Entry - *114020 - CADHERIN 2; CDH2 - OMIM
 
* 114020

CADHERIN 2; CDH2


Alternative titles; symbols

CADHERIN, NEURONAL
N-CADHERIN; NCAD
CALCIUM-DEPENDENT ADHESION PROTEIN, NEURONAL; CDHN


HGNC Approved Gene Symbol: CDH2

Cytogenetic location: 18q12.1     Genomic coordinates (GRCh38): 18:27,932,879-28,177,130 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
18q12.1 ?Attention deficit-hyperactivity disorder 8 619957 AR 3
Agenesis of corpus callosum, cardiac, ocular, and genital syndrome 618929 AD 3
Arrhythmogenic right ventricular dysplasia 14 618920 AD 3

TEXT

Description

The CDH2 gene encodes N (neuronal)-cadherin, which has an essential role in the early steps of brain morphogenesis (summary by Halperin et al., 2021).


Cloning and Expression

Reid and Hemperly (1990) obtained a full-length NCAD clone from a Kelly neuroblastoma library. The NCAD gene encodes a 907-amino acid protein that includes a 159-amino acid signal sequence. Human and mouse nucleotide sequences are 96% identical.


Gene Structure

Miyatani et al. (1992) showed that the mouse Ncad gene consists of 16 exons dispersed over more than 200 kb of genomic DNA. The large size of the N-cadherin gene, compared with its cDNA (4.3 kb), was ascribed to the fact that the first and second introns are 34.2 kb and more than 100 kb long, respectively. Miyatani et al. (1992) compared the NCAD, liver cell adhesion molecule (LCAM; see 192090), and PCAD (CDH3) genes and showed that the exon-intron boundaries are fully conserved between them, except that the first exon of P-cadherin includes the first and second exons of the other 2 genes. Also, the second intron, which is equivalent to the first intron in P-cadherin, is exceptionally large and this structural feature is conserved in all 3 of these genes.

Wallis et al. (1994) demonstrated that the human N-cadherin gene contains 16 exons and its sequence is highly similar to both the mouse NCAD gene (including the large first and second introns) and other cadherin genes.


Mapping

In Southern analysis of a panel of somatic cell hybrids, Walsh et al. (1990) mapped the NCAD gene to chromosome 18. By interspecific backcross analysis, Miyatani et al. (1992) found that the gene in the mouse is located in the proximal region of chromosome 18.

By in situ hybridization, Wallis et al. (1994) refined the map position of N-cadherin to 18q11.2.

Stumpf (2020) mapped the CDH2 gene to chromosome 18q12.1 based on an alignment of the CDH2 sequence (GenBank BC036470) with the genomic sequence (GRCh38).


Gene Family

The cadherin gene family encode proteins that mediate calcium-ion-dependent adhesion (Takeichi, 1987). Three members of this family are E-cadherin, N-cadherin, and P-cadherin. E-cadherin appears to be identical to the protein called uvomorulin. N-cadherin is expressed in the brain and skeletal and cardiac muscle.


Gene Function

Hermiston and Gordon (1995) noted that the mouse intestinal epithelium expresses a sequence of 'developmental events'--proliferation, lineage allocation, migration, differentiation, and death--throughout life. Proliferation is confined to the crypts of Lieberkuhn. The crypt's multipotent stem cell gives rise to enterocytes, mucus-producing goblet cells, enteroendocrine cells, and Paneth cells. Cells of these 4 lineages differentiate during an orderly migration and are frequently eliminated by apoptosis and exfoliation or phagocytosis. Renewal is rapid (3 to 20 days). Results from cell culture studies indicate that cadherin-catenin complexes regulate cell polarity, formation of junctional complexes, migration, and proliferation. Hermiston and Gordon (1995) transfected embryonic stem cells with a dominant-negative N-cadherin mutant under the control of promoters active in small intestinal epithelial cells and introduced them into C57BL/6 blastocysts. Analysis of adult chimeric mice revealed that expression of the mutant along the entire crypt-villus axis, but not in the villus epithelium alone, produced an inflammatory bowel disease resembling Crohn disease (see 266600). The mutation perturbed proliferation, migration, and death patterns in crypts, leading to adenomas. The model provided insights into cadherin function in an adult organ and the factors underlying inflammatory bowel disease and intestinal neoplasia.

Within the bilaterally symmetric vertebrate body plan, many organs develop asymmetrically. Garcia-Castro et al. (2000) demonstrated that the cell adhesion molecule N-cadherin is one of the earliest proteins to be asymmetrically expressed in the chicken embryo and that its activity is required during gastrulation for proper establishment of the left-right axis. Blocking N-cadherin function randomizes heart looping and alters the expression of Snail (604238) and Pitx2 (601542), later components of the molecular cascade that regulates left-right asymmetry. However, the expression of other components of this cascade, Nodal (601265) and Lefty (see 601877), was unchanged after blocking N-cadherin function, suggesting the existence of parallel pathways in the establishment of left-right morphogenesis. Garcia-Castro et al. (2000) concluded that their results suggest that N-cadherin-mediated cell adhesion events are required for establishment of left-right asymmetry.

At certain central nervous system synapses, pre- to postsynaptic adhesion is mediated at least in part by CDH2. Tanaka et al. (2000) demonstrated that upon depolarization of cultured hippocampal neurons by potassium treatment, or the application of N-methyl-D-aspartate or alpha-latrotoxin, synaptic CDH2 dimerizes and becomes markedly protease resistant. The resistance persisted for at least 2 hours while other surface molecules, including other cadherins, were completely degraded. The acquisition of protease resistance and dimerization of CDH2 was not dependent on new protein synthesis. Tanaka et al. (2000) concluded that synaptic adhesion is dynamically and locally controlled and is modulated by synaptic activity.

Arregui et al. (2000) demonstrated that cell-permeable (Trojan) peptides containing the third helix of the antennapedia homeodomain fused to a peptide mimicking the juxtamembrane (JMP) region of the cytoplasmic domain of CDH2 result in the inhibition of both CDH2 and beta-1 integrin (ITGB1; 135630) function. Microscopic analysis showed that expression of JMP, which binds to the cytoplasmic domain of CDH2, results in a reduction of neurite outgrowth on cadherin substrates. Treatment of cells with JMP resulted in the release of FER (176942) from the cadherin complex and its accumulation in the integrin complex. The accumulation of FER in the integrin complex and the inhibitory effects of JMP could be reversed with a peptide that mimics the first coiled-coil domain of FER. The results suggested that FER mediates crosstalk between CDH2 and ITGB1.

Van Aken et al. (2002) studied the cadherin-catenin complex in retinoblastoma and normal retina tissues. In both cases, they found that N-cadherin was associated with alpha- and beta-catenin (116805; 116806) but not with E-cadherin (CDH1; 192090) or P-cadherin (CDH3; 114021), retinoblastoma cells, in contrast with normal retina, expressed an N-cadherin/catenin complex that was irregularly distributed and weakly linked to the cytoskeleton. In retinoblastoma, this complex acted as an invasion promoter.

In cultured rat hippocampal neurons, Togashi et al. (2002) showed that blockade of the known hippocampal cadherins (cadherin-8, 603008; cadherin-11, 600023; and N-cadherin) resulted in alterations of dendritic spine morphology, such as filopodia-like elongation of the spine and bifurcation of its head structure, along with concomitant disruption of the distribution of postsynaptic proteins and perturbation of synaptic vesicle recycling. The findings suggested that cadherins regulate dendritic spine morphogenesis and related synaptic functions.

Rubinek et al. (2003) studied the role of pituicyte cell-cell contact mediated by CDH2 and NCAM1 (116930) in the regulation of GH (139250) secretion. RT-PCR showed CDH2 mRNA expression in 8 of 12 GH-secreting adenomas compared with 1 of 7 prolactin-cell adenomas. CDH2 and NCAM1 were similarly expressed in adenomas and in adult and fetal normal pituitary tissues. Cell adhesion molecule (CAM) stimulation increased GH secretion from pituitary fetal cultures by 40 to 60% and also from cultured GH adenoma cells by 40 to 75%. Disrupting CDH2 homophilic binding by anti-CDH2 antibody decreased fetal, but not tumorous, GH secretion by 40%. This study indicated that pituitary cell-cell contact mediated by homophilic interactions between adhesion molecules regulates human GH secretion.

Hayashi and Carthew (2004) investigated the physical basis of biologic patterning of the Drosophila retina in vivo. They demonstrated that E-cadherin and N-cadherin mediate apical adhesion between retina epithelial cells. Differential expression of N-cadherin within a subgroup of retinal cells (cone cells) caused them to form an overall shape that minimized their surface contact with surrounding cells. The cells within this group, in both normal and experimentally manipulated conditions, packed together in the same way as soap bubbles do. The shaping of cone cell group and packing of its components precisely imitated the physical tendency for surfaces to be minimized. Hayashi and Carthew (2004) concluded that simple patterned expression of N-cadherin resulted in a complex spatial pattern of cells owing to cellular surface mechanics.

Banh et al. (2009) showed that the first 2 extracellular domains of N-cadherin interacted with the inhibitory receptor KLRG1 (604874), blocked interaction of KLRG1 with E-cadherin, and could regulate KLRG1 signaling.

Tsai et al. (2020) noted that, in zebrafish spinal cord, neural progenitors form stereotypic patterns despite noisy morphogen signaling and large-scale cellular rearrangements during morphogenesis and growth. By directly measuring adhesion forces and preferences for 3 types of endogenous neural progenitors, Tsai et al. (2020) provided evidence for the differential adhesion model, in which differences in intercellular adhesion mediate cell sorting. Cell type-specific combinatorial expression of different classes of cadherins, including N-cadherin, cadherin-11, and protocadherin-19 (PCDH19; 300460), resulted in homotypic preference ex vivo and patterning robustness in vivo. The differential adhesion code was regulated by the shh (600725) morphogen gradient. Tsai et al. (2020) proposed that robust patterning during tissue morphogenesis results from interplay between adhesion-based self-organization and morphogen-directed patterning.


Molecular Genetics

Arrhythmogenic Right Ventricular Dysplasia 14

In affected members of a 3-generation South African family segregating autosomal dominant arrhythmogenic right ventricular dysplasia (ARVD14; 618920), Mayosi et al. (2017) identified heterozygosity for a missense mutation in the CDH2 gene (Q229P; 114020.0001) that segregated fully with disease. Screening of 73 additional ARVD probands revealed 1 patient with another missense mutation in CDH2 (D407N; 114020.0002).

Agenesis of Corpus Callosum, Cardiac, Ocular, and Genital Syndrome

In 9 unrelated patients with agenesis of corpus callosum, cardiac, ocular, and genital syndrome (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for de novo mutations in the CDH2 gene (see, e.g., 114020.0003-114020.0007). Functional analysis indicated that the mutations impair the cell adhesion function of N-cadherin by affecting self-binding as well as trans-binding with wildtype N-cadherin. Noting the neurodevelopmental features observed in the affected individuals, including agenesis of the corpus callosum, periventricular nodular heterotopias, hyposmia, mirror movements, Duane anomaly, and abnormal shoulder muscle innervation, the authors suggested that CDH2 plays a critical role in neuronal migration and axon pathfinding.

In 4 unrelated patients with ACOGS, Reis et al. (2020) identified heterozygosity for mutations in the CDH2 gene (see, e.g., 114020.0008-114020.0009).

Attention Deficit-Hyperactivity Disorder 8

In 3 sibs, born of consanguineous Bedouin parents, with attention deficit-hyperactivity disorder-8 (ADHD8; 619957), Halperin et al. (2021) identified a homozygous missense mutation in the CDH2 gene (H150Y; 114020.0010). The mutation, which was found by a combination of homozygosity mapping and whole-exome sequencing and confirmed by Sanger sequencing, segregated with the disorder in the family. It was not present in the gnomAD database. In vitro functional expression studies showed that the mutation interfered with CDH2 protein processing, causing decreased proteolytic cleavage efficacy and defective N-cadherin protein maturation. Mutant mice with a homozygous H150Y mutation in the Cdh2 gene demonstrated motor hyperactivity, with greater traveling distance, increased velocity, and prolonged mobility time in the open-field exploratory test compared to controls (see ANIMAL MODEL).


Animal Model

Halperin et al. (2021) found that mutant mice homozygous for the H150Y mutation in the Cdh2 gene demonstrated hyperactive behavior. Mutant mice also showed an elevated startle amplitude in the acoustic startle reflex test (ASR) compared to controls, suggesting deficits in sensorimotor integration. Detailed analysis of the neurons from brains of mutant mice showed that homozygosity for the Cdh2 mutation was associated with a decrease in the size of the presynaptic vesicle cluster, a decrease in the readily releasable pool size of synaptic vesicles in neurons, and an attenuation of synaptic vesicle release compared to controls. The frequency of spontaneous synaptic release was also decreased compared to controls. There was reduced tyrosine hydroxylase expression and dopamine levels in the midbrain and prefrontal cortex. The authors suggested that deficits in synaptic formation due to aberrant processing of N-cadherin is the underlying mechanism for the clinical manifestations.


ALLELIC VARIANTS ( 10 Selected Examples):

.0001 ARRHYTHMOGENIC RIGHT VENTRICULAR DYSPLASIA, FAMILIAL, 14

CDH2, GLN229PRO
  
RCV001194670

In affected members of a 3-generation white South African family (ACM2) with arrhythmogenic right ventricular dysplasia (ARVD14; 618920), Mayosi et al. (2017) identified heterozygosity for a c.686A-C transversion in the CDH2 gene, resulting in a gln229-to-pro (Q229P) substitution at a highly conserved residue within the EC1 domain. The mutation segregated fully with disease in the family and was not found in 200 white South African control chromosomes or in the 1000 Genomes or ExAC databases.


.0002 ARRHYTHMOGENIC RIGHT VENTRICULAR DYSPLASIA, FAMILIAL, 14

CDH2, ASP407ASN (rs568089577)
  
RCV001194671...

In a white South African man (ACM11) with arrhythmogenic right ventricular dysplasia (ARVD14; 618920), Mayosi et al. (2017) identified heterozygosity for a c.1219G-A transition in the CDH2 gene, resulting in an asp407-to-asn (D407N) substitution at a highly conserved residue within the EC3 domain. The mutation was not found in his unaffected mother or in 200 white South African control chromosomes or the Helmholtz database; however, it was present once in the 1000 Genomes database (minor allele frequency, 0.0002) and once in the ExAC database (MAF, 0.000008).


.0003 AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, ASP597ASN
  
RCV001007453...

In an 9-year-old boy (patient 2) with agenesis of the corpus callosum, tricuspid regurgitation, Duane anomaly and right ptosis, and micropenis (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo c.1789G-A transition (c.1789G-A, NM_001792.5) in the CDH2 gene, resulting in an asp597-to-asn (D597N) substitution at a highly conserved residue in the calcium-binding site within the EC4-EC5 linker region. The mutation was not found in the gnomAD database. Analysis in transfected L cells showed that cells expressing the D597N variant displayed reduced aggregation compared to wildtype N-cadherin, forming smaller aggregates and resulting in a significantly lower index of aggregation compared to wildtype. In addition, mixed aggregation assay showed that fewer mixed aggregates were formed with the D597N mutant compared to wildtype. The authors concluded that the D597N mutation impairs the cell adhesion function of N-cadherin by affecting self-binding as well as trans-binding with wildtype N-cadherin.


.0004 AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, ASP597TYR
  
RCV001007454...

In an infant girl (patient 3) who died at 2 months of age with agenesis of the corpus callosum, atrioventricular canal defect, and roving eye movements with saccadic intrusions (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo c.1789G-T transversion (c.1789G-T, NM_001792.5) in the CDH2 gene, resulting in an asp597-to-tyr (D597Y) substitution at a highly conserved residue in the calcium-binding site within the EC4-EC5 linker region. The mutation was not found in the gnomAD database. At 8 weeks of life, the proband developed progressive shortness of breath with cardiomegaly, pulmonary edema, and ultimately pulseless bradycardia, for which resuscitation attempts were unsuccessful.


.0005 AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, TYR676CYS
  
RCV000991214...

In a 24-year-old man (patient 7) with agenesis of corpus callosum, cardiac, ocular, and genital syndrome (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo c.2027A-G transition (c.2027A-G, NM_001792.5) in the CDH2 gene, resulting in a tyr676-to-cys (Y676C) substitution at a highly conserved residue within the EC5 domain. The mutation was not found in his unaffected parents or 2 sibs, or in the gnomAD database. Analysis in transfected L cells showed that cells expressing the Y676C variant displayed reduced aggregation compared to wildtype N-cadherin, forming smaller aggregates and resulting in a significantly lower index of aggregation compared to wildtype. In addition, mixed aggregation assay showed that fewer mixed aggregates were formed with the Y676C mutant compared to wildtype. The authors concluded that the Y676C mutation impairs the cell adhesion function of N-cadherin by affecting self-binding as well as trans-binding with wildtype N-cadherin.


.0006 AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, 2-BP DEL, 2563CT
  
RCV001007459...

In a 37-year-old woman (patient 8) with agenesis of corpus callosum, cardiac, ocular, and genital syndrome (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo 2-bp deletion (c.2563_2564delCT, NM_001792.5) in the CDH2 gene, causing a frameshift predicted to result in a premature termination codon (Leu855ValfsTer4) and a shortened cytoplasmic tail.


.0007 AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, 4-BP DUP, NT2564
  
RCV001007460...

In a 12.75-year-old boy (patient 9) with agenesis of corpus callosum, cardiac, ocular, and genital syndrome (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo 4-bp duplication (c.2564_2567dup) in the CDH2 gene, causing a frameshift predicted to result in a premature termination codon (Leu856PhefsTer5) and a shortened cytoplasmic tail.


.0008 AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, IVSDS, G-A, +1
  
RCV001195097...

In a boy (patient 1) with agenesis of the corpus callosum, coarctation of the aorta, and Peters anomaly (ACOGS; 618929), Reis et al. (2020) identified heterozygosity for a de novo splicing variant (c.702+1G-A) in the CDH2 gene, predicted to cause loss of the donor splice site and result in a premature termination codon. The mutation was not found in the gnomAD database.


.0009 AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, VAL162ASP
  
RCV001195098

In a boy of Jamaican descent (patient 2) with agenesis of the corpus callosum, Peters anomaly, and bilateral cryptorchidism (ACOGS; 618929), who was previously reported as patient 6 by Reis et al. (2008), Reis et al. (2020) identified heterozygosity for a c.485T-A transversion in the CDH2 gene, resulting in a val162-to-asp (V162D) substitution. The mutation was not found in the gnomAD database.


.0010 ATTENTION DEFICIT-HYPERACTIVITY DISORDER 8 (1 family)

CDH2, HIS150TYR
  

In 3 sibs, born of consanguineous Bedouin parents, with attention deficit-hyperactivity disorder-8 (ADHD8; 619957), Halperin et al. (2021) identified a homozygous c.355C-T transition (c.355C-T, NM_001792.4) in the CDH2 gene, resulting in a his150-to-tyr (H150Y) substitution at a highly conserved residue in the catalytic pocket of the furin protease active site. The mutation, which was found by a combination of homozygosity mapping and whole-exome sequencing and confirmed by Sanger sequencing, segregated with the disorder in the family. It was not present in the gnomAD database. In vitro functional expression studies showed that the mutation interfered with CDH2 protein processing, causing decreased proteolytic cleavage efficacy and defective N-cadherin protein maturation. Mutant mice with a homozygous H150Y mutation in the Cdh2 gene demonstrated motor hyperactivity, with greater traveling distance, increased velocity, and prolonged mobility time in the open-field exploratory test compared to controls. Mutant mice also showed an elevated startle amplitude in the acoustic startle reflex test (ASR) compared to controls, suggesting deficits in sensorimotor integration. Detailed analysis of the neurons from brains of mutant mice showed that homozygosity for the Cdh2 mutation was associated with a decrease in the size of the presynaptic vesicle cluster, a decrease in the readily releasable pool size of synaptic vesicles in neurons, and an attenuation of synaptic vesicle release compared to controls. The frequency of spontaneous synaptic release was also decreased compared to controls. There was reduced tyrosine hydroxylase expression and dopamine levels in the midbrain and prefrontal cortex. The authors suggested that deficits in synaptic formation due to aberrant processing of N-cadherin is the underlying mechanism for the clinical manifestations.


REFERENCES

  1. Accogli, A., Calabretta, S., St-Onge, J., Boudrahem-Addour, N., Dionne-Laporte, A., Joset, P., Azzarello-Burri, S., Rauch, A., Krier, J., Fieg, E., Pallais, J. C., Undiagnosed Diseases Network, and 21 others. De novo pathogenic variants in N-cadherin cause a syndromic neurodevelopmental disorder with corpus collosum [sic], axon, cardiac, ocular, and genital defects. Am. J. Hum. Genet. 105: 854-868, 2019. [PubMed: 31585109, images, related citations] [Full Text]

  2. Arregui, C., Pathre, P., Lilien, J., Balsamo, J. The nonreceptor tyrosine kinase Fer mediates cross-talk between N-cadherin and beta-1-integrins. J. Cell Biol. 149: 1263-1273, 2000. [PubMed: 10851023, images, related citations] [Full Text]

  3. Banh, C., Fugere, C., Brossay, L. Immunoregulatory functions of KLRG1 cadherin interactions are dependent on forward and reverse signaling. Blood 114: 5299-5306, 2009. [PubMed: 19855082, images, related citations] [Full Text]

  4. Garcia-Castro, M. I., Vielmetter, E., Bronner-Fraser, M. N-cadherin, a cell adhesion molecule involved in establishment of embryonic left-right asymmetry. Science 288: 1047-1051, 2000. [PubMed: 10807574, related citations] [Full Text]

  5. Halperin, D., Stavsky, A., Kadir, R., Drabkin, M., Wormser, O., Yogev, Y., Dolgin, V., Proskorovski-Ohayon, R., Perez, Y., Nudelman, H., Stoler, O., Rotblat, B., Lifschytz, T., Lotan, A., Meiri, G., Gitler, D., Birk, O. S. CDH2 mutation affecting N-cadherin function causes attention-deficit hyperactivity disorder in humans and mice. Nature Commun. 12: 6187, 2021. [PubMed: 34702855, images, related citations] [Full Text]

  6. Hayashi, T., Carthew, R. W. Surface mechanics mediate pattern formation in the developing retina. Nature 431: 647-652, 2004. [PubMed: 15470418, related citations] [Full Text]

  7. Hermiston, M. L., Gordon, J. I. Inflammatory bowel disease and adenomas in mice expressing a dominant negative N-cadherin. Science 270: 1203-1206, 1995. [PubMed: 7502046, related citations] [Full Text]

  8. Mayosi, B. M., Fish, M., Shaboodien, G., Mastantuono, E., Kraus, S., Wieland, T., Kotta, M.-C., Chin, A., Laing, N., Ntusi, N. B. A., Chong, M., Horsfall, C., Pimstone, S. N., Gentilini, D., Parati, G., Strom, T.-M., Meitinger, T., Pare, G., Schwartz, P. J., Crotti, L. Identification of cadherin 2 (CDH2) mutations in arrhythmogenic right ventricular cardiomyopathy. Circ. Cardiovasc. Genet. 10: e001605, 2017. Note: Electronic Article. [PubMed: 28280076, related citations] [Full Text]

  9. Miyatani, S., Copeland, N. G., Gilbert, D. J., Jenkins, N. A., Takeichi, M. Genomic structure and chromosomal mapping of the mouse N-cadherin gene. Proc. Nat. Acad. Sci. 89: 8443-8447, 1992. [PubMed: 1528849, related citations] [Full Text]

  10. Reid, R. A., Hemperly, J. J. Human N-cadherin: nucleotide and deduced amino acid sequence. Nucleic Acids Res. 18: 5896 only, 1990. [PubMed: 2216790, related citations] [Full Text]

  11. Reis, L. M., Houssin, N. S., Zamora, C., Abdul-Rahman, O., Kalish, J. M., Zackai, E. H., Plageman, T. F., Jr. Novel variants in CDH2 are associated with a new syndrome including Peters anomaly. Clin. Genet. 97: 502-508, 2020. [PubMed: 31650526, images, related citations] [Full Text]

  12. Reis, L. M., Tyler, R. C., Abdul-Rahman, O., Trapane, P., Wallerstein, R., Broome, D., Hoffman, J., Khan, A., Paradiso, C., Ron, N., Bergner, A., Semina, E. V. Mutation analysis of B3GALTL in Peters plus syndrome. Am. J. Med. Genet. 146A: 2603-2610, 2008. [PubMed: 18798333, images, related citations] [Full Text]

  13. Rubinek, T., Yu, R., Hadani, M., Barkai, G., Nass, D., Melmed, S., Shimon, I. The cell adhesion molecules N-cadherin and neural cell adhesion molecule regulate human growth hormone: a novel mechanism for regulating pituitary hormone secretion. J. Clin. Endocr. Metab. 88: 3724-3730, 2003. [PubMed: 12915661, related citations] [Full Text]

  14. Stumpf, A. M. Personal Communication. Baltimore, Md. 06/23/2020.

  15. Takeichi, M. Cadherins: a molecular family essential for selective cell-cell adhesion and animal morphogenesis. Trends Genet. 3: 213-217, 1987.

  16. Tanaka, H., Shan, W., Phillips, G. R., Arndt, K., Bozdagi, O., Shapiro, L., Huntley, G. W., Benson, D. L., Colman, D. R. Molecular modification of N-cadherin in response to synaptic activity. Neuron 25: 93-107, 2000. [PubMed: 10707975, related citations] [Full Text]

  17. Togashi, H., Abe, K., Mizoguchi, A., Takaoka, K., Chisaka, O., Takeichi, M. Cadherin regulates dendritic spine morphogenesis. Neuron 35: 77-89, 2002. [PubMed: 12123610, related citations] [Full Text]

  18. Tsai, T. Y.-C., Sikora, M., Xia, P., Colak-Champollion, T., Knaut, H., Heisenberg, C.-P., Megason, S. G. An adhesion code ensures robust pattern formation during tissue morphogenesis. Science 370: 113-116, 2020. [PubMed: 33004519, images, related citations] [Full Text]

  19. Van Aken, E. H., Papeleu, P., De Potter, P., Bruyneel, E., Philippe, J., Seregard, S., Kvanta, A., De Laey, J.-J., Mareel, M. M. Structure and function of the N-cadherin/catenin complex in retinoblastoma. Invest. Ophthal. Vis. Sci. 43: 595-602, 2002. [PubMed: 11867572, related citations]

  20. Wallis, J., Fox, M. F., Walsh, F. S. Structure of the human N-cadherin gene: YAC analysis and fine chromosomal mapping to 18q11.2. Genomics 22: 172-179, 1994. [PubMed: 7959764, related citations] [Full Text]

  21. Walsh, F. S., Barton, C. H., Putt, W., Moore, S. E., Kelsell, D., Spurr, N., Goodfellow, P. N. N-cadherin gene maps to human chromosome 18 and is not linked to the E-cadherin gene. J. Neurochem. 55: 805-812, 1990. [PubMed: 2384753, related citations] [Full Text]


Cassandra L. Kniffin - updated : 07/12/2022
Ada Hamosh - updated : 12/07/2020
Marla J. F. O'Neill - updated : 06/26/2020
Marla J. F. O'Neill - updated : 06/23/2020
Paul J. Converse - updated : 11/30/2011
Ada Hamosh - updated : 1/27/2005
John A. Phillips, III - updated : 10/14/2004
Anne M. Stumpf - updated : 10/14/2004
Cassandra L. Kniffin - updated : 2/20/2003
Jane Kelly - updated : 11/7/2002
Paul J. Converse - updated : 8/1/2000
Paul J. Converse - updated : 5/17/2000
Ada Hamosh - updated : 5/11/2000
Creation Date:
Victor A. McKusick : 7/24/1991
alopez : 07/13/2022
ckniffin : 07/12/2022
mgross : 12/07/2020
alopez : 06/26/2020
alopez : 06/23/2020
mgross : 11/30/2011
carol : 10/24/2008
wwang : 2/7/2005
wwang : 1/31/2005
terry : 1/27/2005
alopez : 10/15/2004
alopez : 10/14/2004
alopez : 10/14/2004
alopez : 10/14/2004
cwells : 2/27/2003
ckniffin : 2/20/2003
carol : 11/7/2002
mgross : 8/1/2000
mgross : 5/17/2000
alopez : 5/11/2000
terry : 5/11/2000
psherman : 8/27/1998
carol : 8/6/1998
mark : 11/16/1995
terry : 8/8/1994
carol : 5/12/1994
carol : 10/7/1992
supermim : 3/16/1992
carol : 11/6/1991

* 114020

CADHERIN 2; CDH2


Alternative titles; symbols

CADHERIN, NEURONAL
N-CADHERIN; NCAD
CALCIUM-DEPENDENT ADHESION PROTEIN, NEURONAL; CDHN


HGNC Approved Gene Symbol: CDH2

Cytogenetic location: 18q12.1     Genomic coordinates (GRCh38): 18:27,932,879-28,177,130 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
18q12.1 ?Attention deficit-hyperactivity disorder 8 619957 Autosomal recessive 3
Agenesis of corpus callosum, cardiac, ocular, and genital syndrome 618929 Autosomal dominant 3
Arrhythmogenic right ventricular dysplasia 14 618920 Autosomal dominant 3

TEXT

Description

The CDH2 gene encodes N (neuronal)-cadherin, which has an essential role in the early steps of brain morphogenesis (summary by Halperin et al., 2021).


Cloning and Expression

Reid and Hemperly (1990) obtained a full-length NCAD clone from a Kelly neuroblastoma library. The NCAD gene encodes a 907-amino acid protein that includes a 159-amino acid signal sequence. Human and mouse nucleotide sequences are 96% identical.


Gene Structure

Miyatani et al. (1992) showed that the mouse Ncad gene consists of 16 exons dispersed over more than 200 kb of genomic DNA. The large size of the N-cadherin gene, compared with its cDNA (4.3 kb), was ascribed to the fact that the first and second introns are 34.2 kb and more than 100 kb long, respectively. Miyatani et al. (1992) compared the NCAD, liver cell adhesion molecule (LCAM; see 192090), and PCAD (CDH3) genes and showed that the exon-intron boundaries are fully conserved between them, except that the first exon of P-cadherin includes the first and second exons of the other 2 genes. Also, the second intron, which is equivalent to the first intron in P-cadherin, is exceptionally large and this structural feature is conserved in all 3 of these genes.

Wallis et al. (1994) demonstrated that the human N-cadherin gene contains 16 exons and its sequence is highly similar to both the mouse NCAD gene (including the large first and second introns) and other cadherin genes.


Mapping

In Southern analysis of a panel of somatic cell hybrids, Walsh et al. (1990) mapped the NCAD gene to chromosome 18. By interspecific backcross analysis, Miyatani et al. (1992) found that the gene in the mouse is located in the proximal region of chromosome 18.

By in situ hybridization, Wallis et al. (1994) refined the map position of N-cadherin to 18q11.2.

Stumpf (2020) mapped the CDH2 gene to chromosome 18q12.1 based on an alignment of the CDH2 sequence (GenBank BC036470) with the genomic sequence (GRCh38).


Gene Family

The cadherin gene family encode proteins that mediate calcium-ion-dependent adhesion (Takeichi, 1987). Three members of this family are E-cadherin, N-cadherin, and P-cadherin. E-cadherin appears to be identical to the protein called uvomorulin. N-cadherin is expressed in the brain and skeletal and cardiac muscle.


Gene Function

Hermiston and Gordon (1995) noted that the mouse intestinal epithelium expresses a sequence of 'developmental events'--proliferation, lineage allocation, migration, differentiation, and death--throughout life. Proliferation is confined to the crypts of Lieberkuhn. The crypt's multipotent stem cell gives rise to enterocytes, mucus-producing goblet cells, enteroendocrine cells, and Paneth cells. Cells of these 4 lineages differentiate during an orderly migration and are frequently eliminated by apoptosis and exfoliation or phagocytosis. Renewal is rapid (3 to 20 days). Results from cell culture studies indicate that cadherin-catenin complexes regulate cell polarity, formation of junctional complexes, migration, and proliferation. Hermiston and Gordon (1995) transfected embryonic stem cells with a dominant-negative N-cadherin mutant under the control of promoters active in small intestinal epithelial cells and introduced them into C57BL/6 blastocysts. Analysis of adult chimeric mice revealed that expression of the mutant along the entire crypt-villus axis, but not in the villus epithelium alone, produced an inflammatory bowel disease resembling Crohn disease (see 266600). The mutation perturbed proliferation, migration, and death patterns in crypts, leading to adenomas. The model provided insights into cadherin function in an adult organ and the factors underlying inflammatory bowel disease and intestinal neoplasia.

Within the bilaterally symmetric vertebrate body plan, many organs develop asymmetrically. Garcia-Castro et al. (2000) demonstrated that the cell adhesion molecule N-cadherin is one of the earliest proteins to be asymmetrically expressed in the chicken embryo and that its activity is required during gastrulation for proper establishment of the left-right axis. Blocking N-cadherin function randomizes heart looping and alters the expression of Snail (604238) and Pitx2 (601542), later components of the molecular cascade that regulates left-right asymmetry. However, the expression of other components of this cascade, Nodal (601265) and Lefty (see 601877), was unchanged after blocking N-cadherin function, suggesting the existence of parallel pathways in the establishment of left-right morphogenesis. Garcia-Castro et al. (2000) concluded that their results suggest that N-cadherin-mediated cell adhesion events are required for establishment of left-right asymmetry.

At certain central nervous system synapses, pre- to postsynaptic adhesion is mediated at least in part by CDH2. Tanaka et al. (2000) demonstrated that upon depolarization of cultured hippocampal neurons by potassium treatment, or the application of N-methyl-D-aspartate or alpha-latrotoxin, synaptic CDH2 dimerizes and becomes markedly protease resistant. The resistance persisted for at least 2 hours while other surface molecules, including other cadherins, were completely degraded. The acquisition of protease resistance and dimerization of CDH2 was not dependent on new protein synthesis. Tanaka et al. (2000) concluded that synaptic adhesion is dynamically and locally controlled and is modulated by synaptic activity.

Arregui et al. (2000) demonstrated that cell-permeable (Trojan) peptides containing the third helix of the antennapedia homeodomain fused to a peptide mimicking the juxtamembrane (JMP) region of the cytoplasmic domain of CDH2 result in the inhibition of both CDH2 and beta-1 integrin (ITGB1; 135630) function. Microscopic analysis showed that expression of JMP, which binds to the cytoplasmic domain of CDH2, results in a reduction of neurite outgrowth on cadherin substrates. Treatment of cells with JMP resulted in the release of FER (176942) from the cadherin complex and its accumulation in the integrin complex. The accumulation of FER in the integrin complex and the inhibitory effects of JMP could be reversed with a peptide that mimics the first coiled-coil domain of FER. The results suggested that FER mediates crosstalk between CDH2 and ITGB1.

Van Aken et al. (2002) studied the cadherin-catenin complex in retinoblastoma and normal retina tissues. In both cases, they found that N-cadherin was associated with alpha- and beta-catenin (116805; 116806) but not with E-cadherin (CDH1; 192090) or P-cadherin (CDH3; 114021), retinoblastoma cells, in contrast with normal retina, expressed an N-cadherin/catenin complex that was irregularly distributed and weakly linked to the cytoskeleton. In retinoblastoma, this complex acted as an invasion promoter.

In cultured rat hippocampal neurons, Togashi et al. (2002) showed that blockade of the known hippocampal cadherins (cadherin-8, 603008; cadherin-11, 600023; and N-cadherin) resulted in alterations of dendritic spine morphology, such as filopodia-like elongation of the spine and bifurcation of its head structure, along with concomitant disruption of the distribution of postsynaptic proteins and perturbation of synaptic vesicle recycling. The findings suggested that cadherins regulate dendritic spine morphogenesis and related synaptic functions.

Rubinek et al. (2003) studied the role of pituicyte cell-cell contact mediated by CDH2 and NCAM1 (116930) in the regulation of GH (139250) secretion. RT-PCR showed CDH2 mRNA expression in 8 of 12 GH-secreting adenomas compared with 1 of 7 prolactin-cell adenomas. CDH2 and NCAM1 were similarly expressed in adenomas and in adult and fetal normal pituitary tissues. Cell adhesion molecule (CAM) stimulation increased GH secretion from pituitary fetal cultures by 40 to 60% and also from cultured GH adenoma cells by 40 to 75%. Disrupting CDH2 homophilic binding by anti-CDH2 antibody decreased fetal, but not tumorous, GH secretion by 40%. This study indicated that pituitary cell-cell contact mediated by homophilic interactions between adhesion molecules regulates human GH secretion.

Hayashi and Carthew (2004) investigated the physical basis of biologic patterning of the Drosophila retina in vivo. They demonstrated that E-cadherin and N-cadherin mediate apical adhesion between retina epithelial cells. Differential expression of N-cadherin within a subgroup of retinal cells (cone cells) caused them to form an overall shape that minimized their surface contact with surrounding cells. The cells within this group, in both normal and experimentally manipulated conditions, packed together in the same way as soap bubbles do. The shaping of cone cell group and packing of its components precisely imitated the physical tendency for surfaces to be minimized. Hayashi and Carthew (2004) concluded that simple patterned expression of N-cadherin resulted in a complex spatial pattern of cells owing to cellular surface mechanics.

Banh et al. (2009) showed that the first 2 extracellular domains of N-cadherin interacted with the inhibitory receptor KLRG1 (604874), blocked interaction of KLRG1 with E-cadherin, and could regulate KLRG1 signaling.

Tsai et al. (2020) noted that, in zebrafish spinal cord, neural progenitors form stereotypic patterns despite noisy morphogen signaling and large-scale cellular rearrangements during morphogenesis and growth. By directly measuring adhesion forces and preferences for 3 types of endogenous neural progenitors, Tsai et al. (2020) provided evidence for the differential adhesion model, in which differences in intercellular adhesion mediate cell sorting. Cell type-specific combinatorial expression of different classes of cadherins, including N-cadherin, cadherin-11, and protocadherin-19 (PCDH19; 300460), resulted in homotypic preference ex vivo and patterning robustness in vivo. The differential adhesion code was regulated by the shh (600725) morphogen gradient. Tsai et al. (2020) proposed that robust patterning during tissue morphogenesis results from interplay between adhesion-based self-organization and morphogen-directed patterning.


Molecular Genetics

Arrhythmogenic Right Ventricular Dysplasia 14

In affected members of a 3-generation South African family segregating autosomal dominant arrhythmogenic right ventricular dysplasia (ARVD14; 618920), Mayosi et al. (2017) identified heterozygosity for a missense mutation in the CDH2 gene (Q229P; 114020.0001) that segregated fully with disease. Screening of 73 additional ARVD probands revealed 1 patient with another missense mutation in CDH2 (D407N; 114020.0002).

Agenesis of Corpus Callosum, Cardiac, Ocular, and Genital Syndrome

In 9 unrelated patients with agenesis of corpus callosum, cardiac, ocular, and genital syndrome (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for de novo mutations in the CDH2 gene (see, e.g., 114020.0003-114020.0007). Functional analysis indicated that the mutations impair the cell adhesion function of N-cadherin by affecting self-binding as well as trans-binding with wildtype N-cadherin. Noting the neurodevelopmental features observed in the affected individuals, including agenesis of the corpus callosum, periventricular nodular heterotopias, hyposmia, mirror movements, Duane anomaly, and abnormal shoulder muscle innervation, the authors suggested that CDH2 plays a critical role in neuronal migration and axon pathfinding.

In 4 unrelated patients with ACOGS, Reis et al. (2020) identified heterozygosity for mutations in the CDH2 gene (see, e.g., 114020.0008-114020.0009).

Attention Deficit-Hyperactivity Disorder 8

In 3 sibs, born of consanguineous Bedouin parents, with attention deficit-hyperactivity disorder-8 (ADHD8; 619957), Halperin et al. (2021) identified a homozygous missense mutation in the CDH2 gene (H150Y; 114020.0010). The mutation, which was found by a combination of homozygosity mapping and whole-exome sequencing and confirmed by Sanger sequencing, segregated with the disorder in the family. It was not present in the gnomAD database. In vitro functional expression studies showed that the mutation interfered with CDH2 protein processing, causing decreased proteolytic cleavage efficacy and defective N-cadherin protein maturation. Mutant mice with a homozygous H150Y mutation in the Cdh2 gene demonstrated motor hyperactivity, with greater traveling distance, increased velocity, and prolonged mobility time in the open-field exploratory test compared to controls (see ANIMAL MODEL).


Animal Model

Halperin et al. (2021) found that mutant mice homozygous for the H150Y mutation in the Cdh2 gene demonstrated hyperactive behavior. Mutant mice also showed an elevated startle amplitude in the acoustic startle reflex test (ASR) compared to controls, suggesting deficits in sensorimotor integration. Detailed analysis of the neurons from brains of mutant mice showed that homozygosity for the Cdh2 mutation was associated with a decrease in the size of the presynaptic vesicle cluster, a decrease in the readily releasable pool size of synaptic vesicles in neurons, and an attenuation of synaptic vesicle release compared to controls. The frequency of spontaneous synaptic release was also decreased compared to controls. There was reduced tyrosine hydroxylase expression and dopamine levels in the midbrain and prefrontal cortex. The authors suggested that deficits in synaptic formation due to aberrant processing of N-cadherin is the underlying mechanism for the clinical manifestations.


ALLELIC VARIANTS 10 Selected Examples):

.0001   ARRHYTHMOGENIC RIGHT VENTRICULAR DYSPLASIA, FAMILIAL, 14

CDH2, GLN229PRO
SNP: rs965753331, gnomAD: rs965753331, ClinVar: RCV001194670

In affected members of a 3-generation white South African family (ACM2) with arrhythmogenic right ventricular dysplasia (ARVD14; 618920), Mayosi et al. (2017) identified heterozygosity for a c.686A-C transversion in the CDH2 gene, resulting in a gln229-to-pro (Q229P) substitution at a highly conserved residue within the EC1 domain. The mutation segregated fully with disease in the family and was not found in 200 white South African control chromosomes or in the 1000 Genomes or ExAC databases.


.0002   ARRHYTHMOGENIC RIGHT VENTRICULAR DYSPLASIA, FAMILIAL, 14

CDH2, ASP407ASN ({dbSNP rs568089577})
SNP: rs568089577, gnomAD: rs568089577, ClinVar: RCV001194671, RCV002508294

In a white South African man (ACM11) with arrhythmogenic right ventricular dysplasia (ARVD14; 618920), Mayosi et al. (2017) identified heterozygosity for a c.1219G-A transition in the CDH2 gene, resulting in an asp407-to-asn (D407N) substitution at a highly conserved residue within the EC3 domain. The mutation was not found in his unaffected mother or in 200 white South African control chromosomes or the Helmholtz database; however, it was present once in the 1000 Genomes database (minor allele frequency, 0.0002) and once in the ExAC database (MAF, 0.000008).


.0003   AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, ASP597ASN
SNP: rs1599011050, ClinVar: RCV001007453, RCV001195092, RCV001261827

In an 9-year-old boy (patient 2) with agenesis of the corpus callosum, tricuspid regurgitation, Duane anomaly and right ptosis, and micropenis (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo c.1789G-A transition (c.1789G-A, NM_001792.5) in the CDH2 gene, resulting in an asp597-to-asn (D597N) substitution at a highly conserved residue in the calcium-binding site within the EC4-EC5 linker region. The mutation was not found in the gnomAD database. Analysis in transfected L cells showed that cells expressing the D597N variant displayed reduced aggregation compared to wildtype N-cadherin, forming smaller aggregates and resulting in a significantly lower index of aggregation compared to wildtype. In addition, mixed aggregation assay showed that fewer mixed aggregates were formed with the D597N mutant compared to wildtype. The authors concluded that the D597N mutation impairs the cell adhesion function of N-cadherin by affecting self-binding as well as trans-binding with wildtype N-cadherin.


.0004   AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, ASP597TYR
SNP: rs1599011050, ClinVar: RCV001007454, RCV001195093

In an infant girl (patient 3) who died at 2 months of age with agenesis of the corpus callosum, atrioventricular canal defect, and roving eye movements with saccadic intrusions (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo c.1789G-T transversion (c.1789G-T, NM_001792.5) in the CDH2 gene, resulting in an asp597-to-tyr (D597Y) substitution at a highly conserved residue in the calcium-binding site within the EC4-EC5 linker region. The mutation was not found in the gnomAD database. At 8 weeks of life, the proband developed progressive shortness of breath with cardiomegaly, pulmonary edema, and ultimately pulseless bradycardia, for which resuscitation attempts were unsuccessful.


.0005   AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, TYR676CYS
SNP: rs199984052, gnomAD: rs199984052, ClinVar: RCV000991214, RCV001007458, RCV001195094, RCV001261829, RCV002462248

In a 24-year-old man (patient 7) with agenesis of corpus callosum, cardiac, ocular, and genital syndrome (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo c.2027A-G transition (c.2027A-G, NM_001792.5) in the CDH2 gene, resulting in a tyr676-to-cys (Y676C) substitution at a highly conserved residue within the EC5 domain. The mutation was not found in his unaffected parents or 2 sibs, or in the gnomAD database. Analysis in transfected L cells showed that cells expressing the Y676C variant displayed reduced aggregation compared to wildtype N-cadherin, forming smaller aggregates and resulting in a significantly lower index of aggregation compared to wildtype. In addition, mixed aggregation assay showed that fewer mixed aggregates were formed with the Y676C mutant compared to wildtype. The authors concluded that the Y676C mutation impairs the cell adhesion function of N-cadherin by affecting self-binding as well as trans-binding with wildtype N-cadherin.


.0006   AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, 2-BP DEL, 2563CT
SNP: rs1598982488, ClinVar: RCV001007459, RCV001195095, RCV001261830

In a 37-year-old woman (patient 8) with agenesis of corpus callosum, cardiac, ocular, and genital syndrome (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo 2-bp deletion (c.2563_2564delCT, NM_001792.5) in the CDH2 gene, causing a frameshift predicted to result in a premature termination codon (Leu855ValfsTer4) and a shortened cytoplasmic tail.


.0007   AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, 4-BP DUP, NT2564
SNP: rs1598982483, ClinVar: RCV001007460, RCV001195096, RCV001261831

In a 12.75-year-old boy (patient 9) with agenesis of corpus callosum, cardiac, ocular, and genital syndrome (ACOGS; 618929), Accogli et al. (2019) identified heterozygosity for a de novo 4-bp duplication (c.2564_2567dup) in the CDH2 gene, causing a frameshift predicted to result in a premature termination codon (Leu856PhefsTer5) and a shortened cytoplasmic tail.


.0008   AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, IVSDS, G-A, +1
SNP: rs2013040501, ClinVar: RCV001195097, RCV002290630

In a boy (patient 1) with agenesis of the corpus callosum, coarctation of the aorta, and Peters anomaly (ACOGS; 618929), Reis et al. (2020) identified heterozygosity for a de novo splicing variant (c.702+1G-A) in the CDH2 gene, predicted to cause loss of the donor splice site and result in a premature termination codon. The mutation was not found in the gnomAD database.


.0009   AGENESIS OF CORPUS CALLOSUM, CARDIAC, OCULAR, AND GENITAL SYNDROME

CDH2, VAL162ASP
SNP: rs2013111940, ClinVar: RCV001195098

In a boy of Jamaican descent (patient 2) with agenesis of the corpus callosum, Peters anomaly, and bilateral cryptorchidism (ACOGS; 618929), who was previously reported as patient 6 by Reis et al. (2008), Reis et al. (2020) identified heterozygosity for a c.485T-A transversion in the CDH2 gene, resulting in a val162-to-asp (V162D) substitution. The mutation was not found in the gnomAD database.


.0010   ATTENTION DEFICIT-HYPERACTIVITY DISORDER 8 (1 family)

CDH2, HIS150TYR

In 3 sibs, born of consanguineous Bedouin parents, with attention deficit-hyperactivity disorder-8 (ADHD8; 619957), Halperin et al. (2021) identified a homozygous c.355C-T transition (c.355C-T, NM_001792.4) in the CDH2 gene, resulting in a his150-to-tyr (H150Y) substitution at a highly conserved residue in the catalytic pocket of the furin protease active site. The mutation, which was found by a combination of homozygosity mapping and whole-exome sequencing and confirmed by Sanger sequencing, segregated with the disorder in the family. It was not present in the gnomAD database. In vitro functional expression studies showed that the mutation interfered with CDH2 protein processing, causing decreased proteolytic cleavage efficacy and defective N-cadherin protein maturation. Mutant mice with a homozygous H150Y mutation in the Cdh2 gene demonstrated motor hyperactivity, with greater traveling distance, increased velocity, and prolonged mobility time in the open-field exploratory test compared to controls. Mutant mice also showed an elevated startle amplitude in the acoustic startle reflex test (ASR) compared to controls, suggesting deficits in sensorimotor integration. Detailed analysis of the neurons from brains of mutant mice showed that homozygosity for the Cdh2 mutation was associated with a decrease in the size of the presynaptic vesicle cluster, a decrease in the readily releasable pool size of synaptic vesicles in neurons, and an attenuation of synaptic vesicle release compared to controls. The frequency of spontaneous synaptic release was also decreased compared to controls. There was reduced tyrosine hydroxylase expression and dopamine levels in the midbrain and prefrontal cortex. The authors suggested that deficits in synaptic formation due to aberrant processing of N-cadherin is the underlying mechanism for the clinical manifestations.


REFERENCES

  1. Accogli, A., Calabretta, S., St-Onge, J., Boudrahem-Addour, N., Dionne-Laporte, A., Joset, P., Azzarello-Burri, S., Rauch, A., Krier, J., Fieg, E., Pallais, J. C., Undiagnosed Diseases Network, and 21 others. De novo pathogenic variants in N-cadherin cause a syndromic neurodevelopmental disorder with corpus collosum [sic], axon, cardiac, ocular, and genital defects. Am. J. Hum. Genet. 105: 854-868, 2019. [PubMed: 31585109] [Full Text: https://doi.org/10.1016/j.ajhg.2019.09.005]

  2. Arregui, C., Pathre, P., Lilien, J., Balsamo, J. The nonreceptor tyrosine kinase Fer mediates cross-talk between N-cadherin and beta-1-integrins. J. Cell Biol. 149: 1263-1273, 2000. [PubMed: 10851023] [Full Text: https://doi.org/10.1083/jcb.149.6.1263]

  3. Banh, C., Fugere, C., Brossay, L. Immunoregulatory functions of KLRG1 cadherin interactions are dependent on forward and reverse signaling. Blood 114: 5299-5306, 2009. [PubMed: 19855082] [Full Text: https://doi.org/10.1182/blood-2009-06-228353]

  4. Garcia-Castro, M. I., Vielmetter, E., Bronner-Fraser, M. N-cadherin, a cell adhesion molecule involved in establishment of embryonic left-right asymmetry. Science 288: 1047-1051, 2000. [PubMed: 10807574] [Full Text: https://doi.org/10.1126/science.288.5468.1047]

  5. Halperin, D., Stavsky, A., Kadir, R., Drabkin, M., Wormser, O., Yogev, Y., Dolgin, V., Proskorovski-Ohayon, R., Perez, Y., Nudelman, H., Stoler, O., Rotblat, B., Lifschytz, T., Lotan, A., Meiri, G., Gitler, D., Birk, O. S. CDH2 mutation affecting N-cadherin function causes attention-deficit hyperactivity disorder in humans and mice. Nature Commun. 12: 6187, 2021. [PubMed: 34702855] [Full Text: https://doi.org/10.1038/s41467-021-26426-1]

  6. Hayashi, T., Carthew, R. W. Surface mechanics mediate pattern formation in the developing retina. Nature 431: 647-652, 2004. [PubMed: 15470418] [Full Text: https://doi.org/10.1038/nature02952]

  7. Hermiston, M. L., Gordon, J. I. Inflammatory bowel disease and adenomas in mice expressing a dominant negative N-cadherin. Science 270: 1203-1206, 1995. [PubMed: 7502046] [Full Text: https://doi.org/10.1126/science.270.5239.1203]

  8. Mayosi, B. M., Fish, M., Shaboodien, G., Mastantuono, E., Kraus, S., Wieland, T., Kotta, M.-C., Chin, A., Laing, N., Ntusi, N. B. A., Chong, M., Horsfall, C., Pimstone, S. N., Gentilini, D., Parati, G., Strom, T.-M., Meitinger, T., Pare, G., Schwartz, P. J., Crotti, L. Identification of cadherin 2 (CDH2) mutations in arrhythmogenic right ventricular cardiomyopathy. Circ. Cardiovasc. Genet. 10: e001605, 2017. Note: Electronic Article. [PubMed: 28280076] [Full Text: https://doi.org/10.1161/CIRCGENETICS.116.001605]

  9. Miyatani, S., Copeland, N. G., Gilbert, D. J., Jenkins, N. A., Takeichi, M. Genomic structure and chromosomal mapping of the mouse N-cadherin gene. Proc. Nat. Acad. Sci. 89: 8443-8447, 1992. [PubMed: 1528849] [Full Text: https://doi.org/10.1073/pnas.89.18.8443]

  10. Reid, R. A., Hemperly, J. J. Human N-cadherin: nucleotide and deduced amino acid sequence. Nucleic Acids Res. 18: 5896 only, 1990. [PubMed: 2216790] [Full Text: https://doi.org/10.1093/nar/18.19.5896]

  11. Reis, L. M., Houssin, N. S., Zamora, C., Abdul-Rahman, O., Kalish, J. M., Zackai, E. H., Plageman, T. F., Jr. Novel variants in CDH2 are associated with a new syndrome including Peters anomaly. Clin. Genet. 97: 502-508, 2020. [PubMed: 31650526] [Full Text: https://doi.org/10.1111/cge.13660]

  12. Reis, L. M., Tyler, R. C., Abdul-Rahman, O., Trapane, P., Wallerstein, R., Broome, D., Hoffman, J., Khan, A., Paradiso, C., Ron, N., Bergner, A., Semina, E. V. Mutation analysis of B3GALTL in Peters plus syndrome. Am. J. Med. Genet. 146A: 2603-2610, 2008. [PubMed: 18798333] [Full Text: https://doi.org/10.1002/ajmg.a.32498]

  13. Rubinek, T., Yu, R., Hadani, M., Barkai, G., Nass, D., Melmed, S., Shimon, I. The cell adhesion molecules N-cadherin and neural cell adhesion molecule regulate human growth hormone: a novel mechanism for regulating pituitary hormone secretion. J. Clin. Endocr. Metab. 88: 3724-3730, 2003. [PubMed: 12915661] [Full Text: https://doi.org/10.1210/jc.2003-030090]

  14. Stumpf, A. M. Personal Communication. Baltimore, Md. 06/23/2020.

  15. Takeichi, M. Cadherins: a molecular family essential for selective cell-cell adhesion and animal morphogenesis. Trends Genet. 3: 213-217, 1987.

  16. Tanaka, H., Shan, W., Phillips, G. R., Arndt, K., Bozdagi, O., Shapiro, L., Huntley, G. W., Benson, D. L., Colman, D. R. Molecular modification of N-cadherin in response to synaptic activity. Neuron 25: 93-107, 2000. [PubMed: 10707975] [Full Text: https://doi.org/10.1016/s0896-6273(00)80874-0]

  17. Togashi, H., Abe, K., Mizoguchi, A., Takaoka, K., Chisaka, O., Takeichi, M. Cadherin regulates dendritic spine morphogenesis. Neuron 35: 77-89, 2002. [PubMed: 12123610] [Full Text: https://doi.org/10.1016/s0896-6273(02)00748-1]

  18. Tsai, T. Y.-C., Sikora, M., Xia, P., Colak-Champollion, T., Knaut, H., Heisenberg, C.-P., Megason, S. G. An adhesion code ensures robust pattern formation during tissue morphogenesis. Science 370: 113-116, 2020. [PubMed: 33004519] [Full Text: https://doi.org/10.1126/science.aba6637]

  19. Van Aken, E. H., Papeleu, P., De Potter, P., Bruyneel, E., Philippe, J., Seregard, S., Kvanta, A., De Laey, J.-J., Mareel, M. M. Structure and function of the N-cadherin/catenin complex in retinoblastoma. Invest. Ophthal. Vis. Sci. 43: 595-602, 2002. [PubMed: 11867572]

  20. Wallis, J., Fox, M. F., Walsh, F. S. Structure of the human N-cadherin gene: YAC analysis and fine chromosomal mapping to 18q11.2. Genomics 22: 172-179, 1994. [PubMed: 7959764] [Full Text: https://doi.org/10.1006/geno.1994.1358]

  21. Walsh, F. S., Barton, C. H., Putt, W., Moore, S. E., Kelsell, D., Spurr, N., Goodfellow, P. N. N-cadherin gene maps to human chromosome 18 and is not linked to the E-cadherin gene. J. Neurochem. 55: 805-812, 1990. [PubMed: 2384753] [Full Text: https://doi.org/10.1111/j.1471-4159.1990.tb04563.x]


Contributors:
Cassandra L. Kniffin - updated : 07/12/2022
Ada Hamosh - updated : 12/07/2020
Marla J. F. O'Neill - updated : 06/26/2020
Marla J. F. O'Neill - updated : 06/23/2020
Paul J. Converse - updated : 11/30/2011
Ada Hamosh - updated : 1/27/2005
John A. Phillips, III - updated : 10/14/2004
Anne M. Stumpf - updated : 10/14/2004
Cassandra L. Kniffin - updated : 2/20/2003
Jane Kelly - updated : 11/7/2002
Paul J. Converse - updated : 8/1/2000
Paul J. Converse - updated : 5/17/2000
Ada Hamosh - updated : 5/11/2000

Creation Date:
Victor A. McKusick : 7/24/1991

Edit History:
alopez : 07/13/2022
ckniffin : 07/12/2022
mgross : 12/07/2020
alopez : 06/26/2020
alopez : 06/23/2020
mgross : 11/30/2011
carol : 10/24/2008
wwang : 2/7/2005
wwang : 1/31/2005
terry : 1/27/2005
alopez : 10/15/2004
alopez : 10/14/2004
alopez : 10/14/2004
alopez : 10/14/2004
cwells : 2/27/2003
ckniffin : 2/20/2003
carol : 11/7/2002
mgross : 8/1/2000
mgross : 5/17/2000
alopez : 5/11/2000
terry : 5/11/2000
psherman : 8/27/1998
carol : 8/6/1998
mark : 11/16/1995
terry : 8/8/1994
carol : 5/12/1994
carol : 10/7/1992
supermim : 3/16/1992
carol : 11/6/1991