FLT1 antibodies are engineered to recognize specific epitopes on the extracellular domain of VEGFR-1. Key structural features include:
Epitope Region (Amino Acid Positions) | Antibody Clone | Species Reactivity | Subclass |
---|---|---|---|
1–338 (N-terminal domain) | KM1732 | Human | Mouse IgG1 |
100–204 | KM1750 | Human | Mouse IgG2b |
Extracellular domain (1–251) | FLT-19 | Human | Mouse IgG1 |
These antibodies inhibit VEGF binding to FLT1 with high specificity, showing no cross-reactivity with VEGFR-2 (KDR) or VEGFR-3 . The 21B3 clone, developed via phage display from immunized llamas, exhibits enhanced affinity for both human and mouse FLT1 orthologs, enabling cross-species therapeutic studies .
FLT1 antibodies exert their effects through:
VEGF Binding Inhibition: Blocks VEGF-A and placental growth factor (PlGF) from activating FLT1, reducing angiogenesis signals .
Biological Activity Suppression: Prevents endothelial cell migration and tube formation (IC₅₀ values: 0.5–10 nM in in vitro assays) .
sFlt-1 Neutralization: Lowers free soluble FLT1 levels, increasing bioavailable VEGF to improve vascular perfusion .
Reduces tumor angiogenesis and metastasis in solid cancers (e.g., breast, lung) by 40–60% in murine models .
Synergizes with chemotherapy to enhance drug delivery via vascular normalization .
Mitigates retinal neovascularization in diabetic retinopathy and retinopathy of prematurity by 70% in preclinical studies .
In mdx mice (Duchenne muscular dystrophy model), 21B3 increased muscle perfusion by 30% and improved grip strength by 25% .
Commercial FLT1 antibodies are widely used in diagnostics and research:
Supplier | Clone | Applications | Conjugate | Price (USD) |
---|---|---|---|---|
Miltenyi Biotec | REAfinity™ | Flow Cytometry | PE-Vio® 615 | $160 |
Cusabio | CSB-RA940669A0HU | WB, IHC, FC | Unconjugated | $210 |
Novus Biologicals | FLT1/1663 | IF, IHC, Flow Cyt | Unconjugated | $490 |
Source: Biocompare, Cusabio, and Sigma-Aldrich catalogs .
FLT1 recombinant antibodies are generated through:
Phage Display: Llama-derived VH/VL chains are cloned into human IgG backbones for high-affinity variants .
Hybridoma Technology: Mouse splenocytes immunized with FLT1 extracellular domains yield clones like KM1732 and FLT-19 .
Mammalian Expression: Antibodies are produced in CHO or HEK293 cells and purified via affinity chromatography (>95% purity) .
Quality control includes:
Binding Affinity: Surface plasmon resonance (SPR) confirms KD values <1 nM .
Functional Assays: Endothelial cell migration inhibition and Western blot validation .
Study Model | Outcome | Citation |
---|---|---|
mdx mice (DMD) | 35% increase in capillary density | |
Tumor xenografts | 55% reduction in metastasis | |
Retinopathy models | 70% decrease in neovascular lesions |
The FLT1 recombinant monoclonal antibody is produced through an in vitro process using synthetic genes. This methodology involves the retrieval of FLT1 antibody genes from B cells of immunoreactive rabbits, followed by their amplification and cloning into suitable phage vectors. These vectors are then introduced into mammalian cell lines, enabling the production of functional antibodies in significant quantities. Subsequently, the FLT1 recombinant monoclonal antibody is purified from the culture supernatant of the transfected cell lines through affinity chromatography. This antibody is ideal for a wide range of applications, including ELISA, Western Blot, Immunohistochemistry, and Flow Cytometry, enabling the precise detection of human and mouse FLT1 proteins.
FLT1, also known as VEGFR-1, functions as a receptor for VEGF and PlGF. Upon binding to its ligands, FLT1 initiates intracellular signaling pathways crucial for angiogenesis, the process of forming new blood vessels. By transducing these signals, FLT1 plays a vital role in regulating vascular development, endothelial cell proliferation, migration, and survival, ultimately influencing processes like wound healing, embryonic development, and pathological conditions such as tumor angiogenesis.
FLT1 is a tyrosine-protein kinase that acts as a cell-surface receptor for VEGFA, VEGFB, and PGF. It plays a crucial role in the development of embryonic vasculature, the regulation of angiogenesis, cell survival, cell migration, macrophage function, chemotaxis, and cancer cell invasion. FLT1 also serves as a positive regulator of postnatal retinal hyaloid vessel regression. It may play an essential role as a negative regulator of embryonic angiogenesis by inhibiting excessive proliferation of endothelial cells. In adulthood, FLT1 can promote endothelial cell proliferation, survival, and angiogenesis. Its function in promoting cell proliferation appears to be cell-type specific. It promotes PGF-mediated proliferation of endothelial cells, proliferation of certain types of cancer cells, but does not promote the proliferation of normal fibroblasts (in vitro). FLT1 possesses a very high affinity for VEGFA and relatively low protein kinase activity; it may function as a negative regulator of VEGFA signaling by limiting the amount of free VEGFA and preventing its binding to KDR. It modulates KDR signaling by forming heterodimers with KDR. Ligand binding leads to the activation of several signaling cascades. Activation of PLCG leads to the production of the cellular signaling molecules diacylglycerol and inositol 1,4,5-trisphosphate and the activation of protein kinase C. FLT1 mediates phosphorylation of PIK3R1, the regulatory subunit of phosphatidylinositol 3-kinase, leading to the activation of phosphatidylinositol kinase and the downstream signaling pathway. It also mediates activation of MAPK1/ERK2, MAPK3/ERK1 and the MAP kinase signaling pathway, as well as the AKT1 signaling pathway. FLT1 phosphorylates SRC and YES1, and may also phosphorylate CBL. It promotes phosphorylation of AKT1 at 'Ser-473'. Additionally, it promotes phosphorylation of PTK2/FAK1, PLCG, and may function as a decoy receptor for VEGFA. FLT1 has a truncated kinase domain; it increases phosphorylation of SRC at 'Tyr-418' by unknown means and promotes tumor cell invasion.
FLT1, also known as VEGFR-1, functions as a receptor for VEGF and PlGF. Upon ligand binding, FLT1 initiates intracellular signaling pathways crucial for angiogenesis. It plays a pivotal role in regulating vascular development, endothelial cell proliferation, migration, and survival, influencing processes such as wound healing, embryonic development, and pathological conditions including tumor angiogenesis . FLT1 can also function as a negative regulator of embryonic angiogenesis by inhibiting excessive proliferation of endothelial cells, while promoting endothelial cell proliferation and survival in adulthood . This dual regulatory role makes FLT1 a particularly valuable research target for understanding vascular biology and developing therapeutic approaches.
FLT1 recombinant monoclonal antibodies are generated through sophisticated in vitro processes using synthetic genes. The production methodology involves:
Retrieval of FLT1 antibody genes from B cells sourced from immunoreactive rabbits
Amplification and cloning of these genes into appropriate phage vectors
Introduction of vectors into mammalian cell lines for antibody expression
Purification of the antibodies from culture supernatant through affinity chromatography
This recombinant approach offers significantly improved consistency compared to traditional hybridoma-derived antibodies, as it eliminates batch-to-batch variation through precise genetic control of antibody production .
FLT1 recombinant monoclonal antibodies demonstrate versatility across multiple research applications. The table below summarizes common applications and recommended dilutions:
For optimal results, researchers should validate antibody performance in their specific experimental system, as dilution requirements may vary based on sample type and detection method .
When designing experiments for optimal FLT1 detection specificity, implement a multi-faceted approach:
First, validate antibody specificity through Western blot analysis comparing target samples with appropriate positive and negative controls. For example, when using anti-FLT1 monoclonal antibodies, you might observe that they detect human VEGFR1/Flt-1 in direct ELISAs and Western blots without cross-reacting with recombinant mouse VEGFR1, human VEGFR2, VEGFR3, or VEGFR4 .
Second, determine optimal antibody concentration through titration experiments. For Western blots, begin testing within the recommended 1:500-1:2000 dilution range, while for flow cytometry, start with approximately 2.5 μg/10^6 cells . Include appropriate controls in each experiment, such as isotype control antibodies (e.g., MAB002) for flow cytometry applications to identify non-specific binding .
Third, consider the influence of sample preparation conditions. Some FLT1 antibodies may only detect the protein under specific conditions—for example, non-reducing conditions are required for certain Western blot applications . This specificity in detection conditions stems from the conformational dependency of epitope recognition.
The experimental approach differs substantially when targeting membrane-bound versus soluble FLT1 variants due to their distinct biological properties and experimental accessibility:
For membrane-bound FLT1, flow cytometry represents an optimal methodology as it preserves cellular integrity. When using this approach with HUVEC cells, researchers should optimize staining protocols using approximately 2.5 μg antibody per 10^6 cells, follow with appropriate fluorophore-conjugated secondary antibodies, and always include matched isotype controls . Confocal microscopy provides complementary spatial information on receptor localization and clustering.
In contrast, soluble FLT1 variants (such as sFlt1-14 found in conditions like preeclampsia) require detection in fluid samples through immunoblotting or ELISA techniques. Western blot analysis of serum samples using specialized FLT1 variant antibodies can detect specific variants at their expected molecular weights—approximately 90 kDa for sFlt1-14 in preeclampsia serum samples . When designing immunoblotting experiments, researchers must pay particular attention to the distinction between reducing and non-reducing conditions, as some epitopes are only accessible under non-reducing conditions .
Additionally, antibody selection must account for FLT1 variant-specific epitopes. For instance, antibodies targeting the Glu706-Leu721 region can specifically detect the sFlt1-14 variant prevalent in primate placental tissues and preeclampsia .
Designing experiments to study FLT1 involvement in angiogenesis requires a multifaceted approach combining cellular, molecular, and functional assays:
Begin with expression profiling through quantitative Western blot and flow cytometry analysis using FLT1 recombinant monoclonal antibodies at validated dilutions (1:500-1:2000 for WB; 1:50-1:200 for FC) . This establishes baseline expression levels across relevant cell types such as HUVECs, which naturally express FLT1 and serve as excellent positive controls .
For functional pathway analysis, implement neutralization assays using FLT1 antibodies capable of blocking ligand-receptor interactions . This approach allows direct assessment of FLT1's contribution to downstream signaling cascades, including PLCG activation, phosphatidylinositol 3-kinase pathway induction, and MAPK/ERK signaling .
To connect molecular events with cellular behaviors, incorporate tube formation assays, migration assays, and proliferation studies coupled with FLT1 modulation through antibody-mediated neutralization or genetic approaches. When monitoring endothelial cells' angiogenic capacity, precisely quantify morphological parameters (branch points, tube length) and correlate these with FLT1 expression levels or activation status as determined by immunostaining or biochemical methods .
For comprehensive understanding, consider including comparative analyses with other VEGF receptors (VEGFR2/KDR) to distinguish their unique and overlapping functions, as FLT1 can modulate KDR signaling by forming heterodimers .
Several technical challenges can arise when using FLT1 recombinant monoclonal antibodies in Western blotting:
The most critical issue involves sample preparation conditions. Many FLT1 antibodies detect the protein only under non-reducing conditions due to epitope conformation dependency . For example, antibody MAB321 specifically detects recombinant human VEGFR1/Flt-1 Fc Chimera under non-reducing conditions only, failing to recognize the denatured protein when reducing agents disrupt disulfide bonds . To address this, researchers should prepare parallel samples under both reducing and non-reducing conditions when first optimizing detection protocols.
Another common challenge is cross-reactivity assessment. While some antibodies show excellent specificity for human FLT1 without cross-reacting with mouse VEGFR1 or human VEGFR2-4 , others may exhibit unexpected cross-reactivity. Researchers should validate specificity using appropriate controls, particularly when studying related receptor tyrosine kinases.
Protein molecular weight determination can be confusing due to FLT1's extensive post-translational modifications and multiple isoforms. Full-length FLT1 appears at approximately 180-185 kDa, while soluble variants like sFlt1-14 appear at approximately 90 kDa . Glycosylation patterns further complicate size estimation, requiring careful comparison with validated positive controls.
Additionally, antibody concentration optimization is essential. Starting with recommended dilutions (1:500-1:2000) and performing titration experiments helps identify the optimal concentration that maximizes specific signal while minimizing background.
To overcome weak or inconsistent FLT1 detection in immunohistochemistry applications, implement these methodological refinements:
Optimize antigen retrieval methods: Since FLT1 epitope accessibility may be compromised during fixation, compare heat-induced epitope retrieval using citrate buffer (pH 6.0) versus EDTA buffer (pH 9.0) to determine which most effectively unmasks the FLT1 epitope without compromising tissue morphology.
Refine antibody concentration: While the recommended dilution range is 1:50-1:200 , systematic titration within this range is essential. Prepare serial dilutions and evaluate both signal intensity and background levels to identify the optimal concentration for your specific tissue type and processing method.
Extend primary antibody incubation: Consider overnight incubation at 4°C rather than shorter incubations at room temperature to enhance antibody penetration and epitope binding, particularly in tissues with lower FLT1 expression.
Implement signal amplification: For tissues with naturally low FLT1 expression, employ tyramide signal amplification or polymer-based detection systems that provide signal enhancement without increasing background.
Validate specificity with appropriate controls: Include both positive controls (HUVEC cells or tissues known to express FLT1, such as placental tissue) and negative controls (isotype-matched irrelevant antibodies) in each experiment to confirm detection specificity .
Consider alternative detection systems: Different chromogens or fluorophores may provide better signal-to-noise ratios depending on tissue autofluorescence and endogenous peroxidase activity levels.
Optimize blocking conditions: Extend blocking time or test alternative blocking agents (normal serum, BSA, commercial blocking solutions) to reduce non-specific binding that may mask specific signals.
Optimizing FLT1 recombinant monoclonal antibody use in flow cytometry requires systematic attention to several critical parameters:
First, establish appropriate antibody concentration through careful titration. While 2.5 μg/10^6 cells represents a recommended starting point , optimal concentration varies by cell type and antibody lot. Perform a titration experiment using serial dilutions (e.g., 5, 2.5, 1.25, 0.6 μg/10^6 cells) to identify the concentration that maximizes positive population separation while minimizing background staining.
Second, implement proper controls: Include an unstained control for autofluorescence assessment, a secondary-only control to evaluate non-specific binding of detection antibodies, and critically, an isotype-matched control antibody (e.g., MAB002 when using mouse IgG-derived anti-FLT1) to distinguish specific binding from Fc receptor interactions.
Third, optimize cell preparation protocols. For endothelial cells like HUVECs, gentle enzymatic dissociation methods preserve surface epitopes better than harsh mechanical dissociation. Cell fixation (if required) should be validated to ensure it doesn't obscure the FLT1 epitope recognized by your specific antibody clone.
Fourth, select appropriate fluorophores based on your cytometer configuration and experimental design. For detecting potentially low-abundance FLT1, bright fluorophores like PE or APC often provide better resolution than FITC .
Fifth, when analyzing results, use fluorescence-minus-one (FMO) controls for accurate gating, particularly in multicolor panels. These controls help distinguish real FLT1 expression from spectral overlap artifacts.
Accurate interpretation of FLT1 expression data across different experimental platforms requires understanding platform-specific considerations and implementing appropriate normalization strategies:
For Western blot analysis, quantitative comparison requires normalization to loading controls (β-actin, GAPDH) and validation using recombinant standards of known concentration. Remember that FLT1 detection may be condition-specific—some antibodies detect the protein only under non-reducing conditions , potentially leading to false-negative results if inappropriate sample preparation methods are used.
In flow cytometry, express FLT1 detection as mean/median fluorescence intensity (MFI) relative to isotype controls . For multi-platform comparison, convert raw MFI values to molecules of equivalent soluble fluorochrome (MESF) using calibration beads. This provides a standardized measurement independent of instrument settings or antibody fluorophores.
For immunohistochemistry, implement standardized scoring systems (H-score, Allred score) that account for both staining intensity and percentage of positive cells. Digital pathology quantification tools can provide more objective measurements across different experimental conditions.
When comparing results across platforms, consider that each method detects FLT1 in different contexts—Western blotting measures total protein regardless of localization, flow cytometry primarily detects surface-expressed receptor, and immunohistochemistry provides spatial information but may detect both membrane-bound and soluble forms.
Additionally, remember that different FLT1 antibody clones may recognize distinct epitopes, potentially detecting different subsets of FLT1 variants . This is particularly important when studying alternative splice variants like sFlt1-14, which requires specialized antibodies targeting unique regions (e.g., Glu706-Leu721) .
Rigorous validation of FLT1 antibody specificity requires a multi-method approach that confirms target recognition while excluding cross-reactivity:
The gold standard validation combines genetic approaches with immunological detection. This involves comparing antibody reactivity in wild-type samples versus those with genetic FLT1 knockdown/knockout or overexpression. In flow cytometry or immunoblotting applications, the antibody signal should correlate directly with FLT1 expression manipulation. For instance, CRISPR-Cas9 mediated FLT1 knockout cells would serve as definitive negative controls.
Cross-reactivity testing is essential, particularly given the structural similarity between FLT1 and other VEGF receptors. Validated FLT1 antibodies should demonstrate specific binding to human FLT1 without recognizing related proteins like VEGFR2, VEGFR3, or VEGFR4 . This specificity testing should extend to orthologs from different species when performing cross-species experiments, as some antibodies might detect human but not mouse VEGFR1 .
Epitope mapping provides mechanistic validation by confirming antibody binding to the expected protein region. For specialized applications detecting specific FLT1 variants, epitope-specific antibodies like those targeting the Glu706-Leu721 region for sFlt1-14 detection require validation using synthetic peptides or recombinant protein fragments.
Orthogonal detection methods offer complementary validation. When the same pattern of FLT1 expression is observed using different antibody clones targeting distinct epitopes, or when antibody-based detection aligns with mRNA expression data from qPCR or RNA-seq, confidence in specificity increases substantially.
Mass spectrometry-based verification represents the highest level of validation, confirming that immunoprecipitated proteins recognized by the antibody contain FLT1 peptide sequences.
FLT1 recombinant monoclonal antibodies provide powerful tools for investigating preeclampsia mechanisms through multiple research approaches:
For detection of soluble FLT1 variants, specialized antibodies targeting variant-specific epitopes (e.g., sFlt1-14) can be employed in Western blot analysis of serum samples from preeclamptic patients . These antibodies detect the characteristic 90 kDa sFlt1-14 variant that appears to be unique to primates and is produced in excess during preeclampsia . When analyzing patient samples, implementing standardized protocols with non-reducing conditions is crucial, as some FLT1 epitopes are only accessible under these conditions .
For placental tissue analysis, immunohistochemistry using FLT1 antibodies at optimized dilutions (1:50-1:200) can visualize the distribution and expression levels of FLT1 variants in degenerative syncytiotrophoblasts within syncytial knots, which are key sites of sFlt1-14 production in preeclampsia . This approach can be combined with markers of placental stress or hypoxia to correlate FLT1 dysregulation with other pathological features.
In mechanistic studies, neutralizing FLT1 antibodies can be employed to block specific FLT1 functions in cell culture models, helping distinguish which aspects of preeclampsia pathophysiology are directly attributable to FLT1 upregulation versus other placental factors . Flow cytometry analysis of circulating endothelial cells using FLT1 antibodies can also provide insights into vascular dysfunction associated with preeclampsia .
For translational research, developing assays that quantify the ratio between different FLT1 variants may provide improved biomarkers for preeclampsia risk assessment. Recombinant monoclonal antibodies offer the consistency and specificity required for such clinical applications .
FLT1 recombinant monoclonal antibodies offer sophisticated approaches to angiogenesis inhibition studies through multiple strategic applications:
As neutralizing agents, select FLT1 antibodies can block the interaction between FLT1 and its ligands (VEGF, PlGF), effectively disrupting downstream signaling pathways critical for angiogenesis . This approach allows researchers to distinguish between FLT1-dependent and FLT1-independent angiogenic processes in various model systems. When implementing this strategy, titration experiments are essential to determine the minimum antibody concentration required for effective neutralization without off-target effects.
For therapeutic development research, FLT1 antibodies serve as valuable tools for target validation studies. By comparing the effects of FLT1 antibody-mediated inhibition with genetic knockdown approaches and small molecule inhibitors, researchers can build comprehensive evidence for FLT1's role in pathological angiogenesis and evaluate the therapeutic potential of targeting this pathway.
In complex angiogenesis models, FLT1 antibodies enable mechanistic dissection of receptor interactions. Since FLT1 can modulate KDR signaling through heterodimer formation , using antibodies that specifically disrupt this interaction helps delineate the relative contributions of different VEGF receptor complexes to angiogenic responses.
For quantitative assessment of intervention effects, combining FLT1 antibody treatments with high-content imaging of endothelial tube formation, 3D sprouting assays, or in vivo angiogenesis models provides multidimensional data on how FLT1 inhibition affects vessel morphology, branching patterns, and functional perfusion beyond simple proliferation metrics.
When utilizing FLT1 recombinant monoclonal antibodies in tumor microenvironment research, researchers must address several complex considerations:
First, cell-type heterogeneity within tumors necessitates multiplexed approaches. While FLT1 is classically associated with endothelial cells, it is also expressed by tumor-associated macrophages, some cancer cells, and other stromal components . For accurate cellular attribution, combine FLT1 antibodies (at validated dilutions of 1:50-1:200 for IHC or flow cytometry) with lineage-specific markers in multiplexed immunofluorescence or multi-parameter flow cytometry.
Second, the functional duality of FLT1 requires careful experimental design. FLT1 can promote proliferation in some cell types while inhibiting it in others . When studying tumors, assess whether FLT1 signaling has pro-tumorigenic or anti-tumorigenic effects by combining antibody detection with functional assays specific to each cell population.
Third, soluble versus membrane-bound FLT1 variants play distinct roles in the tumor microenvironment. Soluble FLT1 can function as a VEGF trap, potentially limiting angiogenesis . To distinguish these effects, employ antibodies specific to membrane-bound forms versus those that detect both membrane and soluble variants, or use specialized antibodies targeting specific soluble variants .
Fourth, spatial relationships are critical in tumor biology. Implement FLT1 antibody-based imaging that preserves spatial context, such as multiplex immunohistochemistry or imaging mass cytometry. These approaches allow visualization of FLT1-expressing cells relative to hypoxic regions, immune infiltrates, and vascular structures, providing insights into how FLT1 distribution influences tumor progression.
Finally, consider the temporal dynamics of FLT1 expression throughout tumor evolution. Serial sampling and analysis using consistent antibody protocols helps track how FLT1 patterns change during tumor progression or in response to therapies.
Researchers can effectively combine FLT1 antibodies with complementary molecular tools to achieve comprehensive signaling pathway analysis through several strategic approaches:
Implement phospho-specific antibody arrays in conjunction with FLT1 recombinant monoclonal antibodies to simultaneously monitor receptor activation and downstream effector phosphorylation. After stimulating cells with VEGF or PlGF, use FLT1 antibodies for immunoprecipitation, followed by phospho-tyrosine blotting to assess receptor activation. In parallel, analyze phosphorylation of known downstream targets in the MAPK/ERK, PI3K/AKT, and PLCγ pathways . This approach creates a temporal map of signaling events initiated by FLT1 activation.
Combine genetic manipulation with antibody-based detection for mechanistic validation. Using CRISPR-Cas9 to introduce specific mutations in FLT1 signaling domains, researchers can then employ domain-specific antibodies to correlate structural alterations with functional outcomes. For example, mutations in the kinase domain versus the ligand-binding domain would differentially affect signaling outputs, which can be monitored using phospho-specific antibodies against downstream targets.
Leverage proximity ligation assays (PLA) with FLT1 antibodies to visualize and quantify protein-protein interactions in situ. This technique requires two primary antibodies (one targeting FLT1 and another targeting a potential interaction partner) and generates fluorescent signals only when the proteins are in close proximity (<40nm). This approach is particularly valuable for investigating FLT1 heterodimerization with KDR or interactions with intracellular signaling adaptors .
Integrate transcriptional reporters with FLT1 antibody-based interventions to connect receptor activation to gene expression outcomes. After modulating FLT1 activity with neutralizing antibodies, analyze activation of transcription factors (e.g., STAT3, NF-κB) and expression of target genes to establish the transcriptional programs regulated by FLT1 signaling in specific cellular contexts.
For systems biology approaches, combine antibody-based proteomics with transcriptomics and metabolomics after FLT1 modulation. This multi-omics strategy provides a comprehensive view of how FLT1 signaling reconfigures cellular physiology beyond canonical pathway activation.