CDH11 belongs to the type II classical cadherin family. Key structural features include:
CDH11 facilitates cell-cell adhesion through homophilic interactions and regulates intracellular signaling via β-catenin/Wnt and RhoA pathways .
CDH11 exhibits dual roles depending on cancer type:
Rheumatoid Arthritis (RA): Mediates fibroblast-like synoviocyte (FLS) invasion into cartilage via IL-17 upregulation .
Calcific Aortic Valve Disease (CAVD): Drives valve calcification through RhoA/Sox9 signaling .
Skin and Metabolic Diseases: Linked to tissue remodeling and fibrosis .
Wnt/β-catenin Inhibition: CDH11 sequesters β-catenin, reducing TCF/LEF-mediated proliferation and downregulating CCND1, c-MYC, and MMP7 .
AKT/RhoA Suppression: Inhibits tumor cell migration and epithelial-mesenchymal transition (EMT) .
Pro-Inflammatory Effects in RA: CDH11-Fc fusion protein activates PDGFR-α, amplifying PI3K/Akt and MAPK pathways in FLS .
Promoter methylation silences CDH11 in 60–80% of nasopharyngeal, esophageal, and breast cancers .
Demethylating agents (e.g., 5-aza-2'-deoxycytidine) restore CDH11 expression, reversing tumorigenicity .
Contradictory Roles: CDH11’s pro-metastatic function in breast cancer contrasts with its tumor-suppressive effects in colorectal and oral cancers .
Mechanistic Gaps: The molecular basis of CDH11’s tissue-specific behavior remains unclear.
Clinical Translation: No CDH11-targeted therapies have reached Phase III trials, necessitating further preclinical validation .
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSMGWV WNQFFVIEEY TGPDPVLVGR LHSDIDSGDG NIKYILSGEG AGTIFVIDDK SGNIHATKTL DREERAQYTL MAQAVDRDTN RPLEPPSEFI VKVQDINDNP PEFLHETYHA NVPERSNVGT SVIQVTASDA DDPTYGNSAK LVYSILEGQP YFSVEAQTGI IRTALPNMDR EAKEEYHVVI QAKDMGGHMG GLSGTTKVMI TLTDVNDNPP KFPQSVYQMS VSEAAVPGEE VGRVKAKDPD IGENGLVTYN IVDGDGMESF EITTDYETQE GVIKLKKPVD FETKRAYSLK VEAANVHIDP KFISNGPFKD TVTVKIAVED ADEPPMFLAP SYIHEVQENA AAGTVVGRVH AKDPDAANSP IRYSIDRHTD LDRFFTINPE DGFIKTTKPL DREETAWLNI TVFAAEIHNR HQEAKVPVAI RVLDVNDNAP KFAAPYEGFI CESDQTKPLS NQPIVTISAD DKDDTANGPR FIFSLPPEII HNPNFTVRDN RDNTAGVYAR RGGFSRQKQD LYLLPIVISD GGIPPMSSTN TLTIKVCGCD VNGALLSCNA EAYILNAGLS T.
CDH11, also known as cadherin-11 or OB-cadherin, is a type-II classical cadherin that mediates calcium-dependent cell-cell adhesion. Unlike type-I classical cadherins (such as E-cadherin/CDH1) that primarily maintain cohesive tissue integrity, CDH11 plays a more intricate role in tissue orchestration and is involved in functions such as migration and differentiation .
Methodologically, CDH11 can be distinguished from other cadherins by:
Its unique amino acid sequence in the adhesive interface
Formation of weaker adhesive bonds with a higher turnover rate compared to type-I cadherins
Distinct expression patterns in mesenchymal tissues and developing neural structures
Specific antibody recognition patterns using validated antibodies targeting the C-terminus
For reliable detection of human CDH11 in research settings:
Use validated antibodies such as mouse IgG1 clone 5B2H5 (Thermo Fisher #32-1700) at 2 μg/ml concentration
Include appropriate loading controls and positive control cell lines
Compare against CDH11 knockdown/knockout controls when possible
Multiple antibodies have been validated including goat anti-CDH11 (Santa Cruz sc-6461) and mouse anti-CDH11
Optimal working dilutions typically range from 3-4 μg/ml
Co-staining with membrane or junction markers helps confirm localization
Typically requires 7 μg of antibody per reaction
Verify specificity through reverse co-immunoprecipitation approaches
CDH11 exerts lineage-specific effects on mesenchymal stem cell (MSC) differentiation:
CDH11 is indispensable for adipogenic differentiation of human MSCs
Knockdown of CDH11 significantly inhibits adipogenic differentiation in both monolayer and aggregate cultures
This effect appears consistent across different culture dimensionalities
The role of CDH11 appears to be context-dependent
In aggregate cultures, CDH11 may influence mineralized matrix deposition
CDH11 knockdown has been shown to decrease expression of type VI collagen, potentially affecting osteogenic capacity
Methodologically, researchers should assess differentiation through multiple complementary approaches:
Histological staining (Oil Red O for adipogenic, Alizarin Red S for osteogenic)
Gene expression analysis of lineage-specific markers
Protein analysis of differentiation-associated factors
CDH11 regulates extracellular matrix (ECM) composition through several mechanisms:
CDH11 modulates the TGFβ1 pathway through SMAD2/3 signaling
This regulation occurs in a temporal manner, affecting early and late ECM production differently
The pathway represents a key mechanism by which CDH11 controls ECM synthesis despite lacking intrinsic signaling activity
Cadherin-11 regulates both collagen and fibronectin expression
These effects can be observed using Western blotting and immunofluorescence analysis
Analysis should be performed at both early and late time points to capture temporal dynamics
CDH11 influences receptor tyrosine kinase (RTK) signaling, particularly through PDGFRβ
These changes in RTK profiles lead to downstream alterations in MAPK pathway activation
The RTK-MAPK axis represents another mechanism by which CDH11 influences cell behavior and potentially matrix production
For experimental approaches, researchers should:
Combine genetic manipulation (CDH11 knockdown/knockout) with pathway inhibition studies
Assess both intracellular signaling (phospho-SMAD2/3, MAPK components) and extracellular matrix outcomes
Consider temporal dynamics by analyzing multiple time points
shRNA-mediated stable knockdown has been effectively used to create sh-CDH11 cell lines
Typical knockdown efficiency should be validated at both mRNA and protein levels
Multiple target sequences should be tested to identify optimal knockdown
Cdh11 knockout mice (Cdh11^tm1Mta^/HensJ available from Jackson Laboratory) provide a complete loss-of-function model
CRISPR-Cas9 gene editing can be employed for human cell lines
Inducible knockout systems allow for temporal control of CDH11 expression
Re-expression of CDH11 in knockout/knockdown models should be used to confirm specificity of observed phenotypes
Domain-specific mutants can identify functional regions responsible for specific activities
When studying CDH11 in different culture dimensions:
Compare 2D monolayers vs. 3D aggregates using the same cell numbers
Include CDH11 knockdown/knockout in both 2D and 3D systems
Analyze cadherin expression patterns in both culture systems, as they may differ significantly
Test scaffold-free aggregates vs. cells embedded in biomaterials (e.g., alginate hydrogels)
Compare systems with and without cell-ECM interaction motifs (e.g., RGD peptides)
Vary aggregate size while maintaining consistent cell numbers
Assess both morphological outcomes and molecular signaling changes
Quantify differentiation markers across culture systems
Measure CDH11 and partner protein levels in different dimensional contexts
Research has demonstrated that "cell culture dimensionality influences cell fate through cadherin-2 and cadherin-11," indicating that the dimensional environment itself affects cadherin function and subsequent cellular behaviors .
CDH11 plays significant roles in neural development with implications for autism spectrum disorder (ASD):
Cdh11^-/-^ knockout mice exhibit increased dendritic complexity in hippocampal neurons
This altered morphology correlates with changes in neuronal and synaptic activity
The effects suggest CDH11 normally acts as a constraint on excessive dendritic branching
Loss of CDH11 leads to increased levels of excitatory synaptic markers, including:
Neuroligin-1 (an excitatory synaptic marker)
Postsynaptic density protein-95 (PSD-95)
These changes suggest CDH11 influences excitatory synapse formation or stability
CDH11 knockout leads to compensatory upregulation of cadherin-8 (CDH8)
This relationship mirrors expression patterns observed in autism, where CDH8 is upregulated and CDH11 is downregulated
The reciprocal regulation suggests interconnected roles in neurodevelopment
Synaptic Marker | Expression in CDH11 KO | Potential Functional Impact |
---|---|---|
Neuroligin-1 | Increased | Enhanced excitatory synapse formation |
PSD-95 | Increased | Expanded postsynaptic densities |
Cadherin-8 | Significantly elevated | Potential compensatory mechanism |
These findings suggest that alterations in CDH11 expression may contribute to the neural circuit abnormalities observed in autism spectrum disorder .
Induced pluripotent stem cell (iPSC)-derived cortical neural precursor cells (NPCs) from individuals with autism show altered CDH11 expression
These cells exhibit downregulated CDH11 and upregulated CDH8, mirroring patterns in mouse models
iPSC-derived models allow for patient-specific investigation of cadherin dysregulation
3D cortical organoids generated from individuals with autism also display CDH11/CDH8 expression changes
These complex models recapitulate aspects of human cortical development
Organoids allow assessment of cadherin function in a physiologically relevant 3D context
Compare findings between rodent models and human cellular systems
Validate mechanisms identified in animal models using human-derived cells
Correlate expression patterns with specific clinical phenotypes in ASD patients
When utilizing these models, researchers should:
Establish isogenic control lines when possible
Compare multiple patient-derived lines to account for genetic heterogeneity
Assess both cellular phenotypes and molecular signatures
CDH11 knockout/knockdown cells or tissues serve as negative controls
Overexpression systems provide positive controls
Competing peptides can confirm specificity of antibody binding
Multiple antibodies targeting different epitopes should yield consistent results
Recommended Antibodies and Dilutions:
Based on published research, validated antibodies include:
Mouse monoclonal (IgG1) antibody clone 5B2H5 targeting C-terminus (Thermo Fisher #32-1700)
Western blot: 2 μg/ml
Immunoprecipitation: 7 μg per reaction
Goat polyclonal antibody targeting C-terminus (Santa Cruz sc-6461)
Western blot: 1:500 dilution
Mouse antibody targeting N-terminus (DSHB CAD8-1)
Maintain consistent culture conditions (passage number, confluence, serum batch)
Establish baseline expression profiles before experimental manipulation
Include multiple reference genes for qPCR normalization
Culture dimensionality significantly affects cadherin expression and function
Cell-cell contact density influences cadherin clustering and activity
Media composition, particularly calcium concentration, affects cadherin function
Consider temporal dynamics by analyzing multiple time points
Use absolute quantification methods where possible
Normalize protein expression to multiple loading controls
Employ digital PCR for more precise mRNA quantification
Combine mRNA and protein level measurements to confirm changes
By implementing these comprehensive controls and standardization approaches, researchers can obtain more reliable and reproducible results in CDH11 studies across various experimental systems.
Cadherin-11 is a type II classical cadherin, characterized by a large N-terminal extracellular domain, a single membrane-spanning domain, and a small, highly conserved C-terminal cytoplasmic domain . The protein mediates calcium-dependent cell-cell adhesion, which is essential for maintaining the structural integrity of tissues .
Cadherin-11 is expressed in embryonic mesodermal tissues and contributes to the morphogenesis of the nervous and skeletal systems . In adults, it is predominantly expressed on osteoblasts, where it promotes the differentiation of both osteoblasts and chondrocytes . This makes it a critical player in bone development and maintenance.
Cadherin-11 is up-regulated in certain types of cancer, including breast and prostate cancers, which preferentially metastasize to bone . Its expression is also increased in fibrotic diseases such as systemic sclerosis and chronic inflammatory arthritis . These associations make Cadherin-11 a potential target for therapeutic interventions in these conditions.
Recombinant Human Cadherin-11 is produced using various expression systems, including NS0 mouse myeloma cells and HEK293 cells . The recombinant protein is typically purified to high levels of purity and is used in various research applications, including studies on cell adhesion, cancer metastasis, and bone development.
The recombinant form of Cadherin-11 is often tagged with polyhistidine or other tags to facilitate purification and detection . It is available in lyophilized form and can be reconstituted in sterile buffers for use in laboratory experiments .
Recombinant Human Cadherin-11 is used in a variety of research applications, including: