NECTIN3 produced in Sf9 Insect cells is a single, glycosylated polypeptide chain containing 355 amino acids (58-404 a.a.) and having a molecular mass of 39.1kDa (Molecular size on SDS-PAGE will appear at approximately 40-57kDa).
NECTIN3 is expressed with an 8 amino acid His tag at C-Terminus and purified by proprietary chromatographic techniques.
NECTIN3, also referred to as Nectin Cell Adhesion Molecule 3, is a member of the nectin family. This protein plays a crucial role in initiating cell-to-cell adhesion, which in turn promotes cell attachment and facilitates the subsequent formation of intercellular junctions based on JAM and cadherin. NECTIN3 triggers the down-regulation of PVR from the cell surface through endocytosis, leading to a decrease in cell movement and proliferation. The activity of Nectin-3 further strengthens the junction through trans-interaction with various molecules.
Produced in Sf9 Insect cells, NECTIN3 is a single, glycosylated polypeptide chain consisting of 355 amino acids (58-404 a.a.) and possessing a molecular mass of 39.1kDa. It's important to note that on SDS-PAGE, the molecular size will appear approximately between 40-57kDa. The protein is expressed with an 8 amino acid His tag at the C-Terminus and purified using proprietary chromatographic techniques.
A colorless solution that has undergone sterile filtration.
The NECTIN3 protein solution is provided at a concentration of 0.25mg/ml and contains Phosphate Buffered Saline (pH 7.4), 20% glycerol, and 1mM DTT.
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For extended storage, freezing at -20°C is recommended. To further enhance long-term stability, consider adding a carrier protein (0.1% HSA or BSA). Repeated freezing and thawing should be avoided.
The purity of the protein is determined to be greater than 90.0% based on SDS-PAGE analysis.
Nectin-3, CDw113, Nectin cell adhesion molecule 3, Poliovirus receptor-related protein 3, CD113, PVRL3, PRR3, NECTIN-3, PVRR3, PPR3.
Sf9, Insect cells.
GPIIVEPHVT AVWGKNVSLK CLIEVNETIT QISWEKIHGK SSQTVAVHHP QYGFSVQGEY QGRVLFKNYS LNDATITLHN IGFSDSGKYI CKAVTFPLGN AQSSTTVTVL VEPTVSLIKG PDSLIDGGNE TVAAICIAAT GKPVAHIDWE GDLGEMESTT TSFPNETATI ISQYKLFPTR FARGRRITCV VKHPALEKDI RYSFILDIQY APEVSVTGYD GNWFVGRKGV NLKCNADANP PPFKSVWSRL DGQWPDGLLA SDNTLHFVHP LTFNYSGVYI CKVTNSLGQR SDQKVIYISD PPTTTTLQPT IQWHPSTADI EDLATEPKKL PFPLSTLATI KDDTIATLEH HHHHH.
NECTIN3, also known as Nectin Cell Adhesion Molecule 3 (previously designated as PVRL3), is a member of the nectin family of proteins that function as adhesion molecules at adherens junctions. It plays a critical role in cell-cell adhesion through heterophilic trans-interactions with nectin-like proteins or other nectins, such as NECTIN2 at Sertoli-spermatid junctions. These interactions induce activation of CDC42 and RAC small G proteins through common signaling molecules including SRC and RAP1 .
At the cellular level, NECTIN3 is involved in several key processes:
Formation of cell-cell junctions, including adherens junctions and synapses
Regulation of directional cell motility
Modulation of cell proliferation and survival
Endocytosis-mediated down-regulation of PVR from the cell surface
When designing experiments to study NECTIN3's basic functions, researchers should consider its interactions with other adhesion molecules and downstream signaling pathways that may influence experimental outcomes.
NECTIN3 expression shows distinct developmental regulation patterns and responds to environmental stressors. Studies have demonstrated that:
Expression levels of NECTIN3 are developmentally regulated in the dentate gyrus (DG) of the hippocampus
Early postnatal stress exposure significantly reduces NECTIN3 expression in adult mice
These reductions correlate with cognitive deficits observed in stress models
For researchers investigating developmental regulation, it's important to note that NECTIN3 plays a critical role in ocular development involving the ciliary body, and mutations in this gene may result in congenital ocular defects . When designing developmental studies, time-course experiments should account for these stage-specific expression patterns.
Methodologically, quantitative PCR (Q-PCR) and immunohistochemistry are effective techniques for measuring changes in NECTIN3 expression across developmental stages or following stress exposure. Researchers should include appropriate time points after stressor application (immediate, short-term, and long-term) to fully characterize the temporal dynamics of NECTIN3 regulation .
Several experimental models have been validated for investigating NECTIN3 function:
Cellular models:
Human breast cancer cell lines (e.g., MDA-MB-231) for studying invasion and migration
Human endothelial cell lines (e.g., HECV) for barrier function studies
Primary neuronal cultures for examining synaptic development
Animal models:
When selecting an appropriate model, researchers should consider that tissue-specific expression patterns of NECTIN3 may influence experimental outcomes. For knockdown studies, both AAV (adeno-associated virus) and RV (retrovirus) vectors have been successfully used to suppress NECTIN3 expression in dentate gyrus neurons, with differential targeting capabilities: AAV can affect all neuron populations, while RV can specifically target newly generated neurons .
NECTIN3 plays a significant role in neuronal structural plasticity with direct implications for cognitive function. Research has demonstrated that:
NECTIN3 knockdown in the dentate gyrus results in:
Increased density of doublecortin-immunoreactive differentiating cells under proliferation
Increased calretinin-immunoreactive immature neurons
Decreased calbindin immunoreactivity
Reduction of dendritic spines, especially thin spines, in newly generated DG neurons
These structural changes correlate with specific cognitive impairments:
Impaired long-term spatial memory
Deficits in temporal order memory
For researchers investigating NECTIN3's role in cognition, it's important to distinguish between different memory types in experimental design. While NECTIN3 knockdown in all DG neurons impairs both spatial and temporal order memory, selective knockdown in newly generated neurons only affects spatial memory, suggesting cell population-specific functions .
Methodologically, combining behavioral testing (Y-maze, spatial object recognition, temporal order memory tests) with histological analysis of neuronal morphology provides the most comprehensive assessment of NECTIN3's impact on structural plasticity and cognition.
NECTIN3 has emerged as a significant factor in cancer progression, with particularly important roles in breast cancer:
Q-PCR analysis of human breast tissue has revealed distinct reductions in NECTIN3 expression in node-positive tumors and in patients with poor outcomes
While NECTIN1 and NECTIN2 show increased expression in patients with metastatic disease, NECTIN3 and NECTIN4 expression is reduced
Immunohistochemistry demonstrates clear changes in NECTIN3 distribution patterns between normal and cancerous cells
Functional studies have demonstrated that:
NECTIN3 overexpression in MDA-MB-231 breast cancer cells reduces invasive and migratory capabilities, even when stimulated with Hepatocyte Growth Factor (HGF)
Cells overexpressing NECTIN3 show reduced change in resistance after HGF treatment compared to control cells
NECTIN3-transformed endothelial cells exhibit significantly increased adhesion (irrespective of HGF treatment) and reduced growth
These findings suggest NECTIN3 may function as a metastasis suppressor through regulation of tight junctions and cell adhesion. When designing cancer-related NECTIN3 studies, researchers should consider examining both expression levels and cellular distribution patterns, as both appear to be clinically relevant.
Several approaches for modulating NECTIN3 expression have been validated in research settings:
Viral vector-mediated knockdown:
Overexpression systems:
Essential controls include:
Scrambled shRNA sequences for knockdown experiments
Empty vector controls for overexpression studies
Verification of knockdown/overexpression efficiency via Western blot or qPCR
Both gain- and loss-of-function approaches to confirm bidirectional effects
For in vivo studies, stereotaxic coordinates are critical. For dentate gyrus targeting in adult mice: 1.8 mm posterior to bregma, 1.2 mm lateral from midline, and 1.65 mm dorsoventral from dura. A 4-week recovery period post-injection is typically required before behavioral testing to allow sufficient viral infection .
NECTIN3 functions within a complex network of cell adhesion molecules to regulate junction integrity:
NECTIN3 interacts with other nectin-like proteins and with afadin, a filamentous actin-binding protein
It forms heterophilic trans-interactions with molecules such as NECTIN2 at specialized junctions
These interactions activate small G proteins (CDC42 and RAC) through signaling molecules like SRC and RAP1
To experimentally assess junction integrity mediated by NECTIN3:
Barrier function assays:
Transepithelial/endothelial electrical resistance (TEER) measurements before and after treatments that modulate NECTIN3 (e.g., HGF treatment)
Permeability assays using fluorescently labeled dextrans or other tracers
Adhesion assays:
Quantitative cell-cell adhesion assays to measure changes in adhesive properties
Cell detachment assays under standardized conditions
Junction protein localization:
Immunofluorescence to visualize co-localization of NECTIN3 with other junction proteins
Live-cell imaging to track dynamic changes in junction formation and stability
Molecular interaction studies:
When interpreting results, researchers should consider that NECTIN3 functions within a larger adhesion complex, and compensatory mechanisms involving other cell adhesion molecules may influence experimental outcomes.
For reliable detection of NECTIN3 across different tissues and experimental contexts:
Validated antibodies:
Human NECTIN3 Antibody (AF3064) from R&D Systems has been validated for research applications
Look for antibodies targeting the extracellular domain (Leu56-Asp400) of NECTIN3 for surface labeling
For western blotting, antibodies recognizing the cytoplasmic domain may provide better specificity
Tissue-specific considerations:
Expression patterns differ between normal tissues and cancer samples
In breast tissue, both expression levels and distribution patterns should be assessed
In neural tissue, region-specific expression (e.g., dentate gyrus vs. other hippocampal regions) is important to consider
Detection methods:
Immunohistochemistry works well for tissue distribution studies
Western blotting for quantitative protein expression
qPCR for transcript level analysis
Flow cytometry for cell surface expression quantification
When designing experiments, include appropriate positive and negative controls, and validate antibody specificity in your specific tissue or cell type. Different fixation methods may affect epitope accessibility, so optimization may be required for immunohistochemistry applications.
Designing experiments to differentiate between developmental and functional roles of NECTIN3 requires careful temporal control and specific targeting approaches:
For developmental roles:
Temporal knockdown/knockout strategies:
Developmental trajectory analysis:
Time-course expression studies across developmental stages
Correlation of expression patterns with developmental milestones
Analysis of developmental defects in NECTIN3-deficient models
For acute functional roles:
Acute manipulation in mature systems:
Functional readouts:
Electrophysiological recordings to assess synaptic function
Behavioral testing for cognitive or social deficits
Time-lapse imaging to track dynamic changes in cellular morphology
A particularly effective approach is to compare the effects of constitutive NECTIN3 deficiency with acute knockdown in adult animals. Differences in phenotypes would suggest developmental versus acute functional roles. Additionally, rescue experiments (restoring NECTIN3 expression at different timepoints) can help distinguish between these roles.
To effectively study NECTIN3's role in stress-induced cognitive impairments, researchers should consider the following optimal parameters:
Stress paradigms:
Early-life stress models show consistent effects on NECTIN3 expression
Standard protocols include maternal separation or limited nesting material for early-life stress
Adult stress protocols might include chronic unpredictable stress or social defeat stress
Control for stress intensity and duration to ensure reproducibility
Timing considerations:
NECTIN3 expression levels are developmentally regulated
Early postnatal stress has been shown to reduce NECTIN3 expression in adult mice
Include both immediate and long-term (adult) assessment timepoints
Cognitive assessment:
Include multiple memory tests to distinguish between different cognitive domains
Y-maze for spatial working memory
Spatial object recognition for long-term spatial memory
Temporal order memory tests
Control for potential anxiety effects by including anxiety assessments (open field, light-dark box, elevated plus maze)
Molecular and cellular analyses:
Quantify NECTIN3 expression via qPCR and western blotting
Assess dendritic spine density and morphology
Examine adult neurogenesis markers (doublecortin, calretinin, calbindin)
Consider region-specific effects (dorsal versus ventral hippocampus)
For maximum translational value, combine behavioral, molecular, and morphological analyses in the same animals when possible. This approach allows for direct correlation between behavioral phenotypes and underlying biological changes.
When encountering conflicting data on NECTIN3 expression across cancer types or studies, researchers should consider several factors in their interpretation:
Potential sources of discrepancy:
Tissue-specific differences:
Methodological variations:
Sample preparation differences (fresh vs. fixed tissue)
Antibody specificity and epitope accessibility
RNA vs. protein level measurements
Bulk tissue vs. single-cell analysis
Cancer heterogeneity:
Resolution strategies:
Perform comprehensive profiling across multiple cancer types using standardized methods
Include both protein and transcript level analyses
Assess both expression levels and subcellular localization
Correlate with clinical parameters (stage, grade, outcome)
Validate findings with functional studies to determine biological significance
The apparent discrepancy that NECTIN1/2 show increased expression in metastatic disease while NECTIN3/4 are reduced suggests complementary or opposing roles within the nectin family. This pattern indicates researchers should consider analyzing multiple nectin family members simultaneously rather than focusing exclusively on NECTIN3 .
When extrapolating findings from NECTIN3 knockdown studies in animal models to human neuropsychiatric conditions, researchers should carefully consider:
Translational limitations:
Species differences:
Neural development timelines differ between rodents and humans
Complexity of human brain circuits and behaviors
Potential differences in compensation mechanisms
Manipulation specificity:
Phenotypic complexity:
Strengthening translational relevance:
Combine gene manipulation with environmental factors (stress, enrichment)
Validate findings across multiple behavioral domains
Correlate with human genetic or expression data when available
Consider developmental timing in both model systems and human conditions
Examine sex differences, which are prominent in many neuropsychiatric disorders
As noted in the research, "nectin-3 knockdown-induced effects may be only responsible for the reduction of a small fraction of spines," suggesting that NECTIN3 is likely part of a larger constellation of molecular changes in complex disorders. Additionally, "nectin-3 knockdown resulted in molecular and morphological phenotypes that were not prominent in neonatally stressed adult mice," highlighting the importance of considering the broader context when extrapolating findings .
Distinguishing direct effects of NECTIN3 manipulation from compensatory mechanisms requires careful experimental design and interpretation:
Experimental approaches to identify direct effects:
Acute vs. chronic manipulation:
Use inducible systems for temporal control
Compare short-term vs. long-term effects after manipulation
Rapid effects are more likely direct consequences
Molecular pathway analysis:
Targeted rescue experiments:
Restore NECTIN3 function selectively
Manipulate specific downstream pathways
Use structure-function mutants to identify critical domains
Identifying compensatory mechanisms:
Expression profiling:
Combined manipulations:
Simultaneously target NECTIN3 and potential compensatory molecules
If phenotypes are exacerbated, compensation is likely occurring
Developmental considerations:
Early manipulation may trigger more robust compensation
Adult manipulation may reveal more direct effects
As noted in the research literature, "compensatory effects" from other cell adhesion molecules may explain why some phenotypes observed in NECTIN3 knockdown models are not seen in stress models. For example, "nectin-3 knockdown-induced effects may be only responsible for the reduction of a small fraction of spines" since "adherens junctions are present in 33% of dendritic spines in CA1 pyramidal neurons" .
Several cutting-edge technologies hold promise for advancing our understanding of NECTIN3's role in neural circuit development and function:
Single-cell technologies:
Single-cell RNA sequencing to identify cell type-specific NECTIN3 expression patterns
Single-cell ATAC-seq to examine regulatory mechanisms controlling NECTIN3 expression
Patch-seq for correlating NECTIN3 expression with electrophysiological properties
Advanced imaging techniques:
Super-resolution microscopy to visualize NECTIN3 at the synapse with nanometer precision
Expansion microscopy to examine NECTIN3 distribution in preserved neural circuits
Live-cell imaging with genetically encoded NECTIN3 fusion proteins to track dynamics
Circuit manipulation tools:
Optogenetic control of NECTIN3-expressing neurons
Chemogenetic approaches to modulate activity in NECTIN3 circuits
CRISPR-based approaches for precise genomic manipulation of NECTIN3
Computational approaches:
Machine learning to identify patterns in NECTIN3 expression across brain regions
Predictive modeling of NECTIN3 interactions with other adhesion molecules
Systems biology approaches to integrate NECTIN3 into larger signaling networks
These technologies could address current knowledge gaps, such as how NECTIN3 regulates specific synaptic connections, its role in activity-dependent plasticity, and how it interacts with other cell adhesion molecules in vivo. For example, combining selective NECTIN3 manipulation with in vivo calcium imaging could reveal how NECTIN3 influences neural circuit function during learning and memory processes.
NECTIN3 pathway modulation presents promising therapeutic avenues for both stress-related cognitive disorders and cancer:
For stress-related cognitive disorders:
Potential approaches:
Small molecule enhancers of NECTIN3 signaling to reverse stress-induced deficits
Gene therapy to restore NECTIN3 expression in specific brain regions
Targeting downstream effectors of NECTIN3 signaling (CDC42/RAC pathways)
Peptide mimetics of NECTIN3 binding domains
Therapeutic rationale:
Challenges to address:
Region-specific delivery to avoid off-target effects
Developmental timing considerations
Integration with existing therapeutic approaches
For cancer treatment:
Potential approaches:
NECTIN3 overexpression or stabilization to reduce metastatic potential
Targeting the NECTIN3 interaction with HGF signaling pathways
Developing biomarkers based on NECTIN3 expression patterns
Therapeutic rationale:
Challenges to address:
Tissue-specific effects of NECTIN3 modulation
Potential compensatory mechanisms
Integration with existing cancer therapies
For both applications, therapeutic development should consider that NECTIN3 functions within a complex network of adhesion molecules, and isolated manipulation may trigger compensatory responses. Combination approaches targeting multiple components of adhesion and signaling pathways may offer the most promising therapeutic strategy.
Understanding how diverse environmental factors beyond stress influence NECTIN3 expression and function represents an important frontier in research:
Potential environmental modulators:
Enriched environment:
Environmental enrichment might upregulate NECTIN3 to enhance synaptic plasticity
Physical exercise could influence NECTIN3 in hippocampal neurogenesis
Social enrichment may affect NECTIN3 in regions mediating social behavior
Nutrition and metabolism:
Dietary factors might influence NECTIN3 expression during development
Metabolic disorders could alter NECTIN3 function in multiple tissues
Nutritional interventions might rescue stress-induced NECTIN3 deficits
Inflammation and immune activation:
Inflammatory cytokines may regulate NECTIN3 at the blood-brain barrier
Maternal immune activation could affect developmental NECTIN3 expression
Chronic inflammation in cancer might modulate NECTIN3's role in tumor progression
Aging-related factors:
Age-related changes in NECTIN3 might contribute to cognitive decline
Interactions with age-related pathologies (e.g., neurodegeneration)
Potential protective factors against age-related NECTIN3 dysfunction
Methodological approaches:
Longitudinal studies examining NECTIN3 expression across the lifespan
Multi-factorial experimental designs combining different environmental exposures
Epigenetic profiling to identify environmentally sensitive regulatory regions
Comparative studies across different genetic backgrounds to identify gene-environment interactions
This research direction could provide insights into resilience factors that protect against stress-induced NECTIN3 downregulation and identify novel environmental interventions to promote NECTIN3 function in both neurodevelopmental contexts and cancer progression.
Nectin Cell Adhesion Molecule 3, also known as Nectin-3 or NECTIN3, is a protein encoded by the NECTIN3 gene in humans. It belongs to the family of immunoglobulin-like cellular adhesion molecules that are involved in calcium-independent cellular adhesion in various tissues during development . Nectin-3 plays a crucial role in the formation and maintenance of cell-cell junctions, particularly in the nervous system and epithelial tissues.
Nectin-3 has three splicing variants: nectin-3α, nectin-3β, and nectin-3γ. The largest variant, nectin-3α, is abundantly expressed in the testis and is also found in the heart, brain, liver, and kidney . Nectin-3 is characterized by its immunoglobulin-like domains, which facilitate its role in cell adhesion.
Nectin-3 functions as a homophilic cell adhesion molecule, meaning it can bind to itself to mediate cell-cell adhesion. It is also involved in heterophilic interactions with other nectins, such as nectin-1 and nectin-2. These interactions are crucial for the formation of adherens junctions, which are essential for maintaining the structural integrity of tissues .
In the nervous system, nectin-3 is involved in synapse formation and maintenance. It has been shown to play a role in the development of cortical circuits by regulating dendritic spine densities, which are indicative of synaptic connections . Proper levels of nectin-3 are required for normal synapse formation, and both overexpression and knockdown of nectin-3 can lead to abnormalities in synaptic density .
Mutations or dysregulation of nectin-3 can lead to various developmental disorders and diseases. For example, alterations in nectin-3 expression have been linked to intellectual disabilities and other neurodevelopmental disorders. Additionally, nectin-3 is involved in the entry of certain viruses, such as herpes simplex virus, into host cells .
Human recombinant nectin-3 is produced using recombinant DNA technology. This involves inserting the NECTIN3 gene into an expression vector, which is then introduced into a host cell line, such as E. coli or CHO cells. The host cells express the nectin-3 protein, which is subsequently purified using various chromatographic techniques. Recombinant nectin-3 is used in research to study its role in cell adhesion and synapse formation, as well as in the development of therapeutic interventions for diseases associated with nectin-3 dysfunction.