Human FLT1 D4 (sVEGFR-1(D4)) is a non-chimeric, monomeric protein containing only the first 4 extracellular domains of the VEGFR-1 receptor. These domains contain all necessary information for VEGF binding. The receptor monomers have a molecular weight of approximately 55 kDa and contain 457 amino acid residues. The full VEGFR-1 receptor normally contains seven immunoglobulin-like extracellular domains, a single transmembrane region, and an intracellular split tyrosine kinase domain, but the D4 variant includes only the first four Ig-like domains . This structure allows FLT1 D4 to maintain high binding affinity for VEGF without signal transduction capability.
FLT1 D4 represents a truncated form of the full VEGFR-1 receptor, containing only domains D1-D4 of the extracellular portion. The full-length human VEGFR1 contains 1312 amino acids forming a mature ~180 kDa glycoprotein with seven immunoglobulin-like domains, a transmembrane domain, and a cytoplasmic domain . While the full-length receptor mediates cellular signaling upon VEGF binding, FLT1 D4 functions primarily as a ligand trap. FLT1 D4 retains the high-affinity binding to VEGF-A (~2-10 picomolar affinity), VEGF-B, and PlGF, but lacks signaling capability due to the absence of transmembrane and intracellular domains . Naturally occurring soluble VEGFR-1 is generated through alternative splicing of the flt-1 mRNA and serves as an endogenous regulator of angiogenesis .
FLT1 D4 binds to several members of the VEGF family with varying affinities:
VEGF-A: Binds with very high affinity (~2-10 picomolar Kd), which is much higher than VEGFR2
VEGF-B: Binds specifically to VEGFR1/FLT1 and not other VEGF receptors
PlGF (Placental Growth Factor): Binds with an affinity of ~170 picomolar (Kd)
This selective binding profile makes FLT1 D4 an important tool for studying differential VEGF signaling pathways and developing targeted therapeutic approaches to modulate specific VEGF-dependent processes.
The biological activity of sVEGFR-1(D4) can be determined by measuring its ability to inhibit VEGF-A-induced proliferation of Human Umbilical Vein Endothelial Cells (HUVECs) . A methodological approach includes:
Culture HUVECs in appropriate growth medium with reduced serum (0.5-2%)
Pre-incubate various concentrations of recombinant FLT1 D4 with a fixed concentration of VEGF-A (10-20 ng/mL) for 30-60 minutes
Add the mixture to HUVEC cultures and incubate for 24-72 hours
Assess proliferation using standard methods (MTT/MTS assay, BrdU incorporation, or cell counting)
Calculate IC50 values by plotting inhibition percentage against FLT1 D4 concentration
Compare results with a reference standard of known activity
Effective FLT1 D4 will show dose-dependent inhibition of VEGF-A-stimulated proliferation, confirming its ability to sequester VEGF and prevent receptor activation.
For optimal experimental results, proper handling of lyophilized FLT1 D4 is essential. Based on standard recombinant protein protocols:
Reconstitution: Dissolve lyophilized sVEGFR-1(D4) in sterile, buffered solution (PBS, pH 7.4) to a concentration of 0.1-1.0 mg/mL
Gentle mixing: Avoid vigorous shaking or vortexing to prevent protein denaturation
Short-term storage: Store reconstituted protein at 4°C for up to 1 week
Long-term storage: Prepare small aliquots and store at -20°C to -80°C
Avoid freeze-thaw cycles: Repeated freezing and thawing can reduce protein activity
Working solutions: Dilute in appropriate buffer containing carrier protein (0.1-0.5% BSA) to prevent adhesion to tubes and loss of activity
Following these guidelines helps maintain the structural integrity and biological activity of FLT1 D4 for experimental applications.
Several cellular models can be employed to study FLT1 D4 function, each offering distinct advantages:
HUVECs (Human Umbilical Vein Endothelial Cells):
Monocytes/Macrophages:
Trophoblast cells:
Muscular dystrophy cell models (e.g., mdx mouse-derived cells):
Reporter cell lines:
Engineered cells with VEGF-responsive elements driving reporter gene expression
Allow quantitative measurement of VEGF sequestration by FLT1 D4
The choice of model should align with specific research questions, considering the native expression patterns of VEGF receptors and the cellular responses being studied.
FLT1 D4 exhibits context-dependent effects on pathological angiogenesis across different disease models:
Duchenne Muscular Dystrophy (DMD):
Endothelial cell-specific Flt1 deletion resulted in increased vascular density in mdx mice (DMD model)
Improved muscle histology and function were observed
FLT1 inhibition using anti-FLT1 peptides or monoclonal antibodies blocking VEGF-FLT1 binding ameliorated the muscular dystrophy phenotype
Suggests that reducing FLT1 activity may be beneficial in DMD by promoting functional angiogenesis
Pre-eclampsia:
Elevated sFlt1 (including D4 domain) is implicated in pre-eclampsia pathogenesis
Adenovirus-mediated sFlt-1 overexpression in mice caused pre-eclampsia-like symptoms
Histological changes included severe endotheliosis and occlusion of capillary lumens
Co-administration of VEGF adenovirus with sFlt-1 adenovirus rescued the phenotype
Suggests that excess sFlt1 contributes to pre-eclampsia through VEGF sequestration
Corneal Neovascularization:
These findings highlight the dual nature of FLT1 D4 in angiogenesis regulation—its inhibition may benefit certain conditions (DMD) while its overexpression contributes to others (pre-eclampsia).
The high-affinity binding between VEGF and FLT1 D4 is determined by specific structural features:
Domain | Amino Acid Residues (Human) | Function in VEGF Binding |
---|---|---|
Ig-like 1 | 27-123 | Initial contact with VEGF |
Ig-like 2 | 129-227 | Primary binding interface; highest contribution to affinity |
Ig-like 3 | 230-327 | Stabilizes binding complex |
Ig-like 4 | 335-421 | Enhances binding stability and specificity |
VEGF-A binds to VEGFR1 with approximately 10-fold higher affinity (~2–10 picomolar Kd) compared to VEGFR2, despite VEGFR2 being the primary signaling receptor . This paradox is explained by:
The Ig-like domain 2 of VEGFR1 forms a hydrophobic binding pocket that accommodates VEGF with high complementarity
Key residues within domains 2-3 create specific hydrogen bonds and salt bridges with VEGF
The orientation of the domains creates an optimal binding configuration
Understanding these structural determinants enables rational design of modified FLT1 D4 variants with altered binding properties for therapeutic applications.
FLT1 D4's potential as a therapeutic agent or target derives from its ability to sequester VEGF family ligands with high affinity. Research-based therapeutic approaches include:
For conditions with excessive VEGF signaling:
Recombinant FLT1 D4 administration as a VEGF trap
Gene therapy approaches to increase local FLT1 D4 production
Small molecules that mimic FLT1 D4 binding pocket structure
For conditions with insufficient angiogenesis:
Disease-specific applications:
Duchenne Muscular Dystrophy: FLT1 inhibition using anti-FLT1 peptides or monoclonal antibodies improved muscle histology and function in mdx mice
Pre-eclampsia: Reducing circulating sFlt-1 levels alleviates symptoms in mouse models
Cancer: Modulation of tumor angiogenesis through targeted FLT1 D4 delivery
Translational development requires careful consideration of tissue specificity, timing of intervention, and potential off-target effects due to the widespread role of VEGF signaling in physiological processes.
Several complementary techniques can be employed to detect and quantify FLT1 D4:
Enzyme-Linked Immunosorbent Assay (ELISA):
Commercial kits available for human sFLT1 detection
Sensitivity typically in the pg/mL range
Can be developed with antibodies specific to domains D1-D4
Western Blotting:
Detect FLT1 D4 protein using antibodies against the extracellular domain
Distinguish from full-length receptor by molecular weight (~55 kDa vs. ~180 kDa)
Useful for semi-quantitative analysis in tissue or cell lysates
Flow Cytometry:
Measure cell-bound FLT1 D4 using fluorescent-labeled antibodies
Assess binding to cell surface receptors or internalization
Surface Plasmon Resonance (SPR):
Real-time measurement of binding kinetics between FLT1 D4 and VEGF variants
Determine association/dissociation constants
Useful for comparing engineered FLT1 D4 variants
Mass Spectrometry:
Identify and quantify FLT1 D4 with high specificity
Distinguish between different splicing variants
Characterize post-translational modifications
These methods can be combined for comprehensive analysis of FLT1 D4 in experimental and clinical samples.
Several animal models have proven valuable for investigating FLT1 D4 functions:
Genetic Models:
Disease-Specific Models:
Molecular Tool-Based Models:
Anti-FLT1 peptide or monoclonal antibody administration
Adenoviral co-expression of VEGF and sFlt-1 for rescue experiments
CRISPR/Cas9-engineered variants of Flt1 with specific domain modifications
When designing in vivo studies, consider:
Temporal control of Flt1 modification (development vs. adult)
Tissue specificity of intervention
Physiological vs. pathological contexts
Relevant endpoints (vascular density, function, disease parameters)
Each model offers distinct advantages for addressing specific research questions related to FLT1 D4 biology.
Investigating FLT1 D4 within the complex VEGF signaling network requires multi-level approaches:
Protein-Protein Interaction Analysis:
Co-immunoprecipitation of FLT1 D4 with VEGF family members
Biolayer interferometry or SPR for kinetic binding parameters
Proximity ligation assays to detect interactions in situ
Competitive binding assays with different VEGF family members
Signaling Pathway Analysis:
Phosphorylation status of downstream effectors (ERK1/2, AKT, p38)
Gene expression profiling following FLT1 D4 modulation
Calcium flux measurement in response to VEGF with/without FLT1 D4
Real-time analysis of signaling using FRET-based biosensors
Functional Cellular Responses:
Endothelial cell proliferation, migration, and tube formation assays
Analysis of tip/stalk cell differentiation in sprouting assays
Integration with other angiogenic pathways (Notch, Angiopoietin)
Effects on pericyte recruitment and vessel maturation
Computational Approaches:
Systems biology modeling of VEGF/VEGFR interactions
Prediction of FLT1 D4 effects on VEGF gradient formation
Network analysis of transcriptional responses to FLT1 D4 modulation
These methods collectively provide a comprehensive understanding of how FLT1 D4 integrates into and modulates the broader VEGF signaling network across different cellular contexts.
Hypoxia is a key regulator of FLT1 expression through multiple mechanisms:
Transcriptional Regulation:
Alternative Splicing:
Hypoxic conditions can alter the splicing patterns of FLT1 mRNA
This may affect the ratio of full-length receptor to soluble forms including FLT1 D4
Splicing factors regulated by hypoxia contribute to this process
Methodological approaches to study hypoxic regulation:
Compare FLT1 D4 expression in normoxic vs. hypoxic conditions (1-5% O₂)
Use chemical mimetics of hypoxia (CoCl₂, DMOG, DFO)
Employ HIF-1α knockout or knockdown models
Analyze HIF-1 binding to the VEGFR1 promoter using ChIP assays
Investigate splicing factor activity under hypoxic conditions
Understanding hypoxic regulation of FLT1 D4 has implications for numerous pathological conditions where hypoxia and aberrant angiogenesis coincide, including cancer, ischemic diseases, and pre-eclampsia.
Research on FLT1 D4 has revealed apparent contradictions that highlight its context-dependent roles:
Opposing effects in different diseases:
In muscular dystrophy (mdx mice), inhibition of FLT1 is beneficial, improving vascular density and muscle function
In pre-eclampsia models, increased sFLT1 (including D4 domain) is detrimental, causing endothelial dysfunction and other symptoms
These contradictory findings suggest tissue-specific and context-dependent roles
Molecular paradoxes:
VEGFR1/FLT1 has higher affinity for VEGF (~2-10 pM) than VEGFR2 (~100-125 pM) but weaker signaling activity
VEGFR1 knockout is embryonically lethal, but tyrosine kinase domain-deficient VEGFR1 mice develop normally
This suggests that the ligand-binding function (present in FLT1 D4) is more critical than signaling activity
Resolving contradictions:
Consider tissue-specific VEGF dependency and receptor expression patterns
Examine temporal aspects of FLT1 D4 function during development vs. adult homeostasis
Investigate concentration-dependent effects (partial vs. complete inhibition)
Analyze compensatory mechanisms that may differ between acute intervention and genetic models
These apparent contradictions reflect the complex biology of FLT1 D4 and highlight the importance of precisely defined experimental conditions and careful interpretation of results across models.
Emerging single-cell technologies offer unprecedented insights into FLT1 D4 biology:
Single-cell RNA sequencing (scRNA-seq):
Reveals heterogeneity in FLT1 expression across endothelial subtypes
Identifies cell populations that predominantly express soluble vs. membrane-bound forms
Maps correlations between FLT1 D4 expression and other angiogenic regulators
Captures dynamic changes during development or disease progression
Single-cell protein analysis:
Mass cytometry (CyTOF) with FLT1 D4-specific antibodies
High-parameter flow cytometry to correlate FLT1 D4 with cellular phenotypes
Imaging mass cytometry for spatial context of FLT1 D4 expression
Spatial transcriptomics:
Maps FLT1 D4 expression in tissue context
Correlates with vascular patterns and disease features
Reveals microenvironmental influences on FLT1 D4 expression
Functional single-cell assays:
Microfluidic platforms to assess individual cell responses to FLT1 D4
Live-cell imaging of VEGF gradient formation with variable FLT1 D4 levels
Single-cell secretion analysis to quantify FLT1 D4 release
Vascular Endothelial Growth Factor Receptor-1 (VEGFR-1), also known as Fms-like tyrosine kinase-1 (FLT-1), is a receptor tyrosine kinase that plays a crucial role in angiogenesis, the process of forming new blood vessels from pre-existing ones. This receptor is part of the VEGF family, which includes several proteins such as VEGF-A, VEGF-B, VEGF-C, VEGF-D, and placental growth factor (PlGF). VEGFR-1 is primarily involved in the regulation of vascular development and permeability.
VEGFR-1 consists of an extracellular domain, a transmembrane domain, and an intracellular tyrosine kinase domain. The extracellular domain is responsible for binding to its ligands, VEGF-A, VEGF-B, and PlGF. The binding of these ligands to VEGFR-1 triggers a cascade of intracellular signaling pathways that regulate various cellular processes, including cell migration, survival, and proliferation.
VEGFR-1 is known for its high affinity for VEGF-A, which is a key mediator of angiogenesis. Interestingly, VEGFR-1 can act as a “decoy” receptor by sequestering VEGF-A and preventing it from binding to VEGFR-2, another receptor that mediates angiogenic signaling. This decoy function helps to fine-tune the angiogenic response and maintain vascular homeostasis .
Recombinant VEGFR-1 D4 refers to a specific domain of the VEGFR-1 protein that has been produced using recombinant DNA technology. This domain includes the extracellular region of the receptor, which is crucial for ligand binding. The recombinant form is often used in research to study the interactions between VEGFR-1 and its ligands, as well as to develop therapeutic agents that target angiogenesis-related diseases.
The production of recombinant VEGFR-1 D4 typically involves cloning the gene encoding the extracellular domain into an expression vector, which is then introduced into a host cell line such as HEK293 cells. The host cells express the recombinant protein, which can be purified using techniques like affinity chromatography .
Recombinant VEGFR-1 D4 is widely used in various research applications, including:
VEGFR-1 is a promising target for anti-angiogenic therapies, which aim to inhibit the formation of new blood vessels in diseases characterized by excessive angiogenesis, such as cancer and diabetic retinopathy. By blocking VEGFR-1 signaling, it is possible to reduce tumor growth and metastasis, as well as to alleviate symptoms of other angiogenesis-related conditions .