Neuropilin-1 (NRP1) is a transmembrane glycoprotein critical for vascular development, neuronal guidance, and immune regulation. Mouse models of NRP1 (Mus musculus Nrp1) serve as essential tools for studying its multifaceted roles in physiology and disease. These models include global knockouts, conditional knockouts, and hypomorphic alleles that enable tissue-specific functional analyses .
Embryonic Lethality: Homozygous Nrp1 −/− mice exhibit lethality at E10–E12.5 due to defective yolk sac vascularization and abnormal vessel sprouting .
Vascular Phenotypes: Overexpression of NRP1 causes hypervascularization and hemorrhagic vessels, while endothelial-specific deletion results in mild brain defects .
Reduced NRP1 Expression: The Y297A mutation disrupts VEGF binding and reduces NRP1 protein levels by ~70% in homozygous mice .
Postnatal Defects: These mice show increased mortality (50% survival by 6 weeks), growth retardation, and impaired angiogenesis .
Phenotype | Y297A/Y297A Mice | Wild-Type Mice |
---|---|---|
Survival at 6 weeks | 50% | 100% |
Body weight (P7) | 25% reduction | Normal |
Angiogenesis | Delayed vessel maturation | Normal |
Regulatory T Cells (Tregs): NRP1 stabilizes intratumoral Tregs, suppressing antitumor immunity. Conditional deletion in Tregs enhances CD8+ T cell responses without compromising peripheral tolerance .
CD8+ T Cell Activation: NRP1 marks highly activated CD8+ T cells in cerebral malaria (Plasmodium berghei) and lymphocytic choriomeningitis virus (LCMV) infections. Its ablation reduces cytotoxic T cell infiltration and pathology .
NRP1 is expressed on tumor-associated macrophages and dendritic cells, though its functional role remains less characterized compared to lymphoid cells .
Endothelial-Specific Knockouts (iECKO): Show increased vascular leakage in response to VEGF-A, indicating NRP1 stabilizes endothelial junctions .
Smooth Muscle-Specific Knockouts (Nrp1SMKO): Exhibit reduced contractile protein expression (e.g., smMHC) and impaired intestinal motility .
NRP1+ Tregs are enriched in tumors and associated with poor prognosis. Preclinical studies suggest NRP1 blockade could enhance checkpoint inhibitor efficacy .
Mutant glycyl-tRNA synthetase sequesters NRP1, disrupting VEGF signaling and contributing to Charcot-Marie-Tooth disease .
Cerebral Malaria: NRP1-deficient CD8+ T cells reduce blood-brain barrier leakage and parasite sequestration .
LCMV Infection: NRP1 ablation mitigates liver pathology by dampening cytotoxic T cell responses .
While NRP1 mouse models have clarified its roles in immunity and development, key gaps remain:
Tissue-specific functions of NRP1 in myeloid cells.
Therapeutic window for NRP1 inhibition without compromising vascular homeostasis.
NRP1 is abundantly expressed on the surface of various cell types in mice. In vascular tissue, it's predominantly expressed in endothelial cells where it forms heterocomplexes with VEGFR2. NRP1 is also expressed in smooth muscle cells, particularly in gastrointestinal smooth muscle. Additionally, NRP1 expression has been documented in immune cells and neuronal tissues.
Expression can be detected through:
Immunofluorescent staining of tissues (effective in ear skin, back skin, and other organs)
Western blotting of tissue lysates (lung lysates provide reliable detection)
qPCR for mRNA expression analysis
Protein detection typically shows NRP1 at approximately 120-140 kDa molecular weight, though an 80 kDa protein of unknown origin may also be detected by some antibodies .
Confirmation of NRP1 deletion should involve multiple methods:
Protein level verification:
Western blot analysis of tissue lysates (lung tissue provides reliable results)
Quantification shows significant reduction in NRP1 protein levels in knockout mice
Tissue-specific verification:
Immunofluorescent staining of target tissues
Complete loss of NRP1 can be visualized in tissues such as ear and back skin
Functional verification:
Assessing phenotypic changes characteristic of NRP1 deletion
For endothelial knockout, examining vascular permeability responses
For smooth muscle knockout, evaluating contractility parameters
For inducible models, verification should be performed after the induction period to confirm temporal deletion efficiency .
The effect of NRP1 deletion on VEGFA-induced vascular permeability shows remarkable tissue specificity, presenting a complex research area:
Tissue-dependent responses:
Ear dermis: Endothelial-specific NRP1 deletion (NRP1 iECKO) increases VEGFA-induced vascular leakage
Trachea and back skin: NRP1 deletion reduces VEGFA-induced permeability by approximately 75%
Kidney, skeletal muscle, and heart: NRP1 appears to play a passive role in VEGFA-induced leakage
Quantitative analysis methods:
Measure lag period (time from stimulation to leakage onset)
Assess rate of dextran extravasation
Analyze disruption of endothelial cell-cell junctions
Paradoxical findings between approaches:
Global pharmacological NRP1 inhibition using blocking antibodies reduces vascular leakage in ear dermis
This contrasts with increased leakage observed in endothelial-specific genetic deletion
Global genetic deletion (NRP1 iKO) results match antibody blocking rather than EC-specific deletion
These findings demonstrate that NRP1 can function as a positive regulator, negative regulator, or passive component in vascular permeability depending on tissue context and experimental approach .
NRP1 plays a crucial role in maintaining gastrointestinal smooth muscle contractility, particularly in aging mice:
Key phenotypic changes in NRP1 SMKO mice:
Significant reduction in intestinal length by 6 months of age
Development of severe constipation by 18 months
Intestinal enlargement consistent with chronic intestinal pseudo-obstruction
Thinning of intestinal smooth muscle layers
Molecular mechanisms:
Reduced expression of contractile proteins in smooth muscle cells
Specifically decreased expression of smooth muscle myosin heavy chain (smMHC) isoform SMB
Significant increase in small-conductance calcium-activated potassium channel 3 (SK3/KCa2.3)
SK3/KCa2.3 is known to negatively regulate smooth muscle contraction
Experimental approaches for assessment:
Contractility assays of isolated intestinal segments
Protein expression analysis of contractile apparatus components
Histological assessment of muscle layer thickness
Functional transit studies to measure GI motility
The age-dependent nature of this phenotype suggests NRP1 is essential for maintaining, rather than establishing, the contractile phenotype of visceral smooth muscle cells .
The discrepancies between global and tissue-specific NRP1 deletion outcomes present a methodological challenge:
Contrasting phenotypes:
In ear skin, endothelial-specific deletion increases VEGFA-induced permeability
Global deletion (both genetic and antibody-mediated) decreases permeability
In trachea and back skin, both approaches reduce permeability
Methodological considerations:
Developmental compensation: Global knockouts may activate compensatory mechanisms absent in tissue-specific models
Cell-cell interactions: NRP1 on non-endothelial cells may influence endothelial function through juxtacrine signaling
Temporal aspects: Acute (antibody) versus chronic (genetic) loss of function yields different results
Experimental design recommendations:
Combine multiple approaches (genetic deletion, antibody blocking)
Include both global and tissue-specific models
Employ acute inducible systems to minimize developmental compensation
Analyze multiple tissue beds to account for organotypic differences
This research challenge highlights the importance of using complementary approaches and careful interpretation of seemingly contradictory results .
NRP1 serves important functions in immune regulation, particularly in the context of autoimmune diseases:
NRP1 in experimental autoimmune encephalomyelitis (EAE):
Tissue-specific deletion of NRP1 in CD4+ T cells results in increased EAE severity
NRP1-deficient mice exhibit preferential TH-17 lineage commitment
Regulatory T cell (Treg) functionality is decreased in NRP1-deficient models
Mechanistic insights:
NRP1-expressing CD4+ T cells suppress effector T-cell proliferation
NRP1-mediated suppression can be inhibited by TGF-β blockade
NRP1-mediated suppression is independent of IL-10 signaling
Experimental approaches:
Generation of retroviral GFP vector containing mouse Nrp1 cDNA
Isolation of CD4+ T cells from naïve myelin basic protein (MBP)-specific T cells
Assessment of T cell differentiation patterns and cytokine production
In vivo and in vitro suppression assays
These findings suggest NRP1 is essential for maintaining peripheral tolerance, and its absence can lead to unchecked autoimmune responses .
Contradictory findings regarding NRP1 function, particularly in vascular biology, can be approached methodologically:
Sources of experimental variability:
Model differences: Global versus conditional knockouts yield different results
Tissue specificity: NRP1 functions differently in ear skin versus trachea or back skin
Temporal factors: Acute versus chronic loss of function produces different phenotypes
Cell-type interactions: Non-autonomous effects when multiple cell types express NRP1
Reconciliation strategies:
Side-by-side comparisons: Test multiple models under identical conditions
Rescue experiments: Re-express NRP1 in knockout backgrounds to confirm specificity
Domain-specific mutations: Target specific functional domains rather than whole protein
Single-cell approaches: Analyze cell-type specific responses within heterogeneous tissues
Data interpretation framework:
Consider NRP1 as a context-dependent modulator rather than a simple positive/negative regulator
Analyze both kinetic parameters (response onset, duration) and magnitude of effects
Map tissue-specific receptor and ligand expression patterns
For example, when studying vascular permeability, researchers should employ both antibody blockade and genetic deletion approaches in the same tissue beds under identical stimulation conditions .
Several experimental approaches are particularly valuable for studying NRP1 function in vivo:
Vascular permeability assessment:
Intravital microscopy using fluorescent dextran tracers
Quantitative analysis of both temporal aspects (lag period) and magnitude (rate of extravasation)
Miles assay for macroscopic assessment of vascular leakage
Smooth muscle function evaluation:
Intestinal transit assays using fluorescent tracers
Ex vivo contractility measurements of isolated muscle segments
Histological assessment of muscle layer thickness and organization
Immune function analysis:
T cell proliferation and suppression assays
Cytokine production profiling
Adoptive transfer experiments with NRP1-deficient versus wild-type T cells
Molecular interaction studies:
Co-immunoprecipitation to detect NRP1-VEGFR2 complexes
Proximity ligation assays for detecting protein-protein interactions in situ
FRET-based approaches for monitoring receptor interactions in live cells
The optimal approach depends on the specific research question, but combining multiple methodologies provides the most robust results .
The cytoplasmic domain of NRP1 plays specific roles in receptor trafficking and signaling:
Structure and interactions:
Contains a C-terminal SEA motif
Functions as a PDZ binding domain
Mediates binding to synectin (GIPC1)
Functional consequences:
Regulates endocytic trafficking of both NRP1 and VEGFR2
Removal delays VEGFR2 endocytosis following VEGFA binding
Leads to enhanced surface retention of VEGFR2
Reduces phosphorylation of tyrosine (Y)1175 in VEGFR2
Experimental approaches:
Generation of cytoplasmic domain deletion mutants
Analysis of receptor internalization kinetics
Assessment of downstream signaling pathway activation
Comparison with full NRP1 knockouts to distinguish domain-specific functions
Understanding the cytoplasmic domain's function is particularly important because it reveals how NRP1 acts as a modulator of VEGFR2 activation and downstream signaling pathways rather than simply as a ligand-binding co-receptor .
The role of NRP1 in olfactory system development, particularly regarding glomerular positioning, remains controversial:
The NRP1 gradient model:
Original hypothesis proposed NRP1 forms an anterior-low to posterior-high gradient in the olfactory bulb
OR-derived cAMP signals were thought to determine NRP1 expression levels
This gradient was proposed to determine anterior-posterior patterning of glomeruli
Contradictory findings:
Recent studies using conditional NRP1 knockout mice observed various configurations for M71 glomeruli
Glomerular positions do not undergo the simple anterior shift previously reported
These findings contradict the original model of NRP1-dependent anterior-posterior patterning
Current research approaches:
Use of gene-targeted mouse strains with reporters for specific odorant receptors
Analysis of glomerular positioning in conditional NRP1 knockout backgrounds
3D reconstruction of glomerular maps in the olfactory bulb
The current evidence suggests that while NRP1 may influence olfactory system development, its role in glomerular positioning is more complex than initially proposed .
Understanding NRP1's complex role in vascular biology suggests potential therapeutic applications:
Targeting considerations:
Tissue-specific effects require precise targeting approaches
Global inhibition may produce different effects than cell-type specific approaches
Temporal aspects of intervention may determine efficacy
Potential therapeutic applications:
Vascular permeability modulation: NRP1 blocking antibodies reduce VEGFA-induced vascular leakage in specific tissues
Tumor angiogenesis: Several reports link increased NRP1 expression in tumors to poor prognosis
Inflammatory conditions: NRP1's role in immune regulation suggests potential for targeting inflammatory disorders
Experimental therapeutic approaches:
Antibody-based blocking strategies
Small molecule inhibitors targeting specific NRP1 domains
Cell-type specific delivery systems
Understanding the relationship between NRP1 and VEGFA-VEGFR2 signaling has potential therapeutic benefit, particularly in conditions involving pathological vascular permeability or angiogenesis .
Researchers face several technical challenges when investigating NRP1:
Genetic model limitations:
Global knockout is embryonic lethal, necessitating conditional approaches
Potential off-target effects of Cre expression in conditional models
Incomplete recombination in conditional systems
Tissue-specific variability:
Divergent phenotypes between tissue beds complicate interpretation
Requires analysis of multiple tissues to establish comprehensive understanding
Need for standardized methodologies across different vascular beds
Protein detection challenges:
Some antibodies may detect an 80 kDa protein of unknown origin
Confirmation of specific signal requires knockout controls
Recommended use of multiple detection methods (Western blot, immunofluorescence)
Functional assay standardization:
Vascular permeability assays require precise quantification of both temporal and magnitude parameters
Ex vivo contractility measurements need standardized tension and stimulation protocols
Immune function assays benefit from defined cell populations and culture conditions
Addressing these challenges requires rigorous experimental design, appropriate controls, and combining multiple complementary approaches .
When faced with contradictory results between global and tissue-specific knockouts:
Analytical framework:
Consider non-cell-autonomous effects of NRP1 in global knockouts
Evaluate potential developmental compensation mechanisms
Assess timeline differences between acute and chronic loss of function
Experimental validation approaches:
Complement genetic models with pharmacological interventions
Use inducible systems to minimize developmental effects
Perform tissue-specific rescue experiments
Interpretation guidelines:
View contradictions as revealing of complex biology rather than experimental failures
Map tissue-specific expression of NRP1 interaction partners
Consider microenvironmental factors that might influence NRP1 function
The contrasting phenotypes observed between global and endothelial-specific NRP1 deletion in ear skin vascular permeability exemplify this challenge, suggesting important juxtacrine signaling between endothelial and non-endothelial cells .
Neuropilin-1 consists of several distinct domains:
The extracellular domain of mouse Neuropilin-1 shares a high degree of similarity with its counterparts in other species, including humans, rats, horses, cows, and dogs . This conservation across species underscores the protein’s fundamental role in physiological processes.
Recombinant mouse Neuropilin-1 is typically produced in a mouse myeloma cell line (NS0) and includes a C-terminal 6-His tag for purification purposes . The recombinant protein is often used in research to study its interactions with various ligands, such as VEGF and SEMA3A, and to explore its role in different biological processes.
The recombinant form of Neuropilin-1 is available in a carrier-free formulation, which means it does not contain bovine serum albumin (BSA). This is particularly useful for applications where the presence of BSA could interfere with experimental results .
Neuropilin-1 is a subject of extensive research due to its involvement in critical biological processes. Studies have shown that inhibiting Neuropilin-1 can lead to improved tumor growth inhibition in vivo, highlighting its potential as a therapeutic target in cancer treatment . Researchers have developed monoclonal antibodies that specifically bind to different domains of Neuropilin-1, blocking its interactions with ligands like VEGF and SEMA3A .