PLGF1 Human, HEK is synthesized using HEK293 cells, ensuring proper glycosylation and folding . Critical steps include:
Transfection: HEK293 cells are transfected with plasmids encoding PLGF1 sequences (e.g., residues 19–149) .
Purification: Proprietary chromatographic techniques (e.g., affinity chromatography using His tags) .
Stability: Stable at 4°C for 2–4 weeks or -20°C long-term with carrier proteins (e.g., 0.1% HSA) .
PLGF1 binds VEGF receptor-1 (VEGFR-1) and neuropilin-1 (NRP1), activating pathways critical for angiogenesis and cell survival:
Angiogenic Signaling: Promotes endothelial cell proliferation and migration via PI3K/Akt and ERK pathways .
Antiapoptotic Effects: Upregulates Bcl-2 and suppresses Bax in rheumatoid arthritis fibroblasts .
Cross-Talk with VEGF: Enhances VEGF activity through heterodimer formation, amplifying angiogenic responses .
Hyperglycemia-Induced Angiogenesis: Hyperglycemia suppresses PI3K/Akt, elevating PLGF1 levels via FOXO1, contributing to diabetic vascular complications .
Preeclampsia: Low PLGF1 levels correlate with impaired placental angiogenesis .
Rheumatoid Arthritis: PLGF1 from HEK293-conditioned media blocks apoptosis in fibroblast-like synoviocytes (FLS), driving joint hyperplasia .
Embryo Implantation: Endometrial PLGF1 enhances epithelial adhesion to fibronectin, facilitating blastocyst attachment .
Cancer: PLGF1 overexpression correlates with tumor growth and metastasis via angiogenesis .
Hematologic Disorders: Modulates PI3K/Akt in sickle cell disease, contributing to inflammation .
Diagnostic Kits: Patent US8741287B2 details PLGF1 immunoassays for disease monitoring .
PIGF, PGF, PLGF-1.
HEK293 cells.
LPAVPPQQWA LSAGNGSSEV EVVPFQEVWG RSYCRALERL VDVVSEYPSE VEHMFSPSCV SLLRCTGCCG DENLHCVPVE TANVTMQLLK IRSGDRPSYV ELTFSQHVRC ECRPLREKMK PERCGDAVPR RHHHHHH.
PLGF1 is the predominant soluble isoform of human Placental Growth Factor and differs structurally from other isoforms in significant ways. Unlike PLGF2, PLGF1 lacks a heparin-binding domain, which is necessary for binding to the neuropilin-1 coreceptor. This structural difference profoundly affects its binding properties and biological activities . In hypoxic conditions, human endothelial cells upregulate both PLGF1 and PLGF2, with a preference for the soluble PLGF1 isoform. This differential expression begins approximately 12 hours after exposure to reduced oxygen conditions . When designing experiments involving multiple PLGF isoforms, it's critical to account for these binding differences, particularly when studying receptor interactions or downstream signaling pathways.
In HEK293 cells under normoxic conditions, PLGF1 expression is primarily regulated through nuclear factor κB (NF-κB) dependent mechanisms . The baseline expression is typically modest compared to hypoxic conditions. When establishing experimental protocols for studying PLGF1, researchers should include appropriate normoxic controls and consider the influence of cell confluence and passage number, as these factors can affect basal PLGF1 expression. Quantitative RT-PCR remains the gold standard for measuring expression levels, with primers designed specifically to distinguish PLGF1 from other isoforms. For protein quantification, ELISA assays calibrated with recombinant human PLGF1 standards provide reliable detection with sensitivity in the picogram range.
When designing experiments to investigate PLGF1 in HEK cell systems, several critical controls should be incorporated:
Time-course controls: Include multiple time points (3, 6, 12, and 24 hours) to capture the temporal dynamics of PLGF1 expression, especially under stimulatory conditions like hypoxia .
Isoform specificity controls: Use primers that can distinguish between PLGF1 and other isoforms to ensure specificity when quantifying expression.
Parallel VEGF-A measurements: VEGF-A serves as an established positive control for hypoxic response, with expression increasing earlier (typically at 3 hours) than PLGF1 .
HIF-1α verification: Western blot analysis to confirm HIF-1α stabilization is essential when studying hypoxia-induced PLGF1 expression .
Non-targeting siRNA controls: When conducting knockdown experiments, include non-targeting siRNA controls to account for transfection-related effects .
To effectively study hypoxia-induced PLGF1 expression in HEK cells, researchers should implement a comprehensive methodological approach:
Hypoxia induction protocol: Expose cells to 1% O₂ in a controlled hypoxic chamber with appropriate CO₂ balance. For reproducible results, ensure consistent cell density (70-80% confluence) at the onset of hypoxia.
Time-dependent analysis: Collect samples at multiple time points (3, 6, 12, and 24 hours) to capture the complete kinetics of PLGF1 upregulation. Research indicates significant PLGF1 mRNA increases beginning at approximately 12 hours of hypoxia exposure, with continued elevation through 24 hours .
Dual RNA/protein analysis: Perform parallel quantitative RT-PCR and ELISA assays to correlate transcriptional changes with protein secretion. ELISA analysis of culture medium after 24 hours of hypoxia typically reveals a 4-11 fold increase in secreted PLGF, depending on the cell system .
HIF stabilization verification: Conduct western blot analysis to confirm HIF-1α and/or HIF-2α stabilization as essential validation of the hypoxic response pathway activation .
Isoform-specific quantification: Design PCR primers that can distinguish between PLGF isoforms to determine their differential regulation under hypoxic conditions .
The epigenetic regulation of PLGF1 under hypoxic conditions involves sophisticated chromatin remodeling mechanisms:
Hypoxia induces robust hyperacetylation of histones H3 and H4 specifically in the second intron region of the PLGF gene, where previously uncharacterized hypoxia responsive elements (HREs) are located . Notably, this epigenetic modification occurs without changes in DNA methylation at the PLGF CpG-island. Chromatin immunoprecipitation (ChIP) analyses reveal that HIF-1α, but not HIF-2α, directly binds to these intronic HREs following hypoxia exposure .
For investigating these mechanisms, researchers should:
Perform ChIP assays using anti-HIF-1α and anti-HIF-2α antibodies targeting the second intron region (containing HREs) and known promoter regions.
Include positive controls such as the canonical VEGF-A promoter region containing active HREs.
Analyze histone acetylation levels using ChIP assays with antibodies against acetylated histones H3 and H4.
Conduct DNA methylation analysis using bisulfite sequencing or methylation-specific PCR to confirm the absence of methylation changes at the PLGF CpG-island.
siRNA-mediated knockdown experiments reveal that HIF-1α, rather than HIF-2α, plays the predominant role in regulating PLGF1 expression under hypoxic conditions . To effectively study this relationship:
Transfect HEK cells with siRNAs specifically targeting HIF-1α, HIF-2α, or non-targeting controls.
Verify knockdown efficiency using western blot analysis for the respective proteins.
Subject transfected cells to hypoxic conditions (1% O₂).
Quantify isoform-specific PLGF mRNA levels using RT-PCR with primers distinguishing between PLGF1 and PLGF2.
The experimental evidence indicates that HIF-1α silencing completely abrogates hypoxia-induced PLGF upregulation, establishing a direct functional link between HIF-1α activity and PLGF expression . This finding highlights the isoform-specific regulation mechanisms that could be targeted in therapeutic approaches.
For precise quantification of PLGF1 secretion from HEK cells, researchers should consider these methodological approaches:
ELISA-based quantification: Commercial ELISA kits designed specifically for human PLGF can detect secreted PLGF1 in culture medium. For optimal results, collect medium from confluent cultures after 24 hours of treatment and concentrate if necessary. Standard curves should be prepared using recombinant human PLGF1 to ensure accurate quantification .
Western blot analysis: When performing western blot analysis for PLGF1, use antibodies that can discriminate between PLGF isoforms. For secreted proteins, TCA precipitation of culture medium may be necessary before SDS-PAGE separation.
Immunofluorescence labeling: For visualization of intracellular PLGF1, fix cells with 4% paraformaldehyde and permeabilize with 0.1% Triton X-100 before antibody incubation. Co-staining with endoplasmic reticulum markers can help visualize the secretory pathway.
Metabolic labeling: For pulse-chase experiments to study secretion kinetics, incorporate 35S-methionine for protein labeling followed by immunoprecipitation with PLGF1-specific antibodies.
Differentiating between endogenous and exogenously expressed PLGF1 in HEK systems requires strategic experimental design:
Epitope tagging: Incorporate small epitope tags (HA, FLAG, or V5) to the N-terminus of exogenous PLGF1 (after the signal peptide) or the C-terminus to enable specific detection without significantly altering function.
Species-specific detection: Express murine PLGF in human HEK cells and use species-specific antibodies to distinguish exogenous from endogenous protein .
Knockdown-rescue experiments: Deplete endogenous PLGF1 using siRNA targeting the 3'UTR, then rescue with an exogenous expression construct lacking the targeted 3'UTR sequence.
Isotope labeling: For mass spectrometry applications, express exogenous PLGF1 in media containing heavy isotope-labeled amino acids (SILAC approach) to distinguish from endogenous protein.
Reporter fusion proteins: Generate PLGF1 fusions with fluorescent proteins (ensuring the fusion does not disrupt trafficking or function) for live-cell imaging studies.
For advanced in vivo imaging applications, PLGF1 produced in HEK systems can be labeled through several sophisticated approaches:
Radioisotope labeling: PLGF1 can be successfully labeled with 89Zr for PET imaging studies. Research demonstrates that 89Zr-labeled anti-PLGF antibodies (such as RO5323441) show time-, dose-, and PLGF-dependent tumor uptake, making this approach feasible for visualizing PLGF in vivo .
Fluorescent labeling: Purified PLGF1 can be conjugated with fluorescent dyes such as Cy5 using N-hydroxysuccinimide ester chemistry. An optimal labeling ratio of approximately 2.7:1 (dye:protein) has been reported to maintain binding activity while providing sufficient signal .
Validation protocols: The specificity of labeled PLGF1 should be validated through competition assays with unlabeled protein and through binding studies with recombinant cells expressing membrane-tagged PLGF receptors .
Pharmacokinetic considerations: When designing in vivo studies, consider that the biodistribution of labeled PLGF1 may vary depending on the dose administered. In xenograft models, differential uptake has been observed between tumors with high versus low PLGF expression .
When investigating tumor microenvironment interactions, the experimental approach for PLGF1 must differ from PLGF2 studies due to their distinct binding properties:
Receptor binding analysis: Since PLGF1 lacks the heparin-binding domain present in PLGF2, it cannot interact with neuropilin-1 coreceptors . Therefore, experimental designs must account for this difference when studying receptor engagement and downstream signaling.
Cell type considerations: PLGF1 shows differential binding to macrophages compared to PLGF2, particularly when forming immune complexes with antibodies. In vitro experiments with RAW264.7 macrophages demonstrate that PLGF1 preferentially forms immune complexes that bind to Fcγ receptors, followed by phagocytosis .
Matrix interaction studies: Unlike PLGF2, PLGF1 does not bind effectively to extracellular matrix components. When designing 3D culture systems or studying tissue penetration, these distinct properties must be considered.
Hypoxia response elements: Both isoforms are regulated by hypoxia through HIF-1α binding to HREs in the second intron of the PLGF gene, but with differential expression patterns favoring PLGF1 .
While hypoxia-induced PLGF1 expression through HIF-1α-dependent mechanisms is well-characterized, other cellular stressors also influence PLGF1 expression through chromatin remodeling:
Oxidative stress response: Oxidative stress activates metal responsive transcription factor 1 (MTF-1), which has been implicated in PLGF upregulation in transformed mouse embryonic fibroblasts . Researchers should evaluate histone modifications at both promoter and intronic regions following oxidative stress induction.
Inflammatory stimuli: NF-κB activation in response to inflammatory signals contributes to PLGF regulation in HEK293 cells . Experimental designs should include ChIP analysis targeting NF-κB binding sites alongside histone modification analysis following treatment with inflammatory cytokines.
Combined stress conditions: To study the interplay between different stress pathways, researchers can design experiments with sequential or simultaneous exposure to hypoxia, inflammatory stimuli, and oxidative stress, analyzing the resulting chromatin modifications and transcription factor binding patterns.
Differential histone modification patterns: While hypoxia induces hyperacetylation of histones H3 and H4 at the second intron of PLGF without affecting DNA methylation , other stressors may induce distinct epigenetic signatures that should be characterized through comprehensive epigenomic profiling.
Cutting-edge technologies are transforming PLGF1 research in several key areas:
CRISPR-Cas9 genome editing: Precise modification of endogenous PLGF regulatory elements, including the newly identified HREs in the second intron, allows for detailed functional analysis of these regulatory regions.
Single-cell transcriptomics: This approach reveals heterogeneity in PLGF1 expression within HEK cell populations and identifies subpopulations with differential response to hypoxia or other stimuli.
Advanced imaging techniques: Super-resolution microscopy and proximity labeling methods (BioID, APEX) are enabling detailed analysis of PLGF1 trafficking and secretion pathways in live cells.
Computational modeling: Integration of epigenomic, transcriptomic, and proteomic data allows for predictive modeling of PLGF1 expression under various conditions, guiding experimental design.
Organoid systems: Three-dimensional HEK organoid models provide more physiologically relevant systems for studying PLGF1 biology compared to traditional monolayer cultures.
Placental Growth Factor-1 (PLGF-1) is a member of the vascular endothelial growth factor (VEGF) family, which plays a crucial role in angiogenesis and vascular development. The human recombinant form of PLGF-1, produced in HEK293 cells, is a valuable tool for studying the molecular mechanisms of angiogenic signaling and exploring its potential therapeutic applications.
PLGF-1 is a homodimeric glycoprotein with a distinctive structural configuration essential for its interactions with VEGF receptors. The human recombinant form allows for controlled exploration of the three-dimensional intricacies of PLGF-1, providing insights into its binding affinities and conformational dynamics. Understanding its structure is fundamental for deciphering how PLGF-1 orchestrates angiogenic responses in different physiological and pathological contexts .
The human recombinant PLGF-1 is produced in HEK293 cells, a human embryonic kidney cell line. This production method ensures that the protein is glycosylated, which is important for its biological activity. The recombinant protein is typically fused to a 6 amino acid His-tag at the C-terminus and purified using proprietary chromatographic techniques .
PLGF-1 primarily functions by inducing angiogenesis through its binding to VEGF receptors on endothelial cells. This interaction triggers a cascade of signaling pathways that promote the formation of new blood vessels. The biological activity of PLGF-1 is measured by its binding ability in a functional ELISA with Human VEGFR1/Flt-1 .
The recombinant form of PLGF-1 is widely used in research to dissect the molecular intricacies of angiogenic signaling. It has significant implications in both physiological vascular development and pathological conditions such as cancer. By studying PLGF-1, scientists aim to deepen their understanding of angiogenesis and develop innovative strategies for vascular-targeted therapies .