FLT1 (Fms-related tyrosine kinase 1), also known as Vascular Endothelial Growth Factor Receptor 1 (VEGFR1), is a receptor tyrosine kinase critical for angiogenesis, vascular development, and cellular signaling. It binds ligands such as VEGF-A, VEGF-B, and placental growth factor (PlGF), playing dual roles in promoting and inhibiting angiogenesis depending on context . FLT1 exists as a full-length transmembrane receptor and a soluble isoform (sFLT1), both of which are involved in regulating vascular and immune responses . This article synthesizes structural, functional, and clinical research on FLT1, emphasizing its therapeutic relevance in diseases ranging from cancer to sickle cell anemia.
FLT1 binds VEGF with high affinity but exhibits weaker kinase activity compared to VEGFR2 (KDR/Flk-1) .
Soluble FLT1 (sFLT1) acts as a VEGF trap, inhibiting excessive angiogenesis and maintaining vascular homeostasis .
FLT1 is expressed on monocytes/macrophages and regulates their chemotaxis and migration .
Promotes osteoclast differentiation in hematopoietic stem cells .
Essential for embryonic vasculogenesis; knockout models show lethal vascular defects .
Enhancer elements in the FLT1 locus (e.g., enFLT1) are critical for cardiovascular development .
PARP Inhibitor Resistance: FLT1 activation in BRCA1-mutant breast cancer drives resistance via AKT survival pathways and immunosuppression. High FLT1 expression correlates with poor patient survival .
Glioblastoma: FLT1 promotes tumor proliferation, migration, and metastasis via SHH signaling .
Preeclampsia: Overproduction of sFLT1-14 (a soluble isoform) in placental cells disrupts VEGF signaling, contributing to hypertension and organ damage .
Longevity: The FLT1 rs3794396-GG genotype is associated with protection against hypertension-related mortality .
Sickle Cell Disease: FLT1 variants (e.g., rs115695442) modulate fetal hemoglobin (HbF) levels under hypoxic conditions, offering therapeutic potential .
Axitinib: A tyrosine kinase inhibitor that blocks FLT1 signaling, resensitizing PARPi-resistant tumors .
Anti-FLT1 Antibodies: Neutralizing monoclonal antibodies (e.g., MAB6564) suppress VEGF-induced monocyte migration .
Recombinant Form | Source | Size (kDa) | Application | Reference |
---|---|---|---|---|
Soluble FLT1 (baculovirus) | Insect cells | 96 | VEGF inhibition assays | |
His-tagged FLT1 (Ig-like domains 1–3) | E. coli | 43 | Structural studies, ligand binding |
Deletion of the enFLT1 enhancer disrupts extracellular matrix organization and blood vessel morphogenesis in human endothelial cells .
FLT1 expression declines with age in mouse skeletal muscle, though direct links to human aging remain unclear .
MVSYWDTGVL LCALLSCLLL TGSSSGSKLK DPELSLKGTQ HIMQAGQTLH LQCRGEAAHK WSLPEMVSKE SERLSITKSA CGRNGKQFCS TLTLNTAQAN HTGFYSCKYL AVPTSKKKET ESAIYIFISD TGRPFVEMYS EIPEIIHMTE GRELVIPCRV TSPNITVTLK KFPLDTLIPD GKRIIWDSRK GFIISNATYK EIGLLTCEAT VNGHLYKTNY LTHRQTNTII DVQISTPRPV KLLRGHTLVL NCTATTPLNT RVQMTWSYPD EKNKRASVRR RIDQSNSHAN IFYSVLTIDK MQNKDKGLYT CRVRSGPSFK SVNTSVHIYD KAFITVKHRK QQVLETVAGK RSYRLSMKVK AFPSPEVVWL KDGLPATEKS ARYLTRGYSL IIKDVTEEDA GNYTILLSIK QSNVFKNLTA TLIVNVKPQI YEKAVSSFPD PALYPLGSRQ ILTCTAYGIP QPTIKWFWHP CNHNHSEARC DFCSNNEESF ILDADSNMGN RIESITQRMA IIEGKNKMAS TLVVADSRIS GIYICIASNK VGTVGRNISF YITDVPNGFH VNLEKMPTEG EDLKLSCTVN KFLYRDVTWI LLRTVNNRTM HYSISKQKMA ITKEHSITLN LTIMNVSLQD SGTYACRARN VYTGEEILQK KEITIRGEHC NKKAVFSRIS KFKSTRNDCT TQSNVKH
FLT1, also known as VEGFR1, is a tyrosine kinase receptor predominantly expressed in vascular endothelial cells. It functions as a key regulator of blood vessel development (angiogenesis), cell proliferation, migration, differentiation, and cell survival . In humans, FLT1 is most abundantly expressed in the placenta, with placental FLT1 mRNA levels 40-50 times higher than in other tissues . FLT1 binds vascular endothelial growth factor (VEGF) with high affinity and serves as a critical regulator of vascular development.
The protein exists in both membrane-bound and soluble forms (sFLT1), with the latter functioning as an inhibitor of VEGF signaling. Dysregulation of FLT1 expression or function has been implicated in several pathological conditions, including preeclampsia and coronary artery disease .
The human FLT1 gene contains regulatory elements that control its expression in different tissues. Research has identified a highly conserved enhancer within intron 10 of the FLT1 locus termed enFLT1, which directs expression throughout the cardiovascular system . The gene produces multiple isoforms through alternative splicing and polyadenylation.
For FLT1 mRNA, just three isoforms account for more than 94% of all transcripts in the human placenta . These include the full-length membrane-bound form and soluble variants. One soluble isoform, designated sFlt1-14 or sFlt1-e15a, encodes a 733 amino acid (95-135 kDa) protein that is produced primarily by non-endothelial cells, particularly degenerative syncytiotrophoblasts in the placenta . This form appears to be unique to primates and is overproduced during preeclampsia .
FLT1 expression is controlled through multiple mechanisms, including:
Transcriptional regulation: The FLT1 gene contains several regulatory elements that bind transcription factors involved in cardiovascular development. The enhancer element enFLT1 has been shown to be bound by multiple transcription factors, including NKX2-5, which is critical for cardiac development .
Enhancer elements: The human enFLT1 enhancer directs gene expression throughout the developing cardiovascular system. In transgenic medaka fish models, this enhancer drives GFP reporter expression specifically in cardiovascular tissues .
Alternative polyadenylation: Polyadenylation site sequencing (PAS-Seq) has revealed that FLT1 produces alternatively polyadenylated mRNA isoforms, particularly in the placenta. These different polyadenylation patterns contribute to the production of soluble FLT1 variants .
Single nucleotide polymorphisms: Functional SNPs within the FLT1 gene can affect its transcriptional regulation. For example, rs74412485, located in intron 1 of FLT1, enhances its transcriptional level and has been associated with coronary artery disease .
Soluble FLT1 (sFLT1) plays critical roles in both normal physiology and pathological conditions:
Normal function: sFLT1 serves as a natural antagonist to VEGF signaling by binding VEGF with high affinity but not transducing signals, thus regulating angiogenesis during development.
Preeclampsia: sFLT1 is significantly overproduced in preeclampsia, particularly early-onset preeclampsia (symptom onset <34 weeks). These anti-angiogenic factors originate primarily from the placenta and contribute to maternal symptoms . Placental FLT1 mRNA levels strongly correlate with maternal blood pressure, consistent with sFLT1 overproduction being a significant driver of clinical symptoms .
Biomarker potential: Detection of sFLT1 variants such as Flt1-14 in human serum can serve as a biomarker for preeclampsia. Western blot analysis using specific antibodies can detect these variants in serum samples from preeclamptic patients .
Therapeutic target: The three predominant FLT1 mRNA isoforms represent potential targets for therapeutic RNA interference (RNAi) in both early and late-onset preeclampsia .
Based on published research, the following methodology has proven effective for FLT1 knockdown in human endothelial cells:
Cell isolation and culture: Human fetoplacental endothelial cells should be isolated from placentas of full-term, uncomplicated pregnancies after scheduled Cesarean section .
siRNA transfection:
Use validated siRNA sequences targeting FLT1
Include appropriate control siRNAs (non-targeting)
Transfect cells using a lipid-based transfection reagent according to manufacturer's protocols
Optimize transfection conditions to minimize toxicity while maximizing knockdown efficiency
Validation of knockdown efficiency:
Functional assays: Following confirmed knockdown, cells can be subjected to various functional assays:
Statistical analysis: Perform experiments in triplicate using cells from at least three separate subjects. Use one-way ANOVA with Tukey post-hoc testing for statistical analysis .
CRISPR/Cas9-mediated gene editing has been successfully employed to delete the FLT1 enhancer (enFLT1) to study its function. The following methodology is recommended:
Guide RNA design:
Cell model selection:
Validation of enhancer deletion:
Functional characterization:
In vivo validation:
Several methodologies can be employed to reliably measure sFLT1 levels in clinical samples:
Western blotting:
ELISA:
Commercial ELISA kits specifically designed for sFLT1 detection provide quantitative measurement
Ensure the kit can detect the specific isoforms of interest
Follow manufacturer's protocols carefully, particularly regarding sample preparation
Mass spectrometry:
For more detailed analysis of specific sFLT1 variants
Requires specialized equipment but provides high specificity and sensitivity
Useful for distinguishing between closely related isoforms
Polyadenylation site sequencing (PAS-Seq):
Real-time PCR:
Design primers specific to FLT1 variants
Use for relative quantification of FLT1 isoform expression
Particularly useful for tissue samples rather than serum
Genetic variations in FLT1 have been associated with vascular disease risk, particularly coronary artery disease (CAD):
Single nucleotide polymorphisms (SNPs):
rs9319428 in FLT1 has shown a convincing association with CAD in a Japanese population (P=5.98 × 10^-8)
Further fine mapping revealed that another SNP (rs74412485) had an even more profound genetic effect for CAD (P=2.85 × 10^-12)
This SNP is located in intron 1 of FLT1 and enhances the transcriptional level of the gene
Functional consequences of genetic variations:
The functional SNP rs74412485 leads to increased expression of FLT1
This genetically coded increase in FLT1 expression may activate inflammatory cascades that contribute to CAD development
RNA interference experiments have shown that suppression of FLT1 results in decreased expression of inflammatory adhesion molecules
Tissue-specific effects:
Researchers face several challenges when investigating FLT1's role in preeclampsia:
Complexity of FLT1 regulation:
Multiple isoforms with distinct functions
Complex regulatory networks involving enhancers and other cis-regulatory elements
Tissue-specific expression patterns that may vary during pregnancy
Heterogeneity of preeclampsia:
Methodological challenges:
Need for appropriate cellular models that recapitulate placental function
Difficulty in obtaining human placental samples at early stages of disease
Challenges in creating animal models that fully mimic human preeclampsia
Therapeutic targeting:
Developing specific inhibitors for sFLT1 without affecting membrane-bound FLT1 function
Designing RNAi approaches that target specific isoforms
Determining the optimal timing for intervention
Biomarker validation:
Establishing reliable cutoff values for sFLT1 levels in different populations
Integrating sFLT1 measurements with other biomarkers for improved prediction
Standardizing measurement techniques across clinical laboratories
FLT1 signaling shows distinct patterns between normal and pathological states:
Normal signaling:
FLT1 functions as a regulator of blood vessel development and angiogenesis
It maintains balance between pro-angiogenic and anti-angiogenic factors
Soluble FLT1 serves as a natural VEGF antagonist to fine-tune angiogenic responses
FLT1 regulates extracellular matrix organization and blood vessel morphogenesis
Pathological signaling in preeclampsia:
Excessive production of sFLT1 disrupts angiogenic balance
Three predominant FLT1 mRNA isoforms account for >94% of all transcripts and are upregulated
Increased transcription of the entire FLT1 locus occurs in both early-onset and late-onset preeclampsia
sFLT1 acts as a "VEGF sponge," preventing normal VEGF signaling
Pathological signaling in coronary artery disease:
Regulatory networks:
To effectively study FLT1 enhancer activity across different tissue contexts, researchers should consider these methodological approaches:
Reporter gene assays in transgenic models:
Clone the human enhancer sequence (enFLT1) into reporter vectors (e.g., modified ZED vector)
Generate stable transgenic lines in model organisms like medaka fish
Characterize reporter gene expression patterns throughout development
This approach has successfully demonstrated that human enFLT1 drives GFP expression throughout the cardiovascular system in medaka
In vitro enhancer activity assays:
Transfect enhancer-reporter constructs into relevant cell types
Use minimal promoters (e.g., SCP1) to isolate enhancer effects
Measure reporter gene expression under different conditions
Mutate specific transcription factor binding sites to identify crucial regulatory interactions
CRISPR/Cas9-mediated enhancer deletion:
Generate enhancer knockout cell lines using CRISPR/Cas9
Differentiate cells into relevant lineages (e.g., endothelial cells)
Compare gene expression profiles between wildtype and enhancer-deleted cells
This approach revealed that deletion of enFLT1 resulted in 211 differentially expressed genes compared to wildtype cells
Chromatin immunoprecipitation (ChIP):
Chromosome conformation capture techniques:
Investigate long-range interactions between the enhancer and promoter
Determine how these interactions may differ across tissue types
Identify other genomic regions that may interact with the FLT1 enhancer
When studying FLT1 using gene silencing or editing approaches, researchers should implement these strategies to address off-target effects:
For RNA interference experiments:
Use multiple siRNA sequences targeting different regions of FLT1
Include appropriate negative controls (non-targeting siRNA)
Verify specificity by confirming that expression of related receptors (e.g., KDR/VEGFR2) remains stable
Validate knockdown at both mRNA and protein levels
Published protocols have successfully demonstrated specific FLT1 knockdown without affecting KDR expression
For CRISPR/Cas9 gene editing:
Use prediction tools to select guide RNAs with minimal off-target potential
Sequence multiple clones to confirm specific targeting
Perform whole-genome sequencing to identify potential off-target modifications
Include appropriate controls, such as targeting different regions of the gene
Create rescue experiments by reintroducing the deleted sequence
Validation of phenotypes:
Confirm that observed phenotypes can be rescued by reintroduction of FLT1
Compare phenotypes between different knockdown/knockout strategies
Use multiple experimental approaches to validate findings
In studies of FLT1 enhancer deletion, comparing results with gene deletion helps distinguish enhancer-specific effects from general loss of function
Cross-validation in different models:
For optimal detection of low-abundance FLT1 variants, researchers should consider these methodological strategies:
Sample preparation optimization:
Enrich for the cellular compartment where the variant is expected (membrane, cytosol, etc.)
Use appropriate extraction buffers to maximize protein recovery
Consider sample concentration techniques for low-abundance variants
Western blot optimization:
Use high-sensitivity detection systems (e.g., chemiluminescent substrates)
Optimize antibody concentrations and incubation conditions
Consider non-reducing conditions for certain FLT1 variants
Use appropriate blocking reagents to minimize background
For FLT1-14 detection, non-reducing conditions and specific immunoblot buffer groups have been recommended
PCR-based approaches:
Design primers specific to unique regions of FLT1 variants
Use nested PCR approaches for low-abundance transcripts
Consider digital PCR for absolute quantification of rare variants
Optimize annealing temperatures and cycle numbers
Next-generation sequencing:
Single-cell approaches:
Consider single-cell RNA-Seq to identify cell populations expressing specific variants
Use in situ hybridization techniques to localize variant expression in tissues
Employ immunofluorescence with variant-specific antibodies for protein localization
Several therapeutic strategies targeting FLT1 show promise for treating preeclampsia and cardiovascular diseases:
RNA interference approaches:
Enhancer-targeted therapies:
Modulating the activity of the FLT1 enhancer (enFLT1) could provide a more nuanced approach than targeting the gene itself
Enhancer-targeting compounds could fine-tune FLT1 expression without completely abolishing it
The 211 differentially expressed genes identified after enhancer deletion provide additional potential targets
Anti-inflammatory approaches for CAD:
Since increased FLT1 expression due to genetic variants activates inflammatory cascades in CAD
Anti-inflammatory therapies targeting these pathways could benefit patients with FLT1-associated CAD risk
RNA interference experiments have shown that suppression of FLT1 decreases expression of inflammatory adhesion molecules
sFLT1 neutralization:
Developing antibodies or decoy receptors that specifically bind sFLT1 without affecting membrane-bound FLT1
This approach could restore angiogenic balance in preeclampsia
Careful timing would be crucial to avoid interfering with normal placental development
Personalized medicine approaches:
Genetic screening for FLT1 variants (e.g., rs74412485) could identify individuals at higher risk for CAD
These patients might benefit from earlier or more aggressive preventive interventions
Monitoring sFLT1 levels during pregnancy could help identify women at risk for preeclampsia before symptom onset
Several promising research frontiers are emerging in FLT1 biology:
Comprehensive enhancer characterization:
Further investigation of the FLT1 enhancer network beyond enFLT1
Understanding tissue-specific enhancer activities and how they are integrated
Exploring the three-dimensional chromatin structure around the FLT1 locus
Single-cell transcriptomics:
Characterizing FLT1 isoform expression at single-cell resolution in the placenta and other tissues
Identifying specific cell populations responsible for sFLT1 production in normal and pathological conditions
Mapping the cellular heterogeneity of FLT1 expression throughout development
Systems biology approaches:
Integrating genomics, transcriptomics, and proteomics data to build comprehensive models of FLT1 regulation
Analyzing the 211 differentially expressed genes affected by enhancer deletion to identify key regulatory networks
Developing predictive models for how genetic variants affect FLT1 expression and function
Evolutionary perspectives:
Precision medicine applications:
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 the regulation of angiogenesis and vasculogenesis. It is primarily expressed on endothelial cells and is involved in various physiological and pathological processes, including tumor growth, wound healing, and cardiovascular diseases .
VEGFR-1 is a member of the VEGF receptor family, which includes VEGFR-2 and VEGFR-3. It is composed of an extracellular ligand-binding domain, a single transmembrane region, and an intracellular tyrosine kinase domain. VEGFR-1 can exist in both membrane-bound and soluble forms due to alternative splicing events .
The receptor is widely expressed in normal and pathological tissues, including endothelial cells lining blood vessels, stromal cells, and various human cancer cells. Immunohistochemical studies have shown frequent VEGFR-1 expression in multiple malignancies, such as breast, lung, prostate, pancreas, ovarian, and colon cancers .
VEGFR-1 interacts with several ligands, including Vascular Endothelial Growth Factor-A (VEGF-A), VEGF-B, and Placental Growth Factor (PlGF). While VEGF-A can bind to both VEGFR-1 and VEGFR-2, VEGFR-1 is the only known receptor for VEGF-B and PlGF . The binding of these ligands to VEGFR-1 triggers a cascade of downstream signaling pathways that regulate endothelial cell proliferation, migration, and survival.
VEGFR-1 plays a dual role in angiogenesis. It acts as a decoy receptor by sequestering VEGF-A, thereby preventing its interaction with VEGFR-2, which is a more potent mediator of angiogenic signaling. This decoy function helps to fine-tune the angiogenic response and prevent excessive blood vessel formation .
In addition to its role as a decoy receptor, VEGFR-1 also promotes angiogenesis by activating endothelial cells and supporting cell types, such as stromal and hematopoietic cells. This activation is crucial for the formation of new blood vessels during embryonic development, wound healing, and tumor growth .
Recombinant human VEGFR-1 (rhVEGFR-1) is a biotechnologically engineered form of the receptor that is used in research and therapeutic applications. It is typically produced in Chinese hamster ovary (CHO) cells and purified using protein-A affinity chromatography . The recombinant protein retains the ability to bind VEGF-A and PlGF with high affinity, making it a valuable tool for studying VEGF signaling and developing anti-angiogenic therapies.
The therapeutic potential of targeting VEGFR-1 has been explored in various preclinical and clinical studies. For instance, IMC-18F1, a fully human IgG1 antibody that binds to VEGFR-1, has shown promise in inhibiting cancer growth in multiple in vitro and human tumor xenograft models . Recombinant VEGFR-1 has also been demonstrated to inhibit angiogenesis in vivo, making it a potential candidate for anti-angiogenic therapies in cancer and other diseases characterized by abnormal blood vessel formation .