaffected members of a 3-generation consanguineous Israeli family of Muslim Arab origin with hereditary lymphedema, Spiegel et al. (2006) identified heterozygosity for a missense mutation in the VEGFR3 gene (136352.0010).

Connell et al. (2009) identified mutations in the FLT4 gene, including 14 novel mutations, in 22 (42%) of 52 patients with primary lymphedema. Mutation prevalence was 75% in patients with a typical Milroy phenotype and a positive family history, and 68% if positive family history was not a diagnostic criterion. No mutations were found outside the kinase domains, showing that analysis of nonkinase domains of FLT4 is not useful for Milroy disease patients. No mutations were identified in the VEGFC gene (601528), which encodes the FLT4 ligand.

In a Hispanic female with congenital lymphedema who was born of first-cousin parents, Ghalamkarpour et al. (2009) identified homozygosity for a missense mutation in the ATP-binding domain of the FLT4 gene (136352.0012). Her unaffected parents were heterozygous for the hypomorphic mutation, which was not found in 110 controls.

Congenital Heart Defects, Multiple Types, 7

In a cohort of 2,871 probands with congenital heart disease, comprising 2,645 parent-offspring trios and 226 singletons, Jin et al. (2017) performed whole-exome sequencing and identified 10 probands who were heterozygous for frameshift or nonsense mutations in the FLT4 gene. In 2 probands the mutations appeared to have arisen de novo and in 2 probands the mutations were inherited from an affected parent; however, in 6 probands, the mutation was inherited from an unaffected parent. Cardiac diagnoses in affected individuals included tetralogy of Fallot, pulmonary stenosis or atresia, absent pulmonary valve, right aortic arch, double aortic arch, and major aortopulmonary collateral arteries. The authors stated that FLT4 mutation carriers had no extracardiac malformations (unspecified), but elsewhere in the text stated that 1 proband had an extracardiac congenital anomaly.

Reuter et al. (2019) sequenced 175 adult patients with tetralogy of Fallot and 56 with other congenital cardiac anomalies for loss-of-function and deleterious mutations in FLT4 and other VEGF pathway genes. They identified 9 (5.1%) probands with novel FLT4 variants, all of whom came from the group of individuals with tetralogy of Fallot. Seven of the variants were predicted to have a loss-of-function effect, implicating haploinsufficiency, and comprised 2 stopgain, 3 frameshift, 1 splice site, and 1 multiexon deletion mutation; the other 2 mutations, a missense and an in-frame deletion, were predicted to be deleterious. In addition to variants in FLT4, multiple variants were also identified in the KDR (191306) gene.


Animal Model

The Chy mouse mutant, characterized by accumulation of chylous ascites and swelling of the limbs, was obtained by ethylnitrosourea-induced mutagenesis (Lyon and Glenister (1984, 1986)). The phenotype is linked to mouse chromosome 11. Karkkainen et al. (2001) sequenced the Vegfr3 candidate gene on chromosome 11 in Chy mice and found a heterozygous 3157A-T mutation resulting in an ile1053-to-phe (I1053F) substitution in the tyrosine kinase domain. This mutation was located in a highly conserved catalytic domain of the receptor, in close proximity to the VEGFR3 mutations in human primary lymphedema. The I1053F mutant receptor was tyrosine kinase inactive. Although lymphedema patients with heterozygous missense mutations of VEGFR3 retain some receptor activity because of the presence of the wildtype allele (Karkkainen et al., 2000), the mutant VEGFR3 can be classified as a dominant-negative receptor similar to certain mutant KIT receptors in piebaldism (172800) and RET receptors (164761) in Hirschsprung disease (142623). Karkkainen et al. (2001) found that neuropilin-2 (NRP2; 602070) bound VEGFC and was expressed in the visceral, but not in the cutaneous, lymphatic endothelia. This may explain why hypoplastic cutaneous, but not visceral, lymphatic vessels were found in the Chy mice. Using virus-mediated VEGFC gene therapy, Karkkainen et al. (2001) generated functional lymphatic vessels in the lymphedema mice. The results suggested that growth factor gene therapy is applicable to human lymphedema as well and provided a paradigm for other diseases associated with mutant receptors, i.e., ligand therapy.


ALLELIC VARIANTS 17 Selected Examples):

.0001   MOVED TO 136352.0005


.0002   LYMPHATIC MALFORMATION 1

FLT4, GLY857ARG
SNP: rs267606818, ClinVar: RCV000017647, RCV003441720

In a family with lymphatic malformation-1 (LMPHM1; 153100) in members of 3 generations, Karkkainen et al. (2000) identified a heterozygous G-A transition in the FLT4 gene, resulting in a gly857-to-arg (G857R) substitution.


.0003   LYMPHATIC MALFORMATION 1

FLT4, ARG1041PRO
SNP: rs121909650, ClinVar: RCV000017648

In a family with hereditary lymphedema (LMPHM1; 153100) in at least 4 generations, Karkkainen et al. (2000) identified a heterozygous mutation in the FLT4 gene, resulting in an arg1041-to-pro (R1041P) substitution.


.0004   LYMPHATIC MALFORMATION 1

FLT4, LEU1044PRO
SNP: rs121909651, ClinVar: RCV000017649

In a large family with autosomal dominant lymphedema (LMPHM1; 153100) in 5 generations and many different sibships, Karkkainen et al. (2000) identified heterozygosity for a transition in the FLT4 gene, resulting in a leu1044-to-pro (L1044P) substitution.


.0005   LYMPHATIC MALFORMATION 1

FLT4, PRO1114LEU
SNP: rs121909652, ClinVar: RCV000017650

In a mother and 2 daughters with primary lymphedema (LMPHM1; 153100), Karkkainen et al. (2000) identified a heterozygous pro1114-to-leu (P1114L) missense mutation of the FLT4 gene.

Ferrell et al. (1998) had originally described the mutation in this family as a 3360G-A transition in the FLT4 gene, predicted to cause a nonconservative PRO1126LEU (P1126L) substitution in the mature receptor (Karkkainen et al., 2000).


.0006   LYMPHATIC MALFORMATION 1

FLT4, HIS1035ARG
SNP: rs121909653, ClinVar: RCV000017651

In a family in which the father and 4 of 7 children had congenital lymphedema (LMPHM1; 153100), Irrthum et al. (2000) demonstrated a heterozygous his1035-to-arg (H1035R) missense mutation in the FLT4 gene.


.0007   HEMANGIOMA, CAPILLARY INFANTILE, SOMATIC

FLT4, PRO954SER
SNP: rs34255532, gnomAD: rs34255532, ClinVar: RCV000017652, RCV000249222, RCV000860875, RCV003323277

In 1 of 15 infantile hemangioma (602089) specimens, Walter et al. (2002) found a pro954-to-ser (P954S) missense mutation in the kinase insert of the FLT4 gene. This result, and the finding of a somatic missense mutation in the VEGFR2 gene (191306.0001) in another of the 15 specimens, suggested that alteration of the FLT4 signaling pathway in endothelial and/or pericytic cells may be a mechanism involved in hemangioma formation.


.0008   LYMPHATIC MALFORMATION 1

FLT4, VAL878MET
SNP: rs121909654, ClinVar: RCV000017653

In 4 affected individuals of a family with autosomal dominant lymphedema (LMPHM1; 153100), Ghalamkarpour et al. (2006) identified a heterozygous 2632G-A transition in the FLT4 gene, resulting in a val878-to-met (V878M) substitution in the tyrosine kinase domain I of the protein. One affected family member was a 22-week-old fetus who was found to have fetal hydrops with bilateral leg edema, pleural effusions, hydrothorax, and pulmonary hypoplasia on ultrasound. The pregnancy was terminated. Other affected family members had congenital lymphedema of the legs with variable severity.


.0009   LYMPHATIC MALFORMATION 1

FLT4, ILE1086THR
SNP: rs121909655, ClinVar: RCV000017654, RCV003398529

In affected members of a family with autosomal dominant lymphedema (LMPHM1; 153100) spanning 5 generations, Ghalamkarpour et al. (2006) identified a heterozygous 3257T-C transition in the FLT4 gene, resulting in an ile1086-to-thr (I1086T) substitution in the tyrosine kinase II domain of the protein. The proband had a severe phenotype with elephantiasis up to the inguinal ligament bilaterally, chronic venous ulcerations, cellulitis, and papillomatosis.


.0010   LYMPHATIC MALFORMATION 1

FLT4, GLU1106LYS
SNP: rs121909656, ClinVar: RCV000017655

In 14 affected and 2 unaffected members of a 3-generation consanguineous Israeli family of Muslim Arab origin with hereditary lymphedema (LMPHM1; 153100), Spiegel et al. (2006) identified heterozygosity for a 3316G-A transition in the FLT4 gene, resulting in a glu1106-to-lys (E1106K) substitution. The mutation was not found in 110 control individuals.


.0011   LYMPHATIC MALFORMATION 1

FLT4, 3-BP DEL, 3323TCT
SNP: rs587776833, ClinVar: RCV000017656, RCV002254267

In affected members of a family with hereditary lymphedema (LMPHM1; 153100), Evans et al. (2003) identified a heterozygous 3-bp in-frame deletion (3323delTCT), resulting in the deletion of residue phe1108 in the tyrosine kinase II domain.


.0012   LYMPHATIC MALFORMATION 1

FLT4, ALA855THR
SNP: rs121909657, gnomAD: rs121909657, ClinVar: RCV000017657, RCV003387726

In a Hispanic female with congenital lymphedema (LMPHM1; 153100) who was born of first-cousin parents, Ghalamkarpour et al. (2009) identified homozygosity for a 2563G-A transition in exon 18 of the FLT4 gene, resulting in an ala855-to-thr (A855T) substitution in the ATP-binding domain of the receptor. The unaffected parents were heterozygous for the mutation, which was not found in 110 controls. Assessment of receptor function showed reduced phosphorylation of the receptor, with impaired ligand-induced internalization and defective downstream signaling.


.0013   CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, TYR361TER
ClinVar: RCV001003426

In a mother and son (family 1-04970), both with tetralogy of Fallot (CHTD7; 618780), Jin et al. (2017) identified a tyrosine-to-stop substitution at codon 361 of the FLT4 protein (Y361X). The son additionally had right-sided aortic arch. Hamosh (2020) noted that this variant was absent from the gnomAD database on February 17, 2020.


.0014   CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, 2-BP DEL
SNP: rs755445139, gnomAD: rs755445139, ClinVar: RCV001003427, RCV001585912

In 2 unrelated probands (1-03410 and 1-05967) with tetralogy of Fallot, pulmonary atresia, and major aortopulmonary collateral arteries (CHTD7; 618780), Jin et al. (2017) identified deletion of a C nucleotide in the FLT4 gene resulting in a proline-to-arginine substitution at codon 30, followed by a premature termination codon 3 amino acids downstream (Pro30ArgfsTer3). In family 1-03410, the mutation occurred as a de novo event. The proband also had right-sided aortic arch. In family 1-05967, the proband had inherited the mutation from her unaffected mother. She additionally had double aortic arch.

Reuter et al. (2019) identified this mutation in a patient (CGC-034) with tetralogy of Fallot, pulmonary atresia, and major aortopulmonary collateral arteries, as well as congenital lymphedema. She had inherited the mutation from her mother, who had normal echocardiography results. The mother's father had bradycardia. Reuter et al. (2019) recorded the mutation as c.89delC (c.89delC, NM_182925.4).


.0015   CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, EX25-29DEL
ClinVar: RCV001003428

In a 23-year-old female (TOF293) with tetralogy of Fallot, right-sided aortic arch, and absent pulmonary valve (CHTD7; 618780), Reuter et al. (2019) identified heterozygous deletion of exons 25 through 29 of the FLT4 gene (chr5:180,031,767-180,040,470del).


.0016   CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, GLN1192TER
SNP: rs1581616817, ClinVar: RCV001003429

In a 79-year-old man (TOF158) with tetralogy of Fallot, right-sided aortic arch, and atrial flutter requiring ablation (CHTD7; 618780), and in his daughter with tetralogy of Fallot, Reuter et al. (2019) identified a C-to-T transition at nucleotide 3574 (c.3574C-T, NM_182925.4) of the FLT4 gene, resulting in a glutamine-to-termination substitution at codon 1192 (Q1192X). The variant was absent from gnomAD.


.0017   CONGENITAL HEART DEFECTS, MULTIPLE TYPES, 7

FLT4, 1-BP DUP, 1622G
SNP: rs1581655293, ClinVar: RCV001003430

In a 29-year-old man (TOF284) with tetralogy of Fallot, major aortopulmonary collateral arteries, and aortic valve replacement (CHTD7; 618780), Reuter et al. (2019) identified duplication of a G at nucleotide 1622 (c.1622dupG, NM_182925.4) of the FLT4 gene, resulting in a frameshift and premature termination (Gln542ProfsTer3). The variant was absent from gnomAD.


See Also:

Milroy (1892); Offori et al. (1993)

REFERENCES

  1. Aprelikova, O., Pajusola, K., Partanen, J., Armstrong, E., Alitalo, R., Bailey, S. K., McMahon, J., Wasmuth, J., Huebner, K., Alitalo, K. FLT4, a novel class III receptor tyrosine kinase in chromosome 5q33-qter. Cancer Res. 52: 746-748, 1992. [PubMed: 1310071]

  2. Benedito, R., Rocha, S. F., Woeste, M., Zamykal, M., Radtke, F., Casanovas, O., Duarte, A., Pytowski, B., Adams, R. H. Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF-VEGFR2 signalling. Nature 484: 110-114, 2012. [PubMed: 22426001] [Full Text: https://doi.org/10.1038/nature10908]

  3. Connell, F. C., Ostergaard, P., Carver, C., Brice, G., Williams, N., Mansour, S., Mortimer, P. S., Jeffery, S., Lymphoedema Consortium. Analysis of the coding regions of VEGFR3 and VEGFC in Milroy disease and other primary lymphoedemas. Hum. Genet. 124: 625-631, 2009. Note: Erratum: Hum. Genet. 125: 237 only, 2009. [PubMed: 19002718] [Full Text: https://doi.org/10.1007/s00439-008-0586-5]

  4. Cursiefen, C., Chen, L., Saint-Geniez, M., Hamrah, P., Jin, Y., Rashid, S., Pytowksi, B., Persaud, K., Wu, Y., Streilein, J. W., Dana, R. Nonvascular VEGF receptor 3 expression by corneal epithelium maintains avascularity and vision. Proc. Nat. Acad. Sci. 103: 11405-11410, 2006. [PubMed: 16849433] [Full Text: https://doi.org/10.1073/pnas.0506112103]

  5. Dumont, D. J., Jussila, L., Taipale, J., Lymboussaki, A., Mustonen, T., Pajusola, K., Breitman, M., Alitalo, K. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science 282: 946-949, 1998. [PubMed: 9794766] [Full Text: https://doi.org/10.1126/science.282.5390.946]

  6. Evans, A. L., Bell, R., Brice, G., Comeglio, P., Lipede, C., Jeffery, S., Mortimer, P., Sarfarazi, M., Child, A. H. Identification of eight novel VEGFR-3 mutations in families with primary lymphoedema. J. Med. Genet. 40: 697-703, 2003. [PubMed: 12960217] [Full Text: https://doi.org/10.1136/jmg.40.9.697]

  7. 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. Molec. Genet. 7: 2073-2078, 1998. [PubMed: 9817924] [Full Text: https://doi.org/10.1093/hmg/7.13.2073]

  8. Galland, F., Karamysheva, A., Mattei, M.-G., Rosnet, O., Marchetto, S., Birnbaum, D. Chromosomal localization of FLT4, a novel receptor-type tyrosine kinase gene. Genomics 13: 475-478, 1992. [PubMed: 1319394] [Full Text: https://doi.org/10.1016/0888-7543(92)90277-y]

  9. 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. 46: 399-404, 2009. [PubMed: 19289394] [Full Text: https://doi.org/10.1136/jmg.2008.064469]

  10. 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. 70: 330-335, 2006. [PubMed: 16965327] [Full Text: https://doi.org/10.1111/j.1399-0004.2006.00687.x]

  11. Hamosh, A. Personal Communication. Baltimore, Md. 02/17/2020.

  12. 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. 67: 295-301, 2000. [PubMed: 10856194] [Full Text: https://doi.org/10.1086/303019]

  13. Jin, S. C., Homsy, J., Zaidi, S., Lu, Q., Morton, S., DePalma, S. R., Zeng, X., Qi, H., Chang, W., Sierant, M. C., Hung, W.-C., Haider, S., and 33 others. Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands. Nature Genet. 49: 1593-1601, 2017. [PubMed: 28991257] [Full Text: https://doi.org/10.1038/ng.3970]

  14. Kaipainen, A., Korhonen, J., Mustonen, T., van Hinsbergh, V. W. M., Fang, G.-H., Dumont, D., Breitman, M., Alitalo, K. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc. Nat. Acad. Sci. 92: 3566-3570, 1995. [PubMed: 7724599] [Full Text: https://doi.org/10.1073/pnas.92.8.3566]

  15. 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. Nature Genet. 25: 153-159, 2000. [PubMed: 10835628] [Full Text: https://doi.org/10.1038/75997]

  16. Karkkainen, M. J., Saaristo, A., Jussila, L., Karila, K. A., Lawrence, E. C., Pajusola, K., Bueler, H., Eichmann, A., Kauppinen, R., Kettunen, M. I., Yla-Herttuala, S., Finegold, D. N., Ferrell, R. E., Alitalo, K. A model for gene therapy of human hereditary lymphedema. Proc. Nat. Acad. Sci. 98: 12677-12682, 2001. [PubMed: 11592985] [Full Text: https://doi.org/10.1073/pnas.221449198]

  17. Kim, H., Dumont, D. J. Molecular mechanisms in lymphangiogenesis: model systems and implications in human disease. Clin. Genet. 64: 282-292, 2003. [PubMed: 12974730] [Full Text: https://doi.org/10.1034/j.1399-0004.2003.00152.x]

  18. Lee, J., Gray, A., Yuan, J., Luoh, S.-M., Avraham, H., Wood, W. I. Vascular endothelial growth factor-related protein: a ligand and specific activator of the tyrosine kinase receptor Flt4. Proc. Nat. Acad. Sci. 93: 1988-1992, 1996. [PubMed: 8700872] [Full Text: https://doi.org/10.1073/pnas.93.5.1988]

  19. Lorenz, L., Axnick, J., Buschmann, T., Henning, C., Urner, S., Fang, S., Nurmi, H., Eichhorst, N., Holtmeier, R., Bodis, K., Hwang, J.-H., Mussig, K., Eberhard, D., Stypmann, J., Kuss, O., Roden, M., Alitalo, K., Haussinger, D., Lammert, E. Mechanosensing by beta-1 integrin induces angiocrine signals for liver growth and survival. Nature 562: 128-132, 2018. [PubMed: 30258227] [Full Text: https://doi.org/10.1038/s41586-018-0522-3]

  20. Lyon, M. F., Glenister, P. H. New Mutants. Mouse Newsletter 71: 26 only, 1984.

  21. Lyon, M. F., Glenister, P. H. Gene order of Chy-vt-Re on chromosome 11. Mouse Newsletter 74: 96 only, 1986.

  22. Milroy, W. F. An undescribed variety of hereditary oedema. New York Med. J. 56: 505-508, 1892.

  23. Offori, T. W., Platt, C. C., Stephens, M., Hopkinson, G. B. Angiosarcoma in congenital hereditary lymphoedema (Milroy's disease): diagnostic beacons and a review of the literature. Clin. Exp. Derm. 18: 174-177, 1993. [PubMed: 8482001] [Full Text: https://doi.org/10.1111/j.1365-2230.1993.tb01008.x]

  24. Pajusola, K., Aprelikova, O., Korhonen, J., Kaipainen, A., Pertovaara, L., Alitalo, R., Alitalo, K. FLT4 receptor tyrosine kinase contains seven immunoglobulin-like loops and is expressed in multiple human tissues and cell lines. Cancer Res. 52: 5738-5743, 1992. Note: Erratum: Cancer Res. 53: 3845 only, 1993. [PubMed: 1327515]

  25. Pajusola, K., Aprelikova, O., Pelicci, G., Weich, H., Claesson-Welsh, L., Alitalo, K. Signalling properties of FLT4, a proteolytically processed receptor tyrosine kinase related to two VEGF receptors. Oncogene 9: 3545-3555, 1994. [PubMed: 7970715]

  26. Reuter, M. S., Jobling, R., Chaturvedi, R. R., Manshaei, R., Costain, G., Heung, T., Curtis, M., Hosseini, S. M., Liston, E., Lowther, C., Oechslin, E., Sticht, H., and 9 others. Haploinsufficiency of vascular endothelial growth factor related signaling genes is associated with tetralogy of Fallot. Genet. Med. 21: 1001-1007, 2019. [PubMed: 30232381] [Full Text: https://doi.org/10.1038/s41436-018-0260-9]

  27. Siekmann, A. F., Lawson, N. D. Notch signalling limits angiogenic cell behaviour in developing zebrafish arteries. Nature 445: 781-784, 2007. [PubMed: 17259972] [Full Text: https://doi.org/10.1038/nature05577]

  28. Spiegel, R., Ghalamkarpour, A., Daniel-Spiegel, E., Vikkula, M., Shalev, S. A. Wide clinical spectrum in a family with hereditary lymphedema type I due to a novel missense mutation in VEGFR3. J. Hum. Genet. 51: 846-850, 2006. [PubMed: 16924388] [Full Text: https://doi.org/10.1007/s10038-006-0031-3]

  29. Tammela, T., Zarkada, G., Wallgard, E., Murtomaki, A., Suchting, S., Wirzenius, M., Waltari, M., Hellstrom, M., Schomber, T., Peltonen, R., Freitas, C., Duarte, A., and 9 others. Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation. Nature 454: 656-660, 2008. [PubMed: 18594512] [Full Text: https://doi.org/10.1038/nature07083]

  30. Walter, J. W., North, P. E., Waner, M., Mizeracki, A., Blei, F., Walker, J. W. T., Reinisch, J. F., Marchuk, D. A. Somatic mutation of vascular endothelial growth factor receptors in juvenile hemangioma. Genes Chromosomes Cancer 33: 295-303, 2002. [PubMed: 11807987] [Full Text: https://doi.org/10.1002/gcc.10028]

  31. Wang, Y., Nakayama, M., Pitulescu, M. E., Schmidt, T. S., Bochenek, M. L., Sakakibara, A., Adams, S., Davy, A., Deutsch, U., Luthi, U., Barberis, A., Benjamin, L. E., Makinen, T., Nobes, C. D., Adams, R. H. Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Nature 465: 483-486, 2010. [PubMed: 20445537] [Full Text: https://doi.org/10.1038/nature09002]

  32. Warrington, J. A., Bailey, S. K., Armstrong, E., Aprelikova, O., Alitalo, K., Dolganov, G. M., Wilcox, A. S., Sikela, J. M., Wolfe, S. F., Lovett, M., Wasmuth, J. J. A radiation hybrid map of 18 growth factor, growth factor receptor, hormone receptor, or neurotransmitter receptor genes on the distal region of the long arm of chromosome 5. Genomics 13: 803-808, 1992. [PubMed: 1322355] [Full Text: https://doi.org/10.1016/0888-7543(92)90156-m]

  33. Zhang, Y., Lu, Y., Ma, L., Cao, X., Xiao, J., Chen, J., Jiao, S., Gao, Y., Liu, C., Duan, Z., Li, D., He, Y., Wei, B., Wang, H. Activation of vascular endothelial growth factor receptor-3 in macrophages restrains TLR4-NF-kappa-beta signaling and protects against endotoxin shock. Immunity 40: 501-514, 2014. [PubMed: 24656836] [Full Text: https://doi.org/10.1016/j.immuni.2014.01.013]


Contributors:
Ada Hamosh - updated : 02/17/2020
Ada Hamosh - updated : 09/13/2019
Marla J. F. O'Neill - updated : 04/17/2018
Paul J. Converse - updated : 9/19/2014
Ada Hamosh - updated : 4/24/2012
Ada Hamosh - updated : 6/30/2010
Cassandra L. Kniffin - updated : 6/10/2009
Cassandra L. Kniffin - updated : 2/25/2009
Ada Hamosh - updated : 10/20/2008
Cassandra L. Kniffin - updated : 4/18/2008
Ada Hamosh - updated : 6/26/2007
Marla J. F. O'Neill - updated : 12/29/2006
Cassandra L. Kniffin - updated : 11/1/2006
Marla J. F. O'Neill - updated : 9/29/2006
Cassandra L. Kniffin - reorganized : 11/19/2003
Victor A. McKusick - updated : 11/4/2003
Victor A. McKusick - updated : 3/14/2002
Victor A. McKusick - updated : 1/14/2002
Victor A. McKusick - updated : 10/3/2000
Victor A. McKusick - updated : 5/25/2000
Victor A. McKusick - updated : 2/10/1999
Victor A. McKusick - updated : 1/6/1999
Victor A. McKusick - updated : 11/10/1998
Victor A. McKusick - updated : 10/27/1998

Creation Date:
Victor A. McKusick : 6/1/1992

Edit History:
alopez : 06/25/2020
alopez : 02/18/2020
carol : 02/18/2020
alopez : 02/17/2020
alopez : 02/17/2020
alopez : 09/13/2019
carol : 12/18/2018
alopez : 11/07/2018
carol : 04/18/2018
carol : 04/17/2018
mgross : 09/29/2014
carol : 9/22/2014
mcolton : 9/19/2014
carol : 1/30/2013
carol : 1/29/2013
carol : 1/2/2013
alopez : 4/25/2012
terry : 4/24/2012
alopez : 6/30/2010
terry : 6/30/2010
wwang : 9/28/2009
wwang : 7/1/2009
ckniffin : 6/10/2009
wwang : 6/1/2009
ckniffin : 5/20/2009
wwang : 3/5/2009
ckniffin : 2/25/2009
alopez : 10/22/2008
terry : 10/20/2008
wwang : 4/18/2008
alopez : 7/2/2007
terry : 6/26/2007
wwang : 12/29/2006
ckniffin : 11/6/2006
carol : 11/6/2006
ckniffin : 11/1/2006
wwang : 9/29/2006
wwang : 9/29/2006
carol : 11/19/2003
ckniffin : 11/12/2003
tkritzer : 11/6/2003
terry : 11/4/2003
cwells : 3/20/2002
cwells : 3/18/2002
terry : 3/14/2002
terry : 3/8/2002
carol : 1/20/2002
mcapotos : 1/14/2002
mcapotos : 1/14/2002
carol : 10/5/2000
terry : 10/3/2000
alopez : 5/25/2000
alopez : 11/18/1999
terry : 3/9/1999
carol : 3/3/1999
carol : 2/10/1999
terry : 2/10/1999
carol : 1/18/1999
terry : 1/7/1999
terry : 1/6/1999
carol : 11/18/1998
carol : 11/17/1998
terry : 11/16/1998
terry : 11/10/1998
dkim : 11/6/1998
alopez : 10/29/1998
terry : 10/27/1998
dkim : 7/2/1998
carol : 7/20/1992
carol : 6/1/1992