NRP1 Rat

Neuropilin 1 Rat Recombinant
Shipped with Ice Packs
In Stock

Description

NRP1 Rat Recombinant produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 842 amino acids (22-855 a.a.) and having a molecular mass of 94.8kDa (Migrates at 100-150kDa on SDS-PAGE under reducing conditions).
NRP1 is expressed with an 8 amino acid His tag at C-Terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Neuropilin 1 (Nrp1) is a transmembrane glycoprotein that acts as a co-receptor for various extracellular ligands. These ligands include class III/IV semaphorins, specific isoforms of vascular endothelial growth factor (VEGF), and transforming growth factor beta. Nrp1 binds to VEGF-A and is thought to function as a coreceptor for kinase insert domain-containing receptor (KDR). It achieves this by interacting with KDR and amplifying VEGF signaling. Notably, Nrp1 serves as a marker for regulatory T cells.
Description
Recombinant Rat NRP1, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain. It comprises 842 amino acids (22-855 a.a.) and has a molecular weight of 94.8kDa. Under reducing conditions on SDS-PAGE, it migrates at 100-150kDa. The protein is expressed with an 8 amino acid His tag at the C-terminus and purified using proprietary chromatographic methods.
Physical Appearance
A sterile, colorless solution.
Formulation
The NRP1 protein solution is provided at a concentration of 0.25mg/ml. It is prepared in Phosphate Buffered Saline (pH 7.4) and contains 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure long-term stability, adding a carrier protein (0.1% HSA or BSA) is advisable. Avoid repeated freeze-thaw cycles.
Purity
The purity of the NRP1 protein is determined to be greater than 90.0% by SDS-PAGE analysis.
Synonyms
Neuropilin-1, Vascular endothelial cell growth factor 165 receptor, CD304, Nrp1.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
FRSDKCGGTI KIENPGYLTS PGYPHSYHPS EKCEWLIQAP EPYQRIMINF NPHFDLEDRD CKYDYVEVID GENEGGRLWG KFCGKIAPSP VVSSGPFLFI KFVSDYETHG AGFSIRYEIF KRGPECSQNY TAPTGVIKSP GFPEKYPNSL ECTYIIFAPK MSEIILEFES FDLEQDSNPP GGVFCRYDRL EIWDGFPEVG PHIGRYCGQK TPGRIRSSSG ILSMVFYTDS AIAKEGFSAN YSVLQSSISE DFKCMEALGM ESGEIHSDQI TASSQYGTNW SVERSRLNYP ENGWTPGEDS YREWIQVDLG LLRFVTAVGT QGAISKETKK KYYVKTYRVD ISSNGEDWIT LKEGNKAIIF QGNTNPTDVV FGVFPKPLIT RFVRIKPASW ETGISMRFEV YGCKITDYPC SGMLGMVSGL ISDSQITASN QGDRNWMPEN IRLVTSRTGW ALPPSPHPYI NEWLQVDLGD EKIVRGVIIQ GGKHRENKVF MRKFKIAYSN NGSDWKMIMD DSKRKAKSFE GNNNYDTPEL RAFTPLSTRF IRIYPERATH SGLGLRMELL GCEVEVPTAG PTTPNGNPVD ECDDDQANCH SGTGDDFQLT GGTTVLATEK PTIIDSTIQS EFPTYGFNCE FGWGSHKTFC HWEHDSHAQL RWRVLTSKTG PIQDHTGDGN FIYSQADENQ KGKVARLVSP VVYSQSSAHC MTFWYHMSGS HVGTLRVKLH YQKPEEYDQL VWMVVGHQGD HWKEGRVLLH KSLKLYQVIF EGEIGKGNLG GIAVDDISIN NHIPQEDCAK PTDLDKKNTE IKIDETGSTP GYEEGKGDKN ISRKPGNVLK TLDPLEHHHH HH

Q&A

What is NRP1 and what are its primary functions in rats?

Neuropilin-1 (NRP1) is a transmembrane receptor protein that functions as a critical co-receptor for Vascular Endothelial Growth Factor Receptor 2 (VEGFR2) and plays essential roles in angiogenesis and vascular development in rats and other mammals. In the developing central nervous system, NRP1 is prominently expressed in endothelial cells, where it is particularly enriched in tip cells during vessel sprouting and on filopodia, similar to VEGFR2 expression patterns . Beyond endothelial cells, NRP1 is also expressed in neural progenitors of the ventricular zone that attract growing vessels into the brain, as well as in hindbrain tissue macrophages that promote anastomosis of nascent vessel sprouts . More recent research has identified NRP1 as a mechanosensitive molecule that responds to mechanical stress in various cell types, suggesting its involvement in mechanotransduction pathways particularly relevant in hypertrophic scar models in rats . The protein's widespread expression across multiple cell types involved in vascular development makes it a compelling target for research on angiogenesis, neural development, and tissue repair in rat models.

How does NRP1 expression in rats compare to other species?

NRP1 shows considerable conservation across mammalian species, with similar expression patterns and functional roles in rats compared to mice and humans. In rat models such as the tail-scar model, NRP1 is expressed in dermal endothelial cells and responds significantly to mechanical compression, mirroring observations in human hypertrophic scar tissues . The fundamental role of NRP1 in angiogenesis appears conserved, as demonstrated by studies showing that mechanical compression significantly promotes NRP1 expression in rat tail-scar dermal endothelial cells in a manner consistent with human tissues . While mouse models have been more extensively used for genetic manipulation studies (such as conditional knockout approaches using Cre/Lox systems), rat models offer advantages for certain physiological studies particularly related to wound healing and scar formation . The conservation of NRP1's mechanosensitive properties across species suggests that findings from rat models may translate effectively to human applications, especially in the context of pressure therapies for hypertrophic scarring and related conditions involving aberrant angiogenesis.

What techniques are commonly used to detect and measure NRP1 in rat tissues?

Researchers employ several complementary techniques to detect and quantify NRP1 expression in rat tissues, each with specific advantages depending on the research question. Immunofluorescence staining represents a primary method for visualizing NRP1 expression patterns in tissue sections, allowing for co-localization studies with endothelial markers (such as isolectin B4) and other cell-type specific markers (like IBA1 for macrophages) . Western blotting provides quantitative assessment of NRP1 protein levels in tissue lysates or cell cultures derived from rat models, often used to confirm knockdown efficiency in shRNA experiments . For gene expression analysis, quantitative PCR (qPCR) enables measurement of Nrp1 mRNA levels. In specialized applications, laser capture microdissection can isolate specific cell populations from rat tissues before RNA or protein analysis to determine cell-type specific expression. Flow cytometry using fluorescently-labeled antibodies against NRP1 allows for quantification of the protein on the surface of dissociated cells from rat tissues. For researchers comparing expression across multiple genes, RNA-sequencing of rat tissues provides comprehensive transcriptomic data, as evidenced by studies analyzing differentially expressed genes under mechanical stress conditions .

What rat models are most suitable for studying NRP1 in angiogenesis?

The rat tail-scar model has emerged as a particularly valuable system for studying NRP1's role in angiogenesis, especially in the context of mechanical stress responses and hypertrophic scar formation. This model involves creating a full-thickness skin wound on rat tails, allowing initial healing, and then applying controlled compression through pressure garments . The controlled environment enables researchers to study how mechanical forces influence NRP1 expression and subsequent angiogenic responses. For developmental angiogenesis studies, embryonic and neonatal rat models examining brain vascularization have proven informative, as NRP1 is highly expressed during the formation of the subventricular vascular plexus that supplies the neural progenitor zone . Rat hind-limb ischemia models provide insights into NRP1's role in therapeutic angiogenesis under pathological conditions. For tumor angiogenesis, rat xenograft models with implanted tumor cells (with or without NRP1 manipulation) help evaluate NRP1's contribution to pathological vessel formation. To study specific cellular mechanisms, primary cultures of rat dermal microvascular endothelial cells under various mechanical stress conditions offer a controlled system for examining how NRP1 mediates mechanobiological effects through pathways such as LATS1/YAP signaling .

How does NRP1 knockdown affect endothelial cell function in rat models?

NRP1 knockdown in rat endothelial cells produces significant alterations in fundamental angiogenic processes, demonstrating the protein's essential role in vascular development. When NRP1 is depleted using shRNA approaches in human dermal microvascular endothelial cells (HDMECs), which serve as an in vitro model comparable to rat endothelial cells, researchers observe marked reduction in proliferation as evidenced by decreased Ki67 expression . This proliferation defect persists regardless of whether mechanical forces are applied, suggesting that NRP1 functions as a critical mediator of both baseline and mechanically-stimulated endothelial proliferation. Beyond proliferation, NRP1 knockdown significantly impairs tube formation capacity, with notable reductions in the number of junctions and branches formed in angiogenesis assays . Importantly, the differential response to mechanical forces seen in control cells is abolished in NRP1-deficient cells, indicating that NRP1 transduces mechanical signals that normally regulate angiogenic behaviors. At the molecular level, NRP1 knockdown alters the expression and activity of downstream effectors in the Hippo pathway, particularly increasing LATS1 expression while decreasing YAP, a transcriptional regulator of angiogenic genes . These findings suggest that therapeutic strategies targeting NRP1 in rat models could effectively modulate pathological angiogenesis by disrupting both baseline endothelial functions and mechanosensitive responses.

What signaling pathways interact with NRP1 during mechanical stress response in rat models?

Recent investigations using rat models have revealed that NRP1 mediates mechanical stress responses through intricate interactions with the Hippo-YAP signaling axis, representing a novel mechanotransduction pathway in vascular biology. Under mechanical compression conditions in the rat tail-scar model, NRP1 expression increases significantly in dermal endothelial cells, concurrent with elevated LATS1 expression and reduced nuclear YAP localization . Western blot analyses demonstrate that 30 mmHg compression triggers concurrent increases in multiple Hippo pathway components, including LATS1, MST1, Salvador family WW domain-containing protein 1 (SAV1), and Mps one binder 1 (MOB1), while reducing YAP expression . This pressure-induced modulation appears to function through a mechanism where NRP1 regulates LATS1, which subsequently phosphorylates YAP, preventing its nuclear translocation and transcriptional activity. Functionally, this pathway mediates pressure therapy's anti-angiogenic effects, as compression therapy significantly reduces epidermal thickness, collagen deposition, and vascular density in rat hypertrophic scars . Interestingly, this mechanical stress response appears conserved across multiple cell types beyond endothelial cells, as bioinformatic analyses of GEO datasets (GSE137210 for glioblastoma cells and GSE120194 for hepatocellular cells) identified NRP1 as consistently upregulated by mechanical stress . The identification of this NRP1-LATS1-YAP axis provides valuable insights for optimizing compression therapy parameters in both experimental rat models and potential clinical applications.

How do results from NRP1 studies in rat models compare with knockout mouse models?

Comparative analysis between rat models and genetic mouse models reveals complementary insights into NRP1 function while highlighting important cross-species considerations. In mouse models, complete knockout of Nrp1 results in embryonic lethality between E10.5 and E14.5 with severe vascular defects, particularly in the brain and spinal cord, demonstrating the protein's essential role in developmental angiogenesis . Conditional knockout approaches using Tie2-Cre to selectively delete Nrp1 in mouse endothelial cells recapitulate these devastating vascular phenotypes, confirming NRP1's cell-autonomous requirement in endothelium . Interestingly, more targeted genetic approaches in mice, such as specific disruption of VEGF-NRP1 binding through point mutations (VEGF- Nrp1), yield unexpectedly mild phenotypes with normal vasculature and survival to adulthood, challenging previously held assumptions about NRP1's mechanism of action . In contrast, rat models typically employ different experimental approaches—rather than genetic knockouts, they utilize knockdown techniques or physiological manipulations such as mechanical compression in the rat tail-scar model . These rat studies have been particularly valuable for elucidating NRP1's role in mechanotransduction pathways involving LATS1/YAP signaling that weren't initially identified in mouse knockout studies . The rat tail-scar model specifically offers advantages for studying wound healing and pressure therapy effects that complement the developmental insights from mouse models, together providing a more comprehensive understanding of NRP1 biology across different physiological contexts.

What are the technical challenges in studying NRP1 ligand interactions in rat tissues?

Researchers face several technical challenges when investigating NRP1 ligand interactions in rat tissues, necessitating specialized approaches to overcome these obstacles. One fundamental challenge involves distinguishing between VEGF-dependent and VEGF-independent functions of NRP1, as the receptor interacts with multiple ligand families including class 3 semaphorins and various VEGF isoforms . Unlike mouse models where specific genetic mutations can selectively disrupt particular ligand interactions (such as the VEGF- Nrp1 mutation that abolishes VEGF-NRP1 binding), similar genetic tools are less developed in rat systems . Researchers must instead rely on pharmacological approaches using selective blocking antibodies or peptides, which may have variable specificity. Another significant challenge involves visualizing ligand-receptor interactions in native rat tissues, as standard immunohistochemistry detects protein presence but not active binding events. Advanced techniques such as proximity ligation assays (PLA) or fluorescence resonance energy transfer (FRET) microscopy can address this limitation but require careful optimization for rat tissues. For mechanical stress studies, maintaining physiologically relevant force application while simultaneously monitoring molecular interactions presents technical difficulties that researchers have addressed through custom pressure culture systems . Additionally, the dynamic nature of NRP1 trafficking between cellular compartments in response to ligand binding or mechanical stimulation necessitates live imaging approaches that are technically demanding in primary rat tissue samples. Overcoming these challenges requires integrated approaches combining multiple complementary techniques to comprehensively characterize NRP1 ligand interactions in physiologically relevant rat model systems.

What is the optimal protocol for immunostaining NRP1 in rat brain tissue sections?

The immunostaining of NRP1 in rat brain tissue sections requires careful attention to fixation, antigen retrieval, and antibody selection to achieve optimal results with minimal background and maximal specific signal. Begin with transcardial perfusion of the rat with 4% paraformaldehyde in phosphate-buffered saline, followed by post-fixation of dissected brains for 24 hours at 4°C to ensure proper tissue preservation while maintaining antigen integrity. Following cryoprotection in 30% sucrose, prepare 30-40 μm thick cryosections mounted on positively charged slides. For antigen retrieval, immerse sections in citrate buffer (pH 6.0) at 95°C for 20 minutes, as this has been empirically determined to effectively expose the NRP1 epitope in rat brain tissue. Block sections with 10% normal serum (matching the secondary antibody host) containing 0.3% Triton X-100 for 2 hours at room temperature to reduce nonspecific binding and facilitate antibody penetration . For primary antibody incubation, use goat anti-NRP1 antibody (R&D Systems, AF566) at 1:100 dilution in blocking solution overnight at 4°C, as this antibody has shown reliable specificity in rat tissues based on previous studies . To visualize multiple markers simultaneously, co-stain with Isolectin B4 (IB4) conjugated to Alexa Fluor 488 (1:200) to label endothelial cells and/or anti-IBA1 antibody (1:500) to identify macrophages/microglia . Following thorough washing, apply appropriate fluorescently-conjugated secondary antibodies at 1:500 dilution for 2 hours at room temperature, counterstain nuclei with DAPI, and mount with anti-fade medium. This protocol enables clear visualization of NRP1 expression in various cell types within the rat brain vasculature, including endothelial tip and stalk cells, with particularly strong detection in tip cell filopodia comparable to VEGFR2 staining patterns .

How can researchers effectively knockdown NRP1 in rat endothelial cells?

Researchers can employ several complementary approaches to effectively knockdown NRP1 in rat endothelial cells, with selection depending on experimental duration, level of suppression required, and in vitro versus in vivo context. For in vitro studies with primary rat endothelial cells, lentiviral delivery of short hairpin RNA (shRNA) targeting rat Nrp1 represents a highly effective approach, achieving sustained knockdown with approximately 70-85% reduction in protein expression as confirmed by western blot analysis . Design multiple shRNA sequences targeting different regions of rat Nrp1 mRNA to identify the most effective construct, and include a non-targeting shRNA control to account for non-specific effects. For transient knockdown with potentially higher efficiency but shorter duration, synthesize small interfering RNA (siRNA) duplexes targeting rat Nrp1 and deliver using lipid-based transfection reagents optimized for endothelial cells, typically achieving peak knockdown 48-72 hours post-transfection. For in vivo applications, consider adeno-associated virus (AAV) vectors with endothelial-specific promoters (such as Tie2 or VE-cadherin) to deliver shRNA constructs directly to rat vasculature through tail vein injection, focusing on serotypes with endothelial tropism such as AAV9. To validate knockdown efficiency in both in vitro and in vivo models, employ multiple complementary methods including western blotting for protein levels, qPCR for mRNA expression, and immunofluorescence to assess spatial patterns of remaining NRP1 expression . When interpreting functional outcomes following knockdown, consider potential compensatory upregulation of related family members such as NRP2, which may partially mask phenotypes, particularly in chronic knockdown scenarios.

What experimental design is recommended for studying NRP1's role in rat hypertrophic scar models?

The optimal experimental design for investigating NRP1's role in rat hypertrophic scar models requires careful attention to wound creation, pressure application parameters, and comprehensive outcome measurements. Establish experimental groups including: control (no pressure therapy), pressure therapy only, NRP1 knockdown without pressure, and NRP1 knockdown with pressure therapy, with minimum sample sizes of 6-8 rats per group to ensure statistical power . Create standardized full-thickness wounds (6×6 mm) on rat tails, secured with steel rings to create tension conducive to hypertrophic scarring, allowing initial healing for 14 days before intervention . For pressure therapy, design custom compression garments calibrated to deliver 20-30 mmHg pressure consistently to the scarred region, with precise pressure monitoring using specialized sensors to ensure reproducibility. Implement NRP1 manipulation through local injection of shRNA-expressing viral vectors directly into the developing scar tissue at approximately day 14, with contralateral control injections containing non-targeting constructs to provide within-subject controls. Conduct comprehensive outcome assessments at day 28-35 including: macroscopic scar measurements (height, redness, pliability using durometer), histological analysis (epidermal thickness, collagen organization using Masson's trichrome staining), immunohistochemical evaluation (NRP1, LATS1, YAP, Ki67 expression patterns), vascular density quantification, and molecular analysis of excised tissue (western blotting, qPCR for angiogenic factors and mechanical stress response genes) . Additionally, consider incorporating intravital imaging of labeled endothelial cells in select animals to capture dynamic vessel remodeling processes. This integrated experimental approach enables robust assessment of NRP1's mechanistic role in pressure therapy's effects on hypertrophic scarring while controlling for key variables that might confound interpretation.

How should researchers interpret contradictory findings between NRP1 knockout and knockdown studies?

Researchers should systematically evaluate several factors when confronting contradictory findings between NRP1 knockout and knockdown studies, recognizing that these discrepancies often reveal important biological nuances rather than experimental failures. First, consider developmental compensation mechanisms that may be activated in germline knockout models but not in acute knockdown approaches—genetic knockout from embryonic stages allows developmental adaptation through upregulation of related pathways or NRP2, while acute knockdown in adult tissues reveals immediate requirements without compensatory changes . Second, evaluate the degree and specificity of NRP1 reduction, as knockout approaches completely eliminate the protein while knockdown typically achieves partial reduction (70-90%), potentially preserving threshold-dependent functions while disrupting others . Third, assess cell type specificity, as global knockouts affect all NRP1-expressing cells while many knockdown approaches target specific cell populations; mouse studies clearly demonstrate that endothelial-specific deletion of NRP1 recapitulates the vascular phenotype of global knockouts, confirming its cell-autonomous requirement in endothelium . Fourth, analyze domain-specific functions, as knockdown reduces all NRP1 domains proportionally while some knockout strategies selectively disrupt specific interactions (such as VEGF binding) while preserving others (such as SEMA3 binding), revealing unexpected functional separations . Finally, compare model systems carefully, as mouse knockout findings may not directly translate to rat knockdown studies due to species-specific differences in vascular development, wound healing processes, and the molecular context in which NRP1 functions . By systematically evaluating these factors, researchers can transform apparent contradictions into mechanistic insights about NRP1's context-dependent functions and identify the most appropriate model systems for specific research questions.

How is NRP1 expression regulated under different physiological conditions in rats?

NRP1 expression in rat tissues demonstrates dynamic regulation across various physiological contexts, with mechanical forces emerging as particularly significant modulators. Under mechanical stress conditions, NRP1 expression increases significantly in multiple cell types including dermal endothelial cells, as demonstrated in both the rat tail-scar model under compression therapy and in pressure-cultured human dermal microvascular endothelial cells serving as in vitro models . This mechanosensitive upregulation appears consistent across diverse cell types, as bioinformatic analysis of gene expression datasets from glioblastoma cells (GSE137210) and hepatocellular cells (GSE120194) similarly identified NRP1 as a mechanical stress-responsive gene . During development, NRP1 expression in rat brain vasculature follows a spatiotemporal pattern with particularly strong expression in endothelial tip cells and their filopodia during active angiogenesis, suggesting developmental regulation aligned with vessel sprouting programs . The protein shows differential expression across endothelial subtypes, with enrichment in tip cells versus stalk cells, indicating potential regulation by tip cell specification factors such as Notch signaling components . In tissue macrophages, NRP1 expression increases during development, with higher levels observed at E11.5 compared to E10.5 in mouse models (with rat macrophages presumed to follow similar developmental patterns), particularly in processes extending toward endothelial tip cells, suggesting active regulation during vascular anastomosis . These diverse regulatory patterns highlight NRP1's role as an integrator of multiple physiological signals, with expression levels responsive to mechanical forces, developmental timing, and cell type-specific differentiation programs in rat model systems.

What is the relationship between NRP1 and VEGFR2 expression in rat endothelial cells?

The relationship between NRP1 and VEGFR2 in rat endothelial cells represents a complex interplay beyond simple co-receptor function, with emerging evidence indicating that NRP1 actively regulates VEGFR2 surface expression and signaling dynamics. Immunofluorescence studies demonstrate that both proteins are co-expressed in endothelial cells during brain vascularization, with particularly strong expression in tip cells and their filopodia, suggesting coordinated regulation and function during active angiogenesis . Beyond mere co-expression, recent findings indicate that NRP1 functionally regulates VEGFR2 availability, as VEGF−Nrp1-deficient vessels (mutants with disrupted VEGF-NRP1 binding) show reduced VEGFR2 surface expression in vivo, demonstrating that NRP1 controls the levels of its co-receptor VEGFR2 . This regulatory relationship appears independent of direct VEGF-NRP1 binding, as mice with mutations that specifically abolish VEGF-NRP1 interactions (VEGF−Nrp1) survive to adulthood with normal vasculature, contrasting sharply with the severe vascular defects and embryonic lethality observed in complete Nrp1 knockouts . In rat models examining mechanical stress responses, pressure application concurrent with increased NRP1 expression leads to altered downstream signaling patterns typically associated with VEGFR2 activation, suggesting NRP1-mediated modulation of VEGFR2 signaling under mechanical stress . The emerging model suggests that rather than primarily functioning through direct VEGF binding, NRP1 guides developmental angiogenesis by regulating VEGFR2 trafficking, surface presentation, and signaling dynamics in rat endothelial cells . This relationship has significant implications for interpreting experimental manipulations of either protein in rat models and suggests therapeutic approaches targeting NRP1-VEGFR2 interactions rather than just VEGF binding.

How does mechanical stress modulate NRP1 expression and function in rat models?

Mechanical stress exerts profound effects on both NRP1 expression and function in rat models, establishing this transmembrane receptor as a critical mechanotransducer in vascular biology. In the rat tail-scar model, applied compression therapy significantly increases NRP1 expression in dermal endothelial cells as visualized by immunofluorescence analysis, indicating direct transcriptional or post-transcriptional upregulation in response to mechanical forces . This mechanosensitive expression pattern appears conserved across multiple cell types beyond endothelial cells, as demonstrated by bioinformatic analysis of gene expression datasets from mechanically stressed glioblastoma and hepatocellular cells, suggesting a fundamental cellular response rather than an endothelial-specific phenomenon . Functionally, mechanical compression simultaneously enhances LATS1 expression and reduces YAP nuclear localization in rat tail-scar tissues, establishing a signaling axis through which NRP1 transduces mechanical cues . This pathway mediates the therapeutic effects of compression therapy, as pressure application significantly reduces epidermal thickness, collagen deposition, and vascular density in hypertrophic scars—effects that are mimicked by NRP1 knockdown even without mechanical stimulation . At the molecular level, applied pressure of 30 mmHg increases expression of multiple Hippo pathway components (LATS1, MST1, SAV1, MOB1) while reducing YAP expression and enhancing YAP phosphorylation, indicating that the NRP1-LATS1-YAP axis constitutes a primary mechanotransduction pathway in this context . These findings establish NRP1 as a mechanically-sensitive molecular switch that integrates physical cues into biochemical signals controlling angiogenesis and tissue remodeling in rat models, with potential therapeutic implications for pressure-based treatments of hypertrophic scarring.

What is the current understanding of NRP1's role in rat developmental angiogenesis?

Current understanding of NRP1's role in rat developmental angiogenesis integrates insights from multiple model systems, revealing this receptor's multifaceted functions in vascular patterning. During brain vascularization, NRP1 is strongly expressed in endothelial cells forming the subventricular vascular plexus that supplies the neural progenitor zone, with particularly prominent expression in tip cells and their filopodia . This expression pattern suggests a specialized function in guiding sprouting angiogenesis, consistent with findings from genetic mosaic analyses in mouse models demonstrating that NRP1-expressing endothelial cells preferentially attain the tip cell position when competing with NRP1-negative cells in chimeric vessel sprouts . Beyond simple expression patterns, functional studies across rodent models indicate that NRP1 promotes brain angiogenesis cell-autonomously in endothelium, independent of heterotypic interactions with nonendothelial cells, as selective targeting of Nrp1 in neural progenitors or macrophages does not disrupt brain vascularization while endothelial deletion recapitulates the severe vascular defects of global knockouts . Mechanistically, recent findings challenge the traditional view that NRP1 functions primarily through direct VEGF binding, as mouse mutants specifically lacking VEGF-NRP1 binding capabilities (VEGF−Nrp1) develop normal vasculature, suggesting alternative mechanisms . The emerging model proposes that NRP1 regulates VEGFR2 surface expression and availability, thereby controlling VEGF signaling indirectly rather than through direct ligand binding . These integrated insights suggest that in rat developmental angiogenesis, NRP1 functions as a key endothelial regulator that promotes tip cell functionality and vessel sprouting through modulation of VEGFR2 dynamics rather than primarily serving as a VEGF co-receptor.

How can NRP1 targeting be optimized in rat models of pathological angiogenesis?

Optimizing NRP1 targeting in rat models of pathological angiogenesis requires strategic considerations spanning delivery mechanisms, molecular specificity, and therapeutic timing to maximize efficacy while minimizing off-target effects. Based on recent findings regarding NRP1's mechanistic role, researchers should consider combination approaches targeting both NRP1 and its downstream effectors in the LATS1/YAP pathway, as knockdown studies demonstrate that NRP1 inhibition reduces YAP expression and inhibits endothelial cell proliferation and tube formation . For delivery strategies in rat models, locally administered adeno-associated viral vectors expressing shRNA against NRP1 offer prolonged knockdown with endothelial specificity when employing appropriate serotypes and promoters, while nanoparticle-encapsulated siRNA provides shorter-term suppression with potentially greater initial potency. Timing of intervention requires careful consideration, as NRP1 functions differ between developmental and pathological angiogenesis—for models of tumor angiogenesis or retinopathy, intervention after initial vessel network establishment but during active pathological sprouting likely provides optimal therapeutic windows . Given findings that NRP1 regulates VEGFR2 surface expression independently of direct VEGF binding, targeting the NRP1-VEGFR2 interaction interface rather than the VEGF binding domain might prove more effective, suggesting development of peptides or small molecules that disrupt receptor complex formation . For hypertrophic scar models, combining NRP1 inhibition with optimized mechanical pressure (20-30 mmHg) may provide synergistic effects, as both interventions appear to modulate the same LATS1/YAP pathway . Additionally, considering NRP1's expression in multiple cell types including macrophages, cell-type specific targeting approaches may refine therapeutic outcomes by selectively inhibiting endothelial NRP1 while preserving potential beneficial functions in other cellular contexts .

What are the potential translational implications of rat NRP1 studies for human disease?

Findings from rat NRP1 studies offer significant translational potential for human disease management, particularly in conditions involving pathological angiogenesis or aberrant tissue repair. The identification of NRP1 as a mechanosensitive regulator of the LATS1/YAP pathway in rat hypertrophic scar models directly informs optimization of pressure garment therapy (PGT), a standard clinical intervention for human hypertrophic scarring . Current pressure recommendations range from 15-40 mmHg, but mechanistic understanding from rat studies suggests that targeting 30 mmHg may optimize NRP1-mediated effects on angiogenesis inhibition while potentially improving patient comfort and compliance . Beyond pressure therapy, the molecular pathway involving NRP1-LATS1-YAP identified in rat models provides new pharmaceutical targets for anti-scarring therapies, potentially enabling development of topical agents that mimic mechanical compression's effects without requiring continuous pressure application . For oncology applications, the cell-autonomous requirement for NRP1 in endothelial tip cells during angiogenesis, demonstrated in rat and mouse models, supports ongoing development of NRP1-targeted anti-angiogenic therapies for human cancers, with the important refinement that targeting NRP1-VEGFR2 interactions may prove more effective than blocking VEGF-NRP1 binding based on genetic studies . In cerebrovascular diseases, insights from developmental studies showing NRP1's critical role in brain angiogenesis inform potential regenerative approaches for stroke recovery, where promoting controlled angiogenesis through modulation rather than complete inhibition of NRP1 function might enhance therapeutic outcomes . Additionally, the conservation of NRP1's mechanical stress responses across multiple cell types suggests broader applications in fibrotic diseases affecting the liver, kidney, and lung, where mechanical forces similarly influence disease progression through comparable molecular pathways .

What emerging technologies could advance NRP1 research in rat models?

Emerging technologies across multiple disciplines promise to significantly advance NRP1 research in rat models, enabling more precise manipulation and analysis of this receptor's complex functions. CRISPR-Cas9 genome editing technology is increasingly being optimized for rat models, potentially enabling generation of domain-specific NRP1 mutations comparable to those developed in mice (such as selective disruption of VEGF-NRP1 binding) to dissect functional domains with unprecedented precision . High-resolution intravital imaging approaches, including two-photon microscopy with genetically encoded fluorescent reporters, would allow real-time visualization of NRP1-expressing cells during angiogenesis in living rat tissues, capturing dynamic processes such as tip cell competition and filopodia extension that static analyses cannot reveal . For mechanobiology studies, microfluidic devices and magnetically actuated materials enabling precisely controlled application of mechanical forces to rat endothelial cells while simultaneously monitoring molecular responses will refine understanding of NRP1's mechanosensitive properties . Single-cell RNA sequencing of rat tissues under various conditions (development, injury, mechanical stress) would provide comprehensive transcriptional profiles of NRP1-expressing cells, revealing previously unrecognized heterogeneity and potential new interaction partners. Spatial transcriptomics and proteomics technologies maintain tissue architecture while providing molecular resolution, allowing correlation of NRP1 expression with specific microenvironmental features in complex rat tissues. For translational applications, biomaterials that enable controlled release of NRP1-targeting agents specifically within rat hypertrophic scars could improve therapeutic delivery while mimicking mechanical compression effects . Finally, systems biology approaches integrating multi-omic data from rat models could generate comprehensive interaction networks centered on NRP1, predicting new functions and pathway interactions to guide hypothesis generation for future experimental validation.

What are the key unresolved questions about NRP1 function in rat models?

Despite significant advances, several fundamental questions regarding NRP1 function in rat models remain unresolved, representing critical directions for future research. First, the precise molecular mechanism by which NRP1 senses mechanical forces remains unclear—whether through conformational changes in its extracellular domains, interactions with mechanosensitive membrane components, or alterations in its cytoplasmic domain associations requires investigation using biophysical approaches in rat endothelial cells . Second, the temporal dynamics of NRP1-VEGFR2 interactions during different phases of angiogenesis (initiation, elongation, anastomosis, maturation) remain poorly characterized in rat models, necessitating time-course studies with methods capable of detecting protein complexes in situ . Third, while NRP1's importance in brain angiogenesis is established, its relative contribution to vascular development in other rat organs remains incompletely documented, requiring systematic comparative analyses across tissue beds with potentially different regulatory mechanisms . Fourth, the potential functional redundancy or compensation between NRP1 and its homolog NRP2 in rat models requires clarification through combined knockdown approaches and detailed expression mapping, particularly in contexts where NRP1 manipulation produces milder than expected phenotypes. Fifth, the cross-talk between NRP1-mediated mechanotransduction and classical growth factor signaling pathways remains to be fully elucidated—specifically how mechanical forces might modify NRP1's response to ligands such as VEGF-A or semaphorins in rat tissues . Finally, the long-term consequences of NRP1 manipulation in rat models, particularly regarding vascular stability, regression, and potential compensatory angiogenesis, remain poorly understood, necessitating extended time-course studies with reversible manipulation systems. Addressing these questions will require integrating advanced genetic tools, high-resolution imaging, and systems biology approaches to comprehensively understand NRP1's multifaceted functions in rat model systems.

Product Science Overview

Structure and Function

Neuropilin-1 consists of several distinct domains:

  • Extracellular Domain (ECD): This domain contains two N-terminal CUB domains, two F5/8 type C domains with homology to coagulation factors V and VIII, and a MAM (meprin) domain . The ECD is responsible for binding to various ligands, including vascular endothelial growth factor (VEGF) and semaphorins, which are critical for its role in angiogenesis and axon guidance .
  • Transmembrane Domain ™: This domain anchors the protein to the cell membrane.
  • Cytoplasmic Domain: Although relatively short, this domain is essential for intracellular signaling.
Recombinant Rat Neuropilin-1

Recombinant rat Neuropilin-1 is typically produced using expression systems such as mouse myeloma cell lines (NS0) or insect cell lines (Sf21) infected with baculovirus . The recombinant protein is often engineered to include tags, such as a 6-His tag, to facilitate purification and detection .

Applications

Recombinant Neuropilin-1 is widely used in research to study its role in:

  • Angiogenesis: By binding to VEGF, Neuropilin-1 promotes the formation of new blood vessels, which is crucial in both normal physiological processes and pathological conditions like cancer .
  • Axon Guidance: Neuropilin-1 interacts with semaphorins to guide the growth of axons during neural development .
  • Cancer Research: Due to its role in angiogenesis, Neuropilin-1 is a target for cancer therapy research. Inhibiting its interaction with VEGF can potentially reduce tumor growth and metastasis .
Stability and Storage

Recombinant Neuropilin-1 is typically lyophilized and should be reconstituted in sterile PBS. It is stable for up to 12 months when stored at -20 to -70°C and should be handled to avoid repeated freeze-thaw cycles .

Neuropilin-1’s multifaceted roles in critical biological processes make it a significant protein for research in developmental biology, neurobiology, and oncology.

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.