Entry - *601614 - NETRIN 1; NTN1 - OMIM

 
* 601614

NETRIN 1; NTN1


Alternative titles; symbols

NETRIN 1, MOUSE, HOMOLOG OF
NETRIN 1-LIKE; NTN1L


HGNC Approved Gene Symbol: NTN1

Cytogenetic location: 17p13.1     Genomic coordinates (GRCh38): 17:9,003,087-9,244,000 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
17p13.1 Mirror movements 4 618264 AD 3

TEXT

Description

The NTN1 gene encodes netrin-1, an extracellular secreted protein that mediates axon guidance in the spinal cord during development. It is also involved in synaptogenesis, apoptosis, cell migration, and angiogenesis (summary by Meneret et al., 2017).

Embryologic experiments in both vertebrates and invertebrates provide evidence that developing axons are guided to their targets in the nervous system by the combined actions of attractive and repulsive guidance cues (Tessier-Lavigne and Goodman, 1996). One family of putative guidance cues for developing axons are the netrins, large (approximately 70-80 kD), soluble proteins that show homology in their amino termini to portions of the extracellular matrix molecule laminin (see 150320) and that have been implicated in axon guidance through distinct and complementary lines of evidence in worms, flies, and vertebrates. Netrin-1 is a diffusible protein made by floor plate cells; it can attract spinal commissural axons and repel trochlear axons in vitro (summary by Serafini et al., 1996).


Cloning and Expression

Serafini et al. (1996) isolated cDNA clones comprising the entire coding region of murine netrin-1. PCR was performed on reverse-transcribed mouse brain poly(A)+ RNA using primers based upon sequence homologies between chick netrin-1 and C. elegans Unc6.

Using degenerate PCR primers derived from highly conserved C-terminal domains of chicken netrin-1, netrin-2 (see 602349), and Unc6 proteins, followed by screening of a human brainstem cDNA library, Meyerhardt et al. (1999) isolated NTN1 cDNA. The NTN1 gene encodes a 604-amino acid protein with 98% identity to mouse netrin-1 and 50% identity to C. elegans Unc6. Northern blot analysis detected a 5.0-kb NTN1 transcript in most normal adult human tissues studied. Markedly reduced or absent NTN1 expression was seen in approximately 50% of brain tumors and neuroblastomas by RNase protection assays.


Gene Function

Using Xenopus, Hopker et al. (1999) demonstrated that laminin-1-beta (150240) from the extracellular matrix converts netrin-mediated attraction into repulsion. A soluble peptide fragment of laminin-1-beta (YIGSR) mimicked this laminin-induced conversion. Low levels of cAMP in growth cones also led to the conversion of netrin-induced attraction into repulsion, and Hopker et al. (1999) showed that the amount of cAMP decreases in the presence of laminin-1 or YIGSR, suggesting a possible mechanism for laminin's effect. At the netrin-1-rich optic nerve head, where axons turn sharply to leave the eye, laminin-1 is confined to the retinal surface. Repulsion from the region in which laminin and netrin are coexpressed may help to drive axons into the region where only netrin is present, providing a mechanism for their escape from the retinal surface. Hopker et al. (1999) concluded that extracellular matrix molecules not only promote axon outgrowth, but also modify the behavior of growth cones in response to diffusible guidance cues.

Meyerhardt et al. (1999) showed that netrin-1 protein could be crosslinked to DCC (120470) protein on the cell surface, but did not immunoprecipitate with DCC in the absence of crosslinking and failed to bind to a soluble fusion protein containing the entire DCC extracellular domain. Meyerhardt et al. (1999) concluded that the binding of netrin-1 to DCC appears to depend on the presence of a coreceptor or accessory proteins.

The axonal chemoattractant netrin-1 guides spinal commissural axons by activating its receptor DCC. Galko and Tessier-Lavigne (2000) found that chemical inhibitors of metalloproteases potentiate netrin-mediated axon outgrowth in vitro. Galko and Tessier-Lavigne (2000) also found that DCC is a substrate for metalloprotease-dependent ectodomain shedding, and that the inhibitors block proteolytic processing of DCC and cause an increase in DCC protein levels on axons within spinal cord explants. Thus, Galko and Tessier-Lavigne (2000) suggested that potentiation of netrin activity by inhibitors may result from stabilization of DCC on the axons, and proteolytic activity may regulate axon migration by controlling the number of functional extracellular axon guidance receptors.

Corset et al. (2000) showed that DCC interacts with the membrane-associated adenosine A2b receptor (600466), a G protein-coupled receptor that induces cAMP accumulation on binding adenosine. Corset et al. (2000) showed that adenosine A2b receptor is actually a netrin-1 receptor and induces cAMP accumulation on binding netrin-1, and that netrin-1-dependent outgrowth of dorsal spinal cord axons directly involves A2b. Corset et al. (2000) concluded that the growth-promoting function of netrin-1 may require a receptor complex containing DCC and A2b.

Stein et al. (2001) demonstrated that netrin-1 binds DCC and that the DCC cytoplasmic domain fused to a heterologous receptor ectodomain can mediate guidance through a mechanism involving derepression of cytoplasmic domain multimerization. Activation of the adenosine A2B receptor, proposed to contribute to netrin effects on axons, is not required for rat commissural axon outgrowth or Xenopus spinal axon attraction to netrin-1. Thus, Stein et al. (2001) concluded that DCC plays a central role in netrin signaling of axon growth and guidance independent of A2B receptor activation. Note that an expression of concern was published for the article by Stein et al. (2001).

Axonal growth cones that cross the nervous system midline change their responsiveness to midline guidance cues: they become repelled by the repellent Slit (603746) and simultaneously lose responsiveness to the attractant netrin. These mutually reinforcing changes help to expel growth cones from the midline by making a once-attractive environment appear repulsive. Stein and Tessier-Lavigne (2001) provided evidence that these 2 changes are causally linked: in the growth cones of embryonic Xenopus spinal axons, activation of the Slit receptor Roundabout (Robo; 602430) silences the attractive effect of netrin-1, but not its growth-stimulatory effect, through direct binding of the cytoplasmic domain of Robo to that of the netrin receptor DCC. Biologically, this hierarchical silencing mechanism helps to prevent a tug-of-war between attractive and repulsive signals in the growth cone that might cause confusion. Molecularly, silencing is enabled by a modular and interlocking design of the cytoplasmic domains of these potentially antagonistic receptors that predetermines the outcome of their simultaneous activation.

In a 'whole-optic pathway' preparation in Xenopus devised to assess the behavior of retinal growth cones at 4 defined points along the optic pathway, Shewan et al. (2002) found a gradual change in axonal response to netrin-1, from attraction to repulsion at progressively distal points along the pathway. Axons aged in culture underwent similar changes which correlated with a decline in cAMP and netrin-1 receptor (ADORA2B; 600446) expression, suggesting that responsiveness is intrinsically and developmentally regulated, and also suggesting a possible molecular basis for the altered responsiveness.

Forcet et al. (2002) showed that in embryonic kidney cells expressing full-length, but not cytoplasmic domain-truncated, DCC (120470), NTN1 causes increased transient phosphorylation and activity of ERK1 (601795) and ERK2 (176948), but not of JNK1 (601158), JNK2 (602896), or p38 (MAPK14; 600289). This phosphorylation was mediated by MEK1 (MAP2K1; 176872) and/or MEK2 (MAP2K2; 601263). NTN1 also activated the transcription factor ELK1 (311040) and serum response element-regulated gene expression. Immunoprecipitation analysis showed interaction of full-length DCC with MEK1/2 in the presence or absence of NTN1. Forcet et al. (2002) showed that activation of Dcc by Ntn1 in rat embryonic day-13 dorsal spinal cord stimulates and is required for the outgrowth of commissural axons and Erk1/2 activation. Immunohistochemical analysis demonstrated expression of activated Erk1/2 in embryonic commissural axons, and this expression was diminished in Dcc or Ntn1 knockout animals. Forcet et al. (2002) concluded that the MAPK pathway is involved in responses to NTN1 and proposed that ERK activation affects axonal growth by phosphorylation of microtubule-associated proteins and neurofilaments.

Ming et al. (2002) found that axonal growth cones of cultured frog spinal neurons exhibit adaptation during chemotactic migration, undergoing consecutive phases of desensitization and resensitization in the presence of increasing basal concentrations of the guidance factors Ntn1 or Bdnf1 (113505). Ntn1- or Bdnf1-specific desensitization was accompanied by a reduction of calcium signaling, whereas resensitization required activation of Mapk and local protein synthesis. Ming et al. (2002) suggested that the protracted nature of this process allows adaptive changes in the sensitivity of the growth cone and that the adaptive behavior exemplifies the image of the growth cone as a chemotaking amoeba, as proposed in the 19th century by Ramon y Cajal.

Nishiyama et al. (2003) reported that the ratio of cyclic AMP to cyclic GMP activities sets the polarity of netrin-1-induced axon guidance: high ratios favor attraction, whereas low ratios favor repulsion. Whole-cell recordings of calcium currents in Xenopus spinal neuron growth cones indicated that cyclic nucleotide signaling directly modulates the activity of L-type calcium channels in axonal growth cones. Furthermore, cyclic GMP signaling activated by an arachidonate 12-lipoxygenase metabolite suppressed L-type calcium channel activity triggered by netrin-1, and was required for growth cone repulsion mediated by the DCC-UNC5 (see 603610) receptor complex. By linking cyclic AMP and cyclic GMP signaling and modulation of calcium channel activity in growth cones, these findings delineated an early membrane-associated event responsible for signal transduction during bidirectional axon guidance.

Mehlen et al. (1998) showed that DCC induces apoptosis conditionally: by functioning as a dependence receptor, DCC induces apoptosis unless it is engaged by its ligand netrin-1. Mazelin et al. (2004) demonstrated that inhibition of cell death by enforced expression of netrin-1 in mouse gastrointestinal tract led to the spontaneous formation of hyperplastic and neoplastic lesions. Moreover, in the adenomatous polyposis coli mutant background associated with adenoma formation, enforced expression of netrin-1 engendered aggressive adenocarcinomatous malignancies. Mazelin et al. (2004) concluded that netrin-1 can promote intestinal tumor development, probably by regulating cell survival. Thus, a netrin-1 receptor or receptors function as conditional tumor suppressors.

Blood vessels and nerves often follow with parallel trajectories, suggesting that distal targets use common cues that induce vascularization and innervation. Netrins are secreted by the floor plate and attract commissural axons toward the midline of the neural tube. Park et al. (2004) showed that netrin-1 is also a potent vascular mitogen. Netrin-1 stimulates proliferation, induces migration, and promotes adhesion of endothelial cells and vascular smooth muscle cells with a specific activity comparable to vascular endothelial growth factor (PEGF; 192240) and platelet-derived growth factor (PDGF; see 173430). The authors presented evidence indicating that the netrin receptor neogenin (NEO1; 601907) mediates netrin signaling in vascular smooth muscle cells, but suggested that an unidentified receptor mediates the proangiogenic effects of netrin-1 on endothelial cells. Netrin-1 also stimulates angiogenesis in vivo and augments the response to vascular endothelial growth factor. Park et al. (2004) concluded that netrin-1 is a secreted neural guidance cue with the unique ability to attract both blood vessels and axons and that other cues may also function as vascular endothelial growth factors.

Netrin proteins play a role in the developing nervous system by promoting both axonal outgrowth and axonal guidance in pathfinding. Liu et al. (2004), Li et al. (2004), and Ren et al. (2004) simultaneously reported a complex network of intracellular signaling downstream from netrin-1 involving DCC (120470), focal adhesion kinase (FAK; 600758), and FYN (137025), a member of the SRC family kinases (see 190090). In neurons cultured from rat cerebral cortex, Liu et al. (2004) found that netrin-1 induced tyrosine phosphorylation of FAK and FYN, and coimmunoprecipitation studies showed direct interaction of FAK and FYN with DCC. Inhibition of FYN inhibited FAK phosphorylation, and FYN mutants inhibited the attractive turning responses to netrin. Neurons lacking the FAK gene showed reduced axonal outgrowth and attractive turning responses to netrin. In cultured neurons from chick and mouse, Li et al. (2004) found that netrin increased tyrosine phosphorylation of DCC and FAK. Coimmunoprecipitation studies showed that DCC interacted directly with FAK and SRC to form a complex and that FAK and SRC cooperated to stimulate DCC phosphorylation by SRC. Li et al. (2004) suggested that phosphorylated DCC acts as a kinase-coupled receptor and that FAK and SRC act downstream of DCC in netrin signaling. Ren et al. (2004) found that inhibition of FAK phosphorylation inhibited netrin-1-induced axonal outgrowth and guidance. The authors suggested that FAK may also function as a scaffolding protein and play a role in cytoskeletal reorganization that is necessary for neurite outgrowth and turning.

Lu et al. (2004) showed that the repulsive netrin receptor UNC5B (607870) is expressed by endothelial tip cells of the vascular system. Disruption of the Unc5b gene in mice or of Unc5b or netrin-1A in zebrafish led to aberrant extension of endothelial tip cell filopodia, excessive vessel branching, and abnormal navigation. Netrin-1 caused endothelial filopodial retraction, but only when Unc5b was present. Thus, Lu et al. (2004) concluded that UNC5B functions as a repulsive netrin receptor in endothelial cells controlling morphogenesis of the vascular system. An Editorial Expression of Concern was published for the article by Lu et al. (2004) because image integrity issues had been raised about some of the images. The editors found that some panels of Supplementary Figure 2E appeared to have been duplicated, but noted that the authors provided contemporaneous replicates that confirmed the validity of the data presented in the figure.

Larrivee et al. (2007) found that Unc5b was downregulated in quiescent adult mouse vasculature, but it was reexpressed during sprouting angiogenesis in 3-dimensional gels and in tumor implants. Stimulation of Unc5b-expressing neovessels with netrin-1 inhibited sprouting angiogenesis, and genetic loss of Unc5b reduced netrin-1-mediated inhibition of angiogenesis. Expression of Unc5b triggered endothelial cell repulsion in response to netrin-1 in vitro, whereas a truncated Unc5b lacking the intracellular signaling domain failed to induce repulsion. Larrivee et al. (2007) concluded that activation of UNC5B by netrin-1 inhibits sprouting angiogenesis.

Wang and Poo (2005) reported that TRP (transient receptor potential)-like channel activity exists in the growth cones of cultured Xenopus neurons and can be modulated by exposure to netrin-1 and brain-derived neurotrophic factor (BDNF; 113505), 2 chemoattractants for axon guidance. Whole-cell recording from growth cones showed that netrin-1 induced a membrane depolarization, part of which remained after all major voltage-dependent channels were blocked. Furthermore, the membrane depolarization was sensitive to blockers of TRP channels. Pharmacologic blockade of putative TRP currents or downregulation of Xenopus TRP1 (xTRPC1) expression with a specific morpholino oligonucleotide abolished the growth cone turning and calcium ion elevation induced by a netrin-1 gradient. Thus, Wang and Poo (2005) concluded that TRPC currents reflect early events in the growth cone's detection of some extracellular guidance signal, resulting in membrane depolarization and cytoplasmic calcium ion elevation that mediates the turning of growth cones.

Wilson et al. (2006) demonstrated that netrins stimulate proliferation, migration, and tube formation of human endothelial cells in vitro and that this stimulation is independent of known netrin receptors. Suppression of netrin-1a mRNA in zebrafish inhibited vascular sprouting, implying a proangiogenic role for netrins during vertebrate development. Wilson et al. (2006) also showed that netrins accelerate neovascularization in an in vivo model of ischemia and that they reverse neuropathy and vasculopathy in a diabetic murine model. Wilson et al. (2006) found that vectors expressing netrin-1 and netrin-4 (NTN4; 610401) are comparable in effectiveness to vectors expressing VEGF (192240) and promoting neovascularization and reperfusion.

Colon-Ramos et al. (2007) showed that connectivity between 2 interneurons in C. elegans, AIY and RIA, is orchestrated by a pair of glial cells that express UNC6 (netrin-1). In the postsynaptic neuron RIA, the netrin receptor UNC40 (DCC; 120470) plays a conventional guidance role, directing outgrowth of the RIA process ventrally toward the glia. The authors demonstrated that in the presynaptic neuron AIY, UNC40 plays an unexpected and theretofore uncharacterized role: it cell-autonomously promotes assembly of presynaptic terminals in the immediate vicinity of the glial cell endfeet. Colon-Ramos et al. (2007) concluded that netrin can be used both for guidance and local synaptogenesis and suggested that glial cells can function as guideposts during the assembly of neural circuits in vivo.

Using cultured rat and frog spinal neurons, Ly et al. (2008) showed that Dscam (602523) could mediate the turning response to netrin-1 both alone and in collaboration with the netrin receptor Dcc.

Using primary neurons derived from rat, mouse, and chicken embryos, Furne et al. (2008) showed that Ntn1, in addition to behaving as an attractive cue, functioned in cell survival by inhibiting the proapoptotic activity of Dcc.

By quantitative RT-PCR of 51 primary breast tumors, Fitamant et al. (2008) found significantly increased expression of netrin-1 in a high proportion of lymph node-positive tumors and tumors from metastatic patients. Netrin-1-expressing mammary metastatic tumor cell lines underwent apoptosis when netrin-1 expression was experimentally decreased or when decoy soluble netrin receptor ectodomains were added. Such treatments prevented lung metastasis of a mammary cancer cell line and a xenografted human breast tumor in mice. Fitamant et al. (2008) concluded that netrin-1 expression confers a selective advantage for tumor cell survival.

Moore et al. (2009) showed that traction on immobilized netrin-1 was sufficient to reorient axons.

Delloye-Bourgeois et al. (2009) showed that autocrine production of NTN1 conveyed a selective advantage in tumor growth and dissemination in aggressive forms of neuroblastoma (NB; see 256700) by blocking the proapoptotic activity of the UNC5H receptors. NTN1 upregulation appeared to be a marker for poor prognosis in infants with stage 4S and stage 4 NB. NTN1 disruption blocked NB metastasis in animals, and Delloye-Bourgeois et al. (2009) proposed disrupting NTN1 expression as an anticancer strategy.


Biochemical Features

Crystal Structure

Xu et al. (2014) determined the structures of a functional NTN1 region, alone and in complexes with NEO1 (601907) and DCC (120470). NTN1 has a rigid elongated structure containing 2 receptor-binding sites at opposite ends through which it brings together receptor molecules. The ligand/receptor complexes reveal 2 distinct architectures: a 2:2 heterotetramer and a continuous ligand/receptor assembly. The differences result from different lengths of the linker connecting receptor domains fibronectin type III domain 4 (FN4) and FN5, which differs among DCC and NEO1 splice variants, providing a basis for diverse signaling outcomes.


Mapping

Meyerhardt et al. (1999) mapped the human NTN1 gene to chromosome 17p13-p12 by FISH.


Molecular Genetics

In members of 2 unrelated families and in an unrelated patient with mirror movements-4 (MRMV4; 618264), Meneret et al. (2017) identified heterozygous mutations in the NTN1 gene (601614.0001-601614.0003). The mutations, which were found by exome sequencing and confirmed by Sanger sequencing, segregated with the condition in the 2 families; the patient in the third family had sporadic occurrence and the mutation occurred de novo. All mutations occurred in the NTR domain and were predicted to affect protein structure and/or stability. In vitro functional expression studies in HeLa and HEK293 cells showed that unlike wildtype, the mutant proteins were almost exclusively found in the intracellular compartments and could not be detected in the supernatant. Since netrin-1 is a diffusible extracellular cue, the pathophysiology probably involved a loss of function. The patients were ascertained from a cohort of 47 index cases with congenital mirror movements in whom mutations in other MRMV-associated genes were excluded.


Animal Model

Serafini et al. (1996) used the method of Skarnes et al. (1995) in embryonic stem cells to selectively recover mutations in genes encoding proteins with signal sequences by virtue of creating fusions of N-terminal portions of these proteins with an exogenous transmembrane domain. In this way Serafini et al. (1996) isolated a mutated murine netrin-1 gene and transmitted it through the germline. Homozygotes were born but apparently did not suckle and died within a few days. Netrin-1-deficient mice exhibited defects in spinal commissural axon projections that were considered consistent with netrin-1 guiding these axons. Defects in several forebrain commissures were also observed, suggesting additional guidance roles for netrin-1.

Poon et al. (2008) demonstrated that the axon guidance cue unc6/netrin and its receptor unc5 (607869) act throughout development to exclude synaptic vesicle and active zone proteins from the dendrite of the C. elegans motor neuron DA9, which is proximal to a source of unc6/netrin. In unc6/netrin and unc5 loss-of-function mutants, presynaptic components mislocalized to the DA9 dendrite. In addition, ectopically expressed unc6/netrin, acting through unc5, was sufficient to exclude endogenous synapses from adjacent subcellular domains within the DA9 axon. Furthermore, this antisynaptogenic activity was interchangeable with that of lin44/Wnt despite being transduced through different receptors, suggesting that extracellular cues such as netrin and Wnts not only guide axon navigation but also regulate the polarized accumulation of presynaptic components through local exclusion.


ALLELIC VARIANTS ( 3 Selected Examples):

.0001 MIRROR MOVEMENTS 4

NTN1, CYS601ARG
  
RCV000735954

In 3 affected members of a French family (family 1) with mirror movements-4 (MRMV4; 618264), Meneret et al. (2017) identified a heterozygous c.1801T-C transition (c.1801T-C, NM_004822) in exon 3 of the NTN1 gene, resulting in a cys601-to-arg (C601R) substitution at a highly conserved residue in the netrin domain. The mutation, which was found by exome sequencing, was not found in the ExAC database. There were 2 asymptomatic mutation carriers, indicating incomplete penetrance. In vitro functional expression studies in HeLa and HEK293 cells showed that unlike wildtype, the mutant protein was almost exclusively found in the intracellular compartments and could not be detected in the supernatant.


.0002 MIRROR MOVEMENTS 4

NTN1, 3-BP DEL, NT1552
  
RCV000735955

In 3 affected members of a family (family 2) with mirror movements-4 (MRMV4; 618264), Meneret et al. (2017) identified a heterozygous 3-bp deletion (c.1552_1554del, NM_004822) in exon 3 of the NTN1 gene, resulting in the deletion of conserved residue ile518 (Ile518del) in the netrin domain. The mutation, which was found by exome sequencing, was not found in the ExAC database. In vitro functional expression studies in HeLa and HEK293 cells showed that unlike wildtype, the mutant protein was almost exclusively found in the intracellular compartments and could not be detected in the supernatant. This family was previously reported as family C in Franz et al. (2015).


.0003 MIRROR MOVEMENTS 4

NTN1, CYS601SER
  
RCV000735956

In a patient (family 3) with sporadic occurrence of mirror movements-4 (MRMV4; 618264), Meneret et al. (2017) identified a heterozygous c.1802G-C transversion (c.1802G-C, NM_004822) in exon 3 of the NTN1 gene, resulting in a cys601-to-ser (C601S) substitution at a highly conserved residue in the netrin domain. The mutation, which was found by exome sequencing, was not found in the ExAC database. In vitro functional expression studies in HeLa and HEK293 cells showed that unlike wildtype, the mutant protein was almost exclusively found in the intracellular compartments and could not be detected in the supernatant.


REFERENCES

  1. Colon-Ramos, D. A., Margeta, M. A., Shen, K. Glia promote local synaptogenesis through UNC-6 (netrin) signaling in C. elegans. Science 318: 103-106, 2007. [PubMed: 17916735, images, related citations] [Full Text]

  2. Corset, V., Nguyen-Ba-Charvet, K. T., Forcet, C., Moyse, E., Chedotal, A., Mehlen, P. Netrin-1-mediated axon outgrowth and cAMP production requires interaction with adenosine A2b receptor. Nature 407: 747-750, 2000. [PubMed: 11048721, related citations] [Full Text]

  3. Delloye-Bourgeois, C., Fitamant, J., Paradisi, A., Cappellen, D., Douc-Rasy, S., Raquin, M.-A., Stupack, D., Nakagawara, A., Rousseau, R., Combaret, V., Puisieux, A., Valteau-Couanet, D., Benard, J., Bernet, A., Mehlen, P. Netrin-1 acts as a survival factor for aggressive neuroblastoma. J. Exp. Med. 206: 833-847, 2009. [PubMed: 19349462, images, related citations] [Full Text]

  4. Fitamant, J., Guenebeaud, C., Coissieux, M.-M., Guix, C., Treilleux, I., Scoazec, J.-Y., Bachelot, T., Bernet, A., Mehlen, P. Netrin-1 expression confers a selective advantage for tumor cell survival in metastatic breast cancer. Proc. Nat. Acad. Sci. 105: 4850-4855, 2008. [PubMed: 18353983, images, related citations] [Full Text]

  5. Forcet, C., Stein, E., Pays, L., Corset, V., Llambi, F., Tessier-Lavigne, M., Mehlen, P. Netrin-1-mediated axon outgrowth requires deleted in colorectal cancer-dependent MAPK activation. Nature 417: 443-447, 2002. [PubMed: 11986622, related citations] [Full Text]

  6. Franz, E. A., Chiaroni-Clarke, R., Woodrow, S., Glendining, K. A., Jasoni, C. L., Robertson, S. P., Gardner, R. J. M., Markie, D. Congenital mirror movements: phenotypes associated with DCC and RAD51 mutations. J. Neurol. Sci. 351: 140-145, 2015. [PubMed: 25813273, related citations] [Full Text]

  7. Furne, C., Rama, N., Corset, V., Chedotal, A., Mehlen, P. Netrin-1 is a survival factor during commissural neuron navigation. Proc. Nat. Acad. Sci. 105: 14465-14470, 2008. [PubMed: 18796601, images, related citations] [Full Text]

  8. Galko, M. J., Tessier-Lavigne, M. Function of an axonal chemoattractant modulated by metalloprotease activity. Science 289: 1365-1367, 2000. [PubMed: 10958786, related citations] [Full Text]

  9. Hopker, V. H., Shewan, D., Tessier-Lavigne, M., Poo, M., Holt, C. Growth-cone attraction to netrin-1 is converted to repulsion by laminin-1. Nature 401: 69-73, 1999. [PubMed: 10485706, related citations] [Full Text]

  10. Larrivee, B., Freitas, C., Trombe, M., Lv, X., DeLafarge, B., Yuan, L., Bouvree, K., Breant, C., Del Toro, R., Brechot, N., Germain, S., Bono, F., Dol, F., Claes, F., Fischer, C., Autiero, M., Thomas, J.-L., Carmeliet, P., Tessier-Lavigne, M., Eichmann, A. Activation of the UNC5B receptor by netrin-1 inhibits sprouting angiogenesis. Genes Dev. 21: 2433-2447, 2007. [PubMed: 17908930, images, related citations] [Full Text]

  11. Li, W., Lee, J., Vikis, H. G., Lee, S.-H., Liu, G., Aurandt, J., Shen, T.-L., Fearon, E. R., Guan, J.-L., Han, M., Rao, Y., Hong, K., Guan, K.-L. Activation of FAK and Src are receptor-proximal events required for netrin signaling. Nature Neurosci. 7: 1213-1221, 2004. [PubMed: 15494734, images, related citations] [Full Text]

  12. Liu, G., Beggs, H., Jurgensen, C., Park, H.-T., Tang, H., Gorski, J., Jones, K. R., Reichardt, L. F., Wu, J., Rao, Y. Netrin requires focal adhesion kinase and Src family kinases for axon outgrowth and attraction. Nature Neurosci. 7: 1222-1232, 2004. [PubMed: 15494732, images, related citations] [Full Text]

  13. Lu, X., le Noble, F., Yuan, L., Jiang, Q., de Lafarge, B., Sugiyama, D., Breant, C., Claes, F., De Smet, F., Thomas, J.-L., Autiero, M., Carmeliet, P., Tessier-Lavigne, M., Eichmann, A. The netrin receptor UNC5B mediates guidance events controlling morphogenesis of the vascular system. Nature 432: 179-186, 2004. Note: Editorial Expression of Concern. Nature 625: E12, 2024. [PubMed: 15510105, related citations] [Full Text]

  14. Ly, A., Nikolaev, A., Suresh, G., Zheng, Y., Tessier-Lavigne, M., Stein, E. DSCAM is a netrin receptor that collaborates with DCC in mediating turning responses to netrin-1. Cell 133: 1241-1254, 2008. [PubMed: 18585357, images, related citations] [Full Text]

  15. Mazelin, L., Bernet, A., Bonod-Bidaud, C., Pays, L., Arnaud, S., Gespach, C., Bredesen, D. E., Scoazec, J.-Y., Mehlen, P. Netrin-1 controls colorectal tumorigenesis by regulating apoptosis. Nature 431: 80-84, 2004. [PubMed: 15343335, related citations] [Full Text]

  16. Mehlen, P., Rabizadeh, S., Snipas, S. J., Assa-Munt, N., Salvesen, G. S., Bredesen, D. E. The DCC gene product induces apoptosis by a mechanism requiring receptor proteolysis. Nature 395: 801-804, 1998. [PubMed: 9796814, related citations] [Full Text]

  17. Meneret, A., Franz, E. A., Trouillard, O., Oliver, T. C., Zagar, Y., Robertson, S. P., Weiniarz, Q., Gardner, R. J. M., Gallea, C., Srour, M., Depienne, C., Jasoni, C. L., and 15 others. Mutations in the netrin-1 gene cause congenital mirror movements. J. Clin. Invest. 127: 3923-3936, 2017. [PubMed: 28945198, images, related citations] [Full Text]

  18. Meyerhardt, J. A., Caca, K., Eckstrand, B. C., Hu, G., Lengauer, C., Banavali, S., Look, A. T., Fearon, E. R. Netrin-1: interaction with deleted in colorectal cancer (DCC) and alterations in brain tumors and neuroblastomas. Cell Growth Diff. 10: 35-42, 1999. [PubMed: 9950216, related citations]

  19. Ming, G., Wong, S. T., Henley, J., Yuan, X., Song, H., Spitzer, N. C., Poo, M. Adaptation in the chemotactic guidance of nerve growth cones. Nature 417: 411-418, 2002. [PubMed: 11986620, related citations] [Full Text]

  20. Moore, S. W., Biais, N., Sheetz, M. P. Traction on immobilized netrin-1 is sufficient to reorient axons. Science 325: 166 only, 2009. [PubMed: 19589994, related citations] [Full Text]

  21. Nishiyama, M., Hoshino, A., Tsai, L., Henley, J. R., Goshima, Y., Tessier-Lavigne, M., Poo, M., Hong, K. Cyclic AMP/GMP-dependent modulation of Ca(2+) channels sets the polarity of nerve growth-cone turning. Nature 423: 990-995, 2003. [PubMed: 12827203, related citations] [Full Text]

  22. Park, K. W., Crouse, D., Lee, M., Karnik, S. K., Sorensen, L. K., Murphy, K. J., Kuo, C. J., Li, D. Y. The axonal attractant netrin-1 is an angiogenic factor. Proc. Nat. Acad. Sci. 101: 16210-16215, 2004. [PubMed: 15520390, images, related citations] [Full Text]

  23. Poon, V. Y., Klassen, M. P., Shen, K. UNC-6/netrin and its receptor UNC-5 locally exclude presynaptic components from dendrites. Nature 455: 669-673, 2008. [PubMed: 18776887, images, related citations] [Full Text]

  24. Ren, X., Ming, G., Xie, Y., Hong, Y., Sun, D., Zhao, Z., Feng, Z., Wang, Q., Shim, S., Chen, Z., Song, H., Mei, L., Xiong, W. Focal adhesion kinase in netrin-1 signaling. Nature Neurosci. 7: 1204-1212, 2004. [PubMed: 15494733, related citations] [Full Text]

  25. Serafini, T., Colamarino, S. A., Leonardo, E. D., Wang, H., Beddington, R., Skarnes, W. C., Tessier-Lavigne, M. Netrin-1 is required for commissural axon guidance in the developing vertebrate nervous system. Cell 87: 1001-1014, 1996. [PubMed: 8978605, related citations] [Full Text]

  26. Shewan, D., Dwivedy, A., Anderson, R., Holt, C. E. Age-related changes underlie switch in netrin-1 responsiveness as growth cones advance along visual pathway. Nature Neurosci. 5: 955-962, 2002. [PubMed: 12352982, related citations] [Full Text]

  27. Skarnes, W. C., Moss, J. E., Hurtley, S. M., Beddington, R. S. P. Capturing genes encoding membrane and secreted proteins important for mouse development. Proc. Nat. Acad. Sci. 92: 6592-6596, 1995. [PubMed: 7604039, related citations] [Full Text]

  28. Stein, E., Tessier-Lavigne, M. Hierarchical organization of guidance receptors: silencing of netrin attraction by Slit through a Robo/DCC receptor complex. Science 291: 1928-1938, 2001. Note: Expression of Concern: Science 378: 1284 only, 2022. [PubMed: 11239147, related citations] [Full Text]

  29. Stein, E., Zou, Y., Poo, M., Tessier-Lavigne, M. Binding of DCC by netrin-1 to mediate axon guidance independent of adenosine A2B receptor activation. Science 291: 1976-1982, 2001. Note: Expression of Concern: Science 378: 1284 only, 2022. [PubMed: 11239160, related citations] [Full Text]

  30. Tessier-Lavigne, M., Goodman, C. S. The molecular biology of axon guidance. Science 274: 1123-1133, 1996. [PubMed: 8895455, related citations] [Full Text]

  31. Wang, G. X., Poo, M. Requirement of TRPC channels in netrin-1-induced chemotropic turning of nerve growth cones. Nature 434: 898-904, 2005. [PubMed: 15758951, related citations] [Full Text]

  32. Wilson, B. D., Li, M., Park, K. W., Suli, A., Sorensen, L. K., Larrieu-Lahargue, F., Urness, L. D., Suh, W., Asai, J., Kock, G. A. H., Thorne, T., Silver, M., Thomas, K. R., Chien, C.-B., Losordo, D. W., Li, D. Y. Netrins promote developmental and therapeutic angiogenesis. Science 313: 640-644, 2006. [PubMed: 16809490, related citations] [Full Text]

  33. Xu, K., Wu, Z., Renier, N., Antipenko, A., Tzvetkova-Robev, D., Xu, Y., Minchenko, M., Nardi-Dei, V., Rajashankar, K. R., Himanen, J., Tessier-Lavigne, M., Nikolov, D. B. Structures of netrin-1 bound to two receptors provide insight into its axon guidance mechanism. Science 344: 1275-1279, 2014. [PubMed: 24876346, images, related citations] [Full Text]


Cassandra L. Kniffin - updated : 01/02/2019
Ada Hamosh - updated : 7/7/2014
Paul J. Converse - updated : 11/12/2009
Patricia A. Hartz - updated : 8/31/2009
Ada Hamosh - updated : 8/14/2009
Ada Hamosh - updated : 11/5/2008
Patricia A. Hartz - updated : 9/10/2008
Patricia A. Hartz - updated : 8/13/2008
Patricia A. Hartz - updated : 11/2/2007
Ada Hamosh - updated : 10/26/2007
Ada Hamosh - updated : 6/27/2007
Anne M. Stumpf - updated : 9/8/2006
Ada Hamosh - updated : 9/6/2006
Ada Hamosh - updated : 9/19/2005
Cassandra L. Kniffin - updated : 2/8/2005
Victor A. McKusick - updated : 12/30/2004
Ada Hamosh - updated : 11/10/2004
Ada Hamosh - updated : 7/8/2003
Cassandra L. Kniffin - updated : 9/30/2002
Paul J. Converse - updated : 5/6/2002
Ada Hamosh - updated : 3/27/2001
Ada Hamosh - updated : 3/27/2001
Ada Hamosh - updated : 10/18/2000
Ada Hamosh - updated : 9/5/2000
Ada Hamosh - updated : 2/14/2000
Creation Date:
Victor A. McKusick : 1/8/1997
carol : 04/16/2024
carol : 04/15/2024
carol : 01/26/2024
carol : 01/21/2023
carol : 01/09/2019
carol : 01/08/2019
ckniffin : 01/02/2019
carol : 04/24/2018
alopez : 07/07/2014
alopez : 7/7/2014
carol : 6/20/2012
terry : 1/20/2010
mgross : 11/17/2009
terry : 11/12/2009
mgross : 9/4/2009
terry : 8/31/2009
alopez : 8/18/2009
terry : 8/14/2009
alopez : 11/18/2008
alopez : 11/18/2008
terry : 11/5/2008
mgross : 9/12/2008
terry : 9/10/2008
mgross : 8/13/2008
mgross : 11/2/2007
terry : 11/2/2007
alopez : 11/1/2007
alopez : 11/1/2007
terry : 10/26/2007
alopez : 7/5/2007
terry : 6/27/2007
wwang : 9/13/2006
alopez : 9/13/2006
alopez : 9/8/2006
alopez : 9/8/2006
terry : 9/6/2006
alopez : 9/20/2005
terry : 9/19/2005
wwang : 2/17/2005
ckniffin : 2/8/2005
tkritzer : 1/21/2005
terry : 12/30/2004
tkritzer : 11/10/2004
mgross : 7/14/2003
terry : 7/8/2003
tkritzer : 6/19/2003
alopez : 11/4/2002
carol : 10/21/2002
ckniffin : 9/30/2002
alopez : 6/6/2002
mgross : 5/6/2002
alopez : 3/27/2001
alopez : 3/27/2001
alopez : 10/18/2000
alopez : 9/5/2000
alopez : 2/14/2000
alopez : 4/30/1999
alopez : 6/22/1998
alopez : 6/22/1998
jamie : 1/17/1997
mark : 1/16/1997
jamie : 1/15/1997
jamie : 1/9/1997
jamie : 1/9/1997
mark : 1/8/1997

* 601614

NETRIN 1; NTN1


Alternative titles; symbols

NETRIN 1, MOUSE, HOMOLOG OF
NETRIN 1-LIKE; NTN1L


HGNC Approved Gene Symbol: NTN1

Cytogenetic location: 17p13.1     Genomic coordinates (GRCh38): 17:9,003,087-9,244,000 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
17p13.1 Mirror movements 4 618264 Autosomal dominant 3

TEXT

Description

The NTN1 gene encodes netrin-1, an extracellular secreted protein that mediates axon guidance in the spinal cord during development. It is also involved in synaptogenesis, apoptosis, cell migration, and angiogenesis (summary by Meneret et al., 2017).

Embryologic experiments in both vertebrates and invertebrates provide evidence that developing axons are guided to their targets in the nervous system by the combined actions of attractive and repulsive guidance cues (Tessier-Lavigne and Goodman, 1996). One family of putative guidance cues for developing axons are the netrins, large (approximately 70-80 kD), soluble proteins that show homology in their amino termini to portions of the extracellular matrix molecule laminin (see 150320) and that have been implicated in axon guidance through distinct and complementary lines of evidence in worms, flies, and vertebrates. Netrin-1 is a diffusible protein made by floor plate cells; it can attract spinal commissural axons and repel trochlear axons in vitro (summary by Serafini et al., 1996).


Cloning and Expression

Serafini et al. (1996) isolated cDNA clones comprising the entire coding region of murine netrin-1. PCR was performed on reverse-transcribed mouse brain poly(A)+ RNA using primers based upon sequence homologies between chick netrin-1 and C. elegans Unc6.

Using degenerate PCR primers derived from highly conserved C-terminal domains of chicken netrin-1, netrin-2 (see 602349), and Unc6 proteins, followed by screening of a human brainstem cDNA library, Meyerhardt et al. (1999) isolated NTN1 cDNA. The NTN1 gene encodes a 604-amino acid protein with 98% identity to mouse netrin-1 and 50% identity to C. elegans Unc6. Northern blot analysis detected a 5.0-kb NTN1 transcript in most normal adult human tissues studied. Markedly reduced or absent NTN1 expression was seen in approximately 50% of brain tumors and neuroblastomas by RNase protection assays.


Gene Function

Using Xenopus, Hopker et al. (1999) demonstrated that laminin-1-beta (150240) from the extracellular matrix converts netrin-mediated attraction into repulsion. A soluble peptide fragment of laminin-1-beta (YIGSR) mimicked this laminin-induced conversion. Low levels of cAMP in growth cones also led to the conversion of netrin-induced attraction into repulsion, and Hopker et al. (1999) showed that the amount of cAMP decreases in the presence of laminin-1 or YIGSR, suggesting a possible mechanism for laminin's effect. At the netrin-1-rich optic nerve head, where axons turn sharply to leave the eye, laminin-1 is confined to the retinal surface. Repulsion from the region in which laminin and netrin are coexpressed may help to drive axons into the region where only netrin is present, providing a mechanism for their escape from the retinal surface. Hopker et al. (1999) concluded that extracellular matrix molecules not only promote axon outgrowth, but also modify the behavior of growth cones in response to diffusible guidance cues.

Meyerhardt et al. (1999) showed that netrin-1 protein could be crosslinked to DCC (120470) protein on the cell surface, but did not immunoprecipitate with DCC in the absence of crosslinking and failed to bind to a soluble fusion protein containing the entire DCC extracellular domain. Meyerhardt et al. (1999) concluded that the binding of netrin-1 to DCC appears to depend on the presence of a coreceptor or accessory proteins.

The axonal chemoattractant netrin-1 guides spinal commissural axons by activating its receptor DCC. Galko and Tessier-Lavigne (2000) found that chemical inhibitors of metalloproteases potentiate netrin-mediated axon outgrowth in vitro. Galko and Tessier-Lavigne (2000) also found that DCC is a substrate for metalloprotease-dependent ectodomain shedding, and that the inhibitors block proteolytic processing of DCC and cause an increase in DCC protein levels on axons within spinal cord explants. Thus, Galko and Tessier-Lavigne (2000) suggested that potentiation of netrin activity by inhibitors may result from stabilization of DCC on the axons, and proteolytic activity may regulate axon migration by controlling the number of functional extracellular axon guidance receptors.

Corset et al. (2000) showed that DCC interacts with the membrane-associated adenosine A2b receptor (600466), a G protein-coupled receptor that induces cAMP accumulation on binding adenosine. Corset et al. (2000) showed that adenosine A2b receptor is actually a netrin-1 receptor and induces cAMP accumulation on binding netrin-1, and that netrin-1-dependent outgrowth of dorsal spinal cord axons directly involves A2b. Corset et al. (2000) concluded that the growth-promoting function of netrin-1 may require a receptor complex containing DCC and A2b.

Stein et al. (2001) demonstrated that netrin-1 binds DCC and that the DCC cytoplasmic domain fused to a heterologous receptor ectodomain can mediate guidance through a mechanism involving derepression of cytoplasmic domain multimerization. Activation of the adenosine A2B receptor, proposed to contribute to netrin effects on axons, is not required for rat commissural axon outgrowth or Xenopus spinal axon attraction to netrin-1. Thus, Stein et al. (2001) concluded that DCC plays a central role in netrin signaling of axon growth and guidance independent of A2B receptor activation. Note that an expression of concern was published for the article by Stein et al. (2001).

Axonal growth cones that cross the nervous system midline change their responsiveness to midline guidance cues: they become repelled by the repellent Slit (603746) and simultaneously lose responsiveness to the attractant netrin. These mutually reinforcing changes help to expel growth cones from the midline by making a once-attractive environment appear repulsive. Stein and Tessier-Lavigne (2001) provided evidence that these 2 changes are causally linked: in the growth cones of embryonic Xenopus spinal axons, activation of the Slit receptor Roundabout (Robo; 602430) silences the attractive effect of netrin-1, but not its growth-stimulatory effect, through direct binding of the cytoplasmic domain of Robo to that of the netrin receptor DCC. Biologically, this hierarchical silencing mechanism helps to prevent a tug-of-war between attractive and repulsive signals in the growth cone that might cause confusion. Molecularly, silencing is enabled by a modular and interlocking design of the cytoplasmic domains of these potentially antagonistic receptors that predetermines the outcome of their simultaneous activation.

In a 'whole-optic pathway' preparation in Xenopus devised to assess the behavior of retinal growth cones at 4 defined points along the optic pathway, Shewan et al. (2002) found a gradual change in axonal response to netrin-1, from attraction to repulsion at progressively distal points along the pathway. Axons aged in culture underwent similar changes which correlated with a decline in cAMP and netrin-1 receptor (ADORA2B; 600446) expression, suggesting that responsiveness is intrinsically and developmentally regulated, and also suggesting a possible molecular basis for the altered responsiveness.

Forcet et al. (2002) showed that in embryonic kidney cells expressing full-length, but not cytoplasmic domain-truncated, DCC (120470), NTN1 causes increased transient phosphorylation and activity of ERK1 (601795) and ERK2 (176948), but not of JNK1 (601158), JNK2 (602896), or p38 (MAPK14; 600289). This phosphorylation was mediated by MEK1 (MAP2K1; 176872) and/or MEK2 (MAP2K2; 601263). NTN1 also activated the transcription factor ELK1 (311040) and serum response element-regulated gene expression. Immunoprecipitation analysis showed interaction of full-length DCC with MEK1/2 in the presence or absence of NTN1. Forcet et al. (2002) showed that activation of Dcc by Ntn1 in rat embryonic day-13 dorsal spinal cord stimulates and is required for the outgrowth of commissural axons and Erk1/2 activation. Immunohistochemical analysis demonstrated expression of activated Erk1/2 in embryonic commissural axons, and this expression was diminished in Dcc or Ntn1 knockout animals. Forcet et al. (2002) concluded that the MAPK pathway is involved in responses to NTN1 and proposed that ERK activation affects axonal growth by phosphorylation of microtubule-associated proteins and neurofilaments.

Ming et al. (2002) found that axonal growth cones of cultured frog spinal neurons exhibit adaptation during chemotactic migration, undergoing consecutive phases of desensitization and resensitization in the presence of increasing basal concentrations of the guidance factors Ntn1 or Bdnf1 (113505). Ntn1- or Bdnf1-specific desensitization was accompanied by a reduction of calcium signaling, whereas resensitization required activation of Mapk and local protein synthesis. Ming et al. (2002) suggested that the protracted nature of this process allows adaptive changes in the sensitivity of the growth cone and that the adaptive behavior exemplifies the image of the growth cone as a chemotaking amoeba, as proposed in the 19th century by Ramon y Cajal.

Nishiyama et al. (2003) reported that the ratio of cyclic AMP to cyclic GMP activities sets the polarity of netrin-1-induced axon guidance: high ratios favor attraction, whereas low ratios favor repulsion. Whole-cell recordings of calcium currents in Xenopus spinal neuron growth cones indicated that cyclic nucleotide signaling directly modulates the activity of L-type calcium channels in axonal growth cones. Furthermore, cyclic GMP signaling activated by an arachidonate 12-lipoxygenase metabolite suppressed L-type calcium channel activity triggered by netrin-1, and was required for growth cone repulsion mediated by the DCC-UNC5 (see 603610) receptor complex. By linking cyclic AMP and cyclic GMP signaling and modulation of calcium channel activity in growth cones, these findings delineated an early membrane-associated event responsible for signal transduction during bidirectional axon guidance.

Mehlen et al. (1998) showed that DCC induces apoptosis conditionally: by functioning as a dependence receptor, DCC induces apoptosis unless it is engaged by its ligand netrin-1. Mazelin et al. (2004) demonstrated that inhibition of cell death by enforced expression of netrin-1 in mouse gastrointestinal tract led to the spontaneous formation of hyperplastic and neoplastic lesions. Moreover, in the adenomatous polyposis coli mutant background associated with adenoma formation, enforced expression of netrin-1 engendered aggressive adenocarcinomatous malignancies. Mazelin et al. (2004) concluded that netrin-1 can promote intestinal tumor development, probably by regulating cell survival. Thus, a netrin-1 receptor or receptors function as conditional tumor suppressors.

Blood vessels and nerves often follow with parallel trajectories, suggesting that distal targets use common cues that induce vascularization and innervation. Netrins are secreted by the floor plate and attract commissural axons toward the midline of the neural tube. Park et al. (2004) showed that netrin-1 is also a potent vascular mitogen. Netrin-1 stimulates proliferation, induces migration, and promotes adhesion of endothelial cells and vascular smooth muscle cells with a specific activity comparable to vascular endothelial growth factor (PEGF; 192240) and platelet-derived growth factor (PDGF; see 173430). The authors presented evidence indicating that the netrin receptor neogenin (NEO1; 601907) mediates netrin signaling in vascular smooth muscle cells, but suggested that an unidentified receptor mediates the proangiogenic effects of netrin-1 on endothelial cells. Netrin-1 also stimulates angiogenesis in vivo and augments the response to vascular endothelial growth factor. Park et al. (2004) concluded that netrin-1 is a secreted neural guidance cue with the unique ability to attract both blood vessels and axons and that other cues may also function as vascular endothelial growth factors.

Netrin proteins play a role in the developing nervous system by promoting both axonal outgrowth and axonal guidance in pathfinding. Liu et al. (2004), Li et al. (2004), and Ren et al. (2004) simultaneously reported a complex network of intracellular signaling downstream from netrin-1 involving DCC (120470), focal adhesion kinase (FAK; 600758), and FYN (137025), a member of the SRC family kinases (see 190090). In neurons cultured from rat cerebral cortex, Liu et al. (2004) found that netrin-1 induced tyrosine phosphorylation of FAK and FYN, and coimmunoprecipitation studies showed direct interaction of FAK and FYN with DCC. Inhibition of FYN inhibited FAK phosphorylation, and FYN mutants inhibited the attractive turning responses to netrin. Neurons lacking the FAK gene showed reduced axonal outgrowth and attractive turning responses to netrin. In cultured neurons from chick and mouse, Li et al. (2004) found that netrin increased tyrosine phosphorylation of DCC and FAK. Coimmunoprecipitation studies showed that DCC interacted directly with FAK and SRC to form a complex and that FAK and SRC cooperated to stimulate DCC phosphorylation by SRC. Li et al. (2004) suggested that phosphorylated DCC acts as a kinase-coupled receptor and that FAK and SRC act downstream of DCC in netrin signaling. Ren et al. (2004) found that inhibition of FAK phosphorylation inhibited netrin-1-induced axonal outgrowth and guidance. The authors suggested that FAK may also function as a scaffolding protein and play a role in cytoskeletal reorganization that is necessary for neurite outgrowth and turning.

Lu et al. (2004) showed that the repulsive netrin receptor UNC5B (607870) is expressed by endothelial tip cells of the vascular system. Disruption of the Unc5b gene in mice or of Unc5b or netrin-1A in zebrafish led to aberrant extension of endothelial tip cell filopodia, excessive vessel branching, and abnormal navigation. Netrin-1 caused endothelial filopodial retraction, but only when Unc5b was present. Thus, Lu et al. (2004) concluded that UNC5B functions as a repulsive netrin receptor in endothelial cells controlling morphogenesis of the vascular system. An Editorial Expression of Concern was published for the article by Lu et al. (2004) because image integrity issues had been raised about some of the images. The editors found that some panels of Supplementary Figure 2E appeared to have been duplicated, but noted that the authors provided contemporaneous replicates that confirmed the validity of the data presented in the figure.

Larrivee et al. (2007) found that Unc5b was downregulated in quiescent adult mouse vasculature, but it was reexpressed during sprouting angiogenesis in 3-dimensional gels and in tumor implants. Stimulation of Unc5b-expressing neovessels with netrin-1 inhibited sprouting angiogenesis, and genetic loss of Unc5b reduced netrin-1-mediated inhibition of angiogenesis. Expression of Unc5b triggered endothelial cell repulsion in response to netrin-1 in vitro, whereas a truncated Unc5b lacking the intracellular signaling domain failed to induce repulsion. Larrivee et al. (2007) concluded that activation of UNC5B by netrin-1 inhibits sprouting angiogenesis.

Wang and Poo (2005) reported that TRP (transient receptor potential)-like channel activity exists in the growth cones of cultured Xenopus neurons and can be modulated by exposure to netrin-1 and brain-derived neurotrophic factor (BDNF; 113505), 2 chemoattractants for axon guidance. Whole-cell recording from growth cones showed that netrin-1 induced a membrane depolarization, part of which remained after all major voltage-dependent channels were blocked. Furthermore, the membrane depolarization was sensitive to blockers of TRP channels. Pharmacologic blockade of putative TRP currents or downregulation of Xenopus TRP1 (xTRPC1) expression with a specific morpholino oligonucleotide abolished the growth cone turning and calcium ion elevation induced by a netrin-1 gradient. Thus, Wang and Poo (2005) concluded that TRPC currents reflect early events in the growth cone's detection of some extracellular guidance signal, resulting in membrane depolarization and cytoplasmic calcium ion elevation that mediates the turning of growth cones.

Wilson et al. (2006) demonstrated that netrins stimulate proliferation, migration, and tube formation of human endothelial cells in vitro and that this stimulation is independent of known netrin receptors. Suppression of netrin-1a mRNA in zebrafish inhibited vascular sprouting, implying a proangiogenic role for netrins during vertebrate development. Wilson et al. (2006) also showed that netrins accelerate neovascularization in an in vivo model of ischemia and that they reverse neuropathy and vasculopathy in a diabetic murine model. Wilson et al. (2006) found that vectors expressing netrin-1 and netrin-4 (NTN4; 610401) are comparable in effectiveness to vectors expressing VEGF (192240) and promoting neovascularization and reperfusion.

Colon-Ramos et al. (2007) showed that connectivity between 2 interneurons in C. elegans, AIY and RIA, is orchestrated by a pair of glial cells that express UNC6 (netrin-1). In the postsynaptic neuron RIA, the netrin receptor UNC40 (DCC; 120470) plays a conventional guidance role, directing outgrowth of the RIA process ventrally toward the glia. The authors demonstrated that in the presynaptic neuron AIY, UNC40 plays an unexpected and theretofore uncharacterized role: it cell-autonomously promotes assembly of presynaptic terminals in the immediate vicinity of the glial cell endfeet. Colon-Ramos et al. (2007) concluded that netrin can be used both for guidance and local synaptogenesis and suggested that glial cells can function as guideposts during the assembly of neural circuits in vivo.

Using cultured rat and frog spinal neurons, Ly et al. (2008) showed that Dscam (602523) could mediate the turning response to netrin-1 both alone and in collaboration with the netrin receptor Dcc.

Using primary neurons derived from rat, mouse, and chicken embryos, Furne et al. (2008) showed that Ntn1, in addition to behaving as an attractive cue, functioned in cell survival by inhibiting the proapoptotic activity of Dcc.

By quantitative RT-PCR of 51 primary breast tumors, Fitamant et al. (2008) found significantly increased expression of netrin-1 in a high proportion of lymph node-positive tumors and tumors from metastatic patients. Netrin-1-expressing mammary metastatic tumor cell lines underwent apoptosis when netrin-1 expression was experimentally decreased or when decoy soluble netrin receptor ectodomains were added. Such treatments prevented lung metastasis of a mammary cancer cell line and a xenografted human breast tumor in mice. Fitamant et al. (2008) concluded that netrin-1 expression confers a selective advantage for tumor cell survival.

Moore et al. (2009) showed that traction on immobilized netrin-1 was sufficient to reorient axons.

Delloye-Bourgeois et al. (2009) showed that autocrine production of NTN1 conveyed a selective advantage in tumor growth and dissemination in aggressive forms of neuroblastoma (NB; see 256700) by blocking the proapoptotic activity of the UNC5H receptors. NTN1 upregulation appeared to be a marker for poor prognosis in infants with stage 4S and stage 4 NB. NTN1 disruption blocked NB metastasis in animals, and Delloye-Bourgeois et al. (2009) proposed disrupting NTN1 expression as an anticancer strategy.


Biochemical Features

Crystal Structure

Xu et al. (2014) determined the structures of a functional NTN1 region, alone and in complexes with NEO1 (601907) and DCC (120470). NTN1 has a rigid elongated structure containing 2 receptor-binding sites at opposite ends through which it brings together receptor molecules. The ligand/receptor complexes reveal 2 distinct architectures: a 2:2 heterotetramer and a continuous ligand/receptor assembly. The differences result from different lengths of the linker connecting receptor domains fibronectin type III domain 4 (FN4) and FN5, which differs among DCC and NEO1 splice variants, providing a basis for diverse signaling outcomes.


Mapping

Meyerhardt et al. (1999) mapped the human NTN1 gene to chromosome 17p13-p12 by FISH.


Molecular Genetics

In members of 2 unrelated families and in an unrelated patient with mirror movements-4 (MRMV4; 618264), Meneret et al. (2017) identified heterozygous mutations in the NTN1 gene (601614.0001-601614.0003). The mutations, which were found by exome sequencing and confirmed by Sanger sequencing, segregated with the condition in the 2 families; the patient in the third family had sporadic occurrence and the mutation occurred de novo. All mutations occurred in the NTR domain and were predicted to affect protein structure and/or stability. In vitro functional expression studies in HeLa and HEK293 cells showed that unlike wildtype, the mutant proteins were almost exclusively found in the intracellular compartments and could not be detected in the supernatant. Since netrin-1 is a diffusible extracellular cue, the pathophysiology probably involved a loss of function. The patients were ascertained from a cohort of 47 index cases with congenital mirror movements in whom mutations in other MRMV-associated genes were excluded.


Animal Model

Serafini et al. (1996) used the method of Skarnes et al. (1995) in embryonic stem cells to selectively recover mutations in genes encoding proteins with signal sequences by virtue of creating fusions of N-terminal portions of these proteins with an exogenous transmembrane domain. In this way Serafini et al. (1996) isolated a mutated murine netrin-1 gene and transmitted it through the germline. Homozygotes were born but apparently did not suckle and died within a few days. Netrin-1-deficient mice exhibited defects in spinal commissural axon projections that were considered consistent with netrin-1 guiding these axons. Defects in several forebrain commissures were also observed, suggesting additional guidance roles for netrin-1.

Poon et al. (2008) demonstrated that the axon guidance cue unc6/netrin and its receptor unc5 (607869) act throughout development to exclude synaptic vesicle and active zone proteins from the dendrite of the C. elegans motor neuron DA9, which is proximal to a source of unc6/netrin. In unc6/netrin and unc5 loss-of-function mutants, presynaptic components mislocalized to the DA9 dendrite. In addition, ectopically expressed unc6/netrin, acting through unc5, was sufficient to exclude endogenous synapses from adjacent subcellular domains within the DA9 axon. Furthermore, this antisynaptogenic activity was interchangeable with that of lin44/Wnt despite being transduced through different receptors, suggesting that extracellular cues such as netrin and Wnts not only guide axon navigation but also regulate the polarized accumulation of presynaptic components through local exclusion.


ALLELIC VARIANTS 3 Selected Examples):

.0001   MIRROR MOVEMENTS 4

NTN1, CYS601ARG
SNP: rs1567750186, ClinVar: RCV000735954

In 3 affected members of a French family (family 1) with mirror movements-4 (MRMV4; 618264), Meneret et al. (2017) identified a heterozygous c.1801T-C transition (c.1801T-C, NM_004822) in exon 3 of the NTN1 gene, resulting in a cys601-to-arg (C601R) substitution at a highly conserved residue in the netrin domain. The mutation, which was found by exome sequencing, was not found in the ExAC database. There were 2 asymptomatic mutation carriers, indicating incomplete penetrance. In vitro functional expression studies in HeLa and HEK293 cells showed that unlike wildtype, the mutant protein was almost exclusively found in the intracellular compartments and could not be detected in the supernatant.


.0002   MIRROR MOVEMENTS 4

NTN1, 3-BP DEL, NT1552
SNP: rs1567749982, ClinVar: RCV000735955

In 3 affected members of a family (family 2) with mirror movements-4 (MRMV4; 618264), Meneret et al. (2017) identified a heterozygous 3-bp deletion (c.1552_1554del, NM_004822) in exon 3 of the NTN1 gene, resulting in the deletion of conserved residue ile518 (Ile518del) in the netrin domain. The mutation, which was found by exome sequencing, was not found in the ExAC database. In vitro functional expression studies in HeLa and HEK293 cells showed that unlike wildtype, the mutant protein was almost exclusively found in the intracellular compartments and could not be detected in the supernatant. This family was previously reported as family C in Franz et al. (2015).


.0003   MIRROR MOVEMENTS 4

NTN1, CYS601SER
SNP: rs1567750187, ClinVar: RCV000735956

In a patient (family 3) with sporadic occurrence of mirror movements-4 (MRMV4; 618264), Meneret et al. (2017) identified a heterozygous c.1802G-C transversion (c.1802G-C, NM_004822) in exon 3 of the NTN1 gene, resulting in a cys601-to-ser (C601S) substitution at a highly conserved residue in the netrin domain. The mutation, which was found by exome sequencing, was not found in the ExAC database. In vitro functional expression studies in HeLa and HEK293 cells showed that unlike wildtype, the mutant protein was almost exclusively found in the intracellular compartments and could not be detected in the supernatant.


REFERENCES

  1. Colon-Ramos, D. A., Margeta, M. A., Shen, K. Glia promote local synaptogenesis through UNC-6 (netrin) signaling in C. elegans. Science 318: 103-106, 2007. [PubMed: 17916735] [Full Text: https://doi.org/10.1126/science.1143762]

  2. Corset, V., Nguyen-Ba-Charvet, K. T., Forcet, C., Moyse, E., Chedotal, A., Mehlen, P. Netrin-1-mediated axon outgrowth and cAMP production requires interaction with adenosine A2b receptor. Nature 407: 747-750, 2000. [PubMed: 11048721] [Full Text: https://doi.org/10.1038/35037600]

  3. Delloye-Bourgeois, C., Fitamant, J., Paradisi, A., Cappellen, D., Douc-Rasy, S., Raquin, M.-A., Stupack, D., Nakagawara, A., Rousseau, R., Combaret, V., Puisieux, A., Valteau-Couanet, D., Benard, J., Bernet, A., Mehlen, P. Netrin-1 acts as a survival factor for aggressive neuroblastoma. J. Exp. Med. 206: 833-847, 2009. [PubMed: 19349462] [Full Text: https://doi.org/10.1084/jem.20082299]

  4. Fitamant, J., Guenebeaud, C., Coissieux, M.-M., Guix, C., Treilleux, I., Scoazec, J.-Y., Bachelot, T., Bernet, A., Mehlen, P. Netrin-1 expression confers a selective advantage for tumor cell survival in metastatic breast cancer. Proc. Nat. Acad. Sci. 105: 4850-4855, 2008. [PubMed: 18353983] [Full Text: https://doi.org/10.1073/pnas.0709810105]

  5. Forcet, C., Stein, E., Pays, L., Corset, V., Llambi, F., Tessier-Lavigne, M., Mehlen, P. Netrin-1-mediated axon outgrowth requires deleted in colorectal cancer-dependent MAPK activation. Nature 417: 443-447, 2002. [PubMed: 11986622] [Full Text: https://doi.org/10.1038/nature748]

  6. Franz, E. A., Chiaroni-Clarke, R., Woodrow, S., Glendining, K. A., Jasoni, C. L., Robertson, S. P., Gardner, R. J. M., Markie, D. Congenital mirror movements: phenotypes associated with DCC and RAD51 mutations. J. Neurol. Sci. 351: 140-145, 2015. [PubMed: 25813273] [Full Text: https://doi.org/10.1016/j.jns.2015.03.006]

  7. Furne, C., Rama, N., Corset, V., Chedotal, A., Mehlen, P. Netrin-1 is a survival factor during commissural neuron navigation. Proc. Nat. Acad. Sci. 105: 14465-14470, 2008. [PubMed: 18796601] [Full Text: https://doi.org/10.1073/pnas.0803645105]

  8. Galko, M. J., Tessier-Lavigne, M. Function of an axonal chemoattractant modulated by metalloprotease activity. Science 289: 1365-1367, 2000. [PubMed: 10958786] [Full Text: https://doi.org/10.1126/science.289.5483.1365]

  9. Hopker, V. H., Shewan, D., Tessier-Lavigne, M., Poo, M., Holt, C. Growth-cone attraction to netrin-1 is converted to repulsion by laminin-1. Nature 401: 69-73, 1999. [PubMed: 10485706] [Full Text: https://doi.org/10.1038/43441]

  10. Larrivee, B., Freitas, C., Trombe, M., Lv, X., DeLafarge, B., Yuan, L., Bouvree, K., Breant, C., Del Toro, R., Brechot, N., Germain, S., Bono, F., Dol, F., Claes, F., Fischer, C., Autiero, M., Thomas, J.-L., Carmeliet, P., Tessier-Lavigne, M., Eichmann, A. Activation of the UNC5B receptor by netrin-1 inhibits sprouting angiogenesis. Genes Dev. 21: 2433-2447, 2007. [PubMed: 17908930] [Full Text: https://doi.org/10.1101/gad.437807]

  11. Li, W., Lee, J., Vikis, H. G., Lee, S.-H., Liu, G., Aurandt, J., Shen, T.-L., Fearon, E. R., Guan, J.-L., Han, M., Rao, Y., Hong, K., Guan, K.-L. Activation of FAK and Src are receptor-proximal events required for netrin signaling. Nature Neurosci. 7: 1213-1221, 2004. [PubMed: 15494734] [Full Text: https://doi.org/10.1038/nn1329]

  12. Liu, G., Beggs, H., Jurgensen, C., Park, H.-T., Tang, H., Gorski, J., Jones, K. R., Reichardt, L. F., Wu, J., Rao, Y. Netrin requires focal adhesion kinase and Src family kinases for axon outgrowth and attraction. Nature Neurosci. 7: 1222-1232, 2004. [PubMed: 15494732] [Full Text: https://doi.org/10.1038/nn1331]

  13. Lu, X., le Noble, F., Yuan, L., Jiang, Q., de Lafarge, B., Sugiyama, D., Breant, C., Claes, F., De Smet, F., Thomas, J.-L., Autiero, M., Carmeliet, P., Tessier-Lavigne, M., Eichmann, A. The netrin receptor UNC5B mediates guidance events controlling morphogenesis of the vascular system. Nature 432: 179-186, 2004. Note: Editorial Expression of Concern. Nature 625: E12, 2024. [PubMed: 15510105] [Full Text: https://doi.org/10.1038/nature03080]

  14. Ly, A., Nikolaev, A., Suresh, G., Zheng, Y., Tessier-Lavigne, M., Stein, E. DSCAM is a netrin receptor that collaborates with DCC in mediating turning responses to netrin-1. Cell 133: 1241-1254, 2008. [PubMed: 18585357] [Full Text: https://doi.org/10.1016/j.cell.2008.05.030]

  15. Mazelin, L., Bernet, A., Bonod-Bidaud, C., Pays, L., Arnaud, S., Gespach, C., Bredesen, D. E., Scoazec, J.-Y., Mehlen, P. Netrin-1 controls colorectal tumorigenesis by regulating apoptosis. Nature 431: 80-84, 2004. [PubMed: 15343335] [Full Text: https://doi.org/10.1038/nature02788]

  16. Mehlen, P., Rabizadeh, S., Snipas, S. J., Assa-Munt, N., Salvesen, G. S., Bredesen, D. E. The DCC gene product induces apoptosis by a mechanism requiring receptor proteolysis. Nature 395: 801-804, 1998. [PubMed: 9796814] [Full Text: https://doi.org/10.1038/27441]

  17. Meneret, A., Franz, E. A., Trouillard, O., Oliver, T. C., Zagar, Y., Robertson, S. P., Weiniarz, Q., Gardner, R. J. M., Gallea, C., Srour, M., Depienne, C., Jasoni, C. L., and 15 others. Mutations in the netrin-1 gene cause congenital mirror movements. J. Clin. Invest. 127: 3923-3936, 2017. [PubMed: 28945198] [Full Text: https://doi.org/10.1172/JCI95442]

  18. Meyerhardt, J. A., Caca, K., Eckstrand, B. C., Hu, G., Lengauer, C., Banavali, S., Look, A. T., Fearon, E. R. Netrin-1: interaction with deleted in colorectal cancer (DCC) and alterations in brain tumors and neuroblastomas. Cell Growth Diff. 10: 35-42, 1999. [PubMed: 9950216]

  19. Ming, G., Wong, S. T., Henley, J., Yuan, X., Song, H., Spitzer, N. C., Poo, M. Adaptation in the chemotactic guidance of nerve growth cones. Nature 417: 411-418, 2002. [PubMed: 11986620] [Full Text: https://doi.org/10.1038/nature745]

  20. Moore, S. W., Biais, N., Sheetz, M. P. Traction on immobilized netrin-1 is sufficient to reorient axons. Science 325: 166 only, 2009. [PubMed: 19589994] [Full Text: https://doi.org/10.1126/science.1173851]

  21. Nishiyama, M., Hoshino, A., Tsai, L., Henley, J. R., Goshima, Y., Tessier-Lavigne, M., Poo, M., Hong, K. Cyclic AMP/GMP-dependent modulation of Ca(2+) channels sets the polarity of nerve growth-cone turning. Nature 423: 990-995, 2003. [PubMed: 12827203] [Full Text: https://doi.org/10.1038/nature01751]

  22. Park, K. W., Crouse, D., Lee, M., Karnik, S. K., Sorensen, L. K., Murphy, K. J., Kuo, C. J., Li, D. Y. The axonal attractant netrin-1 is an angiogenic factor. Proc. Nat. Acad. Sci. 101: 16210-16215, 2004. [PubMed: 15520390] [Full Text: https://doi.org/10.1073/pnas.0405984101]

  23. Poon, V. Y., Klassen, M. P., Shen, K. UNC-6/netrin and its receptor UNC-5 locally exclude presynaptic components from dendrites. Nature 455: 669-673, 2008. [PubMed: 18776887] [Full Text: https://doi.org/10.1038/nature07291]

  24. Ren, X., Ming, G., Xie, Y., Hong, Y., Sun, D., Zhao, Z., Feng, Z., Wang, Q., Shim, S., Chen, Z., Song, H., Mei, L., Xiong, W. Focal adhesion kinase in netrin-1 signaling. Nature Neurosci. 7: 1204-1212, 2004. [PubMed: 15494733] [Full Text: https://doi.org/10.1038/nn1330]

  25. Serafini, T., Colamarino, S. A., Leonardo, E. D., Wang, H., Beddington, R., Skarnes, W. C., Tessier-Lavigne, M. Netrin-1 is required for commissural axon guidance in the developing vertebrate nervous system. Cell 87: 1001-1014, 1996. [PubMed: 8978605] [Full Text: https://doi.org/10.1016/s0092-8674(00)81795-x]

  26. Shewan, D., Dwivedy, A., Anderson, R., Holt, C. E. Age-related changes underlie switch in netrin-1 responsiveness as growth cones advance along visual pathway. Nature Neurosci. 5: 955-962, 2002. [PubMed: 12352982] [Full Text: https://doi.org/10.1038/nn919]

  27. Skarnes, W. C., Moss, J. E., Hurtley, S. M., Beddington, R. S. P. Capturing genes encoding membrane and secreted proteins important for mouse development. Proc. Nat. Acad. Sci. 92: 6592-6596, 1995. [PubMed: 7604039] [Full Text: https://doi.org/10.1073/pnas.92.14.6592]

  28. Stein, E., Tessier-Lavigne, M. Hierarchical organization of guidance receptors: silencing of netrin attraction by Slit through a Robo/DCC receptor complex. Science 291: 1928-1938, 2001. Note: Expression of Concern: Science 378: 1284 only, 2022. [PubMed: 11239147] [Full Text: https://doi.org/10.1126/science.1058445]

  29. Stein, E., Zou, Y., Poo, M., Tessier-Lavigne, M. Binding of DCC by netrin-1 to mediate axon guidance independent of adenosine A2B receptor activation. Science 291: 1976-1982, 2001. Note: Expression of Concern: Science 378: 1284 only, 2022. [PubMed: 11239160] [Full Text: https://doi.org/10.1126/science.1059391]

  30. Tessier-Lavigne, M., Goodman, C. S. The molecular biology of axon guidance. Science 274: 1123-1133, 1996. [PubMed: 8895455] [Full Text: https://doi.org/10.1126/science.274.5290.1123]

  31. Wang, G. X., Poo, M. Requirement of TRPC channels in netrin-1-induced chemotropic turning of nerve growth cones. Nature 434: 898-904, 2005. [PubMed: 15758951] [Full Text: https://doi.org/10.1038/nature03478]

  32. Wilson, B. D., Li, M., Park, K. W., Suli, A., Sorensen, L. K., Larrieu-Lahargue, F., Urness, L. D., Suh, W., Asai, J., Kock, G. A. H., Thorne, T., Silver, M., Thomas, K. R., Chien, C.-B., Losordo, D. W., Li, D. Y. Netrins promote developmental and therapeutic angiogenesis. Science 313: 640-644, 2006. [PubMed: 16809490] [Full Text: https://doi.org/10.1126/science.1124704]

  33. Xu, K., Wu, Z., Renier, N., Antipenko, A., Tzvetkova-Robev, D., Xu, Y., Minchenko, M., Nardi-Dei, V., Rajashankar, K. R., Himanen, J., Tessier-Lavigne, M., Nikolov, D. B. Structures of netrin-1 bound to two receptors provide insight into its axon guidance mechanism. Science 344: 1275-1279, 2014. [PubMed: 24876346] [Full Text: https://doi.org/10.1126/science.1255149]


Contributors:
Cassandra L. Kniffin - updated : 01/02/2019
Ada Hamosh - updated : 7/7/2014
Paul J. Converse - updated : 11/12/2009
Patricia A. Hartz - updated : 8/31/2009
Ada Hamosh - updated : 8/14/2009
Ada Hamosh - updated : 11/5/2008
Patricia A. Hartz - updated : 9/10/2008
Patricia A. Hartz - updated : 8/13/2008
Patricia A. Hartz - updated : 11/2/2007
Ada Hamosh - updated : 10/26/2007
Ada Hamosh - updated : 6/27/2007
Anne M. Stumpf - updated : 9/8/2006
Ada Hamosh - updated : 9/6/2006
Ada Hamosh - updated : 9/19/2005
Cassandra L. Kniffin - updated : 2/8/2005
Victor A. McKusick - updated : 12/30/2004
Ada Hamosh - updated : 11/10/2004
Ada Hamosh - updated : 7/8/2003
Cassandra L. Kniffin - updated : 9/30/2002
Paul J. Converse - updated : 5/6/2002
Ada Hamosh - updated : 3/27/2001
Ada Hamosh - updated : 3/27/2001
Ada Hamosh - updated : 10/18/2000
Ada Hamosh - updated : 9/5/2000
Ada Hamosh - updated : 2/14/2000

Creation Date:
Victor A. McKusick : 1/8/1997

Edit History:
carol : 04/16/2024
carol : 04/15/2024
carol : 01/26/2024
carol : 01/21/2023
carol : 01/09/2019
carol : 01/08/2019
ckniffin : 01/02/2019
carol : 04/24/2018
alopez : 07/07/2014
alopez : 7/7/2014
carol : 6/20/2012
terry : 1/20/2010
mgross : 11/17/2009
terry : 11/12/2009
mgross : 9/4/2009
terry : 8/31/2009
alopez : 8/18/2009
terry : 8/14/2009
alopez : 11/18/2008
alopez : 11/18/2008
terry : 11/5/2008
mgross : 9/12/2008
terry : 9/10/2008
mgross : 8/13/2008
mgross : 11/2/2007
terry : 11/2/2007
alopez : 11/1/2007
alopez : 11/1/2007
terry : 10/26/2007
alopez : 7/5/2007
terry : 6/27/2007
wwang : 9/13/2006
alopez : 9/13/2006
alopez : 9/8/2006
alopez : 9/8/2006
terry : 9/6/2006
alopez : 9/20/2005
terry : 9/19/2005
wwang : 2/17/2005
ckniffin : 2/8/2005
tkritzer : 1/21/2005
terry : 12/30/2004
tkritzer : 11/10/2004
mgross : 7/14/2003
terry : 7/8/2003
tkritzer : 6/19/2003
alopez : 11/4/2002
carol : 10/21/2002
ckniffin : 9/30/2002
alopez : 6/6/2002
mgross : 5/6/2002
alopez : 3/27/2001
alopez : 3/27/2001
alopez : 10/18/2000
alopez : 9/5/2000
alopez : 2/14/2000
alopez : 4/30/1999
alopez : 6/22/1998
alopez : 6/22/1998
jamie : 1/17/1997
mark : 1/16/1997
jamie : 1/15/1997
jamie : 1/9/1997
jamie : 1/9/1997
mark : 1/8/1997