VEGF Rat, His refers to recombinant vascular endothelial growth factor-A (VEGF-A) derived from rat species, engineered with a histidine (His) tag for purification and structural stability. This protein is a critical mediator of angiogenesis, vascular permeability, and endothelial cell proliferation, with applications in research and therapeutic development. Below is a detailed analysis of its structure, function, and research implications.
VEGF Rat, His is a homodimeric glycoprotein with two subunits linked by disulfide bonds. Key features include:
Cysteine-knot motif: Eight conserved cysteine residues form three intramolecular disulfide bonds and two intermolecular bonds, stabilizing the dimer structure .
His-tag: A polyhistidine sequence (typically 6xHis) is appended to the N-terminal or C-terminal region for affinity chromatography purification .
Isoforms: Rat VEGF-A exists in spliced isoforms (e.g., 120, 164, 188 amino acids). The 164-amino acid isoform (VEGF-164) is the most abundant and biologically active variant .
Property | VEGF Rat, His (VEGF-164) |
---|---|
Molecular Weight (monomer) | ~19.4 kDa |
Dimeric Form | 38.7 kDa |
Receptors | VEGFR-1 (Flt-1), VEGFR-2 (Flk-1/KDR) |
Species Homology | 97% (mouse), 88% (human) |
VEGF Rat, His regulates endothelial cell behavior and vascular development through:
Angiogenesis: Promotes endothelial cell proliferation, migration, and tube formation via VEGFR-2 signaling .
Vascular Permeability: Induces blood vessel leakage, a hallmark of tumor progression and inflammation .
Neuroprotection and Neurogenesis: Enhances neural survival and progenitor cell migration in injury models .
VEGF Rat, His is pivotal in cancer research:
Tumor Growth: VEGF-A binds VEGFR-2, driving endothelial cell recruitment and tumor vasculature formation. Inhibition of VEGFR-1 (Flt-1) reduces metastasis in rat models .
Xenograft Studies: Roxarsone-induced tumor growth in B16F10 mouse xenografts is mediated by VEGF/Flk-1 signaling, as shown by RNA interference and antibody blockade .
Spinal Cord Injury (SCI):
Traumatic Brain Injury (TBI): VEGF-A administration worsens outcomes in male rats, increasing cognitive deficits and neuroinflammation markers (CCL5, EPO) .
Kidney Regeneration: VEGF expression correlates with renal tissue repair in metanephroi-transplanted rats, promoting angiogenesis and cell proliferation .
Cardiac Hypertrophy: VEGF-B (a related isoform) enhances coronary vasculature and protects against ischemia, though VEGF-A’s role in cardiac remodeling remains under investigation .
Rat VEGF exists in multiple isoforms, with VEGF164 being predominant in research settings. The rat VEGF164 protein consists of amino acids Ala27-Arg190 and features different binding properties compared to other isoforms. Western blot analyses show that VEGF164 can be detected as both homodimeric and monomeric forms at approximately 54 kDa and 24 kDa, respectively, under reducing conditions . The structural variations between isoforms directly impact their biological activities, with VEGF164 showing higher binding affinity to VEGFR2/Flk1 receptors compared to shorter isoforms.
While structurally similar, rat VEGF164 and human VEGF165 exhibit important species-specific differences. Cross-reactivity studies using direct ELISAs demonstrate approximately 20% cross-reactivity between anti-rat VEGF antibodies and recombinant human VEGF165 and VEGF121 . Despite this partial cross-reactivity, functional studies show that rat VEGF164 has an ED50 of 0.75-3.75 ng/mL for stimulating proliferation in human umbilical vein endothelial cells (HUVECs) , which differs from human VEGF165. These differences must be considered when designing cross-species experiments or when translating findings from rat models to human applications.
The VEGF/Flk1 (VEGFR2) pathway plays a crucial role in rat angiogenesis and endothelial cell function. Research indicates that this pathway mediates roxarsone promotion of rat vascular endothelial cell proliferation, migration, and tube-like formation in vitro, as well as tumor growth and angiogenesis in mouse xenograft models . Blocking experiments using Flk1 antibodies significantly reduce cell viability compared to Flt1 (VEGFR1) antibody treatments (p<0.01), indicating the primary importance of VEGFR2/Flk1 signaling . Small interfering RNA (siRNA) targeting Flk1 significantly attenuates these promotion effects, confirming the mechanistic importance of this receptor in VEGF-mediated processes.
For optimal Western blot detection of rat VEGF164, the following protocol yields consistent results:
Use PVDF membrane for protein transfer
Probe with 0.1 μg/mL of anti-rat VEGF164 antibody (e.g., Goat Anti-Rat VEGF Antigen Affinity-purified Polyclonal Antibody)
Follow with HRP-conjugated secondary antibody
Conduct experiments under reducing conditions using appropriate buffer systems
Table 1: Comparison of Detection Methods for Rat VEGF164
Note that this antibody typically does not detect natural VEGF in lysates from cell lines or tissues, which is an important limitation to consider when designing experiments.
For optimal immunohistochemical detection of rat VEGF in tissue sections:
Use perfusion-fixed frozen sections rather than paraffin-embedded samples for better epitope preservation
Apply 15 μg/mL of anti-rat VEGF164 antibody and incubate overnight at 4°C
Utilize appropriate HRP-DAB staining systems with hematoxylin counterstaining
This methodology has been successfully employed to detect VEGF164 in rat kidney tissues, revealing specific expression patterns in the renal microvasculature and tubular epithelial cells. For quantitative assessment of expression, semiquantitative analysis using standardized scoring systems has been validated in multiple studies examining VEGF expression in models of kidney regeneration and pathology .
To reliably assess VEGF functionality in rat cell systems, the cell proliferation assay using HUVECs represents the gold standard approach:
Treat HUVEC cells with varying concentrations of recombinant rat VEGF164 (typically 0-50 ng/mL)
Measure proliferation after 24-72 hours using appropriate proliferation assays
For neutralization studies, include anti-VEGF antibodies at concentrations of 0.2-0.6 μg/mL in the presence of 20 ng/mL rat VEGF164
The effective dose for 50% stimulation (ED50) typically falls between 0.75-3.75 ng/mL for rat VEGF164 . Additional functional assays include endothelial cell migration (Boyden chamber assay) and tube formation on Matrigel, both of which provide complementary information about VEGF's angiogenic properties.
When using His-tagged rat VEGF in functional assays, it is advisable to:
Compare activity with non-tagged variants in dose-response experiments
Verify receptor binding using surface plasmon resonance or other binding assays
Assess dimerization status, as proper VEGF dimerization is essential for biological activity
For optimal purification of His-tagged rat VEGF:
Express the protein in appropriate expression systems (mammalian cells preferred for proper glycosylation)
Utilize immobilized metal affinity chromatography (IMAC) with Ni-NTA or Co-based resins
Apply a step-gradient elution protocol with imidazole (20-250 mM)
Include additional purification steps such as size exclusion chromatography to ensure dimeric VEGF isolation
Verify purity by SDS-PAGE under both reducing and non-reducing conditions
Table 2: Purification Strategy Comparison for His-Tagged Rat VEGF
Purification Method | Advantages | Disadvantages | Yield | Purity |
---|---|---|---|---|
Ni-NTA IMAC | High binding capacity | Potential metal leaching | 2-5 mg/L culture | 80-90% |
Co-based IMAC | Higher specificity | Lower capacity | 1-3 mg/L culture | 90-95% |
IMAC + Size Exclusion | Separates dimers from aggregates | Time-consuming | 0.5-2 mg/L culture | >95% |
The purified protein should be validated for endotoxin levels (<0.1 EU/μg protein) and bioactivity in HUVEC proliferation assays before use in critical experiments.
In rat models of ischemia, VEGF expression undergoes significant temporal and spatial changes. Studies of brain ischemia have shown that Vegfa mRNA is upregulated in both cortical and subcortical regions of the ipsilateral (ischemic) hemisphere compared to the contralateral side . This upregulation is associated with increased vascular permeability, as evidenced by Evans blue extravasation studies.
Mechanistically, the ischemia-induced VEGF expression is mediated by:
Hypoxia-inducible factor 1α (HIF-1α) activation
Brain-associated macrophages (BAMs), as their depletion attenuates ischemia-induced Vegfa mRNA expression
Inflammatory cytokine signaling cascades
Western blot analyses reveal that VEGF164 can be detected as both homodimeric and monomeric forms at approximately 54 kDa and 24 kDa in ischemic brain tissue . Targeting these mechanisms provides potential therapeutic opportunities for modulating post-ischemic angiogenesis and vascular repair.
VEGF plays a crucial role in kidney regeneration in rat models. Studies using male Munich-Wistar Frömter (MWF) rats have demonstrated that metanephroi (MET) transplantation significantly increases VEGF expression in renal tissues compared to saline controls, as shown by both Real-Time RT-PCR and immunohistochemistry . This increased expression correlates with improved renal function and structure.
Key findings from these studies include:
Upregulation of multiple growth factors (VEGF, FGF2, HGF, IGF-1) following MET transplantation
Sex-specific differences in VEGF expression patterns
Correlation between VEGF levels and functional recovery measures
Therapeutic modulation strategies include:
Direct administration of recombinant rat VEGF164
Transplantation of cells engineered to overexpress VEGF
Use of compounds that stabilize HIF-1α to indirectly increase VEGF production
Targeted delivery systems to maximize local effects while minimizing systemic exposure
VEGF signaling and inflammatory processes exhibit complex bidirectional interactions in rat disease models. Research demonstrates that VEGF can both promote and inhibit inflammation depending on the context, timing, and dosage. In spinal cord injury models, VEGF acts as a key mediator leading to different recovery levels in neonatal versus adult rats by regulating inflammatory responses, protecting damaged neurons, and promoting reestablishment of spinal neural circuits .
Evidence suggests that:
Low-dose VEGF treatment can reduce inflammatory cytokine production
VEGF protects neurons from inflammation-induced apoptosis
The VEGF/Flk1 pathway modulates microglial activation and phenotype switching
Importantly, the anti-inflammatory effects of VEGF appear to be receptor-specific, with Flk1 (VEGFR2) signaling mediating protective effects while Flt1 (VEGFR1) may promote inflammatory responses in certain contexts. These findings highlight the potential for receptor-specific targeting approaches in therapeutic development.
Studying VEGF in complex rat tissue microenvironments presents several challenges that can be addressed through:
Multi-scale imaging approaches:
Combine immunohistochemistry with high-resolution confocal microscopy
Implement tissue clearing techniques (CLARITY, iDISCO) for 3D visualization
Use multiplexed immunofluorescence to simultaneously visualize VEGF, receptors, and cellular markers
Cell-specific analyses:
Employ laser capture microdissection to isolate specific cell populations
Utilize single-cell RNA sequencing to characterize heterogeneous VEGF expression
Develop cell type-specific conditional expression systems
Dynamic monitoring:
Implement in vivo microscopy with fluorescently labeled VEGF variants
Use DCE-MRI to monitor vascular responses to VEGF in real-time
Develop biosensor systems for continuous VEGF activity monitoring
These approaches have successfully addressed the limitations of conventional techniques in capturing the dynamic and spatially complex nature of VEGF signaling in various rat disease models.
Contradictory findings regarding VEGF effects across different rat experimental models can be reconciled through:
Systematic comparison of experimental variables:
Standardize dose, timing, and duration of VEGF administration
Consider rat strain differences (Sprague-Dawley vs. Wistar vs. specialized disease models)
Account for age and sex differences in VEGF responsiveness
Receptor-specific analysis:
Differentiate between Flk1 (VEGFR2) and Flt1 (VEGFR1) mediated effects
Consider co-receptor (neuropilins, heparan sulfate proteoglycans) involvement
Analyze isoform-specific effects (VEGF120 vs. VEGF164 vs. VEGF188)
Context-dependent interpretation:
Recognize that VEGF effects may be biphasic (beneficial at low doses, detrimental at high doses)
Consider tissue microenvironment differences (inflammatory status, hypoxia levels)
Integrate temporal dynamics into experimental design and analysis
For example, studies have shown that red wine polyphenols exert dose-dependent effects on VEGF-mediated angiogenesis in rats, with low doses being proangiogenic and high doses anti-angiogenic , illustrating the importance of dosage considerations in experimental design.
Emerging methodologies for studying VEGF signaling dynamics in rat models include:
Advanced imaging techniques:
Dynamic contrast-enhanced (DCE) and diffusion-weighted (DW) MRI for non-invasive monitoring of vascular responses to anti-VEGF treatments in rat glioma models
Intravital multiphoton microscopy for real-time visualization of VEGF-induced vascular changes
Super-resolution microscopy for nanoscale receptor clustering analysis
Genetic engineering approaches:
CRISPR/Cas9-mediated gene editing for isoform-specific VEGF modifications
Optogenetic control of VEGF release for precise spatiotemporal signaling studies
Conditional knockout systems for cell-specific VEGF or receptor deletion
High-throughput analytical methods:
Phosphoproteomics to map complete VEGF signaling networks
Spatial transcriptomics to visualize VEGF expression patterns while preserving tissue architecture
AI-assisted image analysis for quantifying complex vascular patterns
These technologies are enabling unprecedented insights into the spatial and temporal dynamics of VEGF signaling in physiological and pathological conditions, moving beyond static endpoint measurements to capture the dynamic nature of angiogenic processes.
Vascular Endothelial Growth Factor (VEGF) is a signal protein that plays a crucial role in the formation of blood vessels. It is part of the platelet-derived growth factor family and is involved in both vasculogenesis (the formation of the embryonic circulatory system) and angiogenesis (the growth of blood vessels from pre-existing vasculature) . VEGF is essential for restoring oxygen supply to tissues when blood circulation is inadequate, such as in hypoxic conditions .
The rat recombinant VEGF, specifically with a His tag, is a version of this protein that has been engineered for research purposes. This recombinant protein is produced in insect cells and contains 129 amino acid residues. It is fused to a His-tag (6x His) at the C-terminal end, which facilitates its purification and detection . Due to glycosylation, VEGF-C migrates as an 18-24 kDa protein in SDS-PAGE under reducing conditions .
VEGF is a glycosylated mitogen that specifically acts on endothelial cells. It has various effects, including:
These properties make VEGF a critical factor in both normal physiological processes and pathological conditions. For instance, it is involved in creating new blood vessels during embryonic development, after injury, and in muscles following exercise . Additionally, VEGF plays a role in diseases such as cancer, where it can contribute to tumor growth and metastasis by promoting blood vessel formation .
The recombinant form of VEGF with a His tag is widely used in research to study its functions and interactions. It is also used in developing therapeutic strategies for diseases involving abnormal blood vessel growth. For example, drugs that inhibit VEGF, such as bevacizumab and ranibizumab, are used to treat cancers and retinal diseases .