VEGF Mouse, His

Vascular Endothelial Growth Factor Mouse Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Production and Purification

Mouse VEGF is commonly produced in heterologous systems:

Production SystemKey FeaturesSource
E. coliHigh yield, cost-effective. Used for VEGF-120 (14.2 kDa monomer / 28.4 kDa dimer) .Irvine Scientific
Yeast (Saccharomyces cerevisiae)Proper disulfide bonding. Produces VEGF-165 (40 kDa homodimer) .Prospec Bio
Baculovirus/Sf21 cellsAuthentic post-translational modifications. Used for VEGF-164 (Ala27-Arg190) .R&D Systems

Purification Methods

  • Lyophilization: Commonly lyophilized from PBS or TFA-containing solutions .

  • Chromatography: Proprietary techniques for yeast-derived VEGF .

Angiogenesis and Tumor Studies

Mouse VEGF is pivotal in tumor angiogenesis models. For example:

  • Humanized VEGF-A Mice: Engineered to express humanized VEGF (hum-X VEGF), enabling cross-reactivity with anti-VEGF antibodies (e.g., bevacizumab) .

  • VEGF Trap: Binds both mouse and human VEGF, used to study tumor-derived vs. host-derived VEGF contributions .

Pharmacological and Toxicological Evaluations

  • Anti-VEGF Antibodies: Higher-affinity antibodies (e.g., Y0317, G6–31) show faster clearance in hum-X VEGF mice but similar efficacy in tumor suppression .

  • Glomerulosclerosis Risk: Prolonged anti-VEGF therapy correlates with kidney damage .

Metabolic and Inflammatory Research

  • Adipose Tissue: VEGF-C overexpression in subcutaneous adipose tissue (SWAT) induces obesity, insulin resistance, and inflammation .

Table 1: Mouse VEGF Isoforms and Properties

IsoformAmino AcidsHeparin BindingMolecular Mass (kDa)Key Applications
VEGF-120121No14.2 (monomer) / 28.4 (dimer)Skeletal development
VEGF-164164Yes40.0 (homodimer) Tumor angiogenesis
VEGF-188188Strong22.5 (monomer) / 45.0 (dimer)ECM interaction

Table 2: Production Systems and Formulations

SystemFormulationReconstitution
E. coliLyophilized with 0.1% TFA Sterile water (0.1 mg/mL)
YeastLyophilized in PBS (pH 7.4) 100 µg/mL in sterile H₂O
Baculovirus/Sf21 cellsLyophilized with BSA (carrier) PBS with 0.1% BSA

Key Research Findings

  1. Species-Specific Interactions: Humanized VEGF-A mice allow cross-species antibody testing, revealing no consistent correlation between antibody affinity and in vivo efficacy .

  2. Tumor Microenvironment: VEGF-Trap studies show tumor-derived VEGF contributes minimally to total circulating VEGF in mouse models .

  3. Metabolic Impact: VEGF-C overexpression in SWAT promotes obesity and insulin resistance via macrophage infiltration .

Product Specs

Introduction
Vascular endothelial growth factor (VEGF) is a crucial signaling protein involved in the formation of new blood vessels (vasculogenesis and angiogenesis). While primarily known for its effects on vascular endothelial cells, VEGF also influences other cell types such as immune cells, neurons, cancer cells, and kidney epithelial cells. VEGF promotes blood vessel formation, growth, and permeability, stimulates cell migration, and inhibits cell death. In laboratory settings, VEGF has been observed to enhance the growth and movement of endothelial cells. VEGF also acts as a vasodilator, widening blood vessels, and was initially known as vascular permeability factor due to its ability to increase blood vessel permeability. Elevated levels of VEGF are associated with POEMS syndrome, also called Crow-Fukase syndrome. Genetic mutations in the VEGF gene have been linked to diabetic retinopathy, a condition affecting the eyes.
Description
Recombinant Mouse VEGF, produced in E. coli bacteria, is a single, non-glycosylated polypeptide chain comprising 141 amino acids (corresponding to amino acids 205-324 of the full-length protein). It has a molecular weight of 16.3 kDa. This Mouse VEGF variant is fused to a 20 amino acid Histidine (His) tag at its N-terminus to facilitate purification, which is carried out using specialized chromatographic methods.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
Mouse VEGF is supplied in a solution containing 20mM Tris-HCl buffer at pH 8.0 and 10% glycerol.
Stability
For optimal storage, refrigerate at 4°C if the entire vial will be used within 2-4 weeks. For longer-term storage, freeze at -20°C. To enhance stability during long-term storage, consider adding a carrier protein such as 0.1% HSA or BSA. Repeated freezing and thawing should be avoided.
Purity
Purity exceeds 90.0% as determined by SDS-PAGE analysis.
Biological Activity
Biological activity is assessed through a cell proliferation assay utilizing NIH-3T3 mouse embryonic fibroblasts. The ED50, which represents the concentration at which 50% of the maximum effect is observed, is within the range of 0.5-1.5 ng/ml.
Synonyms
Vascular endothelial growth factor A, VEGF-A, Vascular permeability factor, VPF, VEGF, Vegf120, Vegf164, Vegf188, Vegfa.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MAPTTEGEQK SHEVIKFMDV YQRSYCRPIE TLVDIFQEYP DEIEYIFKPS CVPLMRCAGC CNDEALECVP TSESNITMQI MRIKPHQSQH IGEMSFLQHS RCECRPKKDR TKPEKCDKPR R.

Q&A

What is mouse VEGF-A and why is it important in research?

Mouse Vascular Endothelial Growth Factor A (VEGF-A) is a signaling protein critical for both vasculogenesis (de novo formation of blood vessels) and angiogenesis (formation of new blood vessels from pre-existing ones). It serves as an important mediator of physiological and pathological angiogenesis by stimulating endothelial cell mitogenesis, promoting cell migration, and inhibiting apoptosis . VEGF-A is essential during embryonic development, with even heterozygous VEGF-A knockout mice showing embryonic lethality due to multiple defects in vascular structure formation .

In research contexts, mouse VEGF-A is widely used to study:

  • Mechanisms of blood vessel formation and remodeling

  • Tumor angiogenesis and potential cancer therapies

  • Wound healing processes

  • Ischemic diseases including myocardial infarction

  • Vascular permeability regulation

  • Embryonic development of the vascular system

Mouse VEGF-A studies provide valuable translational insights applicable to human research due to the high conservation of VEGF pathways across species.

What is the structure and biological activity of recombinant His-tagged mouse VEGF-A?

Recombinant His-tagged mouse VEGF-A is a protein engineered to contain a histidine tag (typically 6-20 histidine residues) at either the N-terminus or C-terminus of the native mouse VEGF-A sequence. According to the available product information, a typical His-tagged mouse VEGF-A has the following characteristics:

  • Molecular weight: Approximately 16.3 kDa

  • Structure: Single, non-glycosylated polypeptide chain containing 141 amino acids (positions 205-324 of the full sequence)

  • Expression system: Commonly produced in E. coli

  • Purity: >90% by SDS-PAGE

  • Biological activity: Measured in cell proliferation assays using HUVEC (Human Umbilical Vein Endothelial Cells), with an ED50 typically ≤15 ng/ml

The His-tag facilitates efficient protein purification through metal affinity chromatography while generally maintaining the biological function of the native protein. In vitro studies confirm that properly folded His-tagged mouse VEGF-A retains its ability to bind VEGF receptors and stimulate endothelial cell proliferation .

How does mouse VEGF-A differ from human VEGF-A, and what implications does this have for research?

Mouse and human VEGF-A share approximately 88% amino acid sequence homology but exhibit several important differences that researchers must consider:

  • Isoform distribution: While both species express multiple VEGF-A isoforms through alternative splicing (designated by their amino acid lengths), the predominant isoforms and their relative expression levels differ between mice (VEGF120, VEGF164, VEGF188) and humans (VEGF121, VEGF165, VEGF189) .

  • Receptor binding specificity: Though both mouse and human VEGF-A bind to the same receptors (VEGFR-1 and VEGFR-2), subtle differences in binding affinity exist that may affect downstream signaling intensity.

  • Species cross-reactivity: Mouse VEGF-A binds effectively to mouse receptors, while human VEGF-A has lower affinity for mouse receptors. This species-specificity is important when designing experiments using recombinant proteins .

  • Antibody recognition: Many anti-human VEGF-A antibodies do not recognize mouse VEGF-A, which has led to the development of humanized VEGF-A mouse models for testing human-specific therapeutics .

These differences have significant research implications:

  • When testing anti-human VEGF antibodies in mouse models, researchers may need to use humanized VEGF-A mice

  • For xenograft tumor studies, the contribution of both tumor-derived (possibly human) and host-derived (mouse) VEGF must be considered

  • Species-specific reagents (antibodies, ELISA kits) must be selected appropriately

What are the optimal storage and handling conditions for His-tagged mouse VEGF-A?

Proper storage and handling are crucial for maintaining the stability and biological activity of His-tagged mouse VEGF-A. Based on manufacturer recommendations:

Storage conditions:

  • Store undiluted protein at 2-8°C for up to two weeks for active research use

  • For long-term storage, aliquot and store at -20°C or preferably -70°C

  • Avoid repeated freeze-thaw cycles as they can significantly reduce biological activity

Buffer composition:

  • Typically supplied in 20 mM Tris-HCl buffer (pH 8.0) containing 10% glycerol

  • The glycerol functions as a cryoprotectant during freezing

Handling recommendations:

  • Thaw frozen aliquots rapidly at 37°C and place immediately on ice

  • Dilute only the amount needed for immediate use

  • Use sterile technique when handling the protein

  • Consider adding carrier protein (BSA) to dilute solutions to prevent adsorption to tubes

  • For cell culture applications, filter-sterilize through a 0.22 μm filter

Stability:

  • Shelf life is typically one year from date of manufacture when stored properly

  • Monitor activity if used beyond the recommended shelf life

How can researchers effectively design experiments comparing the activities of VEGF-A variants and isoforms in mouse models?

Designing robust experiments to compare VEGF-A variants requires careful consideration of multiple factors:

Experimental design considerations:

  • Selection of appropriate mouse strains:

    • Wild-type mice for baseline studies

    • Genetically modified mice (knockout, knock-in, or conditional expression models)

    • Humanized VEGF-A mice for testing human-specific therapies

  • VEGF-A isoform selection:

    • Use purified recombinant proteins of specific isoforms

    • Consider testing multiple isoforms (VEGF120, VEGF164, VEGF188) individually and in combination

    • Include appropriate controls for each isoform

  • Dose determination and administration route:

    • Perform dose-response studies (typical range: 1-100 ng/ml in vitro; 1-10 μg/kg in vivo)

    • Consider systemic (intravenous, intraperitoneal) versus local (subcutaneous, intramuscular) delivery

    • Use osmotic pumps for continuous delivery or bolus injections for pulsatile exposure

  • Assessment parameters:

    • Vascular response metrics (vessel density, permeability, diameter)

    • Cellular responses (proliferation, migration, survival)

    • Molecular readouts (receptor phosphorylation, downstream signaling)

    • Functional outcomes (blood flow, tissue oxygenation)

Methodological approach:

When comparing VEGF-A variants in angiogenesis models:

For example, when studying the migration of VEGFR-positive cells in response to different VEGF-A variants, researchers have successfully employed in vitro migration assays using soft agar gels containing varying concentrations (0-100 ng/ml) of VEGF variants placed at defined positions, with sorted VEGFR-positive cells applied centrally and migration quantified over time .

What methodological approaches are most effective for studying VEGF-A/VEGFR signaling dynamics using His-tagged mouse proteins?

Investigating VEGF-A/VEGFR signaling dynamics requires sophisticated methodological approaches. The following strategies have proven effective:

Cell-based signaling studies:

  • Receptor binding assays:

    • Radiolabeled or fluorescently-labeled His-tagged VEGF-A

    • Surface plasmon resonance (SPR) for real-time binding kinetics

    • Competition binding assays with unlabeled variants

  • Receptor activation analysis:

    • Phosphorylation-specific antibodies for Western blotting

    • Phospho-flow cytometry for single-cell resolution

    • FRET-based biosensors for real-time activation dynamics

    • Immunoprecipitation of receptor complexes

  • Downstream signaling pathway analysis:

    • Protein arrays for comprehensive pathway mapping

    • Reporter gene assays for transcriptional responses

    • Live-cell imaging with fluorescent pathway sensors

Methodological considerations:

For robust signaling studies using His-tagged mouse VEGF-A:

  • Verify that the His-tag does not interfere with receptor binding or activation

  • Include positive controls (non-tagged VEGF-A) to confirm comparable activity

  • Use serum-free conditions to eliminate confounding factors

  • Time-course experiments to capture both immediate and delayed responses

  • Dose-response studies to identify threshold and saturation concentrations

  • Consider receptor heterodimerization effects (VEGFR1/VEGFR2)

A combination of approaches is recommended. For example, researchers have successfully used His-tagged VEGF-A to study migration of VEGFR-positive cells isolated from mouse embryos at E7.5, revealing directional migration toward VEGF-A sources that could be blocked by specific inhibitors (SU5416) or neutralizing antibodies .

How can researchers resolve contradictory data when comparing in vitro versus in vivo effects of His-tagged mouse VEGF-A?

Discrepancies between in vitro and in vivo results with His-tagged mouse VEGF-A are common and resolving these contradictions requires systematic investigation:

Common sources of contradiction:

  • Microenvironment complexity:

    • In vivo systems contain multiple cell types and extracellular matrix components

    • Presence of endogenous VEGF and other growth factors in vivo

    • Differential receptor expression across tissues

  • Pharmacokinetic/pharmacodynamic factors:

    • Protein half-life differences between in vitro and in vivo settings

    • Unexpected binding to plasma proteins altering bioavailability

    • Different concentration-time relationships

  • Technical considerations:

    • His-tag interference with protein function in specific contexts

    • Buffer components affecting protein activity

    • Differences in recombinant protein folding and post-translational modifications

Resolution strategies:

Contradiction TypeInvestigation ApproachAnalysis Method
Potency differencesParallel dose-response studiesEC50/IC50 comparison across systems
Binding affinity discrepanciesIn vitro binding followed by in vivo validationScatchard analysis, competition assays
Signaling pathway divergencePathway inhibitor studiesWestern blot, phospho-flow cytometry
Functional outcome disparitiesEndpoint validation across modelsHistology, functional assays
  • Developing humanized VEGF-A mouse models

  • Systematically testing antibodies with different affinities

  • Evaluating both short-term and long-term effects

  • Assessing both efficacy (tumor growth inhibition) and safety (glomerulosclerosis) endpoints

This approach revealed that higher-affinity antibodies were more likely to cause glomerulosclerosis during long-term treatment despite not showing clear superiority in efficacy .

What are the molecular mechanisms underlying VEGF-A heterodimerization, and how can researchers account for this in experimental design?

VEGF-A heterodimerization represents an important regulatory mechanism that adds complexity to VEGF signaling. Understanding and accounting for this phenomenon is crucial for accurate experimental interpretation:

Molecular basis of heterodimerization:

VEGF-A can form heterodimers with other VEGF family members, particularly VEGF-B . This occurs through:

  • Intermolecular disulfide bond formation between conserved cysteine residues

  • Co-expression of multiple VEGF family members in the same cell

  • Post-translational assembly in the endoplasmic reticulum

Functional implications:

VEGF-A/VEGF-B heterodimers exhibit distinct properties compared to homodimers:

  • Modified receptor binding profiles and affinities

  • Altered signaling pathway activation

  • Different biological activities and potencies

  • Potentially important for fine-tuning angiogenic responses

Experimental strategies to account for heterodimerization:

  • Heterodimer detection:

    • Co-immunoprecipitation with isoform-specific antibodies

    • Sandwich ELISA using antibodies against different VEGF family members

    • Mass spectrometry for precise molecular characterization

  • Comparative activity assessment:

    • Side-by-side testing of homodimers versus heterodimers

    • Receptor binding competition assays

    • Downstream signaling response profiles

  • Genetic approaches:

    • Single and double knockout models

    • Knockin models expressing only specific dimers

    • Conditional expression systems

When designing experiments, researchers should be aware that "part of the mitogenic activity reported may be due to VEGF-B/VEGF-A heterodimers" . This underscores the importance of using purified, well-characterized proteins and appropriate controls. In migration assays, researchers have observed that at low concentrations (10 ng/ml) of VEGF-A, the effects of VEGF-A and PlGF (another VEGF family member) were partially additive (30% increase), while at higher doses (40-100 ng/ml), additive effects were hardly detected .

What are the most reliable methods for quantifying the biological activity of His-tagged mouse VEGF-A in different experimental systems?

Accurately quantifying the biological activity of His-tagged mouse VEGF-A is essential for consistent and reproducible research. Different experimental contexts require specific approaches:

In vitro activity assays:

  • Endothelial cell proliferation:

    • HUVEC proliferation assay (gold standard for potency)

    • MTT/XTT colorimetric assays

    • BrdU incorporation

    • Standardized ED50 determination (typically ≤15 ng/ml)

  • Migration assays:

    • Transwell/Boyden chamber assays

    • Scratch wound healing assays

    • Time-lapse microscopy tracking

    • Soft agar gel migration assays with defined VEGF gradients

  • Tube formation:

    • Matrigel tube formation assay

    • 3D fibrin gel angiogenesis assay

    • Spheroid sprouting assays

Ex vivo systems:

  • Aortic ring assay:

    • Quantification of vessel sprouting from aortic explants

    • Visualization of branching complexity

    • Analysis of sprout length and number

  • Embryoid body differentiation:

    • Assessment of vascular differentiation from stem cells

    • Quantification of VEGFR-positive cell migration

In vivo quantification:

  • Directed angiogenesis assays:

    • Corneal pocket assay

    • Matrigel plug assay

    • Directed cell migration in embryo models

  • Functional readouts:

    • Laser Doppler blood flow measurement

    • Contrast-enhanced ultrasound

    • Oxygen tension measurement

Standardization approaches:

For reliable cross-laboratory comparison, researchers should:

  • Include international standard VEGF preparations as reference

  • Express activity in International Units rather than mass units

  • Validate activity in multiple assay systems

  • Report detailed methodological parameters (passage number of cells, culture conditions, etc.)

For example, researchers investigating embryonic cell migration established a reliable quantification method where VEGFR2-positive cells were labeled with rhodamine, seeded on cover glass, and placed next to cells from anterior or posterior regions of embryos. Migration was then quantified after 16 hours, revealing that VEGFR-positive cells migrated specifically toward cells from the anterior portion due to abundant VEGF-A expression, and this migration could be blocked with neutralizing antibodies or receptor inhibitors .

How should researchers design mouse models to study specific VEGF-A functions while minimizing confounding variables?

Designing mouse models for VEGF-A research requires careful consideration to isolate specific functions while controlling for potential confounding factors:

Types of mouse models for VEGF-A research:

  • Genetic modification approaches:

    • Global knockout models (note: VEGF-A homozygous and even heterozygous knockouts are embryonic lethal)

    • Conditional knockout using tissue-specific Cre/loxP systems

    • Knock-in models with modified VEGF-A (e.g., humanized VEGF-A)

    • Isoform-specific models (e.g., mice expressing only single isoforms)

    • Reporter models (e.g., VEGF-GFP fusion proteins)

  • Humanized VEGF-A mouse models:

    • Particularly valuable for testing human-specific therapeutics

    • Created through gene replacement technology to express humanized VEGF-A (hum-X VEGF) recognized by anti-human VEGF antibodies

    • Useful for preclinical evaluation of anti-angiogenic therapies

Minimizing confounding variables:

  • Genetic background considerations:

    • Use isogenic backgrounds to reduce variability

    • Consider backcrossing to standard backgrounds (C57BL/6, BALB/c)

    • Include littermate controls whenever possible

  • Experimental controls:

    • Include both positive and negative controls

    • Use multiple control types (vehicle, isotype control antibodies)

    • Consider the use of contralateral control (e.g., untreated limb)

  • Environmental factors:

    • Standardize housing conditions (temperature, light cycles)

    • Control for age and sex differences

    • Maintain consistent diet (some diets can affect angiogenesis)

  • Analytical approaches:

    • Blind observers to experimental groups during analysis

    • Use multiple complementary analytical methods

    • Establish quantitative scoring systems

    • Apply appropriate statistical tests for sample size

The humanized VEGF-A mouse model described in the research demonstrates an effective approach to minimize confounding variables when testing anti-VEGF antibodies. These mice express VEGF-A that has "biochemical and biological properties comparable with WT mouse and human VEGF-A" while allowing for the binding of human-specific antibodies, thus enabling direct comparison between antibodies with different affinities for blocking host VEGF.

What are the key considerations for using His-tagged mouse VEGF-A in angiogenesis and tumor growth studies?

Using His-tagged mouse VEGF-A in angiogenesis and tumor studies requires attention to several critical factors:

Key experimental design considerations:

  • Protein activity verification:

    • Confirm biological activity post-purification

    • Verify that His-tag doesn't interfere with receptor binding

    • Compare activity with non-tagged VEGF-A

  • Delivery method optimization:

    • Systemic administration (intravenous, intraperitoneal)

    • Local delivery (intratumoral, subcutaneous)

    • Sustained release systems (osmotic pumps, biodegradable polymers)

    • Gene transfer approaches (viral vectors, plasmids)

  • Dosing considerations:

    • Establish dose-response relationships

    • Consider physiological versus pharmacological concentrations

    • Account for protein half-life in circulation

    • Determine frequency of administration

  • Tumor model selection:

    • Syngeneic versus xenograft models

    • Orthotopic versus heterotopic implantation

    • Consider VEGF dependence of specific tumor types

    • Account for endogenous VEGF production by tumor cells

Assessment parameters:

Parameter CategorySpecific MeasurementsTechniques
AngiogenesisMicrovessel densityImmunohistochemistry for CD31/PECAM-1
Vessel morphologyConfocal microscopy
Vascular permeabilityEvans blue extravasation
Perfused vessel functionalityLectin perfusion
Tumor responseGrowth kineticsCaliper measurements, imaging
Invasiveness/metastasisHistopathology, PCR detection
Hypoxia levelsPimonidazole staining, HIF-1α IHC
Molecular responsesVEGFR activationPhospho-specific antibodies
Downstream signalingWestern blotting, immunohistochemistry

Potential pitfalls and solutions:

  • Interference from endogenous VEGF: Use VEGF-deficient tumor cells or neutralizing antibodies

  • Heterodimer formation: Account for potential VEGF-A/VEGF-B heterodimers in analysis

  • Species specificity issues: Consider using humanized VEGF-A mice for testing human-specific therapies

  • Tumor heterogeneity: Use multiple tumor models and assessment methods

Research has demonstrated that when studying anti-VEGF antibodies, there may not be a "consistent correlation between antibody affinity and the ability to inhibit tumor growth and angiogenesis in most animal models" , highlighting the complexity of these systems and the need for comprehensive experimental design.

How can researchers effectively use His-tagged mouse VEGF-A to study embryonic development and cell migration?

His-tagged mouse VEGF-A is a valuable tool for investigating embryonic vascular development and cell migration processes, but requires specific methodological approaches:

Embryonic development studies:

  • Ex vivo whole embryo culture systems:

    • Allow manipulation of VEGF signaling in intact embryos

    • Enable time-lapse imaging of vascular development

    • Permit application of recombinant proteins or inhibitors

    • Support assessment of cell migration in physiological context

  • In vivo approaches:

    • Conditional genetic manipulation at specific developmental stages

    • Microinjection of His-tagged VEGF-A into specific embryonic regions

    • Chimeric embryo analysis (wild-type/knockout combinations)

    • Lineage tracing of VEGFR-positive cells

Cell migration assay methodologies:

  • In vitro migration systems:

    • Soft agar gel assays with defined VEGF-A gradients (0-100 ng/ml)

    • Transwell migration assays

    • Microfluidic gradient chambers

    • Time-lapse microscopy tracking

  • Ex vivo approaches:

    • Isolation of VEGFR2-positive cells from embryos

    • Rhodamine labeling for tracking migration

    • Co-culture with cells from different embryonic regions

    • Analysis of directional migration toward VEGF sources

Technical optimizations:

For embryonic cell migration studies using His-tagged VEGF-A:

  • Isolate VEGFR2-positive cells from embryos at E7.5 without heads

  • Label cells with fluorescent markers for tracking

  • Seed cells adjacent to potential VEGF-A source cells

  • Include controls with neutralizing antibodies or receptor inhibitors

  • Quantify migration distance and directionality over time (16-24 hours)

This approach has revealed that "VEGFR-positive cells migrated toward cells obtained from the anterior portion of embryos, and this migration was eliminated by a neutralizing anti-mouse VEGF-A antibody" . The migration could also be blocked by VEGFR kinase inhibitors (SU5416) or soluble VEGFR1, confirming the specificity of the VEGF-A-directed migration.

These techniques have demonstrated that VEGF-A protein concentrated in the anterior region plays an important role in guiding VEGFR-positive cells from the posterior portion to the head region during embryonic development .

What are effective strategies for comparing the effects of different VEGF family members (VEGF-A, VEGF-B, PlGF) using His-tagged recombinant proteins?

Comparing different VEGF family members requires careful experimental design to accurately assess their distinct and overlapping functions:

Comparative study design approaches:

  • Parallel protein characterization:

    • Standardize expression and purification protocols across proteins

    • Verify protein quality (purity, aggregation state, tag position)

    • Characterize receptor binding profiles for each protein

    • Determine protein stability under experimental conditions

  • Receptor binding analysis:

    • Compare binding affinities to VEGFR1, VEGFR2, and neuropilin-1

    • Conduct competition binding assays between family members

    • Assess receptor dimerization patterns induced by each ligand

    • Evaluate binding to soluble receptor forms

  • Functional comparison assays:

    • Cell proliferation in multiple endothelial cell types

    • Migration assays with varying concentration gradients

    • Survival assays under stress conditions

    • Tube formation capacity

Experimental design considerations:

ParameterRecommendationRationale
Concentration rangeTest wide range (1-100 ng/ml)Captures threshold and saturation effects
Cell typesUse multiple endothelial cell sourcesAccounts for heterogeneity in receptor expression
Combination testingTest proteins individually and in combinationReveals additive, synergistic, or antagonistic effects
Time courseAssess both acute and chronic responsesDistinguishes between immediate and adaptive effects

Methodological approaches for combination studies:

When testing combinations of VEGF family members:

  • Use equimolar concentrations for direct comparison

  • Test varying ratios to model physiological contexts

  • Include appropriate controls for each protein individually

  • Consider potential heterodimer formation

Research has found that when combining PlGF with VEGF-A in migration assays, "at a low concentration (10 ng/ml) of VEGF-A, the scores were partially additive (30% increase). At higher doses of VEGF-A (40 and 100 ng/ml), additive effects were hardly detected" . Similarly, when testing PlGF with VEGF-E, only minor additive effects were observed at high doses.

The distinct roles of different family members are highlighted by knockout studies: "PlGF null mutant mice were reported to have no clear abnormalities during the early embryonic stage" whereas VEGF-A knockouts are embryonic lethal. Similarly, VEGF-B knockout mice (Vegfb−/−) "are healthy and fertile" unlike Vegfa knockout mice, but show "reduced size and display vascular dysfunction after coronary occlusion and impaired recovery from experimentally induced myocardial ischemia" .

What are common technical challenges when working with His-tagged mouse VEGF-A and how can they be overcome?

Working with His-tagged mouse VEGF-A presents several technical challenges that can affect experimental outcomes. Recognizing and addressing these issues is essential for reliable results:

Challenge 1: Protein solubility and aggregation

Problem: Recombinant VEGF-A may form aggregates during storage or upon dilution in experimental buffers.

Solutions:

  • Use buffers containing 10% glycerol to improve stability

  • Maintain pH between 7.5-8.5 (typically 20 mM Tris-HCl, pH 8.0)

  • Centrifuge solutions briefly before use to remove potential aggregates

  • Consider adding carrier protein (0.1% BSA) to dilute solutions

  • Filter through 0.22 μm filters immediately before use in cell culture

Challenge 2: His-tag interference with function

Problem: The His-tag may occasionally interfere with protein folding or receptor binding.

Solutions:

  • Compare activity with commercial non-tagged VEGF-A as reference

  • Consider using cleavable His-tags with specific protease sites

  • Test both N-terminal and C-terminal His-tagged versions

  • Verify receptor binding capability using surface plasmon resonance

  • Include proper controls in all functional assays

Challenge 3: Batch-to-batch variability

Problem: Different protein preparations may show variable activity levels.

Solutions:

  • Standardize purification protocols rigidly

  • Quantify specific activity for each batch using bioassays

  • Create internal reference standards for normalization

  • Use multiple complementary activity assays

  • Document and track lot numbers with corresponding activity data

Challenge 4: Short half-life in experimental systems

Problem: VEGF-A can be rapidly degraded or sequestered in biological systems.

Solutions:

  • Monitor protein stability over time in experimental media

  • Consider pulsed administration or continuous perfusion

  • Use protease inhibitors when appropriate

  • Quantify VEGF-A levels at multiple timepoints during experiments

  • Consider using sustained-release formulations for in vivo applications

Challenge 5: Endotoxin contamination

Problem: E. coli-derived proteins may contain endotoxin that affects cell responses.

Solutions:

  • Use endotoxin-free purification systems

  • Test each batch with Limulus Amebocyte Lysate (LAL) assay

  • Include polymyxin B or endotoxin removal steps in purification

  • Use endotoxin-resistant cell lines for certain experiments

  • Include endotoxin-matched controls in experiments

Researchers have successfully addressed these challenges by implementing rigorous quality control protocols and standardized handling procedures, enabling reliable experiments with His-tagged mouse VEGF-A across various applications from in vitro cell assays to complex in vivo models .

How can researchers accurately interpret conflicting results between different VEGF-A assay systems?

Conflicting results between different VEGF-A assay systems are common and require systematic analysis to resolve:

Sources of conflicting results:

  • Assay-specific sensitivities:

    • Different cell types used (HUVECs vs. mouse endothelial cells)

    • Varied endpoints (proliferation vs. migration vs. survival)

    • Distinct detection methods (direct vs. indirect measurements)

  • Technical variables:

    • Serum components affecting VEGF-A activity

    • Substrate differences (2D plastic vs. 3D matrices)

    • Presence of co-factors or inhibitors in specific systems

    • Receptor expression levels across cell types

  • Biological context:

    • Acute vs. chronic responses to VEGF-A

    • Simple vs. complex cellular environments

    • Presence of compensatory mechanisms in some systems

Systematic resolution approach:

  • Comparative analysis workflow:

    • Perform side-by-side testing of multiple assays

    • Use identical protein preparations across systems

    • Implement standardized positive and negative controls

    • Generate complete dose-response curves for each system

  • Reconciliation strategies:

    • Identify assay-specific thresholds and saturation points

    • Determine if conflicts are quantitative or qualitative

    • Test whether conflicts are concentration-dependent

    • Assess time-dependency of observed effects

  • Integrative interpretation:

    • Prioritize functional over proxy readouts

    • Consider the biological relevance of each assay system

    • Develop integrative models that account for system-specific factors

    • When possible, validate in vivo to resolve in vitro conflicts

This systematic approach is illustrated by research comparing in vitro and in vivo effects of anti-VEGF antibodies, where "in vitro studies clearly showed a correlation between binding affinity and potency at blocking endothelial cell proliferation stimulated by VEGF," but "in vivo experiments failed to document any consistent correlation between antibody affinity and the ability to inhibit tumor growth and angiogenesis in most animal models" . This apparent contradiction was resolved through comprehensive testing in humanized VEGF-A mouse models, revealing that higher-affinity antibodies showed increased risk of side effects (glomerulosclerosis) despite similar efficacy .

What are the best methods for validating the specificity of observed effects when using His-tagged mouse VEGF-A?

Validating the specificity of observed effects is crucial when working with His-tagged mouse VEGF-A to ensure that results truly reflect VEGF-A biology rather than experimental artifacts:

Validation strategies:

  • Control protein comparisons:

    • Non-tagged VEGF-A (gold standard positive control)

    • Denatured His-tagged VEGF-A (structure-dependent control)

    • Irrelevant His-tagged protein (tag-specific control)

    • Other VEGF family members (specificity control)

  • Blocking approaches:

    • Neutralizing antibodies against VEGF-A

    • Soluble VEGF receptors (VEGFR1/R2)

    • Small molecule VEGFR inhibitors (e.g., SU5416)

    • Receptor-blocking antibodies

  • Genetic validation:

    • VEGFR1/R2 knockdown or knockout systems

    • Signaling pathway component inhibition

    • Dominant negative receptor expression

    • CRISPR/Cas9 receptor modification

  • Signal transduction verification:

    • Demonstrate receptor phosphorylation

    • Show activation of canonical downstream pathways

    • Inhibit specific signaling nodes (PI3K, MAPK, etc.)

    • Perform comprehensive pathway profiling

Example validation protocol for cell migration:

  • Demonstrate dose-dependent migration toward His-tagged VEGF-A

  • Show absence of migration toward control proteins

  • Block migration using anti-VEGF-A antibodies

  • Inhibit migration with receptor tyrosine kinase inhibitors

  • Show migration can be blocked by soluble VEGFR1

  • Verify receptor phosphorylation correlates with migration

Research has successfully employed these validation approaches, demonstrating that VEGFR-positive cells migrated toward cells obtained from the anterior portion of embryos (which express high levels of VEGF-A), and this migration was specifically eliminated by a neutralizing anti-mouse VEGF-A antibody . Further validation showed the migration could also be blocked by applying SU5416 (a VEGFR inhibitor) or soluble VEGFR1, confirming the effect was specifically mediated by VEGF-A binding to its receptors .

Product Science Overview

Introduction

Vascular Endothelial Growth Factor (VEGF) is a potent mediator of angiogenesis and vasculogenesis, playing a crucial role in both embryonic development and adult physiology. The recombinant form of VEGF, specifically the mouse variant with a His tag, is widely used in research to study its biological functions and therapeutic potential.

Structure and Expression

The recombinant mouse VEGF protein with a His tag is typically expressed in HEK 293 cells, ensuring high purity and biological activity. The His tag, a sequence of histidine residues, facilitates the purification of the protein through affinity chromatography. This recombinant protein is often produced with a purity greater than 95% and an endotoxin level of less than 1 EU/µg, making it suitable for various experimental applications .

Biological Functions

VEGF is essential for the proliferation, migration, and survival of endothelial cells. It binds to specific receptors on the surface of these cells, such as VEGFR-2 (Flk-1/KDR) and VEGFR-3 (Flt4), triggering a cascade of signaling events that promote angiogenesis. In addition to its role in blood vessel formation, VEGF also increases vascular permeability, which is critical during wound healing and in pathological conditions like tumor growth .

Applications in Research

Recombinant mouse VEGF with a His tag is used in a variety of research applications, including:

  • Angiogenesis Studies: Investigating the mechanisms of blood vessel formation and identifying potential therapeutic targets for diseases characterized by abnormal angiogenesis, such as cancer and diabetic retinopathy.
  • Drug Screening: Evaluating the efficacy of anti-angiogenic drugs that target VEGF signaling pathways.
  • Cell Culture: Supplementing cell culture media to study the effects of VEGF on endothelial cell behavior and function.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.