CDH2 Human

Cadherin 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Neural Development

  • Synaptogenesis: CDH2 stabilizes dendritic spines and regulates synaptic vesicle clustering, impacting neurotransmitter release (e.g., dopamine) .

  • Neurulation: Essential for neural tube closure and radial glial cell migration .

  • Neurodevelopmental Disorders: Mutations (e.g., p.H150Y) disrupt presynaptic function and dopamine signaling, linking CDH2 to ADHD and autism .

Cardiac Function

  • Intercalated Discs: Maintains electrical and mechanical coupling between cardiomyocytes. Dysregulation correlates with dilated cardiomyopathy .

Stem Cell Biology

  • Pluripotency: In mouse epiblast stem cells (mEpiSCs), CDH2 stabilizes FGFR1, enhancing FGF2-mediated differentiation .

Neurodevelopmental Disorders

MutationPhenotypeMechanism
p.H150YADHD, hyperactivity in miceImpaired synaptic vesicle clustering .
N706S/N845SOCD/Tourette disorder susceptibilityReduced protein stability and adhesion .

Cardiac and Syndromic Disorders

  • Dilated Cardiomyopathy: Altered CDH2 expression disrupts intercalated disc integrity .

  • Agenesis of Corpus Callosum: Linked to CDH2 mutations affecting neural migration .

Cancer

  • Metastasis: Overexpression promotes epithelial-mesenchymal transition (EMT) and anoikis resistance .

Tissue Distribution

CDH2 is highly expressed in:

  • Neural tissues (hippocampus, cerebral cortex) .

  • Cardiac muscle .

  • Liver, kidney, and adrenal glands .

Therapeutic Insights

  • ADHD: CRISPR-edited mice with CDH2 p.H150Y showed reduced dopamine levels, rescued by methylphenidate .

  • Cancer: Targeting CDH2-mediated adhesion may inhibit metastasis .

Research Gaps and Future Directions

  • Mechanistic Links: How CDH2 variants precisely alter synaptic plasticity in ADHD remains unclear.

  • Therapeutic Strategies: Small molecules modulating CDH2-catenin interactions could treat cardiomyopathy or neurodevelopmental disorders .

Product Specs

Introduction

Cadherin-2 isoform 1, also known as CDH2, is a transmembrane glycoprotein involved in cell-to-cell adhesion. It belongs to the calcium-dependent cell adhesion molecule family. Initially found in neurons, CDH2 has also been identified in cardiac muscle and implicated in cancer metastasis. Loss of CDH2 can promote tumor development by releasing membrane-bound β-catenin, which activates Wnt signaling. While CDH2's role in tumorigenesis is not fully understood, it appears to be particularly significant in adrenocortical tumors (ACTs).

Description

Recombinant human CDH2, produced in Sf9 insect cells using a baculovirus expression system, is a single, glycosylated polypeptide chain. This protein comprises 574 amino acids (residues 160-724), including a C-terminal 6-His tag, and has a molecular weight of 62.9 kDa (appears as 70-100 kDa on SDS-PAGE). Purification is achieved through proprietary chromatographic methods.

Physical Appearance
Sterile, colorless solution.
Formulation

The CDH2 protein solution is provided at a concentration of 0.5 mg/ml in Phosphate Buffered Saline (pH 7.4) containing 20% glycerol.

Stability

For short-term storage (2-4 weeks), the product can be stored at 4°C. For longer periods, store frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.

Purity

Purity is determined to be greater than 90% by SDS-PAGE analysis.

Synonyms

Cadherin 2, Cadherin 2, Type 1, N-Cadherin (Neuronal), Neural Cadherin, N-Cadherin, CDw325, NCAD, CDHN, Calcium-Dependent Adhesion Protein, Neuronal, CD325 Antigen, N-Cadherin 1, Cadherin-2, CD325.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADPDWVIPPI NLPENSRGPF PQELVRIRSD RDKNLSLRYS VTGPGADQPP TGIFIINPIS GQLSVTKPLD REQIARFHLR AHAVDINGNQ VENPIDIVIN VIDMNDNRPE FLHQVWNGTV PEGSKPGTYV MTVTAIDADD PNALNGMLRY RIVSQAPSTP SPNMFTINNE TGDIITVAAG LDREKVQQYT LIIQATDMEG NPTYGLSNTA TAVITVTDVN DNPPEFTAMT FYGEVPENRV DIIVANLTVT DKDQPHTPAW NAVYRISGGD PTGRFAIQTD PNSNDGLVTV VKPIDFETNR MFVLTVAAEN QVPLAKGIQH PPQSTATVSV TVIDVNENPY FAPNPKIIRQ EEGLHAGTML TTFTAQDPDR YMQQNIRYTK LSDPANWLKI DPVNGQITTI AVLDRESPNV KNNIYNATFL ASDNGIPPMS GTGTLQIYLL DINDNAPQVL PQEAETCETP DPNSINITAL DYDIDPNAGP FAFDLPLSPV TIKRNWTITR LNGDFAQLNL KIKFLEAGIY EVPIIITDSG NPPKSNISIL RVKVCQCDSN GDCTDVDRIV GAGLGTGAHH HHHH.

Q&A

What is the molecular structure of CDH2 and how does it facilitate cellular adhesion?

N-cadherin (encoded by CDH2) is a transmembrane adhesion molecule featuring an N-terminal region with five extracellular cadherin domains (EC1-EC5), a single transmembrane domain, and a C-terminal cytoplasmic region (approximately 150 amino acids) . The extracellular domains mediate calcium-dependent homophilic interactions through both cis (same cell surface) and trans (adjacent cells) dimerization . The cytoplasmic domain connects to the actin cytoskeleton via sequential binding of catenin proteins . This architecture enables N-cadherin to establish stable intercellular junctions critical for tissue development and maintenance.

How does CDH2 contribute to neural development?

CDH2 expression appears during neural induction, accompanied by a parallel decrease in CDH1 (E-cadherin) mRNA levels . It plays essential roles in:

  • Maintaining neuroepithelial integrity during neural tube formation

  • Supporting radial glial progenitor cell (RGPC) function and stemness via β-catenin and Notch signaling promotion

  • Regulating the proliferation/differentiation balance of neural progenitors

  • Facilitating proper neuronal migration during cortical development

  • Establishing appropriate synaptic connections

Disruption of these processes through CDH2 mutation or deletion results in severe neurodevelopmental abnormalities including cortical lamination defects .

What animal models are most effective for investigating CDH2 function?

Several complementary model systems have proven valuable for CDH2 research:

  • Complete knockout mice: Result in early embryonic (E10) lethality due to cardiac developmental defects, limiting utility for neural studies

  • Zebrafish models: Nonsense mutations in CDH2 produce the "parachute" (pac) phenotype, allowing development to relatively mature stages before lethality (48 hours post-fertilization)

  • Conditional knockout mice: Using tissue-specific Cre drivers (e.g., Emx1-Cre) enables targeted CDH2 deletion in specific brain regions at defined developmental stages

  • CRISPR/Cas9 knock-in models: Introducing specific human disease-associated mutations enables precise modeling of pathogenic variants

Comparing results across these models can help resolve contradictory findings and elucidate developmental stage-specific functions of CDH2.

What approaches are recommended for functional characterization of CDH2 variants?

Comprehensive characterization requires multiple complementary methods:

  • In silico analysis: Tools like PMut can predict pathological relevance of novel variants

  • Cell aggregation assays: Quantify adhesive properties of different CDH2 variants

  • Protein maturation studies: Assess whether mutations affect protein processing and trafficking

  • Electrophysiological recordings: Measure effects on synaptic transmission (impaired presynaptic vesicle clustering, attenuated transmitter release)

  • Behavioral testing: Evaluate how CDH2 variants affect complex behaviors in animal models and response to pharmaceutical interventions like methylphenidate

What evidence links CDH2 mutations to ADHD?

Research has demonstrated familial ADHD caused by a missense mutation in CDH2 that affects N-cadherin protein maturation . CRISPR/Cas9-generated knock-in mice carrying the human mutation recapitulated core behavioral features of hyperactivity, which were modifiable by methylphenidate treatment . The mutation led to:

  • Impaired presynaptic vesicle clustering

  • Attenuated evoked neurotransmitter release

  • Decreased spontaneous release

  • Reduced tyrosine hydroxylase expression and dopamine levels in both ventral midbrain and prefrontal cortex

These findings delineate specific roles for CDH2-related pathways in ADHD pathophysiology.

How do rare CDH2 variants contribute to OCD and Tourette syndrome?

Exon sequencing of CDH2 identified four non-synonymous SNPs (A118T, V289I, N706S, N845S) with potential relevance to OCD and Tourette syndrome (TD) :

  • N706S, located between EC5 and the transmembrane region, was found in three individuals with OCD or TD but absent in controls

  • N845S, in the cytoplasmic domain, showed significant association with OCD/TD comorbidity (33.3% of OCD patients with N845S had comorbid TD vs. only 7.7% without this variant)

  • V289I, located in extracellular domain EC2, was found in TD probands with additional comorbidities including ADHD

While these variants aren't necessarily disease-causing by themselves, they may represent risk factors in specific genetic or environmental contexts .

What is ACOG syndrome and how is it related to CDH2?

ACOG syndrome (agenesis of corpus callosum, axon pathfinding, cardiac, ocular, and genital defects) is associated with de novo heterozygous pathogenic CDH2 variants . Patients present with:

  • Global developmental delay and intellectual disability

  • Axonal pathfinding defects

  • Cardiac malformations

  • Ocular abnormalities

  • Genital defects

Six different missense mutations affecting the extracellular domain and two frameshift mutations in the cytoplasmic region have been identified . Extracellular domain mutations (particularly Asp353Asn affecting Ca²⁺-binding) result in weaker adhesion, while cytoplasmic mutations disrupt interactions with catenins and the actin cytoskeleton .

How is CDH2 regulated post-translationally?

CDH2 undergoes regulated proteolytic processing by:

  • ADAM10 (A Disintegrin and metalloproteinase domain-containing protein 10) acting as an α-secretase for initial cleavage

  • Presenilin 1 (PSEN1) functioning as a γ-secretase to complete processing

This sequential cleavage generates fragments including the cytoplasmic CTF1. Loss of ADAM10 results in reduced CTF1 generation and phenotypes resembling CDH2 deficiency, including disrupted cortical lamination and decreased subpallium size . Radial glia-specific disruption of Adam10 results in perinatal lethality due to vascular hemorrhages in the brain, highlighting the importance of this regulatory pathway .

How does CDH2 interact with other signaling pathways?

CDH2 participates in multiple signaling networks:

  • Promotes β-catenin signaling, affecting both adhesion and transcriptional regulation

  • Interfaces with Notch pathway to maintain stemness of radial glial progenitor cells

  • Influences intermediate progenitor cell generation through regulation of PAX6, TBR2, and TBR1 expression

The timing and context of these interactions appear critical, as developmental stage-specific effects have been observed in different experimental paradigms .

How can contradictory findings about CDH2's effect on neural progenitor proliferation be reconciled?

Studies have reported opposing effects of CDH2 disruption on neural progenitor proliferation:

  • shRNA knockdown at E12.5 decreased proliferation of precursors

  • Emx1-Cre-induced loss of CDH2 increased proliferation resulting in cortical heterotopia

These contradictions might be explained by:

  • Methodological differences (acute knockdown vs. constitutive knockout)

  • Timing of CDH2 loss (early neuroepithelial cells vs. later radial glia)

  • Developmental stage-specific usage of intracellular α-catenins (αE-catenin in early ventricular zone vs. αN-catenin in later subventricular zone)

  • Different compensatory mechanisms being activated in each scenario

What challenges exist in translating CDH2 findings to clinical applications?

Major obstacles include:

  • Complete CDH2 knockout lethality necessitating creative conditional approaches

  • Relatively small cohorts of patients with rare CDH2 variants limiting statistical power

  • Complex interactions between CDH2 variants and other genetic/environmental factors

  • Neurodevelopmental timing differences between model organisms and humans

  • Challenges in directly studying CDH2 function in developing human brain

What experimental approaches might better elucidate CDH2's role in neurodevelopmental disorders?

Promising strategies include:

  • Single-cell technologies to map cell type-specific CDH2 expression and function

  • Human induced pluripotent stem cell (iPSC) models carrying patient-specific CDH2 mutations

  • Brain organoids to study CDH2's role in 3D human neural development

  • Large-scale genetic studies to identify additional rare CDH2 variants and modifier genes

  • Longitudinal studies of patients with CDH2 variants to characterize developmental trajectories

What potential therapeutic approaches might target CDH2-related pathways?

Several strategies warrant investigation:

  • Small molecules modulating N-cadherin adhesive functions

  • Targeted approaches to regulate ADAM10-mediated cleavage

  • Gene editing technologies to correct specific CDH2 mutations

  • Modulators of downstream signaling pathways (β-catenin, Notch) to compensate for CDH2 dysfunction

  • Early intervention strategies based on genetic screening and developmental monitoring

What are the recommended approaches for analyzing CDH2 expression in human brain samples?

Optimal techniques include:

  • RNAscope in situ hybridization for sensitive detection of CDH2 mRNA with cellular resolution

  • Immunohistochemistry with antibodies targeting specific domains of N-cadherin

  • Western blotting to detect both full-length protein and cleavage products

  • Co-immunoprecipitation to assess interactions with binding partners

  • Cross-validation with multiple antibodies to ensure specificity

How should researchers design experiments to investigate developmental time-specific functions of CDH2?

Effective experimental design should incorporate:

  • Inducible genetic systems (e.g., tamoxifen-inducible CreERT2) for temporal control of CDH2 manipulation

  • Developmental time course analyses across multiple stages

  • Cell type-specific promoters to target distinct neural progenitor populations

  • Careful consideration of compensatory mechanisms that may activate following CDH2 disruption

  • Cross-species validation to identify evolutionarily conserved versus species-specific functions

Product Science Overview

Structure and Function

Cadherin 2 is a single-pass transmembrane protein that mediates homophilic cell-cell adhesion, meaning it binds to the same type of cadherin on adjacent cells. This adhesion is calcium-dependent, requiring calcium ions to maintain its structural integrity and adhesive function . The protein is composed of an extracellular domain, a transmembrane domain, and a cytoplasmic domain. The extracellular domain contains five cadherin repeats, which are responsible for binding calcium ions and mediating cell-cell adhesion .

Role in Development and Physiology

Cadherin 2 is expressed in various tissues, including neural tissue, cardiac muscle, and endothelial cells. It plays a pivotal role in several physiological processes:

  1. Neural Development: Initially named Neural cadherin, Cadherin 2 is essential for the development and maintenance of the nervous system. It facilitates the formation of synaptic connections and is involved in synaptic plasticity, which is crucial for learning and memory .

  2. Cardiac Muscle: In cardiac muscle, Cadherin 2 is a key component of adherens junctions at intercalated discs. These junctions mechanically and electrically couple adjacent cardiomyocytes, ensuring coordinated contraction of the heart muscle .

  3. Cancer Metastasis: Cadherin 2 is also implicated in cancer metastasis. Changes in its expression or function can lead to increased cell motility and invasiveness, contributing to the spread of cancer cells .

Clinical Significance

Cadherin 2 has significant clinical implications. Its role in cancer metastasis makes it a potential target for cancer therapy. Additionally, mutations in the CDH2 gene, which encodes Cadherin 2, have been linked to various diseases, including arrhythmogenic right ventricular dysplasia (ARVD) and other cardiac disorders .

Recombinant Cadherin 2

Recombinant Cadherin 2 (Human) is a form of the protein produced through recombinant DNA technology. This allows for the production of large quantities of the protein for research and therapeutic purposes. Recombinant Cadherin 2 is used in various biomedical research applications, including studies on cell adhesion, cancer metastasis, and cardiac function .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.