FLT1 (VEGFR1) is a tyrosine kinase receptor activated by VEGF-A, VEGF-B, and placental growth factor (PGF) . The His-tagged recombinant form retains critical biological functions:
VEGF Signaling Modulation: Acts as a decoy receptor, inhibiting VEGF-A-driven endothelial cell proliferation .
Structural Insights: The Ig-like domains 1–3 are sufficient for ligand binding, enabling studies on VEGF receptor interactions .
Disease Relevance: Elevated FLT1 expression correlates with pre-eclampsia, cancer progression, and PARP inhibitor resistance .
VEGF Interaction: FLT1 Human, His binds VEGF-A (KD: ~10–100 pM), validated via surface plasmon resonance (SPR) and ELISA .
SARS-CoV-2 Spike Protein: Binds spike-RBD, suggesting a role in COVID-19 pathogenesis .
Cancer Resistance: FLT1 activation in tumor cells promotes PARP inhibitor resistance via AKT survival pathways and immune evasion .
Hypertension and Longevity: The FLT1 SNP rs3794396 ameliorates mortality risk in hypertensive individuals .
FLT1 signaling in BRCA1-mutant tumors activates AKT and reduces CD8+ T-cell infiltration, enabling resistance .
Axitinib (FLT1 inhibitor) restores PARP inhibitor sensitivity in preclinical models .
FLT1 binds SARS-CoV-2 spike-RBD, facilitating viral entry into A549 and MDA-MB231 cells .
Anti-spike antibodies from vaccinated individuals recognize FLT1-spike complexes, implicating FLT1 in immune responses .
Human FLT1 is a tyrosine kinase receptor containing seven immunoglobulin-like domains in its extracellular region followed by a transmembrane domain and an intracellular tyrosine kinase domain. The gene consists of 30 exons and produces two main products: full-length FLT1 and a soluble form (sFLT1) derived from alternative splicing involving the first 13 exons with an intron-13-derived tail of 31 amino acids . The soluble form contains the first six Ig-like domains without the transmembrane or tyrosine kinase domains. When adding a His-tag for purification purposes, researchers should consider placing it at the N-terminus rather than the C-terminus to avoid interfering with signaling functions. Importantly, functionality assays comparing tagged versus untagged protein should be performed to ensure the tag does not disrupt binding to ligands such as VEGF-A.
When conducting RNA interference experiments targeting FLT1, validation requires multiple complementary approaches:
mRNA quantification: Real-time PCR should show significant inhibition of FLT1 mRNA levels without affecting related receptors such as KDR (p<0.01 considered significant) .
Protein verification: Western blotting using specific antibodies (e.g., monoclonal antibody against FLT1 at 1:500 dilution, V4262, Sigma-Aldrich) should confirm reduced protein expression (p<0.001 considered significant) .
Specificity controls: Demonstrate no off-target effects on related receptors; for example, KDR expression should remain stable when FLT1 is knocked down and vice versa .
Functional assays: Include tube formation assays, wound healing/migration assays, and proliferation tests to confirm biological effects of the knockdown .
For optimal transfection efficiency in primary human fetoplacental endothelial cells, researchers have successfully used commercially available siRNA targeting FLT1 with a recommended concentration of 20 nM .
FLT1 plays a dominant role in fetoplacental endothelial cell migration and tube formation compared to KDR, contradicting observations in other vascular beds where KDR is typically the primary mediator of angiogenesis. Research using RNA interference demonstrates that:
FLT1 knockdown significantly impairs tube formation as early as 2 hours after plating, with maximum impairment at 12 hours post-plating. Quantitatively, FLT1-ablated cells show significantly decreased branch points (p<0.001) and total tube length (p<0.001) .
KDR knockdown, in contrast, does not significantly affect tube formation at any time point, suggesting a secondary role in fetoplacental angiogenesis .
FLT1 appears to mediate migration-dependent processes rather than proliferation in these cells, as evidenced by wound scratch assays .
This differential regulation suggests that therapeutic approaches targeting angiogenesis in placental disorders should prioritize FLT1-mediated pathways rather than KDR-mediated pathways, contrary to approaches used in cancer angiogenesis.
FLT1 genetic variants show a significant association with longevity, particularly through interactions with hypertension status. The SNP rs3794396 exhibits the most statistically significant association with longevity (p=0.0007) . Research data demonstrate:
Cox model | Genetic model | Hypertension (n=2639) | Normotension (n=945) |
---|---|---|---|
1 | GG vs GC/CC | 0.91 (0.82-1.02), p=0.097 | 1.11 (0.93-1.32), p=0.25 |
2 | GG vs GC/CC | 0.88 (0.78-0.99), p=0.029 | 1.18 (0.96-1.46), p=0.11 |
1 | GC vs GG/CC | 1.15 (1.03-1.28), p=0.013 | 0.87 (0.73-1.04), p=0.13 |
2 | GC vs GG/CC | 1.22 (1.08-1.37), p=0.0014 | 0.81 (0.66-1.01), p=0.059 |
Model 1 is adjusted for age, while Model 2 is adjusted for additional factors including BMI, glucose levels, and lifestyle factors .
The data reveal that in hypertensive individuals, the GG genotype confers a 12% lower mortality risk compared to GC/CC genotypes, while individuals with GG or CC genotypes have a 22% lower mortality risk compared to those with the GC genotype . This protective effect is not observed in normotensive individuals, suggesting that FLT1 genetic variants specifically modulate the mortality risk associated with hypertension.
The human FLT1 regulatory element (enFLT1) is a critical cis-regulatory element (CRE) involved in cardiovascular development. To effectively study this element:
When designing these experiments, researchers should consider evolutionary conservation of the enhancer sequence as an indicator of functional importance and validate findings across multiple model systems.
For studying FLT1 in primary human endothelial cells, the following methodological approach has been validated:
Protein extraction: Use M-PER Mammalian Protein Extraction Reagent (Thermo Fisher) supplemented with phosphatase and protease inhibitors (Cell Signaling Technologies) . Extract supernatant by centrifugation and determine protein concentration via BCA protein assay.
SDS-PAGE resolution: Load 10 μg of whole cell lysate on 4-15% precise gels before transferring to polyvinylidene difluoride (PVDF) membranes .
Antibody selection and dilution:
Signaling studies: For investigating VEGFA-stimulated signaling, starve cells for 1 hour after siRNA transfection, then treat with VEGFA (R&D, 293-VE) at 60ng/ml for varying time points (0, 5, 10, and 60 min) in serum- and growth factor-free medium .
Downstream signaling detection: For pathway analysis, use the following antibodies:
This methodology allows for reliable quantification of both FLT1 protein levels and its signaling effects in primary human endothelial cells.
To effectively distinguish between the functions of full-length FLT1 and soluble FLT1 (sFLT1) in pathological conditions, researchers should implement the following experimental design strategies:
Genetic models: Utilize Flt-1 TK−/− mice, which are defective only in FLT1 signaling while maintaining the VEGF-trapping function of sFLT1. These models have been successfully used to demonstrate slower growth of transplanted cancer cells and reduced metastasis compared to wild-type mice, highlighting the role of FLT1 signaling in tumor progression .
Specific inhibition: Employ RNA interference with sequence-specific siRNAs targeting either:
Functional assays: Apply tube formation, wound scratch, and MTT assays after specific knockdown to assess the individual contributions of each form to angiogenic processes .
Plasma level measurements: In conditions such as preeclampsia, measure plasma sFLT1 levels at different time points to establish temporal relationships between sFLT1 elevation and symptom onset. Studies have shown that patients affected by preeclampsia have increased plasma sFLT1 levels early in pregnancy, before detectable signs of the condition appear .
Complementation experiments: Perform rescue experiments with recombinant proteins or expression constructs of either full-length FLT1 or sFLT1 following knockdown of the endogenous proteins to distinguish their specific functions.
For analyzing interactions between FLT1 genotypes and disease states in longitudinal studies, researchers should follow these methodological approaches:
Genotyping strategy: Select tagging SNPs (tSNPs) that capture most of the genetic variation in FLT1, including 5 kb of the 5' and 3' flanking DNA. Target SNPs with significant associations after correction for multiple testing, such as rs3794396 which has shown strong association with longevity (p=0.0007) .
Statistical models: Test multiple genetic models to identify the most significant associations:
Interaction analysis: Employ Cox proportional hazards models with interaction terms to assess how FLT1 variants modify disease effects on mortality. Include adjustment for relevant covariates such as:
Stratified analysis: Analyze data separately for subjects with and without specific conditions (e.g., hypertension vs. normotension) to identify condition-specific effects of genotypes, as shown in this interaction effect table:
Disease | GG vs. GC/CC | GC vs. GG/CC | ||
---|---|---|---|---|
Beta | p | Beta | p | |
Hypertension | -0.305 | 0.011 | 0.405 | 0.001 |
Diabetes | -0.175 | 0.135 | 0.137 | 0.253 |
Stroke | -0.466 | 0.156 | 0.401 | 0.242 |
CHD | -0.160 | 0.221 | 0.162 | 0.225 |
Survival analysis: Generate Kaplan-Meier survival curves for each genotype stratified by disease state to visually represent differential effects of genotypes on survival .
FLT1 exhibits a fascinating dual role in angiogenesis: a negative role in early embryogenesis and a positive role in pathological conditions like cancer. To reconcile these opposing functions for therapeutic targeting:
Context-specific signaling: Investigate the molecular switches that determine whether FLT1 acts as a positive or negative regulator in different tissues or disease states. This includes identifying specific co-receptors, ligand concentrations, and downstream signaling modulators .
Domain-specific targeting: Design therapeutic agents that specifically target domains responsible for pathological activation while preserving physiological functions. This requires detailed structural and functional mapping of FLT1 domains involved in different signaling cascades .
Temporal targeting strategies: Develop temporally controlled therapeutic interventions that account for the developmental stage-specific roles of FLT1. For example, VEGF-VEGFR inhibitors used in cancer treatment cause hypertension and proteinuria, reminiscent of preeclampsia symptoms, highlighting the importance of understanding temporal aspects of FLT1 function .
Cell-type specific approaches: Distinguish between FLT1 functions in different cell types, as FLT1 signaling in macrophages may have different implications than in endothelial cells. Studies have shown that wild-type mice carrying Flt-1 TK−/− bone marrow exhibit slower growth of transplanted tumors, suggesting an important role for FLT1 in bone marrow-derived cells .
When studying the enFLT1 enhancer in cardiovascular development disorders, researchers should consider these methodological approaches:
Evolutionary conservation analysis: Prioritize highly conserved regions within the enhancer, as this conservation suggests functional importance across species. The human enFLT1 has demonstrated conserved function in driving reporter expression in the cardiovascular system of model organisms .
CRISPR-based enhancer editing: Rather than complete deletion, consider designing precise edits that modify specific transcription factor binding sites within the enhancer to dissect its functional components. This approach can reveal which aspects of the enhancer contribute to different developmental processes .
Single-cell transcriptomics: Apply single-cell RNA sequencing before and after enhancer manipulation to identify cell-type specific effects on gene expression programs that might be masked in bulk tissue analysis .
4D genome analysis: Employ chromosome conformation capture (Hi-C or similar) techniques to understand the three-dimensional interactions between enFLT1 and its target promoters across developmental time points .
Multi-species validation: Test enhancer function across multiple model systems (zebrafish, medaka, mouse) to distinguish conserved functions from species-specific roles, particularly important given the differences in FLT1 gene structure across vertebrates .
Vascular Endothelial Growth Factor Receptor-1 (VEGFR-1), also known as Fms-like tyrosine kinase 1 (FLT1), is a high-affinity tyrosine kinase receptor for Vascular Endothelial Growth Factor (VEGF). VEGFR-1 plays a crucial role in the regulation of angiogenesis, the process by which new blood vessels form from pre-existing vessels. This receptor is involved in various physiological and pathological processes, including embryonic development, wound healing, and tumor growth.
VEGFR-1 is a transmembrane protein that consists of an extracellular ligand-binding domain, a single transmembrane helix, and an intracellular tyrosine kinase domain. The extracellular domain of VEGFR-1 binds to VEGF-A, VEGF-B, and Placental Growth Factor (PGF), mediating the activation of signaling pathways that promote endothelial cell proliferation, migration, and survival .
VEGFR-1 is considered a decoy receptor because it binds VEGF with high affinity but has a weaker tyrosine kinase activity compared to VEGFR-2. This binding limits the availability of VEGF for VEGFR-2, thereby modulating angiogenic signaling. VEGFR-1 also plays a role in macrophage migration through the activation of phospholipase C gamma (PLCγ) and phosphoinositide 3-kinase (PI3K) signaling pathways .
Recombinant human VEGFR-1 is produced using various expression systems, including mammalian cells and bacteria. The recombinant protein typically includes a His tag at the C-terminus to facilitate purification using nickel affinity chromatography. The His tag allows for efficient isolation of the protein from the expression system, ensuring high purity and yield.
The recombinant VEGFR-1 protein encompasses amino acids 27-756, corresponding to the extracellular domain of the receptor. This construct may also include additional tags, such as an Fc domain of human IgG1 or an Avi-tag™, to enhance stability and facilitate detection .
Recombinant VEGFR-1 is widely used in research to study angiogenesis and related signaling pathways. It serves as a valuable tool for investigating the mechanisms of VEGF-mediated endothelial cell functions and for screening potential therapeutic agents targeting VEGF signaling. Additionally, recombinant VEGFR-1 is utilized in various assays, including binding studies, cell migration assays, and in vivo angiogenesis models .