Entry - *601725 - NEUROGENIC DIFFERENTIATION 2; NEUROD2 - OMIM
 
* 601725

NEUROGENIC DIFFERENTIATION 2; NEUROD2


HGNC Approved Gene Symbol: NEUROD2

Cytogenetic location: 17q12     Genomic coordinates (GRCh38): 17:39,603,768-39,607,920 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
17q12 Developmental and epileptic encephalopathy 72 618374 AD 3

TEXT

Description

The NEUROD2 gene encodes a basic helix-loop-helix (bHLH) protein that is a transcription factor involved in central and peripheral nervous system development (summary by Sega et al., 2019).


Cloning and Expression

Lee et al. (1995) described a bHLH protein, NEUROD (NEUROD1; 601724), that functions during neurogenesis. McCormick et al. (1996) described the cloning and characterization of 2 additional NEUROD genes, NEUROD2 and NEUROD3 (601726). Sequences for the mouse and human homologs were presented. NEUROD2 shows a high degree of homology to the bHLH region of NEUROD, whereas NEUROD3 is more distantly related.


Gene Function

McCormick et al. (1996) found that mouse neuroD2 was initially expressed at embryonic day 11, with persistent expression in the adult nervous system. Similar to neuroD, neuroD2 appears to mediate neuronal differentiation.

Yang et al. (2009) found that the major mitotic E3 ubiquitin ligase Cdc20 (603618)-anaphase-promoting complex (Cdc20-APC; see ANAPC1, 608473) regulates presynaptic differentiation in primary postmitotic mammalian neurons and in the rat cerebellar cortex. Cdc20-APC triggered the degradation of the transcription factor NeuroD2 and thereby promoted presynaptic differentiation. The NeuroD2 target gene encoding complexin-2 (CPLX2; 605033), which acts locally at presynaptic sites, mediated the ability of NeuroD2 to suppress presynaptic differentiation. Yang et al. (2009) concluded that their findings defined a Cdc20-APC ubiquitin signaling pathway that governs presynaptic development.

Yoo et al. (2011) demonstrated that expression of miR9/9* (see 611186) and miR-124 (609327) in human fibroblasts induced their conversion into neurons, a process facilitated by NEUROD2. Further addition of neurogenic transcription factors ASCL1 (100790) and MYT1L (613084) enhanced the rate of conversion and the maturation of the converted neurons, whereas expression of these transcription factors alone without the aforementioned microRNAs was ineffective. Yoo et al. (2011) concluded that the genetic circuitry involving miR9-1 through miR9-3 and miR124 can have an instructive role in neural fate determination.


Mapping

Tamimi et al. (1997) mapped human NEUROD2 to 17q12 by fluorescence in situ hybridization. They mapped the mouse homolog to chromosome 11.


Molecular Genetics

In 2 unrelated children with developmental and epileptic encephalopathy-72 (DEE72; 618374), Sega et al. (2019) identified de novo heterozygous missense mutations in the NEUROD2 gene (E130Q, 601725.0001 and M134T, 601725.0002). The mutations, which were found by whole-exome sequencing and confirmed by Sanger sequencing, were not found in the gnomAD database. Both mutations occurred in the DNA-binding domain. Overexpression of wildtype Neurod2 in X. laevis induces nonneuronal cells to differentiate into neurons and promotes the ectopic expression of neurons expressing mature neural markers. In contrast, expression of the missense mutations into X. laevis resulted in impaired (M134T) or absent (E130Q) ectopic neurons compared to wildtype, suggesting a loss of function.


Animal Model

Ince-Dunn et al. (2006) found that Neurod2 -/- mice were born at a normal mendelian ratio, but most died between 4 to 5 weeks of age. Both Neurod2 -/- and Neurod2 +/- mice showed altered brain organization compared with wildtype littermates, including decreased brain size, slightly smaller and rounder hippocampus, and absence of corpus callosum. Thalamocortical axon terminals of Neurod2 -/- mice failed to segregate in the somatosensory cortex, and the postsynaptic barrel organization was disrupted. Synaptic transmission was defective at thalamocortical synapses in Neurod2 -/- mice, with reduced total excitatory synaptic currents in layer IV due to reduced contribution of AMPA receptors (see GR1AI; 138248) compared with NMDA receptors (see GRIN1; 138249).

Sega et al. (2019) found that knockdown of the Neurod2 orthologs in X. tropicalis tadpoles caused abnormal swimming behavior and seizures followed by periods of immobility.


ALLELIC VARIANTS ( 2 Selected Examples):

.0001 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 72

NEUROD2, GLU130GLN
  
RCV000766210

In a 3.5-year-old girl with infantile epileptic encephalopathy-72 (DEE72; 618374), Sega et al. (2019) identified a de novo heterozygous c.388G-C transversion (c.388G-C, NM_006160.3) in the NEUROD2 gene, resulting in a glu130-to-gln (E130Q) substitution at a highly conserved residue in the DNA-binding domain. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the gnomAD database. Expression of the E130Q mutations into X. laevis resulted in absent production of ectopic neurons compared to wildtype, suggesting a complete loss of function. At 5 months of age, the patient had onset of refractory infantile spasms associated with hypsarrhythmia on EEG.


.0002 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 72

NEUROD2, MET134THR
  
RCV000766211

In a 2.5-year-old boy with infantile epileptic encephalopathy-72 (DEE72; 618374), Sega et al. (2019) identified a de novo heterozygous c.401T-C transition (c.401T-C, NM_006160.3) in the NEUROD2 gene, resulting in a met134-to-thr (M134T) substitution at a highly conserved residue in the DNA-binding domain. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the gnomAD database. Expression of the M134T mutation into X. laevis resulted in impaired production of ectopic neurons compared to wildtype, suggesting a partial loss of function. At 5 months of age, the patient had onset of refractory infantile spasms associated with hypsarrhythmia on EEG.


REFERENCES

  1. Ince-Dunn, G., Hall, B. J., Hu, S.-C., Ripley, B., Huganir, R. L., Olson, J. M., Tapscott, S. J., Ghosh, A. Regulation of thalamocortical patterning and synaptic maturation by NeuroD2. Neuron 49: 683-695, 2006. [PubMed: 16504944, related citations] [Full Text]

  2. Lee, J. E., Hollenberg, S. M., Snider, L., Turner, D. L., Lipnick, N., Weintraub, H. Conversion of Xenopus ectoderm into neurons by NeuroD, a basic helix-loop-helix protein. Science 268: 836-844, 1995. [PubMed: 7754368, related citations] [Full Text]

  3. McCormick, M. B., Tamimi, R. M., Snider, L., Asakura, A., Bergstrom, D., Tapscott, S. J. neuroD2 and neuroD3: distinct expression patterns and transcriptional activation potentials within the neuroD gene family. Molec. Cell. Biol. 16: 5792-5800, 1996. [PubMed: 8816493, related citations] [Full Text]

  4. Sega, A. G., Mis, E. K., Lindsrom, K., Mercimek-Andrews, S., Ji, W., Cho, M. T., Juusola, J., Konstantino, M., Jeffries, L., Khokha, M. K., Lakhani, S. A. Do novo pathogenic variants in neuronal differentiation factor 2 (NEUROD2) cause a form of early infantile epileptic encephalopathy. J. Med. Genet. 56: 113-122, 2019. [PubMed: 30323019, related citations] [Full Text]

  5. Tamimi, R. M., Steingrimsson, E., Montgomery-Dyer, K., Copeland, N. G., Jenkins, N. A., Tapscott, S. J. NEUROD2 and NEUROD3 genes map to human chromosomes 17q12 and 5q23-q31 and mouse chromosomes 11 and 13, respectively. Genomics 40: 355-357, 1997. [PubMed: 9119405, related citations] [Full Text]

  6. Yang, Y., Kim, A. H., Yamada, T., Wu, B., Bilimoria, P. M., Ikeuchi, Y., de la Iglesia, N., Shen, J., Bonni, A. A Cdc20-APC ubiquitin signaling pathway regulates presynaptic differentiation. Science 326: 575-578, 2009. [PubMed: 19900895, images, related citations] [Full Text]

  7. Yoo, A. S., Sun, A. X., Li, L., Shcheglovitov, A., Portmann, T., Li, Y., Lee-Messer, C., Dolmetsch, R. E., Tsien, R. W., Crabtree, G. R. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 476: 228-231, 2011. [PubMed: 21753754, images, related citations] [Full Text]


Cassandra L. Kniffin - updated : 04/02/2019
Ada Hamosh - updated : 8/24/2011
Patricia A. Hartz - updated : 2/11/2011
Ada Hamosh - updated : 11/10/2009
Creation Date:
Mark H. Paalman : 3/27/1997
alopez : 11/25/2020
joanna : 11/13/2020
carol : 04/04/2019
ckniffin : 04/02/2019
alopez : 10/27/2014
alopez : 8/25/2011
terry : 8/24/2011
mgross : 2/15/2011
terry : 2/11/2011
alopez : 11/10/2009
terry : 11/10/2009
joanna : 10/29/2009
mark : 3/31/1997
terry : 3/27/1997

* 601725

NEUROGENIC DIFFERENTIATION 2; NEUROD2


HGNC Approved Gene Symbol: NEUROD2

Cytogenetic location: 17q12     Genomic coordinates (GRCh38): 17:39,603,768-39,607,920 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
17q12 Developmental and epileptic encephalopathy 72 618374 Autosomal dominant 3

TEXT

Description

The NEUROD2 gene encodes a basic helix-loop-helix (bHLH) protein that is a transcription factor involved in central and peripheral nervous system development (summary by Sega et al., 2019).


Cloning and Expression

Lee et al. (1995) described a bHLH protein, NEUROD (NEUROD1; 601724), that functions during neurogenesis. McCormick et al. (1996) described the cloning and characterization of 2 additional NEUROD genes, NEUROD2 and NEUROD3 (601726). Sequences for the mouse and human homologs were presented. NEUROD2 shows a high degree of homology to the bHLH region of NEUROD, whereas NEUROD3 is more distantly related.


Gene Function

McCormick et al. (1996) found that mouse neuroD2 was initially expressed at embryonic day 11, with persistent expression in the adult nervous system. Similar to neuroD, neuroD2 appears to mediate neuronal differentiation.

Yang et al. (2009) found that the major mitotic E3 ubiquitin ligase Cdc20 (603618)-anaphase-promoting complex (Cdc20-APC; see ANAPC1, 608473) regulates presynaptic differentiation in primary postmitotic mammalian neurons and in the rat cerebellar cortex. Cdc20-APC triggered the degradation of the transcription factor NeuroD2 and thereby promoted presynaptic differentiation. The NeuroD2 target gene encoding complexin-2 (CPLX2; 605033), which acts locally at presynaptic sites, mediated the ability of NeuroD2 to suppress presynaptic differentiation. Yang et al. (2009) concluded that their findings defined a Cdc20-APC ubiquitin signaling pathway that governs presynaptic development.

Yoo et al. (2011) demonstrated that expression of miR9/9* (see 611186) and miR-124 (609327) in human fibroblasts induced their conversion into neurons, a process facilitated by NEUROD2. Further addition of neurogenic transcription factors ASCL1 (100790) and MYT1L (613084) enhanced the rate of conversion and the maturation of the converted neurons, whereas expression of these transcription factors alone without the aforementioned microRNAs was ineffective. Yoo et al. (2011) concluded that the genetic circuitry involving miR9-1 through miR9-3 and miR124 can have an instructive role in neural fate determination.


Mapping

Tamimi et al. (1997) mapped human NEUROD2 to 17q12 by fluorescence in situ hybridization. They mapped the mouse homolog to chromosome 11.


Molecular Genetics

In 2 unrelated children with developmental and epileptic encephalopathy-72 (DEE72; 618374), Sega et al. (2019) identified de novo heterozygous missense mutations in the NEUROD2 gene (E130Q, 601725.0001 and M134T, 601725.0002). The mutations, which were found by whole-exome sequencing and confirmed by Sanger sequencing, were not found in the gnomAD database. Both mutations occurred in the DNA-binding domain. Overexpression of wildtype Neurod2 in X. laevis induces nonneuronal cells to differentiate into neurons and promotes the ectopic expression of neurons expressing mature neural markers. In contrast, expression of the missense mutations into X. laevis resulted in impaired (M134T) or absent (E130Q) ectopic neurons compared to wildtype, suggesting a loss of function.


Animal Model

Ince-Dunn et al. (2006) found that Neurod2 -/- mice were born at a normal mendelian ratio, but most died between 4 to 5 weeks of age. Both Neurod2 -/- and Neurod2 +/- mice showed altered brain organization compared with wildtype littermates, including decreased brain size, slightly smaller and rounder hippocampus, and absence of corpus callosum. Thalamocortical axon terminals of Neurod2 -/- mice failed to segregate in the somatosensory cortex, and the postsynaptic barrel organization was disrupted. Synaptic transmission was defective at thalamocortical synapses in Neurod2 -/- mice, with reduced total excitatory synaptic currents in layer IV due to reduced contribution of AMPA receptors (see GR1AI; 138248) compared with NMDA receptors (see GRIN1; 138249).

Sega et al. (2019) found that knockdown of the Neurod2 orthologs in X. tropicalis tadpoles caused abnormal swimming behavior and seizures followed by periods of immobility.


ALLELIC VARIANTS 2 Selected Examples):

.0001   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 72

NEUROD2, GLU130GLN
SNP: rs1323339153, gnomAD: rs1323339153, ClinVar: RCV000766210

In a 3.5-year-old girl with infantile epileptic encephalopathy-72 (DEE72; 618374), Sega et al. (2019) identified a de novo heterozygous c.388G-C transversion (c.388G-C, NM_006160.3) in the NEUROD2 gene, resulting in a glu130-to-gln (E130Q) substitution at a highly conserved residue in the DNA-binding domain. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the gnomAD database. Expression of the E130Q mutations into X. laevis resulted in absent production of ectopic neurons compared to wildtype, suggesting a complete loss of function. At 5 months of age, the patient had onset of refractory infantile spasms associated with hypsarrhythmia on EEG.


.0002   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 72

NEUROD2, MET134THR
SNP: rs1567841596, ClinVar: RCV000766211

In a 2.5-year-old boy with infantile epileptic encephalopathy-72 (DEE72; 618374), Sega et al. (2019) identified a de novo heterozygous c.401T-C transition (c.401T-C, NM_006160.3) in the NEUROD2 gene, resulting in a met134-to-thr (M134T) substitution at a highly conserved residue in the DNA-binding domain. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the gnomAD database. Expression of the M134T mutation into X. laevis resulted in impaired production of ectopic neurons compared to wildtype, suggesting a partial loss of function. At 5 months of age, the patient had onset of refractory infantile spasms associated with hypsarrhythmia on EEG.


REFERENCES

  1. Ince-Dunn, G., Hall, B. J., Hu, S.-C., Ripley, B., Huganir, R. L., Olson, J. M., Tapscott, S. J., Ghosh, A. Regulation of thalamocortical patterning and synaptic maturation by NeuroD2. Neuron 49: 683-695, 2006. [PubMed: 16504944] [Full Text: https://doi.org/10.1016/j.neuron.2006.01.031]

  2. Lee, J. E., Hollenberg, S. M., Snider, L., Turner, D. L., Lipnick, N., Weintraub, H. Conversion of Xenopus ectoderm into neurons by NeuroD, a basic helix-loop-helix protein. Science 268: 836-844, 1995. [PubMed: 7754368] [Full Text: https://doi.org/10.1126/science.7754368]

  3. McCormick, M. B., Tamimi, R. M., Snider, L., Asakura, A., Bergstrom, D., Tapscott, S. J. neuroD2 and neuroD3: distinct expression patterns and transcriptional activation potentials within the neuroD gene family. Molec. Cell. Biol. 16: 5792-5800, 1996. [PubMed: 8816493] [Full Text: https://doi.org/10.1128/MCB.16.10.5792]

  4. Sega, A. G., Mis, E. K., Lindsrom, K., Mercimek-Andrews, S., Ji, W., Cho, M. T., Juusola, J., Konstantino, M., Jeffries, L., Khokha, M. K., Lakhani, S. A. Do novo pathogenic variants in neuronal differentiation factor 2 (NEUROD2) cause a form of early infantile epileptic encephalopathy. J. Med. Genet. 56: 113-122, 2019. [PubMed: 30323019] [Full Text: https://doi.org/10.1136/jmedgenet-2018-105322]

  5. Tamimi, R. M., Steingrimsson, E., Montgomery-Dyer, K., Copeland, N. G., Jenkins, N. A., Tapscott, S. J. NEUROD2 and NEUROD3 genes map to human chromosomes 17q12 and 5q23-q31 and mouse chromosomes 11 and 13, respectively. Genomics 40: 355-357, 1997. [PubMed: 9119405] [Full Text: https://doi.org/10.1006/geno.1996.4578]

  6. Yang, Y., Kim, A. H., Yamada, T., Wu, B., Bilimoria, P. M., Ikeuchi, Y., de la Iglesia, N., Shen, J., Bonni, A. A Cdc20-APC ubiquitin signaling pathway regulates presynaptic differentiation. Science 326: 575-578, 2009. [PubMed: 19900895] [Full Text: https://doi.org/10.1126/science.1177087]

  7. Yoo, A. S., Sun, A. X., Li, L., Shcheglovitov, A., Portmann, T., Li, Y., Lee-Messer, C., Dolmetsch, R. E., Tsien, R. W., Crabtree, G. R. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 476: 228-231, 2011. [PubMed: 21753754] [Full Text: https://doi.org/10.1038/nature10323]


Contributors:
Cassandra L. Kniffin - updated : 04/02/2019
Ada Hamosh - updated : 8/24/2011
Patricia A. Hartz - updated : 2/11/2011
Ada Hamosh - updated : 11/10/2009

Creation Date:
Mark H. Paalman : 3/27/1997

Edit History:
alopez : 11/25/2020
joanna : 11/13/2020
carol : 04/04/2019
ckniffin : 04/02/2019
alopez : 10/27/2014
alopez : 8/25/2011
terry : 8/24/2011
mgross : 2/15/2011
terry : 2/11/2011
alopez : 11/10/2009
terry : 11/10/2009
joanna : 10/29/2009
mark : 3/31/1997
terry : 3/27/1997