NECTIN1 (also known as Poliovirus receptor-related protein-1 or PVRL1) belongs to the Nectin family, which consists of calcium-independent immunoglobulin (Ig)-like cell adhesion molecules that organize intercellular junctions . This family comprises four primary members (NECTIN1-4) and five nectin-like molecules, all sharing structural homology with poliovirus receptors . Human NECTIN1 is widely expressed across different tissues and performs essential functions in establishing and maintaining cell-cell contacts .
NECTIN1 has garnered particular research interest due to its multifaceted roles in developmental processes, viral entry mechanisms, and pathological conditions. The protein's ability to mediate both homophilic (self-binding) and heterophilic (binding to other nectins) interactions enables it to fulfill diverse biological functions .
Characteristic | Description |
---|---|
Alternative names | PVRL1, CD111, PRR1, HVEC, Poliovirus receptor-related protein-1 |
Protein family | Nectin family of cell adhesion molecules |
Interaction type | Calcium-independent |
Structural classification | Immunoglobulin superfamily (IgSF) |
Amino acid homology | 93% with mouse NECTIN1, 94% with rat NECTIN1 |
Gene location | Chromosome 11 (human) |
Known splice variants | Two variants (alternate cytoplasmic domain and soluble form) |
NECTIN1 engages in an intricate network of molecular interactions that underpin its biological functions. These interactions can be broadly categorized into homophilic binding, heterophilic binding with other nectins, interactions with viral proteins, and associations with intracellular signaling molecules.
NECTIN1 forms homodimers in cis (on the same cell surface), followed by interactions in trans (between adjacent cells) with NECTIN1, NECTIN3, or NECTIN4 . These interactions are fundamental to NECTIN1's role in cell adhesion. The binding between NECTIN1 and NECTIN3 is particularly important in chemical synapses, where they localize asymmetrically with NECTIN1 primarily on the axonal side and NECTIN3 on the dendritic side .
NECTIN1 serves as a primary receptor for several alphaherpesviruses by binding to viral glycoprotein D (gD) . This interaction mediates viral entry into vaginal mucosa, sensory neurons, and fibroblasts . The binding site for herpesvirus gD is located on the NECTIN1 V-domain, which comprises the canonical interface shared by nectins for cell adhesion .
NECTIN1 interacts with CD96 on natural killer (NK) cells, though with lower affinity than for other nectins . The binding site for CD96 is also located on the NECTIN1 V-domain. The affinity of NECTIN1 for CD96 is comparable to its affinity for herpesvirus glycoprotein D but lower than its affinity for NECTIN3 and NECTIN1 itself .
Recent research has revealed that the third Ig module of NECTIN1 (NECTIN1 Ig3) directly interacts with various isoforms of fibroblast growth factor receptor (FGFR) . This interaction induces phosphorylation of FGFR1c and promotes neuronal differentiation and survival .
The cytoplasmic domain of NECTIN1 binds to afadin, a scaffolding protein that links NECTIN1 to the actin cytoskeleton . This interaction is crucial for the formation and stability of adherens junctions.
Binding Partner | Interaction Type | Affinity | Functional Outcome |
---|---|---|---|
NECTIN1 | Homophilic (cis and trans) | High | Cell-cell adhesion, junction formation |
NECTIN3 | Heterophilic (trans) | High | Synapse formation, cell-cell adhesion |
NECTIN4 | Heterophilic (trans) | Moderate | Cell-cell adhesion |
Herpesvirus gD | Virus-host | Micromolar | Viral entry into host cells |
CD96 (NK cells) | Immune recognition | Micromolar | Potential immune cell activation |
FGFR | Signal transduction | Detected by SPR | Neuronal differentiation and survival |
Afadin | Intracellular scaffold | High | Linkage to actin cytoskeleton |
NECTIN1 plays a fundamental role in establishing and maintaining cell-cell contacts by forming homophilic or heterophilic trans-dimers . These interactions initiate cell-cell adhesion, recruiting alpha v beta 3 integrin extracellularly and cadherins intracellularly through afadin and other junctional proteins . By organizing the cytoskeleton, NECTIN1 strengthens attachment to basement membrane and promotes further cell-cell connections .
In epithelial tissues, NECTIN1 localizes to adherens junctions, while in neuronal tissues, it contributes to the formation of chemical synapses . This versatility in junction formation is essential for tissue integrity and proper cell communication.
NECTIN1 serves as a crucial entry receptor for multiple alphaherpesviruses, including herpes simplex virus (HSV) . It binds viral glycoprotein D to mediate herpesvirus entry into vaginal mucosa, sensory neurons, and fibroblasts .
During HSV1 infection, glycoprotein D (gD) has been implicated in downregulation of NECTIN1 . This process requires virus protein synthesis rather than incoming virus particles and does not involve virus-induced host shutoff . Studies have demonstrated that gD is both necessary and sufficient for NECTIN1 depletion .
The downregulation of NECTIN1 by HSV1 gD contributes to superinfection exclusion—the ability of an established virus infection to block subsequent infection by the same or related viruses . This mechanism is important for viral infection dynamics but does not affect natural killer cell activation, which is regulated by other mechanisms .
Interestingly, recent research has identified NECTIN1 as a novel restriction factor for flavivirus infection, suggesting a broader role in antiviral immunity beyond its function as a herpesvirus receptor .
NECTIN1 is prominently expressed in the brain, particularly in the cerebellum and hippocampus . It is required for proper development of ectodermal structures, and mutations in the NECTIN1 gene can cause mental retardation in severe cases .
The third Ig module of NECTIN1 (NECTIN1 Ig3) induces neurite outgrowth in primary cultures of hippocampal and cerebellar granule neurons . This effect is mediated through direct interaction with and activation of fibroblast growth factor receptor (FGFR) .
NECTIN1 Ig3 also promotes survival of cerebellar granule neurons induced to undergo apoptosis . A peptide called "nectide," constructed by employing in silico modeling of various FGFR ligand-binding sites, was found to mimic these neuritogenic and survival-promoting effects of NECTIN1 Ig3 .
Deficiency of NECTIN1 can result in cleft lip/palate ectodermal dysplasia type 1 (CLPED1) . This condition is characterized by craniofacial, skin, and digit abnormalities, likely caused by defects in cell-cell adhesion during development . In genetic knockout mice, the lack of NECTIN1 results in microphthalmia and dental abnormalities .
Mutations in the NECTIN1 gene can cause mental retardation in severe cases, emphasizing the importance of this protein in central nervous system development .
NECTIN1 down-regulation in epithelial cancers, mediated in part by ectodomain shedding, may contribute to increased invasiveness . A significant study found that loss of NECTIN1 stimulates melanoma cell migration in vitro and spreading in vivo in both zebrafish and human tumors, specifically in response to decreased insulin-like growth factor 1 (IGF1) signaling .
In human melanoma biopsy specimens, adherens junctions were observed exclusively in areas with low IGF1 levels but were absent in NECTIN1-deficient tumors . This suggests that NECTIN1 acts as a major determinant of melanoma dissemination and highlights a genetic control of the response to microenvironmental signals in cancer progression.
As a primary receptor for alphaherpesviruses, NECTIN1 plays a critical role in viral infections . During herpes simplex virus 1 (HSV1) infection, glycoprotein D downregulates NECTIN1 from the cell surface . This downregulation contributes to superinfection exclusion but does not affect natural killer cell activation .
NECTIN1 has also been identified as a novel restriction factor for flavivirus infection, indicating a complex role in virus-host interactions that extends beyond its function as an entry receptor .
Condition | Mechanism | Clinical Features | Research Status |
---|---|---|---|
Cleft lip/palate ectodermal dysplasia (CLPED1) | NECTIN1 deficiency | Craniofacial abnormalities, skin and digit abnormalities | Well-characterized genetic disorder |
Neurological developmental disorders | NECTIN1 gene mutations | Mental retardation (in severe cases) | Active area of research |
Melanoma metastasis | NECTIN1 loss in low IGF1 conditions | Increased tumor cell migration and dissemination | Recent discovery with potential therapeutic implications |
Alphaherpesvirus infections | NECTIN1 acts as viral receptor | Herpes simplex virus infections | Well-established role |
Flavivirus restriction | NECTIN1 inhibits viral replication | Potential protection against flavivirus infections | Recently identified function |
Several commercial sources provide recombinant NECTIN1 proteins for research purposes . For example, recombinant human NECTIN1 with various tags (His-tag, Avi-tag, hFc-tag) has been developed and characterized . These tools are valuable for studying NECTIN1 functions and interactions in vitro.
The protein can be expressed in different systems including HEK293 cells, and its activity can be assessed through binding assays with known partners such as NECTIN3 . Recombinant NECTIN1 proteins typically show high purity (>95%) as determined by various analytical methods .
Understanding NECTIN1's role in viral entry has implications for developing antiviral treatments targeting herpesvirus infections . By blocking the interaction between viral glycoprotein D and NECTIN1, it may be possible to prevent viral entry and infection.
The neuritogenic and survival-promoting effects of NECTIN1 Ig3 mediated through FGFR activation suggest potential therapeutic applications for neurological conditions . A peptide called "nectide," which mimics the effects of NECTIN1 Ig3, represents a promising starting point for developing neuroprotective and neuritogenic agents .
The recent identification of NECTIN1 as a restriction factor for flavivirus infection opens new avenues for exploring antiviral strategies against flaviviruses .
In cancer therapy, the role of NECTIN1 in melanoma dissemination in response to IGF1 signaling suggests that targeting this pathway might help prevent metastasis in NECTIN1-deficient tumors .
Nectin-1, also known as ED4 or poliovirus receptor-related 1 protein, belongs to the Nectin family. This protein facilitates cell-to-cell connections by creating symmetrical (homophilic) or asymmetrical (heterophilic) pairings across cells. Specifically, Nectin-1 can form heterophilic interactions with PVRL4 (Nectin-4) and PVRL3 (Nectin-3). Beyond its role in cell adhesion, Nectin-1 acts as a receptor for viruses like herpes simplex virus and pseudorabies virus, allowing them to enter cells. Additionally, Nectin-1 has been shown to promote the growth of neurites, which are projections from neurons.
This product consists of the human NECTIN1 protein, recombinantly produced in Sf9 insect cells (using a baculovirus expression system). It is a single polypeptide chain, containing 334 amino acids (specifically, amino acids 31-355 of the full protein sequence), and has a molecular weight of 37.3 kDa. This NECTIN1 protein is glycosylated, meaning it has sugar molecules attached to it. A 9 amino acid Histidine tag (His-tag) has been added to the C-terminus of the protein to facilitate purification, which was carried out using specialized chromatographic techniques.
This NECTIN1 solution is provided at a concentration of 1mg/ml and is prepared in a buffer consisting of Phosphate-Buffered Saline (pH 7.4) with 10% glycerol.
For short-term storage (up to 2-4 weeks), keep the product refrigerated at 4°C. For extended storage, freeze the solution at -20°C. It is highly recommended to supplement the solution with a carrier protein (like 0.1% HSA or BSA) before freezing for long-term stability. Repeated freezing and thawing of the product should be avoided.
This product exhibits a purity greater than 95%, as determined by SDS-PAGE analysis.
PVRL1, CD111, CLPED1, ED4, HIgR, HVIS, HVEC, Nectin-1, OFC7, PRR, PRR1, PVRR, PVRR1, SK-12.
Sf9, Baculovirus cells.
ADPQVVQVND SMYGFIGTDV VLHCSFANPL PSVKITQVTW QKSTNGSKQN VAIYNPSMGV SVLAPYRERV EFLRPSFTDG TIRLSRLELE DEGVYICEFA TFPTGNRESQ LNLTVMAKPT NWIEGTQAVL RAKKGQDDKV LVATCTSANG KPPSVVSWET RLKGEAEYQE IRNPNGTVTV ISRYRLVPSR EAHQQSLACI VNYHMDRFKE SLTLNVQYEP EVTIEGFDGN WYLQRMDVKL
TCKADANPPA TEYHWTTLNG SLPKGVEAQN RTLFFKGPIN YSLAGTYICE ATNPIGTRSG QVEVNITEFP YTPSPPEHGR RAGPVPTAHH HHHH
NECTIN1 (also known as CD111, HVEC, PRR1, or PVRL1) is a type I transmembrane glycoprotein with a molecular weight of approximately 110 kDa that belongs to the nectin family within the immunoglobulin (Ig) superfamily . The name derives from the Latin word "necto" meaning "to connect," reflecting its primary role in calcium-independent cell-cell adhesion .
The primary functions of NECTIN1 include:
Promotion of cell-cell contacts through formation of homophilic or heterophilic trans-dimers
Development of the nervous system, particularly in axon guidance
Involvement in synaptogenesis
Promotion of neurite outgrowth
Formation of adherens junctions
Mediation of heterotypic adhesion between hair cells and supporting cells in the auditory epithelium
NECTIN1 forms homodimers in cis, then interacts in trans with NECTIN1, NECTIN3, or NECTIN4 . These interactions are essential for establishing proper cellular connections during development and in mature tissues.
Human NECTIN1 isoform 1 is a 517 amino acid protein with a well-defined domain architecture consisting of:
A 30 amino acid signal sequence
A 325 amino acid extracellular domain (ECD)
A 21 amino acid transmembrane segment
The extracellular domain contains three immunoglobulin-like domains:
Alternative splicing produces multiple isoforms, including:
Isoform 2 (also called HigR): 458 amino acids with alternate transmembrane and cytoplasmic sequences
Isoform 3: A shorter variant with different functional properties
The V-domain (residues 59-133) constitutes the primary interaction interface with herpes simplex virus glycoprotein D (HSV gD) and is critically important for viral entry .
NECTIN1 serves as a primary entry receptor for several alphaherpesviruses, including herpes simplex virus 1 (HSV-1), herpes simplex virus 2 (HSV-2), and pseudorabies virus (PRV) . Research indicates that NECTIN1 constitutes the major receptor for HSV-1 entry into host cells .
The mechanism of NECTIN1-mediated viral entry involves:
Direct interaction between the V-domain of NECTIN1 (residues 59-133) and the viral glycoprotein D (gD)
This interaction facilitates viral attachment to the cell surface
Subsequent conformational changes in viral envelope proteins enable membrane fusion and viral entry
Experimental evidence supporting NECTIN1's role as an entry mediator includes:
Ectopic expression of human NECTIN1 in otherwise resistant CHO-K1 cells confers susceptibility to VZV infection, with dose-dependent increases in viral glycoprotein expression
Soluble recombinant forms of NECTIN1 can block viral entry when added during infection but have no effect when added post-infection
Knockdown of NECTIN1 reduces viral infectivity in human neurons
These findings highlight NECTIN1's critical role in the initial stages of viral infection, making it an important target for antiviral research.
Targeting NECTIN1-virus interactions represents a promising strategy for developing novel antivirals, particularly against herpes simplex viruses. Several methodological approaches have demonstrated efficacy:
Competitive inhibition using soluble NECTIN1 proteins:
Peptide-based inhibitors:
Peptides derived from the NECTIN1 V-domain that constitutes the interaction interface with HSV gD (residues 59-133) show promising antiviral activity
In particular, peptide N1 (residues 76-90) demonstrated significant protection against both HSV-1 (66.57%) and HSV-2 (71.12%) infections in CPE inhibition assays
Peptidomimetic approaches:
The success of these approaches suggests that disrupting NECTIN1-mediated viral entry represents a viable therapeutic strategy for addressing herpesvirus infections, especially in light of emerging resistance to existing antiviral treatments.
Several validated methods exist for detecting and quantifying NECTIN1 expression in experimental settings:
ELISA-based detection:
Flow cytometry:
Monoclonal antibodies against human NECTIN1 (e.g., clone 610835) can detect NECTIN1 expression on cell surfaces
Demonstrated applications include detection in U937 human histiocytic lymphoma cell lines
Protocol involves staining cells with anti-NECTIN1 antibody followed by fluorophore-conjugated secondary antibody (e.g., Allophycocyanin-conjugated Anti-Mouse IgG F(ab')2)
Reverse transcription-quantitative PCR (RT-qPCR):
Western blotting:
These methods can be employed individually or in combination to comprehensively characterize NECTIN1 expression under various experimental conditions.
Optimizing NECTIN1 knockdown in neuronal models requires careful consideration of several methodological factors:
siRNA transfection strategy:
Single transfections may achieve significant mRNA reduction (71% reduction observed 3 days post-transfection) but may not efficiently reduce protein levels
Serial transfection of siRNAs every 4 days is recommended to achieve substantial protein knockdown
Include appropriate controls (e.g., GAPDH siRNA) to validate transfection efficiency
Verification of knockdown efficacy:
RT-qPCR should be performed to assess mRNA levels (ideally 3 days post-transfection)
Western blotting and/or flow cytometry should be used to confirm protein reduction
Consider the half-life of the NECTIN1 protein when designing experimental timelines
Cell model selection:
Human pluripotent stem cell-derived neurons offer advantages for studying VZV neurotropism
These models have yielded insights regarding viral gene expression, neurovirulence, intracellular viral transport, and latency/reactivation
Consider using established human ESC-derived neuronal models to ensure reproducibility
Functional validation:
Test the impact of knockdown on viral infection using appropriate viral strains
Quantify infection through viral protein expression (e.g., gE for VZV)
Include rescue experiments by expressing siRNA-resistant NECTIN1 constructs to confirm specificity
By implementing these methodological considerations, researchers can effectively reduce NECTIN1 expression in neuronal models and reliably investigate its functional significance.
NECTIN1 participates in both homophilic (NECTIN1-NECTIN1) and heterophilic (NECTIN1 with other nectins) interactions, each with distinct functional implications:
Homophilic interactions:
Heterophilic interactions:
NECTIN1 forms heterophilic trans-dimers with NECTIN3 and NECTIN4
NECTIN1-NECTIN3 interactions:
Heterophilic interactions generally display stronger binding affinities than homophilic interactions
These interactions are crucial for establishing asymmetrical junctions between different cell types
Understanding the structural basis of NECTIN heterophilic interactions has been challenging. While structural characterizations of homophilic NECTIN interactions are available, the structural basis of heterophilic interactions has remained unclear for decades. Recent research has made progress in this area, such as determining the structure of a related Necl (Nectin-like) heterodimer comprising the ectodomains of mouse Necl4 and Necl1 .
The functional differences between these interaction types highlight NECTIN1's versatile role in tissue organization and cell-type specific adhesion patterns.
Species-specific differences in NECTIN1 have significant implications for viral entry studies, particularly for viruses with strict human tropism like varicella-zoster virus (VZV):
Structural and sequence differences:
Human NECTIN1 differs from mouse NECTIN1 in key residues that affect viral binding
Antibodies against human NECTIN1 show no cross-reactivity with recombinant mouse NECTIN1 in direct ELISAs, highlighting structural differences
These differences affect the utility of animal models for studying human-specific viral infections
Post-translational modifications:
Post-translational modifications of NECTIN1 appear critical for its function as a viral entry receptor
Soluble NECTIN1 produced in different expression systems shows variable effects on viral entry:
Methodological considerations for research:
Human pluripotent stem cell-derived neurons have become increasingly utilized to study VZV neurotropism due to species barriers
When designing anti-viral peptides or inhibitors based on NECTIN1, human-specific sequences must be prioritized
Ectopic expression of human NECTIN1 in resistant cell lines (like CHO-K1) can create useful experimental systems for studying viral entry mechanisms
Human NECTIN1 expression confers susceptibility to VZV infection in otherwise resistant cell lines
These species-specific differences underscore the importance of using appropriate human cell models or humanized systems when studying NECTIN1-mediated viral entry, particularly for strictly human-tropic viruses.
Optimizing NECTIN1-derived peptides for antiviral efficacy involves a systematic approach combining in silico prediction, design optimization, and experimental validation:
Identification of binding interfaces:
In silico modeling and optimization:
Use computational tools to model peptide structure and binding:
These tools help predict peptide-viral protein interactions and enable iterative design improvements
Peptidomimetic approaches:
Modification strategies to enhance stability and efficacy include:
Experimental validation workflow:
Synthesize candidate peptides using solid-phase peptide synthesis
Evaluate binding affinity to viral proteins using biophysical techniques (SPR, ITC)
Assess antiviral activity using CPE inhibition assays against relevant viruses
Determine mechanism of action (competition assays, time-of-addition studies)
Evaluate pharmacokinetic properties and stability in biological fluids
This multifaceted approach to peptide optimization offers the potential to develop highly effective antivirals targeting the NECTIN1-virus interaction with improved stability and efficacy compared to first-generation peptide inhibitors.
Targeting NECTIN1 for therapeutic purposes presents several methodological challenges that researchers must address:
Balancing specificity with physiological functions:
NECTIN1 plays critical roles in normal cell adhesion, neural development, and synaptogenesis
Therapeutic interventions must selectively block viral interactions without disrupting essential physiological functions
Research approaches should include:
Detailed mapping of interaction domains specific to viral binding versus cellular adhesion
Development of targeted compounds that specifically block viral binding epitopes
Careful evaluation of potential developmental effects in safety studies
Delivery challenges:
Peptide-based inhibitors face delivery obstacles including:
Limited oral bioavailability
Potential immunogenicity
Poor membrane permeability
Rapid clearance and proteolytic degradation
Methodological solutions include:
Development of non-peptidic small molecule mimetics
Exploration of alternative delivery systems (nanoparticles, liposomes)
Local delivery approaches for accessible infections (e.g., topical formulations)
Timing of intervention:
NECTIN1-targeting strategies are most effective when administered during viral entry
Studies show soluble NECTIN1 effectively blocks infection when added during viral infection but has no effect when added post-infection
This necessitates:
Early therapeutic intervention
Potential prophylactic applications
Combination approaches with antivirals targeting different stages of viral replication
Cross-reactivity and resistance concerns:
Different viruses (HSV-1, HSV-2, VZV) may interact with NECTIN1 through slightly different mechanisms
Viral mutations could potentially lead to resistance
Research strategies should include:
Comprehensive testing against multiple viral strains
Resistance monitoring in experimental settings
Targeting highly conserved interaction domains
Addressing these methodological challenges requires interdisciplinary approaches combining structural biology, medicinal chemistry, drug delivery technology, and virology to develop effective NECTIN1-targeting therapeutics with minimal off-target effects.
Nectin Cell Adhesion Molecule 1, also known as Nectin-1, is a member of the nectin subfamily of immunoglobulin-like adhesion molecules. These molecules play a crucial role in calcium-independent cell-cell adhesion. Nectin-1 is encoded by the NECTIN1 gene and is involved in the formation and maintenance of adherens junctions and tight junctions in epithelial and endothelial cells .
Nectin-1 is characterized by its structure, which includes three extracellular immunoglobulin-like loops, a single transmembrane domain, and a cytoplasmic region. This structure allows Nectin-1 to form homophilic (same molecule) or heterophilic (different molecules) trans-dimers, facilitating cell-cell adhesion . Nectin-1 interacts with the actin cytoskeleton through the adaptor protein afadin, which is essential for the formation of adherens junctions .
Mutations in the NECTIN1 gene are associated with several genetic disorders, including cleft lip and palate/ectodermal dysplasia 1 syndrome (CLPED1) and non-syndromic cleft lip with or without cleft palate (CL/P). These mutations can disrupt the normal function of Nectin-1, leading to developmental abnormalities .
Research on Nectin-1 has revealed its importance in various biological processes. For instance, studies have shown that mice lacking Nectin-1 exhibit defective enamel formation in their incisor teeth, highlighting its role in dental development . Additionally, Nectin-1 is involved in the regulation of desmosome assembly, which is crucial for maintaining the integrity of epithelial tissues .